1
|
Pan S, Yuan H, Zhai Q, Zhang Y, He H, Yin T, Tang X, Gou J. The journey of nanoparticles in the abdominal cavity: Exploring their in vivo fate and impact factors. J Control Release 2024; 376:266-285. [PMID: 39396710 DOI: 10.1016/j.jconrel.2024.10.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 10/03/2024] [Accepted: 10/05/2024] [Indexed: 10/15/2024]
Abstract
Peritoneal carcinomatosis (PC) is caused by metastasis of primary tumor cells from intra-abdominal organs to the peritoneal surface. Intraperitoneal (IP) chemotherapy allows close contact of high concentrations of therapeutic agents with cancer cells in the peritoneal cavity to prolong patient survival. However, conventional IP chemotherapy is prone to rapid elimination from the peritoneal cavity and lacks specificity towards cancer cells. To address these challenges, there is an imperative demand for exploiting novel drug delivery systems to enhance drug retention in the peritoneal cavity and target PC cells. Therefore, in this review, we first recapitulate the physiological structures and barriers associated with IP drug delivery, highlighting the in vivo fate of nanoparticles (NPs) after IP administration. Furthermore, the influence of physicochemical properties (particle size, charge, surface modification, and carrier composition) on the in vivo fate of NPs is discussed. Perspectives on the rational design of NPs for IP therapy and recent clinical progress are also provided.
Collapse
Affiliation(s)
- Shu Pan
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, Liaoning, PR China
| | - Haoyang Yuan
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, Liaoning, PR China
| | - Qiyao Zhai
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, Liaoning, PR China
| | - Yu Zhang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, Liaoning, PR China
| | - Haibing He
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, Liaoning, PR China
| | - Tian Yin
- School of Functional Food and Wine, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, Liaoning, PR China
| | - Xing Tang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, Liaoning, PR China.
| | - Jingxin Gou
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, Liaoning, PR China.
| |
Collapse
|
2
|
Berger JM, Preusser M, Berghoff AS, Bergen ES. Malignant ascites: Current therapy options and treatment prospects. Cancer Treat Rev 2023; 121:102646. [PMID: 39492370 DOI: 10.1016/j.ctrv.2023.102646] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 10/18/2023] [Accepted: 10/22/2023] [Indexed: 11/05/2024]
Abstract
Ascites formation is a common complication of cancer with a significant symptomatic burden for patients. Malignant ascites (MA) is defined by the presence of tumor cells within the ascitic fluid. It does not only cause substantial morbidity, but is also associated with impaired survival. Considering the frequent occurrence of MA, it still represents a clinical challenge for physicians with limited therapy options, mainly comprising of the treatment of the primary tumor and effusion drainage. Particularly the lack of pathophysiological insight limits the development of effective, causative therapies. Causes of MA development such as lymphatic vessel obstruction and the effects of tumor secreted vascular endothelial growth factor (VEGF) have been known for decades. Novel research suggests that the intraperitoneal immune system may also induce and maintain MA accumulation. In this review, we assess current knowledge on the pathophysiology of MA and summarize available evidence of treatment approaches. Also, factors contributing to ascites formation without proof of tumor cells in the peritoneal cavity, defined as paramalignant ascites, with potential treatment strategies are discussed. We further focus on novel findings in the pathophysiology of MA that might lead to treatment improvement in the near future and discussed relevant knowledge gaps in this field.
Collapse
Affiliation(s)
- Julia M Berger
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria; Christian Doppler Laboratory for Personalized Immunotherapy, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Matthias Preusser
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria; Christian Doppler Laboratory for Personalized Immunotherapy, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Anna S Berghoff
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria; Christian Doppler Laboratory for Personalized Immunotherapy, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Elisabeth S Bergen
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
3
|
Liu K, Xu P, Lv J, Ge H, Yan Z, Huang S, Li B, Xu H, Yang L, Xu Z, Zhang D. Peritoneal high-fat environment promotes peritoneal metastasis of gastric cancer cells through activation of NSUN2-mediated ORAI2 m5C modification. Oncogene 2023:10.1038/s41388-023-02707-5. [PMID: 37130916 DOI: 10.1038/s41388-023-02707-5] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 04/19/2023] [Accepted: 04/21/2023] [Indexed: 05/04/2023]
Abstract
Peritoneal metastasis (PM) is an important metastatic modality of gastric cancer (GC).It is associated with poor prognosis. The underlying molecular mechanism of PM remains elusive. 5-Methylcytosine (m5C), a posttranscriptional RNA modification, involves in the progression of many tumors. However, its role in GC peritoneal metastasis remains unclear. In our study, transcriptome results suggested that NSUN2 expression was significantly upregulated in PM. And patients with high NSUN2 expression of PM predicted a worse prognosis. Mechanistically, NSUN2 regulates ORAI2 mRNA stability by m5C modification, thereby promoting ORAI2 expression and further promoting peritoneal metastasis and colonization of GC. YBX1 acts as a "reader" by binding to the ORAI2 m5C modification site. Following the uptake of fatty acids from omental adipocytes by GC cells, the transcription factor E2F1 was upregulated, which further promoted the expression of NSUN2 through cis-element. Briefly, these results revealed that peritoneal adipocytes provide fatty acid for GC cells, thus contributing to the elevation of E2F1 and NSUN2 through AMPK pathway, and upregulated NSUN2 activates the key gene ORAI2 through m5C modification, thereby promoting peritoneal metastasis and colonization of gastric cancer.
Collapse
Affiliation(s)
- Kanghui Liu
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Peng Xu
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Jialun Lv
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Han Ge
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Zhengyuan Yan
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
- Department of Surgery, Nanjing Lishui People's Hospital, Nanjing, 211200, China
| | - Shansong Huang
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Bowen Li
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Hao Xu
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Li Yang
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Zekuan Xu
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Diancai Zhang
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China.
| |
Collapse
|
4
|
Al-Marzouki L, Stavrakos VS, Pal S, Giannias B, Bourdeau F, Rayes R, Bertos N, Najmeh S, Spicer JD, Cools-Lartigue J, Bailey SD, Ferri L, Sangwan V. Soluble factors in malignant ascites promote the metastatic adhesion of gastric adenocarcinoma cells. Gastric Cancer 2023; 26:55-68. [PMID: 36059037 DOI: 10.1007/s10120-022-01338-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 08/25/2022] [Indexed: 02/07/2023]
Abstract
BACKGROUND Adenocarcinoma of the proximal stomach is the fastest rising malignancy in North America. It is commonly associated with peritoneal accumulation of malignant ascites (MA), a fluid containing cancer and inflammatory cells and soluble proteins. Peritoneal metastasis (PM) is the most common site of gastric cancer (GC) progression after curative-intent surgery and is the leading cause of death among GC patients. METHODS/RESULTS Using a panel of gastric adenocarcinoma cell lines (human: MKN 45, SNU-5; murine: NCC-S1M), we demonstrate that prior incubation of GC cells with MA results in a significant (> 1.7-fold) increase in the number of cells capable of adhering to human peritoneal mesothelial cells (HPMC) (p < 0.05). We then corroborate these findings using an ex vivo PM model and show that MA also significantly enhances the ability of GC cells to adhere to strips of human peritoneum (p < 0.05). Using a multiplex ELISA, we identify MIF and VEGF as consistently elevated across MA samples from GC patients (p < 0.05). We demonstrate that agents that block the effects of MIF or VEGF abrogate the ability of MA to stimulate the adhesion of GC cells to adhere to human peritoneum and promote both ex vivo and in vivo metastases. CONCLUSION Agents targeting MIF or VEGF may be relevant to the treatment or prevention of PM in GC patients.
Collapse
Affiliation(s)
- Luai Al-Marzouki
- Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Division of Experimental Surgery, Department of Medicine, McGill University, Montreal, QC, Canada
| | - Vivian S Stavrakos
- Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Division of Experimental Surgery, Department of Medicine, McGill University, Montreal, QC, Canada
| | - Sanjima Pal
- Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Betty Giannias
- Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - France Bourdeau
- Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Roni Rayes
- Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Nicholas Bertos
- Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Sara Najmeh
- Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Division of Experimental Surgery, Department of Medicine, McGill University, Montreal, QC, Canada
- Division of Thoracic and Upper GI Surgery, Montreal General Hospital, 1650 Cedar Avenue, Room L8-325, Montreal, QC, H3G 1A4, Canada
| | - Jonathan D Spicer
- Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Division of Experimental Surgery, Department of Medicine, McGill University, Montreal, QC, Canada
- Division of Thoracic and Upper GI Surgery, Montreal General Hospital, 1650 Cedar Avenue, Room L8-325, Montreal, QC, H3G 1A4, Canada
| | - Jonathan Cools-Lartigue
- Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Division of Experimental Surgery, Department of Medicine, McGill University, Montreal, QC, Canada
- Division of Thoracic and Upper GI Surgery, Montreal General Hospital, 1650 Cedar Avenue, Room L8-325, Montreal, QC, H3G 1A4, Canada
| | - Swneke D Bailey
- Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Division of Experimental Surgery, Department of Medicine, McGill University, Montreal, QC, Canada
| | - Lorenzo Ferri
- Research Institute of the McGill University Health Centre, Montreal, QC, Canada.
- Division of Experimental Surgery, Department of Medicine, McGill University, Montreal, QC, Canada.
- Division of Thoracic and Upper GI Surgery, Montreal General Hospital, 1650 Cedar Avenue, Room L8-325, Montreal, QC, H3G 1A4, Canada.
| | - Veena Sangwan
- Research Institute of the McGill University Health Centre, Montreal, QC, Canada.
- Division of Experimental Surgery, Department of Medicine, McGill University, Montreal, QC, Canada.
| |
Collapse
|
5
|
ADAM10 and ADAM17 as Biomarkers Linked to Inflammation, Metabolic Disorders and Colorectal Cancer. Curr Issues Mol Biol 2022; 44:4517-4527. [PMID: 36286024 PMCID: PMC9600049 DOI: 10.3390/cimb44100309] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 09/15/2022] [Accepted: 09/22/2022] [Indexed: 11/18/2022] Open
Abstract
ADAM10 and ADAM17 have a role in inflammation and diseases associated with inflammation, such as diabetes, cardiovascular diseases (CVD) or cancer, e.g., colorectal cancer (CRC). The aim of this study was to evaluate whether ADAM10 and ADAM17 could be biomarkers of CRC. To achieve this goal, CRC tumors and a surgical margin from 72 patients with CRC were collected. The concentration of ADAM proteins was measured by the ELISA method. Results were analyzed statistically and compared with selected clinical parameters. We found that ADAM17 protein concentration in the tumor samples was higher in patients with diabetes mellitus type 2 (DMT2) (0.28 vs. 0.2 ng/µg protein; p = 0.01) and in the surgical margin was higher both in patients with coexisting DMT2 (0.22 vs. 0.16 ng/µg protein; p < 0.05) and CVD (0.21 vs. 0.13 ng/µg protein; p < 0.01). The concentration of ADAM10 was higher in the surgical margin than in the tumor (249.34 vs. 228.82 pg/µg protein), and the concentration of ADAM17 was higher in the tumor than in the margin (0.23 vs. 0.18 ng/µg protein), but results were not statistically significant. In conclusion, the results of our study indicate that ADAM10 and ADAM17 may be potential biomarkers in cancer linked with DMT2 and CVD as diseases associated with inflammation.
