1
|
Karalis JD, Ju MR, Feig R, Estrella R, Pettigrew MF, Alterio RE, Abreu AA, Farah E, Sawas T, Sanford NN, Sanjeevaiah A, Hammer STG, Porembka MR, Wang SC. Intensifying supportive care is associated with improved survival in gastric cancer patients with malignant ascites. J Surg Oncol 2024; 129:718-727. [PMID: 38063245 DOI: 10.1002/jso.27556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 11/07/2023] [Accepted: 11/25/2023] [Indexed: 02/17/2024]
Abstract
BACKGROUND Gastric cancer patients with malignant ascites often have poor functional status and malnutrition that preclude receipt of systemic therapies. Thus, these patients have a very poor prognosis. Beginning in 2019, our multidisciplinary gastric cancer disease-oriented team implemented a more aggressive supportive care plan for gastric cancer patients with malignant ascites. The initiative included measures such as supplemental enteral nutrition, ascites drainage, and initiation of chemotherapy on an inpatient basis. We compared outcomes for gastric cancer patients who presented with synchronous malignant ascites treated before and after the implementation of the care plan. METHODS We performed a retrospective review of our institutional database to identify patients diagnosed with gastric adenocarcinoma and synchronous malignant ascites between 2010 and 2022. We compared overall survival (OS) between patients diagnosed from 2010 to 2018, which will be referred to as the historical control era and patients diagnosed from 2019 to 2022, which will be called the aggressive supportive care era. RESULTS Fifty-four patients were included in our analysis; 31 patients were treated in the historical control time frame, and 23 patients were treated during the aggressive supportive care era. Demographic, clinical, and pathologic characteristics were similar between groups. 3% of historical controls received supplemental tube feeds at diagnosis as compared to 30% of the aggressive supportive care cohort (p < 0.01). 3% of historical controls received their first cycle of chemotherapy in the inpatient setting versus 39% of patients treated during the aggressive supportive care era (p < 0.01). The median number of chemotherapy cycles received was 5 among historical controls and 9.5 among aggressive supportive care era patients (p = 0.02). There was no difference in the number of days spent as an inpatient between the two groups. The median OS for historical control patients was 5.4 months as compared with 10.4 months for patients treated during aggressive supportive care era (p = 0.04). CONCLUSIONS Gastric cancer patients with synchronous malignant ascites treated during a timeframe when our multidisciplinary team implemented more aggressive supportive care measures had improved OS as compared with historic controls. Our results suggest that aggressive supportive measures for these patients with highly challenging clinical issues and poor prognosis can prolong survival. Specifically, initiation of chemotherapy in the inpatient setting and supplemental nutrition should be considered for patients at high risk for treatment intolerance.
Collapse
Affiliation(s)
- John D Karalis
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Michelle R Ju
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Rachel Feig
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | | | - Morgan F Pettigrew
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Rodrigo E Alterio
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Andres A Abreu
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Emile Farah
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Tarek Sawas
- Division of Digestive and Liver Diseases, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Nina N Sanford
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Aravind Sanjeevaiah
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Suntrea T G Hammer
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Matthew R Porembka
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Sam C Wang
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
2
|
Ueda A, Yuki S, Ando T, Hosokawa A, Nakada N, Kito Y, Motoo I, Ito K, Sakumura M, Nakayama Y, Ueda Y, Kajiura S, Nakashima K, Harada K, Kawamoto Y, Komatsu Y, Yasuda I. CA125 Kinetics as a Potential Biomarker for Peritoneal Metastasis Progression following Taxane-Plus-Ramucirumab Administration in Patients with Advanced Gastric Cancer. Cancers (Basel) 2024; 16:871. [PMID: 38473233 DOI: 10.3390/cancers16050871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 02/12/2024] [Accepted: 02/20/2024] [Indexed: 03/14/2024] Open
Abstract
Currently, no established marker exists for predicting peritoneal metastasis progression during chemotherapy, although they are major interruptive factors in sequential chemotherapy in patients with advanced gastric cancer (AGC). This multicenter retrospective study was conducted from June 2015 to July 2019, analyzing 73 patients with AGC who underwent taxane-plus-ramucirumab (TAX/RAM) therapy and had their serum carbohydrate antigen 125 (CA125) concentrations measured. Of 31 patients with elevated CA125 levels above a cutoff of 35 U/mL, 25 (80.6%) had peritoneal metastasis. The CA125 concentrations before TAX/RAM treatment were associated with ascites burden. The overall survival was significantly shorter in the CA125-elevated group. CA125 kinetics, measured at a median of 28 days after chemotherapy, were associated with the ascites response (complete or partial response: -1.86%/day; stable disease: 0.28%/day; progressive disease: 2.33%/day). Progression-free survival in the CA125-increased group, defined by an increase of 0.0067%/day using receiver operating characteristic curve analysis, was significantly poorer among patients with peritoneal metastases. In conclusion, this study highlights that CA125 kinetics can serve as an early predictor for the progression of peritoneal metastasis during TAX/RAM treatment.
