1
|
Ma C, Gao L, Song K, Gu B, Wang B, Pu W, Chen H. Exploring the therapeutic potential of diterpenes in gastric cancer: Mechanisms, efficacy, and clinical prospects. BIOMOLECULES & BIOMEDICINE 2024; 25:1-15. [PMID: 39151097 PMCID: PMC11647260 DOI: 10.17305/bb.2024.10887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 08/13/2024] [Accepted: 08/13/2024] [Indexed: 08/18/2024]
Abstract
Gastric cancer (GC) remains a significant global health challenge, particularly prevalent in East Asia. Despite advancements in various treatment modalities, the prognosis for patients, especially those in advanced stages, remains poor, highlighting the need for innovative therapeutic approaches. This review explores the promising potential of diterpenes, naturally occurring compounds with robust anticancer properties, derived from diverse sources such as plants, marine organisms, and fungi. Diterpenes have shown the ability to influence reactive oxygen species (ROS) generation, ferroptosis, and autophagy, positioning them as attractive candidates for novel cancer therapies. This review explores the mechanisms of action of diterpenes and their clinical implications for the treatment of GC. Additionally, it addresses the challenges in translating these compounds from preclinical studies to clinical applications, emphasizing the need for further research to enhance their therapeutic profiles and minimize potential side effects. The discussion underscores the importance of diterpenes in future anticancer strategies, particularly in the fight against gastric cancer.
Collapse
Affiliation(s)
- Chenhui Ma
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Lei Gao
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Kewei Song
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Baohong Gu
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Bofang Wang
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Weigao Pu
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Hao Chen
- Gansu Provincial Key Laboratory of Environmental Oncology, Lanzhou, China
- Department of Tumor Surgery, Lanzhou University Second Hospital, Lanzhou, China
| |
Collapse
|
2
|
Proskuriakova E, Balamurali V, Hooda A, Khosla P. Metastatic gastric adenocarcinoma discovered in the bone marrow. BMJ Case Rep 2024; 17:e260217. [PMID: 39306333 DOI: 10.1136/bcr-2024-260217] [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] [Indexed: 01/26/2025] Open
Abstract
Gastric cancer primarily metastasizes to the peritoneum, liver and lungs, with bone marrow involvement being a rare occurrence, found in less than 1% of cases. Disseminated carcinomatosis of the bone marrow (DCBM) is characterised by widespread infiltration of cancer cells into the bone marrow, leading to haematological disorders such as disseminated intravascular coagulation and thrombocytopenia. We present a unique case of a man in his late 50s with acute thrombocytopenia as the initial symptom, subsequently diagnosed with gastric cancer on bone marrow examination. Despite receiving chemotherapy, the patient's condition deteriorated rapidly, emphasising the challenging management and poor prognosis associated with DCBM. This case underscores the need for improved diagnostic strategies and therapeutic approaches to enhance patient outcomes in DCBM associated with gastric cancer.
Collapse
Affiliation(s)
| | - Vaisny Balamurali
- Ross University School of Medicine, Two Tree Hill St. Michael, Barbados
| | - Anuradha Hooda
- Haematology and Oncology, Mount Sinai Hospital, Chicago, Illinois, USA
| | - Paramjeet Khosla
- Haematology and Oncology, Mount Sinai Hospital, Chicago, Illinois, USA
| |
Collapse
|
3
|
Wang Y, Gan X, Cheng X, Jia Y, Wang G, Tang X, Du H, Li X, Liu X, Xing X, Ji J, Li Z. ABCC2 induces metabolic vulnerability and cellular ferroptosis via enhanced glutathione efflux in gastric cancer. Clin Transl Med 2024; 14:e1754. [PMID: 39095325 PMCID: PMC11296884 DOI: 10.1002/ctm2.1754] [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: 03/01/2024] [Revised: 05/30/2024] [Accepted: 06/16/2024] [Indexed: 08/04/2024] Open
Abstract
BACKGROUND Although it is traditionally believed that ATP binding cassette subfamily C member 2 (ABCC2) is a multidrug resistance-associated protein correlated with a worse prognosis, our previous and several other studies demonstrated the contrary to be true in gastric cancer (GC). We aim to explore the underlying mechanism of this discovery. METHODS Our study utilized whole-exome sequencing (WES), RNA sequencing, and droplet digital PCR (ddPCR) analysis of 80 gastric cancer samples, along with comprehensive immunohistochemical (IHC) analysis of 1044 human GC tissue samples.By utilizing CRISPRCas9 to genetically modify cell lines with the ABCC2-24C > T (rs717620) point mutation and conducting dual-luciferase reporter assays, we identified that transcription factors SOX9 and ETS1 serve as negative regulators of ABCC2 expression. Seahorse assay and mass spectrometry were used to discover altered metabolic patterns. Gain and loss-of-function experiments in GC cell lines and preclinical models were carried out to validate ABCC2 biological function. RESULTS ABCC2 high expression correlated with better prognosis, and rs717620 can influence ABCC2 expression by disrupting the binding of ETS1 and SOX9. Gain and loss-of-function experiments in GC cell lines demonstrated amino acid deprivation reduces proliferation, migration, and drug resistance in ABCC2-high GC cells. ABCC2 leads to reduced intracellular amino acid pools and disruption of cellular energy metabolism. This phenomenon depended on ABCC2-mediated GSH extrusion, resulting in alterations in redox status, thereby increasing the cell's susceptibility to ferroptosis. Furthermore, patient-derived organoids and patient-derived tumor-like cell clusters were used to observe impact of ABCC2 on therapeutic effect. In the xenograft model with high ABCC2 expression, we observed that constricting amino acid intake in conjunction with GPX4 inactivation resulted in notable tumor regression. CONCLUSIONS Our findings demonstrate a significant role of ABCC2 in amino acid metabolism and ferroptosis by mediating GSH efflux in GC. This discovery underlines the potential of combining multiple ferroptosis targets as a promising therapeutic strategy for GC with high ABCC2 expression. HIGHLIGHTS ABCC2 plays a crucial role in inducing metabolic vulnerability and ferroptosis in gastric cancer through enhanced glutathione efflux. The ABCC2 24C > T polymorphism is a key factor influencing its expression. These results highlight the potential of ABCC2 as a predictive biomarker and therapeutic target in gastric cancer.
Collapse
Affiliation(s)
- Yiding Wang
- Department of Gastrointestinal Cancer Translational ResearchKey Laboratory of Carcinogenesis and Translational Research (Ministry of Education)Peking University Cancer Hospital & InstituteBeijingP.R. China
- Department of Gastrointestinal Cancer CenterWard IPeking University Cancer Hospital & InstituteBeijingP.R. China
| | - Xuejun Gan
- Department of Gastrointestinal Cancer Translational ResearchKey Laboratory of Carcinogenesis and Translational Research (Ministry of Education)Peking University Cancer Hospital & InstituteBeijingP.R. China
- Department of Gastrointestinal Cancer CenterWard IPeking University Cancer Hospital & InstituteBeijingP.R. China
| | - Xiaojing Cheng
- Department of Gastrointestinal Cancer Translational ResearchKey Laboratory of Carcinogenesis and Translational Research (Ministry of Education)Peking University Cancer Hospital & InstituteBeijingP.R. China
| | - Yongning Jia
- Department of Gastrointestinal Cancer CenterWard IPeking University Cancer Hospital & InstituteBeijingP.R. China
| | - Gangjian Wang
- Department of Gastrointestinal Cancer Translational ResearchKey Laboratory of Carcinogenesis and Translational Research (Ministry of Education)Peking University Cancer Hospital & InstituteBeijingP.R. China
- Department of Gastrointestinal Cancer CenterWard IPeking University Cancer Hospital & InstituteBeijingP.R. China
| | - Xiaohuan Tang
- Department of Gastrointestinal Cancer Translational ResearchKey Laboratory of Carcinogenesis and Translational Research (Ministry of Education)Peking University Cancer Hospital & InstituteBeijingP.R. China
- Department of Gastrointestinal Cancer CenterWard IPeking University Cancer Hospital & InstituteBeijingP.R. China
| | - Hong Du
- Department of Gastrointestinal Cancer Translational ResearchKey Laboratory of Carcinogenesis and Translational Research (Ministry of Education)Peking University Cancer Hospital & InstituteBeijingP.R. China
| | - Xiaomei Li
- Department of Gastrointestinal Cancer Translational ResearchKey Laboratory of Carcinogenesis and Translational Research (Ministry of Education)Peking University Cancer Hospital & InstituteBeijingP.R. China
| | - Xijuan Liu
- Department of Central LaboratoryKey Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing)Peking University Cancer Hospital & InstituteBeijingChina
| | - Xiaofang Xing
- Department of Gastrointestinal Cancer Translational ResearchKey Laboratory of Carcinogenesis and Translational Research (Ministry of Education)Peking University Cancer Hospital & InstituteBeijingP.R. China
| | - Jiafu Ji
- Department of Gastrointestinal Cancer Translational ResearchKey Laboratory of Carcinogenesis and Translational Research (Ministry of Education)Peking University Cancer Hospital & InstituteBeijingP.R. China
- Department of Gastrointestinal Cancer CenterWard IPeking University Cancer Hospital & InstituteBeijingP.R. China
| | - Ziyu Li
- Department of Gastrointestinal Cancer Translational ResearchKey Laboratory of Carcinogenesis and Translational Research (Ministry of Education)Peking University Cancer Hospital & InstituteBeijingP.R. China
- Department of Gastrointestinal Cancer CenterWard IPeking University Cancer Hospital & InstituteBeijingP.R. China
| |
Collapse
|
4
|
Kim HJ, Cho YB, Heo K, Kim JW, Shin HG, Lee EB, Park SM, Park JB, Lee S. Targeting cell surface glucose-regulated protein 94 in gastric cancer with an anti-GRP94 human monoclonal antibody. BMB Rep 2024; 57:188-193. [PMID: 38449302 PMCID: PMC11058359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 11/22/2023] [Accepted: 02/14/2024] [Indexed: 03/08/2024] Open
Abstract
Gastric cancer (GC), a leading cause of cancer-related mortality, remains a significant challenge despite recent therapeutic advancements. In this study, we explore the potential of targeting cell surface glucose-regulated protein 94 (GRP94) with antibodies as a novel therapeutic approach for GC. Our comprehensive analysis of GRP94 expression across various cancer types, with a specific focus on GC, revealed a substantial overexpression of GRP94, highlighting its potential as a promising target. Through in vitro and in vivo efficacy assessments, as well as toxicological analyses, we found that K101.1, a fully human monoclonal antibody designed to specifically target cell surface GRP94, effectively inhibits GC growth and angiogenesis without causing in vivo toxicity. Furthermore, our findings indicate that K101.1 promotes the internalization and concurrent downregulation of cell surface GRP94 on GC cells. In conclusion, our study suggests that cell surface GRP94 may be a potential therapeutic target in GC, and that antibody-based targeting of cell surface GRP94 may be an effective strategy for inhibiting GRP94-mediated GC growth and angiogenesis. [BMB Reports 2024; 57(4): 188-193].
Collapse
Affiliation(s)
- Hyun Jung Kim
- Department of Biopharmaceutical Chemistry, Kookmin University, Seoul 02707, Korea
| | - Yea Bin Cho
- Department of Chemistry, Kookmin University, Seoul 02707, Korea
| | - Kyun Heo
- Department of Biopharmaceutical Chemistry, Kookmin University, Seoul 02707, Korea
- Department of Chemistry, Kookmin University, Seoul 02707, Korea
- Antibody Research Institute, Kookmin University, Seoul 02707, Korea
| | - Ji Woong Kim
- Department of Biopharmaceutical Chemistry, Kookmin University, Seoul 02707, Korea
| | - Ha Gyeong Shin
- Department of Biopharmaceutical Chemistry, Kookmin University, Seoul 02707, Korea
| | - Eun-bi Lee
- Department of Biopharmaceutical Chemistry, Kookmin University, Seoul 02707, Korea
| | - Seong-Min Park
- Department of Cancer Biomedical Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang 10408, Korea
| | - Jong Bae Park
- Department of Cancer Biomedical Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang 10408, Korea
| | - Sukmook Lee
- Department of Biopharmaceutical Chemistry, Kookmin University, Seoul 02707, Korea
- Department of Chemistry, Kookmin University, Seoul 02707, Korea
- Antibody Research Institute, Kookmin University, Seoul 02707, Korea
| |
Collapse
|
5
|
Bilan F, Amini M, Doustvandi MA, Tohidast M, Baghbanzadeh A, Hosseini SS, Mokhtarzadeh A, Baradaran B. Simultaneous suppression of miR-21 and restoration of miR-145 in gastric cancer cells; a promising strategy for inhibition of cell proliferation and migration. BIOIMPACTS : BI 2023; 14:27764. [PMID: 38505672 PMCID: PMC10945301 DOI: 10.34172/bi.2023.27764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 06/13/2023] [Accepted: 06/25/2023] [Indexed: 03/21/2024]
Abstract
Introduction Gastric cancer (GC) is the third leading cause of cancer-related death worldwide. microRNAs are a group of regulatory non-coding RNAs that are involved in GC progression. miR-145 as a tumor suppressor and miR-21 as an oncomiR were shown to be dysregulated in many cancers including GC. This research aimed to enhance the expression of miR-145 while reducing the expression of miR-21 and examine their impact on the proliferation, apoptosis, and migration of GC cells. Methods KATO III cells with high expression levels of miR-21-5p and low expression of miR-145-5p were selected. These cells were then transfected with either miR-145-5p mimics or anti-miR-21-5p, alone or in combination. Afterward, the cell survival rate was determined using the MTT assay, while apoptosis induction was investigated through V-FITC/PI and DAPI staining. Additionally, cell migration was examined using the wound healing assay, and cell cycle progression was analyzed through flow cytometry. Furthermore, gene expression levels were quantified utilizing the qRT-PCR technique. Results The study's findings indicated that the co-replacement of miR-145-5p and anti-miR-21-5p led to a decrease in cell viability and the induction of apoptosis in GC cells. This was achieved via modulating the expression of Bax and Bcl-2, major cell survival regulators. Additionally, the combination therapy significantly increased sub-G1 cell cycle arrest and reduced cell migration by downregulating MMP-9 expression as an epithelial-mesenchymal transition marker. This study provides evidence for the therapeutic possibility of the combination of miR-145-5p and anti-miR-21-5p and also suggests that they could inhibit cell proliferation by modulating the PTEN/AKT1 signaling pathway. Conclusion Our research revealed that utilizing miR-145-5p and anti-miR-21-5p together could be a promising therapeutic approach for treating GC.
Collapse
Affiliation(s)
- Farzaneh Bilan
- Department of Biological Science, Faculty of Basic Science, Higher Education Institute of Rab-Rashid, Tabriz, Iran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Amini
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Maryam Tohidast
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Baghbanzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Ahad Mokhtarzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
6
|
CT-derived body composition measurements as predictors for neoadjuvant treatment tolerance and survival in gastroesophageal adenocarcinoma. ABDOMINAL RADIOLOGY (NEW YORK) 2023; 48:211-219. [PMID: 36209446 DOI: 10.1007/s00261-022-03695-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 09/21/2022] [Accepted: 09/26/2022] [Indexed: 01/21/2023]
Abstract
PURPOSE Treatment for gastroesophageal adenocarcinomas can result in significant morbidity and mortality. The purpose of this study is to supplement methods for choosing treatment strategy by assessing the relationship between CT-derived body composition, patient, and tumor features, and clinical outcomes in this population. METHODS Patients with neoadjuvant treatment, biopsy-proven gastroesophageal adenocarcinoma, and initial staging CTs were retrospectively identified from institutional clinic encounters between 2000 and 2019. Details about patient, disease, treatment, and outcomes (including therapy tolerance and survival) were extracted from electronic medical records. A deep learning semantic segmentation algorithm was utilized to measure cross-sectional areas of skeletal muscle (SM), visceral fat (VF), and subcutaneous fat (SF) at the L3 vertebra level on staging CTs. Univariate and multivariate analyses were performed to assess the relationships between predictors and outcomes. RESULTS 142 patients were evaluated. Median survival was 52 months. Univariate and multivariate analysis showed significant associations between treatment tolerance and SM and VF area, SM to fat and VF to SF ratios, and skeletal muscle index (SMI) (p = 0.004-0.04). Increased survival was associated with increased body mass index (BMI) (p = 0.01) and increased SMI (p = 0.004). A multivariate Cox model consisting of BMI, SMI, age, gender, and stage demonstrated that patients in the high-risk group had significantly lower survival (HR = 1.77, 95% CI = 1.13-2.78, p = 0.008). CONCLUSION CT-based measures of body composition in patients with gastroesophageal adenocarcinoma may be independent predictors of treatment complications and survival and can supplement methods for assessing functional status during treatment planning.
