1
|
El Mashad SN, Kandil MAEH, Talab TAEH, Saied Abd El Naby AEN, Sultan MM, Sohaib A, Hemida AS. Gastric Carcinoma with low ROR alpha, low E- Cadherin and High LAPTM4B Immunohistochemical Profile; is associated with unfavorable prognosis in Egyptian patients. J Immunoassay Immunochem 2024; 45:50-72. [PMID: 38031398 DOI: 10.1080/15321819.2023.2279639] [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: 12/01/2023]
Abstract
In view of multiplicity of carcinogenic pathways of gastric carcinoma (GC), poor survival and chemotherapy resistance, more analysis of these pathways is required for prediction of prognosis and developing new therapeutic targets. Knocking down of RORα; induces tumor cell proliferation and epithelial-mesenchymal transition (EMT). LAPTM4B has been suggested to be associated with EMT which promote tumor invasion. This work aimed to investigate prognostic role of RORα, LAPTM4B, and E-Cadherin expression in GC. This retrospective immunohistochemical study assesses the expression of RORα, LAPTM4B, and E-Cadherin in 73 primary gastric carcinomas. Low RORα and high LAPTM4B expression in GC cases were associated with unfavorable prognostic factors such as positive lymph nodes, and high tumor budding. E-Cadherin heterogeneous staining was associated with poor prognostic criteria, such as diffuse type GC and high tumor budding. Low RORα, high LAPTM4B, and heterogeneous E-Cadherin were the most common immunohistochemical profile in GC cases. Low RORα expression showed poor prognostic impact on overall patient survival. In conclusion, RORα and LAPTM4B may have crucial role in GC aggressiveness. The predominance of low RORα, high LAPTM4B, and heterogeneous or negative E-Cadherin immunohistochemical profile in GC cases with unfavorable pathological parameters suggested that this profile may predict tumor behavior.
Collapse
Affiliation(s)
| | | | | | | | - Mervat Mahmoud Sultan
- Pathology Department, National Liver Institute, Menoufia University, Shebin El Kom, Egypt
| | - Ahmed Sohaib
- Clinical Oncology& Nuclear medicine Department, Faculty of Medicine, Menoufia University, Shebin El Kom, Egypt
| | - Aiat Shaban Hemida
- Pathology Department, Faculty of Medicine, Menoufia University, Shebin El Kom, Egypt
| |
Collapse
|
2
|
Jung YJ, Woo JS, Hwang SH, Yang S, Kim SJ, Jhun J, Lee SY, Lee KH, Cho ML, Song KY. Effect of IL-10-producing B cells in peripheral blood and tumor tissue on gastric cancer. Cell Commun Signal 2023; 21:320. [PMID: 37946227 PMCID: PMC10634038 DOI: 10.1186/s12964-023-01174-5] [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: 03/08/2023] [Accepted: 05/25/2023] [Indexed: 11/12/2023] Open
Abstract
BACKGROUND Interleukin (IL)-10-producing B (B10) cells are generated in response to signals from the tumor microenvironment and promote tumor growth by interacting with B10 cells. We investigated the distributions of immune cells in peripheral blood and tumor tissue samples from patients with gastric cancer (GC). METHODS Patients with GC who underwent radical gastrectomy in Seoul St. Mary's Hospital between August 2020 and May 2021 were enrolled in this study. Forty-two samples of peripheral blood were collected, and a pair of gastric mucosal samples (normal and cancerous mucosa; did not influence tumor diagnosis or staging) was collected from each patient after surgery. B10 cells in peripheral blood and cancer mucosa samples were investigated by flow cytometry and immunofluorescence. AGS cells, gastric cancer cell line, were cultured with IL-10 and measured cell death and cytokine secretion. Also, AGS cells were co-cultured with CD19 + B cells and measured cytokine secretion. RESULTS The population of B10 cells was significantly larger in the blood of patients with GC compared with controls. In confocal images of gastric mucosal tissues, cancerous mucosa contained more B10 cells than normal mucosa. The population of B10 cells in cancerous mucosa increased with cancer stage. When AGS cells were cultured under cell-death conditions, cellular necrosis was significantly decreased, and proliferation was increased, for 1 day after IL-10 stimulation. Tumor necrosis factor (TNF)-α, IL-8, IL-1β, and vascular endothelial growth factor secretion by cancer cells was significantly increased by coculture of AGS cells with GC-derived CD19+ B cells. CONCLUSIONS B cells may be one of the populations that promote carcinogenesis by inducing the production of inflammatory mediators, such as IL-10, in GC. Targeting B10 cells activity could improve the outcomes of antitumor immunotherapy. Video Abstract.
Collapse
Affiliation(s)
- Yoon Ju Jung
- Division of Gastrointestinal Surgery, Department of Surgery, Yeouido St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, 07345, Korea
| | - Jin Seok Woo
- Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
- Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
| | - Sun-Hee Hwang
- Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
- Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
| | - SeungCheon Yang
- Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
- Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
| | - So Jung Kim
- Division of Gastrointestinal Surgery, Department of Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
| | - JooYeon Jhun
- Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
- Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
- Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
| | - Seung Yoon Lee
- Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
- Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
- Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
| | - Kun Hee Lee
- Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
- Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
- Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
| | - Mi-La Cho
- Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea.
- Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea.
- Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea.
- Department of Medical Life Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea.
| | - Kyo Young Song
- Division of Gastrointestinal Surgery, Department of Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea.
| |
Collapse
|
3
|
Han G, Sun C, Cui L, Huang Y, Yu L, Liu S, Tao M. Efficacy of the low dose apatinib plus deep hyperthermia as third-line or later treatment in HER-2 negative advanced gastric cancer. Pathol Oncol Res 2023; 29:1611114. [PMID: 37465317 PMCID: PMC10350525 DOI: 10.3389/pore.2023.1611114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 06/21/2023] [Indexed: 07/20/2023]
Abstract
Aim: To observe the efficacy of the low dose apatinib plus deep hyperthermia as third-line or later treatment for patients with human epidermal growth factor receptor 2 (HER-2) negative advanced gastric cancer. Methods: 80 eligible patients with HER-2 negative advanced gastric cancer admitted to Jingjiang People's Hospital Affiliated with Yangzhou University-from March 2021 to March 2022 were selected, and they were divided into the control group (n = 40, apatinib) and experimental group (n = 40, apatinib plus deep hyperthermia) on the basis of random number table method. The levels of serum carcinoembryonic antigen (CEA), carbohydrate antigen 199 (CA199), and vascular endothelial growth factor (VEGF) were monitored, and the efficacy of the two groups was analyzed by referring to Karnofsky performance status (KPS), overall survival (OS) and disease control rate (DCR) before and after treatment. Results: The levels of CEA, CA199, and VEGF in both groups were lower after treatment than before (p < 0.05), and lower (CEA: 8.85 ± 1.36 vs. 12.87 ± 1.23, CA199: 34.19 ± 4.68 vs. 50.11 ± 5.73, VEGF: 124.8 ± 18.03 vs. 205.9 ± 19.91) in the experimental group than in the control group (p < 0.05). The DCR and KPS of the patients in the experimental group were significantly higher (DCR: 62.50% vs. 40.00%; KPS: 83.25 ± 1.15 vs. 76.25 ± 1.17) than in the control group (p < 0.05). In survival analysis, patients with control group had shorter OS than the experimental group. (median 5.65 vs. 6.50 months; hazard ratio [HR], 1.63 [95% confidence interval (CI) 1.02-2.60], p = 0.0396). Conclusion: The application of low-dose apatinib plus deep hyperthermia for patients with HER-2 negative gastric cancer who failed second-line treatment should be a promising option.