Collapse
Affiliation(s)
- Akira Ueda
- Third Department of Internal Medicine, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
| | - Satoshi Yuki
- Department of Gastroenterology and Hepatology, Hokkaido University Hospital, Kita14, Nishi 5, Kita-ku, Sapporo 060-8648, Japan
| | - Takayuki Ando
- Third Department of Internal Medicine, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
| | - Ayumu Hosokawa
- Department of Clinical Oncology, University of Miyazaki Hospital, 5200 Kihara, Kiyotake-cho, Miyazaki 889-1692, Japan
| | - Naokatsu Nakada
- Department of Internal Medicine, Itoigawa Sogo Hospital, 457-1 Takegahana, Itoigawa 941-8502, Japan
| | - Yosuke Kito
- Department of Medical Oncology, Ishikawa Prefectural Central Hospital, 2-1 Kuratuki Higashi, Kanazawa 920-8530, Japan
| | - Iori Motoo
- Third Department of Internal Medicine, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
| | - Ken Ito
- Department of Gastroenterology, Tomakomai City Hospital, 1-5-20 Shimizucho, Tomakomai 053-8567, Japan
| | - Miho Sakumura
- Third Department of Internal Medicine, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
| | - Yurika Nakayama
- Third Department of Internal Medicine, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
| | - Yuko Ueda
- Third Department of Internal Medicine, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
| | - Shinya Kajiura
- Third Department of Internal Medicine, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
| | - Koji Nakashima
- Department of Clinical Oncology, University of Miyazaki Hospital, 5200 Kihara, Kiyotake-cho, Miyazaki 889-1692, Japan
| | - Kazuaki Harada
- Department of Gastroenterology and Hepatology, Hokkaido University Hospital, Kita14, Nishi 5, Kita-ku, Sapporo 060-8648, Japan
| | - Yasuyuki Kawamoto
- Division of Cancer Center, Hokkaido University Hospital, Kita 14, Nishi 5, Kita-ku, Sapporo 060-8648, Japan
| | - Yoshito Komatsu
- Division of Cancer Center, Hokkaido University Hospital, Kita 14, Nishi 5, Kita-ku, Sapporo 060-8648, Japan
| | - Ichiro Yasuda
- Third Department of Internal Medicine, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
| |
Collapse
|
3
|
Development and validation of an artificial neural network model for non-invasive gastric cancer screening and diagnosis. Sci Rep 2022; 12:21795. [PMID: 36526664 PMCID: PMC9758153 DOI: 10.1038/s41598-022-26477-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022] Open
Abstract
Non-invasive and cost-effective diagnosis of gastric cancer is essential to improve outcomes. Aim of the study was to establish a neural network model based on patient demographic data and serum biomarker panels to aid gastric cancer diagnosis. A total of 295 patients hospitalized in Nanjing Drum Tower hospital diagnosed with gastric cancer based on tissue biopsy, and 423 healthy volunteers were included in the study. Demographical information and tumor biomarkers were obtained from Hospital Information System (HIS) as original data. Pearson's correlation analysis was applied on 574 individuals' data (training set, 229 patients and 345 healthy volunteers) to analyze the relationship between each variable and the final diagnostic result. And independent sample t test was used to detect the differences of the variables. Finally, a neural network model based on 14 relevant variables was constructed. The model was tested on the validation set (144 individuals including 66 patients and 78 healthy volunteers). The predictive ability of the proposed model was compared with other common machine learning models including logistic regression and random forest. Tumor markers contributing significantly to gastric cancer screening included CA199, CA125, AFP, and CA242 were identified, which might be considered as important inspection items for gastric cancer screening. The accuracy of the model on validation set was 86.8% and the F1-score was 85.0%, which were better than the performance of other models under the same condition. A non-invasive and low-cost artificial neural network model was developed and proved to be a valuable tool to assist gastric cancer diagnosis.
Collapse
|
4
|
Ren J, Dai Y, Chao F, Tang D, Gu J, Niu G, Xia J, Wang X, Song T, Hu Z, Hong R, Ke C. A Nomogram for Predicting the Cancer-Specific Survival of Patients with Initially Diagnosed Metastatic Gastric Cancer. Clin Med Insights Oncol 2022; 16:11795549221142095. [DOI: 10.1177/11795549221142095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 11/13/2022] [Indexed: 12/15/2022] Open
Abstract
Background: There are few models to predict the survival of patients of different ethnicities initially diagnosed with metastatic gastric cancer (mGC). Therefore, the aim of this study was to construct a nomogram to predict the cancer-specific survival (CSS) of these patients. Methods: Data for 994 patients initially diagnosed with mGC between 2000 and 2013 were extracted from the Surveillance, Epidemiology, and End Results database. Patients were randomly classified into a training (n = 696) or internal validation (n = 298) cohort, and a cohort of 133 patients from Fudan cohort was used for external validation. A nomogram to predict the CSS of mGC patients was derived and validated using a concordance index (C-index), calibration curves, and decision-curve analysis (DCA). Results: Multivariate Cox regression indicated that five factors were independent predictors of CSS: differentiation grade, T stage, N stage, metastatic site at diagnosis, and with or without chemotherapy. Thus, these factors were integrated into the nomogram model. The C-index value of the nomogram model was 0.63 (95% CI: 0.60–0.65), and those of the internal and external validation cohorts were 0.60 (95%: CI 0.55–0.64) and 0.63 (95%: CI 0.57–0.69), respectively. The calibration curves showed good consistency between the actual and predicted survival rates in both the internal and external validation cohorts. The DCA also showed the clinical utility of the nomogram model. Conclusions: We established a practical nomogram to predict the CSS of patients initially diagnosed with mGC. The nomogram can be used for individualized prediction of survival and to guide clinicians in making treatment decisions.