Collapse
|
6
|
Guo L, Qin X, Xue L, Yang JY, Zhang Y, Zhu S, Ye G, Tang R, Yang W. A novel form of docetaxel polymeric micelles demonstrates anti-tumor and ascites-inhibitory activities in animal models as monotherapy or in combination with anti-angiogenic agents. Front Pharmacol 2022; 13:964076. [PMID: 36091776 PMCID: PMC9449419 DOI: 10.3389/fphar.2022.964076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 07/29/2022] [Indexed: 11/30/2022] Open
Abstract
Malignant ascites (MA) is caused by intraperitoneal spread of solid tumor cells and results in a poor quality of life. Chemotherapy is a common first-line treatment for patients with MA. Taxotere ® (DTX) is widely used in solid tumor therapies. However, the low water solubility and side effects caused by additives in the formulation restrict the clinical application of docetaxel. HT001 is a clinical stage docetaxel micelle developed to overcome the solubility issue with improved safety profiles. To support clinical development and expand clinical application of HT001, this study used in vitro and in vivo approaches to investigate the anti-tumor effects of HT001 when applied as monotherapy or in combination with anti-angiogenic agents. HT001 demonstrated comparable anti-proliferative activities as docetaxel in a broad range of cancer cell lines in vitro. Furthermore, HT001 suppressed tumor growth in a dose-dependent manner in A549, MCF-7, and SKOV-3 xenograft tumor mouse models in vivo. In a hepatocellular carcinoma H22 malignant ascites-bearing mouse model, HT001 presented a dose-dependent inhibition of ascites production, prolonged animal survival, and reduced VEGF levels. When dosed at 20 mg/kg, the HT001-treated group exhibited curative results, with no ascites formation in 80% of mice at the end of the study while all the mice in the vehicle control group succumbed. Similar results were obtained in HT001 treatment of mice bearing malignant ascites produced by human ovarian cancer ES-2 cells. Notably, the combination of HT001 with Endostar not only significantly reduced ascites production but also prolonged survival of H22 ascites-bearing mice. HT001 showed similar PK and tissue distribution profiles as DTX in non-rodent hosts. Collectively, these results demonstrate potent anti-tumor activity of HT001 in multiple solid tumor models or malignant ascites models, and reveal synergistic effects with anti-angiogenic agents, supporting the clinical development and clinical expansion plans for HT001.
Collapse
Affiliation(s)
- Leilei Guo
- State Key Laboratory of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Pharmaceutical Co. Ltd, Nanjing, China
| | - Xiaokang Qin
- State Key Laboratory of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Pharmaceutical Co. Ltd, Nanjing, China
| | - Liting Xue
- State Key Laboratory of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Pharmaceutical Co. Ltd, Nanjing, China
| | - Janine Y. Yang
- Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, United States
| | - Yumei Zhang
- Department of Oncology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Shunwei Zhu
- State Key Laboratory of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Pharmaceutical Co. Ltd, Nanjing, China
| | - Gang Ye
- State Key Laboratory of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Pharmaceutical Co. Ltd, Nanjing, China
| | - Renhong Tang
- State Key Laboratory of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Pharmaceutical Co. Ltd, Nanjing, China
- *Correspondence: WenQing Yang, ; Renhong Tang,
| | - WenQing Yang
- State Key Laboratory of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Pharmaceutical Co. Ltd, Nanjing, China
- *Correspondence: WenQing Yang, ; Renhong Tang,
| |
Collapse
|
7
|
Dong S, Liu B, Hu S, Guo F, Zhong Y, Cai Q, Zhang S, Qian Y, Wang J, Zhou F. A novel oncolytic virus induces a regional cytokine storm and safely eliminates malignant ascites of colon cancer. Cancer Med 2022; 11:4297-4309. [PMID: 35510373 DOI: 10.1002/cam4.4772] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 03/23/2022] [Accepted: 04/12/2022] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Given malignant ascites with a terrible prognosis and a unique immune microenvironment, our purpose is to evaluate whether oncolytic herpes simplex virus type 2(OH2) is able to safely eliminate ascites of colon cancer and through which specific mechanism it exerts antitumor immunity. METHODS We established an ascites mice model through intraperitoneal injection of CT26 cells and obtained an appropriate dose range for in vivo tests. Efficacy and safety of OH2 were detected by weight of ascites, blood routine analysis, histopathological examination, and the survival time of mice. The specific mechanism underlying antitumor immunity was analyzed by cytometric bead array, flow cytometry, and single-cell RNA sequencing. Furthermore, anti-interleukin (IL)-6R antibody tocilizumab was synchronously or sequentially delivered with OH2 to explore the role of the regional cytokine storm, mainly IL-6 hypersecretion. RESULTS OH2 was able to eliminate ascites and significantly prolong the survival of mice-bearing CT26 tumor cells by intraperitoneal injection, without obvious systemic damage to the main organs even though a regional cytokine storm. Hypersecretion of pro-inflammatory cytokines, mainly IL-6, and increased infiltration of CD4+ and CD8+ T cells were observed in ascites mice treated by OH2, compared with those treated by 5-fluorouracil or nonresponders. Furthermore, the initial-stage blocking of the IL-6 pathway was able to considerably suppress antitumor immune responses driven by OH2. Surprisingly, we discovered upregulations of the immune checkpoint genes such as Cd274 and Pdcd1 by single-cell RNA sequencing. CONCLUSIONS OH2 could safely eliminate malignant ascites of colon cancer and convert the cold immune microenvironment by inducing a remarkably regional cytokine storm in ascites, mainly IL-6, in the early stage of antitumor immune responses beyond directed oncolytic virotherapy.
Collapse
Affiliation(s)
- Shuang Dong
- Hubei Key Laboratory of Tumor Biological Behaviors, Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China.,Department of Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Binlei Liu
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei Provincial Cooperative Innovation Center of Industrial Fermentation, Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, Hubei, China
| | - Sheng Hu
- Department of Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Fang Guo
- Department of Pathology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yi Zhong
- Department of Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Qian Cai
- Department of Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Siqi Zhang
- School of Pharmacy, Hubei University of Science and Technology, Xianning, Hubei, China
| | - Yu Qian
- Department of Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jun Wang
- Department of Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Fuxiang Zhou
- Hubei Key Laboratory of Tumor Biological Behaviors, Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
8
|
Kasai S, Kuwayama N, Motoo Y, Kawashima A, Matsumoto K, Yano S, Matsushima K, Yasumoto K. Dual blockade of MET and VEGFR2 signaling pathways as a potential therapeutic maneuver for peritoneal carcinomatosis in scirrhous gastric cancer. Biochem Biophys Res Commun 2022; 600:80-86. [DOI: 10.1016/j.bbrc.2022.02.045] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 02/04/2022] [Accepted: 02/11/2022] [Indexed: 12/15/2022]
|
9
|
Hendrikson J, Liu Y, Ng WH, Lee JY, Lim AH, Loh JW, Ng CC, Ong WS, Tan JWS, Tan QX, Ng G, Shannon NB, Lim WK, Lim TK, Chua C, Wong JSM, Tan GHC, So JBY, Yeoh KG, Teh BT, Chia CS, Soo KC, Kon OL, Tan IB, Chan JY, Teo MCC, Ong CAJ. Ligand-mediated PAI-1 inhibition in a mouse model of peritoneal carcinomatosis. Cell Rep Med 2022; 3:100526. [PMID: 35243423 PMCID: PMC8861959 DOI: 10.1016/j.xcrm.2022.100526] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 11/22/2021] [Accepted: 01/19/2022] [Indexed: 12/28/2022]
Abstract
Peritoneal carcinomatosis (PC) present a ubiquitous clinical conundrum in all intra-abdominal malignancies. Via functional and transcriptomic experiments of ascites-treated PC cells, we identify STAT3 as a key signaling pathway. Integrative analysis of publicly available databases and correlation with clinical cohorts (n = 7,359) reveal putative clinically significant activating ligands of STAT3 signaling. We further validate a 3-biomarker prognostic panel in ascites independent of clinical covariates in a prospective study (n = 149). Via single-cell sequencing experiments, we uncover that PAI-1, a key component of the prognostic biomarker panel, is largely secreted by fibroblasts and mesothelial cells. Molecular stratification of ascites using PAI-1 levels and STAT3 activation in ascites-treated cells highlight a therapeutic opportunity based on a phenomenon of paracrine addiction. These results are recapitulated in patient-derived ascites-dependent xenografts. Here, we demonstrate therapeutic proof of concept of direct ligand inhibition of a prognostic target within an enclosed biological space.
Collapse
Affiliation(s)
- Josephine Hendrikson
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, National Cancer Centre Singapore, 11 Hospital Cresent, Singapore 169610, Singapore
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, Singapore General Hospital, Singapore 169608, Singapore
- Laboratory of Applied Human Genetics, Division of Medical Sciences, National Cancer Centre Singapore, Singapore 169610, Singapore
| | - Ying Liu
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, National Cancer Centre Singapore, 11 Hospital Cresent, Singapore 169610, Singapore
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, Singapore General Hospital, Singapore 169608, Singapore
- Laboratory of Applied Human Genetics, Division of Medical Sciences, National Cancer Centre Singapore, Singapore 169610, Singapore
| | - Wai Har Ng
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, National Cancer Centre Singapore, 11 Hospital Cresent, Singapore 169610, Singapore
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, Singapore General Hospital, Singapore 169608, Singapore
- Laboratory of Applied Human Genetics, Division of Medical Sciences, National Cancer Centre Singapore, Singapore 169610, Singapore
| | - Jing Yi Lee
- Cancer Discovery Hub, National Cancer Centre Singapore, Singapore 169610, Singapore
| | - Abner Herbert Lim
- Cancer Discovery Hub, National Cancer Centre Singapore, Singapore 169610, Singapore
| | - Jui Wan Loh
- Cancer Discovery Hub, National Cancer Centre Singapore, Singapore 169610, Singapore
| | - Cedric C.Y. Ng
- Laboratory of Cancer Epigenome, Division of Medical Sciences, National Cancer Centre Singapore, Singapore 169610, Singapore
| | - Whee Sze Ong
- Division of Clinical Trials and Epidemiological Sciences, National Cancer Centre Singapore, Singapore 169610, Singapore
| | - Joey Wee-Shan Tan
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, National Cancer Centre Singapore, 11 Hospital Cresent, Singapore 169610, Singapore
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, Singapore General Hospital, Singapore 169608, Singapore
- Laboratory of Applied Human Genetics, Division of Medical Sciences, National Cancer Centre Singapore, Singapore 169610, Singapore
| | - Qiu Xuan Tan
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, National Cancer Centre Singapore, 11 Hospital Cresent, Singapore 169610, Singapore
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, Singapore General Hospital, Singapore 169608, Singapore
- Laboratory of Applied Human Genetics, Division of Medical Sciences, National Cancer Centre Singapore, Singapore 169610, Singapore
| | - Gillian Ng
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, National Cancer Centre Singapore, 11 Hospital Cresent, Singapore 169610, Singapore
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, Singapore General Hospital, Singapore 169608, Singapore
- Laboratory of Applied Human Genetics, Division of Medical Sciences, National Cancer Centre Singapore, Singapore 169610, Singapore
| | - Nicholas B. Shannon
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, National Cancer Centre Singapore, 11 Hospital Cresent, Singapore 169610, Singapore
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, Singapore General Hospital, Singapore 169608, Singapore