Collapse
Affiliation(s)
- Guohu Han
- Department of Oncology, Dushu Lake Hospital Affiliated to Soochow University, Suzhou, Jiangsu, China
- Department of Oncology, Jingjiang People’s Hospital, The Seventh Affiliated Hospital of Yangzhou University, Jingjiang, Jiangsu, China
| | - Changchun Sun
- Department of Oncology, Jingjiang People’s Hospital, The Seventh Affiliated Hospital of Yangzhou University, Jingjiang, Jiangsu, China
| | - Lihua Cui
- Department of Oncology, Jingjiang People’s Hospital, The Seventh Affiliated Hospital of Yangzhou University, Jingjiang, Jiangsu, China
| | - Yufeng Huang
- Department of Oncology, Jingjiang People’s Hospital, The Seventh Affiliated Hospital of Yangzhou University, Jingjiang, Jiangsu, China
| | - Lijiang Yu
- Department of Oncology, Jingjiang People’s Hospital, The Seventh Affiliated Hospital of Yangzhou University, Jingjiang, Jiangsu, China
| | - Shenzha Liu
- Department of Oncology, Jingjiang People’s Hospital, The Seventh Affiliated Hospital of Yangzhou University, Jingjiang, Jiangsu, China
| | - Min Tao
- Department of Oncology, Dushu Lake Hospital Affiliated to Soochow University, Suzhou, Jiangsu, China
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
4
|
McDonald HG, Harper MM, Hill K, Gao A, Solomon AL, Bailey CJ, Lin M, Barry-Hundeyin M, Cavnar MJ, Mardini SH, Pandalai PJ, Patel RA, Kolesar JM, Rueckert JA, Hookey L, Ropeleski M, Merchant SJ, Kim J, Gao M. Creation of EGD-Derived Gastric Cancer Organoids to Predict Treatment Responses. Cancers (Basel) 2023; 15:3036. [PMID: 37296998 PMCID: PMC10252567 DOI: 10.3390/cancers15113036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 05/25/2023] [Accepted: 05/31/2023] [Indexed: 06/12/2023] Open
Abstract
Gastric adenocarcinoma (GAd) is the third leading cause of cancer-related deaths worldwide. Most patients require perioperative chemotherapy, yet methods to accurately predict responses to therapy are lacking. Thus, patients may be unnecessarily exposed to considerable toxicities. Here, we present a novel methodology using patient-derived organoids (PDOs) that rapidly and accurately predicts the chemotherapy efficacy for GAd patients. Methods:Endoscopic GAd biopsies were obtained from 19 patients, shipped overnight, and PDOs were developed within 24 h. Drug sensitivity testing was performed on PDO single-cells with current standard-of-care systemic GAd regimens and cell viability was measured. Whole exome sequencing was used to confirm the consistency of tumor-related gene mutations and copy number alterations between primary tumors, PDOs, and PDO single-cells. Results:Overall, 15 of 19 biopsies (79%) were appropriate for PDO creation and single-cell expansion within 24 h of specimen collection and overnight shipment. With our PDO single-cell technique, PDOs (53%) were successfully developed. Subsequently, two PDO lines were subjected to drug sensitivity testing within 12 days from initial biopsy procurement. Drug sensitivity assays revealed unique treatment response profiles for combination drug regimens in both of the two unique PDOs, which corresponded with the clinical response. Conclusions:The successful creation of PDOs within 24 h of endoscopic biopsy and rapid drug testing within 2 weeks demonstrate the feasibility of our novel approach for future applications in clinical decision making. This proof of concept sets the foundation for future clinical trials using PDOs to predict clinical responses to GAd therapies.
Collapse
Affiliation(s)
- Hannah G. McDonald
- Division of Surgical Oncology, Department of General Surgery, University of Kentucky, Lexington, KY 40536, USA; (H.G.M.); (M.G.)
| | - Megan M. Harper
- Division of Surgical Oncology, Department of General Surgery, University of Kentucky, Lexington, KY 40536, USA; (H.G.M.); (M.G.)
| | - Kristen Hill
- College of Pharmacy, University of Kentucky, Lexington, KY 40536, USA
| | - Anqi Gao
- Division of Surgical Oncology, Department of General Surgery, University of Kentucky, Lexington, KY 40536, USA; (H.G.M.); (M.G.)
| | - Angelica L. Solomon
- Division of Surgical Oncology, Department of General Surgery, University of Kentucky, Lexington, KY 40536, USA; (H.G.M.); (M.G.)
| | - Charles J. Bailey
- Division of Surgical Oncology, Department of General Surgery, University of Kentucky, Lexington, KY 40536, USA; (H.G.M.); (M.G.)
| | - Miranda Lin
- Division of Surgical Oncology, Department of General Surgery, University of Kentucky, Lexington, KY 40536, USA; (H.G.M.); (M.G.)
| | - Mautin Barry-Hundeyin
- Division of Surgical Oncology, Department of General Surgery, University of Kentucky, Lexington, KY 40536, USA; (H.G.M.); (M.G.)
| | - Michael J. Cavnar
- Division of Surgical Oncology, Department of General Surgery, University of Kentucky, Lexington, KY 40536, USA; (H.G.M.); (M.G.)
| | - Samuel H. Mardini
- Division of Gastroenterology, Department of Internal Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Prakash J. Pandalai
- Division of Surgical Oncology, Department of General Surgery, University of Kentucky, Lexington, KY 40536, USA; (H.G.M.); (M.G.)
| | - Reema A. Patel
- Division of Medical Oncology, Department of Internal Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Jill M. Kolesar
- College of Pharmacy, University of Kentucky, Lexington, KY 40536, USA
| | - Justin A. Rueckert
- Department of Pathology and Laboratory Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Lawrence Hookey
- Division of Gastroenterology, Department of Internal Medicine, Queen’s University, Kingston, ON K7L 3N6, Canada
| | - Mark Ropeleski
- Division of Gastroenterology, Department of Internal Medicine, Queen’s University, Kingston, ON K7L 3N6, Canada
| | - Shaila J. Merchant
- Division of General Surgery and Surgical Oncology, Queen’s University, Kingston, ON K7L 3N6, Canada
| | - Joseph Kim
- Division of Surgical Oncology, Department of General Surgery, University of Kentucky, Lexington, KY 40536, USA; (H.G.M.); (M.G.)
| | - Mei Gao
- Division of Surgical Oncology, Department of General Surgery, University of Kentucky, Lexington, KY 40536, USA; (H.G.M.); (M.G.)
| |
Collapse
|
5
|
Yu P, Zhu S, Pu Y, Cai B, Ma X, Zhang C. Efficacy and safety evaluation of PSOX, DOF and SOX regimens as neoadjuvant chemotherapy for advanced gastric cancer. Future Oncol 2023; 18:4483-4492. [PMID: 36916454 DOI: 10.2217/fon-2022-0337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/15/2023] Open
Abstract
Aim: To explore the efficacy and safety of paclitaxel+oxaliplatin+S-1 (PSOX), docetaxel+oxaliplatin+fluorouracil (DOF) and oxaliplatin+S-1 (SOX) regimens as neoadjuvant chemotherapy for advanced gastric cancer (GC). Methods: A retrospective analysis was used in 306 patients with GC who underwent neoadjuvant chemotherapy, consisting of 102 from the PSOX group, 100 from the DOF group and 104 from the SOX group. Results: The total effective rates and disease control rates for the PSOX, DOF and SOX groups were 31.4, 18 and 16.3% and 96.1, 94 and 92.3%, respectively. The highest total effective rate and disease control rate were found in the PSOX groups. Moreover, no difference among the PSOX, DOF and SOX groups on the incidence of adverse events was observed (p > 0.05). Conclusion: The PSOX regimen is an alternative neoadjuvant chemotherapy regimen for GC patients.
Collapse
Affiliation(s)
- Pengjie Yu
- Department of Gastrointestinal Oncology Surgery, The Affiliated Hospital of Qinghai University, 810000, Qinghai, China
| | - Shengmao Zhu
- Department of Gastrointestinal Oncology Surgery, The Affiliated Hospital of Qinghai University, 810000, Qinghai, China
| | - Yongqiang Pu
- Department of Gastrointestinal Oncology Surgery, The Affiliated Hospital of Qinghai University, 810000, Qinghai, China
| | - Baojia Cai
- Department of Gastrointestinal Oncology Surgery, The Affiliated Hospital of Qinghai University, 810000, Qinghai, China
| | - Xiaoming Ma
- Department of Gastrointestinal Oncology Surgery, The Affiliated Hospital of Qinghai University, 810000, Qinghai, China
| | - Chengwu Zhang
- Department of Gastrointestinal Oncology Surgery, The Affiliated Hospital of Qinghai University, 810000, Qinghai, China
| |
Collapse
|
6
|
Degu A, Karimi PN, Opanga SA, Nyamu DG. Predictors of survival outcomes among patients with gastric cancer in a leading tertiary, teaching and referral hospital in Kenya. Cancer Med 2023; 12:4147-4160. [PMID: 36172986 PMCID: PMC9972118 DOI: 10.1002/cam4.5275] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 09/07/2022] [Accepted: 09/09/2022] [Indexed: 11/12/2022] Open
Abstract
INTRODUCTION The incidence of gastrointestinal malignancies in Kenya is increasing, although there is a paucity of data on survival outcomes among gastric cancer patients. Hence, this study aimed to assess survival outcomes among adult gastric cancer patients at Kenyatta National Hospital. METHODS A retrospective cohort study design was used to assess the survival outcomes among 247 gastric cancer patients. All medical records of adult (≥18 years) gastric cancer patients with complete medical records of diagnosis, stage of cancer, and treatment regimen in the study setting in the last 5 years (2016-2020) were included. A simple random sampling technique was employed to select the study participants. Data were collected using a data abstraction tool composed of socio-demographic and clinical characteristics. Survival outcomes were reported as the percentage of mortality, mean survival estimate, and mean cancer-specific survival. The data were entered and analyzed using version 20.0 SPSS statistical software. The mean survival estimates and predictors of mortality were computed using the Kaplan-Meier and Cox regression analysis. RESULTS The study showed that 33.3% (64) had new distant metastasis, and 42.1% (104) had disease progression. Besides, the mortality rate was high (33.6%), and 14.6% and 7.7% of patients had complete and partial responses, respectively. The five-year survival was 32.7% among gastric cancer patients. Comorbidity (p = 0.014), advanced-stage diseases (p = 0.03), chemotherapy (p = 0.008), and gastrectomy (p = 0.016) were significant determinants of mortality. CONCLUSIONS A significant proportion of patients had distant metastasis, disease progression, and a low five-year survival rate. Hence, early cancer-screening programs are indispensable to circumvent disease progression and improve survival outcomes.
