1
|
Wang F, Zhang X, Tang L, Wu Q, Cai M, Li Y, Qu X, Qiu H, Zhang Y, Ying J, Zhang J, Sun L, Lin R, Wang C, Liu H, Qiu M, Guan W, Rao S, Ji J, Xin Y, Sheng W, Xu H, Zhou Z, Zhou A, Jin J, Yuan X, Bi F, Liu T, Liang H, Zhang Y, Li G, Liang J, Liu B, Shen L, Li J, Xu R. The Chinese Society of Clinical Oncology (CSCO): Clinical guidelines for the diagnosis and treatment of gastric cancer, 2023. Cancer Commun (Lond) 2024; 44:127-172. [PMID: 38160327 PMCID: PMC10794017 DOI: 10.1002/cac2.12516] [Citation(s) in RCA: 75] [Impact Index Per Article: 75.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 12/17/2023] [Accepted: 12/18/2023] [Indexed: 01/03/2024] Open
Abstract
The 2023 update of the Chinese Society of Clinical Oncology (CSCO) Clinical Guidelines for Gastric Cancer focuses on standardizing cancer diagnosis and treatment in China, reflecting the latest advancements in evidence-based medicine, healthcare resource availability, and precision medicine. These updates address the differences in epidemiological characteristics, clinicopathological features, tumor biology, treatment patterns, and drug selections between Eastern and Western gastric cancer patients. Key revisions include a structured template for imaging diagnosis reports, updated standards for molecular marker testing in pathological diagnosis, and an elevated recommendation for neoadjuvant chemotherapy in stage III gastric cancer. For advanced metastatic gastric cancer, the guidelines introduce new recommendations for immunotherapy, anti-angiogenic therapy and targeted drugs, along with updated management strategies for human epidermal growth factor receptor 2 (HER2)-positive and deficient DNA mismatch repair (dMMR)/microsatellite instability-high (MSI-H) patients. Additionally, the guidelines offer detailed screening recommendations for hereditary gastric cancer and an appendix listing drug treatment regimens for various stages of gastric cancer. The 2023 CSCO Clinical Guidelines for Gastric Cancer updates are based on both Chinese and international clinical research and expert consensus to enhance their applicability and relevance in clinical practice, particularly in the heterogeneous healthcare landscape of China, while maintaining a commitment to scientific rigor, impartiality, and timely revisions.
Collapse
Affiliation(s)
- Feng‐Hua Wang
- Department of Medical OncologySun Yat‐sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer MedicineGuangzhouGuangdongP. R. China
| | - Xiao‐Tian Zhang
- Department of Gastrointestinal OncologyKey Laboratory of Carcinogenesis and Translational Research (Ministry of Education)Peking University Cancer HospitalBeijingP. R. China
| | - Lei Tang
- Department of RadiologyPeking University Cancer HospitalBeijingP. R. China
| | - Qi Wu
- Department of Endoscopy CenterPeking University Cancer HospitalBeijingP. R. China
| | - Mu‐Yan Cai
- Department of PathologySun Yat‐sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center of Cancer MedicineGuangzhouGuangdongP. R. China
| | - Yuan‐Fang Li
- Department of Gastric SurgerySun Yat‐sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer MedicineGuangzhouGuangdongP. R. China
| | - Xiu‐Juan Qu
- Department of Medical OncologyThe First Hospital of China Medical UniversityShenyangLiaoningP. R. China
| | - Hong Qiu
- Department of Medical OncologyTongji Hospital Affiliated to Tongji Medical College of Huazhong University of Science and TechnologyWuhanHubeiP. R. China
| | - Yu‐Jing Zhang
- Department of RadiotherapySun Yat‐sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer MedicineGuangzhouGuangdongP. R. China
| | - Jie‐Er Ying
- Department of Medical OncologyZhejiang Cancer HospitalHangzhouZhejiangP. R. China
| | - Jun Zhang
- Department of Medical OncologyRuijin HospitalShanghai Jiaotong University School of MedicineShanghaiP. R. China
| | - Ling‐Yu Sun
- Department of Surgical OncologyThe Fourth Affiliated Hospital of Harbin Medical UniversityHarbinHeilongjiangP. R. China
| | - Rong‐Bo Lin
- Department of Medical OncologyFujian Cancer HospitalFuzhouFujianP. R. China
| | - Chang Wang
- Tumor CenterThe First Hospital of Jilin UniversityChangchunJilinP. R. China
| | - Hao Liu
- Department of General SurgeryNanfang HospitalSouthern Medical UniversityGuangzhouGuangdongP. R. China
| | - Miao‐Zhen Qiu
- Department of Medical OncologySun Yat‐sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer MedicineGuangzhouGuangdongP. R. China
| | - Wen‐Long Guan
- Department of Medical OncologySun Yat‐sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer MedicineGuangzhouGuangdongP. R. China
| | - Sheng‐Xiang Rao
- Department of RadiologyZhongshan HospitalFudan UniversityShanghaiP. R. China
| | - Jia‐Fu Ji
- Department of Gastrointestinal SurgeryPeking University Cancer HospitalBeijingP. R. China
| | - Yan Xin
- Pathology Laboratory of Gastrointestinal TumorThe First Hospital of China Medical UniversityShenyangLiaoningP. R. China
| | - Wei‐Qi Sheng
- Department of PathologyZhongshan Hospital Affiliated to Shanghai Fudan UniversityShanghaiP. R. China
| | - Hui‐Mian Xu
- Department of Gastrointestinal Oncology Surgery. The First Hospital of China Medical UniversityShenyangLiaoningP. R. China
| | - Zhi‐Wei Zhou
- Department of Gastric SurgerySun Yat‐sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center of Cancer MedicineGuangzhouGuangdongP. R. China
| | - Ai‐Ping Zhou
- Department of OncologyNational Cancer CenterNational Clinical Research Center for CancerCancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingP. R. China
| | - Jing Jin
- Department of Radiation OncologyShenzhen hospitalCancer Hospital of Chinese Academy of Medical SciencesBeijingP. R. China
| | - Xiang‐Lin Yuan
- Department of OncologyTongji Hospital Affiliated to Tongji Medical College of Huazhong University of Science and TechnologyWuhanHubeiP. R. China
| | - Feng Bi
- Department of Abdominal OncologyWest China Hospital of Sichuan UniversityChengduSichuanP. R. China
| | - Tian‐Shu Liu
- Department of Medical OncologyZhongshan Hospital Affiliated to Fudan UniversityShanghaiP. R. China
| | - Han Liang
- Department of Gastric SurgeryTianjin Medical University Cancer Institute & HospitalTianjinP. R. China
| | - Yan‐Qiao Zhang
- Department of Medical OncologyCancer Hospital of Harbin Medical UniversityHarbinHeilongjiangP. R. China
| | - Guo‐Xin Li
- Department of General SurgeryNanfang HospitalSouthern Medical UniversityGuangzhouGuangdongP. R. China
| | - Jun Liang
- Department of Medical OncologyPeking University International HospitalBeijingP. R. China
| | - Bao‐Rui Liu
- Department of Medical OncologyNanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical SchoolNanjingP. R. China
| | - Lin Shen
- Department of GI OncologyKey Laboratory of Carcinogenesis and Translational Research (Ministry of Education)Peking University Cancer HospitalBeijingP. R. China
| | - Jin Li
- Department of OncologyEaster Hospital affiliated to Shanghai Tongji UniversityShanghaiP. R. China
| | - Rui‐Hua Xu
- Department of Medical OncologySun Yat‐sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer MedicineGuangzhouGuangdongP. R. China
| |
Collapse
|
2
|
Saito A, Kitayama J, Nagai R, Aizawa K. Anatomical Targeting of Anticancer Drugs to Solid Tumors Using Specific Administration Routes: Review. Pharmaceutics 2023; 15:1664. [PMID: 37376112 DOI: 10.3390/pharmaceutics15061664] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 06/02/2023] [Accepted: 06/04/2023] [Indexed: 06/29/2023] Open
Abstract
Despite remarkable recent progress in developing anti-cancer agents, outcomes of patients with solid tumors remain unsatisfactory. In general, anti-cancer drugs are systemically administered through peripheral veins and delivered throughout the body. The major problem with systemic chemotherapy is insufficient uptake of intravenous (IV) drugs by targeted tumor tissue. Although dose escalation and treatment intensification have been attempted in order to increase regional concentrations of anti-tumor drugs, these approaches have produced only marginal benefits in terms of patient outcomes, while often damaging healthy organs. To overcome this problem, local administration of anti-cancer agents can yield markedly higher drug concentrations in tumor tissue with less systemic toxicity. This strategy is most commonly used for liver and brain tumors, as well as pleural or peritoneal malignancies. Although the concept is theoretically reasonable, survival benefits are still limited. This review summarizes clinical results and problems and discusses future directions of regional cancer therapy with local administration of chemotherapeutants.