Collapse
|
7
|
Identification of Kynurenic Acid-Induced Apoptotic Biomarkers in Gastric Cancer-Derived AGS Cells through Next-Generation Transcriptome Sequencing Analysis. Nutrients 2022; 15:nu15010193. [PMID: 36615849 PMCID: PMC9823332 DOI: 10.3390/nu15010193] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 12/23/2022] [Accepted: 12/26/2022] [Indexed: 01/04/2023] Open
Abstract
Understanding the triggers and therapeutic targets for gastric cancer, one of the most common cancers worldwide, can provide helpful information for the development of therapeutics. RNA sequencing technology can be utilized to identify complex disease targets and therapeutic applications. In the present study, we aimed to establish the pharmacological target of Kynurenic acid (KYNA) for gastric cancer AGS cells and to identify the biological network. RNA sequencing identified differentially expressed genes (DEGs) between KYNA-treated and untreated cells. A total of 278 genes were differentially expressed, of which 120 genes were up-regulated, and 158 genes were down-regulated. Gene ontology results confirmed that KYNA had effects such as a reduction in genes related to DNA replication and nucleosome organization on AGS cells. Protein-protein interaction was confirmed through STRING analysis, and it was confirmed that cancer cell growth and proliferation were inhibited through KEGG, Reactome, and Wiki pathway analysis, and various signaling pathways related to cancer cell death were induced. It was confirmed that KYNA treatment reduced the gene expression of cancer-causing AP-1 factors (Fos, Jun, ATF, and JDP) in AGS cell lines derived from gastric cancer. Overall, using next-generation transcriptome sequencing data and bioinformatics tools, we confirmed that KYNA had an apoptosis effect by inducing changes in various genes, including factor AP-1, in gastric cancer AGS cells. This study can identify pharmacological targets for gastric cancer treatment and provide a valuable resource for drug development.
Collapse
|
8
|
Bai L, Dong K, Tong D, Shi X, Wei S, Cai Y. lncRNA HIT000218960 enhances resistance to 5‑fluorouracil by promoting HMGA2 and activating the AKT/mTOR/P70S6K pathway in gastric cancer cells. Exp Ther Med 2022; 24:527. [PMID: 35837027 DOI: 10.3892/etm.2022.11454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 07/28/2021] [Indexed: 11/06/2022] Open
Affiliation(s)
- Li Bai
- Department of Gastroenterology, The 970th Hospital of The PLA Joint Logistics Support Force, Yantai, Shandong 264001, P.R. China
| | - Kunbo Dong
- Department of Gastroenterology, The 970th Hospital of The PLA Joint Logistics Support Force, Yantai, Shandong 264001, P.R. China
| | - Deyong Tong
- Department of Oncology, The 970th Hospital of The PLA Joint Logistics Support Force, Yantai, Shandong 264001, P.R. China
| | - Xiuna Shi
- Department of Gastroenterology, The 970th Hospital of The PLA Joint Logistics Support Force, Yantai, Shandong 264001, P.R. China
| | - Sirong Wei
- Department of Intervention, The 970th Hospital of The PLA Joint Logistics Support Force, Yantai, Shandong 264001, P.R. China
| | - Yongguo Cai
- Department of Gastroenterology, The 970th Hospital of The PLA Joint Logistics Support Force, Yantai, Shandong 264001, P.R. China
| |
Collapse
|
9
|
Zhao X, Xia X, Wang X, Bai M, Zhan D, Shu K. Deep Learning-Based Protein Features Predict Overall Survival and Chemotherapy Benefit in Gastric Cancer. Front Oncol 2022; 12:847706. [PMID: 35651795 PMCID: PMC9148960 DOI: 10.3389/fonc.2022.847706] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 04/05/2022] [Indexed: 12/22/2022] Open
Abstract
Gastric cancer (GC) is one of the most common malignant tumors with a high mortality rate worldwide and lacks effective methods for prognosis prediction. Postoperative adjuvant chemotherapy is the first-line treatment for advanced gastric cancer, but only a subgroup of patients benefits from it. Here, we used 833 formalin-fixed, paraffin-embedded resected tumor samples from patients with TNM stage II/III GC and established a proteomic subtyping workflow using 100 deep-learned features. Two proteomic subtypes (S-I and S-II) with overall survival differences were identified. S-I has a better survival rate and is sensitive to chemotherapy. Patients in the S-I who received adjuvant chemotherapy had a significant improvement in the 5-year overall survival rate compared with patients who received surgery alone (65.3% vs 52.6%; log-rank P = 0.014), but no improvement was observed in the S-II (54% vs 51%; log-rank P = 0.96). These results were verified in an independent validation set. Furthermore, we also evaluated the superiority and scalability of the deep learning-based workflow in cancer molecular subtyping, exhibiting its great utility and potential in prognosis prediction and therapeutic decision-making.
Collapse
Affiliation(s)
- Xuefei Zhao
- Chongqing Key Laboratory of Big Data for Bio Intelligence, School of Bioinformation, Chongqing University of Posts and Telecommunications, Chongqing, China
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Xia Xia
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Xinyue Wang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Mingze Bai
- Chongqing Key Laboratory of Big Data for Bio Intelligence, School of Bioinformation, Chongqing University of Posts and Telecommunications, Chongqing, China
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Dongdong Zhan
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
- Department of Bioinformatics, Beijing Pineal Diagnostics Co., Ltd., Beijing, China
- *Correspondence: Kunxian Shu, ; Dongdong Zhan,
| | - Kunxian Shu
- Chongqing Key Laboratory of Big Data for Bio Intelligence, School of Bioinformation, Chongqing University of Posts and Telecommunications, Chongqing, China
- *Correspondence: Kunxian Shu, ; Dongdong Zhan,
| |
Collapse
|
10
|
Cao T, Lu Y, Wang Q, Qin H, Li H, Guo H, Ge M, Glass SE, Singh B, Zhang W, Dong J, Du F, Qian A, Tian Y, Wang X, Li C, Wu K, Fan D, Nie Y, Coffey RJ, Zhao X. A CGA/EGFR/GATA2 positive feedback circuit confers chemoresistance in gastric cancer. J Clin Invest 2022; 132:154074. [PMID: 35289315 PMCID: PMC8920335 DOI: 10.1172/jci154074] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 01/26/2022] [Indexed: 12/24/2022] Open
Abstract
De novo and acquired resistance are major impediments to the efficacy of conventional and targeted cancer therapy. In unselected gastric cancer (GC) patients with advanced disease, trials combining chemotherapy and an anti-EGFR monoclonal antibody have been largely unsuccessful. In an effort to identify biomarkers of resistance so as to better select patients for such trials, we screened the secretome of chemotherapy-treated human GC cell lines. We found that levels of CGA, the α-subunit of glycoprotein hormones, were markedly increased in the conditioned media of chemoresistant GC cells, and CGA immunoreactivity was enhanced in GC tissues that progressed on chemotherapy. CGA levels in plasma increased in GC patients who received chemotherapy, and this increase was correlated with reduced responsiveness to chemotherapy and poor survival. Mechanistically, secreted CGA was found to bind to EGFR and activate EGFR signaling, thereby conferring a survival advantage to GC cells. N-glycosylation of CGA at Asn52 and Asn78 is required for its stability, secretion, and interaction with EGFR. GATA2 was found to activate CGA transcription, whose increase, in turn, induced the expression and phosphorylation of GATA2 in an EGFR-dependent manner, forming a positive feedback circuit that was initiated by GATA2 autoregulation upon sublethal exposure to chemotherapy. Based on this circuit, combination strategies involving anti-EGFR therapies or targeting CGA with microRNAs (miR-708-3p and miR-761) restored chemotherapy sensitivity. These findings identify a clinically actionable CGA/EGFR/GATA2 circuit and highlight CGA as a predictive biomarker and therapeutic target in chemoresistant GC.
Collapse
Affiliation(s)
- Tianyu Cao
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| | - Yuanyuan Lu
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| | - Qi Wang
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| | - Hongqiang Qin
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Hongwei Li
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| | - Hao Guo
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China.,State Key Laboratory of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Diagnostics Co., Ltd., Nanjing, China
| | - Minghui Ge
- State Key Laboratory of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Diagnostics Co., Ltd., Nanjing, China
| | - Sarah E Glass
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Bhuminder Singh
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Wenyao Zhang
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| | - Jiaqiang Dong
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| | - Feng Du
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| | - Airong Qian
- Key Laboratory for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Ye Tian
- Key Laboratory for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Xin Wang
- Department of Gastroenterology, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Cunxi Li
- Beijing Institute of Human Reproduction and Genetics Medicine, Beijing, China.,Jiaen Genetics Laboratory, Beijing Jiaen Hospital, Beijing, China
| | - Kaichun Wu
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| | - Daiming Fan
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| | - Yongzhan Nie
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| | - Robert J Coffey
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Xiaodi Zhao
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
11
|
Catenacci DV, Chao J, Muro K, Al‐Batran SE, Klempner SJ, Wainberg ZA, Shah MA, Rha SY, Ohtsu A, Liepa AM, Knoderer H, Chatterjee A, Van Cutsem E. Toward a Treatment Sequencing Strategy: A Systematic Review of Treatment Regimens in Advanced Gastric Cancer/Gastroesophageal Junction Adenocarcinoma. Oncologist 2021; 26:e1704-e1729. [PMID: 34288262 PMCID: PMC8488781 DOI: 10.1002/onco.13907] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 07/02/2021] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Platinum and fluoropyrimidine combinations typically comprise first-line (1L) therapy in advanced gastric cancer or gastroesophageal junction adenocarcinoma (G/GEA), although controversy exists regarding the use of 5doublet versus triplet cytotoxic regimens. Historically, second-line (2L) and third-line or later (3L+) therapy has been fragmented. Recent trials have increased the need for optimal treatment sequencing in advanced G/GEA. MATERIALS AND METHODS We conducted a systematic search of peer-reviewed manuscripts of randomized clinical trials examining 1L, 2L, and 3L+ therapy for advanced G/GEA published from 2009 through November 19, 2019. When available, overall survival, progression-free survival, time to progression, overall response rate, and toxicity were extracted from each and compared descriptively. RESULTS In 1L therapy, chemotherapy triplets demonstrated variable efficacy improvements with invariable increased toxicity compared with platinum/fluoropyrimidine doublets. Currently, the only published report of positive outcomes using biologics in 1L describes adding trastuzumab in HER2-overexpressing advanced G/GEA. In 2L, doublet chemotherapy regimens are not uniformly more efficacious than single-agent taxanes or irinotecan, and ramucirumab has demonstrated improved outcomes both as monotherapy and in combination. CONCLUSION For advanced G/GEA, review of trial results from 2009-2019 support 1L therapy with platinum and fluoropyrimidine and sequencing with taxanes or irinotecan in combination with biologics as effective 2L options. Escalating to a triplet may add some efficacy at the expense of added toxicity. IMPLICATIONS FOR PRACTICE The rapidly changing treatment landscape for advanced gastric cancer includes increasing options for refractory disease. With multiple first-line platinum-based regimens, identification of those with the best benefit-to-risk ratio may provide guidance on treatment sequencing strategies. This article presents findings from the published literature of randomized controlled trials that included a first-line platinum/fluoropyrimidine combination and, for second-line trials, patients with platinum/fluoropyrimidine-refractory disease. This guiding summary could be a tool for clinicians to identify the optimal first-line regimen(s) followed by a strategy for subsequent regimens.
Collapse
Affiliation(s)
- Daniel V. Catenacci
- University of Chicago Medical Center & Biological SciencesChicagoIllinoisUSA
| | - Joseph Chao
- City of Hope Comprehensive Cancer CenterDuarteCaliforniaUSA
| | - Kei Muro
- Aichi Cancer Center HospitalNagoyaJapan
| | | | | | | | | | - Sun Young Rha
- Yonsei Cancer Center, Yonsei University College of MedicineSeoulKorea
| | | | | | | | | | - Eric Van Cutsem
- Digestive Oncology, University Hospitals Gasthuisberg Leuven and KU LeuvenLeuvenBelgium
| |
Collapse
|
12
|
Calcium channel blockers lercanidipine and amlodipine inhibit YY1/ERK/TGF-β mediated transcription and sensitize the gastric cancer cells to doxorubicin. Toxicol In Vitro 2021; 74:105152. [DOI: 10.1016/j.tiv.2021.105152] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 02/24/2021] [Accepted: 03/22/2021] [Indexed: 12/24/2022]
|
13
|
Boonyanugomol W, Rukseree K, Prapatpong P, Reamtong O, Baik SC, Jung M, Shin MK, Kang HL, Lee WK. An In Vitro Anti-Cancer Activity of Ocimum tenuiflorum Essential Oil by Inducing Apoptosis in Human Gastric Cancer Cell Line. ACTA ACUST UNITED AC 2021; 57:medicina57080784. [PMID: 34440988 PMCID: PMC8400819 DOI: 10.3390/medicina57080784] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 07/17/2021] [Accepted: 07/27/2021] [Indexed: 11/16/2022]
Abstract
Background and Objectives: The effects of Ocimum tenuiflorum essential oil (OTEO) against gastric cancer remain unknown and merit investigation. Materials and Methods: In the present study, the anti-cancer activity of OTEO was examined in a human gastric cancer cell line (AGS). After OTEO treatment, AGS cell viability was determined by an MTT assay, and inhibition of metastasis was determined by cell migration and invasion assays. The expression of apoptosis-related genes in treated AGS cells was determined by qRT-PCR. Results: OTEO significantly decreased AGS cell viability in a dose-dependent manner (IC50 163.42 µg/mL) and effectively inhibited cell migration and invasion. Morphological examination demonstrated that OTEO induced cell shrinkage, chromatin condensation, and fragmentation, which are considered typical morphologies of apoptotic cell death. Pro-apoptotic genes (TP53, BAX, and BAK) were significantly up-regulated, while anti-apoptotic genes (BCL-2 and BCL-xL) were significantly down-regulated after treatment with OTEO. In addition, significantly increased gene expression was detected for CASP8, CASP9, and CASP3 in AGS cells exposed to OTEO. GC-MS analysis demonstrated that the major compound of OTEO was caryophyllene (25.85%) and α-pinene (11.66%). Conclusions: This in vitro study demonstrates for the first time that OTEO has potential anti-gastric cancer activity and may induce apoptosis in AGS cells through extrinsic and intrinsic pathways.
Collapse
Affiliation(s)
- Wongwarut Boonyanugomol
- Department of Sciences and Liberal Arts, Amnatcharoen Campus, Mahidol University, Amnatcharoen 37000, Thailand;
- Correspondence:
| | - Kamolchanok Rukseree
- Department of Sciences and Liberal Arts, Amnatcharoen Campus, Mahidol University, Amnatcharoen 37000, Thailand;
| | - Pornpan Prapatpong
- Department of Public Health, Amnatcharoen Campus, Mahidol University, Amnatcharoen 37000, Thailand;
| | - Onrapak Reamtong
- Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand;
| | - Seung-Chul Baik
- Department of Microbiology, Gyeongsang Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Korea; (S.-C.B.); (M.J.); (M.-K.S.); (H.-L.K.); (W.-K.L.)
| | - Myunghwan Jung
- Department of Microbiology, Gyeongsang Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Korea; (S.-C.B.); (M.J.); (M.-K.S.); (H.-L.K.); (W.-K.L.)
| | - Min-Kyoung Shin
- Department of Microbiology, Gyeongsang Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Korea; (S.-C.B.); (M.J.); (M.-K.S.); (H.-L.K.); (W.-K.L.)
| | - Hyung-Lyun Kang
- Department of Microbiology, Gyeongsang Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Korea; (S.-C.B.); (M.J.); (M.-K.S.); (H.-L.K.); (W.-K.L.)
| | - Woo-Kon Lee
- Department of Microbiology, Gyeongsang Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Korea; (S.-C.B.); (M.J.); (M.-K.S.); (H.-L.K.); (W.-K.L.)
| |
Collapse
|
14
|
Bhosale PB, Vetrivel P, Ha SE, Kim HH, Heo JD, Won CK, Kim SM, Kim GS. Iridin Induces G2/M Phase Cell Cycle Arrest and Extrinsic Apoptotic Cell Death through PI3K/AKT Signaling Pathway in AGS Gastric Cancer Cells. Molecules 2021; 26:2802. [PMID: 34068568 PMCID: PMC8126061 DOI: 10.3390/molecules26092802] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 05/06/2021] [Accepted: 05/07/2021] [Indexed: 01/03/2023] Open
Abstract
Iridin is a natural flavonoid found in Belamcanda chinensis documented for its broad spectrum of biological activities like antioxidant, antitumor, and antiproliferative effects. In the present study, we have investigated the antitumor potential of iridin in AGS gastric cancer cells. Iridin treatment decreases AGS cell growth and promotes G2/M phase cell cycle arrest by attenuating the expression of Cdc25C, CDK1, and Cyclin B1 proteins. Iridin-treatment also triggered apoptotic cell death in AGS cells, which was verified by cleaved Caspase-3 (Cl- Caspase-3) and poly ADP-ribose polymerase (PARP) protein expression. Further apoptotic cell death was confirmed by increased apoptotic cell death fraction shown in allophycocyanin (APC)/Annexin V and propidium iodide staining. Iridin also increased the expression of extrinsic apoptotic pathway proteins like Fas, FasL, and cleaved Caspase-8 in AGS cells. On the contrary, iridin-treated AGS cells did not show variations in proteins related to an intrinsic apoptotic pathway such as Bax and Bcl-xL. Besides, Iridin showed inhibition of PI3K/AKT signaling pathways by downregulation of (p-PI3K, p-AKT) proteins in AGS cells. In conclusion, these data suggest that iridin has anticancer potential by inhibiting PI3K/AKT pathway. It could be a basis for further drug design in gastric cancer treatment.