Collapse
Affiliation(s)
- Amsalu Degu
- Department of Pharmaceutics and Pharmacy Practice, School of Pharmacy and Health Sciences, United States International University-Africa, Nairobi, Kenya.,Department of Pharmacy, Faculty of Health Sciences, University of Nairobi, Nairobi, Kenya
| | - Peter N Karimi
- Department of Pharmacy, Faculty of Health Sciences, University of Nairobi, Nairobi, Kenya
| | - Sylvia A Opanga
- Department of Pharmacy, Faculty of Health Sciences, University of Nairobi, Nairobi, Kenya
| | - David G Nyamu
- Department of Pharmacy, Faculty of Health Sciences, University of Nairobi, Nairobi, Kenya
| |
Collapse
|
7
|
Mashad SNE, Kandil MAE, Talab TAE, Naby AENSAE, Sultan MM, Sohaib A, Hemida AS. Gastric Carcinoma with low ROR alpha, low E- Cadherin and High LAPTM4B Immunohistochemical Profile; is associated with poor prognosis in Egyptian patients.. [DOI: 10.21203/rs.3.rs-2123133/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
Abstract
Abstract
Background
Gastric carcinoma (GC) is the tenth most prevalent cancer in both sexes in Egypt. Many pathways have been investigated regarding pathogenesis of GC, including epithelial-mesenchymal transition (EMT) pathway. In view of multiplicity of carcinogenic pathways, poor survival and chemotherapy resistance detected in GC patients, more analysis of these pathways is required for better molecular selection of patients, prediction of prognosis and developing new therapeutic targets. Down-regulation of E-Cadherin is an important EMT stage. RORα is a tumor suppressor gene, expressed in normal epithelial tissues and reduced in a variety of human cancers. Knocking down of RORα; increase cell proliferation, EMT, migration, and invasion. LAPTM4B is a protooncogene and it has been suggested to be strictly associated with EMT induction. Therefore, this work aims to investigate the role of RORα, LAPTM4B and E-Cadherin and its relationship to prognosis of GC.
Methods
This is a retrospective study where the standard immunohistochemical technique was done to assess the expression of RORα, LAPTM4B and E-Cadherin in 167 cases of chronic gastritis (control group) and 73 primary gastric carcinomas (51 of them have available adjacent non tumor tissue).
Results
Low RORα and high LAPTM4B expression in GC cases were associated with unfavorable prognostic factors such as positive lymph nodes, and high tumor budding. E-Cadherin Heterogeneous staining was associated with poor prognostic pathological criteria, such as diffuse type GC and high tumor budding. In GC, there was significant co parallel correlation between RORα and E-Cadherin expression while LAPTM4B showed inverse correlation with E-Cadherin expression. Low RORα, high LAPTM4B, and negative or heterogeneous E-Cadherin were the most common immunohistochemical profile in GC cases. Low RORα expression showed poor prognostic impact on overall patient survival.
Conclusions
Low RORα H-score and increased expression of LAPTM4B were significantly associated with unfavorable prognostic parameters of GC which may indicate their crucial role in tumor aggressiveness. The predominance of low RORα, high LAPTM4B and heterogeneous or negative E-Cadherin immunohistochemical profile in GC cases with unfavorable pathological parameters suggested that this profile may predict tumor behavior and this profile could be linked to EMT molecular subtype of GC
Collapse
|
8
|
Xue C, Xu YH. Trastuzumab combined chemotherapy for the treatment of HER2-positive advanced gastric cancer: A systematic review and meta-analysis of randomized controlled trial. Medicine (Baltimore) 2022; 101:e29992. [PMID: 36042610 PMCID: PMC9410626 DOI: 10.1097/md.0000000000029992] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND This systematic review and meta-analysis aimed to assess the efficacy of trastuzumab combined with chemotherapy for the treatment in HER2-positive advanced gastric cancer (HER2-PAGC). METHODS This systematic review and meta-analysis was designed using randomized controlled trials that compared trastuzumab in combination with chemotherapy and chemotherapy alone. A comprehensive search was conducted in the following databases from their inception onwards: PubMed, EMBASE, Cochrane Library, WANGFANG, and CNKI. We also searched other literature sources to avoid missing relevant studies. Two reviewers independently performed all record selection, data collection, and methodological assessments. Any confusion was resolved by discussion or referral to a third reviewer. If there were ample data from eligible studies, we performed a fixed-effects meta-analysis. Whenever this was not possible, we conducted a narrative synthesis. RESULTS Meta-analysis results showed that trastuzumab in combination with chemotherapy achieved better outcomes on response rate (trastuzumab plus CFC vs CFC: odds ratio [OR] = 1.56, 95% confidence interval [CI] [1.17-2.09], I2 = 0%, P < .003; trastuzumab plus OT vs OT: OR = 2.97, 95% CI [1.74-5.09], I2 = 0%, P < .0001; and trastuzumab plus CC vs CC: OR = 2.62, 95% CI [1.84-3.73], I2 = 0%, P < .0001), and disease control rate (trastuzumab plus CFC vs CFC: OR = 1.61, 95% CI [1.17-2.21], I2 = 0%, P = .004; trastuzumab plus OT vs OT: OR = 4.29, 95% CI [2.33-7.90], I2 = 0%, P < .0001; and trastuzumab plus CC vs CC: OR = 2.99, 95% CI [1.99-4.48], I2 = 0%, P < .0001). However, there were no significant differences in the adverse events. CONCLUSIONS The results of this study revealed that the efficacy of trastuzumab combined with chemotherapy was superior to that of chemotherapy alone for the treatment of HER2-PAGC. The 2 modalities showed similar safety profiles.
Collapse
Affiliation(s)
- Chuan Xue
- Department of Gastroenterology, Sunshine Union Hospital, Weifang, China
| | - Yong-Hong Xu
- Department of Gastroenterology, Sunshine Union Hospital, Weifang, China
- *Correspondence: Yong-Hong Xu, MM, Department of Gastroenterology, Sunshine Union Hospital, No. 9000 Yingqian Street, High-tech District, Weifang, Shandong Province 261061, China (e-mail: )
| |
Collapse
|
9
|
Wang J, Kunzke T, Prade VM, Shen J, Buck A, Feuchtinger A, Haffner I, Luber B, Liu DHW, Langer R, Lordick F, Sun N, Walch A. Spatial metabolomics identifies distinct tumor-specific subtypes in gastric cancer patients. Clin Cancer Res 2022; 28:2865-2877. [PMID: 35395077 DOI: 10.1158/1078-0432.ccr-21-4383] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 03/01/2022] [Accepted: 04/06/2022] [Indexed: 11/16/2022]
Abstract
PURPOSE Current systems of gastric cancer (GC) molecular classification include genomic, molecular, and morphological features. GC classification based on tissue metabolomics remains lacking. This study aimed to define metabolically distinct GC subtypes and identify their clinicopathological and molecular characteristics. EXPERIMENTAL DESIGN Spatial metabolomics by high mass resolution imaging mass spectrometry was performed in 362 GC patients. K-means clustering was used to define tumor and stroma-related subtypes based on tissue metabolites. The identified subtypes were linked with clinicopathological characteristics, molecular features, and metabolic signatures. Responses to trastuzumab treatment were investigated across the subtypes by introducing an independent patient cohort with HER2-positive GC from a multicenter observational study. RESULTS Three tumor- and three stroma-specific subtypes with distinct tissue metabolite patterns were identified. Tumor-specific subtype T1(HER2+MIB+CD3+) positively correlated with HER2, MIB1, DEFA-1, CD3, CD8, FOXP3, but negatively correlated with MMR. Tumor-specific subtype T2(HER2-MIB-CD3-) negatively correlated with HER2, MIB1, CD3, FOXP3, but positively correlated with MMR. Tumor-specific subtype T3(pEGFR+) positively correlated with pEGFR. Patients with tumor subtype T1(HER2+MIB+CD3+) had elevated nucleotide levels, enhanced DNA metabolism, and a better prognosis than T2(HER2-MIB-CD3-) and T3(pEGFR+). An independent validation cohort confirmed that the T1 subtype benefited from trastuzumab therapy. Stroma-specific subtypes had no association with clinicopathological characteristics, however linked to distinct metabolic pathways and molecular features. CONCLUSIONS Patient subtypes derived by tissue-based spatial metabolomics are a valuable addition to existing GC molecular classification systems. Metabolic differences between the subtypes and their associations with molecular features could provide a valuable tool to aid in selecting specific treatment approaches.