Collapse
Affiliation(s)
- Akira Saito
- Department of Surgery, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi 329-0431, Japan
| | - Joji Kitayama
- Department of Surgery, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi 329-0431, Japan
- Division of Translational Research, Clinical Research Center, Jichi Medical University Hospital, Tochigi, Tochigi 329-0498, Japan
| | - Ryozo Nagai
- Department of Medicine, School of Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi 329-0498, Japan
| | - Kenichi Aizawa
- Division of Translational Research, Clinical Research Center, Jichi Medical University Hospital, Tochigi, Tochigi 329-0498, Japan
- Division of Clinical Pharmacology, Department of Pharmacology, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi 329-0498, Japan
| |
Collapse
|
3
|
Graversen M, Rouvelas I, Ainsworth AP, Bjarnesen AP, Detlefsen S, Ellebaek SB, Fristrup CW, Liljefors MG, Lundell L, Nilsson M, Pfeiffer P, Tarpgaard LS, Tsekrekos A, Mortensen MB. Feasibility and Safety of Laparoscopic D2 Gastrectomy in Combination with Pressurized Intraperitoneal Aerosol Chemotherapy (PIPAC) in Patients with Gastric Cancer at High Risk of Recurrence-The PIPAC-OPC4 Study. Ann Surg Oncol 2023. [PMID: 36867174 DOI: 10.1245/s10434-023-13278-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/04/2023]
Abstract
BACKGROUND Patients with gastric adenocarcinoma (GAC) are at high risk of peritoneal recurrence despite perioperative chemotherapy and radical resection. This study evaluated feasibility and safety of laparoscopic D2 gastrectomy in combination with pressurized intraperitoneal aerosol chemotherapy (PIPAC). METHODS This was a prospective, controlled bi-institutional study in patients with GAC at high risk of recurrence treated with PIPAC with cisplatin and doxorubicin (PIPAC C/D) after laparoscopic D2 gastrectomy. High risk was defined as a poorly cohesive subtype with predominance of signet-ring cells, clinical stage ≥ T3 and/or ≥ N2, or positive peritoneal cytology. Peritoneal lavage fluid was collected before and after resection. Cisplatin (10.5 mg/m2) and doxorubicin (2.1 mg/m2) were aerosolized after anastomosis (flow 0.5-0.8 ml/s, maximum pressure 300 PSI). Treatment was feasible and safe if ≤ 20% had Dindo-Clavien ≥ 3b surgical complications or CTCAE ≥ 4 medical adverse events within 30 days. Secondary outcomes were length of stay (LOS), peritoneal lavage cytology, and completion of postoperative systemic chemotherapy. RESULTS Twenty-one patients were treated with a D2 gastrectomy and PIPAC C/D. The median age was 61 years (range 24-76), there were eleven female patients, and 20 patients had preoperative chemotherapy. There was no mortality. Two patients had grade 3b complications that were potentially related to PIPAC C/D (one anastomotic leakage, and one late duodenal blow-out). One patient had severe neutropenia, and nine patients had moderate pain. The LOS was 6 days (4-26). One patient had positive peritoneal lavage cytology before resection, and none were positive after. Fifteen patients had postoperative chemotherapy. CONCLUSIONS Laparoscopic D2 gastrectomy in combination with PIPAC C/D is feasible and safe.
Collapse
Affiliation(s)
- Martin Graversen
- Odense PIPAC Center, Odense University Hospital, Odense, Denmark. .,Department of Surgery, Odense University Hospital, Odense, Denmark. .,OPEN - Open Patient data Explorative Network, Odense University Hospital, Region of Southern Denmark, Odense, Denmark. .,Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark.
| | - I Rouvelas
- Department of Upper Abdominal Diseases, Karolinska University Hospital and Division of Surgery and Oncology, CLINTEC, Karolinska Institutet, Stockholm, Sweden
| | - A P Ainsworth
- Odense PIPAC Center, Odense University Hospital, Odense, Denmark.,Department of Surgery, Odense University Hospital, Odense, Denmark.,Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
| | - A P Bjarnesen
- Odense PIPAC Center, Odense University Hospital, Odense, Denmark.,Department of Surgery, Odense University Hospital, Odense, Denmark
| | - S Detlefsen
- Odense PIPAC Center, Odense University Hospital, Odense, Denmark.,Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark.,Department of Pathology, Odense University Hospital, Odense, Denmark
| | - S B Ellebaek
- Odense PIPAC Center, Odense University Hospital, Odense, Denmark.,Department of Surgery, Odense University Hospital, Odense, Denmark
| | - C W Fristrup
- Odense PIPAC Center, Odense University Hospital, Odense, Denmark.,Department of Surgery, Odense University Hospital, Odense, Denmark
| | - M G Liljefors
- Department of Oncology, Karolinska University Hospital and Division of Surgery and Oncology, CLINTEC, Karolinska Institutet, Stockholm, Sweden
| | - L Lundell
- Department of Upper Abdominal Diseases, Karolinska University Hospital and Division of Surgery and Oncology, CLINTEC, Karolinska Institutet, Stockholm, Sweden
| | - M Nilsson
- Department of Upper Abdominal Diseases, Karolinska University Hospital and Division of Surgery and Oncology, CLINTEC, Karolinska Institutet, Stockholm, Sweden
| | - P Pfeiffer
- Odense PIPAC Center, Odense University Hospital, Odense, Denmark.,Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark.,Department of Oncology, Odense University Hospital, Odense, Denmark
| | - L S Tarpgaard
- Odense PIPAC Center, Odense University Hospital, Odense, Denmark.,Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark.,Department of Oncology, Odense University Hospital, Odense, Denmark
| | - A Tsekrekos
- Department of Upper Abdominal Diseases, Karolinska University Hospital and Division of Surgery and Oncology, CLINTEC, Karolinska Institutet, Stockholm, Sweden
| | - M B Mortensen
- Odense PIPAC Center, Odense University Hospital, Odense, Denmark.,Department of Surgery, Odense University Hospital, Odense, Denmark.,Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
4
|
Zhou Y, Qi M, Zhou YX, Fang SQ. Application of intraperitoneal hyperthermic perfusion chemotherapy in gastric cancer. Shijie Huaren Xiaohua Zazhi 2023; 31:1-7. [DOI: 10.11569/wcjd.v31.i1.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Gastric cancer is one of the most common malignant tumors in China. Intraperitoneal hyperthermic perfusion chemotherapy is a comprehensive therapy that combines intraperitoneal perfusion, hyperthermia, and chemotherapy. It has a good curative effect in peritoneal metastasis of gastric cancer. In recent years, with the continuous progress of technology and the deepening of research, the scope of application of intraperitoneal hyperthermic perfusion chemotherapy is more extensive. In this paper, we discuss intraperitoneal hyperthermic perfusion chemotherapy with regard to its application in gastric cancer, commonly used drugs, safety, and prospects.
Collapse
Affiliation(s)
- Yue Zhou
- Department of Gastroenterology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese medicine, Shanghai 200437, China
| | - Mei Qi
- Department of Gastroenterology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese medicine, Shanghai 200437, China
| | - Yu-Xuan Zhou
- Department of Gastroenterology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese medicine, Shanghai 200437, China
| | - Sheng-Quan Fang
- Department of Gastroenterology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese medicine, Shanghai 200437, China
| |
Collapse
|
5
|
Di Giorgio A, Gerardi C, Abatini C, Melotti G, Bonavina L, Torri V, Santullo F, Garattini S, De Luca M, Rulli E, Rulli E, Pacelli F. Prophylactic surgery plus hyperthermic intraperitoneal chemotherapy (HIPEC CO2) versus standard surgery for gastric carcinoma at high risk of peritoneal carcinomatosis: short and long-term outcomes (GOETH STUDY)-a collaborative randomized controlled trial by ACOI, FONDAZIONE AIOM, SIC, SICE, and SICO. Trials 2022; 23:969. [PMID: 36457115 PMCID: PMC9714394 DOI: 10.1186/s13063-022-06880-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 11/02/2022] [Indexed: 12/03/2022] Open
Abstract
INTRODUCTION At the time of diagnosis, 15-20% of gastric carcinomas are in stage T4 or T4b. Furthermore, 5-20% of patients undergoing potentially curative surgery suffer from synchronous or metachronous peritoneal metastases. To date, neither surgery nor systemic chemotherapy successfully controls peritoneal dissemination, offering a limited impact on survival. Peritoneal metastases are in fact responsible for death in around 60% of gastric cancer patients. Several Eastern studies in the past have focused on hyperthermic intraperitoneal chemotherapy (HIPEC) as a prophylactic measure in patients with serosal extension, nodal involvement, and positive peritoneal fluid cytology. Therefore, a new multimodal therapeutic strategy based on aggressive surgery plus new locoregional treatment may prolong survival in this particular clinical scenario. METHODS This study compares the efficacy of prophylactic surgery (radical gastric resection, appendectomy, resection of the round ligament of the liver, and bilateral adnexectomy) plus hybrid CO2 HIPEC system versus standard surgery in patients with T3-T4 N0-N + gastric adenocarcinoma. Patients will be randomly assigned (1:1 ratio) to the experimental arm or standard surgery. The primary endpoint is to establish the difference in disease-free survival between the groups. The secondary objective is to compare the safety and tolerability of prophylactic surgery plus HIPEC CO2 versus standard surgery. DISCUSSION Considering the poor prognosis of patients with peritoneal dissemination from gastric cancer, a prophylactic strategy to prevent peritoneal metastases may be beneficial. In patients with gastric cancer at high risk of peritoneal carcinomatosis, we propose aggressive surgical treatment with radical gastrectomy, removal of organs at risk of harbouring tumour cells, and HIPEC. TRIAL REGISTRATION ClinicalTrials.gov NCT03917173. Registered on 16 April 2019. PROTOCOL VERSION v1, March 27, 2019. Protocol number: IRFMN-GCC-7813. EudraCT number: 2019-001478-27.