Collapse
Affiliation(s)
- Jun Ren
- Department of General Surgery, Shanghai Fifth People’s Hospital, Fudan University, Shanghai, P.R. China
- Department of General Surgery, Northern Jiangsu People’s Hospital, Clinical Medical School, Yangzhou University, Yangzhou, P.R. China
| | - Yuedi Dai
- Department of Medical Oncology, Minhang Branch, Fudan University Shanghai Cancer Center, Shanghai, P.R. China
| | - Fei Chao
- Department of Anesthesiology, Northern Jiangsu People’s Hospital, Clinical Medical School, Yangzhou University, Yangzhou, P.R. China
| | - Dong Tang
- Department of General Surgery, Northern Jiangsu People’s Hospital, Clinical Medical School, Yangzhou University, Yangzhou, P.R. China
| | - Jiawei Gu
- Department of General Surgery, Shanghai Fifth People’s Hospital, Fudan University, Shanghai, P.R. China
| | - Gengming Niu
- Department of General Surgery, Shanghai Fifth People’s Hospital, Fudan University, Shanghai, P.R. China
| | - Jie Xia
- Department of General Surgery, Shanghai Fifth People’s Hospital, Fudan University, Shanghai, P.R. China
| | - Xin Wang
- Department of General Surgery, Shanghai Fifth People’s Hospital, Fudan University, Shanghai, P.R. China
| | - Tao Song
- Department of General Surgery, Shanghai Fifth People’s Hospital, Fudan University, Shanghai, P.R. China
| | - Zhiqing Hu
- Department of General Surgery, Shanghai Fifth People’s Hospital, Fudan University, Shanghai, P.R. China
| | - Runqi Hong
- Department of General Surgery, Shanghai Fifth People’s Hospital, Fudan University, Shanghai, P.R. China
| | - Chongwei Ke
- Department of General Surgery, Shanghai Fifth People’s Hospital, Fudan University, Shanghai, P.R. China
| |
Collapse
|
5
|
Moya-Salazar J, Cáceres E, Blejer J, Gonzalez C, Contreras-Pulache H. Frequency of allogenic blood transfusion in patients with gastrointestinal cancer: a cross-sectional study in Peru. Ecancermedicalscience 2021; 15:1289. [PMID: 34824612 PMCID: PMC8580600 DOI: 10.3332/ecancer.2021.1289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Indexed: 12/24/2022] Open
Abstract
Background Gastrointestinal cancer demands a high frequency of transfusions, and the high availability of blood products. We aimed to determine the frequency of blood transfusions and the most used blood products according to the type of gastrointestinal cancer. Methods A cross-sectional study was conducted in a Peruvian Type I Hemotherapy and Blood Bank Service of a Private Oncological Clinic during 2016–2018. We included patients with gastrointestinal cancer using the International Code of Diseases. The donations were made in compliance with the requirements of the Programa Nacional de Hemoterapía y Banco de Sangre and in accordance with the Standardised Operational Procedure of the clinic. Results We analysed 3,022 patients, of which 163 (5.4%) had gastrointestinal cancer (67.1 ± 12 years). The 80 (49.1%) men did not show significant differences with the 83 (50.9%) women (p = 0.178). The most frequent neoplasia was the colon (41.7%) and pancreas (37.4%). Three hundred and four blood products were transfused (average 1.8 ± 2.5 units (range: 1–30 units/patient)), of which 81.3% (247 units) were red blood cells concentrated, 8.6% (26 units) were fresh-frozen-plasma (FFP) and 6.6% (20 units) were cryoprecipitate. The type of cancer that most blood products demanded was colon neoplasia (41.8%), followed by pancreatic cancer (26.3%) and liver cancer (10.9%). We determined that ~55% of patients were O Rh(D)+ and in five patients we were poly-transfused. Conclusion Our findings suggested that patients with gastrointestinal cancer require large numbers of transfusions of blood cell concentrate and FFP. Also, we showed that cancer of the colon, pancreas and liver demanded more than 75% of blood products.
Collapse
Affiliation(s)
- Jeel Moya-Salazar
- Pathology Department, Hospital Nacional Docente Madre-Niño San Bartolomé, Lima 15001, Peru.,School of Medicine, Faculties of Health Science, Universidad Norbert Wiener, Lima 15001, Peru
| | - Eulogio Cáceres
- Service of Blood Bank, Department of Clinical Pathology, Oncosalud, Lima 15001, Peru
| | - Jorgelina Blejer
- Transfusion Transmissible Infections Section, Fundación Hemocentro Buenos Aires, Buenos Aires 1407, Argentina
| | - Carlos Gonzalez
- Hemotherapy Department, Hospital de Infecciosas F.J. Muñiz, Buenos Aires 1407, Argentina
| | - Hans Contreras-Pulache
- School of Medicine, Faculties of Health Science, Universidad Norbert Wiener, Lima 15001, Peru
| |
Collapse
|
6
|
Ando T, Ueda A, Ogawa K, Motoo I, Kajiura S, Nakajima T, Hirano K, Okumura T, Tsukada K, Hara T, Suzuki N, Nakada N, Horikawa N, Fujii T, Yasuda I. Prognosis of Immune-related Adverse Events in Patients With Advanced Gastric Cancer Treated With Nivolumab or Pembrolizumab: A Multicenter Retrospective Analysis. In Vivo 2021; 35:475-482. [PMID: 33402499 DOI: 10.21873/invivo.12281] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 10/19/2020] [Accepted: 10/21/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND Immune-checkpoint inhibitors (ICI), including nivolumab and pembrolizumab, are among the standard treatments for previously treated advanced gastric cancer (AGC). This study aimed to evaluate the frequency of immune-related adverse events (irAEs) and the correlation between irAEs and their efficacy in AGC cases. PATIENTS AND METHODS Patients were divided into two groups according to irAE occurrence. The frequency of irAEs and the treatment outcome (response rate [RR], progression-free survival [PFS], and overall survival [OS]) were evaluated. The survival rates were evaluated by landmark analysis considering lead-time bias. RESULTS Among 108 patients who received nivolumab or pembrolizumab, 17 (15.7%) had irAEs. In a 4-week landmark analysis, the RR, median PFS, and median OS were 28.5%, 3.9 months (95% CI=2.8-9.3), and 12.2 months (95% CI=3.8-NA) in patients with irAEs, while 3.0% (2/65), 1.8 months (95% CI=1.4-2.1), and 3.5 months (95% CI, 2.9-5.1) in patients without irAEs, respectively. In multivariate analysis, irAEs were associated with better PFS (HR=2.08, 95% CI=1.34-3.21). CONCLUSION The occurrence of irAEs was associated with a better clinical outcome of ICIs in patients with AGC.