Collapse
Affiliation(s)
- Pritam-Bhagwan Bhosale
- Research Institute of Life Science and College of Veterinary Medicine, Gyeongsang National University, Gazwa, Jinju 52828, Korea; (P.-B.B.); (P.V.); (S.-E.H.); (H.-H.K.); (C.-K.W.)
| | - Preethi Vetrivel
- Research Institute of Life Science and College of Veterinary Medicine, Gyeongsang National University, Gazwa, Jinju 52828, Korea; (P.-B.B.); (P.V.); (S.-E.H.); (H.-H.K.); (C.-K.W.)
| | - Sang-Eun Ha
- Research Institute of Life Science and College of Veterinary Medicine, Gyeongsang National University, Gazwa, Jinju 52828, Korea; (P.-B.B.); (P.V.); (S.-E.H.); (H.-H.K.); (C.-K.W.)
| | - Hun-Hwan Kim
- Research Institute of Life Science and College of Veterinary Medicine, Gyeongsang National University, Gazwa, Jinju 52828, Korea; (P.-B.B.); (P.V.); (S.-E.H.); (H.-H.K.); (C.-K.W.)
| | - Jeong-Doo Heo
- Biological Resources Research Group, Bioenvironmental Science & Toxicology Division, Korea Institute of Toxicology (KIT), 17 Jeigok-gil, Jinju 52834, Korea;
| | - Chung-Kil Won
- Research Institute of Life Science and College of Veterinary Medicine, Gyeongsang National University, Gazwa, Jinju 52828, Korea; (P.-B.B.); (P.V.); (S.-E.H.); (H.-H.K.); (C.-K.W.)
| | - Seong-Min Kim
- Research Institute of Life Science and College of Veterinary Medicine, Gyeongsang National University, Gazwa, Jinju 52828, Korea; (P.-B.B.); (P.V.); (S.-E.H.); (H.-H.K.); (C.-K.W.)
| | - Gon-Sup Kim
- Research Institute of Life Science and College of Veterinary Medicine, Gyeongsang National University, Gazwa, Jinju 52828, Korea; (P.-B.B.); (P.V.); (S.-E.H.); (H.-H.K.); (C.-K.W.)
| |
Collapse
|
15
|
Mezynski MJ, Farrelly AM, Cremona M, Carr A, Morgan C, Workman J, Armstrong P, McAuley J, Madden S, Fay J, Sheehan KM, Kay EW, Holohan C, Elamin Y, Rafee S, Morris PG, Breathnach O, Grogan L, Hennessy BT, Toomey S. Targeting the PI3K and MAPK pathways to improve response to HER2-targeted therapies in HER2-positive gastric cancer. J Transl Med 2021; 19:184. [PMID: 33933113 PMCID: PMC8088633 DOI: 10.1186/s12967-021-02842-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 04/18/2021] [Indexed: 12/24/2022] Open
Abstract
Background Aberrant PI3K signalling is implicated in trastuzumab resistance in HER2-positive gastric cancer (GC). The role of PI3K or MEK inhibitors in sensitising HER2-positive GCs to trastuzumab or in overcoming trastuzumab resistance is unclear. Methods Using mass spectrometry-based genotyping we analysed 105 hotspot, non-synonymous somatic mutations in PIK3CA and ERBB-family (EGFR, ERBB2, ERBB3 and ERBB4) genes in gastric tumour samples from 69 patients. A panel of gastric cell lines (N87, OE19, ESO26, SNU16, KATOIII) were profiled for anti-proliferative response to the PI3K inhibitor copanlisib and the MEK1/2 inhibitor refametinib alone and in combination with anti-HER2 therapies. Results Patients with HER2-positive GC had significantly poorer overall survival compared to HER2-negative patients (15.9 months vs. 35.7 months). Mutations in PIK3CA were only identified in HER2-negative tumours, while ERBB-family mutations were identified in HER2-positive and HER2-negative tumours. Copanlisib had anti-proliferative effects in 4/5 cell lines, with IC50s ranging from 23.4 (N87) to 93.8 nM (SNU16). All HER2-positive cell lines except SNU16 were sensitive to lapatinib (IC50s 0.04 µM–1.5 µM). OE19 cells were resistant to trastuzumab. The combination of lapatinib and copanlisib was synergistic in ESO-26 and OE-19 cells (ED50: 0.83 ± 0.19 and 0.88 ± 0.13, respectively) and additive in NCI-N87 cells (ED50:1.01 ± 0.55). The combination of copanlisib and trastuzumab significantly improved growth inhibition compared to either therapy alone in NCI-N87, ESO26 and OE19 cells (p < 0.05). Conclusions PI3K or MEK inhibition alone or in combination with anti-HER2 therapy may represent an improved treatment strategy for some patients with HER2-positive GC, and warrants further investigation in a clinical trial setting. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-021-02842-1.
Collapse
Affiliation(s)
- M Janusz Mezynski
- Medical Oncology Group, Department of Molecular Medicine, Royal College of Surgeons in Ireland, RCSI Smurfit Building, Beaumont Hospital, Dublin 9, Ireland
| | - Angela M Farrelly
- Medical Oncology Group, Department of Molecular Medicine, Royal College of Surgeons in Ireland, RCSI Smurfit Building, Beaumont Hospital, Dublin 9, Ireland
| | - Mattia Cremona
- Medical Oncology Group, Department of Molecular Medicine, Royal College of Surgeons in Ireland, RCSI Smurfit Building, Beaumont Hospital, Dublin 9, Ireland
| | - Aoife Carr
- Medical Oncology Group, Department of Molecular Medicine, Royal College of Surgeons in Ireland, RCSI Smurfit Building, Beaumont Hospital, Dublin 9, Ireland
| | - Clare Morgan
- Medical Oncology Group, Department of Molecular Medicine, Royal College of Surgeons in Ireland, RCSI Smurfit Building, Beaumont Hospital, Dublin 9, Ireland
| | - Julie Workman
- Medical Oncology Group, Department of Molecular Medicine, Royal College of Surgeons in Ireland, RCSI Smurfit Building, Beaumont Hospital, Dublin 9, Ireland
| | - Paul Armstrong
- Medical Oncology Group, Department of Molecular Medicine, Royal College of Surgeons in Ireland, RCSI Smurfit Building, Beaumont Hospital, Dublin 9, Ireland
| | - Jennifer McAuley
- Medical Oncology Group, Department of Molecular Medicine, Royal College of Surgeons in Ireland, RCSI Smurfit Building, Beaumont Hospital, Dublin 9, Ireland
| | - Stephen Madden
- Data Science Centre, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Joanna Fay
- Department of Histopathology, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Katherine M Sheehan
- Department of Histopathology, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Elaine W Kay
- Department of Histopathology, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Ciara Holohan
- Department of Medical Oncology, St. James's Hospital, Dublin, Ireland
| | - Yasir Elamin
- Medical Oncology Group, Department of Molecular Medicine, Royal College of Surgeons in Ireland, RCSI Smurfit Building, Beaumont Hospital, Dublin 9, Ireland
| | - Shereen Rafee
- Department of Medical Oncology, St. James's Hospital, Dublin, Ireland
| | - Patrick G Morris
- Department of Medical Oncology, Beaumont Hospital, Dublin, Ireland
| | - Oscar Breathnach
- Department of Medical Oncology, Beaumont Hospital, Dublin, Ireland
| | - Liam Grogan
- Department of Medical Oncology, Beaumont Hospital, Dublin, Ireland
| | - Bryan T Hennessy
- Medical Oncology Group, Department of Molecular Medicine, Royal College of Surgeons in Ireland, RCSI Smurfit Building, Beaumont Hospital, Dublin 9, Ireland.,Department of Medical Oncology, Beaumont Hospital, Dublin, Ireland
| | - Sinead Toomey
- Medical Oncology Group, Department of Molecular Medicine, Royal College of Surgeons in Ireland, RCSI Smurfit Building, Beaumont Hospital, Dublin 9, Ireland.
| |
Collapse
|
16
|
Cheng H, Sharen G, Wang Z, Zhou J. LncRNA UCA1 Enhances Cisplatin Resistance by Regulating CYP1B1-mediated Apoptosis via miR-513a-3p in Human Gastric Cancer. Cancer Manag Res 2021; 13:367-377. [PMID: 33469378 PMCID: PMC7813468 DOI: 10.2147/cmar.s277399] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 11/11/2020] [Indexed: 12/18/2022] Open
Abstract
Background Chemoresistance contributes to treatment failure of gastric cancer (GC) patients but the molecular mechanism of chemoresistance in GC is still unclear. Long-chain noncoding RNA (lncRNA) urothelial cancer associated 1 (UCA1) is associated with resistance to chemotherapy drugs. Methods We detected the expression of UCA1 in 53 pairs of GC tumor tissue and adjacent normal tissue, human normal gastric mucosa cells (GES-1) and human GC cells (HGC-27, SNU-5, AGS, SGC-7901, and NCI-N87) using RT-qPCR. Small RNA interference technology was used to knock down the expression of UCA1 in gastric cancer cells. CCK8 solution was used to detect cell viability. Flow cytometry was used to detect apoptosis, and Western blotting was used to detect protein expression. Results UCA1 was highly expressed in GC tissues and cells, and knockdown of UCA1 increased chemosensitivity to cisplatin by inducing cell apoptosis. Furthermore, UCA1 promoted CYP1B1 expression by binding to miR-513a-3p in human GC cells in vitro, and UCA1/CYP1B1 expression was negatively related to miR-513a-3p expression, while UCA1 expression was positively related to CYP1B1 expression in human GC tissues. Moreover, overexpression of miR-513a-3p or knockdown of CYP1B1 increased chemosensitivity to cisplatin, and knockdown of miR-513a-3p or overexpression of CYP1B1 decreased chemosensitivity to cisplatin by inducing cell apoptosis in human GC cells. Importantly, overexpression of CYP1B1 reduced chemosensitivity to cisplatin which increased by knockdown of UCA1, and knockdown of CYP1B1 increased chemosensitivity to cisplatin which decreased by knockdown of miR-513a-3p in human GC cells. Conclusion The lncRNA UCA1/miR-513a-3p/CYP1B1 axis regulates cisplatin resistance in human GC cells; hence, it is a potential target for treating chemoresistance in GC.
Collapse
Affiliation(s)
- Haidong Cheng
- Department of Gastroenterology, Affiliated Hospital of Inner Mongolia Medical University, Hohhot 010059, People's Republic of China
| | - Gaowa Sharen
- Department of Pathological Anatomy, College of Basic Medicine of Inner Mongolia Medical University, Hohhot 010059, People's Republic of China
| | - Zhaoyang Wang
- Department of Gastroenterology, Affiliated Hospital of Inner Mongolia Medical University, Hohhot 010059, People's Republic of China
| | - Jing Zhou
- Department of Pharmacology, College of Basic Medicine of Inner Mongolia Medical University, Hohhot 010059, People's Republic of China
| |
Collapse
|
17
|
Karamoozian A, Baneshi MR, Bahrampour A. Short-term and long-term survival of patients with gastric cancer. GASTROENTEROLOGY AND HEPATOLOGY FROM BED TO BENCH 2021; 14:115-122. [PMID: 33968338 PMCID: PMC8101525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
AIM The aim of this study was to apply the Bayesian mixture cure rate frailty model to determine the factors that influence short-term and long-term survival of patients with gastric cancer. BACKGROUND Determining the risk factors of gastric cancer is currently considered very important, because the disease has become one of the most dangerous types of mortal cancers. Therefore, it is possible to determine the effective risk factors of short-term and long-term survival in patients through utilizing this model. METHODS The present retrospective study was conducted on 339 gastric cancer patients whose data was recorded in hospitals of Kerman province, Iran, during 2001-2015. In the study, the Bayesian mixture cure rate frailty model was used to determine the effective factors of short-term and long-term survival in patients. RESULTS In the present study, the event of interest occurred for 57.5% of patients. Over time, the survival rate of cancer patients reached its lowest point, approximately 0.3 at the end of study. According to the results of the present study, variables of chemotherapy (β=-0.35 (-0.75, -0.03) and OR=1.59 (1.08, 2.19)), morphology (β =-0.98(-1.45, -0.48) and OR=2.99 (1.78, 4.17)), and metastasis (β =0.42(0.10, 0.93) and OR=0.39(0.01, 0.84)) were identified as effective factors in short-term and long-term survival of patients. CONCLUSION The effective factors of long-term and short-term survival can be identified by utilizing the Bayesian mixture cure rate frailty model, while it is impossible through conventional models of survival analysis. Chemotherapy, morphology, and metastasis are the most important effective factors of short-term and long-term survival in patients with gastric cancer.
Collapse
Affiliation(s)
- Ali Karamoozian
- Department of Biostatistics and Epidemiology, Kerman University of Medical Sciences, Kerman, Islamic Republic of Iran,Modeling in Health Research Center, Institute for Futures Studies in Health, Kerman University of Medical Sciences, Kerman, Islamic Republic of Iran
| | - Mohammad Reza Baneshi
- Department of Biostatistics and Epidemiology, Kerman University of Medical Sciences, Kerman, Islamic Republic of Iran,Modeling in Health Research Center, Institute for Futures Studies in Health, Kerman University of Medical Sciences, Kerman, Islamic Republic of Iran
| | - Abbas Bahrampour
- Department of Biostatistics and Epidemiology, Kerman University of Medical Sciences, Kerman, Islamic Republic of Iran,Modeling in Health Research Center, Institute for Futures Studies in Health, Kerman University of Medical Sciences, Kerman, Islamic Republic of Iran
| |
Collapse
|
18
|
Hu G, Zhong K, Wang S, Wang S, Ding Q, Xu F, Chen W, Cheng P, Huang L. Cellular immunotherapy plus chemotherapy ameliorates survival in gastric cancer patients: a meta-analysis. Int J Clin Oncol 2020; 25:1747-1756. [PMID: 32728865 DOI: 10.1007/s10147-020-01750-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 07/03/2020] [Indexed: 01/11/2023]
Abstract
The efficacy of cellular immunotherapy plus chemotherapy in treatment of gastric cancer (GC) remains inconsistent even controversial. Hence, we performed a meta-analysis to better comprehend the clinical value of cellular immunotherapy plus chemotherapy for GC patients. We searched PubMed, Embase and EBSCO databases to identify the studies evaluating the association of cellular immunotherapy plus chemotherapy and overall survival (OS) and/or disease-free survival (DFS) in patients with GC, and then combined relevant data into hazard ratios (HRs) for OS, DFS and clinicopathological features such as TNM stage, etc. with STATA 12.0. Eleven studies with 1244 patients were included in this meta-analysis. We found that cellular immunotherapy plus chemotherapy remarkably improved overall survival (OS) and diseases-free survival (DFS) as compared to the chemotherapy for GC patients. In subgroup analyses, pooled data showed that the combined therapy was significantly associated with better 3-year and 5-year survival rate, but not with 1-year survival rate of patients; the application of cellular immunotherapy based on either CIK or DC-CIK cells could enhance survival as well as NK, γδT and CIK cells-based immunotherapy. More importantly, the addition of cellular immunotherapy considerably improved OS and DFS only in patients with stage III rather than stage II. In addition, we also discovered that the combined therapy did not cause intolerable side effects to patients. Cellular immunotherapy plus chemotherapy ameliorates survival in GC, especially in patients with stage III, implicating that it is a valuable therapeutic strategy for these patients.