Collapse
Affiliation(s)
- A. Di Giorgio
- grid.411075.60000 0004 1760 4193Operational Unit of Peritoneum and Retroperitoneum Surgery, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo A. Gemelli, 8, 00168 Rome, Italy
| | - C. Gerardi
- grid.4527.40000000106678902Istituto Di Ricerche Farmacologiche “Mario Negri” IRCCS, Milan, Italy
| | - C. Abatini
- grid.411075.60000 0004 1760 4193Operational Unit of Peritoneum and Retroperitoneum Surgery, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo A. Gemelli, 8, 00168 Rome, Italy
| | - G. Melotti
- Associazione Chirurghi Ospedalieri Italiani, ACOI, Rome, Italy
| | - L. Bonavina
- grid.419557.b0000 0004 1766 7370Division of General and Foregut Surgery, Department of Biomedical Sciences for Health, IRCCS Policlinico San Donato and University of Milan, Milan, Italy
| | - V. Torri
- grid.4527.40000000106678902Istituto Di Ricerche Farmacologiche “Mario Negri” IRCCS, Milan, Italy
| | - F. Santullo
- grid.411075.60000 0004 1760 4193Operational Unit of Peritoneum and Retroperitoneum Surgery, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo A. Gemelli, 8, 00168 Rome, Italy
| | - S. Garattini
- grid.4527.40000000106678902Istituto Di Ricerche Farmacologiche “Mario Negri” IRCCS, Milan, Italy
| | - M. De Luca
- grid.4527.40000000106678902Istituto Di Ricerche Farmacologiche “Mario Negri” IRCCS, Milan, Italy
| | - Erica Rulli
- grid.4527.40000000106678902Istituto Di Ricerche Farmacologiche “Mario Negri” IRCCS, Milan, Italy
| | - Eliana Rulli
- grid.4527.40000000106678902Istituto Di Ricerche Farmacologiche “Mario Negri” IRCCS, Milan, Italy
| | - F. Pacelli
- grid.411075.60000 0004 1760 4193Operational Unit of Peritoneum and Retroperitoneum Surgery, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo A. Gemelli, 8, 00168 Rome, Italy
| | | |
Collapse
|
6
|
Goetze TO, Al-Batran SE. Perspectives on the Management of Oligometastatic Disease in Esophago-Gastric Cancer. Cancers (Basel) 2022; 14:5200. [PMID: 36358619 PMCID: PMC9658190 DOI: 10.3390/cancers14215200] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 10/17/2022] [Accepted: 10/21/2022] [Indexed: 09/22/2023] Open
Abstract
Gastric adenocarcinoma and esophageal cancer are the fifth and seventh most common cancer types worldwide. At the time of initial diagnosis, up to 50% of esophagogastric cancers present with distant metastatic lesions and are candidates for chemotherapy. Curative surgery in this stage is still an experimental approach. Only a small number of these metastatic patients show an oligometastatic disease with no uniform definition of what oligometastatic means in gastric cancer. Nevertheless, the question remains unanswered as to whether these patients are still candidates for curative concepts. Some studies have attempted to answer this question but have not been adequately designed to address the role of a curative-intended multimodal therapy in this setting. The current FLOT-5 is designed to potentially provide a definitive answer to the question of whether curatively intended surgery plays a role or is a disadvantage in this setting.
Collapse
Affiliation(s)
- Thorsten Oliver Goetze
- Krankenhaus Nordwest gGmbH, Institut of Clinical Cancer Research, UCT—University Cancer Center Frankfurt-Marburg, Steinbacher Hohl 2-26, 60488 Frankfurt, Germany
| | | |
Collapse
|
7
|
Yarema R, Оhorchak М, Hyrya P, Kovalchuk Y, Safiyan V, Oliynyk Y, Rilinh O, Matusyak M. PREDICTIVE NOMOGRAM OF THE RISK OF PERITONEAL RELAPSE FOLLOWING RADICAL GASTRIC CANCER SURGERY. PROCEEDINGS OF THE SHEVCHENKO SCIENTIFIC SOCIETY. MEDICAL SCIENCES 2022; 69. [DOI: 10.25040/ntsh2022.02.03] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2023]
|
8
|
Hama S, Nishi T, Isono E, Itakura S, Yoshikawa Y, Nishimoto A, Suzuki S, Kirimura N, Todo H, Kogure K. Intraperitoneal administration of nanoparticles containing tocopheryl succinate prevents peritoneal dissemination. Cancer Sci 2022; 113:1779-1788. [PMID: 35253340 PMCID: PMC9128176 DOI: 10.1111/cas.15321] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 03/02/2022] [Accepted: 03/03/2022] [Indexed: 11/30/2022] Open
Abstract
Intraperitoneal administration of anticancer nanoparticles is a rational strategy for preventing peritoneal dissemination of colon cancer owing to the prolonged retention of nanoparticles in the abdominal cavity. However, instability of nanoparticles in body fluids causes inefficient retention, reducing its anticancer effects. We have previously developed anticancer nanoparticles containing tocopheryl succinate, which showed high in vivo stability and multifunctional anticancer effects. In the present study, we have demonstrated that peritoneal dissemination derived from colon cancer was prevented by intraperitoneal administration of tocopheryl succinate nanoparticles. The biodistribution of tocopheryl succinate nanoparticles was evaluated by inductively coupled plasma mass spectroscopy and imaging analysis in mice administered quantum dot encapsulated tocopheryl succinate nanoparticles. Intraperitoneal administration of tocopheryl succinate nanoparticles showed longer retention in the abdominal cavity than by its intravenous administration. Moreover, due to effective biodistribution, tumor growth was prevented by intraperitoneal administration of tocopheryl succinate nanoparticles. Furthermore, the anticancer effect was attributed to the inhibition of cancer cell proliferation and improvement of the intraperitoneal microenvironment, such as decrease in the levels of vascular endothelial growth factor A, interleukin 10, and M2-like phenotype of tumor-associated macrophages. Collectively, intraperitoneal administration of tocopheryl succinate nanoparticles is expected to have multifaceted antitumor effects against colon cancer with peritoneal dissemination.
Collapse
Affiliation(s)
- Susumu Hama
- Research Institute of Pharmaceutical Sciences, Faculty of Pharmacy, Musashino University, Tokyo, 202-8585, Japan
| | - Takayuki Nishi
- Department of Biophysical Chemistry, Kyoto Pharmaceutical University, Kyoto, 607-8414, Japan
| | - Eitaro Isono
- Research Institute of Pharmaceutical Sciences, Faculty of Pharmacy, Musashino University, Tokyo, 202-8585, Japan
| | - Shoko Itakura
- Faculty of Pharmacy and Pharmaceutical Sciences, Josai University, Saitama, 350-0295, Japan
| | - Yutaka Yoshikawa
- Department of Health, Sports, and Nutrition, Faculty of Health and Welfare, Kobe Women's University, Kobe, 650-0046, Japan
| | - Akinori Nishimoto
- Department of Biophysical Chemistry, Kyoto Pharmaceutical University, Kyoto, 607-8414, Japan
| | - Satoko Suzuki
- Department of Biophysical Chemistry, Kyoto Pharmaceutical University, Kyoto, 607-8414, Japan
| | - Naoko Kirimura
- Department of Biophysical Chemistry, Kyoto Pharmaceutical University, Kyoto, 607-8414, Japan
| | - Hiroaki Todo
- Faculty of Pharmacy and Pharmaceutical Sciences, Josai University, Saitama, 350-0295, Japan
| | - Kentaro Kogure
- Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, 770-8505, Japan
| |
Collapse
|
9
|
Abstract
Peritoneal surface malignancies comprise a heterogeneous group of primary tumours, including peritoneal mesothelioma, and peritoneal metastases of other tumours, including ovarian, gastric, colorectal, appendicular or pancreatic cancers. The pathophysiology of peritoneal malignancy is complex and not fully understood. The two main hypotheses are the transformation of mesothelial cells (peritoneal primary tumour) and shedding of cells from a primary tumour with implantation of cells in the peritoneal cavity (peritoneal metastasis). Diagnosis is challenging and often requires modern imaging and interventional techniques, including surgical exploration. In the past decade, new treatments and multimodal strategies helped to improve patient survival and quality of life and the premise that peritoneal malignancies are fatal diseases has been dismissed as management strategies, including complete cytoreductive surgery embedded in perioperative systemic chemotherapy, can provide cure in selected patients. Furthermore, intraperitoneal chemotherapy has become an important part of combination treatments. Improving locoregional treatment delivery to enhance penetration to tumour nodules and reduce systemic uptake is one of the most active research areas. The current main challenges involve not only offering the best treatment option and developing intraperitoneal therapies that are equivalent to current systemic therapies but also defining the optimal treatment sequence according to primary tumour, disease extent and patient preferences. New imaging modalities, less invasive surgery, nanomedicines and targeted therapies are the basis for a new era of intraperitoneal therapy and are beginning to show encouraging outcomes.
Collapse
|
10
|
Molecular Cytology by One-Step Nucleic Acid Amplification (OSNA) Assay of Peritoneal Washings during D2 Gastrectomy in Advanced Gastric Cancer Patients: Preliminary Results. J Clin Med 2021; 10:jcm10225230. [PMID: 34830512 PMCID: PMC8621409 DOI: 10.3390/jcm10225230] [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: 07/28/2021] [Revised: 10/31/2021] [Accepted: 11/08/2021] [Indexed: 11/17/2022] Open
Abstract
The presence of peritoneal free cancer cells (FCC) in gastric cancer (GC) patients is a poor prognostic factor. D2 gastrectomy may induce exfoliated FCC spread from the primary tumour or involved lymph nodes (LN). Conventional cytology for FCC detection has several limitations, whereas prophylactic use of extensive intraoperative peritoneal lavage (IPL) does not improve survival. A prospective single-arm observational study was conducted to verify whether D2 gastrectomy causes an intraoperative increase of FCC in peritoneal fluid. Twenty-seven GC patients underwent D2 gastrectomy, followed by objective quantitative measurements of CK19 mRNA level reflecting FCC with One-Step Nucleic Acid Amplification (OSNA) assay. The IPL with 3000 mL of saline was performed twice: (1) after gastrectomy with D2 lymphadenectomy and (2) after alimentary tract reconstruction. The IPL samples were analysed by initial cytology and four (1-4) consecutive OSNA assays. Initial OSNA measurement (1) revealed positive results (≥24.6 cCP/μL) in 7 (29.6%) patients. Subsequent OSNA measurements showed a significant decrease in the FCC level after D2 gastrectomy (1 vs. 2; p = 0.0012). The first IPL induced a non-significant increase in the FCCs (2 vs. 3, p = 0.3300), but the second IPL reversed it to normal levels (3 vs. 4, p = 0.0.0574). The OSNA assay indicates a temporal intraoperative increase in the peritoneal FCC in advanced GC patients undergoing D2 gastrectomy. Two consecutive IPLs are necessary to reverse the increase of CK19 mRNA level in peritoneal washings.