- Laboratory of Applied Human Genetics, Division of Medical Sciences, National Cancer Centre Singapore, Singapore 169610, Singapore
| | - Weng Khong Lim
- SingHealth Duke-NUS Institute of Precision Medicine, National Heart Centre Singapore, Singapore 169609, Singapore
- Cancer and Stem Biology Signature Research Program, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Tony K.H. Lim
- Department of Anatomical Pathology, Singapore General Hospital, Singapore 169856, Singapore
- Pathology Academic Clinical Program, SingHealth Duke-NUS Academic Medical Centre, Singapore 168753, Singapore
| | - Clarinda Chua
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore 169610, Singapore
| | - Jolene Si Min Wong
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, National Cancer Centre Singapore, 11 Hospital Cresent, Singapore 169610, Singapore
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, Singapore General Hospital, Singapore 169608, Singapore
- SingHealth Duke-NUS Oncology Academic Clinical Program, Duke-NUS Medical School, Singapore 169857, Singapore
- SingHealth Duke-NUS Surgery Academic Clinical Program, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Grace Hwei Ching Tan
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, National Cancer Centre Singapore, 11 Hospital Cresent, Singapore 169610, Singapore
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, Singapore General Hospital, Singapore 169608, Singapore
| | - Jimmy Bok Yan So
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119077, Singapore
- Division of Surgical Oncology, National University Cancer Institute, National University Health System, Singapore 119074, Singapore
| | - Khay Guan Yeoh
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119077, Singapore
- Department of Gastroenterology and Hepatology, National University Hospital, Singapore 119074, Singapore
| | - Bin Tean Teh
- Laboratory of Cancer Epigenome, Division of Medical Sciences, National Cancer Centre Singapore, Singapore 169610, Singapore
- Institute of Molecular and Cell Biology, A∗STAR Research Entities, Singapore 138673, Singapore
| | - Claramae Shulyn Chia
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, National Cancer Centre Singapore, 11 Hospital Cresent, Singapore 169610, Singapore
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, Singapore General Hospital, Singapore 169608, Singapore
- SingHealth Duke-NUS Oncology Academic Clinical Program, Duke-NUS Medical School, Singapore 169857, Singapore
- SingHealth Duke-NUS Surgery Academic Clinical Program, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Khee Chee Soo
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, National Cancer Centre Singapore, 11 Hospital Cresent, Singapore 169610, Singapore
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, Singapore General Hospital, Singapore 169608, Singapore
| | - Oi Lian Kon
- Laboratory of Applied Human Genetics, Division of Medical Sciences, National Cancer Centre Singapore, Singapore 169610, Singapore
| | - Iain Beehuat Tan
- Cancer and Stem Biology Signature Research Program, Duke-NUS Medical School, Singapore 169857, Singapore
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore 169610, Singapore
- Laboratory of Applied Cancer Genomics, Genome Institute of Singapore, A∗STAR Research Entities, Singapore 138672, Singapore
| | - Jason Yongsheng Chan
- Cancer Discovery Hub, National Cancer Centre Singapore, Singapore 169610, Singapore
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore 169610, Singapore
| | - Melissa Ching Ching Teo
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, National Cancer Centre Singapore, 11 Hospital Cresent, Singapore 169610, Singapore
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, Singapore General Hospital, Singapore 169608, Singapore
| | - Chin-Ann J. Ong
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, National Cancer Centre Singapore, 11 Hospital Cresent, Singapore 169610, Singapore
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, Singapore General Hospital, Singapore 169608, Singapore
- Laboratory of Applied Human Genetics, Division of Medical Sciences, National Cancer Centre Singapore, Singapore 169610, Singapore
- SingHealth Duke-NUS Oncology Academic Clinical Program, Duke-NUS Medical School, Singapore 169857, Singapore
- SingHealth Duke-NUS Surgery Academic Clinical Program, Duke-NUS Medical School, Singapore 169857, Singapore
- Institute of Molecular and Cell Biology, A∗STAR Research Entities, Singapore 138673, Singapore
| | - on behalf of the Singapore Peritoneal Oncology Study (SPOS) Group and Singapore Gastric Cancer Consortium (SGCC)
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, National Cancer Centre Singapore, 11 Hospital Cresent, Singapore 169610, Singapore
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, Singapore General Hospital, Singapore 169608, Singapore
- Laboratory of Applied Human Genetics, Division of Medical Sciences, National Cancer Centre Singapore, Singapore 169610, Singapore
- Cancer Discovery Hub, National Cancer Centre Singapore, Singapore 169610, Singapore
- Laboratory of Cancer Epigenome, Division of Medical Sciences, National Cancer Centre Singapore, Singapore 169610, Singapore
- Division of Clinical Trials and Epidemiological Sciences, National Cancer Centre Singapore, Singapore 169610, Singapore
- SingHealth Duke-NUS Institute of Precision Medicine, National Heart Centre Singapore, Singapore 169609, Singapore
- Cancer and Stem Biology Signature Research Program, Duke-NUS Medical School, Singapore 169857, Singapore
- Department of Anatomical Pathology, Singapore General Hospital, Singapore 169856, Singapore
- Pathology Academic Clinical Program, SingHealth Duke-NUS Academic Medical Centre, Singapore 168753, Singapore
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore 169610, Singapore
- SingHealth Duke-NUS Oncology Academic Clinical Program, Duke-NUS Medical School, Singapore 169857, Singapore
- SingHealth Duke-NUS Surgery Academic Clinical Program, Duke-NUS Medical School, Singapore 169857, Singapore
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119077, Singapore
- Division of Surgical Oncology, National University Cancer Institute, National University Health System, Singapore 119074, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119077, Singapore
- Department of Gastroenterology and Hepatology, National University Hospital, Singapore 119074, Singapore
- Institute of Molecular and Cell Biology, A∗STAR Research Entities, Singapore 138673, Singapore
- Laboratory of Applied Cancer Genomics, Genome Institute of Singapore, A∗STAR Research Entities, Singapore 138672, Singapore
| |
Collapse
|
10
|
Matsumoto T, Yamamura S, Ikoma T, Kurioka Y, Doi K, Yasuda T, Boku S, Kawai T, Shibata N, Nagai H, Tsuduki T, Shimada T, Matsumoto Y, Tsumura T, Takatani M, Yasui H, Satake H. Real-World Data of Trifluridine/Tipiracil for Patients With Advanced Gastric Cancer: A Multi-Institutional Retrospective Study. CLINICAL MEDICINE INSIGHTS: ONCOLOGY 2022; 16:11795549221137135. [PMID: 36408335 PMCID: PMC9666830 DOI: 10.1177/11795549221137135] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 10/19/2022] [Indexed: 11/16/2022] Open
Abstract
Background: A trial with trifluridine/tipiracil (FTD/TPI) versus placebo in patients with
heavily pretreated metastatic gastric cancer showed that FTD/TPI is
effective with manageable toxicity in these patients. However, real-world
data on the effects of FTD/TPI in patients with advanced gastric cancer
(AGC) are limited. Methods: We retrospectively collected and analyzed the clinicopathological data of
patients with AGC who received FTD/TPI monotherapy at our institutions (Kobe
City Medical Center General Hospital, Osaka Red Cross Hospital, Himeji Red
Cross Hospital, and Kansai Medical University Hospital) between September
2019 and July 2021. Tumor responses were evaluated based on the Response
Evaluation Criteria in Solid Tumors, version 1.1. Overall survival (OS) and
progression-free survival were estimated using the Kaplan-Meier method. Results: A total of 53 patients were included in the study. The median age was 70
(range, 37-85) years; 39 patients (74%) were men; the numbers of patients
with Eastern Cooperative Oncology Group performance status scores of 0, 1,
and 2 were 10 (19%), 39 (74%), and 4 (8%), respectively; and 27 patients
(51%) had diffuse-type histology. A total of 29 patients (56%) had ascites.
Prior nivolumab therapy was administered to 49 patients (92%). The response
rate and disease control rate (DCR) were 2% and 35%, respectively. The
median progression-free survival was 2.4 months, and OS was 5.8 months.
Patients with ascites exhibited significantly shorter OS (8.6 vs 4.7 months,
P = .0291) than those without ascites, and DCR (54% vs
18%, P = .0055) was significantly worse in patients with
ascites. There was no significant difference in the frequency of adverse
events of grade 3 or higher between patients with and without ascites. Conclusion: In a real-world setting, FTD/TPI has similar effectiveness as late-line
chemotherapy for patients with AGC, including those who previously had
received nivolumab.
Collapse
Affiliation(s)
- Toshihiko Matsumoto
- Department of Medical Oncology, Kobe City Medical Center General Hospital, Kobe, Japan
- Department of Internal Medicine, Japanese Red Cross Society Himeji Hospital, Himeji, Japan
- Cancer Treatment Center, Kansai Medical University, Hirakata, Japan
| | - Shogo Yamamura
- Department of Medical Oncology, Kobe City Medical Center General Hospital, Kobe, Japan
| | - Tatsuki Ikoma
- Cancer Treatment Center, Kansai Medical University, Hirakata, Japan
- Department of Clinical Oncology, Osaka Red Cross Hospital, Osaka, Japan
| | - Yusuke Kurioka
- Department of Internal Medicine, Japanese Red Cross Society Himeji Hospital, Himeji, Japan
- Department of Medical Oncology, Kochi Medical School, Nankoku, Japan
| | - Keitaro Doi
- Department of Clinical Oncology, Osaka Red Cross Hospital, Osaka, Japan
| | - Tomoyo Yasuda
- Cancer Treatment Center, Kansai Medical University, Hirakata, Japan
| | - Shogen Boku
- Cancer Treatment Center, Kansai Medical University, Hirakata, Japan
| | - Takashi Kawai
- Department of Surgery, Japanese Red Cross Society Himeji Hospital, Himeji, Japan
| | - Nobuhiro Shibata
- Cancer Treatment Center, Kansai Medical University, Hirakata, Japan
| | - Hiroki Nagai
- Department of Medical Oncology, Kobe City Medical Center General Hospital, Kobe, Japan
| | - Takao Tsuduki
- Department of Internal Medicine, Japanese Red Cross Society Himeji Hospital, Himeji, Japan
| | - Takanobu Shimada
- Department of Clinical Oncology, Osaka Red Cross Hospital, Osaka, Japan
| | - Yusuke Matsumoto
- Department of Surgery, Japanese Red Cross Society Himeji Hospital, Himeji, Japan
| | - Takehiko Tsumura
- Department of Clinical Oncology, Osaka Red Cross Hospital, Osaka, Japan
| | - Masahiro Takatani
- Department of Internal Medicine, Japanese Red Cross Society Himeji Hospital, Himeji, Japan
| | - Hisateru Yasui
- Department of Medical Oncology, Kobe City Medical Center General Hospital, Kobe, Japan
| | - Hironaga Satake
- Department of Medical Oncology, Kobe City Medical Center General Hospital, Kobe, Japan
- Department of Medical Oncology, Kochi Medical School, Nankoku, Japan
| |
Collapse
|
11
|
Adamalizyny jako potencjalne biomarkery w wybranych nowotworach złośliwych przewodu pokarmowego. POSTEP HIG MED DOSW 2021. [DOI: 10.2478/ahem-2021-0020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Abstrakt
Nowotwory złośliwe przewodu pokarmowego zajmują czołowe miejsce zarówno wśród przyczyn zachorowań jak i zgonów z powodu chorób nowotworowych na świecie. Wciąż poszukuje się potencjalnych biomarkerów, które mogłyby posłużyć jako czynniki predykcyjne i prognostyczne w tych nowotworach. Wśród białek, które mogłyby pełnić taką rolę, wymienia się adamalizyny. Liczne białka z tej rodziny są zaangażowane w wielu etapach nowotworzenia, od procesu różnicowania się pojedynczych komórek, wzrost i progresję guza do tworzenia przerzutów odległych. Dzieje się to m.in. poprzez ścieżki sygnałowe związane z aktywacją insulinopodobnych czynników wzrostu, naskórkowych czynników wzrostu czy oddziaływanie na czynnik martwicy nowotworu TNF-α. Szczególnie istotna w wyjaśnieniu patomechanizmu rozwoju raków gruczołowych przewodu pokarmowego wydaje się ścieżka sygnałowa związana z aktywacją cytokin prozapalnych. Przewlekły stan zapalny jest bowiem dobrze udokumentowanym czynnikiem ryzyka rozwoju tej grupy chorób nowotworowych.