Collapse
Affiliation(s)
- Jun Wang
- Research Unit Analytical Pathology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
| | - Thomas Kunzke
- Research Unit Analytical Pathology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
| | - Verena M Prade
- Research Unit Analytical Pathology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
| | - Jian Shen
- Research Unit Analytical Pathology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
| | - Achim Buck
- Research Unit Analytical Pathology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
| | - Annette Feuchtinger
- Research Unit Analytical Pathology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
| | - Ivonne Haffner
- University Cancer Center Leipzig (UCCL), Leipzig University Medical Center, Leipzig, Germany
| | - Birgit Luber
- Technische Universität München, Fakultät für Medizin, Klinikum rechts der Isar, Institut für Allgemeine Pathologie und Pathologische Anatomie, München, Germany
| | - Drolaiz H W Liu
- Department of Pathology, GROW School for Oncology and Reproduction, Maastricht University Medical Center, Maastricht, the Netherlands
- Institute of Clinical Pathology and Molecular Pathology, Kepler University Hospital and Johannes Kepler University, Linz, Austria
| | - Rupert Langer
- Institute of Clinical Pathology and Molecular Pathology, Kepler University Hospital and Johannes Kepler University, Linz, Austria
| | - Florian Lordick
- University Cancer Center Leipzig (UCCL), Leipzig University Medical Center, Leipzig, Germany
- Department of Oncology, Gastroenterology, Hepatology, Pulmonology and Infectious Diseases, Leipzig University Medical Center, Leipzig, Germany
| | - Na Sun
- Research Unit Analytical Pathology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
| | - Axel Walch
- Research Unit Analytical Pathology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
| |
Collapse
|
10
|
TANG J, YANG J, HE J, XIE J, WANG P, WEI S. Assessment of the efficacy and safety of intraperitoneal chemotherapy in patients with advanced gastric cancer in Chinese population: a meta-analysis. FOOD SCIENCE AND TECHNOLOGY 2022. [DOI: 10.1590/fst.34321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Jin TANG
- Affiliated Hospital of North Sichuan Medical College, China
| | | | - Jinsong HE
- Affiliated Hospital of North Sichuan Medical College, China
| | - Jiebin XIE
- Affiliated Hospital of North Sichuan Medical College, China
| | - Pan WANG
- Affiliated Hospital of North Sichuan Medical College, China
| | - Shoujiang WEI
- Affiliated Hospital of North Sichuan Medical College, China
| |
Collapse
|
11
|
Lin C, Fan H, Chen W, Cui L. Palliative Gastrectomy vs. Gastrojejunostomy for Advanced Gastric Cancer: A Systematic Review and Meta-Analysis. Front Surg 2021; 8:723065. [PMID: 34901136 PMCID: PMC8661416 DOI: 10.3389/fsurg.2021.723065] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 10/26/2021] [Indexed: 12/24/2022] Open
Abstract
Background: Advanced gastric cancer is the fifth leading cause of cancer-related deaths. Patients with metastatic advanced gastric cancer commonly develop a gastric outlet obstruction that considerably worsens their quality of life. Surgical interventions such as gastrojejunostomy and palliative gastrectomy are commonly administered to alleviate this obstruction. However, whether one intervention is better than another at improving morbidity- and mortality-related outcomes is unclear. Thus, in this meta-analysis, we compare outcomes of palliative gastrectomy and gastrojejunostomy (overall hospital stay length, time to oral intake, survival, and complication rates) in patients with metastatic advanced gastric cancer to identify the best procedure. Objective: To compare morbidity and mortality outcomes of palliative gastrectomy and gastrojejunostomy in patients with metastatic advanced gastric cancer. Methods: We followed the PRISMA guidelines to systematically search Web of Science, EMBASE, CENTRAL, Scopus, and MEDLINE for relevant studies. We conducted a random-effects meta-analysis to find differential outcomes between palliative gastrectomy and gastrojejunostomy among variables such as time to oral intake, overall hospital stay length, complication rates, and survival in patients with metastatic advanced gastric cancer. Results: From 963 studies, we found 7 eligible studies with 642 patients (70.3 ± 4.7 years) who had undergone palliative gastrectomy or gastrojejunostomy. Our meta-analysis revealed an insignificant (p > 0.05) differences in terms of overall survival duration (Hedge's g, 1.22), complication risks (odds ratio, 1.35), and time to oral intake (g, 0.62) and hospital stay length (g, 0.12) between patients undergoing gastrojejunostomy and palliative gastrectomy. Conclusion: In this present study we observed no statistically significant differences in terms of morbidity and mortality outcomes after palliative gastrectomy and gastrojejunostomy in patients with metastatic advanced gastric cancer. Therefore, no conclusions can be drawn for the variables evaluated. This study provides a preliminary overview of the risks associated with gastrojejunostomy and palliative gastrectomy to help gastroenterologists manage patients with metastatic advanced-stage gastric cancer.
Collapse
Affiliation(s)
- Chunfang Lin
- Department of General Surgery, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Haibo Fan
- Department of Targeted Therapy, Shanxi Cancer Hospital, Taiyuan, China
| | - Wenjun Chen
- Department of General Surgery, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Lingzhi Cui
- Department of Targeted Therapy, Shanxi Cancer Hospital, Taiyuan, China
| |
Collapse
|
12
|
Li Z, Jia Y, Zhu H, Xing X, Pang F, Shan F, Li S, Wang D, Zhao F, Ma T, Wang S, Ji J. Tumor mutation burden is correlated with response and prognosis in microsatellite-stable (MSS) gastric cancer patients undergoing neoadjuvant chemotherapy. Gastric Cancer 2021; 24:1342-1354. [PMID: 34406546 DOI: 10.1007/s10120-021-01207-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 06/22/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND Neoadjuvant chemotherapy (NACT) before radical gastrectomy is preferred for locally advanced gastric cancer (GC). However, clinical practices demonstrate that a considerable proportion of GC patients do not benefit from NACT, largely due to the lack of biomarkers for patient selection and prognosis prediction. A recent study revealed that patients with microsatellite instability-high (MSI-H) may be resistant to NACT, however, most tumors in Chinese GC patients (~ 95%) are characterized by microsatellite stability (MSS). Here, we aimed to discover new molecular biomarkers for this larger population. METHODS We performed whole-exome sequencing on 46 clinical samples (pre- and post-treatment) from 30 stage II/III MSS GC patients whose response to NACT was rigorously defined. Serum tumor markers (TMs), including AFP, CEA, CA199, CA724 and CA242 were measured during the course. RESULTS High tumor mutation burden (TMB-H) and 19q12 amplification (19q12 +) were positively associated with the NACT response. When TMB and 19q12 amplification were jointly analyzed, those with TMB-H or 19q12 + showed favorable response to NACT (p = 0.035). Further, TMB-H was negatively correlated with ypN stage, lymph node metastasis, and macrophage infiltration. Patients with TMB-H showed better disease-free survival (DFS) than those with TMB-L (P = 0.025, HR = 0.1331), and this was further validated using two larger GC datasets: TCGA-STAD (p = 0.004) and ICGC-CN (p = 0.045). CONCLUSION The combination of TMB-H and 19q12 + can serve as an early indicator of response to NACT. Superior to traditional clinical indicators, TMB-H is a robust and easily accessible candidate biomarker associated with better DFS, and can be evaluated at the time of diagnosis.
Collapse
Affiliation(s)
- Ziyu Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery, Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Yongning Jia
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery, Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Honglin Zhu
- Genetron Health (Beijing) Technology, Co. Ltd, Beijing, 102206, China
| | - Xiaofang Xing
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Center for Molecular Diagnostics, Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Fei Pang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery, Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Fei Shan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery, Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Shuangxi Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery, Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Danhua Wang
- Genetron Health (Beijing) Technology, Co. Ltd, Beijing, 102206, China
| | - Fangping Zhao
- Genetron Health (Beijing) Technology, Co. Ltd, Beijing, 102206, China
| | - Tonghui Ma
- Genetron Health (Beijing) Technology, Co. Ltd, Beijing, 102206, China
| | - Sizhen Wang
- Genetron Health (Beijing) Technology, Co. Ltd, Beijing, 102206, China
| | - Jiafu Ji
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery, Peking University Cancer Hospital and Institute, Beijing, 100142, China.