Collapse
|
11
|
Wang F, Zhang X, Li Y, Tang L, Qu X, Ying J, Zhang J, Sun L, Lin R, Qiu H, Wang C, Qiu M, Cai M, Wu Q, Liu H, Guan W, Zhou A, Zhang Y, Liu T, Bi F, Yuan X, Rao S, Xin Y, Sheng W, Xu H, Li G, Ji J, Zhou Z, Liang H, Zhang Y, Jin J, Shen L, Li J, Xu R. The Chinese Society of Clinical Oncology (CSCO): Clinical guidelines for the diagnosis and treatment of gastric cancer, 2021. Cancer Commun (Lond) 2021; 41:747-795. [PMID: 34197702 PMCID: PMC8360643 DOI: 10.1002/cac2.12193] [Citation(s) in RCA: 435] [Impact Index Per Article: 108.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 06/21/2021] [Accepted: 06/23/2021] [Indexed: 02/05/2023] Open
Abstract
There exist differences in the epidemiological characteristics, clinicopathological features, tumor biological characteristics, treatment patterns, and drug selections between gastric cancer patients from the Eastern and Western countries. The Chinese Society of Clinical Oncology (CSCO) has organized a panel of senior experts specializing in all sub-specialties of gastric cancer to compile a clinical guideline for the diagnosis and treatment of gastric cancer since 2016 and renews it annually. Taking into account regional differences, giving full consideration to the accessibility of diagnosis and treatment resources, these experts have conducted expert consensus judgment on relevant evidence and made various grades of recommendations for the clinical diagnosis and treatment of gastric cancer to reflect the value of cancer treatment and meeting health economic indexes in China. The 2021 CSCO Clinical Practice Guidelines for Gastric Cancer covers the diagnosis, treatment, follow-up, and screening of gastric cancer. Based on the 2020 version of the CSCO Chinese Gastric Cancer guidelines, this updated guideline integrates the results of major clinical studies from China and overseas for the past year, focused on the inclusion of research data from the Chinese population for more personalized and clinically relevant recommendations. For the comprehensive treatment of non-metastatic gastric cancer, attentions were paid to neoadjuvant treatment. The value of perioperative chemotherapy is gradually becoming clearer and its recommendation level has been updated. For the comprehensive treatment of metastatic gastric cancer, recommendations for immunotherapy were included, and immune checkpoint inhibitors from third-line to the first-line of treatment for different patient groups with detailed notes are provided.
Collapse
|
12
|
Ukegjini K, Putora PM, Guidi M, Süveg K, Cihoric N, Widmann B, Steffen T. Pressurized Intraperitoneal Aerosol Chemotherapy-Related Clinical Trials in the Treatment of Peritoneal Metastases. Oncology 2021; 99:601-610. [PMID: 34265774 DOI: 10.1159/000516959] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Accepted: 04/29/2021] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Pressurized intraperitoneal aerosol chemotherapy (PIPAC) is a treatment option for patients with peritoneal metastases. We evaluated the current status of ongoing prospective clinical trials investigating PIPAC to provide an overview and predict trends in this field. METHODS All 367,494 records of clinical trials registered at ClinicalTrials.gov were searched for trials dealing with PIPAC. Active or unpublished trials were further analyzed. RESULTS In total, 22 clinical trials were identified and selected for further analyses. Most trials had a single-arm design and were phase I or II. No phase III trials were registered. Academic centers were recorded as primary sponsors in the majority of trials (63.6%). Every year, between 2 and 5 new trials were initiated. In 17 trials (81.8%), PIPAC was used in a palliative setting only, 2 trials performed PIPAC in a neoadjuvant setting, and 2 trials performed PIPAC in an adjuvant setting. Six different drugs (doxorubicin, cisplatin, oxaliplatin, nab-paclitaxel, 5-fluorouracil, and docetaxel) were used in these clinical trials. Most trials investigated the efficacy (n = 15) or safety (n = 7) of PIPAC therapies. CONCLUSIONS The results of ongoing clinical trials will bring specific information on indications for PIPAC as well as the impact of PIPAC on quality of life and overall survival.
Collapse
Affiliation(s)
- Kristjan Ukegjini
- Department of Surgery, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Paul Martin Putora
- Department of Radiation Oncology, Kantonsspital St. Gallen, St. Gallen, Switzerland.,Department of Radiation Oncology, University of Bern, Bern, Switzerland
| | - Marisa Guidi
- Department of Surgery, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Krisztian Süveg
- Department of Radiation Oncology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Nikola Cihoric
- Department of Radiation Oncology, University of Bern, Bern, Switzerland
| | - Bernhard Widmann
- Department of Surgery, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Thomas Steffen
- Department of Surgery, Kantonsspital St. Gallen, St. Gallen, Switzerland
| |
Collapse
|
13
|
Kang WZ, Zhong YX, Ma FH, Tian YT. Progress and controversy in treatment of gastric cancer patients with positive peritoneal lavage cytology. Shijie Huaren Xiaohua Zazhi 2021; 29:269-273. [DOI: 10.11569/wcjd.v29.i6.269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Affiliation(s)
- Wen-Zhe Kang
- Department of Pancreatic and Gastric Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yu-Xin Zhong
- Department of Pancreatic and Gastric Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Fu-Hai Ma
- Department of Pancreatic and Gastric Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yan-Tao Tian
- Department of Pancreatic and Gastric Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| |
Collapse
|
14
|
Kang WZ, Zhong YX, Ma FH, Xue LY, Xiong JP, Ma S, Li Y, Xie YB, Quan X, Tian YT. Survival outcomes and prognostic indicators for gastric cancer patients with positive peritoneal wash cytology but no peritoneal metastasis after radical gastrectomy. World J Gastrointest Oncol 2021; 13:24-36. [PMID: 33510847 PMCID: PMC7805269 DOI: 10.4251/wjgo.v13.i1.24] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 11/03/2020] [Accepted: 11/29/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Positive peritoneal wash cytology with no peritoneal metastasis (CY1P0) is a special type of distant gastric cancer metastasis, which describes a patient with positive peritoneal lavage cytology, but no definitive peritoneal metastasis, and there are no widely accepted treatment guidelines. We enrolled 48 primary CY1P0 gastric cancer patients treated by radical gastrectomy in this study. Our study illustrated the efficacy of radical gastrectomy for CY1P0 gastric cancer patients, and suggested that the pathological N factor and vascular invasion were significant independent risk factors for overall survival (OS). AIM To assess the survival of CY1P0 gastric cancer patient post-radical gastrectomy, and to identify factors associated with long-term prognosis. METHODS Our study included 48 patients with primary CY1P0 gastric cancer who had radical gastrectomies at the Cancer Hospital, Chinese Academy of Medical Sciences, Beijing, China between 2013 and 2018. R0 resection was achieved in all 48 patients. Twelve patients received neoadjuvant chemotherapy. Thirty patients received adjuvant chemotherapy and four received adjuvant chemoradiotherapy. OS statistics were available for 48 patients. Follow-up continued through March 2020. Univariate and multivariate analyses were performed using a Cox proportional hazards model to identify prognostic factors. RESULTS Median OS was 22.0 mo (95% confidence interval: 13.366-30.634 mo) post-surgery. Univariate analyses demonstrated that tumor site (P = 0.021), pathological N factor (P = 0.001), pathological T factor (P = 0.028), vascular invasion (P = 0.046), and the level of CA199 prior to initiating therapy (P = 0.002) were significant risk factors for OS. Multivariate analyses demonstrated that pathological N factor (P = 0.001) and vascular invasion (P = 0.031) were significant independent risk factors for OS. CONCLUSION This study suggested that radical gastrectomy may be efficient for CY1P0 gastric cancer patient post-radical gastrectomy and the pathological N factor and vascular invasion are significant independent risk factors for OS.
Collapse
Affiliation(s)
- Wen-Zhe Kang
- Department of Pancreatic and Gastric Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yu-Xin Zhong
- Department of Pancreatic and Gastric Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Fu-Hai Ma
- Department of Pancreatic and Gastric Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Li-Yan Xue
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Bejing 100021, China
| | - Jian-Ping Xiong
- Department of Pancreatic and Gastric Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Shuai Ma
- Department of Pancreatic and Gastric Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yang Li
- Department of Pancreatic and Gastric Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yi-Bin Xie
- Department of Pancreatic and Gastric Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Xu Quan
- Department of Pancreatic and Gastric Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yan-Tao Tian
- Department of Pancreatic and Gastric Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| |
Collapse
|
15
|
Hundeyin M, Strong VE. Is dilution the solution in gastric cancer? Lancet Gastroenterol Hepatol 2020; 6:85-86. [PMID: 33253660 DOI: 10.1016/s2468-1253(20)30339-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Accepted: 10/19/2020] [Indexed: 10/22/2022]
Affiliation(s)
- Mautin Hundeyin
- Department of Surgery, Gastric and Mixed Tumor Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Vivian E Strong
- Department of Surgery, Gastric and Mixed Tumor Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| |
Collapse
|
16
|
Leigh N, Solomon D, Pletcher E, Labow DM, Magge DR, Sarpel U, Golas BJ. The importance of primary tumor origin in gastrointestinal malignancies undergoing cytoreductive surgery and hyperthermic intraperitoneal chemotherapy. World J Surg Oncol 2020; 18:182. [PMID: 32703239 PMCID: PMC7379772 DOI: 10.1186/s12957-020-01938-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 06/29/2020] [Indexed: 12/14/2022] Open
Abstract
Background Appendiceal and colorectal cancers with peritoneal carcinomatosis (PC) can derive benefit from cytoreductive surgery and hyperthermic intraperitoneal chemotherapy (CRS/HIPEC). However, its role in gastric and small bowel malignancies remains undefined. Methods We retrospectively analyzed 251 gastrointestinal adenocarcinomas with PC which underwent CRS/HIPEC at our institution from 2007 to 2017. We compared outcomes of gastric, small bowel, appendiceal, and colorectal cohorts. Results Thirty-one gastric, 8 small bowel, 91 appendiceal, and 121 colorectal cohorts were included. More gastric cancers (90%) received neoadjuvant chemotherapy than any other cohort, p = 0.002. Although colorectal had the lowest peritoneal cancer index (PCI) (9) and appendiceal the highest (16), all cohorts underwent similar rates of organ resection and complete cytoreduction. Length of stay (p = 0.005) and major perioperative morbidity (Clavien III/IV, p = 0.011) were significantly higher in gastric and small bowel. Median overall survival (OS, p < 0.001) was significantly shorter in gastric (13 months) and small bowel (9 months) than in appendiceal (33 months) and colorectal (42 months) cohorts. On multivariate analysis, complete cytoreduction and PCI score were significant predictors of OS, p < 0.05. Conclusions Primary tumor origin significantly affects outcomes after CRS/HIPEC for gastrointestinal malignancies. Though there was a survival benefit in appendiceal and colorectal, gastric and small bowel survival was comparable to systemic chemotherapy.