Poznanie roli białek z rodziny adamalizyn w rozwoju i patogenezie nowotworów złośliwych przewodu pokarmowego wymaga wciąż dalszych badań. W artykule podjęto próbę syntezy aktualnej wiedzy na temat wykorzystania wybranych białek z rodziny adamalizyn jako biomarkerów nowotworów złośliwych przewodu pokarmowego.
Collapse
|
12
|
Zhang S, Xie B, Wang L, Yang H, Zhang H, Chen Y, Wang F, Liu C, He H. Macrophage-mediated vascular permeability via VLA4/VCAM1 pathway dictates ascites development in ovarian cancer. J Clin Invest 2021; 131:140315. [PMID: 33295887 DOI: 10.1172/jci140315] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 11/25/2020] [Indexed: 12/14/2022] Open
Abstract
The development of ascites correlates with advanced stage disease and poor prognosis in ovarian cancer. Vascular permeability is the key pathophysiological change involved in ascites development. Previously, we provided evidence that perivascular M2-like macrophages protect the vascular barrier through direct contact with endothelial cells (ECs). Here, we investigated the molecular mechanism and its clinical significance in the ovarian cancer setting. We found that upon direct coculture with the endothelium, M2 macrophages tuned down their VLA4 and reduced the levels of VCAM1 in ECs. On the other hand, ectopically overexpressing VLA4 in macrophages or VCAM1 in ECs induced hyperpermeability. Mechanistically, downregulation of VLA4 or VCAM1 led to reduced levels of RAC1 and ROS, which resulted in decreased phosphorylation of PYK2 (p-PYK2) and VE-cadherin (p-VE-cad), hence enhancing cell adhesion. Furthermore, targeting the VLA4/VCAM1 axis augmented vascular integrity and abrogated ascites formation in vivo. Finally, VLA4 expression on the macrophages isolated from ascites dictated permeability ex vivo. Importantly, VLA4 antibody acted synergistically with bevacizumab to further enhance the vascular barrier. Taking these data together, we reveal here that M2 macrophages regulate the vascular barrier though the VCAM1/RAC1/ROS/p-PYK2/p-VE-cad cascade, which provides specific therapeutic targets for the treatment of malignant ascites.
Collapse
Affiliation(s)
- Shibo Zhang
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| | - Bingfan Xie
- Department of Gynaecology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Lijie Wang
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| | - Hua Yang
- Department of Gynaecology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Haopei Zhang
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| | - Yuming Chen
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| | - Feng Wang
- Department of Gynaecology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Changqing Liu
- Department of Gynaecology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Huanhuan He
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| |
Collapse
|
13
|
Chen MH, Lu SN, Chen CH, Lin PC, Jiang JK, D’yachkova Y, Lukanowski M, Cheng R, Chen LT. How May Ramucirumab Help Improve Treatment Outcome for Patients with Gastrointestinal Cancers? Cancers (Basel) 2021; 13:3536. [PMID: 34298750 PMCID: PMC8306041 DOI: 10.3390/cancers13143536] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/24/2021] [Accepted: 07/09/2021] [Indexed: 12/24/2022] Open
Abstract
GI cancers are characterized by high recurrence rates and a dismal prognosis and there is an urgent need for new therapeutic approaches. This is a narrative review designed to provide a summary of the efficacy as measured by overall survival, progression free survival, and safety data from phase 3 randomized controlled GI clinical trials of ramucirumab including those from important pre-specified patient subgroups and evidence from real clinical practice worldwide. Quality of life (QOL) is discussed where data are available. Our aim was to summarize the efficacy and safety of ramucirumab in the treatment of GI cancers using these existing published data with a view to demonstrating how ramucirumab may help improve treatment outcome for patients with GI cancers. The data indicate that ramucirumab is efficacious, safe, and tolerable across the intent-to-treat patient populations as a whole and across several pre-specified subgroups, even those whose disease is traditionally more difficult to treat. Furthermore, survival outcomes observed in real-world clinical practice demonstrate similar data from phase 3 clinical trials even in patients with complications, suggesting that the benefits of ramucirumab translate in actual clinical practice.
Collapse
Affiliation(s)
- Ming-Huang Chen
- Taipei Veterans General Hospital, Taipei 11217, Taiwan; (M.-H.C.); (J.-K.J.)
| | - Sheng-Nan Lu
- Kaohsiung Chang Gung Memorial Hospital, Kaohsiung City 83301, Taiwan;
| | - Chien-Hung Chen
- Department of Internal Medicine, National Taiwan University Hospital, Douliu 64041, Taiwan;
| | - Peng-Chan Lin
- National Cheng Kung University Hospital, National Cheng Kung University, Tainan 70403, Taiwan;
| | - Jeng-Kai Jiang
- Taipei Veterans General Hospital, Taipei 11217, Taiwan; (M.-H.C.); (J.-K.J.)
| | | | - Mariusz Lukanowski
- Global Medical Affairs, Eli Lilly Denmark, Hovedstaden, 2730 Herlev, Denmark;
| | - Rebecca Cheng
- Eli Lilly and Company (Taiwan) Inc., Taipe City 10543, Taiwan;
| | - Li-Tzong Chen
- National Cheng Kung University Hospital, National Cheng Kung University, Tainan 70403, Taiwan;
- National Institute of Cancer Research, National Health Research Institutes, Tainan 70456, Taiwan
- Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung City 80756, Taiwan
| |
Collapse
|
14
|
Anti-Angiogenic Treatment in Pseudomyxoma Peritonei-Still a Strong Preclinical Rationale. Cancers (Basel) 2021; 13:cancers13112819. [PMID: 34198773 PMCID: PMC8201024 DOI: 10.3390/cancers13112819] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 06/02/2021] [Accepted: 06/02/2021] [Indexed: 12/30/2022] Open
Abstract
Simple Summary Patients with pseudomyxoma peritonei that are not cured by the standard treatment (cytoreductive surgery and hyperthermic intraperitoneal chemotherapy) have no efficacious treatment options. Drugs that inhibit formation of new vessels (anti-angiogenic drugs) could be a therapeutic option for these patients. Using patient samples and animal models we show that angiogenesis is important in pseudomyxoma peritonei and that anti-angiogenic drugs may indeed have an effect. Our results support continued efforts to determine the role of anti-angiogenic treatment in pseudomyxoma peritonei. Abstract Pseudomyxoma peritonei (PMP) is a rare, slow-growing cancer characterized by progressive accumulation of intraperitoneal mucinous tumor deposits. Cytoreductive surgery and hyperthermic intraperitoneal chemotherapy (HIPEC) cures approximately 50% of patients, but in unresectable and recurrent cases, treatment options are limited. Anti-angiogenic treatment is being explored as a potential therapeutic option. Using PMP patient samples, microvessel densities (immunostaining for CD31 and CD105) and pro-angiogenic factors were analyzed, and the proliferative response upon incubation with human umbilical cord vascular endothelial cells (HUVEC) was determined. Growth inhibition by anti-angiogenic drugs was analyzed in patient-derived xenograft models of PMP. PMP tumor tissues were found to be highly vascularized and contained key pro-angiogenic factors, in particular related to vascular endothelial growth factor (VEGF) signaling, but interestingly, high levels of fibroblast growth factor 2 were also detected. HUVEC proliferation was stimulated upon incubation with fresh tumor samples and the observed proliferation could be inhibited by VEGF pathway inhibitor bevacizumab. In xenograft models the two VEGF pathway inhibitors, bevacizumab and aflibercept, inhibited tumor growth. This work reemphasizes the importance of angiogenesis as a major driver in PMP and strengthens the preclinical rationale for continued exploration of angiogenesis inhibition in the hope of providing novel treatment to a group of patients that have few other treatment options.
Collapse
|
15
|
Yamaguchi T, Kinoshita J, Saito H, Shimada M, Terai S, Moriyama H, Okamoto K, Makino I, Nakamura K, Tajima H, Ninomiya I, Fushida S. High CD8/CD33 ratio in peritoneal metastatic lesions is associated with favorable prognosis in gastric cancer. Cancer Rep (Hoboken) 2021; 4:e1389. [PMID: 33793095 PMCID: PMC8551992 DOI: 10.1002/cnr2.1389] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 03/02/2021] [Accepted: 03/17/2021] [Indexed: 12/13/2022] Open
Abstract
Background Tumor‐infiltrating lymphocytes (TILs) and other immune cells have been reported as a prognostic factor in several tumors, including gastric cancer, and they play an important role in antitumor effect at the primary site. There were few reports on the immune status in peritoneal metastatic lesions for gastric cancer. Aims The aims of this study were to assess the prognostic significance of TILs (CD4, CD8, CD19, regulatory T cells [Tregs]), and myeloid‐derived suppressor cells (MDSCs) in peritoneal metastatic lesions. Methods We retrospectively investigated 60 patients for gastric cancer with peritoneal metastasis who were treated between 2009 and 2016 in our institute. Immunohistochemistry for CD4, CD8, CD19, FOXP3, and CD33 was performed in the peritoneal metastatic lesions. The absolute numbers of immune cells and ratios were evaluated, and the relationship between immune‐related marker and overall survival (OS) was investigated. Results A high infiltration of CD8+ lymphocytes or high CD8/CD33 ratio was a better prognosis for OS in univariate analysis using all immunologic variables (P = .012, P = .001). In multivariate analysis for clinical and immunologic variables, high CD8/CD33 ratio was identified as an independent prognostic factor for OS (Hazard ratio: 0.291, 95% confidence interval: 0.126‐0.670, P = .004). Conclusion High CD8/CD33 ratio and high infiltration of CD8+ lymphocytes in peritoneal metastatic lesions were favorable prognoses for gastric cancer patients with peritoneal metastasis. It is necessary to modify the immune microenvironment result to increase the level of CD8+ lymphocytes in the peritoneal metastatic lesions.
Collapse
Affiliation(s)
- Takahisa Yamaguchi
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Kanazawa University Graduate School of Medical Science, Kanazawa, Japan
| | - Jun Kinoshita
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Kanazawa University Graduate School of Medical Science, Kanazawa, Japan
| | - Hiroto Saito
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Kanazawa University Graduate School of Medical Science, Kanazawa, Japan
| | - Mari Shimada
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Kanazawa University Graduate School of Medical Science, Kanazawa, Japan
| | - Siro Terai
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Kanazawa University Graduate School of Medical Science, Kanazawa, Japan
| | - Hideki Moriyama
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Kanazawa University Graduate School of Medical Science, Kanazawa, Japan
| | - Koichi Okamoto
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Kanazawa University Graduate School of Medical Science, Kanazawa, Japan
| | - Isamu Makino
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Kanazawa University Graduate School of Medical Science, Kanazawa, Japan
| | - Keishi Nakamura
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Kanazawa University Graduate School of Medical Science, Kanazawa, Japan
| | - Hidehiro Tajima
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Kanazawa University Graduate School of Medical Science, Kanazawa, Japan
| | - Itasu Ninomiya
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Kanazawa University Graduate School of Medical Science, Kanazawa, Japan
| | - Sachio Fushida
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Kanazawa University Graduate School of Medical Science, Kanazawa, Japan
| |
Collapse
|
16
|
Kou F, Gong J, Li Y, Li J, Zhang X, Li J, Shen L. Phase I study of intraperitoneal bevacizumab for treating refractory malignant ascites. J Int Med Res 2021; 49:300060520986664. [PMID: 33616416 PMCID: PMC7903826 DOI: 10.1177/0300060520986664] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVE This prospective, dose-escalation phase I study evaluated the safety and efficacy of intraperitoneal bevacizumab in managing refractory malignant ascites and explored the recommended dose of bevacizumab for further study. METHODS Patients with refractory malignant ascites were enrolled. Bevacizumab was intraperitoneal administered weekly at an initial dose of 2.5 mg/kg, with dose escalation to 5 and 7.5 mg/kg performed following the standard "3 + 3" rule. The total duration of treatment was 2 or 3 weeks. RESULTS Between December 2013 and September 2014, 13 patients (2.5 mg/kg, n = 4; 5 mg/kg, n = 3; 7.5 mg/kg, n = 6) with refractory malignant ascites were enrolled. Bevacizumab was well tolerated, and the most common treatment-related adverse events were abdominal pain (5/13), abdominal distension (2/13), and fatigue (2/13). The dose-limiting toxicity at 7.5 mg/kg was grade 3 bowel obstruction (1/13). The maximum tolerated dose (MTD) was not reached. The overall response and disease control rates were 7.7 and 61.5%, respectively. CONCLUSIONS Intraperitoneal bevacizumab safe and well tolerated for treating malignant ascites, and the MTD was not reached at doses of 2.5 to 7.5 mg/kg. Intraperitoneal bevacizumab at 7.5 mg/kg weekly is recommended for further study to verify its anti-tumor activity.Trial registration: Clinical Trials NCT01852409.