| |
Collapse
|
13
|
Kronenfeld JP, Collier AL, Turgeon MK, Ju M, Alterio R, Wang A, Fernandez M, Porembka MR, Richter H, Lee AY, Russell MC, Merchant NB, Maker AV, Datta J. Attrition during neoadjuvant chemotherapy for gastric adenocarcinoma is associated with decreased survival: A United States Safety-Net Collaborative analysis. J Surg Oncol 2021; 124:1317-1328. [PMID: 34379324 DOI: 10.1002/jso.26638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 07/25/2021] [Accepted: 07/26/2021] [Indexed: 11/07/2022]
Abstract
BACKGROUND Neoadjuvant chemotherapy (NAC) is standard management for localized gastric cancer (GC). Attrition during NAC due to treatment-related toxicity or functional decline is considered a surrogate for worse biologic outcomes; however, data supporting this paradigm are lacking. We investigated factors predicting attrition and its association with overall survival (OS) in GC. METHODS Patients with nonmetastatic GC initiating NAC were identified from the US Safety-Net Collaborative (2012-2014). Patient/treatment-related characteristics were compared between attrition/nonattrition cohorts. Cox models determined factors associated with OS. RESULTS Of 116 patients initiating NAC, attrition during prescribed NAC occurred in 24%. No differences were observed in performance status, comorbidities, treatment at safety-net hospital, or clinicopathologic factors between cohorts. Despite absence of distinguishing factors, attrition was associated with worse OS (median: 11 vs. 37 months; p = 0.01) and was an independent predictor of mortality (hazard ratio [HR]: 4.7, 95% confidence interval [CI]: 1.5-15.2; p = 0.02). Fewer patients with attrition underwent curative-intent surgery (39% vs. 89%; p < 0.001). Even in patients undergoing surgical exploration (n = 89), NAC attrition remained an independent predictor of worse OS (HR: 50.8, 95% CI: 3.6-717.8; p = 0.004) despite similar receipt of adjuvant chemotherapy. CONCLUSION Attrition during NAC for nonmetastatic GC is independently associated with worse OS, even in patients undergoing surgery. Attrition during NAC may reflect unfavorable tumor biology not captured by conventional staging metrics.
Collapse
Affiliation(s)
- Joshua P Kronenfeld
- Division of Surgical Oncology, Department of Surgery, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Amber L Collier
- Division of Surgical Oncology, Department of Surgery, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Michael K Turgeon
- Division of Surgical Oncology, Department of Surgery, Winship Cancer Institute, Emory University, Atlanta, Georgia, USA
| | - Michelle Ju
- Division of Surgical Oncology, Department of Surgery, University of Texas Southwestern Medical School, Dallas, Texas, USA
| | - Rodrigo Alterio
- Division of Surgical Oncology, Department of Surgery, University of Texas Southwestern Medical School, Dallas, Texas, USA
| | - Annie Wang
- Division of Surgical Oncology, Department of Surgery, NYU Langone Health, New York, New York, USA
| | - Manuel Fernandez
- Division of Surgical Oncology, Department of Surgery, University of Illinois College of Medicine, Chicago, Illinois, USA
| | - Matthew R Porembka
- Division of Surgical Oncology, Department of Surgery, University of Texas Southwestern Medical School, Dallas, Texas, USA
| | - Harry Richter
- Division of Surgical Oncology, Department of Surgery, University of Illinois College of Medicine, Chicago, Illinois, USA
| | - Ann Y Lee
- Division of Surgical Oncology, Department of Surgery, NYU Langone Health, New York, New York, USA
| | - Maria C Russell
- Division of Surgical Oncology, Department of Surgery, Winship Cancer Institute, Emory University, Atlanta, Georgia, USA
| | - Nipun B Merchant
- Division of Surgical Oncology, Department of Surgery, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Ajay V Maker
- Division of Surgical Oncology, Department of Surgery, University of California San Francisco, San Francisco, California, USA
| | - Jashodeep Datta
- Division of Surgical Oncology, Department of Surgery, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida, USA
| |
Collapse
|
14
|
Ryu HR, Lee DW, Choi HJ, Kim JH, Ahn H. Scalp metastasis of advanced gastric cancer. Arch Craniofac Surg 2021; 22:157-160. [PMID: 34225408 PMCID: PMC8257450 DOI: 10.7181/acfs.2021.00129] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 04/20/2021] [Indexed: 12/22/2022] Open
Abstract
Head and neck cutaneous metastasis of advanced gastric cancer is uncommon, and scalp metastasis is particularly rare. We present the case of a 60-year-old man who was diagnosed with cutaneous metastasis on the scalp originating from advanced gastric cancer. The patient was referred to the plastic surgery department for a scalp mass near the hairline. He had been diagnosed with advanced gastric cancer and undergone total gastrectomy and Roux esophagojejunostomy 3 years previously. The differential diagnosis for a single flesh-colored nodule on the scalp included benign tumors such as epidermal cyst or lipoma; therefore, the patient underwent excision and biopsy. In the operative field, the mass was found to be located in the frontalis muscle. The biopsy result showed that the mass was a metastatic lesion of advanced gastric cancer. Whole-body computed tomography revealed a gastric tumor with blood vessel infiltration, peritoneal carcinomatosis, liver metastasis, and multiple disseminated subcutaneous metastases. Although scalp metastasis originating from an internal organ is extremely rare, plastic surgeons should always consider a metastatic lesion in the differential diagnosis if a patient with a scalp lesion has a history of malignant cancer.
Collapse
Affiliation(s)
- Hyeong Rae Ryu
- Department of Plastic and Reconstructive Surgery, Soonchunhyang University College of Medicine, Cheonan, Korea
| | - Da Woon Lee
- Department of Plastic and Reconstructive Surgery, Soonchunhyang University College of Medicine, Cheonan, Korea
| | - Hwan Jun Choi
- Department of Plastic and Reconstructive Surgery, Soonchunhyang University College of Medicine, Cheonan, Korea
| | - Jun Hyuk Kim
- Department of Plastic and Reconstructive Surgery, Soonchunhyang University College of Medicine, Cheonan, Korea
| | - Hyein Ahn
- Department of Pathology, Soonchunhyang University College of Medicine, Cheonan, Korea
| |
Collapse
|
15
|
Rabben HL, Kodama Y, Nakamura M, Bones AM, Wang TC, Chen D, Zhao CM, Øverby A. Chemopreventive Effects of Dietary Isothiocyanates in Animal Models of Gastric Cancer and Synergistic Anticancer Effects With Cisplatin in Human Gastric Cancer Cells. Front Pharmacol 2021; 12:613458. [PMID: 33897415 PMCID: PMC8060630 DOI: 10.3389/fphar.2021.613458] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 02/09/2021] [Indexed: 12/15/2022] Open
Abstract
Naturally occurring isothiocyanates (ITCs) from edible vegetables have shown potential as chemopreventive agents against several types of cancer. The aims of the present study were to study the potential of ITCs in chemoprevention and in potentiating the efficacy of cytotoxic drugs in gastric cancer treatment. The chemoprevention was studied in chemically induced mouse model of gastric cancer, namely N-methyl-N-nitrosourea (MNU) in drinking water, and in a genetically engineered mouse model of gastric cancer (the so-called INS-GAS mice). The pharmacological effects of ITCs with or without cisplatin were studied in human gastric cell lines MKN45, AGS, MKN74 and KATO-III, which were derived from either intestinal or diffused types of gastric carcinoma. The results showed that dietary phenethyl isothiocyanate (PEITC) reduced the tumor size when PEITC was given simultaneously with MNU, but neither when administrated after MNU nor in INS-GAS mice. Treatments of gastric cancer cells with ITCs resulted in a time- and concentration-dependent inhibition on cell proliferation. Pretreatment of gastric cancer cells with ITCs enhanced the inhibitory effects of cisplatin (but not 5-fluorouracil) in time- and concentration-dependent manners. Treatments of gastric cancer cells with PEITC plus cisplatin simultaneously at different concentrations of either PEITC or cisplatin exhibited neither additive nor synergetic inhibitory effect. Furthermore, PEITC depleted glutathione and induced G2/M cell cycle arrest in gastric cancer cells. In conclusion, the results of the present study showed that PEITC displayed anti-cancer effects, particularly when given before the tumor initiation, suggesting a chemopreventive effect in gastric cancer, and that pretreatment of PEITC potentiated the anti-cancer effects of cisplatin, possibly by reducing the intracellular pool of glutathione, suggesting a possible combination strategy of chemotherapy with pretreatment with PEITC.