Collapse
Affiliation(s)
- Guoming Hu
- Department of General Surgery (Breast and Thyroid Surgery), Shaoxing People's Hospital, Shaoxing Hospital, Zhejiang University School of Medicine, 568 Zhongxing Road, Shaoxing, 312000, Zhejiang, China.
| | - Kefang Zhong
- Department of General Surgery (Breast and Thyroid Surgery), Shaoxing People's Hospital, Shaoxing Hospital, Zhejiang University School of Medicine, 568 Zhongxing Road, Shaoxing, 312000, Zhejiang, China
| | - Songxiang Wang
- Department of General Surgery (Breast and Thyroid Surgery), Shaoxing People's Hospital, Shaoxing Hospital, Zhejiang University School of Medicine, 568 Zhongxing Road, Shaoxing, 312000, Zhejiang, China
| | - Shimin Wang
- Department of Nephrology, Shaoxing People's Hospital, Shaoxing Hospital, Zhejiang University School of Medicine, 568 Zhongxing Road, Shaoxing, 312000, Zhejiang, China
| | - Qiannan Ding
- Medical Research Center, Shaoxing People's Hospital, Shaoxing Hospital, Zhejiang University School of Medicine, 568 Zhongxing Road, Shaoxing, 312000, Zhejiang, China
| | - Feng Xu
- Department of General Surgery (Breast and Thyroid Surgery), Shaoxing People's Hospital, Shaoxing Hospital, Zhejiang University School of Medicine, 568 Zhongxing Road, Shaoxing, 312000, Zhejiang, China
| | - Wei Chen
- Department of General Surgery (Breast and Thyroid Surgery), Shaoxing People's Hospital, Shaoxing Hospital, Zhejiang University School of Medicine, 568 Zhongxing Road, Shaoxing, 312000, Zhejiang, China
| | - Pu Cheng
- Department of Gynecology, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China.
| | - Liming Huang
- Department of General Surgery (Breast and Thyroid Surgery), Shaoxing People's Hospital, Shaoxing Hospital, Zhejiang University School of Medicine, 568 Zhongxing Road, Shaoxing, 312000, Zhejiang, China.
| |
Collapse
|
19
|
Fernandes E, Freitas R, Ferreira D, Soares J, Azevedo R, Gaiteiro C, Peixoto A, Oliveira S, Cotton S, Relvas-Santos M, Afonso LP, Palmeira C, Oliveira MJ, Ferreira R, Silva AMN, Lara Santos L, Ferreira JA. Nucleolin-Sle A Glycoforms as E-Selectin Ligands and Potentially Targetable Biomarkers at the Cell Surface of Gastric Cancer Cells. Cancers (Basel) 2020; 12:cancers12040861. [PMID: 32252346 PMCID: PMC7226152 DOI: 10.3390/cancers12040861] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 03/24/2020] [Accepted: 03/28/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Gastric cancer (GC) is a major health burden worldwide, with half of patients developing metastases within 5 years after treatment, urging novel biomarkers for diagnosis and efficient therapeutic targeting. Sialyl-Lewis A (SLeA), a terminal glycoepitope of glycoproteins and glycolipids, offers tremendous potential towards this objective. It is rarely expressed in healthy tissues and blood cells, while it is present in highly metastatic cell lines and metastases. SLeA is also involved in E-selectin mediated metastasis, making it an ideal target to control disease dissemination. METHODS AND RESULTS To improve cancer specificity, we have explored the SLeA-glycoproteome of six GC cell models, with emphasis on glycoproteins showing affinity for E-selectin. A novel bioinformatics-assisted algorithm identified nucleolin (NCL), a nuclear protein, as a potential targetable biomarker potentially involved in metastasis. Several immunoassays, including Western blot and in situ proximity ligation reinforced the existence of cell surface NCL-SLeA glycoforms in GC. The NCL-SLeA glycophenotype was associated with decreased survival and was not reflected in relevant healthy tissues. CONCLUSIONS NCL-SLeA is a biomarker of poor prognosis in GC holding potential for precise cancer targeting. This is the first report describing SLeA in preferentially nuclear protein, setting a new paradigm for cancer biomarkers discovery and targeted therapies.
Collapse
Affiliation(s)
- Elisabete Fernandes
- Experimental Pathology and Therapeutics Group, Portuguese Institute of Oncology, 4200-162 Porto, Portugal; (E.F.); (R.F.); (D.F.); (J.S.); (R.A.); (C.G.); (A.P.); (S.O.); (S.C.); (M.R.-S.); (L.P.A.); (C.P.); (L.L.S.)
- Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, 4050-013 Porto, Portugal
- Institute for Research and Innovation in Health (i3S), University of Porto, 4200-135 Porto, Portugal;
- Institute for Biomedical Engineering (INEB), Porto, Portugal, 4200-135 Porto, Portugal
- Digestive Cancer Research Group, 1495-161 Algés, Portugal
| | - Rui Freitas
- Experimental Pathology and Therapeutics Group, Portuguese Institute of Oncology, 4200-162 Porto, Portugal; (E.F.); (R.F.); (D.F.); (J.S.); (R.A.); (C.G.); (A.P.); (S.O.); (S.C.); (M.R.-S.); (L.P.A.); (C.P.); (L.L.S.)
| | - Dylan Ferreira
- Experimental Pathology and Therapeutics Group, Portuguese Institute of Oncology, 4200-162 Porto, Portugal; (E.F.); (R.F.); (D.F.); (J.S.); (R.A.); (C.G.); (A.P.); (S.O.); (S.C.); (M.R.-S.); (L.P.A.); (C.P.); (L.L.S.)
- Institute for Research and Innovation in Health (i3S), University of Porto, 4200-135 Porto, Portugal;
- Institute for Biomedical Engineering (INEB), Porto, Portugal, 4200-135 Porto, Portugal
- Digestive Cancer Research Group, 1495-161 Algés, Portugal
| | - Janine Soares
- Experimental Pathology and Therapeutics Group, Portuguese Institute of Oncology, 4200-162 Porto, Portugal; (E.F.); (R.F.); (D.F.); (J.S.); (R.A.); (C.G.); (A.P.); (S.O.); (S.C.); (M.R.-S.); (L.P.A.); (C.P.); (L.L.S.)
- Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, 4050-013 Porto, Portugal
- REQUIMTE-LAQV, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal;
| | - Rita Azevedo
- Experimental Pathology and Therapeutics Group, Portuguese Institute of Oncology, 4200-162 Porto, Portugal; (E.F.); (R.F.); (D.F.); (J.S.); (R.A.); (C.G.); (A.P.); (S.O.); (S.C.); (M.R.-S.); (L.P.A.); (C.P.); (L.L.S.)
| | - Cristiana Gaiteiro
- Experimental Pathology and Therapeutics Group, Portuguese Institute of Oncology, 4200-162 Porto, Portugal; (E.F.); (R.F.); (D.F.); (J.S.); (R.A.); (C.G.); (A.P.); (S.O.); (S.C.); (M.R.-S.); (L.P.A.); (C.P.); (L.L.S.)
- Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, 4050-013 Porto, Portugal
| | - Andreia Peixoto
- Experimental Pathology and Therapeutics Group, Portuguese Institute of Oncology, 4200-162 Porto, Portugal; (E.F.); (R.F.); (D.F.); (J.S.); (R.A.); (C.G.); (A.P.); (S.O.); (S.C.); (M.R.-S.); (L.P.A.); (C.P.); (L.L.S.)
- Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, 4050-013 Porto, Portugal
- Institute for Research and Innovation in Health (i3S), University of Porto, 4200-135 Porto, Portugal;
- Institute for Biomedical Engineering (INEB), Porto, Portugal, 4200-135 Porto, Portugal
| | - Sara Oliveira
- Experimental Pathology and Therapeutics Group, Portuguese Institute of Oncology, 4200-162 Porto, Portugal; (E.F.); (R.F.); (D.F.); (J.S.); (R.A.); (C.G.); (A.P.); (S.O.); (S.C.); (M.R.-S.); (L.P.A.); (C.P.); (L.L.S.)
| | - Sofia Cotton
- Experimental Pathology and Therapeutics Group, Portuguese Institute of Oncology, 4200-162 Porto, Portugal; (E.F.); (R.F.); (D.F.); (J.S.); (R.A.); (C.G.); (A.P.); (S.O.); (S.C.); (M.R.-S.); (L.P.A.); (C.P.); (L.L.S.)
- Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, 4050-013 Porto, Portugal
| | - Marta Relvas-Santos
- Experimental Pathology and Therapeutics Group, Portuguese Institute of Oncology, 4200-162 Porto, Portugal; (E.F.); (R.F.); (D.F.); (J.S.); (R.A.); (C.G.); (A.P.); (S.O.); (S.C.); (M.R.-S.); (L.P.A.); (C.P.); (L.L.S.)
- Institute for Biomedical Engineering (INEB), Porto, Portugal, 4200-135 Porto, Portugal
- REQUIMTE-LAQV, Department of Chemistry and Biochemistry, Faculty of Sciences of the University of Porto, 4169-007 Porto, Portugal;
| | - Luis Pedro Afonso
- Experimental Pathology and Therapeutics Group, Portuguese Institute of Oncology, 4200-162 Porto, Portugal; (E.F.); (R.F.); (D.F.); (J.S.); (R.A.); (C.G.); (A.P.); (S.O.); (S.C.); (M.R.-S.); (L.P.A.); (C.P.); (L.L.S.)
- Pathology Department, Portuguese Institute of Oncology of Porto, 4200-162 Porto, Portugal
| | - Carlos Palmeira
- Experimental Pathology and Therapeutics Group, Portuguese Institute of Oncology, 4200-162 Porto, Portugal; (E.F.); (R.F.); (D.F.); (J.S.); (R.A.); (C.G.); (A.P.); (S.O.); (S.C.); (M.R.-S.); (L.P.A.); (C.P.); (L.L.S.)
- Immunology Department, Portuguese Institute of Oncology of Porto, 4200-162 Porto, Portugal
- Health Science Faculty, University of Fernando Pessoa, 4249-004 Porto, Portugal
| | - Maria José Oliveira
- Institute for Research and Innovation in Health (i3S), University of Porto, 4200-135 Porto, Portugal;
- Institute for Biomedical Engineering (INEB), Porto, Portugal, 4200-135 Porto, Portugal
| | - Rita Ferreira
- REQUIMTE-LAQV, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal;
| | - André M. N. Silva
- REQUIMTE-LAQV, Department of Chemistry and Biochemistry, Faculty of Sciences of the University of Porto, 4169-007 Porto, Portugal;
| | - Lúcio Lara Santos
- Experimental Pathology and Therapeutics Group, Portuguese Institute of Oncology, 4200-162 Porto, Portugal; (E.F.); (R.F.); (D.F.); (J.S.); (R.A.); (C.G.); (A.P.); (S.O.); (S.C.); (M.R.-S.); (L.P.A.); (C.P.); (L.L.S.)
- Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, 4050-013 Porto, Portugal
- Digestive Cancer Research Group, 1495-161 Algés, Portugal
- Health Science Faculty, University of Fernando Pessoa, 4249-004 Porto, Portugal
- Department of Surgical Oncology, Portuguese Institute of Oncology of Porto, 4200-162 Porto, Portugal
- Department, Porto Comprehensive Cancer Centre (P.ccc), 4200-162 Porto, Portugal
| | - José Alexandre Ferreira
- Experimental Pathology and Therapeutics Group, Portuguese Institute of Oncology, 4200-162 Porto, Portugal; (E.F.); (R.F.); (D.F.); (J.S.); (R.A.); (C.G.); (A.P.); (S.O.); (S.C.); (M.R.-S.); (L.P.A.); (C.P.); (L.L.S.)
- Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, 4050-013 Porto, Portugal
- Department, Porto Comprehensive Cancer Centre (P.ccc), 4200-162 Porto, Portugal
- Correspondence: ; Tel.: +351-225084000 (ext. 5111)
| |
Collapse
|
20
|
Yeh SP, Wang HT. Successful treatment with continuous high-dose 5-fluorouracil infusion, followed by oral capecitabine in a patient with advanced gastric cancer with bone marrow metastasis and microangiopathic hemolytic anemia. JOURNAL OF CANCER RESEARCH AND PRACTICE 2020. [DOI: 10.4103/jcrp.jcrp_20_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
21
|
Kim SH, Lee HJ, Park JH, Choi JH, Park SH, Choe HN, Oh SY, Suh YS, Kong SH, Park DJ, Yang HK. Proposal of a New TNM Classification for Gastric Cancer: Focusing on pN3b and Cytology-Positive (CY1) Disease. J Gastric Cancer 2019; 19:329-343. [PMID: 31598375 PMCID: PMC6769367 DOI: 10.5230/jgc.2019.19.e33] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 09/06/2019] [Accepted: 09/06/2019] [Indexed: 12/15/2022] Open
Abstract
Purpose Gastric cancer with lymph node metastasis (LNM) more than 15 (N3b) was defined as stage IV until the 6th AJCC system. However, it has been reclassified as a localized disease (stage IIb or III) since the 7th system. The aim of this study is to demonstrate that the survival of N3b is comparable to cytology-only positive (CY1-only) stage IV and to propose a new TNM system interpreting N3b as an eligibility criterion for receiving more intensive chemotherapy regimens. Materials and Methods 1,430 patients who underwent gastric cancer surgery at Seoul National University Hospital from 2007 to 2012 were retrospectively analyzed. The 5-year survival rate (5YSR) and 3-year recurrence-free survival (RFS) were evaluated according to the 7th and 8th systems, as well as a new categorization based on N-classification; N0-2 (LNM<7), N3a (LNM 7–15), or N3b (LNM>15). Results The survival of N3b is comparable to that of CY1-only stage IV (log rank test, P=0.671) and is distinct from that of grossly stage IV (log rank test, P<0.001). The survival of the remaining stage IIIc (T4bN3a) was comparable to those of N3b and CY1-only stage IV. Most N3b patients had significantly shorter 3-year RFS and mean RFS than those with IIb–IIIc, as if N3b itself was a higher TNM stage. Conclusions In terms of survival, T4bN3a, N3b, and CY1-only stage IV were unified as stage IVa, while grossly stage IV was defined as stage IVb. N3b can be regarded as an eligibility criterion for undergoing more intensive chemotherapy regimens.
Collapse
Affiliation(s)
- Sa-Hong Kim
- Department of Surgery, Seoul National University Hospital, Seoul, Korea
| | - Hyuk-Joon Lee
- Department of Surgery, Seoul National University Hospital, Seoul, Korea.,Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Ji-Hyeon Park
- Department of Surgery, Seoul National University Hospital, Seoul, Korea
| | - Jong-Ho Choi
- Department of Surgery, Seoul National University Hospital, Seoul, Korea
| | - Shin-Hoo Park
- Department of Surgery, Seoul National University Hospital, Seoul, Korea
| | - Hwi-Nyeong Choe
- Department of Surgery, Seoul National University Hospital, Seoul, Korea
| | - Seung-Young Oh
- Department of Surgery, Seoul National University Hospital, Seoul, Korea
| | - Yun-Suhk Suh
- Department of Surgery, Seoul National University Hospital, Seoul, Korea
| | - Seong-Ho Kong
- Department of Surgery, Seoul National University Hospital, Seoul, Korea
| | - Do-Joong Park
- Department of Surgery, Seoul National University Hospital, Seoul, Korea
| | - Han-Kwang Yang
- Department of Surgery, Seoul National University Hospital, Seoul, Korea.,Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
22
|
Fernandes E, Ferreira D, Peixoto A, Freitas R, Relvas-Santos M, Palmeira C, Martins G, Barros A, Santos LL, Sarmento B, Ferreira JA. Glycoengineered nanoparticles enhance the delivery of 5-fluoroucil and paclitaxel to gastric cancer cells of high metastatic potential. Int J Pharm 2019; 570:118646. [PMID: 31465836 DOI: 10.1016/j.ijpharm.2019.118646] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 07/26/2019] [Accepted: 08/24/2019] [Indexed: 01/08/2023]
Abstract
Gastric cancer is the third leading cause of cancer-related death worldwide, with half of patients developing metastasis within 5 years after curative treatment. Moreover, many patients cannot tolerate or complete systemic treatment due severe side-effects, reducing their effectiveness. Thus, targeted therapeutics are warranted to improve treatment outcomes and reduce toxicity. Herein, poly(lactic-co-glycolic acid) (PLGA) nanoparticles loaded with 5-fluorouracil (5-FU) and paclitaxel were surface-functionalized with a monoclonal antibody targeting sialyl-Lewis A (sLeA), a known glycan mediating hematogenous metastasis. Nanoparticles, ranging from 137 to 330 nm, enabled the controlled release of cytotoxic drugs at neutral and acid pH, supporting potential for intravenous and oral administration. Nanoencapsulation also reduced the initial toxicity of the drugs against gastric cells, suggesting it may constitute a safer administration vehicle. Furthermore, nanoparticle functionalization significantly enhanced targeting to sLeA cells in vitro and ex vivo (over 40% in comparison to non-targeted nanoparticles). In summary, a glycoengineered nano-vehicle was successfully developed to deliver 5-FU and paclitaxel therapeutic agents to metastatic gastric cancer cells. We anticipate that this may constitute an important milestone to establish improved targeted therapeutics against gastric cancer. Given the pancarcinomic nature of the sLeA antigen, the translation of this solution to other models may be also envisaged.