Collapse
Affiliation(s)
- Natasha Leigh
- Division of Surgical Oncology, Icahn School of Medicine at Mount Sinai St. Luke's West Hospital, 425 West 59th Street, Suite 7B, New York, NY, 10019, USA.
| | - Daniel Solomon
- Division of Surgical Oncology, Icahn School of Medicine at Mount Sinai St. Luke's West Hospital, 425 West 59th Street, Suite 7B, New York, NY, 10019, USA
| | - Eric Pletcher
- Division of Surgical Oncology, Icahn School of Medicine at Mount Sinai St. Luke's West Hospital, 425 West 59th Street, Suite 7B, New York, NY, 10019, USA
| | - Daniel M Labow
- Division of Surgical Oncology, Icahn School of Medicine at Mount Sinai St. Luke's West Hospital, 425 West 59th Street, Suite 7B, New York, NY, 10019, USA
| | - Deepa R Magge
- Division of Surgical Oncology, Icahn School of Medicine at Mount Sinai St. Luke's West Hospital, 425 West 59th Street, Suite 7B, New York, NY, 10019, USA
| | - Umut Sarpel
- Division of Surgical Oncology, Icahn School of Medicine at Mount Sinai St. Luke's West Hospital, 425 West 59th Street, Suite 7B, New York, NY, 10019, USA
| | - Benjamin J Golas
- Division of Surgical Oncology, Icahn School of Medicine at Mount Sinai St. Luke's West Hospital, 425 West 59th Street, Suite 7B, New York, NY, 10019, USA
| |
Collapse
|
17
|
Zhao L, Li J, Bai C, Nie Y, Lin G. Multi-Modality Treatment for Patients With Metastatic Gastric Cancer: A Real-World Study in China. Front Oncol 2019; 9:1155. [PMID: 31737573 PMCID: PMC6839024 DOI: 10.3389/fonc.2019.01155] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 10/16/2019] [Indexed: 12/26/2022] Open
Abstract
Introduction: People with metastatic gastric cancer (GC) have a poor prognosis. The study aims to investigate the efficacy of multi-modality treatment for patients with metastatic GC. Methods: We retrospectively identified 267 patients with stage IV gastric cancer who were treated with systemic chemotherapy: 114 received multi-modality treatments, 153 received systematic chemotherapy alone. The survival of these two groups was compared by log rank test, the independent prognostic factors were investigated using univariate and multivariate analyses. Results: The median survival of metastatic GC patients who received multi-modality treatment was significantly longer than those who received systematic chemotherapy alone (18.4 vs. 11.4 months, P < 0.001). Multivariate analysis identified tumor histologic differentiation, CA19–9 level, previous curative resection, palliative gastrectomy, and metastasectomy as independent prognostic factors for overall survival. In the multimodality treatment group, patients who received palliative gastrectomy or metastasectomy had a longer survival than those who only received intraperitoneal chemotherapy or radiotherapy (21.6 vs. 15.2 months, P = 0.014). Conclusion: Multi-modality treatments offer a survival benefit for patients with metastatic GC. Future prospective studies are needed to confirm the result.
Collapse
Affiliation(s)
- Lin Zhao
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiarui Li
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chunmei Bai
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yongdu Nie
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Guole Lin
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
18
|
Wang FH, Shen L, Li J, Zhou ZW, Liang H, Zhang XT, Tang L, Xin Y, Jin J, Zhang YJ, Yuan XL, Liu TS, Li GX, Wu Q, Xu HM, Ji JF, Li YF, Wang X, Yu S, Liu H, Guan WL, Xu RH. The Chinese Society of Clinical Oncology (CSCO): clinical guidelines for the diagnosis and treatment of gastric cancer. Cancer Commun (Lond) 2019; 39:10. [PMID: 30885279 PMCID: PMC6423835 DOI: 10.1186/s40880-019-0349-9] [Citation(s) in RCA: 306] [Impact Index Per Article: 51.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 02/01/2019] [Indexed: 02/08/2023] Open
Abstract
China is one of the countries with the highest incidence of gastric cancer. There are differences in epidemiological characteristics, clinicopathological features, tumor biological characteristics, treatment patterns, and drug selection between gastric cancer patients from the Eastern and Western countries. Non-Chinese guidelines cannot specifically reflect the diagnosis and treatment characteristics for the Chinese gastric cancer patients. The Chinese Society of Clinical Oncology (CSCO) arranged for a panel of senior experts specializing in all sub-specialties of gastric cancer to compile, discuss, and revise the guidelines on the diagnosis and treatment of gastric cancer based on the findings of evidence-based medicine in China and abroad. By referring to the opinions of industry experts, taking into account of regional differences, giving full consideration to the accessibility of diagnosis and treatment resources, these experts have conducted experts' consensus judgement on relevant evidence and made various grades of recommendations for the clinical diagnosis and treatment of gastric cancer to reflect the value of cancer treatment and meeting health economic indexes. This guideline uses tables and is complemented by explanatory and descriptive notes covering the diagnosis, comprehensive treatment, and follow-up visits for gastric cancer.
Collapse
Affiliation(s)
- Feng-Hua Wang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060 Guangdong P. R. China
| | - Lin Shen
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Oncology, Peking University Cancer Hospital and Institute, Beijing, 100142 P. R. China
| | - Jin Li
- Department of Oncology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120 P. R. China
| | - Zhi-Wei Zhou
- Department of Gastric Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060 Guangdong P. R. China
| | - Han Liang
- Department of Gastric Cancer, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center of Cancer, Tianjin’s Clinical Research Cancer for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060 P. R. China
| | - Xiao-Tian Zhang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Oncology, Peking University Cancer Hospital and Institute, Beijing, 100142 P. R. China
| | - Lei Tang
- Medical Imaging Department, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Oncology, Peking University Cancer Hospital and Institute, Beijing, 100142 P. R. China
| | - Yan Xin
- Pathology Laboratory of Gastrointestinal Tumor, The First Hospital of China Medical University, Shenyang, 110001 Liaoning P. R. China
| | - Jing Jin
- Department of Radiation Oncology, National Cancer Center, China and Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021 P. R. China
| | - Yu-Jing Zhang
- Department of Radiotherapy, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060 Guangdong P. R. China
| | - Xiang-Lin Yuan
- Department of Medical Oncology, Tongji Hospital Affiliated to Tongji Medical College of Huazhong University of Science and Technology, Wuhan, 430030 Hubei P. R. China
| | - Tian-Shu Liu
- Department of Medical Oncology, Zhongshan Hospital Affiliated to Fudan University, Shanghai, 200032 P. R. China
| | - Guo-Xin Li
- Department of General Surgery, Nanfang Hospital Affiliated to Southern Medical University, Guangzhou, 510515 Guangdong P. R. China
| | - Qi Wu
- Department of Endoscopy Center, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Oncology, Peking University Cancer Hospital and Institute, Beijing, 100142 P. R. China
| | - Hui-Mian Xu
- Department of Surgical Oncology, The First Hospital of China Medical University, Shenyang, 110001 Liaoning P. R. China
| | - Jia-Fu Ji
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Oncology, Peking University Cancer Hospital and Institute, Beijing, 100142 P. R. China
| | - Yuan-Fang Li
- Department of Gastric Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060 Guangdong P. R. China
| | - Xin Wang
- Department of Radiation Oncology, National Cancer Center, China and Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021 P. R. China
| | - Shan Yu
- Department of Medical Oncology, Zhongshan Hospital Affiliated to Fudan University, Shanghai, 200032 P. R. China
| | - Hao Liu
- Department of General Surgery, Nanfang Hospital Affiliated to Southern Medical University, Guangzhou, 510515 Guangdong P. R. China
| | - Wen-Long Guan
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060 Guangdong P. R. China
| | - Rui-Hua Xu
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060 Guangdong P. R. China
| |
Collapse
|
19
|
Yonemura Y, Sako S, Wakama S, Ishibashi H, Mizumoto A, Takao N, Ichinose M, Noguchi K, Liu Y, Motoi S, Taniguchi K, Fushida S. History of Peritoneal Surface Malignancy Treatment in Japan. Indian J Surg Oncol 2019; 10:3-11. [PMID: 30886486 PMCID: PMC6397123 DOI: 10.1007/s13193-019-00893-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 02/05/2019] [Indexed: 12/13/2022] Open
Abstract
In this review, Japanese experience of cytoreductive surgery and perioperative chemotherapy is described. The new concept of peritoneal metastasis (PM) type, i.e., trans-mesothelial, trans-lymphatic, and superficial growing metastasis type was proposed in 2012. Surgeons should perform peritonectomy according to the type of PM. Since 1980, Japanese surgical oncologists have been spearheading the use of cytoreductive surgery (CRS) plus hyperthermic intraperitoneal chemoperfusion (HIPEC) as treatment for PM from gastric cancer. Two RCTs were conducted to verify the effect of HIPEC for the prophylaxis of peritoneal recurrence after curative resection of advanced gastric cancer. These two studies indicated that HIPEC is effective in preventing peritoneal recurrence of gastric cancer with serosal invasion. In 2002, intraperitoneal chemotherapy using taxans was developed for the treatment of PM from gastric cancer and led to the development of neoadjuvant intraperitoneal/systemic chemotherapy (NIPS), which was reported in 2006. In 2009, extensive intra-operative peritoneal lavage (EIPL) was developed, and contributed to the remarkable improvement in survival of patients with positive lavage cytology as demonstrated by prospective randomized clinical trials. In 2017, the Peritoneal Surface Oncology Group International proposed the value of complete cytoreduction and peritoneal cancer index cut-off as independent prognostic factors after CRS for gastric cancer with PM. Founded in 2016, the Japanese/Asian School of Peritoneal Surface Oncology (JASPSO) trains beginners to perform CRS and HIPEC safely. Sixteen students have already graduated from JASPSO and started to perform the treatment in their home countries.
