201
|
Zhuang J, Liu X, Yang Y, Zhang Y, Guan G. Sulfasalazine, a potent suppressor of gastric cancer proliferation and metastasis by inhibition of xCT: Conventional drug in new use. J Cell Mol Med 2021; 25:5372-5380. [PMID: 33988296 PMCID: PMC8184680 DOI: 10.1111/jcmm.16548] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 03/15/2021] [Accepted: 03/22/2021] [Indexed: 12/17/2022] Open
Abstract
The aim of this study was to explore the role of sulfasalazine on proliferation and metastasis in gastric cancer by inhibition of xCT. The relationships between clinical characteristics and xCT expression were analysed. An immunohistochemical staining assay and Western blot were performed among gastric cancers and normal gastric tissues. qPCR and Western blot were also used to evaluate the mRNA and protein expression in the normal gastric cell and eight gastric cancer cells, respectively. CCK‐8 and colony formation assays were used to evaluate the effect of sulfasalazine on the proliferation and colony formation ability of three gastric cancers. The effect of sulfasalazine on the migration and invasion abilities of three cancer cells was assessed by the Transwell assay. xCT protein is up‐regulated in gastric cancer specimens and cells. Three gastric cancer cells with high, medium and low expression of xCT were selected for the following analyses. CCK‐8 assays revealed that sulfasalazine could attenuate the proliferation of HGC‐27 and AGS. Also, the colony formation assay revealed that sulfasalazine might attenuate the colony formation ability in HGC‐27 and AGS cells. Plus, the Transwell assays demonstrated that sulfasalazine might attenuate the migration and invasion abilities in HGC‐27 and AGS cells. In conclusion, higher expression of xCT is associated with advanced tumour stage and poor overall survival of gastric cancer. Sulfasalazine can attenuate the proliferation, colony formation, metastasis and invasion of gastric cancer in vitro. Further study is required to validate our findings.
Collapse
Affiliation(s)
- Jinfu Zhuang
- Department of Colorectal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Xing Liu
- Department of Colorectal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Yuanfeng Yang
- Department of Colorectal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Yiyi Zhang
- Department of Colorectal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Guoxian Guan
- Department of Colorectal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| |
Collapse
|
202
|
Ma R, Shimura T, Yin C, Okugawa Y, Kitajima T, Koike Y, Okita Y, Ohi M, Uchida K, Goel A, Yao L, Zhang X, Toiyama Y. Antitumor effects of Andrographis via ferroptosis-associated genes in gastric cancer. Oncol Lett 2021; 22:523. [PMID: 34025790 DOI: 10.3892/ol.2021.12784] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Accepted: 03/31/2021] [Indexed: 12/24/2022] Open
Abstract
The overall prognosis of advanced/metastatic gastric cancer (GC) remains poor despite the development of pharmacotherapy. Therefore, other treatment options, such as complementary and alternative medicine, should be considered to overcome this aggressive malignancy. Andrographis, which is a generally unharmful botanical compound, has gained increasing interest for its anticancer effects in multiple malignancies via the regulation of cancer progression-associated signaling pathways. In the present study, a series of in vitro experiments (cell proliferation, colony formation and apoptosis assays) was designed to elucidate the antitumor potential and mechanism of Andrographis in GC cells. The present study demonstrated that Andrographis exerted antitumor effects in GC cell lines (MKN74 and NUGC4) by inhibiting proliferation, reducing colony formation and enhancing apoptotic activity. Furthermore, it was demonstrated that the expression levels of the ferroptosis-associated genes heme oxygenase-1, glutamate-cysteine ligase catalytic and glutamate-cysteine ligase modifier were significantly upregulated after Andrographis treatment in both GC cell lines in reverse transcription-quantitative PCR experiments (P<0.05); this finding was further confirmed by immunoblotting assays (P<0.05). In conclusion, to the best of our knowledge, the present study was the first to demonstrate that Andrographis possessed antitumor properties by altering the expression levels of ferroptosis-associated genes, thereby providing novel insights into the potential of Andrographis as an adjunctive treatment option for patients with metastatic GC.
Collapse
Affiliation(s)
- Ruiya Ma
- Department of Gastrointestinal and Pediatric Surgery, Institute of Life Sciences, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan.,Department of Colorectal Surgery, Tangshan Gongren Hospital, Tangshan, Hebei 063000, P.R. China
| | - Tadanobu Shimura
- Department of Gastrointestinal and Pediatric Surgery, Institute of Life Sciences, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| | - Chengzeng Yin
- Department of Gastrointestinal and Pediatric Surgery, Institute of Life Sciences, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| | - Yoshinaga Okugawa
- Department of Genomic Medicine, Mie University Hospital, Tsu, Mie 514-8507, Japan
| | - Takahito Kitajima
- Department of Gastrointestinal and Pediatric Surgery, Institute of Life Sciences, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| | - Yuhki Koike
- Department of Gastrointestinal and Pediatric Surgery, Institute of Life Sciences, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| | - Yoshiki Okita
- Department of Gastrointestinal and Pediatric Surgery, Institute of Life Sciences, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| | - Masaki Ohi
- Department of Gastrointestinal and Pediatric Surgery, Institute of Life Sciences, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| | - Keiichi Uchida
- Department of Gastrointestinal and Pediatric Surgery, Institute of Life Sciences, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| | - Ajay Goel
- Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA 91016, USA
| | - Li Yao
- Department of Surgery, China-Japan Friendship Hospital, Beijing 100029, P.R. China
| | - Xueming Zhang
- Department of Colorectal Surgery, Tangshan Gongren Hospital, Tangshan, Hebei 063000, P.R. China
| | - Yuji Toiyama
- Department of Gastrointestinal and Pediatric Surgery, Institute of Life Sciences, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| |
Collapse
|
203
|
Baxter MA, Petty RD, Swinson D, Hall PS, O'Hanlon S. Real‑world challenge for clinicians treating advanced gastroesophageal adenocarcinoma (Review). Int J Oncol 2021; 58:22. [PMID: 33760115 PMCID: PMC7979263 DOI: 10.3892/ijo.2021.5202] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 03/04/2021] [Indexed: 12/15/2022] Open
Abstract
Gastroesophageal adenocarcinoma (GOA) is a disease of older people. Incidence is rising in the developed world and the majority of patients present with advanced disease. Based on clinical trial data, systemic chemotherapy in the advanced setting is associated with improvements in quality of life and survival. However, there is a recognised mismatch between trial populations and the patients encountered in clinical practice in terms of age, comorbidity and fitness. Appropriate patient selection is essential to safely deliver effective treatment. In this narrative review, we discuss the challenges faced by clinicians when assessing real‑world patients with advanced GOA for systemic therapy. We also highlight the importance of frailty screening and the current available evidence we can use to guide our management.
Collapse
Affiliation(s)
- Mark A. Baxter
- Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee DD2 1SY, Scotland
- Tayside Cancer Centre, Ninewells Hospital and Medical School, NHS Tayside, Dundee DD1 9SY, UK
| | - Russell D. Petty
- Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee DD2 1SY, Scotland
- Tayside Cancer Centre, Ninewells Hospital and Medical School, NHS Tayside, Dundee DD1 9SY, UK
| | - Daniel Swinson
- Department of Oncology, St. James's Hospital, Leeds Teaching Hospitals NHS Trust, Leeds LS9 7TF, UK
| | - Peter S. Hall
- Edinburgh Cancer Research Centre, University of Edinburgh, Edinburgh EH4 2XR, Scotland, UK
| | - Shane O'Hanlon
- Department of Geriatric Medicine, St. Vincent's University Hospital, Dublin 4, D04 N2E0, Republic of Ireland
| |
Collapse
|
204
|
Development and Validation of a Robust Immune-Related Prognostic Signature for Gastric Cancer. J Immunol Res 2021; 2021:5554342. [PMID: 34007851 PMCID: PMC8110424 DOI: 10.1155/2021/5554342] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 04/11/2021] [Accepted: 04/13/2021] [Indexed: 02/07/2023] Open
Abstract
Background An increasing number of reports have found that immune-related genes (IRGs) have a significant impact on the prognosis of a variety of cancers, but the prognostic value of IRGs in gastric cancer (GC) has not been fully elucidated. Methods Univariate Cox regression analysis was adopted for the identification of prognostic IRGs in three independent cohorts (GSE62254, n = 300; GSE15459, n = 191; and GSE26901, n = 109). After obtaining the intersecting prognostic genes, the three independent cohorts were merged into a training cohort (n = 600) to establish a prognostic model. The risk score was determined using multivariate Cox and LASSO regression analyses. Patients were classified into low-risk and high-risk groups according to the median risk score. The risk score performance was validated externally in the three independent cohorts (GSE26253, n = 432; GSE84437, n = 431; and TCGA, n = 336). Immune cell infiltration (ICI) was quantified by the CIBERSORT method. Results A risk score comprising nine genes showed high accuracy for the prediction of the overall survival (OS) of patients with GC in the training cohort (AUC > 0.7). The risk of death was found to have a positive correlation with the risk score. The univariate and multivariate Cox regression analyses revealed that the risk score was an independent indicator of the prognosis of patients with GC (p < 0.001). External validation confirmed the universal applicability of the risk score. The low-risk group presented a lower infiltration level of M2 macrophages than the high-risk group (p < 0.001), and the prognosis of patients with GC with a higher infiltration level of M2 macrophages was poor (p = 0.011). According to clinical correlation analysis, compared with patients with the diffuse and mixed type of GC, those with the Lauren classification intestinal GC type had a significantly lower risk score (p = 0.00085). The patients' risk score increased with the progression of the clinicopathological stage. Conclusion In this study, we constructed and validated a robust prognostic signature for GC, which may help improve the prognostic assessment system and treatment strategy for GC.