Collapse
Affiliation(s)
- Elisabete Fernandes
- Experimental Pathology and Therapeutics Group, Portuguese Institute of Oncology, 4200-162 Porto, Portugal; Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, 4050-013 Porto, Portugal; Institute for Research and Innovation in Health (i3s), University of Porto, 4200-135 Porto, Portugal; Institute for Biomedical Engineering (INEB), 4200-135 Porto, Portugal; Digestive Cancer Research Group, 1495-161 Algés, Portugal
| | - Dylan Ferreira
- Experimental Pathology and Therapeutics Group, Portuguese Institute of Oncology, 4200-162 Porto, Portugal; Digestive Cancer Research Group, 1495-161 Algés, Portugal
| | - Andreia Peixoto
- Experimental Pathology and Therapeutics Group, Portuguese Institute of Oncology, 4200-162 Porto, Portugal; Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, 4050-013 Porto, Portugal; Institute for Research and Innovation in Health (i3s), University of Porto, 4200-135 Porto, Portugal; Institute for Biomedical Engineering (INEB), 4200-135 Porto, Portugal
| | - Rui Freitas
- Experimental Pathology and Therapeutics Group, Portuguese Institute of Oncology, 4200-162 Porto, Portugal; QOPNA, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Marta Relvas-Santos
- Experimental Pathology and Therapeutics Group, Portuguese Institute of Oncology, 4200-162 Porto, Portugal; Digestive Cancer Research Group, 1495-161 Algés, Portugal
| | - Carlos Palmeira
- Experimental Pathology and Therapeutics Group, Portuguese Institute of Oncology, 4200-162 Porto, Portugal; Immunology Department, Portuguese Institute of Oncology of Porto, 4200-162 Porto, Portugal; Health Science Faculty, University of Fernando Pessoa, 4249-004 Porto, Portugal
| | - Gabriela Martins
- Immunology Department, Portuguese Institute of Oncology of Porto, 4200-162 Porto, Portugal
| | - Anabela Barros
- Digestive Cancer Research Group, 1495-161 Algés, Portugal; Department of Medical Oncology, Coimbra Hospital and University Centre, 3075-075 Coimbra, Portugal
| | - Lúcio Lara Santos
- Experimental Pathology and Therapeutics Group, Portuguese Institute of Oncology, 4200-162 Porto, Portugal; Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, 4050-013 Porto, Portugal; Digestive Cancer Research Group, 1495-161 Algés, Portugal; Department of Surgical Oncology, Portuguese Institute of Oncology of Porto, 4200-162 Porto, Portugal; Porto Comprehensive Cancer Centre (P.ccc), 4200-162 Porto, Portugal
| | - Bruno Sarmento
- Institute for Research and Innovation in Health (i3s), University of Porto, 4200-135 Porto, Portugal; Institute for Biomedical Engineering (INEB), 4200-135 Porto, Portugal; Institute of Research and Advanced Training in Health Sciences and Technologies (CESPU), 4585-116 Gandra PRD, Portugal
| | - José Alexandre Ferreira
- Experimental Pathology and Therapeutics Group, Portuguese Institute of Oncology, 4200-162 Porto, Portugal; Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, 4050-013 Porto, Portugal; Porto Comprehensive Cancer Centre (P.ccc), 4200-162 Porto, Portugal.
| |
Collapse
|
23
|
Hosseini FS, Noroozi Karimabad M, Hajizadeh MR, Khoshdel A, Khanamani Falahati-Pour S, Mirzaei MR, Mirmohamadi SM, Mahmoodi M. Evaluating of Induction of Apoptosis by Cornus mass L. Extract in the Gastric Carcinoma Cell Line (AGS). Asian Pac J Cancer Prev 2019; 20:123-130. [PMID: 30678391 PMCID: PMC6485578 DOI: 10.31557/apjcp.2019.20.1.123] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 12/06/2018] [Indexed: 11/25/2022] Open
Abstract
Aim and objectives: Natural products and derivatives of medicinal vegetation can play an important role to the cure tumor. The Present study was focused to determine the effect of Cornus mass L. extract on the induction of apoptosis in AGS gastric carcinoma cell line in compared to L929 cells. Methods: In this experimental study, AGS and L929 cells were cultured and treated with different concentrations (0–10 mg/ml) of Cornus mass L. extract for 48 and 72 hours. Cell proliferation was assessed by MTT assay. The optical density of the colored solution was quantified at 570 nm wavelengths by an ELISA Reader. Making use of the apoptosis detection kit of Annexin V-FITC, PI and double staining with Annexin V-FITC were carried out for flow cytometry investigations. Data were analyzed by ANOVA. Variations with a P-value less than 0.05 were considered significant. Results: shows a noticeable deviation among various concentrations of extract when cells were treated for 48, 72 h declined cell viability in AGS cell line in comparison L929 cell lines in a dose and time-dependent manner (P < 0.05). This extract also displayed approximately several-fold increased anti-cancer potency in AGS compared to L929 cells. The IC50 value in AGS cells (evaluated after 48,72h) of the extract against AGS cells was 5/44, 2/44 mg/ml (p≤0.05). The analysis results of flow cytometry indicated that apoptosis was induced by the extract in AGS cells treated, compared with L929 cells. Conclusion: Each of our results implicates the reality that Cornus mass L. extract acts as a novel, potent inhibitor of cancer proliferation in in vitro. This may result in developing a promising therapeutic agent for the treatment of indole-sensitive cancers.
Collapse
Affiliation(s)
- Farzaneh Sadat Hosseini
- Molecular Medicine Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
- Department of Clinical Biochemistry, Faculty of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran.
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Small benzothiazole molecule induces apoptosis and prevents metastasis through DNA interaction and c-MYC gene supression in diffuse-type gastric adenocarcinoma cell line. Chem Biol Interact 2018; 294:118-127. [DOI: 10.1016/j.cbi.2018.08.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2018] [Revised: 07/30/2018] [Accepted: 08/10/2018] [Indexed: 12/18/2022]
|
25
|
Wang X, Tang S, Cui X, Yang J, Geng C, Chen C, Zhou N, Li Y. Cytokine-induced killer cell/dendritic cell-cytokine-induced killer cell immunotherapy for the postoperative treatment of gastric cancer: A systematic review and meta-analysis. Medicine (Baltimore) 2018; 97:e12230. [PMID: 30200148 PMCID: PMC6133452 DOI: 10.1097/md.0000000000012230] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Immunotherapy is emerging as a new treatment strategy for gastric cancer(GC). However, the efficacy and safety of this technique remain unclear. This meta-analysis aimed to assess the effect of cytokine-induced killer cell (CIK)/dendritic cell-cytokine-induced killer cell (DC-CIK) treatment for GC after surgery. METHODS Hazard ratio (HR), overall survival (OS) rates, and disease-free survival (DFS) rates were calculated using a Mantel-Haenszel (M-H) fixed-effects model (FEM), and results were displayed using forest plots. Publication bias was assessed by Begg test, and data were presented using funnel plots. Date robustness was assessed by the trim and fill method. Descriptive analysis was performed on T lymphocytes and adverse effects. RESULTS In total, 9 trials, including 1216 patients, were eligible for inclusion in this meta-analysis. Compared with the control group, the HR for OS was 0.712 (95% confidence interval [CI] 0.594-0.854) and 0.66 (95% CI 0.546-0.797) for overall (DFS). The risk ratio (RR) of the 3 and 5-year OS rate was 1.29 (95% CI 1.15-1.46) and 1.73 (95% CI 1.36-2.19), respectively. The RR for the 3 and 5-year DFS rate 1.40 (95% CI 1.19-1.65) and 2.10 (95% CI1.53-2.87), respectively. The proportion of patients who were CD3+, CD4+, and CD4+/CD8+ increased in the cellular therapy groups. No fatal adverse reactions were noted. CONCLUSION Chemotherapy combined with CIK/DC-CIK therapy after surgery resulted in low HR, and significantly increasing OS rates, DFS rates, and T-lymphocyte responses in patients with GC.
Collapse
Affiliation(s)
- Xiang Wang
- Lanzhou University Second Hospital
- Key Laboratory of Digestive System Tumors of Gansu Province
- Medicine School of Kunming University, China
| | - Song Tang
- Lanzhou University Second Hospital
- Key Laboratory of Digestive System Tumors of Gansu Province
| | - Xiang Cui
- Lanzhou University Second Hospital
- Key Laboratory of Digestive System Tumors of Gansu Province
| | - Jinwei Yang
- Lanzhou University Second Hospital
- Key Laboratory of Digestive System Tumors of Gansu Province
| | - Chunyu Geng
- Lanzhou University Second Hospital
- Key Laboratory of Digestive System Tumors of Gansu Province
| | - Cong Chen
- Lanzhou University Second Hospital
- Key Laboratory of Digestive System Tumors of Gansu Province
| | - Ning Zhou
- Key Laboratory of Digestive System Tumors of Gansu Province
- The First People's Hospital of Lanzhou City, Lanzhou
| | - Yumin Li
- Lanzhou University Second Hospital
- Key Laboratory of Digestive System Tumors of Gansu Province
| |
Collapse
|
26
|
Ognjenovic L, Trajkovski G, Gjoshev S, Shumkovski A, Dzambaz D, Hadzi-Manchev D, Volcevski G, Fildishevski I, Nikolova D, Petrushevska G, Janevska V, Janevski V. HER2 Positive Gastric Carcinomas and Their Clinico-Pathological Characteristics. Open Access Maced J Med Sci 2018; 6:1187-1192. [PMID: 30087720 PMCID: PMC6062279 DOI: 10.3889/oamjms.2018.280] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 06/18/2018] [Accepted: 06/19/2018] [Indexed: 12/05/2022] Open
Abstract
BACKGROUND: HER2 protein expression in gastric carcinoma, in correlation with existing, acknowledged prognostic factors which include the parameters that determine the TNM stage of the disease, could become the basis for ongoing research in the field of molecular targeted and personalised therapy. AIM: To determine the expression of the HER2 protein in gastric carcinoma and to correlate the expression of a HER2 protein with clinicopathological characteristics of the disease. MATERIAL AND METHODS: The data of HER2 protein expression and the parameters of the TNM classification were obtained from the histopathological reports of the Institute of Pathology in Skopje, and for the clinical stage we used patient’s files from the University Clinic for Abdominal Surgery in Skopje. RESULTS: The analysis of the correlation of HER2 protein expression and TNM classification parameters pointed out a significant correlation between HER2 protein expression and intragastric localisation of gastric carcinoma (P = 0.005), and the tumour grade of differentiation (P = 0.034). There was also a positive correlation between HER2 protein expression pattern and positive lymph nodes in patients with gastric carcinoma (P = 0.03). The expression pattern of HER2 +++ was significantly more common registered in patients with positive lymph nodes (P = 0.03) CONCLUSION: The expression of HER2 protein could represent a biological marker with prognostic and predictive value in patients with gastric carcinoma. Considering the high mortality rate in patients with gastric carcinoma and lack of international standardised therapeutic approach, research of the role and significance of HER2 overexpression and Trastuzumab therapy may prove useful in the development of new therapeutic strategies.
Collapse
Affiliation(s)
- Ljubomir Ognjenovic
- University Clinic of Abdominal Surgery, Faculty of Medicine, Ss Cyril and Methodius University of Skopje, Skopje, Republic of Macedonia
| | - Gjorgji Trajkovski
- University Clinic of Abdominal Surgery, Faculty of Medicine, Ss Cyril and Methodius University of Skopje, Skopje, Republic of Macedonia
| | - Stojan Gjoshev
- University Clinic of Abdominal Surgery, Faculty of Medicine, Ss Cyril and Methodius University of Skopje, Skopje, Republic of Macedonia
| | - Aleksandar Shumkovski
- University Clinic of Abdominal Surgery, Faculty of Medicine, Ss Cyril and Methodius University of Skopje, Skopje, Republic of Macedonia
| | - Darko Dzambaz
- University Clinic of Abdominal Surgery, Faculty of Medicine, Ss Cyril and Methodius University of Skopje, Skopje, Republic of Macedonia
| | - Dragan Hadzi-Manchev
- University Clinic of Abdominal Surgery, Faculty of Medicine, Ss Cyril and Methodius University of Skopje, Skopje, Republic of Macedonia
| | - Goce Volcevski
- University Clinic of Abdominal Surgery, Faculty of Medicine, Ss Cyril and Methodius University of Skopje, Skopje, Republic of Macedonia
| | - Igor Fildishevski
- University Clinic for Surgery St. Naum Ohridski, Skopje, Republic of Macedonia
| | - Dafina Nikolova
- University Clinic of Gastroenterohepatology, Faculty of Medicine, Ss Cyril and Methodius University of Skopje, Skopje, Republic of Macedonia
| | - Gordana Petrushevska
- Institute of Pathology, Faculty of Medicine, Ss Cyril and Methodius University of Skopje, Skopje, Republic of Macedonia
| | - Vesna Janevska
- Institute of Pathology, Faculty of Medicine, Ss Cyril and Methodius University of Skopje, Skopje, Republic of Macedonia
| | - Vlado Janevski
- University Clinic of Abdominal Surgery, Faculty of Medicine, Ss Cyril and Methodius University of Skopje, Skopje, Republic of Macedonia
| |
Collapse
|
27
|
Abbas M, Faggian A, Sintali DN, Khan GJ, Naeem S, Shi M, Dingding C. Current and future biomarkers in gastric cancer. Biomed Pharmacother 2018; 103:1688-1700. [DOI: 10.1016/j.biopha.2018.04.178] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 04/24/2018] [Accepted: 04/24/2018] [Indexed: 02/06/2023] Open
|
28
|
Shabbir A, Qureshi MA, Khalid AB, Mirza T, Shaikh A, Hasan SM. Gastric adenocarcinoma expressing human epidermal growth factor receptor in South Asian population. Saudi J Gastroenterol 2018; 24:289-293. [PMID: 29806596 PMCID: PMC6151997 DOI: 10.4103/sjg.sjg_23_18] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND/AIM Gastric cancer is the third leading cause of cancer mortality worldwide. Human epidermal growth factor (Her-2/neu) has shown strong therapeutic implication in breast cancer. Although the prevalence of Her-2/neu over-expression in gastric cancer has been reported across the world, it is still unknown from South Asia. The aim of this study is to evaluate Her-2/neu expression in gastric adenocarcinomas and to correlate with various clinicopathological variables. PATIENTS AND METHODS A total of 95 consecutive patients undergoing endoscopic biopsy or gastrectomy were recruited in this study. Clinicopathological parameters of all patients were recorded and hematoxylin and eosin (H and E) staining was performed. Over-expression of Her-2/neu was investigated by immunohistochemistry using α-Her-2 antibody. To quantify Her-2/neu over-expression, the Hofmann validation scoring system was used and further its association was seen with age, gender, histopathological type, grade, and stage of the tumor. Data were entered and analyzed using SPSS version 21. A P value of <0.05 was considered as significant. RESULTS Overall, 21 (22.1%) cases were positive for Her-2/neu overexpression from the total of 95 gastric adenocarcinomas. Her-2/neu was significantly expressed in low-grade gastric cancer (grade I = 50%, grade II = 34.5%, grade III = 14.5%; P = 0.030). Although there was insignificant difference between Her-2/neu over expression and other variables, Her-2/neu score 3+ was predominantly seen in females, age >60 years, Laurens intestinal type, and IIIC stage tumors. CONCLUSION Her-2/neu is over-expressed in a limited group of gastric cancer patients in our population and indicates a significant strong association with low grades of gastric cancer.