Collapse
Affiliation(s)
- Yutaka Yonemura
- Asian School of Peritoneal Surface Malignancy Treatment, Osaka, Japan
- Department of Regional Cancer Therapy, Peritoneal Dissemination Center, Kishiwada Tokushukai Hospital, Kishiwada, Japan
- Department of Regional Cancer Therapy, Peritoneal Dissemination Center, Kusatsu General Hospital, Shiga, Japan
| | - Shouzou Sako
- Asian School of Peritoneal Surface Malignancy Treatment, Osaka, Japan
- Department of Regional Cancer Therapy, Peritoneal Dissemination Center, Kishiwada Tokushukai Hospital, Kishiwada, Japan
| | - Satoshi Wakama
- Department of Regional Cancer Therapy, Peritoneal Dissemination Center, Kishiwada Tokushukai Hospital, Kishiwada, Japan
| | - Haruaki Ishibashi
- Department of Regional Cancer Therapy, Peritoneal Dissemination Center, Kishiwada Tokushukai Hospital, Kishiwada, Japan
| | - Akiyoshi Mizumoto
- Department of Regional Cancer Therapy, Peritoneal Dissemination Center, Kusatsu General Hospital, Shiga, Japan
| | - Nobuyuki Takao
- Department of Regional Cancer Therapy, Peritoneal Dissemination Center, Kusatsu General Hospital, Shiga, Japan
| | - Masumi Ichinose
- Department of Regional Cancer Therapy, Peritoneal Dissemination Center, Kusatsu General Hospital, Shiga, Japan
| | - Kousuke Noguchi
- Department of Regional Cancer Therapy, Peritoneal Dissemination Center, Kusatsu General Hospital, Shiga, Japan
| | - Yang Liu
- Department of Regional Cancer Therapy, Peritoneal Dissemination Center, Kishiwada Tokushukai Hospital, Kishiwada, Japan
| | - Syunsuke Motoi
- Department of Regional Cancer Therapy, Peritoneal Dissemination Center, Kusatsu General Hospital, Shiga, Japan
| | - Keizou Taniguchi
- Department of Surgery, Mizonoguichi Hospital, Teikyou University, Tokyo, Japan
| | - Sachio Fushida
- Department of Surgery, Kanazawa University Hospital, Kanazawa University, Kanazawa, Japan
| |
Collapse
|
20
|
Coccolini F, Fugazzola P, Ansaloni L, Sartelli M, Cicuttin E, Leandro G, De' Angelis GL, Gaiani F, Di Mario F, Tomasoni M, Catena F. Advanced gastric cancer: the value of systemic and intraperitoneal chemotherapy. ACTA BIO-MEDICA : ATENEI PARMENSIS 2018; 89:104-109. [PMID: 30561427 PMCID: PMC6502214 DOI: 10.23750/abm.v89i8-s.7904] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Indexed: 12/20/2022]
Abstract
Several possibilities in treating advanced gastric cancer exist. Radical surgery associated with chemotherapy represents the cornerstone. Which one is more effective among neoadjuvant, adjuvant or perioperative chemotherapy is still a matter of debate. Several innovative results showed the necessity to keep increasingly into consideration the intraperitoneal administration of chemotherapies. Moreover, classical drugs and their ways of administration should be combined with the new ones to improve results. Lastly the prevention of recurrence should be considered: one possibility is to administer intraperitoneal chemotherapy earlier in the therapeutic algorithm.
Collapse
Affiliation(s)
- Federico Coccolini
- Emergency, General and Trauma Surgery dept., Bufalini hospital, Cesena, Italy.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Yepuri N, Bahary N, Jain A, Dhir M. Review and Update on the Role of Peritoneal Cytology in the Treatment of Gastric Cancer. J Surg Res 2018; 235:607-614. [PMID: 30691849 DOI: 10.1016/j.jss.2018.10.049] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Revised: 08/12/2018] [Accepted: 10/26/2018] [Indexed: 02/06/2023]
Abstract
BACKGROUND Positive peritoneal cytology (Cyt+) even in the absence of macroscopic disease is associated with poor prognosis in patients with gastric cancer and deemed as M1 disease. Recent years have seen advancements in the evaluation strategies for peritoneal washings and management of patients with Cyt+. The aim of this review was to describe the newest paradigms in the management of patients with gastric cancer who have Cyt+ without macroscopic peritoneal metastases. METHODS A comprehensive literature review was performed to identify studies on the management of gastric cancer and thereby to summarize relevant information on the accuracy of various diagnostic tests and controversies involved in the treatment of patients with Cyt+. RESULTS Although conventional cytology remains the standard technique for assessment of peritoneal washings, it is limited by low sensitivity. The role of immunohistochemistry and molecular techniques for the assessment of peritoneal washings is evolving. Although systemic chemotherapy remains the standard of care for patients with Cyt+ disease, the role of gastrectomy, intraperitoneal chemotherapy, extensive intraperitoneal saline lavage, and hyperthermic intraperitoneal chemotherapy is being evaluated. CONCLUSIONS Clinical decision-making in patients with Cyt+ remains controversial given the seemingly technical resectable albeit biologically unresectable or aggressive disease that portends an overall poor prognosis. Current management strategies are evolving, and further studies are needed to develop an optimal treatment strategy for these patients.
Collapse
Affiliation(s)
- Natesh Yepuri
- Division of Surgical Oncology, Department of Surgery, SUNY Upstate Medical University, Syracuse, New York
| | - Nathan Bahary
- Division of Oncology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Ajay Jain
- Division of Surgical Oncology, Department of Surgery, SUNY Upstate Medical University, Syracuse, New York
| | - Mashaal Dhir
- Division of Surgical Oncology, Department of Surgery, SUNY Upstate Medical University, Syracuse, New York.
| |
Collapse
|
22
|
Xi Y, Niu J, Li D, He J, Qin L, Peng X. Mixed lineage kinase-4 promotes gastric carcinoma tumorigenesis through suppression of the c-Jun N-terminal kinase signaling pathway. Exp Ther Med 2018; 16:3317-3324. [PMID: 30233678 PMCID: PMC6143876 DOI: 10.3892/etm.2018.6618] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 04/21/2017] [Indexed: 01/08/2023] Open
Abstract
Mixed lineage kinase-4 (MLK-4) is an important member of the mixed-lineage family of kinases that regulates the extracellular signal-regulated kinases and c-Jun N-terminal kinase (JNK) signaling pathways. The functions and mechanisms of MLK-4 in cancer initiation and progression have not been well understood. The present study investigated the expression, function and regulatory mechanism of MLK-4 in gastric carcinoma cells. Biochemical data indicated that normal MLK-4 was downregulated, which exerted dominant negative effects on gastric carcinoma cell viability, migration and invasion. The experimental data demonstrated that MLK-4 supplement abrogated activity of these mutants and induced inhibitory effects on gastric carcinoma cell viabilty, migration and invasion in vitro and in vivo. In addition, to determine the regulatory mechanism of MLK-4, its signaling pathway was assessed in gastric carcinoma cancer cells by regulating MLK-4. The present observations indicated that restoring MLK-4 activity by supplemental MLK-4 reduced gastric carcinoma cell colony formation in vitro and suppressed tumor viability, migration and invasion in vivo. The results of the present study indicated that MLK-4 may be a potential protein for targeting gastric carcinoma by suppressing kinases, which may lead to reduction of JNK signaling and enhance therapeutic efficacy in gastric carcinoma.
Collapse
Affiliation(s)
- Yu Xi
- Department of General Surgery, Tongji Hospital, Tongji Medical School, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
- Department of General Surgery, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, Xinjiang 832008, P.R. China
| | - Jianhua Niu
- Department of General Surgery, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, Xinjiang 832008, P.R. China
| | - Dongmei Li
- Department of General Surgery, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, Xinjiang 832008, P.R. China
| | - Jiagen He
- Department of General Surgery, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, Xinjiang 832008, P.R. China
| | - Le Qin
- Department of General Surgery, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, Xinjiang 832008, P.R. China
| | - Xinyu Peng
- Department of General Surgery, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, Xinjiang 832008, P.R. China
| |
Collapse
|
23
|
Ronellenfitsch U, Ernst K, Mertens C, Trunk MJ, Ströbel P, Marx A, Kienle P, Post S, Nowak K. Extensive intraperitoneal lavage to eliminate intraperitoneal tumor cells in gastrectomy with D2 lymphadenectomy for gastric cancer. TUMORI JOURNAL 2018; 104:361-368. [PMID: 30185117 DOI: 10.1177/0300891618792485] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
INTRODUCTION: Survival in gastric cancer is often limited by peritoneal carcinomatosis, which supposedly develops from serosal tumor infiltration or tumor cell spread during gastrectomy with lymphadenectomy. To eliminate peritoneal tumor cells, extensive intraperitoneal lavage (EIPL) has been suggested. Impressive results have been achieved in Japanese trials. In this trial, we assessed EIPL in Western patients. METHODS: This prospective trial included patients with non-metastatic gastric adenocarcinoma undergoing gastrectomy with D2 lymphadenectomy. Peritoneal fluid samples at laparotomy, after lymphadenectomy, and after EIPL were analyzed for tumor cells using cytology and EpCAM antibodies. The primary endpoint was peritoneal conversion rate (PCR; proportion of patients in whom EIPL eliminated tumor cells after lymphadenectomy). Secondary endpoints were peritoneal release rate (PRR; proportion of patients with peritoneal tumor cells after gastrectomy/lymphadenectomy among all patients without cells before gastrectomy/lymphadenectomy) and prevalence of peritoneal tumor cells before resection. EIPL was considered ineffective if PCR ⩽ 0.2 and warranted further exploration if PCR ⩾ 0.5. Clinicaltrials.gov identifier is NCT01476553. RESULTS: The trial was stopped early because tumor cells after gastrectomy/lymphadenectomy were detected in only 3/27 (11.1%) patients. In none of these did EIPL eliminate tumor cells (PCR 0, 95% confidence interval [CI] 0%-12.5%). In 8/27 (29.6%) patients, tumor cells were detected after EIPL. PRR was 11.1% (95% CI 2.4%-29.2%). There were no perioperative complications higher than Clavien-Dindo grade 3a. CONCLUSIONS: In Western patients, free peritoneal tumor cells after gastrectomy with D2 lymphadenectomy for gastric cancer were detected only sporadically. Although based on few cases, the findings suggest that EIPL spreads tumor cells into the peritoneal cavity, thus being potentially harmful. Therefore, EIPL cannot be generally recommended.