Collapse
Affiliation(s)
- Takayuki Ando
- Third Department of Internal Medicine, University of Toyama, Toyama, Japan;
| | - Akira Ueda
- Department of Medical Oncology, Toyama Red Cross Hospital, Toyama, Japan
| | - Kohei Ogawa
- Department of Medical Oncology, Toyama Prefectural Central Hospital, Toyama, Japan
| | - Iori Motoo
- Third Department of Internal Medicine, University of Toyama, Toyama, Japan
| | - Shinya Kajiura
- Third Department of Internal Medicine, University of Toyama, Toyama, Japan
| | - Takahiko Nakajima
- Department of Diagnostic Pathology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Katsuhisa Hirano
- Department of Surgery and Science, Faculty of Medicine, Academic Assembly, University of Toyama, Toyama, Japan
| | - Tomoyuki Okumura
- Department of Surgery and Science, Faculty of Medicine, Academic Assembly, University of Toyama, Toyama, Japan
| | - Kenichiro Tsukada
- Department of Gastroenterology, Kouseiren Takaoka Hospital, Takaoka, Japan
| | - Takuo Hara
- Department of Surgery, Kouseiren Takaoka Hospital, Takaoka, Japan
| | - Nobuhiro Suzuki
- Department of Gastroenterology, Joetsu General Hospital, Joetsu, Japan
| | - Naokatsu Nakada
- Itoigawa Community Medical Unit, Toyama University Hospital, Itoigawa, Japan
| | - Naoki Horikawa
- Department of Surgery, Takaoka City Hospital, Takaoka, Japan
| | - Tsutomu Fujii
- Department of Surgery and Science, Faculty of Medicine, Academic Assembly, University of Toyama, Toyama, Japan
| | - Ichiro Yasuda
- Third Department of Internal Medicine, University of Toyama, Toyama, Japan
| |
Collapse
|
7
|
Iwasa S, Kudo T, Takahari D, Hara H, Kato K, Satoh T. Practical guidance for the evaluation of disease progression and the decision to change treatment in patients with advanced gastric cancer receiving chemotherapy. Int J Clin Oncol 2020; 25:1223-1232. [PMID: 32347434 PMCID: PMC7329754 DOI: 10.1007/s10147-020-01684-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 04/16/2020] [Indexed: 02/08/2023]
Abstract
After failure of first-line chemotherapy with fluoropyrimidines and platinum compounds for advanced gastric cancer, second-line chemotherapy with ramucirumab plus paclitaxel, which elicits a durable response, and third-line or later chemotherapy with nivolumab have been shown to lead to a more favorable prognosis in advanced gastric cancer patients. As new and more effective drugs are now available, sequential chemotherapy would contribute to prolonged survival. From this point of view, the patient's disease course should be frequently monitored in order to adapt treatment regimens. This review summarizes the points to note in regard to radiological assessment, and discusses the integration of prognostic factors, tumor markers, and clinical symptoms that need to be taken into account to change treatment at an appropriate timing.
Collapse
Affiliation(s)
- Satoru Iwasa
- Gastrointestinal Medical Oncology Division, National Cancer Center Hospital, 5-1-1, Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Toshihiro Kudo
- Department of Medical Oncology, Osaka International Cancer Institute, Osaka, Japan
| | - Daisuke Takahari
- Department of Gastroenterology, Cancer Institute Hospital of the Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Hiroki Hara
- Department of Gastroenterology, Saitama Cancer Center, Saitama, Japan
| | - Ken Kato
- Gastrointestinal Medical Oncology Division, National Cancer Center Hospital, 5-1-1, Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan.
| | - Taroh Satoh
- Frontier Science for Cancer and Chemotherapy, Osaka University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
8
|
|
9
|
Hamamoto Y, Piao Y, Makiyama A. Achieving sequential therapy in advanced gastric cancer: the importance of appropriate patient management for the elderly and/or those with ascites. Gastric Cancer 2020; 23:363-372. [PMID: 32236760 PMCID: PMC7165131 DOI: 10.1007/s10120-020-01067-3] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 03/21/2020] [Indexed: 02/07/2023]
Abstract
Treatment options for patients with advanced gastric cancer (AGC) are limited. One approach to improving survival in patients with AGC is to optimize the available agents via sequential therapy. However, clinical trial reports of first-line chemotherapy indicate that elderly patients and patients with massive ascites are less likely to receive subsequent lines of therapy. In addition, clinical trials of second- and third-line chemotherapy generally exclude these two patient populations because they are likely to have poor performance status and additional issues that are difficult to manage. Good patient management is likely to be key to the successful use of sequential therapy in these two patient populations by minimizing adverse effects to allow patients to derive benefit from the additional treatment. This narrative review summarizes the available information on AGC treatment and patient management in elderly patients and patients with massive ascites. The available data suggest that elderly patients benefit from chemotherapy; however, monitoring toxicity is essential to avoid chemotherapy-related toxicities. Important aspects of patient management for elderly patients include symptom monitoring, nutritional support, and fall prevention. The available data for patients with massive ascites show limited success for a range of treatment approaches, including systemic chemotherapy. The management of ascites is also challenging, with no clear guidance on the preferred strategies. To address these gaps in knowledge, future clinical trials should incorporate more inclusive eligibility criteria to enroll populations of patients with AGC that are more reflective of the real-world population with respect to age, complications, and overall health status.