Collapse
Affiliation(s)
- Furong Kou
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Jifang Gong
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Yan Li
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Jian Li
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Xiaotian Zhang
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Jie Li
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Lin Shen
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| |
Collapse
|
17
|
Yamaguchi T, Fushida S, Kinoshita J, Okazaki M, Ishikawa S, Ohbatake Y, Terai S, Okamoto K, Nakanuma S, Makino I, Nakamura K, Miyashita T, Tajima H, Takamura H, Ninomiya I, Ohta T. Extravasated platelet aggregation contributes to tumor progression via the accumulation of myeloid-derived suppressor cells in gastric cancer with peritoneal metastasis. Oncol Lett 2020; 20:1879-1887. [PMID: 32724431 PMCID: PMC7377031 DOI: 10.3892/ol.2020.11722] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 05/05/2020] [Indexed: 12/24/2022] Open
Abstract
Extravasated platelet aggregation (EPA) serves an important role in the cancer microenvironment during cancer progression, and has been demonstrated to interact with tumor cells in several types of cancer. EPA induces epithelial-mesenchymal transition (EMT) via transforming growth factor-β, and also recruits immunosuppressive cells, including regulatory T (Treg) cells and myeloid-derived suppressor cells (MDSCs). However, the role of EPA in gastric cancer with peritoneal metastasis remains unknown. The present study analyzed the association between EPA and prognosis in patients with gastric cancer with peritoneal metastasis. The present study evaluated 62 patients diagnosed with advanced gastric cancer with peritoneal metastasis between 2001 and 2016. EPA, EMT, Treg cells and MDSCs in peritoneal metastatic lesions were detected by immunohistochemical evaluation of CD42b, SNAIL, FOXP3 and CD33, respectively. CD42b expression was observed in 56.5% (35/62) of peritoneal metastatic lesions. CD42b expression in peritoneal metastatic lesions was associated with poor overall survival compared with lower frequencies (hazard ratio, 2.03; 95% confidence interval, 1.12-3.69; P=0.018). SNAIL, FOXP3 and CD33 expression were not associated with overall survival, but CD33 expression was markedly higher in CD42b-positive patients (P=0.022). These results indicated that EPA affects immunosuppression by recruiting MDSCs in the tumor microenvironment via the secretion of soluble factors, resulting in tumor progression. EPA may be a novel therapeutic target for gastric cancer with peritoneal metastasis.
Collapse
Affiliation(s)
- Takahisa Yamaguchi
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Ishikawa 920-8641, Japan
| | - Sachio Fushida
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Ishikawa 920-8641, Japan
| | - Jun Kinoshita
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Ishikawa 920-8641, Japan
| | - Mitsuyoshi Okazaki
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Ishikawa 920-8641, Japan
| | - Satoko Ishikawa
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Ishikawa 920-8641, Japan
| | - Yoshinao Ohbatake
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Ishikawa 920-8641, Japan
| | - Shiro Terai
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Ishikawa 920-8641, Japan
| | - Koichi Okamoto
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Ishikawa 920-8641, Japan
| | - Shinichi Nakanuma
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Ishikawa 920-8641, Japan
| | - Isamu Makino
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Ishikawa 920-8641, Japan
| | - Keishi Nakamura
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Ishikawa 920-8641, Japan
| | - Tomoharu Miyashita
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Ishikawa 920-8641, Japan
| | - Hidehiro Tajima
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Ishikawa 920-8641, Japan
| | - Hiroyuki Takamura
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Ishikawa 920-8641, Japan
| | - Itasu Ninomiya
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Ishikawa 920-8641, Japan
| | - Tetsuo Ohta
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Ishikawa 920-8641, Japan
| |
Collapse
|
18
|
Zheng LN, Wen F, Xu P, Zhang S. Prognostic significance of malignant ascites in gastric cancer patients with peritoneal metastasis: A systemic review and meta-analysis. World J Clin Cases 2019; 7:3247-3258. [PMID: 31667175 PMCID: PMC6819285 DOI: 10.12998/wjcc.v7.i20.3247] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 08/16/2019] [Accepted: 09/09/2019] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Recent evidence indicates that malignant ascites may be associated with the high malignancy and poor prognosis of gastric cancer (GC) with peritoneal metastasis (PM), but no robust consensus has been reached until now.
AIM To evaluate the prognostic significance of malignant ascites in GC patients with PM.
METHODS Two independent authors conducted database searches. The searches were performed in the EMBASE, PubMed, and Cochrane Library databases, and the terms used to search included stomach neoplasms, GC, ascites, peritoneal effusion, survival, and survival analysis. Outcomes included overall survival and hazard ratios with 95% confidence intervals (CIs). Three pairs of comparisons for measuring survival were made: (1) Patients with ascites vs those without ascites; (2) Patients with massive ascites vs those with mild to moderate ascites; and (3) Patients with massive ascites vs those with no to moderate ascites.
RESULTS Fourteen articles including fifteen studies were considered in the final analysis. Among them, nine studies assessed the difference in prognosis between patients with and without malignant ascites. A pooled HR of 1.63 (95%CI: 1.47-1.82, P < 0.00001) indicated that GC patients with malignant ascites had a relatively poor prognosis compared to patients without ascites. We also found that the prognosis of GC patients with malignant ascites was related to the volume of ascites in the six other studies.
CONCLUSION GC patients with malignant ascites tend to have a worse prognosis, and the volume of ascites has an impact on GC outcomes.
Collapse
Affiliation(s)
- Ling-Nan Zheng
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Feng Wen
- Department of Abdominal Oncology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Ping Xu
- Sichuan University Library, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Shuang Zhang
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| |
Collapse
|
19
|
Yamasaki F, Kolakshyapati M, Takano M, Yonezawa U, Nishibuchi I, Imano N, Taguchi A, Onishi S, Amatya VJ, Takeshima Y, Nagata Y, Kurisu K, Sugiyama K. Effect of bevacizumab against cystic components of brain tumors. Cancer Med 2019; 8:6519-6527. [PMID: 31498567 PMCID: PMC6825995 DOI: 10.1002/cam4.2537] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 08/22/2019] [Accepted: 08/23/2019] [Indexed: 12/23/2022] Open
Abstract
Background Bevacizumab improves symptoms via reducing the peritumoral edema and/or normalizing blood brain barrier, and occasionally via reducing the tumor size. However, the effect against active cystic components has not been documented yet. Materials and Methods Between 2008 and 2018, 139 patients with primary or metastatic brain tumors were treated with bevacizumab (BEV) in our institution. The images and symptoms before and after administration of BEV were examined, and changes in size of cysts were evaluated as follows: CR (complete disappearance), PR (reduction by ≥50%), MR (reduction by ≥25%), SD (size change <25%), PD (increase by ≥25%). The effect of BEV on tumor itself was determined according to Response Assessment in Neuro‐Oncology criteria. Results Of the 139 patients, 21 (15.1%) had cystic components. The best responses of cysts to BEV treatment were as follows: CR 6, PR 7, MR 4, SD 4. The group of patients with progressively increasing cysts prior to BEV treatment had significant cyst size reduction compared to stable cyst size groups, at initial imaging after BEV (mean 62.6% vs 22.5%, P = .0055) and at best response timing (mean 76.3% vs 32.8%, P = .0050). Patients with cysts showed significant improvement in symptoms after the treatment with BEV compared to patients without cysts (P = .0033). However, response rate was not different between patients with or without cysts. Overall survival after starting BEV was not different between glioblastoma patients with or without cysts. Conclusion Bevacizumab is effective against progressively increasing cysts. Although cysts reduction effect and tumor response and/or overall survival are independent, BEV may be effective in patients who are symptomatic due to cyst enlargement.
Collapse
Affiliation(s)
- Fumiyuki Yamasaki
- Department of Neurosurgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Manish Kolakshyapati
- Department of Neurosurgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Motoki Takano
- Department of Neurosurgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Ushio Yonezawa
- Department of Neurosurgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Ikuno Nishibuchi
- Department of Radiation Oncology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Nobuki Imano
- Department of Radiation Oncology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Akira Taguchi
- Department of Neurosurgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Shumpei Onishi
- Department of Neurosurgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Vishwa Jeet Amatya
- Department of Pathology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Yukio Takeshima
- Department of Pathology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Yasushi Nagata
- Department of Radiation Oncology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Kaoru Kurisu
- Department of Neurosurgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Kazuhiko Sugiyama
- Department of Clinical Oncology & Neuro-oncology Program, Hiroshima University Hospital, Hiroshima, Japan
| |
Collapse
|
20
|
Raimondi A, Nichetti F, Peverelli G, Di Bartolomeo M, De Braud F, Pietrantonio F. Genomic markers of resistance to targeted treatments in gastric cancer: potential new treatment strategies. Pharmacogenomics 2018; 19:1047-1068. [PMID: 30041572 DOI: 10.2217/pgs-2018-0077] [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] [Indexed: 12/12/2022] Open
Abstract
Gastric cancer is a highly heterogeneous disease, displaying a complex genomic landscape and an unfavorable outcome with standard therapies. Based on distinctive genomic alterations, novel targeted agents have been developed with the aim of personalizing treatments and improving patient outcome. However, a subgroup of patients is primarily treatment-resistant, and even in the initially sensitive population, secondary resistance emerges, thus limiting therapeutic benefit. In this review, we summarize the clinical data about standard targeted agents in gastric cancer, specifically anti-HER2 treatments and antivascular therapies. We also illustrate the available evidence regarding molecular mechanisms of resistance to these agents and we discuss potential strategies for new targeted treatments that could overcome such resistance.
Collapse
Affiliation(s)
- Alessandra Raimondi
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Federico Nichetti
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Giorgia Peverelli
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Maria Di Bartolomeo
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Filippo De Braud
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy.,Department of Oncology & Hemato-oncology, University of Milan, Italy
| | - Filippo Pietrantonio
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy.,Department of Oncology & Hemato-oncology, University of Milan, Italy
| |
Collapse
|
21
|
Kitayama J, Ishigami H, Yamaguchi H, Sakuma Y, Horie H, Hosoya Y, Lefor AK, Sata N. Treatment of patients with peritoneal metastases from gastric cancer. Ann Gastroenterol Surg 2018; 2:116-123. [PMID: 29863151 PMCID: PMC5881364 DOI: 10.1002/ags3.12060] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 01/14/2018] [Indexed: 12/12/2022] Open
Abstract
Despite recent advances in chemotherapy, outcomes of patients with peritoneal metastases (PM) from gastric cancer are still very poor and standard treatment has not been established. Although oral S‐1 appears to be effective for patients with PM, the effects of systemic chemotherapy are limited. Cytoreductive surgery (CRS) and hyperthermic intraperitoneal chemotherapy (HIPEC) yield fewer benefits in patients with PM from gastric cancer than in patients with PM from other malignancies. In comparison, repeated intraperitoneal chemotherapy (RIPEC) with taxanes using an implantable peritoneal access port has a pharmacokinetic advantage for the control of peritoneal lesions and in combination with systemic chemotherapy can result in surprisingly long‐term survival in patients with PM from gastric cancer. Herein, we review the results of recent clinical studies specifically targeting PM from gastric cancer and discuss future prospects for an intraperitoneal approach to the ideal treatment of patients with gastric cancer with peritoneal involvement.