Collapse
Affiliation(s)
- Hanne-Line Rabben
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.,The Central Norway Regional Health Authority, Stjørdal, Norway
| | - Yosuke Kodama
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Masahiko Nakamura
- Center for Clinical Pharmacy and Clinical Sciences, School of Pharmacy, Kitasato University, Tokyo, Japan
| | - Atle Magnar Bones
- Cell, Molecular Biology and Genomics Group, Department of Biology, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Timothy Cragin Wang
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.,Division of Digestive and Liver Diseases, Columbia University College of Physicians and Surgeons, New York, NY, United States
| | - Duan Chen
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Chun-Mei Zhao
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.,The Central Norway Regional Health Authority, Stjørdal, Norway
| | - Anders Øverby
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.,Center for Clinical Pharmacy and Clinical Sciences, School of Pharmacy, Kitasato University, Tokyo, Japan
| |
Collapse
|
16
|
Huang B, Mu P, Yu Y, Zhu W, Jiang T, Deng R, Feng G, Wen J, Zhu X, Deng Y. Inhibition of EZH2 and activation of ERRγ synergistically suppresses gastric cancer by inhibiting FOXM1 signaling pathway. Gastric Cancer 2021; 24:72-84. [PMID: 32529327 DOI: 10.1007/s10120-020-01097-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Accepted: 06/03/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND Gastric cancer (GC) is a leading cause of cancer-related mortality worldwide, because of the low efficacy of current therapeutic strategies. Estrogen-related receptor γ (ERRγ) was previously showed as a suppressor of GC. However, the mechanism and effective therapeutic method based on ERRγ is yet to be developed. METHODS The expression levels of ERRγ, EZH2, and FOXM1 were detected by immunohistochemistry, qRT-PCR, and western blot. The regulatory mechanisms of ERRγ and FOXM1 were analyzed by ChIP, EMSA, and siRNA. The effects of EZH2 inhibitor (GSK126) or/and ERRγ agonist (DY131) on the tumorigenesis of gastric cancer cell lines were examined by cell proliferation, transwell migration, wound healing, and colony formation assays. Meanwhile, the inhibitory effects of GSK126 or/and DY131 on tumor growth were analyzed by xenograft tumor growth assay. RESULTS The expression of ERRγ was suppressed in tumor tissues of GC patients and positively correlated with prognosis, as opposed to that of EZH2 and FOXM1. EZH2 transcriptionally suppressed ERRγ via H3K27me3, which subsequently activated the expression of master oncogene FOXM1. The combination of GSK126 and DY131 synergistically activated ERRγ expression, which subsequently inhibited the expression of FOXM1 and its regulated pathways. Synergistic combination of GSK126 and DY131 significantly inhibited the tumorigenesis of GC cell lines and suppressed the growth of GC xenograft. CONCLUSION The FOXM1 signaling pathway underlying the ERRγ-mediated gastric cancer suppression was identified. Furthermore, combined treatment with EZH2 inhibitor and ERRγ agonist synergistically suppressed GC progression by inhibiting this signaling pathway, suggesting its high potential in treating GC patients.
Collapse
Affiliation(s)
- Boyan Huang
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Tianhe District, Guangzhou, 510642, Guangdong, P. R. China.,Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou, 510642, Guangdong, P. R. China.,Key Laboratory of Zoonosis of Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou, 510642, Guangdong, P. R. China
| | - Peiqiang Mu
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Tianhe District, Guangzhou, 510642, Guangdong, P. R. China.,Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou, 510642, Guangdong, P. R. China.,Key Laboratory of Zoonosis of Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou, 510642, Guangdong, P. R. China
| | - Yan Yu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Cancer Center, Sun Yat-Sen University, Guangzhou, 510060, Guangdong, P. R. China
| | - Wenya Zhu
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Tianhe District, Guangzhou, 510642, Guangdong, P. R. China.,Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou, 510642, Guangdong, P. R. China.,Key Laboratory of Zoonosis of Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou, 510642, Guangdong, P. R. China
| | - Tianqing Jiang
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Tianhe District, Guangzhou, 510642, Guangdong, P. R. China.,Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou, 510642, Guangdong, P. R. China.,Key Laboratory of Zoonosis of Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou, 510642, Guangdong, P. R. China
| | - Rong Deng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Cancer Center, Sun Yat-Sen University, Guangzhou, 510060, Guangdong, P. R. China
| | - Gongkan Feng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Cancer Center, Sun Yat-Sen University, Guangzhou, 510060, Guangdong, P. R. China
| | - Jikai Wen
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Tianhe District, Guangzhou, 510642, Guangdong, P. R. China.,Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou, 510642, Guangdong, P. R. China.,Key Laboratory of Zoonosis of Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou, 510642, Guangdong, P. R. China
| | - Xiaofeng Zhu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Cancer Center, Sun Yat-Sen University, Guangzhou, 510060, Guangdong, P. R. China.
| | - Yiqun Deng
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Tianhe District, Guangzhou, 510642, Guangdong, P. R. China. .,Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou, 510642, Guangdong, P. R. China. .,Key Laboratory of Zoonosis of Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou, 510642, Guangdong, P. R. China.
| |
Collapse
|
17
|
Karbalaei M, Keikha M. Potential association between the hopQ alleles of Helicobacter pylori and gastrointestinal diseases: A systematic review and meta-analysis. Meta Gene 2020. [DOI: 10.1016/j.mgene.2020.100816] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
|
18
|
Wang SF, Huang KH, Tseng WC, Lo JF, Li AFY, Fang WL, Chen CF, Yeh TS, Chang YL, Chou YC, Hung HH, Lee HC. DNAJA3/Tid1 Is Required for Mitochondrial DNA Maintenance and Regulates Migration and Invasion of Human Gastric Cancer Cells. Cancers (Basel) 2020; 12:cancers12113463. [PMID: 33233689 PMCID: PMC7699785 DOI: 10.3390/cancers12113463] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 11/13/2020] [Accepted: 11/18/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Gastric cancer is a common health issue. Deregulated cellular energetics is regarded as a cancer hallmark and mitochondrial dysfunction might contribute to cancer progression. Tid1, a mitochondrial co-chaperone, may play a role as a tumor suppressor in various cancers, but the role of Tid1 in gastric cancers remains under investigated. METHODS The clinical TCGA online database and immunohistochemical staining for Tid1 expression in tumor samples of gastric cancer patients were analyzed. Tid1 knockdown by siRNA was applied to investigate the role of Tid1 in gastric cancer cells. RESULTS Low Tid1 protein-expressing gastric cancer patients had a poorer prognosis and higher lymph node invasion than high Tid1-expressing patients. Knockdown of Tid1 did not increase cell proliferation, colony/tumor sphere formation, or chemotherapy resistance in gastric cancer cells. However, Tid1 knockdown increased cell migration and invasion. Moreover, Tid1 knockdown reduced the mtDNA copy number of gastric cancer cells. In addition, the Tid1-galectin-7-MMP-9 axis might be associated with Tid1 knockdown-induced cell migration and invasion of gastric cancer cells. CONCLUSIONS Tid1 is required for mtDNA maintenance and regulates migration and invasion of gastric cancer cells. Tid1 deletion may be a poor prognostic factor in gastric cancers and could be further investigated for development of gastric cancer treatments.
Collapse
Affiliation(s)
- Sheng-Fan Wang
- Department of Pharmacy, Taipei Veterans General Hospital, Taipei 112, Taiwan; (S.-F.W.); (Y.-L.C.); (Y.-C.C.)
- School of Pharmacy, Taipei Medical University, Taipei 110, Taiwan
- Department and Institute of Pharmacology, School of Medicine, National Yang-Ming University, Taipei 112, Taiwan; (W.-C.T.); (J.-F.L.)
| | - Kuo-Hung Huang
- School of Medicine, National Yang-Ming University, Taipei 112, Taiwan; (K.-H.H.); (A.F.-Y.L.); (W.-L.F.)
- Department of Surgery, Division of General Surgery, Taipei Veterans General Hospital, Taipei 112, Taiwan
| | - Wei-Chuan Tseng
- Department and Institute of Pharmacology, School of Medicine, National Yang-Ming University, Taipei 112, Taiwan; (W.-C.T.); (J.-F.L.)
| | - Jeng-Fan Lo
- Department and Institute of Pharmacology, School of Medicine, National Yang-Ming University, Taipei 112, Taiwan; (W.-C.T.); (J.-F.L.)
- Department of Dentistry, School of Dentistry, National Yang-Ming University, Taipei 112, Taiwan
- Institute of Oral Biology, National Yang-Ming University, Taipei 112, Taiwan
- Cancer Progression Research Center, National Yang-Ming University, Taipei 112, Taiwan;
| | - Anna Fen-Yau Li
- School of Medicine, National Yang-Ming University, Taipei 112, Taiwan; (K.-H.H.); (A.F.-Y.L.); (W.-L.F.)
- Department of Pathology, Taipei Veterans General Hospital, Taipei 112, Taiwan
| | - Wen-Liang Fang
- School of Medicine, National Yang-Ming University, Taipei 112, Taiwan; (K.-H.H.); (A.F.-Y.L.); (W.-L.F.)
- Department of Surgery, Division of General Surgery, Taipei Veterans General Hospital, Taipei 112, Taiwan
| | - Chian-Feng Chen
- Cancer Progression Research Center, National Yang-Ming University, Taipei 112, Taiwan;
| | - Tien-Shun Yeh
- Institute of Anatomy and Cell Biology, School of Medicine, National Yang-Ming University, Taipei 112, Taiwan;
| | - Yuh-Lih Chang
- Department of Pharmacy, Taipei Veterans General Hospital, Taipei 112, Taiwan; (S.-F.W.); (Y.-L.C.); (Y.-C.C.)