Collapse
|
205
|
Yue C, Yu C, Peng R, Wang J, Li G, Xu L. LINC00665/miR-379-5p/GRP78 regulates cisplatin sensitivity in gastric cancer by modulating endoplasmic reticulum stress. Cytotechnology 2021; 73:413-422. [PMID: 34149174 DOI: 10.1007/s10616-021-00466-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 03/19/2021] [Indexed: 12/25/2022] Open
Abstract
Acquired resistance to cisplatin (DDP)-based chemotherapy greatly hinders the treatment of gastric cancer (GC). LINC00665 serves as an oncogene in GC. Hence, the current study was designed to investigate the regulatory effects of LINC00665 on DDP-resistance of GC. LINC00665 and miR-379-5p expression levels were detected by real-time quantitative polymerase chain reaction (RT-qPCR) and Glucose regulated protein 78 (GRP78) protein level was measured by western blot assay. Interactions between LINC00665 and miR-379-5p or between miR-379-5p and GRP78 were verified by dual luciferase reporter assay. Cell counting kit 8 (CCK-8) assay and flow cytometry assay respectively determine the proliferative ability and apoptosis of GC cells. Western blot analysis was also performed to detect the protein levels of C/EBP-homologous protein (CHOP), X box binding protein (XBP1) and apoptosis-related proteins. In addition, GRP78 expression was evaluated by immunofluorescence. It was observed that the expression levels of LINC00665 and GRP78 were upregulated, and the expression level of miR-379-5p was downregulated in DDP-sensitive and DDP-resistant GC cell lines. What's more, GRP78 expression and the cell growth inhibition rates of DDP-sensitive and DDP-resistant GC cells had a negative correlation. Additionally, miR-379-5p was a target miRNA of LINC00665, and GRP78 was a target mRNA of miR-379-5p. Functional studies revealed that knockdown of LINC00665 inhibited DDP-resistant GC cell proliferation, induced apoptosis as well as suppressed Endoplasmic reticulum (ER) stress. Mechanistically, knockdown of LINC00665 downregulated GRP78 expression by strengthening miR-379-5p. LINC00665 silencing could overcome DPP-resistance of GC cells by downregulating GRP78 via sponging miR-379-5p, indicating that LINC00665 might be a potential therapeutic target for DDP- resistant GC patients.
Collapse
Affiliation(s)
- Chao Yue
- Department of General Surgery, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, 210009 Jiangsu Province China
| | - Chen Yu
- Department of Integrated Traditional Chinese and Western Medicine, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, 210009 Jiangsu Province China
| | - Rui Peng
- Department of General Surgery, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, 210009 Jiangsu Province China
| | - Jian Wang
- Department of General Surgery, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, 210009 Jiangsu Province China
| | - Gang Li
- Department of General Surgery, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, 210009 Jiangsu Province China
| | - Lin Xu
- Department of Thoracic Surgery, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, 210029 Jiangsu Province China
| |
Collapse
|
206
|
Exosomes derived from miR-1228 overexpressing bone marrow-mesenchymal stem cells promote growth of gastric cancer cells. Aging (Albany NY) 2021; 13:11808-11821. [PMID: 33883305 PMCID: PMC8109060 DOI: 10.18632/aging.202878] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 10/27/2020] [Indexed: 01/23/2023]
Abstract
There has been increasing evidence that microRNAs (miRNAs) are related to glioma progression, and that genetically engineered mesenchymal stem cells (MSCs) can inhibit the growth of gliomas. However, the underlying mechanism of bone marrow-MSCs (BM--MSCs) and miRs in gastric cancer still remains unclear. Patients with gastric cancer treated in Shijiazhuang First Hospital as well as healthy individuals undergoing physical examinations were recruited to measure the expression of exosomal miR-1228. Receiver operating characteristic (ROC) curves were plotted and the patients were followed up. BM--MSCs from healthy subjects were collected and exosomes were extracted. The MSC cells were transfected with lentiviral vectors carrying miR-1228 and MMP-14 over-expression sequences and scramble sequence, followed by exosome extraction. The exosomes were co-cultured with SGC-7901 and MGC-823 cells to detect cell proliferation, invasion, apoptosis and migration. The correlation between miR-1228 and MMP-14 was determined by dual-luciferase reporter assay. miR-1228 was highly expressed in serum exosomes of patients with gastric cancer with a area under ROC curve (AUC) of 0.865. The exosomes derived from BM-MSCs are expected to be efficient nanocarriers. Up-regulation of miR-1228 can down-regulate the expression of MMP-14 and effectively hinders the development and progression of gastric cancer.
Collapse
|
207
|
Rimini M, Pecchi A, Prampolini F, Bussei C, Salati M, Forni D, Martelli F, Valoriani F, Canino F, Bocconi A, Gelsomino F, Reverberi L, Benatti S, Piacentini F, Menozzi R, Dominici M, Luppi G, Spallanzani A. The Prognostic Role of Early Skeletal Muscle Mass Depletion in Multimodality Management of Patients with Advanced Gastric Cancer Treated with First Line Chemotherapy: A Pilot Experience from Modena Cancer Center. J Clin Med 2021; 10:jcm10081705. [PMID: 33921004 PMCID: PMC8071389 DOI: 10.3390/jcm10081705] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 04/06/2021] [Accepted: 04/12/2021] [Indexed: 12/29/2022] Open
Abstract
Background: Few data about the link between nutritional status and survival are available in the metastatic gastric cancer (GC) setting. The aim of this work was to evaluate the prognostic role of tissue modifications during treatment and the benefit of a scheduled nutritional assessment in this setting. Methods: Clinical and laboratory variables of 40 metastatic GC patients treated at Modena Cancer Center were retrieved: 20 received a nutritional assessment on the oncology’s discretion, the other 20 received a scheduled nutritional assessment at baseline and every 2–4 weeks. Anthropometric parameters were calculated on Computed Tomography (CT) images at the baseline and after 3 months of chemotherapy. Results: A correlation between baseline Eastern Cooperative Oncology Group Performance Status (ECOG PS), Lymphocyte to Monocyte Ratio (LMR), C-reactive protein (PCR), Prognostic Nutritional Index (PNI) and Overall survival (OS) was highlighted. Among the anthropometric parameters, early skeletal muscle mass depletion (ESMMD) >10% in the first months of treatment significantly impacted on mOS (p = 0.0023). A link between ESMMD and baseline LDH > 460 U/L, baseline CRP > 2.2 mg/dL and weight decrease during treatment emerged. Patients evaluated with a nutritional scheduled support experienced a mean gain in subcutaneous and visceral fat of 11.4% and 10.21%, respectively. Conclusion: We confirm the prognostic impact of ESMMD > 10% during chemotherapy in metastatic GC. The prognostic role of a scheduled nutritional assessment deserves further confirmation in large prospective trials.
Collapse
Affiliation(s)
- Margherita Rimini
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41122 Modena, Italy; (M.R.); (M.S.); (F.C.); (A.B.); (F.G.); (S.B.); (F.P.); (M.D.); (G.L.)
| | - Annarita Pecchi
- Department of Radiology, University Hospital of Modena, 41122 Modena, Italy; (A.P.); (F.P.); (D.F.); (F.M.)
| | - Francesco Prampolini
- Department of Radiology, University Hospital of Modena, 41122 Modena, Italy; (A.P.); (F.P.); (D.F.); (F.M.)
| | - Chiara Bussei
- Division of Metabolic Disease and Clinical Nutrition, University Hospital of Modena, 41122 Modena, Italy; (C.B.); (F.V.); (L.R.); (R.M.)
| | - Massimiliano Salati
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41122 Modena, Italy; (M.R.); (M.S.); (F.C.); (A.B.); (F.G.); (S.B.); (F.P.); (M.D.); (G.L.)
| | - Daniela Forni
- Department of Radiology, University Hospital of Modena, 41122 Modena, Italy; (A.P.); (F.P.); (D.F.); (F.M.)
| | - Francesca Martelli
- Department of Radiology, University Hospital of Modena, 41122 Modena, Italy; (A.P.); (F.P.); (D.F.); (F.M.)
| | - Filippo Valoriani
- Division of Metabolic Disease and Clinical Nutrition, University Hospital of Modena, 41122 Modena, Italy; (C.B.); (F.V.); (L.R.); (R.M.)
| | - Fabio Canino
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41122 Modena, Italy; (M.R.); (M.S.); (F.C.); (A.B.); (F.G.); (S.B.); (F.P.); (M.D.); (G.L.)
| | - Alessandro Bocconi
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41122 Modena, Italy; (M.R.); (M.S.); (F.C.); (A.B.); (F.G.); (S.B.); (F.P.); (M.D.); (G.L.)
| | - Fabio Gelsomino
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41122 Modena, Italy; (M.R.); (M.S.); (F.C.); (A.B.); (F.G.); (S.B.); (F.P.); (M.D.); (G.L.)
| | - Linda Reverberi
- Division of Metabolic Disease and Clinical Nutrition, University Hospital of Modena, 41122 Modena, Italy; (C.B.); (F.V.); (L.R.); (R.M.)
| | - Stefania Benatti
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41122 Modena, Italy; (M.R.); (M.S.); (F.C.); (A.B.); (F.G.); (S.B.); (F.P.); (M.D.); (G.L.)
| | - Federico Piacentini
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41122 Modena, Italy; (M.R.); (M.S.); (F.C.); (A.B.); (F.G.); (S.B.); (F.P.); (M.D.); (G.L.)
| | - Renata Menozzi
- Division of Metabolic Disease and Clinical Nutrition, University Hospital of Modena, 41122 Modena, Italy; (C.B.); (F.V.); (L.R.); (R.M.)
| | - Massimo Dominici
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41122 Modena, Italy; (M.R.); (M.S.); (F.C.); (A.B.); (F.G.); (S.B.); (F.P.); (M.D.); (G.L.)
| | - Gabriele Luppi
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41122 Modena, Italy; (M.R.); (M.S.); (F.C.); (A.B.); (F.G.); (S.B.); (F.P.); (M.D.); (G.L.)
| | - Andrea Spallanzani
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41122 Modena, Italy; (M.R.); (M.S.); (F.C.); (A.B.); (F.G.); (S.B.); (F.P.); (M.D.); (G.L.)
- Correspondence: ; Tel.: +39-05-9422-3310
| |
Collapse
|
208
|
Ge J, Liu T, Lei T, Li X, Song K, Azizi S, Liu H, Tang M. Retrospective Cohort Study of Intraoperative Administration of Sustained-Release 5-Fluorouracil Implants in Advanced Gastric Cancer Patients. Front Pharmacol 2021; 12:659258. [PMID: 33927633 PMCID: PMC8076801 DOI: 10.3389/fphar.2021.659258] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 03/08/2021] [Indexed: 12/13/2022] Open
Abstract
Background: 5-fluorouracil (5-FU) is basically used in the field of postoperative chemotherapy of gastric cancer (GC), the goal of this study was to evaluate improvement of long-term survival rate among GC patients after the 5-FU implants treatment. Methods: The study included 145 patients with gastric cancer who received postoperative chemotherapy with 5-FU implants and had complete follow-up information. According to the sex, age and clinical stage of 5-FU implants group, 74 patients were matched as the control group at the same time. In the study, we compared the 5-year overall survival rate with progression-free survival rate in the two groups, and the drug safety for both groups during the treatment was also compared. Results: The median follow-up time was 85 months (range 60–116 months). 31 patients (21.38%) died of tumor recurrence in 5-FU implants group and 21 (28.38%) in control group. In the control group, metastatic lesions were found in the small intestine, left adrenal gland and peritoneum in three patients. The 5-year progression-free survival (PFS) rate was 79.71% in 5-FU group and 67.12% in control (p = 0.0045). The 5-year overall survival (OS) rate was 77.68% in 5-FU implants group and 64.87% in control (p = 0.0159). Both the 5-years OS and PFS rates in 5-FU group were better than control group without significant side effect. Conclusions: 5-FU implants may improve 5-years OS and PFS rates after surgery in gastric cancer patients, while good safety profile suggests it could be reliable.