Collapse
Affiliation(s)
- Ulrich Ronellenfitsch
- 1 Department of Vascular and Endovascular Surgery, University Hospital Heidelberg, Heidelberg, Germany.,2 Department of Surgery, University Medical Center Mannheim, Medical Faculty Mannheim of the University of Heidelberg, Mannheim, Germany
| | - Kristina Ernst
- 2 Department of Surgery, University Medical Center Mannheim, Medical Faculty Mannheim of the University of Heidelberg, Mannheim, Germany.,3 Department of Gynecology, University Hospital Ulm, Ulm, Germany
| | - Christina Mertens
- 2 Department of Surgery, University Medical Center Mannheim, Medical Faculty Mannheim of the University of Heidelberg, Mannheim, Germany
| | - Marcus J Trunk
- 4 SYNLAB Pathology, Mannheim, Germany.,5 Institute of Pathology, University Medical Center Mannheim, Medical Faculty Mannheim of the University of Heidelberg, Mannheim, Germany
| | - Philipp Ströbel
- 5 Institute of Pathology, University Medical Center Mannheim, Medical Faculty Mannheim of the University of Heidelberg, Mannheim, Germany.,6 Institute of Pathology, University Medical Center Göttingen, Göttingen, Germany
| | - Alexander Marx
- 5 Institute of Pathology, University Medical Center Mannheim, Medical Faculty Mannheim of the University of Heidelberg, Mannheim, Germany
| | - Peter Kienle
- 2 Department of Surgery, University Medical Center Mannheim, Medical Faculty Mannheim of the University of Heidelberg, Mannheim, Germany.,7 Department of General and Abdominal Surgery, Theresienkrankenhaus and St. Hedwig-Klinik GmbH, Mannheim, Germany
| | - Stefan Post
- 2 Department of Surgery, University Medical Center Mannheim, Medical Faculty Mannheim of the University of Heidelberg, Mannheim, Germany
| | - Kai Nowak
- 2 Department of Surgery, University Medical Center Mannheim, Medical Faculty Mannheim of the University of Heidelberg, Mannheim, Germany.,8 Department of Abdominal, Vascular and Thoracic Surgery, Romed Klinikum Rosenheim, Rosenheim, Germany
| |
Collapse
|
24
|
Montori G, Coccolini F, Fugazzola P, Ceresoli M, Tomasoni M, Rubicondo C, Raimondo S, Pinelli D, Colledan M, Frigerio L, Ansaloni L. Cytoreductive surgery and hyperthermic intraperitoneal chemotherapy in ovarian and gastrointestinal peritoneal carcinomatosis: results from a 7-year experience. J Gastrointest Oncol 2018; 9:241-253. [PMID: 29755762 DOI: 10.21037/jgo.2017.12.04] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Background An increasing promising evidence and increasing long-term oncologic outcomes support the use of cytoreductive surgery (CRS) and hyperthermic intraperitoneal chemotherapy (HIPEC) as locoregional treatment for peritoneal carcinosis (PC) especially from ovarian and gastrointestinal tumors, but also for others cancers. Methods A prospective monocentric study was performed in Papa Giovanni XXIII Hospital, Bergamo (Italy). Patients and tumor characteristics were analyzed. Overall survival (OS), disease free survival (DFS) and morbidity were analyzed with Kaplan-Meier curves and log-rank testing. Results A total of 150 patients undergone CRS + HIPEC were analyzed from January 2011 to June 2017. The principal origins of PC were: gastric cancer (GC) (n=40), colon cancer (n=31), appendiceal cancer (AC) (n=18), ovarian cancer (OC) (n=49), others (n=12). Major morbidity [≥3 Common Terminology Criteria for Adverse Events (CTCAE)] and perioperative mortality rates were 38% and 2.7% respectively. Re-operation rate was 15.3%. Median OS is 9, 35, 47, 51, 82 months (29% 3-year OS; 27% 5-year OS; 48% 5-year OS; 40% 5-year OS; 67% 5-year OS respectively) in GC, colorectal cancer (CRC), OC, others tumors and AC respectively. Median DFS is 4, 14, 17, 19, 82 months (32% 3-year DFS; 22% 5-year DFS; 29% 5-year DFS; 11% 5-year DFS; 67% 5-year DFS respectively) in GC, CRC, others tumors, OC and AC respectively. Conclusions A therapeutic approach that combined CRS + HIPEC could achieve long-term survival in selected groups of patients with PC from gastrointestinal, gynecological and others tumors with acceptable morbidity and mortality. A good expertise and a high volume of patients are necessary to manage PC and to further improve results.
Collapse
Affiliation(s)
- Giulia Montori
- Unit of General and Emergency Surgery, ASST Papa Giovanni XXIII, Bergamo, Italy
| | - Federico Coccolini
- Unit of General and Emergency Surgery, ASST Papa Giovanni XXIII, Bergamo, Italy
| | - Paola Fugazzola
- Unit of General and Emergency Surgery, ASST Papa Giovanni XXIII, Bergamo, Italy
| | - Marco Ceresoli
- Unit of General and Emergency Surgery, ASST Papa Giovanni XXIII, Bergamo, Italy
| | - Matteo Tomasoni
- Unit of General and Emergency Surgery, ASST Papa Giovanni XXIII, Bergamo, Italy
| | - Carolina Rubicondo
- Unit of General and Emergency Surgery, ASST Papa Giovanni XXIII, Bergamo, Italy
| | - Stefano Raimondo
- Unit of General and Emergency Surgery, ASST Papa Giovanni XXIII, Bergamo, Italy
| | - Domenico Pinelli
- Unit of Hepato-biliary Surgery and Liver Transplantation, ASST Papa Giovanni XXIII, Bergamo, Italy
| | - Michele Colledan
- Unit of Hepato-biliary Surgery and Liver Transplantation, ASST Papa Giovanni XXIII, Bergamo, Italy
| | - Luigi Frigerio
- Unit of Gynecology and Obstetrics, ASST Papa Giovanni XXIII, Bergamo, Italy
| | - Luca Ansaloni
- Unit of General and Emergency Surgery, ASST Papa Giovanni XXIII, Bergamo, Italy
| |
Collapse
|
25
|
Sugarbaker PH. “Total Mesenteric Peritonectomy for Peritoneal Metastases” (with Video) by Jean-Baptiste Cazauran et al. Ann Surg Oncol 2017; 24:3799-3800. [DOI: 10.1245/s10434-017-6100-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
|
26
|
Ilhan E, Alemdar A, Ureyen O, Bas K. The Importance of Extensive Intraoperative Peritoneal Lavage as a Promising Method in Patients with Gastric Cancer Showing Positive Peritoneal Cytology Without Overt Peritoneal Metastasis and Other Therapeutic Approaches. J INVEST SURG 2017; 30:318-324. [PMID: 27806214 DOI: 10.1080/08941939.2016.1247930] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Peritoneal invasion is more common and has a worse prognosis in gastric cancer than most of other intestinal cancers. Advanced gastric cancers have a poor course in terms of the development of peritoneal carcinomatosis and prognosis, even if the curative resection has been performed. Patients usually die within the first 2 years of the postoperative period mainly due to peritoneal metastasis. It is, therefore, essential to eradicate intraperitoneal free cancer cells to prevent peritoneal recurrences. A standard therapy has not been developed yet for patients with gastric cancer with a positive peritoneal cytology or a gross peritoneal metastasis. Curative resection following neoadjuvant chemotherapy, postoperative oral S-1 chemotherapy, intraoperative intraperitoneal chemotherapy (IPC), and extensive intraoperative peritoneal lavage (EIPL)-IPC are recommended as therapeutic approaches. Although there is a limited number of studies on EIPL, which is a promising and exciting method in this patient population, unexpected results of survival have been demonstrated. We consider that the results of ongoing and further studies would lead to an extensive use of EIPL, which is a simple and easy method which can be applied anywhere and anytime, in patients with advanced gastic cancer and/or peritoneal cytology positive but peritoneal metastasis negative (CY+/P0) gastric cancer.