Collapse
Affiliation(s)
- Asma Shabbir
- Department of Pathology, Sindh Medical College, Jinnah Sindh Medical University, Karachi, Pakistan,Address for correspondence: Dr. Asma Shabbir, Department of Pathology, Sindh Medical College, Jinnah Sindh Medical University, Karachi, Pakistan. E-mail:
| | - Muhammad Asif Qureshi
- Department of Pathology, Dow International Medical College, Dow University of Health Sciences, Karachi, Pakistan
| | - Abdullah Bin Khalid
- Department of Pathology, Dow International Medical College, Dow University of Health Sciences, Karachi, Pakistan
| | - Talat Mirza
- Department of Pathology, Doctor. Ziauddin Hospital and University, Karachi, Pakistan
| | - Asma Shaikh
- Department of Pathology, Sindh Medical College, Jinnah Sindh Medical University, Karachi, Pakistan
| | - Syed Mehmood Hasan
- Department of Pathology, Sindh Medical College, Jinnah Sindh Medical University, Karachi, Pakistan
| |
Collapse
|
29
|
Zhu C, Wang Y, Wang X, Bai C, Su D, Cao B, Xu J. Profiling chemotherapy-associated myelotoxicity among Chinese gastric cancer population receiving cytotoxic conventional regimens: epidemiological features, timing, predictors and clinical impacts. J Cancer 2017; 8:2614-2625. [PMID: 28900499 PMCID: PMC5595091 DOI: 10.7150/jca.17847] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2016] [Accepted: 12/24/2016] [Indexed: 11/05/2022] Open
Abstract
Objectives: We aim to evaluate the epidemiological features, timing, predictors and clinical impacts of chemotherapy-associated myelotoxicity in Chinese gastric cancer population receiving six established cytotoxic conventional regimens (CF/XP, EC(O)F/EC(O)X, DC(O)F/DC(O)X, PC(O)F/PC(O)X, FOLFOX4, or mFOLFOX7/XELOX). Patients and methods: A 4-year multicenter, prospective, observational study was conducted in multiple hospitals/institutes spanning three major regions in China. A total of 1,285 patients with gastric cancer, treated with six selected regimens between 2010 and 2014 were included. Kaplan-meier analysis was applied to estimate the time to develop myelotoxicity events for each regimen. Multivariable logistic regression model was built to identify predictors associated with chemotherapy-induced myelotoxicity, evaluating detailed specific factors of patients, disease and treatment patterns. Results: Triplet regimens were associated with more moderate-to-severe myelotoxicity events than doublet regimens. DC(O)F/DC(O)X group presented with moderate-to-severe anaemia, thrombocytopenia, and leukopenia earlier than other regimen groups, with median time of 3.5, 4.8 and 3.3 cycles, respectively. PC(O)F/PC(O)X group had a shortest time to develop Moderate-to-Severe neutropenia (median time, 3.3 cycles). Multivariate analysis identified several independent predictors for moderate-to-severe myelotoxicity, including: baseline Hb<12.0 g/dL, male gender, KPS<80, previously treated with surgery, tumor located at gastroesophageal junction(GEJ), DC(O)F/DC(O)X regimen, palliative intent, triplet combination therapy and No. of cycles received≥4. Dose reductions≥20% occurred in 16.7% of patients and treatment delays≥7 days presented in 21.1% of patients, resulting in patients receiving an actual average Relative Dose Intensity (RDI) of 0.733. Conclusions: Myelotoxicity events were frequently observed within the gastric cancer population undertaking multicycle polychemotherapy. Predictive models based on risk factors identified for moderate-to-severe myelotoxicity should enable the targeted use of appropriate supportive care in an effort to facilitate the delivery of full chemotherapy doses on schedule.
Collapse
Affiliation(s)
- Ci Zhu
- Beijing 307 People's Liberation Army Hospital Cancer Centre, Beijing, China
| | - Yan Wang
- Beijing 307 People's Liberation Army Hospital Cancer Centre, Beijing, China
| | - Xicheng Wang
- Department of Gastrointestinal Oncology, Beijing Cancer Hospital, Beijing, China
| | - Chunmei Bai
- Department of Gastrointestinal Oncology, Peking Union Medical College Hospital, Beijing, China
| | - Dan Su
- Oncology Department, No. Chinese PLA General Hospital, Beijing, China
| | - Bing Cao
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Jianming Xu
- Beijing 307 People's Liberation Army Hospital Cancer Centre, Beijing, China
| |
Collapse
|
30
|
Ghazala S, Bilal J, Riaz IB. Advanced gastric cancer linitis plastica presented with disseminated intravascular coagulation. BMJ Case Rep 2016; 2016:bcr-2016-217675. [PMID: 27803086 DOI: 10.1136/bcr-2016-217675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
An old man was found unconscious; on admission found to have disseminated intravascular coagulation with concern of upper gastrointestinal bleed after he was found to have melena. Esophagogastroduodenoscopy on admission showed diffuse thickened gastric folds, and biopsy showed mucosal oedema. Bone marrow biopsy concerning for lymphoma was obtained showed adenocarcinoma. MRI of the abdomen was significant for diffuse gastric wall thickening. A repeat endoscopic ultrasound showed a diffuse gastric wall thickening of 15 mm and submucosal tunneling technique biopsy suggested high-grade, invasive, signet ring adenocarcinoma of the stomach. Oncology was consulted to initiate palliative chemotherapy. In retrospect, the patient was questioned regarding gastrointestinal symptoms; he reported gradual early satiety, dysphagia and unintentional weight loss over the course of 4 months.
Collapse
Affiliation(s)
- Sehem Ghazala
- Department of Internal Medicine, University of Arizona, Tucson, Arizona, USA
| | - Jawad Bilal
- Department of Internal Medicine, University of Arizona, Tucson, Arizona, USA
| | - Irbaz Bin Riaz
- Department of Internal Medicine, University of Arizona, Tucson, Arizona, USA
| |
Collapse
|
31
|
Tran P, Nguyen C, Klempner SJ. Targeting the Phosphatidylinositol-3-kinase Pathway in Gastric Cancer: Can Omics Improve Outcomes? Int Neurourol J 2016; 20:S131-140. [PMID: 27915478 PMCID: PMC5169087 DOI: 10.5213/inj.1632740.370] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 10/17/2016] [Indexed: 01/08/2023] Open
Abstract
Phosphatidylinositol-3-kinase (PI3K) pathway signaling is an established oncogenic signal transduction pathway implicated in multiple malignancies. Therapeutic targeting of PI3K pathway components has improved outcomes in chronic lymphocytic leukemia, kidney cancer, breast cancer, and neuroendocrine tumors. Gastric cancers harbor some of the highest rates of oncogenic alterations in PI3K but attempts to translate this genomic observation have met with limited clinical success and novel approaches are needed. In the following review we discuss PI3K signaling, previous preclinical and clinical investigations in gastric cancer, and discuss future strategies aimed at overcoming resistance and improving efficacy. Identification and refinement of molecular tumor subtypes, development of predictive biomarkers along, and rational drug combination strategies are key to capitalizing on the therapeutic potential of PI3K pathway directed therapies in gastric cancers.
Collapse
Affiliation(s)
- Phu Tran
- Division of Hematology-Oncology, University of California Irvine, Orange, CA, USA
| | - Cham Nguyen
- Department of Pharmacy, University of California Irvine, Orange, CA, USA
| | - Samuel J. Klempner
- The Angeles Clinic and Research Institute, Los Angeles, CA, USA
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
32
|
LaBonte MJ, Yang D, Zhang W, Wilson PM, Nagarwala YM, Koch KM, Briner C, Kaneko T, Rha SY, Gladkov O, Urba SG, Sakaeva D, Pishvaian MJ, Hsieh RK, Lee WP, Lenz HJ. A Phase II Biomarker-Embedded Study of Lapatinib plus Capecitabine as First-line Therapy in Patients with Advanced or Metastatic Gastric Cancer. Mol Cancer Ther 2016; 15:2251-8. [PMID: 27325685 DOI: 10.1158/1535-7163.mct-15-0908] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Accepted: 05/05/2016] [Indexed: 02/07/2023]
Abstract
An exploratory phase II biomarker-embedded trial (LPT109747; NCT00526669) designed to determine the association of lapatinib-induced fluoropyrimidine gene changes with efficacy of lapatinib plus capecitabine as first-line treatment for advanced gastric cancer or gastroesophageal junction adenocarcinoma independent of tumor HER2 status. Tumor biopsies obtained before and after 7-day lapatinib (1,250 mg) to analyze changes in gene expression, followed by a 14-day course of capecitabine (1,000 mg/m(2) twice daily, 14/21 days) plus lapatinib 1,250 mg daily. Blood samples were acquired for pharmacokinetic analysis. Primary clinical objectives were response rate (RR) and 5-month progression-free survival (PFS). Secondary objectives were overall survival (OS), PFS, time to response, duration of response, toxicity, and identification of associations between lapatinib pharmacokinetics and biomarker endpoints. Primary biomarker objectives were modulation of 5-FU-pathway genes by lapatinib, effects of germline SNPs on treatment outcome, and trough steady-state plasma lapatinib concentrations. Sixty-eight patients were enrolled; (75% gastric cancer, 25% gastroesophageal junction). Twelve patients (17.9%) had confirmed partial response, 31 (46.3%) had stable disease, and 16 (23.9%) had progressive disease. Median PFS and OS were 3.3 and 6.3 months, respectively. Frequent adverse events included diarrhea (45%), decreased appetite (39%), nausea (36%), and fatigue (36%). Lapatinib induced no changes in gene expression from baseline and no significant associations were found for SNPs analyzed. Elevated baseline HER3 mRNA expression was associated with a higher RR (33% vs. 0%; P = 0.008). Lapatinib plus capecitabine was well tolerated, demonstrating modest antitumor activity in patients with advanced gastric cancer. The association of elevated HER3 and RR warrants further investigation as an important player for HER-targeted regimens in combination with capecitabine. Mol Cancer Ther; 15(9); 2251-8. ©2016 AACR.
Collapse
Affiliation(s)
- Melissa J LaBonte
- Azusa Pacific University, Azusa, California. Queen's University Belfast, Belfast, United Kingdom
| | - Dongyun Yang
- University of Southern California Norris Comprehensive Cancer Center, Los Angeles, California
| | - Wu Zhang
- University of Southern California Norris Comprehensive Cancer Center, Los Angeles, California
| | - Peter M Wilson
- University of Southern California Norris Comprehensive Cancer Center, Los Angeles, California
| | - Yasir M Nagarwala
- GlaxoSmithKline Clinical Development and Medical Affairs Oncology, Collegeville, Pennsylvania
| | - Kevin M Koch
- GlaxoSmithKline Clinical Pharmacology, Durham North Carolina
| | - Colleen Briner
- GlaxoSmithKline Oncology Clinical and Operational Sciences, Collegeville, Pennsylvania
| | - Tomomi Kaneko
- GlaxoSmithKline Oncology Clinical and Operational Sciences, Collegeville, Pennsylvania
| | - Sun-Young Rha
- Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, South Korea
| | - Oleg Gladkov
- Chelyabinsk Regional Clinical Oncology Dispensary, Chelyabinsk, Russia
| | - Susan G Urba
- University of Michigan Cancer Center, Ann Arbor, Michigan
| | - Dina Sakaeva
- Bashkir Republican Clinical Oncology Dispensary, Ufa, Russia
| | | | | | - Wei-Ping Lee
- Taipei Veterans General Hospital and Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei, Taiwan
| | - Heinz-Josef Lenz
- University of Southern California Norris Comprehensive Cancer Center, Los Angeles, California.
| |
Collapse
|
33
|
Tewari M, Kumar A, Mishra RR, Kumar M, Shukla HS. HER2 Expression in Gastric and Gastroesophageal Cancer: Report from a Tertiary Care Hospital in North India. Indian J Surg 2015; 77:447-51. [PMID: 26730043 PMCID: PMC4692956 DOI: 10.1007/s12262-013-0871-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2012] [Accepted: 01/17/2013] [Indexed: 02/05/2023] Open
Abstract
Despite improvements in chemotherapy, survival of metastatic gastric and gastroesophageal junction (GEJ) adenocarcinoma remains poor. Trastuzumab, a monoclonal antibody targeting the human epidermal growth factor receptor 2 (HER2), has shown promise in improving survival of these patients by a recent large phase III trial. HER2 status in gastric and GEJ cancers, although reported from across the world, is yet unknown in India due to lack of published literature from the country. HER2 status in 70 samples of gastric and GEJ adenocarcinomas (Siewert type III) was evaluated by immunohistochemistry (IHC) in this study using the gastric cancer scoring system. It was also correlated with clinic-pathologic factors. Samples with IHC score 2+ and 3+ were taken as HER2 positive. HER2 overexpression was found in 15 (21.4 %) samples, was significantly (p = 0.006) more common in intestinal type (45 %), but it did not correlate with age, gender, stage, or grade of tumor and did not affect the 2-year disease-free survival. HER2 overexpression is found only in a minority of patients with gastric and GEJ cancers in the Indian population. A large cohort of patients with a longer follow-up will be required to assess for any significant statistical association of HER2 expression with prognosis of these patients.
Collapse
Affiliation(s)
- Mallika Tewari
- />Department of Surgical Oncology, Institute of Medical Sciences, Banaras Hindu University, 7 SKG Colony, Lanka, Varanasi, 221005 Uttar Pradesh India
| | - Akhileshwar Kumar
- />Department of Surgical Oncology, Institute of Medical Sciences, Banaras Hindu University, 7 SKG Colony, Lanka, Varanasi, 221005 Uttar Pradesh India
| | - RR Mishra
- />Department of Surgical Oncology, Institute of Medical Sciences, Banaras Hindu University, 7 SKG Colony, Lanka, Varanasi, 221005 Uttar Pradesh India
- />Department of Microbiology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh India
| | - Mohan Kumar
- />Department of Pathology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh India
| | - Hari S Shukla
- />Department of Surgical Oncology, Institute of Medical Sciences, Banaras Hindu University, 7 SKG Colony, Lanka, Varanasi, 221005 Uttar Pradesh India
| |
Collapse
|
34
|
Rajagopal I, Niveditha SR, Sahadev R, Nagappa PK, Rajendra SG. HER 2 Expression in Gastric and Gastro-esophageal Junction (GEJ) Adenocarcinomas. J Clin Diagn Res 2015; 9:EC06-10. [PMID: 25954623 PMCID: PMC4413071 DOI: 10.7860/jcdr/2015/12581.5630] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Accepted: 01/27/2015] [Indexed: 01/18/2023]
Abstract
INTRODUCTION Gastric cancer is one of the leading causes of cancer mortality in the world/India with majority being diagnosed at an advanced stage. Various chemotherapeutic regimens have modestly improved overall survival leading to quest for novel therapeutic agents. Overexpression of HER2 in many gastric cancers has lead to the advent of targeted therapy with anti HER2 antibody like Trastusumab which has improved the overall survival. MATERIALS AND METHODS Sixty cases of gastric adenocarcinomas (44 biopsies and 16 gastrectomies) over the past five years ( June 2009 to June 2014),were included in the study. Diagnosis was confirmed by review of slides and IHC with anti HER2 antibodies was performed using Dako Real Envision Detection system and scoring was done by Hoffmann et al., scoring system. RESULTS Of the 60 cases, majority were males (60%),with a mean age of 65.65 yrs. Tumours in antrum (76.7%) formed the major bulk. HER2 expression was observed in 26.7% of Tumours, predominantly in males (p=0.006) and intestinal type (p= 0.054). HER2 expression correlated with Tumour grade (moderately differentiated and well differentiated, p= 0.042). Tumours of gastro-esophageal junction (GEJ) showed HER2 expression in 45.5% as opposed to 22.4% in gastric location. Poorly differentiated and diffuse type of adenocarcinomas did not express HER2. Two of three Tumours from patients in the age group 31-40 y expressed HER2. CONCLUSION Male gender, intestinal-type and moderately differentiated gastric cancers may be the ones that can be targeted for therapy using Herceptin. Though trastusumab is approved for advanced gastric and GEJ cancers, it's role in adjuvant / neo-adjuvant setting in early stages needs to be evaluated with newer agents like Pertuzumab, Bevacizumab, especially in young patients.