Collapse
Affiliation(s)
- Yasuo Hamamoto
- Keio Cancer Center, Keio University School of Medicine, Tokyo, Japan
| | | | - Akitaka Makiyama
- Cancer Center, Gifu University Hospital, 1-1 Yanagido, Gifu, 501-1194 Japan
| |
Collapse
|
10
|
Park HS, Kwon WS, Park S, Jo E, Lim SJ, Lee CK, Lee JB, Jung M, Kim HS, Beom SH, Park JY, Kim TS, Chung HC, Rha SY. Comprehensive immune profiling and immune-monitoring using body fluid of patients with metastatic gastric cancer. J Immunother Cancer 2019; 7:268. [PMID: 31639056 PMCID: PMC6805480 DOI: 10.1186/s40425-019-0708-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 08/14/2019] [Indexed: 02/08/2023] Open
Abstract
Background The aim of this study is to profile the cytokines and immune cells of body fluid from metastatic gastric cancer (mGC), and evaluate the potential role as a prognostic factor and the feasibility as a predictive biomarker or monitoring source for immune checkpoint inhibitor. Methods Body fluid including ascites and pleural fluid were obtained from 55 mGC patients and 24 matched blood. VEGF-A, IL-10, and TGF-β1 were measured and immune cells were profiled by fluorescence assisted cell sorting (FACS). Results VEGF-A and IL-10 were significantly higher in body fluid than in plasma of mGC. Proportion of T lymphocytes with CD69 or PD-1, memory T cell marked with CD45RO, and number of Foxp3+ T regulatory cells (Tregs) were significantly higher in body fluid than those in blood of mGC. Proportion of CD8 T lymphocyte with memory marker (CD45RO) and activation marker (HLA-DR), CD3 T lymphocyte with PD-1, and number of FoxP3+ Tregs were identified as independent prognostic factors. When patients were classified by molecular subgroups of primary tumor, VEGF-A was significantly higher in genomically stable (GS)-like group than that in chromosomal instability (CIN)-like group while PD-L1 positive tumor cells (%) showed opposite results. Monitoring immune dynamics using body fluid was also feasible. Early activated T cell marked with CD25 was significantly increased in chemotherapy treated group. Conclusions By analyzing cytokines and proportion of immune cells in body fluid, prognosis of patients with mGC can be predicted. Immune monitoring using body fluid may provide more effective treatment for patients with mGC. Electronic supplementary material The online version of this article (10.1186/s40425-019-0708-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hyung Soon Park
- Division of Medical Oncology, Department of Internal Medicine, St. Vincent's Hospital, The Catholic University of Korea, Suwon, South Korea.,Songdang Institute for Cancer Research, Yonsei University College of Medicine, Seoul, South Korea
| | - Woo Sun Kwon
- Songdang Institute for Cancer Research, Yonsei University College of Medicine, Seoul, South Korea.,Brain Korea 21 Project for Medical Sciences, Yonsei University College of Medicine, Seoul, South Korea
| | - Sejung Park
- Songdang Institute for Cancer Research, Yonsei University College of Medicine, Seoul, South Korea.,Brain Korea 21 Project for Medical Sciences, Yonsei University College of Medicine, Seoul, South Korea
| | - Eunji Jo
- Songdang Institute for Cancer Research, Yonsei University College of Medicine, Seoul, South Korea.,Brain Korea 21 Project for Medical Sciences, Yonsei University College of Medicine, Seoul, South Korea
| | - So Jung Lim
- Songdang Institute for Cancer Research, Yonsei University College of Medicine, Seoul, South Korea
| | - Choong-Kun Lee
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, 50-1 Yonsei-Ro, Seodaemun-gu, Seoul, 120-752, South Korea
| | - Jii Bum Lee
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, 50-1 Yonsei-Ro, Seodaemun-gu, Seoul, 120-752, South Korea
| | - Minkyu Jung
- Songdang Institute for Cancer Research, Yonsei University College of Medicine, Seoul, South Korea.,Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, 50-1 Yonsei-Ro, Seodaemun-gu, Seoul, 120-752, South Korea
| | - Hyo Song Kim
- Songdang Institute for Cancer Research, Yonsei University College of Medicine, Seoul, South Korea.,Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, 50-1 Yonsei-Ro, Seodaemun-gu, Seoul, 120-752, South Korea
| | - Seung-Hoon Beom
- Songdang Institute for Cancer Research, Yonsei University College of Medicine, Seoul, South Korea.,Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, 50-1 Yonsei-Ro, Seodaemun-gu, Seoul, 120-752, South Korea
| | - Jun Yong Park
- Division of Gastroenterology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Tae Soo Kim
- Songdang Institute for Cancer Research, Yonsei University College of Medicine, Seoul, South Korea
| | - Hyun Cheol Chung
- Songdang Institute for Cancer Research, Yonsei University College of Medicine, Seoul, South Korea.,Brain Korea 21 Project for Medical Sciences, Yonsei University College of Medicine, Seoul, South Korea.,Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, 50-1 Yonsei-Ro, Seodaemun-gu, Seoul, 120-752, South Korea
| | - Sun Young Rha
- Songdang Institute for Cancer Research, Yonsei University College of Medicine, Seoul, South Korea. .,Brain Korea 21 Project for Medical Sciences, Yonsei University College of Medicine, Seoul, South Korea. .,Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, 50-1 Yonsei-Ro, Seodaemun-gu, Seoul, 120-752, South Korea.
| |
Collapse
|
11
|
Russi S, Verma HK, Laurino S, Mazzone P, Storto G, Nardelli A, Zoppoli P, Calice G, La Rocca F, Sgambato A, Lucci V, Falco G, Ruggieri V. Adapting and Surviving: Intra and Extra-Cellular Remodeling in Drug-Resistant Gastric Cancer Cells. Int J Mol Sci 2019; 20:ijms20153736. [PMID: 31370155 PMCID: PMC6695752 DOI: 10.3390/ijms20153736] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 07/26/2019] [Accepted: 07/29/2019] [Indexed: 02/06/2023] Open
Abstract
Despite the significant recent advances in clinical practice, gastric cancer (GC) represents a leading cause of cancer-related deaths in the world. In fact, occurrence of chemo-resistance still remains a daunting hindrance to effectiveness of the current approach to GC therapy. There is accumulating evidence that a plethora of cellular and molecular factors is implicated in drug-induced phenotypical switching of GC cells. Among them, epithelial-mesenchymal transition (EMT), autophagy, drug detoxification, DNA damage response and drug target alterations, have been reported as major determinants. Intriguingly, resistant GC phenotype may be the result of GC cell-induced tumor microenvironment (TME) remodeling, which is currently emerging as a key player in promoting drug resistance and overcoming cytotoxic effects of drugs. In this review, we discuss the possible mechanisms of drug resistance and their involvement in determining current GC therapies failure.