- Department and Institute of Pharmacology, School of Medicine, National Yang-Ming University, Taipei 112, Taiwan; (W.-C.T.); (J.-F.L.)
- Faculty of Pharmacy, National Yang-Ming University, Taipei 112, Taiwan
| | - Yueh-Ching Chou
- Department of Pharmacy, Taipei Veterans General Hospital, Taipei 112, Taiwan; (S.-F.W.); (Y.-L.C.); (Y.-C.C.)
- School of Pharmacy, Taipei Medical University, Taipei 110, Taiwan
- Department and Institute of Pharmacology, School of Medicine, National Yang-Ming University, Taipei 112, Taiwan; (W.-C.T.); (J.-F.L.)
- Faculty of Pharmacy, National Yang-Ming University, Taipei 112, Taiwan
| | - Hung-Hsu Hung
- School of Medicine, Faculty of Medicine, National Yang-Ming University, Taipei 112, Taiwan
- Department of Medicine, Division of Gastroenterology, Cheng Hsin General Hospital, Taipei 112, Taiwan
- Correspondence: (H.-H.H.); (H.-C.L.); Tel.: +886-2-2826-7327 (H.-C.L.)
| | - Hsin-Chen Lee
- Department and Institute of Pharmacology, School of Medicine, National Yang-Ming University, Taipei 112, Taiwan; (W.-C.T.); (J.-F.L.)
- Correspondence: (H.-H.H.); (H.-C.L.); Tel.: +886-2-2826-7327 (H.-C.L.)
| |
Collapse
|
19
|
Moehler M, Dvorkin M, Boku N, Özgüroğlu M, Ryu MH, Muntean AS, Lonardi S, Nechaeva M, Bragagnoli AC, Coşkun HS, Cubillo Gracian A, Takano T, Wong R, Safran H, Vaccaro GM, Wainberg ZA, Silver MR, Xiong H, Hong J, Taieb J, Bang YJ. Phase III Trial of Avelumab Maintenance After First-Line Induction Chemotherapy Versus Continuation of Chemotherapy in Patients With Gastric Cancers: Results From JAVELIN Gastric 100. J Clin Oncol 2020; 39:966-977. [PMID: 33197226 PMCID: PMC8078426 DOI: 10.1200/jco.20.00892] [Citation(s) in RCA: 155] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The role of maintenance therapy for gastric (GC) or gastroesophageal junction cancer (GEJC) is unclear. We investigated avelumab (anti–programmed death ligand-1 [PD-L1]) maintenance after first-line induction chemotherapy for GC/GEJC.
Collapse
Affiliation(s)
- Markus Moehler
- Department of Internal Medicine, Johannes-Gutenberg University, Mainz, Germany
| | - Mikhail Dvorkin
- Department of Oncology, Budgetary Healthcare Institution of Omsk Region Clinical Oncology Dispensary, Omsk, Russian Federation
| | | | - Mustafa Özgüroğlu
- Clinical Trial Unit, Division of Medical Oncology, Department of Internal Medicine, Cerrahpaşa Medical Faculty, Istanbul University-Cerrahpaşa, Istanbul, Turkey
| | - Min-Hee Ryu
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Alina S Muntean
- Department of Research, Oncology Institute Prof Dr Ion Chiricuţă, Cluj Napoca, Romania
| | - Sara Lonardi
- Dipartimento di Oncologia Clinica e Sperimentale, Istituto Oncologico Veneto, Istituto di Ricovero e Cura a Carattere Scientifico, Padova, Italy
| | - Marina Nechaeva
- Arkhangelsk Clinical Oncological Dispensary, State Budgetary Healthcare Institution of Arkhangelsk Region, Arkhangelsk, Russian Federation
| | | | - Hasan S Coşkun
- Department of Medical Oncology, Akdeniz University Medical Faculty, Antalya, Turkey
| | - Antonio Cubillo Gracian
- Hospital Universitario HM Sanchinarro, Madrid, Spain.,CEU Universidad San Pablo, Madrid, Spain
| | | | - Rachel Wong
- Eastern Health, Box Hill Hospital, Monash University, Melbourne, Victoria, Australia
| | - Howard Safran
- Rhode Island Hospital, Brown University, Providence, RI
| | | | - Zev A Wainberg
- Ronald Reagan University of California Los Angeles Medical Center, Santa Monica, CA
| | - Matthew R Silver
- EMD Serono Research & Development Institute, Inc, Billerica, MA; an affiliate of Merck KGaA, Darmstadt, Germany
| | - Huiling Xiong
- EMD Serono Research & Development Institute, Inc, Billerica, MA; an affiliate of Merck KGaA, Darmstadt, Germany
| | - Janet Hong
- EMD Serono Research & Development Institute, Inc, Billerica, MA; an affiliate of Merck KGaA, Darmstadt, Germany
| | - Julien Taieb
- Department of Gastroenterology and Digestive Oncology, Georges-Pompidou European Hospital, Assistance Publique-Hôpitaux de Paris, Université de Paris, Paris, France
| | - Yung-Jue Bang
- Seoul National University Hospital, Seoul, Republic of Korea
| |
Collapse
|
20
|
Gowthami J, Gururaj N, Mahalakshmi V, Sathya R, Sabarinath TR, Doss DM. Genetic predisposition and prediction protocol for epithelial neoplasms in disease-free individuals: A systematic review. J Oral Maxillofac Pathol 2020; 24:293-307. [PMID: 33456239 PMCID: PMC7802851 DOI: 10.4103/jomfp.jomfp_348_19] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 03/23/2020] [Accepted: 04/24/2020] [Indexed: 01/13/2023] Open
Abstract
Background Epithelial neoplasm is an important global health-care problem, with high morbidity and mortality rates. Early diagnosis and appropriate treatment are essential for increased life survival. Prediction of occurrence of malignancy in a disease-free individual by any means will be a great breakthrough for healthy living. Aims and Objectives The aims and objectives were to predict the genetic predisposition and propose a prediction protocol for epithelial malignancy of various systems in our body, in a disease-free individual. Methods We have searched databases both manually and electronically, published in English language in Cochrane group, Google search, MEDLINE and PubMed from 2000 to 2019. We have included all the published, peer-reviewed, narrative reviews; randomized controlled trials; case-control studies; and cohort studies and excluded the abstract-only articles and duplicates. Specific words such as "etiological factors," "pathology and mutations," "signs and symptoms," "genetics and IHC marker," and "treatment outcome" were used for the search. A total of 1032 citations were taken, and only 141 citations met the inclusion criteria and were analyzed. Results After analyzing various articles, the etiological factors, clinical signs and symptoms, genes and the pathology involved and the commonly used blood and tissue markers were analyzed. A basic investigation strategy using immunohistochemistry markers was established. Conclusion The set of proposed biomarkers should be studied in future to predict genetic predisposition in disease-free individuals.
Collapse
Affiliation(s)
- J Gowthami
- Department of Oral and Maxillofacial Pathology and Microbiology, CSI College of Dental Sciences and Research, Madurai, Tamil Nadu, India
| | - N Gururaj
- Department of Oral and Maxillofacial Pathology and Microbiology, CSI College of Dental Sciences and Research, Madurai, Tamil Nadu, India
| | - V Mahalakshmi
- Department of Oral and Maxillofacial Pathology and Microbiology, CSI College of Dental Sciences and Research, Madurai, Tamil Nadu, India
| | - R Sathya
- Department of Oral and Maxillofacial Pathology and Microbiology, CSI College of Dental Sciences and Research, Madurai, Tamil Nadu, India
| | - T R Sabarinath
- Department of Oral and Maxillofacial Pathology and Microbiology, CSI College of Dental Sciences and Research, Madurai, Tamil Nadu, India
| | - Daffney Mano Doss
- Department of Oral and Maxillofacial Pathology and Microbiology, CSI College of Dental Sciences and Research, Madurai, Tamil Nadu, India
| |
Collapse
|
21
|
Hummell NA, Kirienko NV. Repurposing bioactive compounds for treating multidrug-resistant pathogens. J Med Microbiol 2020; 69:881-894. [PMID: 32163353 DOI: 10.1099/jmm.0.001172] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Introduction. Antimicrobial development is being outpaced by the rising rate of antimicrobial resistance in the developing and industrialized world. Drug repurposing, where novel antibacterial functions can be found for known molecular entities, reduces drug development costs, reduces regulatory hurdles, and increases rate of success.Aim. We sought to characterize the antimicrobial properties of five known bioactives (DMAQ-B1, carboplatin, oxaliplatin, CD437 and PSB-069) that were discovered in a high-throughput phenotypic screen for hits that extend Caenorhabditis elegans survival during exposure to Pseudomonas aeruginosa PA14.Methodology. c.f.u. assays, biofilm staining and fluorescence microscopy were used to assay the compounds' effect on various virulence determinants. Checkerboard assays were used to assess synergy between compounds and conventional antimicrobials. C. elegans-based assays were used to test whether the compounds could also rescue against Enterococcus faecalis and Staphyloccus aureus. Finally, toxicity was assessed in C. elegans and mammalian cells.Results. Four of the compounds rescued C. elegans from a second bacterial pathogen and two of them (DMAQ-B1, a naturally occurring insulin mimetic, and CD437, an agonist of the retinoic acid receptor) rescued against all three. The platinum complexes displayed increased antimicrobial activity against P. aeruginosa. Of the molecules tested, only CD437 showed slight synergy with ampicillin. The two most effective compounds, DMAQ-B1 and CD437, showed toxicity to mammalian cells.Conclusion. Although these compounds' potential for repurposing is limited by their toxicity, our results contribute to this growing field and provide a simple road map for using C. elegans for preliminary testing of known bioactive compounds with predicted antimicrobial activity.