Collapse
Affiliation(s)
- Jie Ge
- Department of Gastrointestinal Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Ting Liu
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China
| | - Tianxiang Lei
- Department of Gastrointestinal Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Xuan Li
- Department of Gastrointestinal Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Kun Song
- Department of Gastrointestinal Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Samim Azizi
- Department of Cardiothoracic and Vascular Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Heli Liu
- Department of Gastrointestinal Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Mimi Tang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
209
|
Tsuji T, Maeda Y, Kita K, Murakami K, Saya H, Takemura H, Inaki N, Oshima M, Oshima H. FOXO3 is a latent tumor suppressor for FOXO3-positive and cytoplasmic-type gastric cancer cells. Oncogene 2021; 40:3072-3086. [PMID: 33795838 PMCID: PMC8084732 DOI: 10.1038/s41388-021-01757-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 03/03/2021] [Accepted: 03/15/2021] [Indexed: 12/12/2022]
Abstract
FOXO3 is a member of the FOXO transcription factors thought to play a tumor-suppressor role in gastrointestinal cancer, while tumor-promoting function of FOXO3 has also been reported. These results suggest a context-dependent function of FOXO3 in tumor development. However, the relationship between the FOXO3 expression pattern and its role in tumorigenesis has not been elucidated. We examined the FOXO3 expression in 65 human primary gastric cancer and patient-derived xenograft tissues by immunohistochemistry and identified three subtypes according to subcellular localization: FOXO3-nuclear accumulated (FOXO3-Nuc), FOXO3-nuclear/cytoplasmic or cytoplasmic distributed (FOXO3-Cyt), and FOXO3-negative. In the FOXO3-Cyt gastric cancer cells, the expression of the constitutive active mutant FOXO3 (Act-ER FOXO3) induced the nuclear accumulation of FOXO3 and significantly suppressed colony formation and proliferation. The inhibition of the PI3K-AKT pathway by inhibitor treatment also suppressed the proliferation of FOXO3-Cyt gastric cancer cells, which was associated with the nuclear accumulation of endogenous FOXO3. Furthermore, the expression of Act-ER FOXO3 by an endogenous promoter significantly suppressed gastric tumorigenesis in Gan mice, a model of gastric cancer. Finally, treatment of FOXO3-Cyt human gastric cancer-derived organoids with an AKT inhibitor significantly suppressed the survival and proliferation. These results indicate that FOXO3 is a latent tumor suppressor for FOXO3-Cyt-type gastric cancer cells and that activation of the PI3K-AKT pathway protects this type of gastric cancer cell from FOXO3-mediated growth suppression via constitutive nuclear export. Thus, the inhibition of the PI3K-AKT pathway and nuclear translocation of endogenous FOXO3 may have therapeutic applications in the treatment of FOXO3-positive and cytoplasmic-type gastric cancer.
Collapse
Affiliation(s)
- Toshikatsu Tsuji
- Division of Genetics, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
- Department of Thoracic, Cardiovascular and General Surgery, Kanazawa University, Kanazawa, Japan
- Department of Gastroenterological Surgery, Ishikawa Prefectural Central Hospital, Kanazawa, Japan
| | - Yusuke Maeda
- Division of Genetics, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
- Division of Gene Regulation, Institute for Advanced Medical Research (IAMR), Keio University School of Medicine, Tokyo, Japan
| | - Kenji Kita
- Central Research Resource Branch, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - Kazuhiro Murakami
- Division of Stem Cell Biology, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - Hideyuki Saya
- Division of Gene Regulation, Institute for Advanced Medical Research (IAMR), Keio University School of Medicine, Tokyo, Japan
| | - Hirofumi Takemura
- Department of Thoracic, Cardiovascular and General Surgery, Kanazawa University, Kanazawa, Japan
| | - Noriyuki Inaki
- Department of Gastroenterological Surgery, Kanazawa University, Kanazawa, Japan
| | - Masanobu Oshima
- Division of Genetics, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
- WPI Nano-Life Science Institute (Nano-LSI), Kanazawa University, Kanazawa, Japan
| | - Hiroko Oshima
- Division of Genetics, Cancer Research Institute, Kanazawa University, Kanazawa, Japan.
- WPI Nano-Life Science Institute (Nano-LSI), Kanazawa University, Kanazawa, Japan.
| |
Collapse
|
210
|
Canbay E, Canbay Torun B, Cosarcan K, Altunal C, Gurbuz B, Bilgic C, Sezgin C, Kaban KK, Yilmaz S, Yazici Z. Surgery with hyperthermic intraperitoneal chemotherapy after response to induction chemotherapy in patients with peritoneal metastasis of gastric cancer. J Gastrointest Oncol 2021; 12:S47-S56. [PMID: 33968425 DOI: 10.21037/jgo-20-121] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Background Gastric cancer (GC) with peritoneal metastases (PM) has a dismal prognosis and to date only a few management options have been reported. Of those, cytoreductive surgery (CRS) and hyperthermic intraperitoneal chemotherapy (HIPEC) after induction bidirectional intraperitoneal and systemic chemotherapy (BIPSC) appear as a promising treatment option for these patients. Outcome data including safety and efficacy of CRS with radical Gastrectomy and HIPEC after response to combination of laparoscopic HIPEC (LHIPEC) with BIPSC as an induction therapy in patients with PM of GC was evaluated in this retrospective observational study. Methods Diagnostic Laparoscopy was performed in 53 patients with PM of GC who admitted to the Center for Treatment of Peritoneal Surface Malignancies, Istanbul, between 2013 and 2016. Peritoneal cancer index (PCI), ascites status and cytology were determined. The patients underwent LHIPEC and then, BIPSC induction chemotherapy using intraperitoneal docetaxel (30 mg/m2) and cisplatin (30 mg/m2) and intravenous Docetaxel/Cisplatin/5-Fluorouracil (DCF) for 3 cycles. In selected patients, CRS with radical gastrectomy and HIPEC were performed after the response to induction therapy. BIPSC was continued for 3 more cycles with a dose reduction in an adjuvant setting. Results All LHIPEC procedures were uneventful with Grade 1-2 side effects (11/53, 20,8%). As a response to induction chemotherapy PCI was reduced from 19.6±8 (range, 6-39) to 13.6±9.8 (range, 1-39) (P<0.001). Ascites was detected in 55% (29 out of 53) and cytology was positive in 51% (27 out of 53) of the patients before induction chemotherapy. Ascites was completely abolished and all cytology became negative. Then, 34 of 53 (64.15%) patients underwent CRS with radical gastrectomy and HIPEC. CC0/1 resection was achieved in 22 (64.70%) of patients (P<0.05). The median survival time was 18.9±13.4 (95% CI: 15.2-22.6 months. Combined surgery and HIPEC related mortality occurred in 1 out of 34 patients (2.9%) due to developed diffuse intravascular coagulation at postoperative day 2. Grade 2 operative complications included biliary fistula in one, and duodenal stump leakage in two patients (8.7%). All of the fistula closed with conservative management. The median survival time was 18.9±13.4 months and the median progression-free survival time was 15.6±12.9 with 1-, 2-, and 5-year survival rates of 82.4%, 59% and 17.6% in patients with PM of GC. Multivariate analysis identified high peritoneal cancer index (P=0.000) and complete resection (P<0.05) as independent predictors for better progression-free and overall survival. Conclusions The best outcomes can be expected with optimal cytoreduction and limited peritoneal dissemination in response to induction chemotherapy. Knowledgeable selection of patients with PM of GC is essential to perform surgery with HIPEC safely with acceptable mortality and morbidity.
Collapse
Affiliation(s)
- Emel Canbay
- Department of General Surgery, NPO Center for Peritoneal Surface Malignancies, Istanbul, Turkey
| | - Bahar Canbay Torun
- Department of General Surgery, Istanbul Haseki Education & Research Hospital, Istanbul, Turkey
| | - Kaan Cosarcan
- Department of Anesthesiology, American Hospital, Istanbul, Turkey
| | - Cetin Altunal
- Department of General Surgery, NPO Center for Peritoneal Surface Malignancies, Istanbul, Turkey
| | - Bulent Gurbuz
- Department of General Surgery, American Hospital, Istanbul, Turkey
| | - Cagri Bilgic
- Department of General Surgery, American Hospital, Istanbul, Turkey
| | - Canfeza Sezgin
- Department of Medical Oncology, American Hospital, Istanbul, Turkey
| | - Kerim Kim Kaban
- Department of Medical Oncology, Faculty of Medicine, Biruni University, Istanbul, Turkey
| | - Serpil Yilmaz
- Department of Pathology, American Hospital, Istanbul, Turkey
| | - Zeliha Yazici
- Department of Pharmacology, Faculty of Medicine, Biruni University, Istanbul, Turkey
| |
Collapse
|
211
|
Kim TH, Do Cho H, Choi YW, Lee HW, Kang SY, Jeong GS, Choi JH, Ahn MS, Sheen SS. Trastuzumab-based palliative chemotherapy for HER2-positive gastric cancer: a single-center real-world data. BMC Cancer 2021; 21:325. [PMID: 33771119 PMCID: PMC7995795 DOI: 10.1186/s12885-021-08058-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 03/15/2021] [Indexed: 12/12/2022] Open
Abstract
Background Since the results of the ToGA trial were published, trastuzumab-based chemotherapy has been used as the standard first-line treatment for HER2-positive recurrent or primary metastatic gastric cancer (RPMGC). However, the real-world data has been rarely reported. Therefore, we investigated the outcomes of trastuzumab-based chemotherapy in a single center. Methods This study analyzed the real-world data of 47 patients with HER2-positive RPMGC treated with trastuzumab-based chemotherapy in a single institution. Results With the median follow-up duration of 18.8 months in survivors, the median overall survival (OS) and progression-free survival were 12.8 and 6.9 months, respectively, and the overall response rate was 64%. Eastern Cooperative Oncology Group performance status 2 and massive amount of ascites were independent poor prognostic factors for OS, while surgical resection before or after chemotherapy was associated with favorable OS, in multivariate analysis. In addition, 5 patients who underwent conversion surgery after chemotherapy demonstrated an encouraging median OS of 30.8 months, all with R0 resection. Conclusions Trastuzumab-based chemotherapy in patients with HER2-positive RPMGC in the real world demonstrated outcomes almost comparable to those of the ToGA trial. Moreover, conversion surgery can be actively considered in fit patients with a favorable response after trastuzumab-based chemotherapy.