Collapse
Affiliation(s)
- Sabino Russi
- Laboratory of Preclinical and Translational Research, IRCCS-Referral Cancer Center of Basilicata (CROB), 85028 Rionero in Vulture (PZ), Italy
| | - Henu Kumar Verma
- Institute of Experimental Endocrinology and Oncology, National Research Council, 80131 Naples, Italy
- Section of Stem Cell and Development, Istituto di Ricerche Genetiche "Gaetano Salvatore" Biogem s.c. a.r.l., 83031 Ariano Irpino, Italy
| | - Simona Laurino
- Laboratory of Preclinical and Translational Research, IRCCS-Referral Cancer Center of Basilicata (CROB), 85028 Rionero in Vulture (PZ), Italy
| | - Pellegrino Mazzone
- Section of Stem Cell and Development, Istituto di Ricerche Genetiche "Gaetano Salvatore" Biogem s.c. a.r.l., 83031 Ariano Irpino, Italy
| | - Giovanni Storto
- Department of Nuclear Medicine, IRCCS-Referral Cancer Center of Basilicata (CROB), 85028 Rionero in Vulture (PZ), Italy
| | - Anna Nardelli
- Istituto di Biostrutture e Bioimmagini, Consiglio Nazionale delle Ricerche, 80145 Napoli, Italy
| | - Pietro Zoppoli
- Laboratory of Preclinical and Translational Research, IRCCS-Referral Cancer Center of Basilicata (CROB), 85028 Rionero in Vulture (PZ), Italy
| | - Giovanni Calice
- Laboratory of Preclinical and Translational Research, IRCCS-Referral Cancer Center of Basilicata (CROB), 85028 Rionero in Vulture (PZ), Italy
| | - Francesco La Rocca
- Laboratory of Clinical Research and Advanced Diagnostics, IRCCS-Referral Cancer Center of Basilicata (CROB), 85028 Rionero in Vulture (PZ), Italy
| | - Alessandro Sgambato
- Scientific Direction, IRCCS-Referral Cancer Center of Basilicata (CROB), 85028 Rionero in Vulture (PZ), Italy
| | - Valeria Lucci
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy
| | - Geppino Falco
- Section of Stem Cell and Development, Istituto di Ricerche Genetiche "Gaetano Salvatore" Biogem s.c. a.r.l., 83031 Ariano Irpino, Italy.
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy.
| | - Vitalba Ruggieri
- Laboratory of Preclinical and Translational Research, IRCCS-Referral Cancer Center of Basilicata (CROB), 85028 Rionero in Vulture (PZ), Italy.
| |
Collapse
|
12
|
A novel YAP1/SLC35B4 regulatory axis contributes to proliferation and progression of gastric carcinoma. Cell Death Dis 2019; 10:452. [PMID: 31175271 PMCID: PMC6555804 DOI: 10.1038/s41419-019-1674-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2019] [Revised: 05/06/2019] [Accepted: 05/13/2019] [Indexed: 12/18/2022]
Abstract
Solute carrier family 35 member B4 (SLC35B4), a nucleotide sugar transporter, is capable of transporting UDP-xylose and UDP-GlcNAc from the cytoplasm to the lumen of the endoplasmic reticulum and Golgi. SLC35B4 has a pivotal role in glycosylation of biological macromolecules. However, its functional roles and regulatory mechanisms in malignant diseases remain unknown. Here, using the cDNA arrays, promoter reporter assays, and chromatin immunoprecipitation assays, we demonstrated that SLC35B4 is directly transactivated by YAP1–TEADs complex in gastric cancer (GC) cells. CCK-8, plate colony formation and soft agar assays revealed that SLC35B4 is essential for survival and proliferation in GC cells and nude mice models. SLC35B4 expression is markedly higher in GC tissues compared with control noncancerous tissues. Immunohistochemistry revealed that SLC35B4 expression is positively correlated with YAP1 expression in human GC tissues, and this correlation is also confirmed in the GC TCGA data set. GC patients with high levels of SLC35B4 expression have poorer prognosis than those with low levels of SLC35B4 expression. Collectively, our findings defined SLC35B4 as an important downstream oncogenic target of YAP1, suggesting that dysregulated signaling of a novel YAP1/SLC35B4 axis promotes GC development and progression, and this axis could be a potential candidate for prognosis and therapeutics in GC.