Collapse
|
22
|
Li J, Han B, Sun G, Zheng Z, Mu Y, Chi J. Chinese patent medicine (Jinlong Capsule) for gastric cancer: Protocol for a systematic review and meta-analysis. Medicine (Baltimore) 2020; 99:e20532. [PMID: 32502010 PMCID: PMC7306397 DOI: 10.1097/md.0000000000020532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND JLC has been widely applied as a promising adjunctive drug for GC. However, the exact effects and safety of JLC have yet to be systematically investigated. We aimed to summarize the efficacy and safety of JLC for the treatment of advanced GC through the meta-analysis, in order to provide scientific reference for the design of future clinical trials. METHODS The protocol followed Preferred Reporting Items for Systematic Reviews and Meta-Analyses Protocols. Relevant randomized controlled trials were searched from Cochrane Library, PubMed, Web of Science (WOS), Excerpt Medica Database (Embase), Chinese Biomedical Literature Database (CBM), China National Knowledge Infrastructure (CNKI), China Scientific Journal Database (VIP), and Wanfang Database. Papers in English or Chinese published from their inception to January 2020 will be included without any restrictions.Study selection and data extraction will be performed independently by 2 investigators. The clinical outcomes including overall response rate, complete response rate, overall survival, Disease-free survival, quality of life (QoL), immune function, and adverse events, were systematically evaluated. Review Manager 5.3 and Stata 14.0 were used for data analysis, and the quality of the studies was also evaluated. RESULTS AND CONCLUSION The findings of this research will be published in a peer-reviewed journal, and provide more evidence-based guidance in clinical practice. INTERNATIONAL PLATFORM OF REGISTERED SYSTEMATIC REVIEW AND META-ANALYSIS PROTOCOLS (INPLASY) REGISTRATION NUMBER:: INPLASY202040105. URL: https://inplasy.com/inplasy-2020-4-0105/.
Collapse
Affiliation(s)
| | | | - Guangzong Sun
- Department of Emergency, People's Hospital of Weifang Binhai Economic and Technological Development Zone
| | - Zhong Zheng
- Department of Gastroenterology, Weifang People's Hospital, Weifang
| | - Ying Mu
- Department of Gastroenterology, Liaocheng People's Hospital, Liaocheng
| | - Jingxia Chi
- Quality Management Office, People's Hospital of Weifang Binhai Economic and Technological Development Zone, Weifang, Shandong Province, China
| |
Collapse
|
23
|
Zhang Y, Lin Y, Duan J, Xu K, Mao M, Wang X. A Population-Based Analysis of Distant Metastasis in Stage IV Gastric Cancer. Med Sci Monit 2020; 26:e923867. [PMID: 32409630 PMCID: PMC7245058 DOI: 10.12659/msm.923867] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Distant metastasis (DM) is a crucial problem in management of patients with gastric cancer. Identification of the risk factors for development of DM and the prognostic factors for patients with DM is essential in development of individualized treatment of patients at the advanced stage with specific metastasis. MATERIAL AND METHODS Records of patients with gastric cancer were extracted from the Surveillance, Epidemiology, and End Results (SEER) database. Survival duration of patients with specific DM was estimated, and the prognostic factors were investigated using the Cox proportional hazard regression model. The logistic regression model was used to reveal the inherent risk factors for development of DM. RESULTS Eventually, 32.6% (11,918 out of 36,588) of gastric cancer patients were diagnosed with DM between 2010 and 2015, among whom 5,361, 1,778, 1,495, and 231 patients were diagnosed with liver, lung, bone, and brain metastasis, respectively. The median overall survival for patients with DM was 5.0 (95% CI: 4.8-5.2) months, with a 5-year survival rate of 3.9%. Primary tumor site, histology types, tumor grade, T stage, N stage, surgery, chemotherapy, and the number of metastases were associated with worse survival. Younger age and higher tumor grade were positively associated with the development of DM. CONCLUSIONS Initial DM was found in 32.6% of patients with gastric cancer. Homogenous and heterogenous predictive factors were identified for patients with a specific metastatic site, which can be used in targeted screening and individualized treatment.
Collapse
Affiliation(s)
- Yiran Zhang
- International Medical Center, Tianjin First Central Hospital, Tianjin, China (mainland)
| | - Yile Lin
- Clinical Medical College of Tianjin Medical University, Tianjin, China (mainland)
| | - Jincai Duan
- Clinical Medical College of Tianjin Medical University, Tianjin, China (mainland)
| | - Ke Xu
- Clinical Medical College of Tianjin Medical University, Tianjin, China (mainland)
| | - Min Mao
- Department of Pathology and Southwest Cancer Center, First Affiliated Hospital, Army Medical University, Chongqing, China (mainland)
| | - Xin Wang
- Department of Epidemiology and Biostatistics, First Affiliated Hospital, Army Medical University, Chongqing, China (mainland)
| |
Collapse
|
24
|
Alshahrani S, Baabbad F, Bahobail M, Hawsawi A, Jastania E, Bamousa S, Shobair A, Zaidi SF. Survival Time in Treatment Modalities of Gastric Carcinoma at King Khalid Hospital- Jeddah Saudi Arabia: a Retrospective Cohort Study. Mater Sociomed 2020; 32:271-276. [PMID: 33628129 PMCID: PMC7879444 DOI: 10.5455/msm.2020.32.271-276] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Background: Gastric cancer (GC) is the fifth most common cancer worldwide and is responsible for 10% of annual cancer deaths. Gastric cancer treatment requires a multimodality approach involving surgery, adjuvant chemotherapy (AC), and neoadjuvant chemotherapy (NAC) for locally advanced tumors. Surgical removal of the tumor is the most common and effective curative approach with a more promising survival rate. Objectives: Due to the scarcity of studies in the kingdom of Saudi Arabia, this study aims to provide an epidemiological background on the subject and compare multiple treatments and their survival outcomes in a tertiary hospital in the western region. Methods: After obtaining the IB approval, data was collected from medical files of all histologically confirmed GC patients (101) between the years 2000-2015. In this study, patients’ demographics, tumor characteristics, treatment types, and patients’ vital status (deceased or alive) were collected. Survival analysis between treatment modalities subgroups was carried out using the Kaplan-Meier test and Cox regression model. Results: Out of 101 patients with GC, 65.35% were males with a mean age of 64.9±19 years and 61.71±17.83 years for female patients. Most patients had a stage IV tumor 47 (74.6%). The adjuvant chemotherapy group had a median survival time of 140 months (95% CI 33.72-246.29), while the neoadjuvant chemotherapy was 69 months. Patients who only underwent surgery had a median survival time of 28 months (95% CI 0-56.14), whereas patients who received no intervention had a median survival time of 8 months (95% CI 1.47-14.53) (log-rank = 0.002). The median overall survival time for all patients was 28 months (95%CI 0-77.68) with an overall five-year survival rate of 19.6%. Cox regression model revealed that treatment type and tumor stage were significant predictors of survival with p values of 0.001 and 0.009, respectively. Conclusion: Adjuvant, neoadjuvant, and palliative chemotherapy constitute the paramount treatment modalities of GC with adjuvant treatment having the highest survival time. However, these modalities necessitate further understanding as to whether other factors play a role in the selection of one treatment with the preferable outcome.
Collapse
Affiliation(s)
- Saad Alshahrani
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Jeddah 22384, Saudi Arabia
| | - Faisal Baabbad
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Jeddah 22384, Saudi Arabia
| | - Majed Bahobail
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Jeddah 22384, Saudi Arabia
| | - Alaa Hawsawi
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Jeddah 22384, Saudi Arabia
| | - Essam Jastania
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Jeddah 22384, Saudi Arabia
| | - Saeed Bamousa
- Princess Norah Oncology Center at King Khalid Hospital, Jeddah 22384, Saudi Arabia
| | - Ammar Shobair
- Gastroenterology and Hepatology Department at King Khalid Hospital, Jeddah 22384, Saudi Arabia
| | - Syed Faisal Zaidi
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Jeddah 22384, Saudi Arabia
| |
Collapse
|