Collapse
Affiliation(s)
- Tae-Hwan Kim
- Department of Hematology-Oncology, Ajou University School of Medicine, 164, World cup-ro, Yeongtong-gu, Suwon, 443-380, South Korea
| | - Hun Do Cho
- Department of Hematology-Oncology, Ajou University School of Medicine, 164, World cup-ro, Yeongtong-gu, Suwon, 443-380, South Korea
| | - Yong Won Choi
- Department of Hematology-Oncology, Ajou University School of Medicine, 164, World cup-ro, Yeongtong-gu, Suwon, 443-380, South Korea
| | - Hyun Woo Lee
- Department of Hematology-Oncology, Ajou University School of Medicine, 164, World cup-ro, Yeongtong-gu, Suwon, 443-380, South Korea
| | - Seok Yun Kang
- Department of Hematology-Oncology, Ajou University School of Medicine, 164, World cup-ro, Yeongtong-gu, Suwon, 443-380, South Korea
| | - Geum Sook Jeong
- Department of Hematology-Oncology, Ajou University School of Medicine, 164, World cup-ro, Yeongtong-gu, Suwon, 443-380, South Korea
| | - Jin-Hyuk Choi
- Department of Hematology-Oncology, Ajou University School of Medicine, 164, World cup-ro, Yeongtong-gu, Suwon, 443-380, South Korea.
| | - Mi Sun Ahn
- Department of Hematology-Oncology, Ajou University School of Medicine, 164, World cup-ro, Yeongtong-gu, Suwon, 443-380, South Korea.
| | - Seung-Soo Sheen
- Department of Pulmonology and Critical Care Medicine, Ajou University School of Medicine, 164, World cup-ro, Yeongtong-gu, Suwon, 443-380, South Korea
| |
Collapse
|
212
|
Yalikong A, Li XQ, Zhou PH, Qi ZP, Li B, Cai SL, Zhong YS. A Triptolide Loaded HER2-Targeted Nano-Drug Delivery System Significantly Suppressed the Proliferation of HER2-Positive and BRAF Mutant Colon Cancer. Int J Nanomedicine 2021; 16:2323-2335. [PMID: 33776436 PMCID: PMC7989962 DOI: 10.2147/ijn.s287732] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 03/02/2021] [Indexed: 12/15/2022] Open
Abstract
Background Colon cancer (CRC) was a malignant tumor and there were about 25% of patients with tumor metastasis at diagnosis stage. Chemotherapeutic agents for metastatic CRC patients were with great side effects and the clinical treatment results of advanced CRC were still not satisfactory. Human epidermal growth factor receptor 2 (HER2) is overexpressed in some CRC patients and is an effective target for CRC patient treatment. Anti-HER2 therapy had a beneficial role in the treatment of HER2-positive metastatic CRC with fewer side effects. CRC patients with BRAF mutations were resistant to HER2 antibodies treatment. Therefore, there was an urgent need to develop new therapeutic agents. Methods HER2 targeted nanoparticles (TPLNP) drug delivery system loading triptolide (TPL) were prepared and identified. The effects of TPLNP and free TPL on cell viability, targeting and cell cycle progression on HT29 (BRAF mutation) with HER2 overexpression, were evaluated by Cell Counting Kit-8 (CCK8), Fluorescence Activating Cell Sorter (FACS) and immunofluorescence methods, respectively. The anti-tumor efficacies of TPLNP were evaluated in subcutaneous xenograft model of colon cancer and the survival rate, tumor volume, liver and kidney indexes of tumor-bearing mice were measured. Results TPLNP was small in nanosize (73.4±5.2nm) with narrow size distribution (PDI=0.15±0.02) and favorable zeta potential (pH=9.6, zeta potential: −57.3±6.69mV; pH=7.0, zeta potential: −28.7±5.1mV; pH=5.6, zeta potential: −21.1±4.73mV). Comparing with free TPL treatment group, TPLNP developed stranger colon cancer-killing efficiency in a dose- and time-dependent manner detected with CCK8 method; achieved good in vitro colon cancer targeting detected with flow cytometry and immunofluorescence experiments; enhanced more HT29-HER2 apoptosis and induced more cell cycle arrested in G1-S phase detected with FACS in vitro. As for in vivo antitumor response, TPLNP remarkably inhibited the growth of colon cancer in the colon cancer xenograft model, significantly improved the survival rate and did not exhibit significant liver and kidney toxicity in contrast with free TPL in vivo. Conclusion TPLNP was effectively against colon cancer with HER2 overexpression and BRAF mutation in pre-clinical models. In summary, the TPLNP appeared to be a promising treatment option for CRC in clinical application based on improved efficacy and the favorable safety profile.
Collapse
Affiliation(s)
- Ayimukedisi Yalikong
- Endoscopy Center, Zhongshan Hospital of Fudan University, Shanghai, 200032, People's Republic of China.,Endoscopy Research Institute of Fudan University, Shanghai, 200032, People's Republic of China
| | - Xu-Quan Li
- Endoscopy Center, Zhongshan Hospital of Fudan University, Shanghai, 200032, People's Republic of China.,Shanghai Henlius Biopharmaceuticals Co., Ltd., Shanghai, 200033, People's Republic of China
| | - Ping-Hong Zhou
- Endoscopy Center, Zhongshan Hospital of Fudan University, Shanghai, 200032, People's Republic of China.,Endoscopy Research Institute of Fudan University, Shanghai, 200032, People's Republic of China
| | - Zhi-Peng Qi
- Endoscopy Center, Zhongshan Hospital of Fudan University, Shanghai, 200032, People's Republic of China.,Endoscopy Research Institute of Fudan University, Shanghai, 200032, People's Republic of China
| | - Bing Li
- Endoscopy Center, Zhongshan Hospital of Fudan University, Shanghai, 200032, People's Republic of China.,Endoscopy Research Institute of Fudan University, Shanghai, 200032, People's Republic of China
| | - Shi-Lun Cai
- Endoscopy Center, Zhongshan Hospital of Fudan University, Shanghai, 200032, People's Republic of China.,Endoscopy Research Institute of Fudan University, Shanghai, 200032, People's Republic of China
| | - Yun-Shi Zhong
- Endoscopy Center, Zhongshan Hospital of Fudan University, Shanghai, 200032, People's Republic of China.,Endoscopy Research Institute of Fudan University, Shanghai, 200032, People's Republic of China
| |
Collapse
|
213
|
Prefoldin subunits (PFDN1-6) serve as poor prognostic markers in gastric cancer. Biosci Rep 2021; 40:221904. [PMID: 31957800 PMCID: PMC7024841 DOI: 10.1042/bsr20192712] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 01/13/2020] [Accepted: 01/15/2020] [Indexed: 12/22/2022] Open
Abstract
Prefoldin subunits (PFDN), primarily known for co-chaperone function associated with cytoskeletal rearrangement, have been found involved in epithelial–mesenchymal transition (EMT) and cancer progression. However, studies focusing on the roles of PFDN in gastric cancer (GC) remain limited. The present study aims to evaluate the prognostic values of PFDN in GC. Prognostic roles of PFDNs were analyzed via the Kaplan–Meier platform, followed by subset analysis within various clinical parameters. High mRNA expression of PFDN2, PFDN3 and PFDN4 displayed poor overall survival (OS) while PFDN5 displayed favorable OS. In HER2+ subset, PFDN2, PFDN3, PFDN4 and PFDN6 displayed poor OS. In human epidermal growth factor receptor 2 (HER2−) subset, PFDN2, PFDN3 and PFDN4 displayed poor OS. In intestinal type subset, PFDN1 and PFDN2 displayed poor OS. In diffuse-type subset, PFDN2 and PFDN6 displayed poor OS. In moderate differentiation type subset, PFDN1 displayed poor OS. In poor differentiation type subset, PFDN2 and PFDN6 displayed poor OS. In metastasis negative subset, PFDN1, PFDN2 and PFDN6 displayed poor OS. In lymph node (LN) positive subset, PFDN2 and PFDN5 displayed poor OS. The present study provided insightful clues into the poor prognostic values of PFDNs in GC patients.
Collapse
|
214
|
Fornaro L, Spallanzani A, de Vita F, D’Ugo D, Falcone A, Lorenzon L, Tirino G, Cascinu S. Beyond the Guidelines: The Grey Zones of the Management of Gastric Cancer. Consensus Statements from the Gastric Cancer Italian Network (GAIN). Cancers (Basel) 2021; 13:1304. [PMID: 33804024 PMCID: PMC8001719 DOI: 10.3390/cancers13061304] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/19/2021] [Accepted: 03/11/2021] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Management of gastric and gastroesophageal junction (GEJ) adenocarcinoma remains challenging, because of the heterogeneity in tumor biology within the upper gastrointestinal tract. Daily clinical practice is full of grey areas regarding the complexity of diagnostic, staging, and therapeutic procedures. The aim of this paper is to provide a guide for clinicians facing challenging situations in routine practice, taking a multidisciplinary consensus approach based on available literature. METHODS The GAIN (GAstric cancer Italian Network) group was established with the aims of reviewing literature evidence, discussing key issues in prevention, diagnosis, and management of gastric and GEJ adenocarcinoma, and offering a summary of statements. A Delphi consensus method was used to obtain opinions from the expert panel of specialists. RESULTS Forty-nine clinical questions were identified in six areas of interest: role of multidisciplinary team; risk factors; diagnosis; management of early gastric cancer and multimodal approach to localized gastric cancer; treatment of elderly patients with locally advanced resectable disease; and treatment of locally advanced and metastatic cancer. CONCLUSIONS The statements presented may guide clinicians in practical management of this disease.
Collapse
Affiliation(s)
- Lorenzo Fornaro
- Department of Translational Medicine, Division of Medical Oncology, AOU Pisana, 56126 Pisa, Italy;
| | - Andrea Spallanzani
- Department of Oncology and Hematology, University Hospital of Modena, 41125 Modena, Italy;
| | - Ferdinando de Vita
- Department of Precision Medicine, Division of Medical Oncology, School of Medicine, University of Campania ‘Luigi Vanvitelli’, 81100 Caserta, Italy; (F.d.V.); (G.T.)
| | - Domenico D’Ugo
- General Surgery Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Catholic University, 00168 Rome, Italy; (D.D.); (L.L.)
| | - Alfredo Falcone
- Department of Translational Medicine, Division of Medical Oncology, University of Pisa, 56126 Pisa, Italy;
| | - Laura Lorenzon
- General Surgery Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Catholic University, 00168 Rome, Italy; (D.D.); (L.L.)
| | - Giuseppe Tirino
- Department of Precision Medicine, Division of Medical Oncology, School of Medicine, University of Campania ‘Luigi Vanvitelli’, 81100 Caserta, Italy; (F.d.V.); (G.T.)
| | - Stefano Cascinu
- Medical Oncology, Università Vita-Salute San Raffaele, 20132 Milan, Italy
| | | |
Collapse
|
215
|
Shreyash N, Sonker M, Bajpai S, Tiwary SK. Review of the Mechanism of Nanocarriers and Technological Developments in the Field of Nanoparticles for Applications in Cancer Theragnostics. ACS APPLIED BIO MATERIALS 2021; 4:2307-2334. [PMID: 35014353 DOI: 10.1021/acsabm.1c00020] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cancer cannot be controlled by the usage of drugs alone, and thus, nanotechnology is an important technique that can provide the drug with an impetus to act more effectively. There is adequate availability of anticancer drugs that are classified as alkylating agents, hormones, or antimetabolites. Nanoparticle (NP) carriers increase the residence time of the drug, thereby enhancing the survival rate of the drug, which otherwise gets washed off owing to the small size of the drug particles by the excretory system. For example, for enhancing the circulation, a coating of nonfouling polymers like PEG and dextran is done. Famous drugs such as doxorubicin (DOX) are commonly encapsulated inside the nanocomposite. The various classes of nanoparticles are used to enhance drug delivery by aiding it to fight against the tumor. Targeted therapy aims to attack the cells with features common to the cancer cells while minimizing damage to the normal cell, and these therapies work in one in four ways. Some block the cancer cells from reproducing newer cells, others release toxic substances to kill the cancer cells, some stimulate the immune system to destroy the cancer cells, and some block the growth of more blood vessels around cancer cells, which starve the cells of the nutrients, which is needed for their growth. This review aims to testify the advancements nanotechnology has brought in cancer therapy, and its statements are supported with recent research findings and clinical trial results.