Collapse
Affiliation(s)
- Indu Rajagopal
- Post Graduate, Department of Pathology, Kempegowda Institute of Medical Sciences, Karnataka, India
| | - S R Niveditha
- Professor, Department of Pathology, Kempegowda Institute of Medical Sciences, Karnataka, India
| | - R Sahadev
- Professor and HOD, Department of Surgical Gastroenterology, Kempegowda Institute of Medical Sciences, Karnataka, India
| | - Preethan Kamagere Nagappa
- Associate Professor, Department of Surgical Gastroenterology, Kempegowda Institute of Medical Sciences, Karnataka, India
| | - Sowmya Goddanakoppal Rajendra
- Senior Resident, Department of Surgical Gastroenterology, Kempegowda Institute of Medical Sciences, Karnataka, India
| |
Collapse
|
35
|
Yuan F, Shi H, Ji J, Cai Q, Chen X, Yu Y, Liu B, Zhu Z, Zhang J. Capecitabine metronomic chemotherapy inhibits the proliferation of gastric cancer cells through anti-angiogenesis. Oncol Rep 2015; 33:1753-62. [PMID: 25634241 DOI: 10.3892/or.2015.3765] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Accepted: 01/12/2015] [Indexed: 11/05/2022] Open
Abstract
To evaluate the inhibitory effect and mechanism of capecitabine metronomic chemotherapy on gastric cancer cells. In vitro, the effects of 5-fluorouracil (Fu) metronomic chemotherapy on proliferation, apoptosis, tube formation ability, and angiogenesis were detected. In vivo, Ki-67, CD34 and VEGF were detected by immunohistochemical staining (IHC). Flow cytometry was used to detect the percentage of circulating endothelial progenitors (CEPs), and VEGF and PDGF were detected by ELISA in the peripheral blood of nude mice. The proliferation of the SGC-7901 and AGS gastric cancer cell lines in the metronomic 5-Fu group was decreased compared with the control group in vitro. The total length of the small tubes and tubular junction numbers were significantly lower in the metronomic group than the control group. The VEGF and PDGF levels in the cell culture supernatants were lower in the metronomic group than the control group. Compared with the control group, the CEP percentage was decreased in the peripheral blood of tumor-bearing nude mice following treatment with metronomic 5-Fu or capecitabine chemotherapy. No significant changes were found in the conventional or control group. In the peripheral blood of tumor-bearing nude mice, the VEGF and PDGF levels were decreased in the metronomic groups. Metronomic 5-Fu inhibited the proliferation of gastric cancer cells in vitro and in vivo, and their antitumor effects were non-inferior to those of conventional dose chemotherapy, with mild side effects. Thus, tumor inhibition may be attributed to anti-angiogenesis.
Collapse
Affiliation(s)
- Fei Yuan
- Department of Pathology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, P.R. China
| | - Hailong Shi
- Department of Surgery, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, P.R. China
| | - Jun Ji
- Department of Surgery, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, P.R. China
| | - Qu Cai
- Department of Surgery, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, P.R. China
| | - Xuehua Chen
- Department of Surgery, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, P.R. China
| | - Yingyan Yu
- Department of Surgery, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, P.R. China
| | - Bingya Liu
- Department of Surgery, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, P.R. China
| | - Zhenggang Zhu
- Department of Surgery, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, P.R. China
| | - Jun Zhang
- Department of Surgery, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, P.R. China
| |
Collapse
|
36
|
Xiong B, Ma L, Huang W, Cheng Y, Zhao Q, Liu J. WITHDRAWN: Clinical effectiveness of neoadjuvant chemotherapy in advanced gastric cancer: An updated meta-analysis of 12 randomized controlled trials. Surg Oncol 2014:S0960-7404(14)00097-8. [PMID: 25515042 DOI: 10.1016/j.suronc.2014.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Revised: 10/16/2014] [Accepted: 11/23/2014] [Indexed: 11/20/2022]
Abstract
This article has been withdrawn at the request of the author(s) and/or editor. The Publisher apologizes for any inconvenience this may cause. The full Elsevier Policy on Article Withdrawal can be found at http://www.elsevier.com/locate/withdrawalpolicy.
Collapse
Affiliation(s)
- Binghong Xiong
- Department of General Surgery, Peking University Shougang Hospital, No 9 Jinyuanzhuang Road, Shijingshan District, Beijing, 100144, PR China.
| | - Li Ma
- Department of Endocrinology, The Third Hospital of Mianyang, Sichuan Province, Mianyang, 621000, PR China
| | - Wei Huang
- Department of General Surgery, The Ninth People's Hospital of Chongqing City, Chongqing 400070, PR China
| | - Yong Cheng
- Department of General Surgery, The First Affiliated Hospital of Chongqing Medical University, No 1 Youyi Road, Yuanjiagang, Yuzhong District, Chongqing, 400016, PR China.
| | - Qikang Zhao
- Department of General Surgery, Peking University Shougang Hospital, No 9 Jinyuanzhuang Road, Shijingshan District, Beijing, 100144, PR China
| | - Jingshan Liu
- Department of General Surgery, Peking University Shougang Hospital, No 9 Jinyuanzhuang Road, Shijingshan District, Beijing, 100144, PR China
| |
Collapse
|
37
|
Trendowski M. Exploiting the cytoskeletal filaments of neoplastic cells to potentiate a novel therapeutic approach. Biochim Biophys Acta Rev Cancer 2014; 1846:599-616. [PMID: 25286320 DOI: 10.1016/j.bbcan.2014.09.007] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2014] [Revised: 09/19/2014] [Accepted: 09/21/2014] [Indexed: 02/06/2023]
Abstract
Although cytoskeletal-directed agents have been a mainstay in chemotherapeutic protocols due to their ability to readily interfere with the rapid mitotic progression of neoplastic cells, they are all microtubule-based drugs, and there has yet to be any microfilament- or intermediate filament-directed agents approved for clinical use. There are many inherent differences between the cytoskeletal networks of malignant and normal cells, providing an ideal target to attain preferential damage. Further, numerous microfilament-directed agents, and an intermediate filament-directed agent of particular interest (withaferin A) have demonstrated in vitro and in vivo efficacy, suggesting that cytoskeletal filaments may be exploited to supplement chemotherapeutic approaches currently used in the clinical setting. Therefore, this review is intended to expose academics and clinicians to the tremendous variety of cytoskeletal filament-directed agents that are currently available for further chemotherapeutic evaluation. The mechanisms by which microfilament directed- and intermediate filament-directed agents damage malignant cells are discussed in detail in order to establish how the drugs can be used in combination with each other, or with currently approved chemotherapeutic agents to generate a substantial synergistic attack, potentially establishing a new paradigm of chemotherapeutic agents.
Collapse
Affiliation(s)
- Matthew Trendowski
- Department of Biology, Syracuse University, 107 College Place, Syracuse, NY 13244, USA.
| |
Collapse
|
38
|
Liu JS, He SC, Zhang ZL, Chen R, Fan L, Qiu GL, Chang S, Li L, Che XM. Anticancer effects of β-elemene in gastric cancer cells and its potential underlying proteins: a proteomic study. Oncol Rep 2014; 32:2635-47. [PMID: 25333415 DOI: 10.3892/or.2014.3490] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2014] [Accepted: 08/26/2014] [Indexed: 01/19/2023] Open
Abstract
Gastric cancer is a common malignancy with a poor prognosis. β-elemene is a broad-spectrum anticancer drug extracted from the traditional Chinese medicinal herb Curcuma wenyujin. In the present study, we investigated the anticancer effects of β-elemene in gastric cancer cells and the potential proteins involved. Human SGC7901 and MKN45 gastric cancer cells were treated with different concentrations of β-elemene. Cell viability, clonogenic survival and apoptotic cell death were assessed. β-elemene inhibited viability and decreased clonogenic survival of gastric cancer cells in a dose-dependent manner. Apoptosis induction contributed to the anticancer effects. We then employed a proteomic method, isobaric tags for relative and absolute quantitation (iTRAQ), to detect the proteins altered by β-elemene. In total, 147 upregulated proteins and 86 downregulated proteins were identified in response to β-elemene treatment in SGC7901 gastric cancer cells. Among them, expression of p21-activated protein kinase‑interacting protein 1 (PAK1IP1), Bcl-2-associated transcription factor 1 (BTF) and topoisomerase 2-α (TOPIIα) were validated by western blot analyses and the trends were consistent with iTRAQ results. Top pathways involved in β-elemene treatment in SGC7901 gastric cancer cells included ribosome signaling, peroxisome proliferator-activated receptors (PPARs) signaling pathway, regulation of actin cytoskeleton, phagosome, biosynthesis and metabolism of some amino acids. Collectively, our results suggest a promising therapeutic role of β-elemene in gastric cancer. The differentially expressed proteins provide further insight into the potential mechanisms involved in gastric cancer treatment using β-elemene.
Collapse
Affiliation(s)
- Jun-Song Liu
- Department of General Surgery, The First Affiliated Hospital of Medical College of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Shi-Cai He
- Department of General Surgery, The First Affiliated Hospital of Medical College of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Zheng-Liang Zhang
- Department of General Surgery, The First Affiliated Hospital of Medical College of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Rui Chen
- Department of General Surgery, The First Affiliated Hospital of Medical College of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Lin Fan
- Department of General Surgery, The First Affiliated Hospital of Medical College of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Guang-Lin Qiu
- Department of General Surgery, The First Affiliated Hospital of Medical College of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Shuai Chang
- Department of General Surgery, The First Affiliated Hospital of Medical College of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Liang Li
- Department of General Surgery, The First Affiliated Hospital of Medical College of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Xiang-Ming Che
- Department of General Surgery, The First Affiliated Hospital of Medical College of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| |
Collapse
|
39
|
Jung HK, Wang K, Jung MK, Kim IS, Lee BH. In vivo near-infrared fluorescence imaging of apoptosis using histone H1-targeting peptide probe after anti-cancer treatment with cisplatin and cetuximab for early decision on tumor response. PLoS One 2014; 9:e100341. [PMID: 24949860 PMCID: PMC4065102 DOI: 10.1371/journal.pone.0100341] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2014] [Accepted: 05/23/2014] [Indexed: 12/29/2022] Open
Abstract
Early decision on tumor response after anti-cancer treatment is still an unmet medical need. Here we investigated whether in vivo imaging of apoptosis using linear and cyclic (disulfide-bonded) form of ApoPep-1, a peptide that recognizes histone H1 exposed on apoptotic cells, at an early stage after treatment could predict tumor response to the treatment later. Treatment of stomach tumor cells with cistplatin or cetuximab alone induced apoptosis, while combination of cisplatin plus cetuximab more efficiently induced apoptosis, as detected by binding with linear and cyclic form of ApoPep-1. However, the differences between the single agent and combination treatment were more remarkable as detected with the cyclic form compared to the linear form. In tumor-bearing mice, apoptosis imaging was performed 1 week and 2 weeks after the initiation of treatment, while tumor volumes and weights were measured 3 weeks after the treatment. In vivo fluorescence imaging signals obtained by the uptake of ApoPep-1 to tumor was most remarkable in the group injected with cyclic form of ApoPep-1 at 1 week after combined treatment with cisplatin plus cetuximab. Correlation analysis revealed that imaging signals by cyclic ApoPep-1 at 1 week after treatment with cisplatin plus cetuximab in combination were most closely related with tumor volume changes (r2 = 0.934). These results demonstrate that in vivo apoptosis imaging using Apopep-1, especially cyclic ApoPep-1, is a sensitive and predictive tool for early decision on stomach tumor response after anti-cancer treatment.
Collapse
Affiliation(s)
- Hyun-Kyung Jung
- Department of Biochemistry and Cell Biology and School of Medicine, Kyungpook National University, Daegu, Korea
- BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, Graduate School, Kyungpook National University, Daegu, Korea
| | - Kai Wang
- Department of Plastic Surgery, Henan Provincial People's Hospital, Zhengzhou, Henan, China
| | - Min Kyu Jung
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Daegu, Korea
| | - In-San Kim
- Department of Biochemistry and Cell Biology and School of Medicine, Kyungpook National University, Daegu, Korea
- BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, Graduate School, Kyungpook National University, Daegu, Korea
| | - Byung-Heon Lee
- Department of Biochemistry and Cell Biology and School of Medicine, Kyungpook National University, Daegu, Korea
- BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, Graduate School, Kyungpook National University, Daegu, Korea
- * E-mail:
| |
Collapse
|
40
|
Rani I, Vaiphei K, Agnihotri N. Supplementation of fish oil augments efficacy and attenuates toxicity of 5-fluorouracil in 1,2-dimethylhydrazine dihydrochloride/dextran sulfate sodium-induced colon carcinogenesis. Cancer Chemother Pharmacol 2014; 74:309-22. [PMID: 24916547 DOI: 10.1007/s00280-014-2497-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2013] [Accepted: 05/22/2014] [Indexed: 12/20/2022]
Abstract
PURPOSE 5-Fluorouracil (5-FU) is used for the treatment of colorectal cancer, but has low therapeutic response rate and severe side effects. Recently, fish oil (FO) rich in n-3 polyunsaturated fatty acids has been preferred to chemosensitize tumor cells to anticancer drugs. Therefore, the current study is designed to evaluate chemotherapeutic efficacy and toxicity profile of 5-FU in combination with FO in 1,2-dimethylhydrazine dihydrochloride/dextran sulfate sodium (DMH/DSS)-induced colon cancer model. METHODS The therapeutic efficacy of 5-FU along with FO was analyzed through assessment of survival rate, tumor burden, volume, serum sialic acid levels, cytokeratin 19 (CK19) expression and index of cell proliferation such as cell cycle progression. Toxicological aspects were evaluated by standard functional and structural parameters related to spleen, gastrointestinal, liver and kidney. RESULTS In the present study, 5-FU in combination with FO increased the survival rate in carcinogen-treated animals. Synergism of 5-FU and FO was also reflected in significant inhibition in tumor growth and serum sialic acid levels in DMH/DSS model. Moreover, the combination dosage significantly augmented the inhibition of cell cycle progression, as shown by CK19 expression. Additionally, FO ameliorated hematologic depression, gastrointestinal, hepatic and renal toxicity caused by 5-FU as substantiated by a marked improvement in structural and functional alterations of these organs. CONCLUSION The supplementation of FO is potentially a promising option for increasing the therapeutic potential and mitigating the side effects of 5-FU.
Collapse
Affiliation(s)
- Isha Rani
- Department of Biochemistry, Panjab University, Chandigarh, 160014, India
| | | | | |
Collapse
|
41
|
Mongan AM, Kalachand R, King S, O'Farrell NJ, Power D, Ravi N, Muldoon C, O'Byrne K, Reynolds JV. Outcomes in gastric and junctional cancer using neoadjuvant and adjuvant chemotherapy (epirubicin, oxaliplatin, and capecitabine) and radical surgery. Ir J Med Sci 2014; 184:417-23. [PMID: 24879337 DOI: 10.1007/s11845-014-1135-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2013] [Accepted: 05/05/2014] [Indexed: 01/12/2023]
Abstract
BACKGROUND The MAGIC/UK Medical Research Council (MRC) trial set the standard of care for treatment of resectable gastric and junctional adenocarcinoma, demonstrating that perioperative chemotherapy with epirubicin, cisplatin and 5-fluorouracil (ECF) confers a survival benefit over surgery alone. The randomized ECF for advanced and locally advanced esophagogastric cancer (REAL-2) trial showed that, in the metastatic setting, the EOX regimen (epirubicin, oxaliplatin and capecitabine) is as effective as ECF, with a favourable toxicity profile. METHODS Consecutive patients with resectable gastric or junctional adenocarcinoma treated with perioperative EOX, between 2007 and 2012, were retrospectively analysed. RESULTS Fifty-nine patients (12 female, 47 male), commenced EOX therapy; 47 underwent surgery. A good pathological response was seen in 34%, (16/47). Disease recurrence occurred in 19 patients (19/47, 40%). Median overall survival was 22 months, with 4-year survival of 47%. Chemotoxicities were consistent with those previously reported for this regimen. CONCLUSION This study in a high-volume centre demonstrates that EOX in resectable gastric and junctional adenocarcinoma is associated with a reasonable safety profile, and efficacy consistent with that reported for ECF.
Collapse
Affiliation(s)
- A M Mongan
- Department of Surgery, Trinity Centre for Health Sciences, St. James's Hospital, Dublin 8, Ireland
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Guo MG, Zheng Q, zhong Di J, Yang Z. Histological complete response to a combined docetaxel/cisplatin/fluorouracil neoadjuvant chemotherapy for T4 stage gastric adenocarcinoma. World J Surg Oncol 2014; 12:150. [PMID: 24885421 PMCID: PMC4032865 DOI: 10.1186/1477-7819-12-150] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2013] [Accepted: 04/20/2014] [Indexed: 12/26/2022] Open
Abstract
Local advanced gastric carcinoma has a very poor prognosis. When a T4 gastric carcinoma has invaded the surrounding tissues and organs, curative resection is unlikely. We present here a case of a 63-year-old woman with a T4 unresectable gastric adenocarcinoma. She underwent two 3-week cycles of docetaxel/cisplatin/fluorouracil chemotherapy, followed by radical gastric resection. Each cycle consisted of 75 mg/m2 docetaxel and 75 mg/m2 cisplatin on day 1, and 200 mg/m2 leucovorin and 500 mg/m2 fluorouracil on days 1 through 5. The patient exhibited a complete histologic response. Our results indicate that docetaxel/cisplatin/fluorouracil neoadjuvant chemotherapy is a promising method of treatment for advanced gastric cancer.