Collapse
Affiliation(s)
- Enver Ilhan
- a Izmir Bozyaka Training and Research Hospital, The Division of General Surgery-A , Karabaglar , 35170 Turkey
| | - Ali Alemdar
- b Okmeydani Training and Research Hospital, The Division of General Surgery , Okmeydani , 34384 Turkey
| | - Orhan Ureyen
- a Izmir Bozyaka Training and Research Hospital, The Division of General Surgery-A , Karabaglar , 35170 Turkey
| | - Koray Bas
- a Izmir Bozyaka Training and Research Hospital, The Division of General Surgery-A , Karabaglar , 35170 Turkey
| |
Collapse
|
27
|
Fugazzola P, Coccolini F, Montori G, Ceresoli M, Baggi P, Costanzo A, Tomasoni M, Gregis F, Nozza S, Ansaloni L. Overall and disease-free survival in patients treated with CRS + HIPEC with cisplatin and paclitaxel for gastric cancer with peritoneal carcinomatosis. J Gastrointest Oncol 2017; 8:572-582. [PMID: 28736644 DOI: 10.21037/jgo.2017.03.11] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Our experience regarding cytoreductive surgery (CRS) and hyperthermic intraperitoneal chemotherapy (HIPEC) was reviewed in terms of overall survival (OS) and disease-free survival (DFS) in patients with synchronous peritoneal carcinomatosis (SPC) and metachronous peritoneal carcinomatosis (MPC) from gastric cancer (GC). METHODS An analysis of prospectively collected data about patients who underwent CRS and HIPEC from July 2011 to July 2016 was carried out. Patients and tumor characteristics were taken into consideration together with pre and post-operative data. The outcomes concerned OS and DFS in both groups. RESULTS A total of 17 cases were reported. All patients of SPC group underwent neoadjuvant chemotherapy, while all patients of MPC group underwent adjuvant chemotherapy subsequently to surgery of the primary tumor. The mean follow up period was 9 months (SD±9.5). Thirteen patients (76.5%) had SPC and four (23.5%) had MPC. The mean total Peritoneal Cancer Index (PCI) was 8.5 (SD±8.4). The mean PCI was 3.75 (SD±4.9) for SPC group and 16 (SD±9.5) for MPC (P=0.003). HIPEC regimen was cisplatin plus paclitaxel for fourteen patients (82.4%) and cisplatin plus mitomycin-C (MMC) for three patients (17.6%). OS was 16 months and 6 months respectively in patients with SPC and MPC (P=0.189). DFS was 11 months and 2 months respectively in the two groups (P=0.156). Patients with SPC patients and PCI ≥12 were significantly different in terms of DFS from SPC with PCI <12 (P=0.001). Overall, twelve patients had postoperative major complications (CTCAE>2), in particular eight (61%) in SPC group while four (100%) in MPC group. Our study showed significantly better DFS for patients aged >60 years (P=0.016). CONCLUSIONS HIPEC and CRS with cisplatin and paclitaxel in patients with PC from GC showed promising results in improving the DFS and the OS, particularly for patients with PCI <12 and for those aged >60. Although a high incidence of complications was revealed, especially in MPC group.
Collapse
Affiliation(s)
- Paola Fugazzola
- Department of General and Emergency Surgery, ASST Papa Giovanni XXIII, Bergamo, Italy
| | - Federico Coccolini
- Department of General and Emergency Surgery, ASST Papa Giovanni XXIII, Bergamo, Italy
| | - Giulia Montori
- Department of General and Emergency Surgery, ASST Papa Giovanni XXIII, Bergamo, Italy
| | - Marco Ceresoli
- Department of General and Emergency Surgery, ASST Papa Giovanni XXIII, Bergamo, Italy
| | - Paolo Baggi
- Department of General and Emergency Surgery, ASST Papa Giovanni XXIII, Bergamo, Italy
| | - Antonio Costanzo
- Department of General and Emergency Surgery, ASST Papa Giovanni XXIII, Bergamo, Italy
| | - Matteo Tomasoni
- Department of General and Emergency Surgery, ASST Papa Giovanni XXIII, Bergamo, Italy
| | - Francesco Gregis
- Department of Farmacology, ASST Papa Giovanni XXIII, Bergamo, Italy
| | - Silvia Nozza
- Department of Farmacology, ASST Papa Giovanni XXIII, Bergamo, Italy
| | - Luca Ansaloni
- Department of General and Emergency Surgery, ASST Papa Giovanni XXIII, Bergamo, Italy
| |
Collapse
|
28
|
Qiu C, Li Y, Liang X, Qi Y, Chen Y, Meng X, Zheng H, Xu Y, Cai S, Cai G, Liu J. A study of peritoneal metastatic xenograft model of colorectal cancer in the treatment of hyperthermic intraperitoneal chemotherapy with Raltitrexed. Biomed Pharmacother 2017; 92:149-156. [PMID: 28535418 DOI: 10.1016/j.biopha.2017.04.053] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Revised: 04/13/2017] [Accepted: 04/13/2017] [Indexed: 01/07/2023] Open
Abstract
Peritoneal metastasis of colorectal cancer is one of the most incident and fateful diseases among relapse cases. It shows a certain resistance to systemic chemotherapy. The perfusion system in clinic is complex and hard to be used in fundamental researches. This study aims at evaluating the effect of an improved hyperthermic intraperitoneal chemotherapy with Raltitrexed used in tumor-bearing mice with peritoneal metastatic colorectal carcinoma. The results showed that no severe adverse effect was observed. All control animals developed extensive peritoneal and mesenteric metastatic nodes. Tumor sites in the treatment groups were reduced significantly. The administration dose of Raltitrexed influenced concentration in systemic blood and peritoneal tissues. Temperature promoted the intracellular absorption of Raltitrexed significantly. Our findings reveal that hyperthermic intraperitoneal chemotherapy is an efficient therapy in treating peritoneal metastatic carcinoma in nude mice. It can effectively reduce the extension of carcinoma cells from macro and micro examination. The combination of hyperthermia and Raltitrexed resulted in an improved therapeutic effect on animal models.
Collapse
MESH Headings
- Absorption, Physiological
- Animals
- Antimetabolites, Antineoplastic/administration & dosage
- Antimetabolites, Antineoplastic/adverse effects
- Antimetabolites, Antineoplastic/pharmacokinetics
- Antimetabolites, Antineoplastic/therapeutic use
- Apoptosis/drug effects
- Carcinoma/drug therapy
- Carcinoma/pathology
- Carcinoma/secondary
- Carcinoma/therapy
- Colorectal Neoplasms/drug therapy
- Colorectal Neoplasms/metabolism
- Colorectal Neoplasms/pathology
- Colorectal Neoplasms/therapy
- Combined Modality Therapy/adverse effects
- Dose-Response Relationship, Drug
- Drug Resistance, Neoplasm
- Female
- Half-Life
- Hot Temperature
- Humans
- Hyperthermia, Induced/adverse effects
- Infusions, Parenteral
- Male
- Mice, Nude
- Peritoneal Neoplasms/drug therapy
- Peritoneal Neoplasms/pathology
- Peritoneal Neoplasms/secondary
- Peritoneal Neoplasms/therapy
- Quinazolines/administration & dosage
- Quinazolines/adverse effects
- Quinazolines/pharmacokinetics
- Quinazolines/therapeutic use
- Random Allocation
- Thiophenes/administration & dosage
- Thiophenes/adverse effects
- Thiophenes/pharmacokinetics
- Thiophenes/therapeutic use
- Tumor Burden
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Cen Qiu
- State Key Laboratory of Bioreactor Engineering & Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Yueqi Li
- State Key Laboratory of Bioreactor Engineering & Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Xin Liang
- State Key Laboratory of Bioreactor Engineering & Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Yingxue Qi
- State Key Laboratory of Bioreactor Engineering & Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Yiyang Chen
- State Key Laboratory of Bioreactor Engineering & Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Xianke Meng
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, PR China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, PR China
| | - Hongtu Zheng
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, PR China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, PR China
| | - Ye Xu
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, PR China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, PR China
| | - Sanjun Cai
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, PR China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, PR China
| | - Guoxiang Cai
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, PR China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, PR China.
| | - Jianwen Liu
- State Key Laboratory of Bioreactor Engineering & Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, PR China.
| |
Collapse
|
29
|
Solass W, Struller F, Horvath P, Königsrainer A, Sipos B, Weinreich FJ. Morphology of the peritoneal cavity and pathophysiological consequences. Pleura Peritoneum 2017; 1:193-201. [PMID: 30911623 DOI: 10.1515/pp-2016-0023] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 11/29/2016] [Indexed: 01/05/2023] Open
Abstract
The peritoneal cavity (cavum peritonei) is incompletely divided into spaces and recessus (or fossae), which are playing an important role in health and disease. Peritoneal subspaces are determined by the parietal attachments of the abdominal organs, the ligaments and mesenteries. These include the splenorenal, the falciform, the triangular, the gastrosplenic, the phrenicocolic and the gastrocolic ligaments; the greater omentum and the lesser omentum (formed by the gastrohepatic and hepatoduodenal ligaments); the small bowel mesenterium and the mesocolon. These ligaments and mesenteries divide the peritoneal cavity into several distinct anatomic and functional regions. The supramesocolic compartment is divided into a bilateral subphrenic space and a subhepatic space continuing into the lesser sac (bursa omentalis). The inframesolic compartment is divided into a left and right region by the mesentery. The right paracolic gutter communicates with the pelvis and with the right suphrenic space. The left paracolic gutter is separated from the left subphrenic space by the phrenocolic ligament. The peritoneal space is virtual, is completely occupied by the intraabdominal organs and can only be visualized by radiological means in the presence of air (organ perforation), liquid (ascites, pus, bile, gastrointestinal fluids) or tumor invasion. Peritoneal morphology has numerous pathophysiological implications: it impacts on the propagation of intraabdominal infections, determines the spreading of peritoneal metastasis and can cause bowel volvulus. Internal hernias can arise at the junction between intraperitoneal and extraperitoneal bowel segments, in particular into the left paraduodenal recessus. Knowledge of peritoneal morphology is a precondition for developing locoregional therapeutic strategies in peritoneal disease and for effective peritoneal dialysis.
Collapse
Affiliation(s)
- Wiebke Solass
- Institute of Pathology, University of Tübingen, Tübingen, Germany
| | - Florian Struller
- Department of General and Transplant Surgery, Experimental Surgery, University of Tübingen, Tübingen, Germany
| | - Philipp Horvath
- Department of General and Transplant Surgery, Experimental Surgery, University of Tübingen, Tübingen, Germany
| | - Alfred Königsrainer
- Department of General and Transplant Surgery, Experimental Surgery, University of Tübingen, Tübingen, Germany
| | - Bence Sipos
- Institute of Pathology, University of Tübingen, Tübingen, Germany
| | - Frank-Jürgen Weinreich
- Department of General and Transplant Surgery, Experimental Surgery, University of Tübingen, Hoppe-Seyler Str. 3, 72076 Tübingen, Germany
| |
Collapse
|