Collapse
|
216
|
Ni Q, Zhang Y, Tao R, Li X, Zhu J. MicroRNA-95-3p serves as a contributor to cisplatin resistance in human gastric cancer cells by targeting EMP1/PI3K/AKT signaling. Aging (Albany NY) 2021; 13:8665-8687. [PMID: 33714198 PMCID: PMC8034895 DOI: 10.18632/aging.202679] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 01/22/2021] [Indexed: 12/11/2022]
Abstract
MicroRNAs (miRNAs) are thought to be involved in the development of cisplatin (DDP) resistance in gastric cancer (GC). Using RNA sequencing analysis (RNA-seq), we found that miR-95-3p is associated with DDP resistance in GC. We discovered that miR-95-3p is highly expressed in DDP-resistant GC tissues and cell lines (SGC7901/DDP and AGS/DDP). Furthermore, results from the BrdU and MTT assays indicated that miR-95-3p promotes GC cell proliferation. Additionally, data from transwell chamber assay, wound healing test and in vivo experiments illustrated that miR-95-3p can effectively promote invasion, migration and tumorigenic capacity, respectively, of DDP-resistant GC cells. Subsequently, results from dual luciferase assay and qRT-PCR collectively indicated that EMP1 is a target of miR-95-3p with inhibitory function through suppression of the EMT process and drug-resistance proteins. Furthermore, PI3K/AKT was identified as a downstream pathway of miR-95-3p, which promotes DDP resistance in GC. In summary, miR-95-3p helped develop DDP-resistance through down-regulation of EMP1 and increasing phosphorylation of the PI3K/Akt pathway in GC.
Collapse
Affiliation(s)
- Qingfeng Ni
- Department of General Surgery, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu, PR China
| | - Yan Zhang
- Department of Chemotherapy, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu, PR China
| | - Ran Tao
- Department of General Surgery, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu, PR China
| | - Xiaolong Li
- Department of General Surgery, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu, PR China
| | - Jianwei Zhu
- Department of General Surgery, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu, PR China
| |
Collapse
|
217
|
Identification and Construction of a Long Noncoding RNA Prognostic Risk Model for Stomach Adenocarcinoma Patients. DISEASE MARKERS 2021; 2021:8895723. [PMID: 33680217 PMCID: PMC7929674 DOI: 10.1155/2021/8895723] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 12/11/2020] [Accepted: 02/09/2021] [Indexed: 02/08/2023]
Abstract
Background Long noncoding RNA-based prognostic biomarkers have demonstrated great potential in the diagnosis and prognosis of cancer patients. However, systematic assessment of a multiple lncRNA-composed prognostic risk model is lacking in stomach adenocarcinoma (STAD). This study is aimed at constructing a lncRNA-based prognostic risk model for STAD patients. Methods RNA sequencing data and clinical information of STAD patients were retrieved from The Cancer Genome Atlas (TCGA) database. Differentially expressed lncRNAs (DElncRNAs) were identified using the R software. Univariate and multivariate Cox regression analyses were performed to construct a prognostic risk model. The survival analysis, C-index, and receiver operating characteristic (ROC) curve were employed to assess the sensitivity and specificity of the model. The results were verified using the GEPIA online tool and our clinical samples. Pearson correlation coefficient analysis, Gene Ontology (GO), and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment were performed to indicate the potential biological functions of the selected lncRNA. Results A total of 1917 DElncRNAs were identified from 343 cases of STAD tissues and 30 cases of noncancerous tissues. According to univariate and multivariable Cox regression analyses, four DElncRNAs (AC129507.1, LINC02407, AL022316.1, and AP000695.2) were selected to establish a prognostic risk model. There was a significant difference in the overall survival between high-risk patients and low-risk patients based on this risk model. The C-index of the model was 0.652. The area under the curve (AUC) for the ROC curve was 0.769. GEPIA results confirmed the expression and prognostic significance of AP000695.2 in STAD. Our clinical data confirmed that upregulated expression of AP000695.2 was correlated with the T stage, distant metastasis, and TNM stage in STAD. GO and KEGG analyses demonstrated that AP000695.2 was closely related to the tumorigenesis process. Conclusions In this study, we constructed a lncRNA-based prognostic risk model for STAD patients. Our study will provide novel insight into the diagnosis and prognosis of STAD patients.
Collapse
|
218
|
Oh SJ. Long-Term Survival of Two Patients with Liver Metastases from Advanced Gastric Cancer Treated with Radiofrequency Ablation and Chemotherapy. Case Rep Oncol 2021; 14:67-72. [PMID: 33776684 PMCID: PMC7983590 DOI: 10.1159/000507849] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 04/10/2020] [Indexed: 12/19/2022] Open
Abstract
Liver metastasis from gastric cancer has a very poor prognosis. Herein, we present two cases of liver metastases (synchronous and metachronous) from advanced gastric cancer. In the first case, the patient underwent radical subtotal gastrectomy. Liver metastases occurred 6 months after surgery while the patient was receiving adjuvant chemotherapy, but two hepatic tumors were successfully removed by radiofrequency ablation (RFA). In the second case, liver metastases occurred 15 months after surgery for gastric cancer. The patient also received RFA for one hepatic tumor, and other suspicious metastatic tumors were treated with systemic chemotherapy. Although these case presentations are limited for the efficacy of RFA treatment with systemic chemotherapy for hepatic metastases from gastric cancer, our findings showed long-term survival (overall survival for 108 and 67 months, respectively) of the affected patients, without recurrence. Therefore, we suggest that RFA treatment with systemic chemotherapy could be an effective alternative treatment modality for hepatic metastases from gastric cancer.
Collapse
Affiliation(s)
- Sung Jin Oh
- Department of Surgery, Haeundae Paik Hospital, Inje University College of Medicine, Busan, Republic of Korea
| |
Collapse
|
219
|
The Efficacy of Ramucirumab in the Treatment of Gastric or Gastroesophageal Junction Cancer: A Meta-Analysis of RCTs. Gastroenterol Res Pract 2021; 2021:8960315. [PMID: 33679971 PMCID: PMC7925040 DOI: 10.1155/2021/8960315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 08/20/2020] [Accepted: 02/01/2021] [Indexed: 11/24/2022] Open
Abstract
Five electronic databases were searched for eligible records. Outcomes were presented and analyzed according to the objective response rate (ORR), progression-free survival (PFS) rate, and overall survival (OS) rate. Five records involving 2,024 participants were included in the study. The pooled analysis of OS and PFS were longer with ramucirumab (RAM) therapy than without RAM for OS (odds ratio (OR) = 0.90, 95% confidence interval (CI) = 0.82–1.00, p = 0.05) and PFS (OR = 0.74, 95%CI = 0.57–0.96, p = 0.02). Moreover, compared with the current first-line chemotherapy, the OS (OR = 0.93, 95%CI = 0.83–1.04, p = 0.19) and PFS (OR = 0.82, 95%CI = 0.64–1.06, p = 0.13) results were not significantly higher with RAM. The ORRs of the patients in the RAM therapy groups were significantly higher than those in the groups without RAM (OR = 1.40, 95%CI = 1.14–1.73, p = 0.001).
Collapse
|
220
|
Wang YN, Chang SY, Hwang JM, Chang YK, Kao WY, Wan HL, Tzeng IS, Wu CC. Evaluating the benefit of adjuvant radiotherapy after extensive lymph node dissection for gastric cancer: a single-institute retrospective study. Tzu Chi Med J 2021; 33:288-293. [PMID: 34386368 PMCID: PMC8323656 DOI: 10.4103/tcmj.tcmj_230_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 10/06/2020] [Accepted: 11/03/2020] [Indexed: 11/17/2022] Open
Abstract
Objective: This study aimed to evaluate whether adjuvant radiotherapy (RT) can improve the treatment outcome of patients with locally advanced gastric cancer who underwent extensive lymph node dissection (ELND). Materials and Methods: This retrospective study included patients with gastric cancer pathological stages IIA–IIIC at Taipei Tzu Chi Hospital between 2008 and 2015. Patients (a) aged >80 years, (b) with distant metastasis at diagnosis, (c) with coexisting malignancies, (d) who did not complete the prescribed RT course, and (e) who died 1 month after surgery were excluded. Among 420 patients diagnosed with gastric cancer, 98 were included. Results: The median follow-up was 24.5 months. Of 39 patients who underwent adjuvant RT, 38 also received adjuvant chemotherapy (CT). Of 59 patients who did not receive adjuvant RT, only 34 received adjuvant CT. ELND was performed in 67.3% of the patients. The 5-year overall survival (OS) rate was 40%. In the univariate analyses, adjuvant CT regimen, 5-fluorouracil + leucovorin, was associated with worst outcome, while TS-1 was associated with better survival outcome (P = 0.018). The number of involved lymph nodes was strongly related to the OS and disease-free survival (DFS) (P < 0.001). We tried using different numbers of involved lymph nodes as a cutoff point and found that adjuvant RT significantly improved both OS and DFS in patients whose involved lymph nodes were ≥4 (OS, P = 0.017; DFS, P = 0.015). In multivariate analyses, better DFS was associated with negative surgical margin (P = 0.04), earlier disease stage (P = 0.001), adjuvant radiotherapy (P = 0.045), and adjuvant CT regimen TS-1 (P = 0.001). Conclusion: Adjuvant RT could improve DFS of patients with locally advanced gastric cancer with or without ELND. When the number of involved lymph nodes is ≥4, adjuvant RT is strongly suggested.