Collapse
|
13
|
Aoki Y, Hamamoto Y, Ugamura A, Togasaki K, Suzuki T, Kawasaki K, Hirata K, Kasuga A, Sukawa Y, Kanai T, Takaishi H. Early administration of systemic chemotherapy should be�considered for scirrhous gastric cancer: A case report. Mol Clin Oncol 2018; 10:113-117. [DOI: 10.3892/mco.2018.1767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 09/06/2018] [Indexed: 11/05/2022] Open
Affiliation(s)
- Yu Aoki
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Keio University School of Medicine, Tokyo 160‑8582, Japan
| | - Yasuo Hamamoto
- Keio Cancer Center, Keio University School of Medicine, Tokyo 160‑8582, Japan
| | - Aya Ugamura
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Keio University School of Medicine, Tokyo 160‑8582, Japan
| | - Kazuhiro Togasaki
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Keio University School of Medicine, Tokyo 160‑8582, Japan
| | - Takeshi Suzuki
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Keio University School of Medicine, Tokyo 160‑8582, Japan
| | - Kenta Kawasaki
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Keio University School of Medicine, Tokyo 160‑8582, Japan
| | - Kenro Hirata
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Keio University School of Medicine, Tokyo 160‑8582, Japan
| | - Akiyoshi Kasuga
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Keio University School of Medicine, Tokyo 160‑8582, Japan
| | - Yasutaka Sukawa
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Keio University School of Medicine, Tokyo 160‑8582, Japan
| | - Takanori Kanai
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Keio University School of Medicine, Tokyo 160‑8582, Japan
| | - Hiromasa Takaishi
- Keio Cancer Center, Keio University School of Medicine, Tokyo 160‑8582, Japan
| |
Collapse
|
14
|
Shi B, Li Q, Ma X, Gao Q, Li L, Chu J. High expression of programmed cell death protein 1 on peripheral blood T-cell subsets is associated with poor prognosis in metastatic gastric cancer. Oncol Lett 2018; 16:4448-4454. [PMID: 30214579 PMCID: PMC6126159 DOI: 10.3892/ol.2018.9190] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2017] [Accepted: 05/24/2018] [Indexed: 12/16/2022] Open
Abstract
Immune checkpoints in solid tumors serve important roles in metastasis. The present study was designed to explore the expression of programmed cell death protein 1 (PD-1) on peripheral blood T-cell subsets and its role in the clinicopathological features and prognosis of patients with metastatic gastric cancer. The expression of PD-1 in peripheral blood T-cell subsets was detected in 100 metastatic gastric cancer patients prior to the first line chemotherapy by flow cytometric analysis. The potential associaton between the peripheral blood T-cell subsets PD-1 level and the clinicopathological features of patients with metastatic gastric cancer and the clinical outcomes was analyzed. The percent of high PD-1 expressed cluster of differentiation (CD)3+, CD3+CD4+ and CD3+CD8+ T-cells was 20.4, 13.0 and 9.4%, respectively in patients with metastatic gastric cancer. The overall survival (OS) and progression-free survival (PFS) rate of the 100 patients with metastatic gastric cancer was 12.2 and 3.9 months, respectively. Kaplan-Meier curve with long-rank analysis indicated that patients with higher PD-1+/CD3+, PD-1+/CD3+CD4+ and PD-1+/CD3+CD8+ levels had a worse prognosis (all P<0.05). Univariate and multivariate analysis revealed that high PD-1+/CD3+ [hazard ratio (HR), 2.145; P=0.015], high PD-1+/CD3+CD4+ (HR, 1.866; P=0.034) and high PD-1+/CD3+CD8+ (HR, 1.817; P=0.033) level in peripheral blood were independent risk factors for predicting the survival time of patients with metastatic gastric cancer. High PD-1+/CD3+, high PD-1+/CD3+CD4+ and high PD-1+/CD3+CD8+ expression conferred a lower overall survival rate in patients with metastatic gastric cancer. These results suggest that high PD-1 expression on peripheral blood T-cell subsets may potentially be novel prognostic biomarker for metastatic gastric cancer.
Collapse
Affiliation(s)
- Bian Shi
- Department of Integrated Traditional Chinese and Western Medical Oncology, Zhengzhou University Affiliated Cancer Hospital, Zhengzhou, Henan 450001, P.R. China
| | - Qiujian Li
- Department of Integrated Traditional Chinese and Western Medical Oncology, Zhengzhou University Affiliated Cancer Hospital, Zhengzhou, Henan 450001, P.R. China
| | - Xuhui Ma
- Department of Integrated Traditional Chinese and Western Medical Oncology, Zhengzhou University Affiliated Cancer Hospital, Zhengzhou, Henan 450001, P.R. China
| | - Qilong Gao
- Department of Integrated Traditional Chinese and Western Medical Oncology, Zhengzhou University Affiliated Cancer Hospital, Zhengzhou, Henan 450001, P.R. China
| | - Lu Li
- Department of Integrated Traditional Chinese and Western Medical Oncology, Zhengzhou University Affiliated Cancer Hospital, Zhengzhou, Henan 450001, P.R. China
| | - Junfeng Chu
- Department of Oncology, Zhengzhou University Affiliated Cancer Hospital, Zhengzhou, Henan 450001, P.R. China
| |
Collapse
|
15
|
Almasi S, Kennedy BE, El-Aghil M, Sterea AM, Gujar S, Partida-Sánchez S, El Hiani Y. TRPM2 channel-mediated regulation of autophagy maintains mitochondrial function and promotes gastric cancer cell survival via the JNK-signaling pathway. J Biol Chem 2018; 293:3637-3650. [PMID: 29343514 DOI: 10.1074/jbc.m117.817635] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 12/22/2017] [Indexed: 12/16/2022] Open
Abstract
A lack of effective treatment is one of the main factors contributing to gastric cancer-related death. Discovering effective targets and understanding their underlying anti-cancer mechanism are key to achieving the best response to treatment and to limiting side effects. Although recent studies have shown that the cation channel transient receptor potential melastatin-2 (TRPM2) is crucial for cancer cell survival, the exact mechanism remains unclear, limiting its therapeutic potential. Here, using molecular and functional assays, we investigated the role of TRPM2 in survival of gastric cancer cells. Our results indicated that TRPM2 knockdown in AGS and MKN-45 cells decreases cell proliferation and enhances apoptosis. We also observed that the TRPM2 knockdown impairs mitochondrial metabolism, indicated by a decrease in basal and maximal mitochondrial oxygen consumption rates and ATP production. These mitochondrial defects coincided with a decrease in autophagy and mitophagy, indicated by reduced levels of autophagy- and mitophagy-associated proteins (i.e. ATGs, LC3A/B II, and BNIP3). Moreover, we found that TRPM2 modulates autophagy through a c-Jun N-terminal kinase (JNK)-dependent and mechanistic target of rapamycin-independent pathway. We conclude that in the absence of TRPM2, down-regulation of the JNK-signaling pathway impairs autophagy, ultimately causing the accumulation of damaged mitochondria and death of gastric cancer cells. Of note, by inhibiting cell proliferation and promoting apoptosis, the TRPM2 down-regulation enhanced the efficacy of paclitaxel and doxorubicin in gastric cancer cells. Collectively, we provide compelling evidence that TRPM2 inhibition may benefit therapeutic approaches for managing gastric cancer.