Collapse
Affiliation(s)
- Joji Kitayama
- Department of Gastrointestinal Surgery Jichi Medical University Shimotsuke Japan
| | | | - Hironori Yamaguchi
- Department of Gastrointestinal Surgery Jichi Medical University Shimotsuke Japan
| | - Yasunaru Sakuma
- Department of Gastrointestinal Surgery Jichi Medical University Shimotsuke Japan
| | - Hisanaga Horie
- Department of Gastrointestinal Surgery Jichi Medical University Shimotsuke Japan
| | - Yoshinori Hosoya
- Department of Gastrointestinal Surgery Jichi Medical University Shimotsuke Japan
| | - Alan Kawarai Lefor
- Department of Gastrointestinal Surgery Jichi Medical University Shimotsuke Japan
| | - Naohiro Sata
- Department of Gastrointestinal Surgery Jichi Medical University Shimotsuke Japan
| |
Collapse
|
22
|
Matsumoto H, Kawazoe A, Shimada K, Fukuoka S, Kuboki Y, Bando H, Kojima T, Ohtsu A, Yoshino T, Doi T, Shitara K. A retrospective study of the safety and efficacy of paclitaxel plus ramucirumab in patients with advanced or recurrent gastric cancer with ascites. BMC Cancer 2018; 18:120. [PMID: 29385993 PMCID: PMC5793350 DOI: 10.1186/s12885-018-4057-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2017] [Accepted: 01/25/2018] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Ramucirumab has recently proved to be effective for advanced or recurrent gastric cancer (AGC). Ascites and peritoneal metastasis are among the most common complications of AGC. However, there are few data on the safety and efficacy of paclitaxel plus ramucirumab in patients with AGC with ascites. The purpose of this retrospective study was to evaluate the safety and efficacy of paclitaxel plus ramucirumab in patients with AGC with ascites. METHODS We retrospectively evaluated the safety and efficacy of paclitaxel plus ramucirumab in patients with AGC with ascites in comparison with patients without ascites in a single institution from June 2015 to May 2016. The median progression-free survival (PFS) and overall survival (OS) were calculated using the Kaplan-Meier method, and differences evaluated using the Log-lank test. The differences in baseline characteristics and response rates of each ascites group were calculated for homogeneity by chi-square tests and for trends by Fisher's exact test. RESULTS Eighty-three patients were analyzed in this study. Ascites was detected in 40 patients, 26 patients (31%) had small to moderate ascites and 14 (17%) had massive ascites. The proportion of patients who started with a reduced dose of paclitaxel was higher for patients with massive ascites than others. The frequencies of any grade 3 or 4 hematological toxicity were 51% in patients without ascites, 77% in patients with small to moderate ascites, and 71% in patients with massive ascites. The frequencies of common ramucirumab-related adverse events were also not significantly different among ascites groups, however one patient had a tumor hemorrhage, and one patient had a gastrointestinal perforation. PFS and OS were shorter in patients with massive ascites than in patients with small or moderate ascites or patients without ascites. CONCLUSIONS The use of paclitaxel and ramucirumab in patients with AGC with large amounts of ascites was tolerable with adequate dose modification. However, we should pay attention to the risks of ramucirumab-related toxicity in patients with bleeding tumors or intestinal stenosis.
Collapse
Affiliation(s)
- Hiroshi Matsumoto
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, 6-5-1, Kashiwanoha, Kashiwa, Chiba 277-8577 Japan
| | - Akihito Kawazoe
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, 6-5-1, Kashiwanoha, Kashiwa, Chiba 277-8577 Japan
| | - Kaoru Shimada
- Department of Diagnostic Radiology, National Cancer Center Hospital East, 6-5-1, Kashiwanoha, Kashiwa, Chiba 277-8577 Japan
| | - Shota Fukuoka
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, 6-5-1, Kashiwanoha, Kashiwa, Chiba 277-8577 Japan
| | - Yasutoshi Kuboki
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, 6-5-1, Kashiwanoha, Kashiwa, Chiba 277-8577 Japan
| | - Hideaki Bando
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, 6-5-1, Kashiwanoha, Kashiwa, Chiba 277-8577 Japan
| | - Takashi Kojima
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, 6-5-1, Kashiwanoha, Kashiwa, Chiba 277-8577 Japan
| | - Atsushi Ohtsu
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, 6-5-1, Kashiwanoha, Kashiwa, Chiba 277-8577 Japan
| | - Takayuki Yoshino
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, 6-5-1, Kashiwanoha, Kashiwa, Chiba 277-8577 Japan
| | - Toshihiko Doi
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, 6-5-1, Kashiwanoha, Kashiwa, Chiba 277-8577 Japan
| | - Kohei Shitara
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, 6-5-1, Kashiwanoha, Kashiwa, Chiba 277-8577 Japan
| |
Collapse
|
23
|
Chen S, Chen X, Nie R, Ou Yang L, Liu A, Li Y, Zhou Z, Chen Y, Peng J. A nomogram to predict prognosis for gastric cancer with peritoneal dissemination. Chin J Cancer Res 2018; 30:449-459. [PMID: 30210225 PMCID: PMC6129562 DOI: 10.21147/j.issn.1000-9604.2018.04.08] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Objective To identify independent prognostic factors to be included in a nomogram to predict the prognosis of gastric cancer patients with peritoneal dissemination. Methods This is a retrospective study on 684 patients with a histological diagnosis of gastric cancer with peritoneal dissemination from the Sun Yat-sen University Cancer Center as the development set, and 62 gastric cancer patients from the Sixth Affiliated Hospital of Sun Yat-sen University as the validation group. Chi-square test and Cox regression analysis were used to compare the clinicopathological variables and the prognosis of gastric cancer patients with peritoneal dissemination. The Harrell’s concordance index (C-index) and calibration curve were determined for comparisons of predictive ability of the nomogram. Results Univariate and multivariate analyses showed that serum carcinoembryonic antigen (CEA) level (P=0.032), ascites grading (P=0.008), presence of extraperitoneal metastasis (P<0.001), seeding status (P=0.016) and performance status (P=0.009) were independent prognostic factors for gastric cancer patients with peritoneal dissemination in the development set. The nomogram model was constructed using these five factors. Internal validation showed that the C-index of the model was 0.641. For the external validation, the C-index of this model was 0.709. Conclusions We developed and validated a nomogram to predict the prognosis for gastric cancer patients with peritoneal dissemination. This nomogram may play an important clinical role in guiding palliative therapy for these types of patients, although it may need more data for optimization.
Collapse
Affiliation(s)
- Shi Chen
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, the Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou 510655, China.,Department of Gastrointestinal Surgery, the Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou 510655, China
| | - Xijie Chen
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, the Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou 510655, China.,Department of Gastrointestinal Surgery, the Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou 510655, China
| | - Runcong Nie
- Department of Gastropancreatic Surgery, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Liying Ou Yang
- Department of Intensive Care, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Aihong Liu
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, the Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou 510655, China.,Department of Gastrointestinal Surgery, the Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou 510655, China
| | - Yuanfang Li
- Department of Gastropancreatic Surgery, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Zhiwei Zhou
- Department of Gastropancreatic Surgery, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Yingbo Chen
- Department of Gastropancreatic Surgery, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Junsheng Peng
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, the Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou 510655, China.,Department of Gastrointestinal Surgery, the Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou 510655, China
| |
Collapse
|
24
|
Predictive Significance of Serum Level of Vascular Endothelial Growth Factor in Gastric Cancer Patients. BIOMED RESEARCH INTERNATIONAL 2016; 2016:8103019. [PMID: 27597973 PMCID: PMC4997027 DOI: 10.1155/2016/8103019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Revised: 07/12/2016] [Accepted: 07/12/2016] [Indexed: 12/28/2022]
Abstract
The study aims to evaluate serum VEGF expression in gastric cancer patients and investigate its relationship with clinicopathological parameters. We also examined the serum VEGF levels in GC patients having received surgery or chemotherapy treatment to assess its predictive and prognostic value as a biomarker. We enrolled 154 GC patients having not received neoadjuvant treatment and 100 healthy controls. In the treatment groups, 13 surgery patients and 15 chemotherapy patients were investigated. 42 chemotherapy patients with different chemotherapy efficacy were recruited as well. The serum VEGF was examined by ELISA. Serum VEGF level was remarkably upregulated in GC group compared with healthy group (p < 0.001). The serum VEGF level of GC group was significantly correlated with tumor cells differentiation degree, clinical stages, tumor infiltration depth, lymph node metastasis, and tumor size. The serum VEGF level of the 1 to 3 days after operation group was much lower than that of the preoperative group (p < 0.001) and the 7 days after operation group (p < 0.001). By contrast, serum VEGF level was decreased significantly after chemotherapy (p = 0.001). Importantly, serum VEGF level in PD+SD group was significantly higher compared to the PR+CR group (p = 0.011). Therefore, serum VEGF was a valuable biomarker in clinically monitoring the condition of GC patients.
Collapse
|
25
|
Jordan K, Luetkens T, Gog C, Killing B, Arnold D, Hinke A, Stahl M, Freier W, Rüssel J, Atanackovic D, Hegewisch-Becker S. Intraperitoneal bevacizumab for control of malignant ascites due to advanced-stage gastrointestinal cancers: A multicentre double-blind, placebo-controlled phase II study – AIO SUP-0108. Eur J Cancer 2016; 63:127-34. [DOI: 10.1016/j.ejca.2016.05.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Revised: 04/22/2016] [Accepted: 05/04/2016] [Indexed: 10/21/2022]
|
26
|
Peptidomimetic suppresses proliferation and invasion of gastric cancer cells by fibroblast growth factor 2 signaling cascade blockage. Anticancer Drugs 2016; 27:164-72. [PMID: 26556626 DOI: 10.1097/cad.0000000000000312] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Fibroblast growth factor 2 (FGF2) is closely involved in a variety of tumors, including gastric cancer (GC). FGF2 inhibitors exert good antitumor activity, but no FGF2 inhibitor has been employed for clinical use. To obtain a low-toxicity, stable peptidomimetic (called P29) target to FGF2, the affinity between P29 and FGF2 was detected by surface plasmon resonance. The stability of P29 was measured by high performance liquid chromatography. MTT assay and transwell assay were used to access the proliferative and invasive ability of GC cells, respectively. Western blot assay and flow cytometric analysis were applied to study the mechanism of P29. P29 possessed high affinity with FGF2 and a longer half-life in vitro. P29 suppressed the FGF2-induced proliferation of GC cells. It also inhibited the phosphorylation of FRS2, ERK1/2, and AKT triggered by FGF2 in GC. In addition, P29 blocked GC cell transformation from the G1/G0 phase to the S phase and weakened the invasive capability of GC cells. In this paper, we present a novel FGF2 inhibitor that could exert improved anticancer effect in GC in vitro.