Collapse
Affiliation(s)
- Yu-Nong Wang
- Department of Radiation Oncology, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei, Taiwan
| | - Shou-Yi Chang
- Department of Hematology/Oncology, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei, Taiwan
| | - Jing-Min Hwang
- Department of Radiation Oncology, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei, Taiwan.,Department of Radiology, School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - You-Kang Chang
- Department of Radiation Oncology, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei, Taiwan.,Department of Radiology, School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Woei-Yau Kao
- Department of Hematology/Oncology, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei, Taiwan.,Department of Oncology, School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Hsiang-Lin Wan
- Department of Hematology/Oncology, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei, Taiwan
| | - I-Shiang Tzeng
- Department of Research, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei, Taiwan
| | - Chao-Chuan Wu
- Department of General Surgery, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei, Taiwan.,Department of Surgery, School of Medicine, Tzu Chi University, Hualien, Taiwan
| |
Collapse
|
221
|
Yang F, Yan Z, Nie W, Cheng X, Liu Z, Wang W, Shao C, Fu G, Yu Y. LACTB induced apoptosis of oxaliplatin-resistant gastric cancer through regulating autophagy-mediated mitochondrial apoptosis pathway. Am J Transl Res 2021; 13:601-616. [PMID: 33594312 PMCID: PMC7868839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 11/30/2020] [Indexed: 06/12/2023]
Abstract
Oxaliplatin (OXA), as a third-generation platinum anticancer drug, is a treatment drug for gastric cancer (GC). However, OXA resistance has become the main reason for OXA treatment failure. Serine beta-lactamase-like protein (LACTB), acts as a mitochondrial protein, can affect multiple cancer processes. Here, we aimed to investigate the function and mechanism of LACTB in OXA-resistant GC. After LACTB overexpression or autophagy activator (RAPA) treatment, cell proliferation, reactive oxygen species (ROS), apoptosis, mitochondrial dysfunction were evaluated through CCK-8 assay, Edu staining, flow cytometry and immunofluorescence assay. Moreover, DNA double-stranded damage and autophagy-related proteins were examined via western blot. We revealed that LACTB was downregulated in OXA-resistant MGC-803 cells, and overexpression of LACTB reduced the resistance of GC cells to OXA. Besides, our results uncovered that overexpression of LACTB induced apoptosis, reduced the mitochondrial membrane potential (MMP) and accelerated ROS accumulation in OXA-resistant MGC-803 (MGC-803/OXA) cells. Meanwhile, we verified that overexpression of LACTB decreased glucose uptake and ATP synthesis, induced mitochondria and DNA damages, and inhibited autophagy of MGC-803/OXA cells. Furthermore, our results certified that RAPA could weaken the function of LACTB on apoptosis and mitochondrial morphology and function in OXA-resistant MGC-803 cells with OXA treatment. Therefore, we demonstrated that LACTB could attenuate the resistance of MGC-803/OXA cells to OXA through autophagy-mediated mitochondrial morphological changes, mitochondrial dysfunction, and apoptosis, suggesting that LACTB, functions as a suppressor, is conducive to the therapy of OXA-resistant GC.
Collapse
Affiliation(s)
- Fang Yang
- Department of Pathology, Guizhou Medical UniversityGuiyang, Guizhou, China
- College of Basic Medical Science, Guizhou Medical UniversityGuiyang, Guizhou, China
- Department of Laboratory Medicine, Guizhou Cancer HospitalGuiyang, Guizhou, China
- School of Clinical Laboratory Science, Guizhou Medical UniversityGuiyang, Guizhou, China
| | - Zhiqiang Yan
- Department of Gastrointestinal Surgery, Affiliated Hospital of Guizhou Medical UniversityGuiyang, Guizhou, China
| | - Wei Nie
- Department of Laboratory Medicine, Guizhou Cancer HospitalGuiyang, Guizhou, China
- School of Clinical Laboratory Science, Guizhou Medical UniversityGuiyang, Guizhou, China
| | - Xingzhen Cheng
- Department of Gastrointestinal Surgery, Affiliated Hospital of Guizhou Medical UniversityGuiyang, Guizhou, China
| | - Zeying Liu
- Department of Laboratory Medicine, Guizhou Cancer HospitalGuiyang, Guizhou, China
- School of Clinical Laboratory Science, Guizhou Medical UniversityGuiyang, Guizhou, China
| | - Wei Wang
- Department of Laboratory Medicine, Guizhou Cancer HospitalGuiyang, Guizhou, China
| | - Chunyan Shao
- Department of Laboratory Medicine, Guizhou Cancer HospitalGuiyang, Guizhou, China
| | - Gui Fu
- Department of Laboratory Medicine, Guizhou Cancer HospitalGuiyang, Guizhou, China
| | - Yanni Yu
- Department of Pathology, Guizhou Medical UniversityGuiyang, Guizhou, China
- College of Basic Medical Science, Guizhou Medical UniversityGuiyang, Guizhou, China
| |
Collapse
|
222
|
Tumor suppressor lnc-CTSLP4 inhibits EMT and metastasis of gastric cancer by attenuating HNRNPAB-dependent Snail transcription. MOLECULAR THERAPY-NUCLEIC ACIDS 2021; 23:1288-1303. [PMID: 33717650 PMCID: PMC7907227 DOI: 10.1016/j.omtn.2021.02.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 02/05/2021] [Indexed: 02/07/2023]
Abstract
Tumor metastasis is a crucial impediment to the treatment of gastric cancer (GC), and the epithelial-to-mesenchymal transition (EMT) program plays a critical role for the initiation of GC metastasis. Thus, the aim of this study is to investigate the regulation of lnc-CTSLP4 in the EMT process during GC progression. We found that lnc-CTSLP4 was significantly downregulated in GC tumor tissues compared with adjacent non-tumor tissues, and its levels in GC tumor tissues were closely correlated with tumor local invasion, TNM stage, lymph node metastasis, and prognosis of GC patients. Loss- and gain-of-function assays indicated that lnc-CTSLP4 inhibited GC cell migration, invasion, and EMT in vitro, as well as peritoneal dissemination in vivo. Mechanistic analysis demonstrated that lnc-CTSLP4 could bind with Hsp90α/heterogeneous nuclear ribonucleoprotein AB (HNRNPAB) complex and recruit E3-ubiquitin ligase ZFP91 to induce the degradation of HNRNPAB, thus suppressing the transcriptional activation of Snail and ultimately reversing EMT of GC cells. Taken together, our results suggest that lnc-CTSLP4 is significantly downregulated in GC tumor tissues and inhibits metastatic potential of GC cells by attenuating HNRNPAB-dependent Snail transcription via interacting with Hsp90α and recruiting E3 ubiquitin ligase ZFP91, which shows that lnc-CTSLP4 could serve as a prognostic biomarker and therapeutic target for metastatic GC.
Collapse
|
223
|
Lu J, Bang H, Kim SM, Cho SJ, Ashktorab H, Smoot DT, Zheng CH, Ryeom SW, Yoon SS, Yoon C, Lee JH. Lymphatic metastasis-related TBL1XR1 enhances stemness and metastasis in gastric cancer stem-like cells by activating ERK1/2-SOX2 signaling. Oncogene 2021; 40:922-936. [PMID: 33288885 DOI: 10.1038/s41388-020-01571-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 10/20/2020] [Accepted: 11/17/2020] [Indexed: 12/20/2022]
Abstract
The poor prognosis of gastric cancer (GC) results largely from metastasis and chemotherapy resistance. Toward novel therapeutic strategies that target or evade these phenomena, we evaluated the function of the transcriptional regulator transducin (β)-like 1 X-linked receptor 1 (TBL1XR1) in GC cells, including stem-like cells. In this study, the correlation of expression of TBL1XR1 and clinical features and GC patients' outcomes was evaluated. Knockdown or exogenous expression of TBL1XR1 was combined with in vitro (2D and 3D cultures) and in vivo (mouse lung and lymphatic metastasis models) assays to evaluate the function of TBL1XR1. TBL1XR1's downstream signaling was delineated by phospho-kinase array and knockdown of candidate mediators. Analysis of clinical data showed that TBL1XR1 overexpression was correlated with worse prognosis. Functional assays showed that TBL1XR1 promoted stemness, epithelial-mesenchymal transition (EMT), and lung and lymphatic metastasis in GC cells. TBL1XR1 activated ERK1/2-Sox2 signaling and was dependent on signaling via PI3K/AKT, in GC stem-like cells distinguished by CD44 expression. Moreover, inhibition of these signaling proteins reversed chemoresistance in in vitro and in vivo models. Taken together, our results indicate that TBL1XR1 promotes stemness and metastasis in GC, making it a potential prognostic indicator. The PI3K/AKT-TBL1XR1-ERK1/2-Sox2 axis may represent a target for the treatment of GC.
Collapse
Affiliation(s)
- Jun Lu
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fujian, China
| | - Heejin Bang
- Department of Pathology, Kangnam Sacred Heart Hospital, Hallym University College of Medicine, Seoul, South Korea
| | - Su Mi Kim
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
- Department of Surgery, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Gyeonggi-do, South Korea
| | - Soo-Jeong Cho
- Department of Internal Medicine, Liver Research Institute, Seoul National University Hospital, Seoul, South Korea
| | | | - Duane T Smoot
- Department of Medicine, Howard University, Washington, DC, USA
| | - Chao-Hui Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fujian, China
| | - Sandra W Ryeom
- Department of Cancer Biology, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Sam S Yoon
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Changhwan Yoon
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | - Jun Ho Lee
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea.
| |
Collapse
|
224
|
Pan S, Li K, Huang B, Huang J, Xu H, Zhu Z. Efficacy and safety of immune checkpoint inhibitors in gastric cancer: a network meta-analysis of well-designed randomized controlled trials. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:290. [PMID: 33708917 PMCID: PMC7944325 DOI: 10.21037/atm-20-6639] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Background Immune checkpoint inhibitors (ICIs) that inhibit the programmed death 1 (PD-1)/programmed death-ligand 1 (PD-L1) and cytotoxic T-lymphocyte antigen 4 (CTLA-4) interactions have shown promising prospects as treatment options for advanced gastric cancer (AGC). This manuscript analyzed well designed clinical trials to evaluate the efficacy and safety of immunotherapy in AGC. Methods PubMed, Embase, the Cochrane Library, and Medline were searched for randomized controlled trials (RCTs) of AGC treatments that were published before April 2020. Progression-free survival (PFS), overall survival (OS), objective response rate (ORR), and treatment-related adverse events (TRAEs) were evaluated to determine the efficacy and safety of ICIs. Network meta-analysis was performed using a random-effects model under the Bayesian framework. The ability of each treatment was ranked using the surface under the cumulative ranking (SUCRA) curve. Results Our analysis included five studies having seven immunotherapy regimens and 1,730 patients. The network meta-analysis showed that nivolumab 1 mg/kg every 3 weeks plus ipilimumab 3 mg/kg every 3 weeks (88.369%) was the regimen most likely to improve PFS. Nivolumab 3 mg/kg every 3 weeks (84.563%) and nivolumab 1 mg/kg every 3 weeks plus ipilimumab 3 mg/kg every 3 weeks (84.556%) were similarly best for OS outcome with excellent tolerance. The regimen of avelumab 10 mg/kg every 2 weeks (91.167%) had the lowest TRAEs. All immunotherapies had similar response rates. Conclusions We recommend nivolumab 3 mg/kg every 2 weeks or nivolumab 1 mg/kg every 3 weeks plus ipilimumab 3 mg/kg every 3 weeks as the preferred regimen due to their high efficacies.