Collapse
Affiliation(s)
| | | | | | - Andra M Sterea
- Physiology, Biophysics Faculty of Life Science, Dalhousie University, Halifax and
| | - Shashi Gujar
- Pathology.,Microbiology and Immunology, and.,the Centre for Innovative and Collaborative Health Services Research, Quality and System Performance, IWK Health Centre, Halifax, Nova Scotia B3H 4R2, Canada
| | - Santiago Partida-Sánchez
- Center for Microbial Pathogenesis, Research Institute at Nationwide Children's Hospital and.,the Department of Pediatrics, College of Medicine, Ohio State University, Columbus, Ohio 43205
| | - Yassine El Hiani
- Physiology, Biophysics Faculty of Life Science, Dalhousie University, Halifax and
| |
Collapse
|
16
|
Xu MD, Wang Y, Weng W, Wei P, Qi P, Zhang Q, Tan C, Ni SJ, Dong L, Yang Y, Lin W, Xu Q, Huang D, Huang Z, Ma Y, Zhang W, Sheng W, Du X. A Positive Feedback Loop of lncRNA- PVT1 and FOXM1 Facilitates Gastric Cancer Growth and Invasion. Clin Cancer Res 2016; 23:2071-2080. [PMID: 27756785 DOI: 10.1158/1078-0432.ccr-16-0742] [Citation(s) in RCA: 197] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Revised: 09/16/2016] [Accepted: 09/20/2016] [Indexed: 02/07/2023]
Abstract
Purpose: The long, noncoding RNA (lncRNA) PVT1 is an important epigenetic regulator with a critical role in human tumors. Here, we aimed to investigate the clinical application and the potential molecular mechanisms of PVT1 in gastric cancer tumorigenesis and progression.Experimental Design: The expression level of PVT1 was determined by RT-qPCR analysis in 190 pairs of gastric cancer tissues and adjacent normal gastric mucosa tissues (ANT). The biologic functions of PVT1 were assessed by in vitro and in vivo functional experiments. RNA protein pull-down assays and LS/MS mass spectrometry analysis were performed to detect and identify the PVT1- interacting protein FOXM1. Protein-RNA immunoprecipitation assays were conducted to examine the interaction of FOXM1 and PVT1 Chromatin immunoprecipitation (ChIP) and luciferase analyses were utilized to identify the binding site of FOXM1 on the PVT1 promoter.Results: The lncRNA PVT1 was significantly upregulated in gastric cancer tissues compared with ANTs. High expression of PVT1 predicted poor prognosis in patients with gastric cancer. PVT1 enhanced gastric cancer cell proliferation and invasion in vitro and in vivoPVT1 directly bound FOXM1 protein and increased FOXM1 posttranslationally. Moreover, PVT1 is also a FOXM1-responsive lncRNA, and FOXM1 directly binds to the PVT1 promoter to activate its transcription. Finally, PVT1 fulfilled its oncogenic functions in a FOXM1-mediated manner.Conclusions: Our study suggests that PVT1 promotes tumor progression by interacting with FOXM1. PVT1 may be a valuable prognostic predictor for gastric cancer, and the positive feedback loop of PVT1-FOXM1 could be a therapeutic target in pharmacologic strategies. Clin Cancer Res; 23(8); 2071-80. ©2016 AACR.
Collapse
Affiliation(s)
- Mi-Die Xu
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Institute of Pathology, Fudan University, Shanghai, China
| | - Yiqin Wang
- Department of Pathology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Weiwei Weng
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Institute of Pathology, Fudan University, Shanghai, China
| | - Ping Wei
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Institute of Pathology, Fudan University, Shanghai, China
| | - Peng Qi
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Institute of Pathology, Fudan University, Shanghai, China
| | - Qiongyan Zhang
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Institute of Pathology, Fudan University, Shanghai, China
| | - Cong Tan
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Institute of Pathology, Fudan University, Shanghai, China
| | - Shu-Juan Ni
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Institute of Pathology, Fudan University, Shanghai, China
| | - Lei Dong
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Institute of Pathology, Fudan University, Shanghai, China
| | - Yusi Yang
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Institute of Pathology, Fudan University, Shanghai, China
| | - Wanrun Lin
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Institute of Pathology, Fudan University, Shanghai, China
| | - Qinghua Xu
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China.,Hangzhou Canhelp Genomics Company Limited, Hangzhou, Zhejiang, China
| | - Dan Huang
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Institute of Pathology, Fudan University, Shanghai, China
| | - Zhaohui Huang
- Wuxi Oncology Institute, the Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China
| | - Yuqing Ma
- Department of Pathology, First Hospital Affiliated to Xinjiang Medical University, Urumqi, China
| | - Wei Zhang
- Department of Pathology, First Hospital Affiliated to Xinjiang Medical University, Urumqi, China
| | - Weiqi Sheng
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China. .,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Institute of Pathology, Fudan University, Shanghai, China
| | - Xiang Du
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China. .,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Institute of Pathology, Fudan University, Shanghai, China.,Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| |
Collapse
|