Collapse
Affiliation(s)
- Ming gao Guo
- Department of Surgery, the Six People's Hospital of Shanghai, Shanghai Jiaotong University, Shanghai 200233, China.
| | | | | | | |
Collapse
|
43
|
Xiong B, Ma L, Cheng Y, Zhang C. Clinical effectiveness of neoadjuvant chemotherapy in advanced gastric cancer: an updated meta-analysis of randomized controlled trials. Eur J Surg Oncol 2014; 40:1321-30. [PMID: 25239442 DOI: 10.1016/j.ejso.2014.01.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2013] [Revised: 12/19/2013] [Accepted: 01/05/2014] [Indexed: 12/19/2022] Open
Abstract
AIMS To assess the efficacy and safety of neoadjuvant chemotherapy (NAC) for advanced gastric cancer (AGC). METHODS By searching electronic databases (PubMed, Embase, Cochrane Library) and ASCO proceedings from 1990 to 2012, all randomized controlled trials (RCTs) which compared the effect of NAC-combined surgery versus surgery alone in AGC were included. All calculations and statistical tests were performed using RevMan 5.0 software. RESULTS 12 RCTs with a total of 1820 patients were included. All patients had locally advanced but resectable gastric cancer and received NAC. NAC can slightly improve the survival rate (OR = 1.32, 95% confidence interval (CI): 1.07-1.64, P = 0.01), with little or no significant benefits in subgroup analyses between either different population or regimens. NAC can significantly improve the 3-year progression-free survival (PFS) (OR: 1.85, 95% CI: 1.39-2.46, p < 0.0001), tumor down-staging rate (OR: 1.71, 95% CI: 1.26, 2.33, p = 0.0006) and R0 resection rate (OR: 1.38, 95% CI: 1.08-1.78, P = 0.01) of patients with AGC. There was no difference between the two arms, in terms of relapse rates (OR: 1.03, 95% CI: 0.60-1.78, p = 0.92), operative complications (OR: 1.20, 95% CI: 0.90-1.58, p = 0.21), perioperative mortality (OR: 1.14, 95% CI: 0.64-2.05, p = 0.65) and grade 3/4 adverse effects: gastrointestinal problem (OR: 0.57, 95% CI: 0.25-1.30, p = 0.18), leukopenia (OR: 0.88, 95% CI: 0.41-1.91, p = 0.75), thrombocytopenia (OR: 1.27, 95% CI: 0.27-5.93, p = 0.76). CONCLUSION NAC is effective and safe. However, further prospective multi-national and multi-center RCTs are still needed in order to investigate the long-term oncological and functional outcomes to define the clinical benefits of NAC and the most effective strategies for AGC.
Collapse
Affiliation(s)
- B Xiong
- Department of General Surgery, The First Affiliated Hospital of Chongqing Medical University, No. 1, Youyi Road, Yuanjiagang, Yuzhong District, Chongqing 400016, PR China.
| | - L Ma
- Department of Internal Medicine, Chongqing Huaxi Hospital, Banan District, Chongqing 400054, PR China
| | - Y Cheng
- Department of General Surgery, The First Affiliated Hospital of Chongqing Medical University, No. 1, Youyi Road, Yuanjiagang, Yuzhong District, Chongqing 400016, PR China.
| | - C Zhang
- Department of General Surgery, The First Affiliated Hospital of Chongqing Medical University, No. 1, Youyi Road, Yuanjiagang, Yuzhong District, Chongqing 400016, PR China.
| |
Collapse
|
44
|
Ina K, Kataoka T, Ando T. The use of lentinan for treating gastric cancer. Anticancer Agents Med Chem 2014; 13:681-8. [PMID: 23092289 PMCID: PMC3664515 DOI: 10.2174/1871520611313050002] [Citation(s) in RCA: 131] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2012] [Revised: 08/23/2012] [Accepted: 10/17/2012] [Indexed: 12/11/2022]
Abstract
Natural compounds containing fungal β-glucans have been used to improve general health for thousands of years in China and Japan. Lentinan, the backbone of β-(1, 3)-glucan with β-(1, 6) branches, is one of the active ingredients purified from Shiitake mushrooms and has been approved as a biological response modifier for the treatment of gastric cancer in Japan. Despite recent advances in chemotherapeutic agents, unresectable or recurrent gastric cancer remains an incurable disease, with survival rates being far from satisfactory. Recent clinical studies have shown that chemo-immunotherapy using lentinan prolongs the survival of patients with advanced gastric cancer, as compared to chemotherapy alone. In addition, trastuzumab, an antibody against HER2/neu growth factor receptor, has been used for the treatment of gastric cancer in combination with cytotoxic chemotherapeutic agents. Lentinan may exert a synergistic action with anti-cancer monoclonal antibodies to activate complement systems through the mechanism of antibody-dependent cellular cytotoxicity and complement dependent cytotoxicity. Because a better understanding of its biological activities should enable us to use lentinan more efficiently in the treatment of gastric cancer, immunological effects provided by β-glucans, a possible mode of action of lentinan, and its clinical application including future potential uses are discussed in the present review.
Collapse
Affiliation(s)
- Kenji Ina
- Department of Medical Oncology, Nagoya Memorial Hospital, Nagoya 468-8520, Japan.
| | | | | |
Collapse
|
45
|
Wang T, Gao J, Yu J, Shen L. Synergistic inhibitory effect of wogonin and low-dose paclitaxel on gastric cancer cells and tumor xenografts. Chin J Cancer Res 2013; 25:505-13. [PMID: 24255573 DOI: 10.3978/j.issn.1000-9604.2013.08.14] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2012] [Accepted: 05/10/2012] [Indexed: 12/31/2022] Open
Abstract
OBJECTIVE To investigate the synergistic inhibitory effects of wogonin (WOG) and chemotherapeutic drugs on growth of gastric cancer cells and tumor xenografts. METHODS The IC50 values of WOG, cisplatin (CDDP) and paclitaxel (PTX) in four gastric cancer cell lines were determined by MTS assay. Hoechst staining and the median effect method of Chou-Talalay were used to assess the apoptosis of cells and the interaction of two drugs, respectively. BGC-823-derived xenografts in nude mice were established to investigate the effects of WOG combined with chemotherapeutic drugs in vivo. RESULTS WOG, CDDP and PTX inhibited the growth of BGC-823, MGC-803, MKN-45 and HGC-27 gastric cancer cells in a dose-dependent manner. WOG combined with CDDP or PTX synergistically inhibited the growth of all gastric cancer cell lines in vitro. In BGC-823, MGC-803, HGC-27 and MKN-45 cell lines, synergisms between WOG and PTX were shown when the fraction affected (Fa) values were <0.45, <0.90, <0.85 and <0.60. While WOG and CDDP had a synergistic inhibitory effect when the Fa values were >0, >0, >0.65 and >0.10. From the results of in vivo experiments using tumor xenografts, WOG and low-dose PTX showed better efficacy than either drug alone. The inhibitory percentages of tumor weight were 61.58%, 20.29%, and 22.28% for the combination, WOG-alone, and low-dose PTX-alone groups, respectively. Notably, WOG combined with CDDP displayed very high toxicity. CONCLUSIONS A synergistic inhibitory effect on growth was observed when WOG was combined with low-dose PTX in gastric cancer cells and tumor xenografts. These findings provide evidence for the design of a clinical trial to test the combination of WOG with low-dose PTX in human gastric cancer.
Collapse
Affiliation(s)
- Tingting Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | | | | | | |
Collapse
|
46
|
Florou D, Patsis C, Ardavanis A, Scorilas A. Effect of doxorubicin, oxaliplatin, and methotrexate administration on the transcriptional activity of BCL-2 family gene members in stomach cancer cells. Cancer Biol Ther 2013; 14:587-96. [PMID: 23792648 DOI: 10.4161/cbt.24591] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Defective apoptosis comprises the main reason for tumor aggressiveness and chemotherapy tolerance in solid neoplasias. Among the BCL-2 family members, whose mRNA or protein expression varies considerably in different human malignancies, BCL2L12 is the one for which we have recently shown its propitious prognostic value in gastric cancer. The purpose of the current work was to investigate the expression behavior of BCL2L12, BAX, and BCL-2 in human stomach adenocarcinoma cells following their exposure to anti-tumor substances. The 3-(4,5-dimethyl thiazol-2-yl)-2,5-diphenyl tetrazolium bromide and trypan blue methods assessed the impact of doxorubicin, oxaliplatin and methotrexate on AGS cells' viability and growth. Following isolation from cells, total RNA was reverse-transcribed to cDNA. Quantification of target genes' expression was performed with real-time PCR using SYBR Green detection system. The relative changes in their mRNA levels between drug-exposed and untreated cells were calculated with the comparative Ct method (2(-ddCt)). All three drugs, as a result of their administration to AGS cancer cells for particular time intervals, provoked substantial fluctuations in the transcriptional levels of the apoptosis-related genes studied. While BAX was principally upregulated, striking similar were the notable changes regarding BCL-2 and BCL2L12 expression in our cellular system. Our findings indicate the growth suppressive effects of doxorubicin, oxaliplatin and methotrexate treatment on stomach carcinoma cells and the implication of BCL2L12, BAX, and BCL-2 expression profiles in the molecular signaling pathways triggered by chemotherapy.
Collapse
Affiliation(s)
- Dimitra Florou
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Athens, Athens, Greece
| | | | | | | |
Collapse
|
47
|
Shah MA, Wainberg ZA, Catenacci DVT, Hochster HS, Ford J, Kunz P, Lee FC, Kallender H, Cecchi F, Rabe DC, Keer H, Martin AM, Liu Y, Gagnon R, Bonate P, Liu L, Gilmer T, Bottaro DP. Phase II study evaluating 2 dosing schedules of oral foretinib (GSK1363089), cMET/VEGFR2 inhibitor, in patients with metastatic gastric cancer. PLoS One 2013; 8:e54014. [PMID: 23516391 PMCID: PMC3597709 DOI: 10.1371/journal.pone.0054014] [Citation(s) in RCA: 162] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2012] [Accepted: 12/05/2012] [Indexed: 01/01/2023] Open
Abstract
PURPOSE The receptors for hepatocyte and vascular endothelial cell growth factors (MET and VEGFR2, respectively) are critical oncogenic mediators in gastric adenocarcinoma. The purpose is to examine the safety and efficacy of foretinib, an oral multikinase inhibitor targeting MET, RON, AXL, TIE-2, and VEGFR2 receptors, for the treatment of metastatic gastric adenocarcinoma. PATIENTS AND METHODS Foretinib safety and tolerability, and objective response rate (ORR) were evaluated in patients using intermittent (240 mg/day, for 5 days every 2 weeks) or daily (80 mg/day) dosing schedules. Thirty evaluable patients were required to achieve alpha = 0.10 and beta = 0.2 to test the alternative hypothesis that single-agent foretinib would result in an ORR of ≥ 25%. Up to 10 additional patients could be enrolled to ensure at least eight with MET amplification. Correlative studies included tumor MET amplification, MET signaling, pharmacokinetics and plasma biomarkers of foretinib activity. RESULTS From March 2007 until October 2009, 74 patients were enrolled; 74% male; median age, 61 years (range, 25-88); 93% had received prior therapy. Best response was stable disease (SD) in 10 (23%) patients receiving intermittent dosing and five (20%) receiving daily dosing; SD duration was 1.9-7.2 months (median 3.2 months). Of 67 patients with tumor samples, 3 had MET amplification, one of whom had SD. Treatment-related adverse events occurred in 91% of patients. Rates of hypertension (35% vs. 15%) and elevated aspartate aminotransferase (23% vs. 8%) were higher with intermittent dosing. In both patients with high baseline tumor phospho-MET (pMET), the pMET:total MET protein ratio decreased with foretinib treatment. CONCLUSION These results indicate that few gastric carcinomas are driven solely by MET and VEGFR2, and underscore the diverse molecular oncogenesis of this disease. Despite evidence of MET inhibition by foretinib, single-agent foretinib lacked efficacy in unselected patients with metastatic gastric cancer.
Collapse
Affiliation(s)
- Manish A Shah
- The Weill Cornell Medical College/New York-Presbyterian Hospital, New York, New York, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Rivera F, Grávalos C, García-Carbonero R. SEOM clinical guidelines for the diagnosis and treatment of gastric adenocarcinoma. Clin Transl Oncol 2012; 14:528-35. [PMID: 22721798 DOI: 10.1007/s12094-012-0836-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Gastric adenocarcinomas are tumours of decreasing incidence in the Western world, although they are still the fourth leading cause of cancer mortality. The purpose of these clinical guidelines is to provide recommendations for the diagnosis and treatment of this disease based on the best available evidence. Regarding resectable gastric cancer, the various potential therapeutic options are discussed (adjuvant or perioperative chemotherapy, and adjuvant or neoadjuvant chemoradiotherapy). With regard to advanced or metastatic disease, different alternative combinations of conventional cytotoxic agents including a platinum agent (cisplatin or oxaliplatin) and a fluoropyrimidine (5-FU, capecitabine or S1), with or without a third drug (epirubicin or docetaxel), as well as their integration with new biological agents (trastuzumab in HER2+ tumours), are discussed. Finally, an outline is provided of the main lines of research and development of therapies for this disease.
Collapse
Affiliation(s)
- Fernando Rivera
- Servicio de Oncología Médica, Hospital Universitario Marqués de Valdecilla, Santander, Spain.
| | | | | | | |
Collapse
|
49
|
Abstract
In the past, patients with advanced or metastatic gastric or gastroesophageal junction cancer have had few treatment options and generally poor survival rates. The human epidermal growth factor receptor 2 (HER2) has been identified as a potential therapeutic target because of its overexpression or gene amplification in 6% to 35% of gastric or gastroesophageal junction cancers, although the methods of assessment and prognostic value of HER2 have been subject to debate. The phase III Trastuzumab for Gastric Cancer (ToGA) trial showed that adding the HER2-targeted humanized monoclonal antibody trastuzumab to chemotherapy significantly improves survival without negatively impacting quality of life in patients with advanced gastric or gastroesophageal junction cancer. As a result, trastuzumab is now the sole HER2-targeted therapy approved in several countries for this indication. The ToGA trial also demonstrated that patients who expressed higher levels of HER2 (determined by immunohistochemical screening) received the greatest benefit from trastuzumab therapy. This finding underlines the importance of accurate HER2 testing. Because of the unique characteristics of gastric cancer, a new gastric cancer-specific scoring system for HER2 expression was proposed during the ToGA trial. The aim of this review is to inform the gastroenterologist of the potential role of HER2-targeted therapy, to discuss the importance of accurate and reliable HER2 testing, and to discuss ongoing studies with HER2-targeted therapies that may have an impact on the future treatment of HER2-positive gastric cancer.
Collapse
|
50
|
Unek IT, Unek T, Oztop I, Akman T, Atilla K, Ellidokuz H, Bora S, Sarioglu S, Yilmaz U. Bimonthly regimen of high-dose leucovorin, infusional 5-fluorouracil, epirubicin and cisplatin (modified ECF) as adjuvant chemotherapy in resected gastric adenocarcinoma. Chemotherapy 2012; 58:233-40. [PMID: 22832016 DOI: 10.1159/000339493] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2011] [Accepted: 05/14/2012] [Indexed: 12/23/2022]
Abstract
BACKGROUND The administration of the de Gramont regimen in combination with cisplatin and epirubicin (modified ECF) has previously been reported as a treatment for advanced gastric cancer, but here we report this regimen combination in an adjuvant setting for the first time. METHODS Forty-eight patients with curatively resected gastric cancer were treated. Each 2-week cycle consisted of epirubicin (50 mg/m(2)), cisplatin (50 mg/m(2)), 5-fluorouracil (5-FU) IV bolus (400 mg/m(2)) and 5-FU IV (2,400 mg/m(2)) over 46 h plus leucovorin IV (400 mg/m(2)) over 2 h. Postoperative chemoradiotherapy was also administered to the patients when indicated. We retrospectively reviewed the patients who were treated with modified ECF. RESULTS The median disease-free survival (DFS) was 40.7 months and the 1-, 3- and 5-year DFS rates were 78.5, 55.7 and 44.6%, respectively. The most common grade 3-4 toxicities were hematological and gastrointestinal. CONCLUSION A modified ECF regimen may be an effective and convenient treatment with tolerable toxicities for the adjuvant treatment of gastric cancer. It may provide an alternative regimen to the standard ECF when a continuous ambulatory infusion pump is not feasible or not preferred by the patient.
Collapse
Affiliation(s)
- Ilkay Tugba Unek
- Division of Medical Oncology, Department of Internal Medicine, Dokuz Eylul University School of Medicine, Izmir, Turkey.
| | | | | | | | | | | | | | | | | |
Collapse
|