Collapse
Affiliation(s)
- Siwei Pan
- Department of Surgical Oncology, First Hospital of China Medical University, Shenyang, China.,Department of Surgical Oncology and General Surgery, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Kai Li
- Department of Surgical Oncology, First Hospital of China Medical University, Shenyang, China.,Department of Surgical Oncology and General Surgery, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Baojun Huang
- Department of Surgical Oncology, First Hospital of China Medical University, Shenyang, China.,Department of Surgical Oncology and General Surgery, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Jinyu Huang
- Department of Surgical Oncology, First Hospital of China Medical University, Shenyang, China.,Department of Surgical Oncology and General Surgery, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Huimian Xu
- Department of Surgical Oncology, First Hospital of China Medical University, Shenyang, China.,Department of Surgical Oncology and General Surgery, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Zhi Zhu
- Department of Surgical Oncology, First Hospital of China Medical University, Shenyang, China.,Department of Surgical Oncology and General Surgery, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
225
|
Pakbin B, Pishkhan Dibazar S, Allahyari S, Javadi M, Farasat A, Darzi S. Probiotic Saccharomyces cerevisiae var. boulardii supernatant inhibits survivin gene expression and induces apoptosis in human gastric cancer cells. Food Sci Nutr 2021; 9:692-700. [PMID: 33598154 PMCID: PMC7866606 DOI: 10.1002/fsn3.2032] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Revised: 10/12/2020] [Accepted: 11/10/2020] [Indexed: 01/17/2023] Open
Abstract
Natural anticancer drug and compounds with other great benefits are of interest recently due to lower side effects than chemotherapy for cancer treatment and prevention. Different natural and synthetic drugs have been suggested to be used for treatment of gastric cancers, the second deadly cancer worldwide. The aim of this study was to investigate anticancer activity of SBS including inducing apoptosis and inhibition of survivin gene expression in gastric cancer cells. We evaluated cell viability, inducing apoptosis and change in survivin gene expression of EPG85-257P (EPG) and EPG85-257RDB (resistant to Daunorubicin, RDB) cell lines under exposure of SBS after 24, 48, and 72 hr. We found that SBS decreased cell viability, induced apoptosis, and reduced survivin gene expression in treated EPG and RDB cells (with the significant IC50 values of 387 and 575 µg/ml after 72 and 48 hr for EPG and RDB cells respectively). However, we observed SBS was more efficient to induce apoptosis in EPG than RDB cells. We strongly suggest SBS be considered as a prospective anticancer agent or in formulation of complementary medication to treat and prevent gastric cancers.
Collapse
Affiliation(s)
- Babak Pakbin
- Department of Food Hygiene and Quality of ControlFaculty of Veterinary MedicineUniversity of TehranTehranIran
| | | | - Samaneh Allahyari
- Department of Food Hygiene and SafetySchool of HealthQazvin University of Medical sciencesQazvinIran
| | - Maryam Javadi
- Children Growth and Development Research CenterResearch Institute for prevention of Non‐ Communicable DiseaseQazvin University of Medical SciencesQazvinIran
- Cellular and Molecular Research CenterResearch Institute for prevention of Non‐Communicable DiseaseQazvin University of Medical SciencesQazvinIran
| | - Alireza Farasat
- Cellular and Molecular Research CenterResearch Institute for prevention of Non‐Communicable DiseaseQazvin University of Medical SciencesQazvinIran
- Department of Medical BiotechnologyQazvin University of Medical SciencesQazvinIran
| | - Sina Darzi
- Health Products Safety Research CenterQazvin University of Medical ScienceQazvinIran
| |
Collapse
|
226
|
Lv H, Zhang J, Sun K, Nie C, Chen B, Wang J, Xu W, Wang S, Liu Y, Chen X. Expression of Human Epidermal Growth Factor Receptor-2 Status and Programmed Cell Death Protein-1 Ligand Is Associated With Prognosis in Gastric Cancer. Front Oncol 2021; 10:580045. [PMID: 33598422 PMCID: PMC7882725 DOI: 10.3389/fonc.2020.580045] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Accepted: 12/14/2020] [Indexed: 11/23/2022] Open
Abstract
Background PD-L1 and HER-2 are routine biomarkers for gastric cancer (GC). However, little research has been done to investigate the correlation among PD-L1, HER-2, immune microenvironment, and clinical features in GC. Methods Between January 2013 and May 2020, a total of 120 GC patients treated with chemotherapy were admitted to Henan Tumor Hospital. We retrospectively identified PD-L1, HER-2 level before chemotherapy and abstracted clinicopathologic features and treatment outcomes. Univariate and multivariate survival analyses were performed to assess the relationship between PD-L1/HER-2 levels and progression-free survival (PFS). The mRNA and tumor microenvironment of 343 patients with GC from The Cancer Genome Atlas (TCGA) were used to explore the underlying mechanism. Results We retrospectively analyzed 120 patients with gastric cancer, including 17 patients with HER-2 positive and 103 patients with HER-2 negative GC. The results showed that the expression of PD-L1 was closely correlated with HER-2 (P = 0.015). Patients with PD-L1/HER-2 positive obtained lower PFS compared to PD-L1/HER-2 negative (mPFS: 6.4 vs. 11.1 months, P = 0.014, mPFS: 5.3 vs. 11.1 months, P = 0.002, respectively), and the PD-L1 negative and HER-2 negative had the best PFS than other groups (P = 0.0008). In a multivariate model, PD-L1 status, HER-2 status, tumor location, and tumor differentiation remained independent prognostic indicators for PFS (P < 0.05). The results of database further analysis showed that the proportion of PD-L1+/CD8A+ in HER-2 negative patients was higher than that in HER-2 positive patients (37.6 vs 20.3%), while PD-L1−/CD8A− was significantly higher in HER-2 positive patients than HER-2 negative patients (57.8 vs. 28.8%). In addition, it showed that not only CD4+T cells, macrophages, and CD8+T cells, but also the associated inflammatory pathways such as IFN-γ/STAT1 were associated with HER-2. Conclusion HER-2 status could predict the efficacy of immune checkpoint inhibitors, and HER-2 status combined with PD-L1 level could predict the prognosis of GC patients.
Collapse
Affiliation(s)
- Huifang Lv
- Department of Oncology, Henan Cancer Hospital, Zhengzhou, China
| | - Junling Zhang
- Medical Department, 3D Medicines Inc., Shanghai, China
| | - Keran Sun
- Department of Oncology, Henan Cancer Hospital, Zhengzhou, China
| | - Caiyun Nie
- Department of Oncology, Henan Cancer Hospital, Zhengzhou, China
| | - Beibei Chen
- Department of Oncology, Henan Cancer Hospital, Zhengzhou, China
| | - Jianzheng Wang
- Department of Oncology, Henan Cancer Hospital, Zhengzhou, China
| | - Weifeng Xu
- Department of Oncology, Henan Cancer Hospital, Zhengzhou, China
| | - Saiqi Wang
- Department of Oncology, Henan Cancer Hospital, Zhengzhou, China
| | - Yingjun Liu
- Department of Surgery, Henan Cancer Hospital, Zhengzhou, China
| | - Xiaobing Chen
- Department of Oncology, Henan Cancer Hospital, Zhengzhou, China
| |
Collapse
|
227
|
Yang X, Cai S, Shu Y, Deng X, Zhang Y, He N, Wan L, Chen X, Qu Y, Yu S. Exosomal miR-487a derived from m2 macrophage promotes the progression of gastric cancer. Cell Cycle 2021; 20:434-444. [PMID: 33522393 DOI: 10.1080/15384101.2021.1878326] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Tumor-associated macrophages contribute to cell growth, development, and metastasis in various cancers. However, the underlying mechanisms of M2 macrophage that modulate the progression of gastric cancer (GC) remain largely unknown. In this study, we detected the ratio of macrophages in GC tissues and found that the proportion of M2 macrophages was increased in GC tissues. We then co-cultured GC cells with M1 and M2 macrophages, respectively, and then assessed cell proliferation and tumorigenicity of GC cells by MTT and colony formation assay. The results indicated that M2 macrophages promoted the proliferation of GC cells, but M1 not. Besides, GW4869, an exosomes inhibitor, reduced the effects induced by M2 macrophage. Then, we isolated and identified exosomes derived from M1 and M2 macrophage, and confirmed that the exosomes could be taken up by GC cells. We demonstrated that M2 macrophage-exosomes could induce the proliferation and tumorigenesis in vitro and in vivo. Moreover, miR-487a was enriched in M2 macrophage-exosomes and further determined that miR-487a exert the functions by targeting TIA1. In conclusion, exosomal miR-487a derived from M2 macrophage promotes the proliferation and tumorigenesis in gastric cancer, and the novel findings might be helpful to the development of novel diagnostic and therapeutic methods in GC.