Collapse
|
27
|
Wu Y, Pan M, Cui S, Ba M, Chen Z, Ruan Q. Efficacy and safety of ultrasound-guided continuous hyperthermic intraperitoneal perfusion chemotherapy for the treatment of malignant ascites: a midterm study of 36 patients. Onco Targets Ther 2016; 9:403-7. [PMID: 26855589 PMCID: PMC4725693 DOI: 10.2147/ott.s85564] [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] [Indexed: 12/04/2022] Open
Abstract
Background This study aimed to evaluate the efficacy and safety of ultrasound-guided continuous hyperthermic intraperitoneal perfusion chemotherapy (CHIPC) for the treatment of malignant ascites (MA). Methods Between July 2011 and June 2013, 36 MA patients were prospectively and consecutively hospitalized for three cycles of elective CHIPC under ultrasound guidance, maintained at a constant flow rate of 400–600 mL/min normal saline containing 5-fluorouracil plus mitomycin or carboplatin and at a constant temperature of 43°C±0.2°C, for 90 minutes. Main outcome measures were ascites resolution, Karnofsky performance status (KPS), and serum tumor biomarkers at 2 weeks after the last cycle of CHIPC. All the patients underwent uneventful CHIPC as scheduled, and vital signs remained stable over CHIPC. Results At 2 weeks after the last cycle of CHIPC, MA completely and partially resolved in 26 (72.2%) patients and eight (22.2%) patients, respectively; mean KPS score increased from pretreatment 61±9 to posttreatment 76±9 (P<0.001), and serum carcinoembryonic antigen and carbohydrate antigens 12-5 and 19-9 significantly decreased (all P<0.01). Conclusion The current study indicated that ultrasound-guided CHIPC is an effective and safe palliative treatment modality for MA with respect to MA resolution, patient’s general well-being, and systemic disease control. The long-term benefit of CHIPC on overall survival remains to be investigated in MA patients.
Collapse
Affiliation(s)
- Yinbing Wu
- Second Department of Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou Medical University, Guangzhou, People's Republic of China; Treatment Center of Body Cavitary Thermo-Perfusion, Cancer Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Mingxin Pan
- Second Department of Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Shuzhong Cui
- Treatment Center of Body Cavitary Thermo-Perfusion, Cancer Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Mingchen Ba
- Treatment Center of Body Cavitary Thermo-Perfusion, Cancer Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Zulong Chen
- Treatment Center of Body Cavitary Thermo-Perfusion, Cancer Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Qiang Ruan
- Treatment Center of Body Cavitary Thermo-Perfusion, Cancer Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, People's Republic of China
| |
Collapse
|
28
|
Tumor-associated macrophages of the M2 phenotype contribute to progression in gastric cancer with peritoneal dissemination. Gastric Cancer 2016; 19:1052-1065. [PMID: 26621525 PMCID: PMC5034006 DOI: 10.1007/s10120-015-0579-8] [Citation(s) in RCA: 204] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Accepted: 11/13/2015] [Indexed: 02/07/2023]
Abstract
BACKGROUND Tumor-associated macrophages (TAMs) of the M2 phenotype are known to promote tumor proliferation and to be associated with a poor prognosis in numerous cancers. Here, we investigated whether M2 macrophages participate in the development of peritoneal dissemination in gastric cancer. METHODS The characteristics of peritoneal macrophages in gastric cancer patients with or without peritoneal dissemination were examined by flow cytometry and the real-time quantitative polymerase chain reaction. The effects of M2 macrophages on phenotypic changes of the gastric cancer cell line MKN45 were assessed with a direct or indirect co-culture system in vitro and an in vivo mouse xenograft model. RESULTS The number of peritoneal macrophages with the M2 phenotype (CD68(+)CD163(+) or CD68(+)CD204(+)) was significantly higher in gastric cancer patients with peritoneal dissemination than in those without peritoneal dissemination. Higher expression of the M2-related messenger RNAs (IL-10, vascular endothelial growth factor A, vascular endothelial growth factor C, matrix metalloproteinase 1, and amphiregulin) and lower expression of M1-related messenger RNAs (TNF-α, CD80, CD86, and IL-12p40) were also confirmed in the TAMs. Macrophage co-culture with gastric cancer cells converted M1 phenotype into M2 phenotype. Moreover, the coexistence of MKN45 cells with M2 macrophages resulted in cancer cell proliferation and an acceleration of tumor growth in the xenograft model. CONCLUSIONS Intraperitoneal TAMs in gastric cancer patients with peritoneal dissemination were polarized to the M2 phenotype, and could contribute to tumor proliferation and progression. Therefore, intraperitoneal TAMs are expected to be a promising target in the treatment of peritoneal dissemination in gastric cancer.
Collapse
|
29
|
Liu R, Zheng H, Li W, Guo Q, He S, Hirasaki Y, Hou W, Hua B, Li C, Bao Y, Gao Y, Qi X, Pei Y, Zhang Y. Anti-tumor enhancement of Fei-Liu-Ping ointment in combination with celecoxib via cyclooxygenase-2-mediated lung metastatic inflammatory microenvironment in Lewis lung carcinoma xenograft mouse model. J Transl Med 2015; 13:366. [PMID: 26597177 PMCID: PMC4656184 DOI: 10.1186/s12967-015-0728-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Accepted: 11/10/2015] [Indexed: 12/19/2022] Open
Abstract
Background Fei-Liu-Ping (FLP) ointment is an oral prescription medication that has been widely applied to treat lung cancer patients in China. Regulation of the metastatic microenvironment is an important therapeutic approach for prevention and treatment of tumor recurrence and metastasis. The advantage of Traditional Chinese Medicine management of lung cancer lies in the prevention of recurrence and metastasis. Our previous study has demonstrated that FLP ointment could regulate lung inflammatory microenvironment in vitro. However, the effects of FLP on the tumor microenvironment in vivo are still poorly understood. The objective of this study is to investigate the effect of FLP alone or in combination with celecoxib in the prevention of lung cancer progression by Cyclooxygenase (Cox)-2 mediated tumor inflammatory microenvironment in vivo. Methods 120 Lewis lung carcinoma xenograft mice were divided equally into four groups: vehicle, FLP, celecoxib, and FLP plus celecoxib. The dynamic growth of the xenografted tumors was observed using an in vivo fluorescence imaging system. Mice were sacrificed on day 14, day 21, and day 28, and tumor specimens and lung tissues were harvested to detect the metastasis-associated protein expression. Results Tumor inhibition rate was 15.4, 44.2, 47.4 % at day 14, 37.3, 34.7, 61.5 % at day 21, and 15.5, 10.3, 32.5 % at day 28 after treatment of FLP, celecoxib, and FLP plus celecoxib, respectively. Upon treatment of FLP and celecoxib together, lung metastasis rate was 30 % (8 metastatic nodules) lower than other groups. FLP inhibited Cox-2 expression in a time-dependent manner. Moreover, FLP inhibited N-cadherin, matrix metalloproteinases (MMP)-9, and Vimentin expression. Treatment of FLP in combination with celecoxib was more effective than FLP or celecoxib alone in inhibiting vascular endothelial growth factor, platelet-derived growth factor receptors β, microsomal Prostaglandin E synthase-1, MMP-2, MMP-9, N-cadherin, and Vimentin expression, but increased E-cadherin expression. Conclusions FLP inhibited tumor growth and metastasis in a Lewis lung xenograft mice model through the Cox-2 pathway. FLP in combination with celecoxib enhanced the antitumor growth and anti-metastasis effects. Traditional Chinese herbs combined with anti-inflammatory drugs might offer a promising strategy to prevent tumor metastasis.
Collapse
Affiliation(s)
- Rui Liu
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Number 5 Beixiange, Xicheng District, Beijing, 100053, China. .,Cancer Institute, China Academy of Chinese Medical Sciences, Beijing, 100053, China.
| | - Honggang Zheng
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Number 5 Beixiange, Xicheng District, Beijing, 100053, China. .,Cancer Institute, China Academy of Chinese Medical Sciences, Beijing, 100053, China.
| | - Weidong Li
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Number 5 Beixiange, Xicheng District, Beijing, 100053, China. .,Cancer Institute, China Academy of Chinese Medical Sciences, Beijing, 100053, China.
| | - Qiujun Guo
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Number 5 Beixiange, Xicheng District, Beijing, 100053, China. .,Cancer Institute, China Academy of Chinese Medical Sciences, Beijing, 100053, China. .,Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Shulin He
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Number 5 Beixiange, Xicheng District, Beijing, 100053, China. .,Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Yoshiro Hirasaki
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Number 5 Beixiange, Xicheng District, Beijing, 100053, China. .,Department of Japanese-Oriental (Kampo) Medicine, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8670, Japan.
| | - Wei Hou
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Number 5 Beixiange, Xicheng District, Beijing, 100053, China. .,Cancer Institute, China Academy of Chinese Medical Sciences, Beijing, 100053, China.
| | - Baojin Hua
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Number 5 Beixiange, Xicheng District, Beijing, 100053, China. .,Cancer Institute, China Academy of Chinese Medical Sciences, Beijing, 100053, China.
| | - Conghuang Li
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Number 5 Beixiange, Xicheng District, Beijing, 100053, China. .,Cancer Institute, China Academy of Chinese Medical Sciences, Beijing, 100053, China.
| | - Yanju Bao
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Number 5 Beixiange, Xicheng District, Beijing, 100053, China. .,Cancer Institute, China Academy of Chinese Medical Sciences, Beijing, 100053, China.
| | - Yebo Gao
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Number 5 Beixiange, Xicheng District, Beijing, 100053, China. .,Cancer Institute, China Academy of Chinese Medical Sciences, Beijing, 100053, China. .,Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Xin Qi
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Number 5 Beixiange, Xicheng District, Beijing, 100053, China. .,Cancer Institute, China Academy of Chinese Medical Sciences, Beijing, 100053, China.
| | - Yingxia Pei
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Number 5 Beixiange, Xicheng District, Beijing, 100053, China. .,Cancer Institute, China Academy of Chinese Medical Sciences, Beijing, 100053, China.
| | - Yun Zhang
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Number 5 Beixiange, Xicheng District, Beijing, 100053, China. .,Cancer Institute, China Academy of Chinese Medical Sciences, Beijing, 100053, China.
| |
Collapse
|
30
|
Li ZR, Li DJ, Jie ZG. Diagnosis and treatment of peritoneal metastasis of gastric cancer. Shijie Huaren Xiaohua Zazhi 2015; 23:3653-3662. [DOI: 10.11569/wcjd.v23.i23.3653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
As the main reason of death in patients with gastric cancer, peritoneal metastasis is still a major problem to be solved. As we all know, peritoneal metastasis is the main form of advanced gastric cancer and gastric cancer recurrence, which involves a complex, multi-stage, multifactorial pathological process. The diagnosis and treatment of peritoneal metastasis of gastric cancer are particularly difficult. The good news is that through active exploration and clinical research, several important achievements have been made and gradually bring the gospel to clinical patients. This paper will review the recent progress in the diagnosis and treatment of peritoneal metastasis of gastric cancer.
Collapse
|
31
|
Kim JS, Gang GW, Lee SR, Sung HJ, Park Y, Kim DS, Choi CW, Kim BS. Bone marrow vascular endothelial growth factor level per platelet count might be a significant predictor for the treatment outcomes of patients with diffuse large B-cell lymphomas. Jpn J Clin Oncol 2015; 45:914-20. [PMID: 26185139 DOI: 10.1093/jjco/hyv102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 06/13/2015] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVE Developing a parameter to predict bone marrow invasion by non-Hodgkin's lymphoma is an important unmet medical need for treatment decisions. This study aimed to confirm the validity of the hypothesis that bone marrow plasma vascular endothelial growth factor level might be correlated with the risk of bone marrow involvement and the prognosis of patients with diffuse large B-cell non-Hodgkin's lymphoma. METHODS Forty-nine diffuse large B-cell lymphoma patients treated with rituximab, cyclophosphamide, daunorubicin, vincristine and prednisolone regimen were enrolled. Vascular endothelial growth factor level was measured with enzyme-linked immunosorbent assay. The validity of bone marrow plasma vascular endothelial growth factor level and bone marrow vascular endothelial growth factor level per platelet count for predicting treatment response and survival after initial rituximab, cyclophosphamide, daunorubicin, vincristine and prednisolone combined chemotherapy was assessed. RESULTS Bone marrow plasma vascular endothelial growth factor level per platelet count was significantly associated with old age (≥ 65 years), poor performance score (≥ 2), high International prognosis index (≥ 3) and bone marrow invasion. The patients with high bone marrow plasma vascular endothelial growth factor level per platelet count (≥ 3.01) showed a significantly lower complete response rate than the others. On Kaplan-Meier survival curves, the patients with high bone marrow plasma vascular endothelial growth factor levels (≥ 655 pg/ml) or high bone marrow plasma vascular endothelial growth factor level per platelet count (≥ 3.01) demonstrated a significantly shorter overall survival and progression-free survival than the others. In the patients without bone marrow involvement, bone marrow plasma vascular endothelial growth factor level per platelet count had a significant relationship with overall survival and progression-free survival. Multivariate analysis revealed that the patients without BM invasion showing high level of bone marrow plasma vascular endothelial growth factor per platelet count had significantly shorter progression-free survival and overall survival. CONCLUSIONS Bone marrow plasma vascular endothelial growth factor level per platelet count might be associated with bone marrow invasion by diffuse large B-cell lymphoma and is correlated with clinical outcomes after treatment.
Collapse
Affiliation(s)
- Jung Sun Kim
- Division of Oncology and Hematology, Department of Internal Medicine, Korea University Medical Center, Seoul, Korea
| | - Ga Won Gang
- Division of Oncology and Hematology, Department of Internal Medicine, Korea University Medical Center, Seoul, Korea
| | - Se Ryun Lee
- Division of Oncology and Hematology, Department of Internal Medicine, Korea University Medical Center, Seoul, Korea
| | - Hwa Jung Sung
- Division of Oncology and Hematology, Department of Internal Medicine, Korea University Medical Center, Seoul, Korea
| | - Young Park
- Division of Oncology and Hematology, Department of Internal Medicine, Korea University Medical Center, Seoul, Korea
| | - Dae Sik Kim
- Division of Oncology and Hematology, Department of Internal Medicine, Korea University Medical Center, Seoul, Korea
| | - Chul Won Choi
- Division of Oncology and Hematology, Department of Internal Medicine, Korea University Medical Center, Seoul, Korea
| | - Byung Soo Kim
- Division of Oncology and Hematology, Department of Internal Medicine, Korea University Medical Center, Seoul, Korea
| |
Collapse
|
32
|
Ni ZT, Liu WT, Yang QM, Yan M, Zhu ZG. State-of-the-art methods in clinical diagnosis and treatment of peritoneal metastasis of gastric cancer. Shijie Huaren Xiaohua Zazhi 2015; 23:2843-2853. [DOI: 10.11569/wcjd.v23.i18.2843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Gastric cancer is one of the most common malignant tumors and presents a very high mortality. The main reason for this situation is metastasis after curative resection, with the most common type being peritoneal metastasis, which accounts for more than 50% of all cases. Once peritoneal carcinomatosis (PC) happens, the pathological stage is stage IV and there is a grim prognosis. Accordingly, early effective prevention and treatment of PC have extremely important clinical significance for the improvement of the prognosis of patients with gastric cancer. This article describes the pathogenesis, clinical diagnosis using serum biomarkers and image examinations, as well as multimodality treatment of peritoneal metastasis of gastric cancer by neoadjuvant intraperitoneal-systemic chemotherapy (NIPS), cytoreductive surgery+hyperthermic intraperitoneal chemotherapy (CRS+HIPEC), early postoperative intraperitoneal chemotherapy (EPIC), extensive intraoperative peritoneal lavage (EIPL), molecular targeting therapy, and usage of drug delivery systems.
Collapse
|
33
|
Li J, Kong D, He Y, Wang X, Gao L, Li J, Yan M, Liu D, Wang Y, Zhang L, Jin X. The impact of inflammatory cells in malignant ascites on small intestinal ICCs' morphology and function. J Cell Mol Med 2015; 19:2118-27. [PMID: 26087333 PMCID: PMC4568916 DOI: 10.1111/jcmm.12575] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Accepted: 02/05/2015] [Indexed: 12/14/2022] Open
Abstract
Malignant ascites is one of the common complication at the late stage of abdominal cancers, which may deteriorate the environment of abdominal cavity and lead to potential damage of functional cells. Interstitial cells of Cajal (ICCs) are mesoderm-derived mesenchymal cells that function normal gastrointestinal motility. The pathological changes of ICCs or the reduced number may lead to the motility disorders of gastrointestinal tract. In this study, through analysis of malignant ascites which were obtained from cancer patients, we found that inflammatory cells, including tumour-infiltrating lymphocytes, accounted for 17.26 ± 1.31% and tumour-associated macrophages, occupied 19.06 ± 2.27% of total cells in the ascites, suggesting these inflammatory cells, in addition to tumour cells, may exert important influence on the tumour environment of abdominal cavity. We further demonstrated that the number of mice ICCs were significant decreased, as well as morphological and functional damage when ICCs were in the simulated tumour microenvironment in vitro. Additionally, we illustrated intestinal myoelectrical activity reduced and irregular with morphological changes of ICCs using the mice model of malignant ascites. In conclusion, our data suggested that inflammatory cells in malignant ascites may damage ICCs of the small intestine and lead to intestinal motility disorders.
Collapse
Affiliation(s)
- Jing Li
- Department of Pathology, Basic Medical Science College, Harbin Medical University, Harbin, China
| | - Dan Kong
- Department of Gynecology, Third Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yan He
- Department of Pathology, Basic Medical Science College, Harbin Medical University, Harbin, China
| | - Xiuli Wang
- Department of Pathology, Basic Medical Science College, Harbin Medical University, Daqing, China
| | - Lei Gao
- Department of Pathology, Basic Medical Science College, Harbin Medical University, Harbin, China
| | - Jiade Li
- Department of Pathology, Basic Medical Science College, Harbin Medical University, Harbin, China
| | - Meisi Yan
- Department of Pathology, Basic Medical Science College, Harbin Medical University, Harbin, China
| | - Duanyang Liu
- Department of Pathology, Basic Medical Science College, Harbin Medical University, Harbin, China
| | - Yufu Wang
- Department of Orthopedics, Second Clinical Hospital, Harbin Medical University, Harbin, China
| | - Lei Zhang
- Department of Pathology, Basic Medical Science College, Harbin Medical University, Harbin, China
| | - Xiaoming Jin
- Department of Pathology, Basic Medical Science College, Harbin Medical University, Harbin, China
| |
Collapse
|
34
|
Yuan P, Yue TH, Xiao YH, Zhu LJ, Li S, Chen BA. Clinical effects of hyperthermic intraperitoneal chemotherapy for gastric cancer with malignant ascites. Shijie Huaren Xiaohua Zazhi 2014; 22:4825-4829. [DOI: 10.11569/wcjd.v22.i31.4825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To assess the clinical effects of hyperthermic intraperitoneal chemotherapy (HIPEC) in the treatment of gastric cancer with malignant ascites.
METHODS: Seventy patients with gastric cancer with malignant ascites were randomly divided into either a study group or a control group. The study group was treated by HIPEC combined with thermal therapy, and the control group was treated by non-hyperthermic peritoneal perfusion chemotherapy. The clinical effects, Kamofsky score and adverse reactions were compared for the two groups. The temperature and vital signs at different points of HIPEC were recorded.
RESULTS: The total effective rate was significantly higher in the experiment group than in the control group (77.14% vs 37.14%, P < 0.05). Posttreatment Kamofsky scores for the two groups were significantly higher than prior-treatment values (77.92 ± 6.83 vs 54.44 ± 5.47, 62.08 ± 6.17 vs 53.89 ± 5.56, P < 0.05). Posttreatment Kamofsky score was significantly higher in the experiment group than in the control group (77.92 ± 6.83 vs 62.08 ± 6.17, P < 0.05). There were no significant difference in the shell temperature, tympanic temperature, rectal temperature, blood pressure, heart rate, breath, or oxyhemoglobin saturation for the experiment group at different time points (36.18 ℃ ± 0.42 ℃ vs 36.42 ℃ ± 0.27 ℃ vs 37.13 ℃ ± 1.72 ℃, 35.66 ℃ ± 0.23 ℃ vs 35.94 ℃ ± 0.37 ℃ vs 36.60 ℃ ± 0.22 ℃, 36.34 ℃ ± 0.12 ℃ vs 36.64 ℃ ± 0.27 ℃ vs 37.10 ℃ ± 0.30 ℃, 117 mmHg ± 6.2 mmHg vs 116 mmHg ± 6.5 mmHg vs 116 mmHg ± 6.4 mmHg, 62 mmHg ± 4.9 mmHg vs 69 mmHg ± 6.8 mmHg vs 72 mmHg ± 5.3 mmHg, 68/min ± 4.3/min vs 72/min ± 5.3/min vs 73/min ± 4.5/min, 14/min ± 2.5/min vs 13/min ± 1.8/min vs 14/min ± 1.7/min, 98% ± 1.8% vs 97% ± 0.9% vs 98% ± 1.3%, P > 0.05). The rate of fatty scleroma for the experiment group was significantly higher than that for the control group (14.29% vs 0.00%, P < 0.05).
CONCLUSION: HIPEC can improve Kamofsky score and has high safety in patients with gastric cancer with malignant ascites.
Collapse
|
35
|
Kikuchi H, Kamiya K, Hiramatsu Y, Miyazaki S, Yamamoto M, Ohta M, Baba S, Konno H. Laparoscopic narrow-band imaging for the diagnosis of peritoneal metastasis in gastric cancer. Ann Surg Oncol 2014; 21:3954-62. [PMID: 24859934 DOI: 10.1245/s10434-014-3781-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2013] [Indexed: 12/20/2022]
Abstract
BACKGROUND Staging laparoscopy (SL) is often used to diagnose peritoneal metastasis in patients with advanced gastric cancer, but accurate detection of metastasis can be difficult. We evaluated the usefulness of laparoscopic narrow-band imaging (NBI) versus conventional laparoscopic white-light imaging (WLI) for the diagnosis of peritoneal metastasis. METHODS We excised 37 white nodules from the parietal peritoneum of 26 patients with gastric cancer and suspected peritoneal metastasis. The WLI and NBI findings were compared with the pathological findings. All the peritoneal lesions examined were observed as white nodules on WLI. Intranodular vessels were evaluated by WLI and NBI for (1) vessel dilatation, (2) vessel tortuousness, (3) vessel heterogeneity, and (4) brown spots. RESULTS Each individual abnormal finding had a diagnostic accuracy of less than 79 % with or without NBI. Detection of any one abnormal finding had a sensitivity, specificity, and accuracy of 47.8, 85.7, and 62.2 %, respectively, on WLI and 91.3, 71.4, and 83.8 %, respectively, on NBI, for detection of peritoneal metastasis. Detection of any one abnormal finding on NBI plus clear demarcation of the nodule on WLI had a sensitivity of 91.3 %, specificity of 92.9 %, and accuracy of 91.9 % for detection of peritoneal metastasis. Pathological examination showed that a brown spot detected on NBI correlated with dilated vessels around cancer cells. Vascular endothelial growth factor was expressed in 76.2 % of peritoneal metastases. CONCLUSIONS NBI was more sensitive for the detection of dilated vessels than WLI. NBI could be a useful tool for the diagnosis of peritoneal metastasis during SL.
Collapse
Affiliation(s)
- Hirotoshi Kikuchi
- Second Department of Surgery, Hamamatsu University School of Medicine, Hamamatsu, Japan,
| | | | | | | | | | | | | | | |
Collapse
|