Collapse
Affiliation(s)
- Xuefeng Yang
- Department of Gastrointestinal Surgery, Affiliated Hospital of Zunyi Medical University , Zunyi, China.,Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Zunyi Medical University , Zunyi, China
| | - Shuang Cai
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Zunyi Medical University , Zunyi, China
| | - Yue Shu
- Key Laboratory of Brain Science, Guizhou Key Laboratory of Anesthesia and Organ Protection, Zunyi Medical University , Zunyi, China
| | - Xun Deng
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Zunyi Medical University , Zunyi, China
| | - Yuanwei Zhang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Zunyi Medical University , Zunyi, China
| | - Nian He
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Zunyi Medical University , Zunyi, China
| | - Lei Wan
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Zunyi Medical University , Zunyi, China
| | - Xu Chen
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Zunyi Medical University , Zunyi, China
| | - Yan Qu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Zunyi Medical University , Zunyi, China
| | - Shouyang Yu
- Key Laboratory of Brain Science, Guizhou Key Laboratory of Anesthesia and Organ Protection, Zunyi Medical University , Zunyi, China
| |
Collapse
|
228
|
Park H, Jin RU, Wang-Gillam A, Suresh R, Rigden C, Amin M, Tan BR, Pedersen KS, Lim KH, Trikalinos NA, Acharya A, Copsey ML, Navo KA, Morton AE, Gao F, Lockhart AC. FOLFIRINOX for the Treatment of Advanced Gastroesophageal Cancers: A Phase 2 Nonrandomized Clinical Trial. JAMA Oncol 2021; 6:1231-1240. [PMID: 32469386 DOI: 10.1001/jamaoncol.2020.2020] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Importance Standard first-line regimens for patients with metastatic gastroesophageal adenocarcinomas have an approximate 40% objective response rate (ORR). The combination of leucovorin, fluorouracil, irinotecan, and oxaliplatin (FOLFIRINOX) has been efficacious as first-line therapy for other gastrointestinal cancers, such as pancreatic and colon cancers. Objective To evaluate the clinical activity and safety of FOLFIRINOX as first-line treatment for patients with advanced gastroesophageal adenocarcinoma. Design, Setting, and Participants This is an open-label, single-arm phase 2 study of first-line FOLFIRINOX in patients with advanced gastroesophageal adenocarcinoma. Estimated sample size included 41 patients with ERBB2-negative disease with 90% power to detect an ORR of 60% or greater with α of .10. No enrollment goal was planned for ERBB2-positive patients, but they were allowed to receive trastuzumab in combination with FOLFIRINOX. Interventions Starting doses were fluorouracil, 400 mg/m2 bolus, followed by 2400 mg/m2 over 46 hours; leucovorin, 400 mg/m2; irinotecan, 180 mg/m2; and oxaliplatin, 85 mg/m2. Trastuzumab was administered as a 6 mg/kg loading dose, followed by 4 mg/kg every 14 days in patients with ERBB2-positive disease. Main Outcomes and Measures The primary end point was ORR by the Response Evaluation Criteria in Solid Tumors, version 1.1. Secondary end points included safety profile, progression-free survival (PFS), overall survival (OS), and duration of response. Results From November 2013 to May 2018, 67 patients were enrolled (median [range] age, 59.0 [34-78] years; including 56 [84%] men), and 26 of 67 (39%) had ERBB2-positive disease. Median follow-up was 17.4 months. The ORR was 61%(95% CI, 44.5%-75.8%) (25 of 41) in the ERBB2-negative group and 85% (95% CI, 65.1%-95.6%) (22 of 26) in the ERBB2-positive group, including 1 patient with complete response. For ERBB2-negative patients, median PFS was 8.4 months and median OS was 15.5 months; for ERBB2-positive patients, median PFS was 13.8 months and median OS was 19.6 months. Fifty-six patients (84%) had dose modifications or treatment delays. The most common toxic effects were neutropenia (91%, n = 61), diarrhea (63%, n = 42), peripheral sensory neuropathy (61%, n = 41), and nausea (48%, n = 32), with no unexpected toxic effects. Conclusions and Relevance The FOLFIRINOX regimen with or without trastuzumab was associated with improved ORR and PFS in patients with advanced gastroesophageal adenocarcinoma in the first-line setting. This regimen may be a reasonable therapeutic option for patients with preserved performance status. Trial Registration ClinicalTrials.gov Identifier: NCT01928290.
Collapse
Affiliation(s)
- Haeseong Park
- Division of Oncology, Washington University School of Medicine in St Louis, St Louis, Missouri
| | - Ramon U Jin
- Division of Oncology, Washington University School of Medicine in St Louis, St Louis, Missouri
| | - Andrea Wang-Gillam
- Division of Oncology, Washington University School of Medicine in St Louis, St Louis, Missouri
| | - Rama Suresh
- Division of Oncology, Washington University School of Medicine in St Louis, St Louis, Missouri
| | - Caron Rigden
- Division of Oncology, Washington University School of Medicine in St Louis, St Louis, Missouri
| | - Manik Amin
- Division of Oncology, Washington University School of Medicine in St Louis, St Louis, Missouri
| | - Benjamin R Tan
- Division of Oncology, Washington University School of Medicine in St Louis, St Louis, Missouri
| | - Katrina S Pedersen
- Division of Oncology, Washington University School of Medicine in St Louis, St Louis, Missouri
| | - Kian-Huat Lim
- Division of Oncology, Washington University School of Medicine in St Louis, St Louis, Missouri
| | - Nikolaos A Trikalinos
- Division of Oncology, Washington University School of Medicine in St Louis, St Louis, Missouri
| | - Abhilasha Acharya
- Division of Oncology, Washington University School of Medicine in St Louis, St Louis, Missouri
| | - Megan L Copsey
- Division of Oncology, Washington University School of Medicine in St Louis, St Louis, Missouri
| | - Katherine A Navo
- Division of Oncology, Washington University School of Medicine in St Louis, St Louis, Missouri
| | - Ashley E Morton
- Division of Oncology, Washington University School of Medicine in St Louis, St Louis, Missouri
| | - Feng Gao
- Division of Public Health Sciences, Washington University School of Medicine in St Louis, St Louis, Missouri
| | - A Craig Lockhart
- Sylvester Comprehensive Cancer Center, Division of Medical Oncology, Department of Internal Medicine, Leonard M. Miller School of Medicine, University of Miami, Miami, Florida
| |
Collapse
|
229
|
Chen W, Zhang K, Yang Y, Guo Z, Wang X, Teng B, Zhao Q, Huang C, Qiu Z. MEF2A-mediated lncRNA HCP5 Inhibits Gastric Cancer Progression via MiR-106b-5p/p21 Axis. Int J Biol Sci 2021; 17:623-634. [PMID: 33613117 PMCID: PMC7893594 DOI: 10.7150/ijbs.55020] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 12/31/2020] [Indexed: 12/16/2022] Open
Abstract
Background: Long non-coding RNAs (lncRNAs) are deemed to be relevant to the tumorigenesis and development of a variety of tumors, containing gastric cancer (GC). The purpose of our investigations is to explore the character of HCP5 in GC. Methods: HCP5 expression was detected by quantitative real-time polymerase chain reaction (qRT-PCR) in 62 matched GC tissues and corresponding para-carcinoma tissues. In vitro and in vivo functional assays were subjected to verify the biological effects of HCP5 after alteration of HCP5. Chromatin immunoprecipitation assay (CHIP) assays were conducted to confirm that myocyte enhancer factor 2A (MEF2A) could bind to HCP5 promoter regions and thereby induce HCP5 expression. Analysis of the latent binding of miR-106b-5p to HCP5 and p21 was made by bioinformatics prediction and luciferase reporter assays. Results: Significant downregulation of HCP5 was detected in GC tissues. Negative correlation was determined between HCP5 expression level and tumor size and overall survival in GC patients. HCP5 depletion had a facilitating impact on proliferation, migration and invasion of GC cells. Consistently, overexpression of HCP5 came into an opposite effect. Moreover, we demonstrated that MEF2A could combine with the promoter region of HCP5 and thereby induce HCP5 transcription. Luciferase reporter assays revealed that HCP5 could compete with miR-106b-5p as a competing endogenous RNA (ceRNA) and upregulated p21 expression in GC. Conclusions: MEF2A-mediated HCP5 could exert an anti-tumor effect among the development of GC via miR-106b-5p/p21 axis, which provides a novel target for GC therapy.
Collapse
Affiliation(s)
- Weiwei Chen
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, 100 Haining Road, Shanghai, 201600, China
| | - Kundong Zhang
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, 100 Haining Road, Shanghai, 201600, China
| | - Yuhan Yang
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, 100 Haining Road, Shanghai, 201600, China
| | - Zengya Guo
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, 100 Haining Road, Shanghai, 201600, China
| | - Xiaofeng Wang
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, 100 Haining Road, Shanghai, 201600, China
| | - Buwei Teng
- Lianyungang Clinical College of Nanjing Medical University/The First People's Hospital of Lianyungang, 6 Zhenhua East Road, Haizhou District, City of Lianyungang, Jiangsu Province, 222061, China
| | - Qian Zhao
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of National Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Chen Huang
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, 100 Haining Road, Shanghai, 201600, China
| | - Zhengjun Qiu
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, 100 Haining Road, Shanghai, 201600, China
| |
Collapse
|
230
|
Miao X, Liu Y, Fan Y, Wang G, Zhu H. LncRNA BANCR Attenuates the Killing Capacity of Cisplatin on Gastric Cancer Cell Through the ERK1/2 Pathway. Cancer Manag Res 2021; 13:287-296. [PMID: 33469371 PMCID: PMC7811444 DOI: 10.2147/cmar.s269679] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Accepted: 11/27/2020] [Indexed: 12/12/2022] Open
Abstract
Purpose Chemotherapy-based comprehensive treatments are the most important therapeutic methods for patients with advanced gastric cancer, but chemoresistance often cause treatment failure. Long non-coding RNA (LncRNA) BRAF-activated non-coding RNA (BANCR) has been shown to participate in many biological behaviors of multiple cancers. However, the biological roles of LncRNA BANCR in chemoresistance of gastric cancer remain unclear. Here, we aimed to evaluate the functions of LncRNA BANCR on the therapy of gastric cancer. Methods In this study, LncRNA BANCR expression was detected in gastric cancer patient samples and cell lines by quantity polymerase chain reaction (qPCR). Cell proliferation and viability in cisplatin-treated cells were measured using clonogenic survival assay and cell counting kit-8. The levels of ERK1/2 pathway molecules were tested with Western blot. Ly3214996, an inhibitor of ERK signal pathway, was administered to assess the effects of BANCR overexpression on gastric cancer cell with cisplatin-treated resistance. Moreover, the role of BANCR in cisplatin resistance of gastric cancer was validated in xenograft mouse models in vivo. Results Our study revealed that LncRNA BANCR expression was also significantly increased in gastric cancer tissues compared with adjacent normal tissues. Furthermore, we found that BANCR overexpression promoted gastric cancer cell resistance to cisplatin in vitro. Ly3214996 treatment abolished the BANCR overexpression-mediated gastric cancer cell cisplatin resistance via regulating the phosphorylation of ERK protein. Knock-down of BANCR significantly delayed tumor growth in xenograft mouse models. Conclusion BANCR promoted cisplatin resistance of gastric cancer cells by activating ERK1/2 pathway. Inhibition of BANCR markedly suppressed the growth of gastric cancer cells in vitro as well as in vivo. These results provided a new strategy for gastric cancer therapy via targeting BANCR.
Collapse
Affiliation(s)
- Xiang Miao
- Department of General Surgery, Lianyungang Municipal Oriental Hospital, Lianyungang, Jiangsu 222042, People's Republic of China
| | - Yixiang Liu
- Department of General Surgery, Lianyungang Municipal Oriental Hospital, Lianyungang, Jiangsu 222042, People's Republic of China
| | - Yuzhu Fan
- Department of General Surgery, Lianyungang Municipal Oriental Hospital, Lianyungang, Jiangsu 222042, People's Republic of China
| | - Guoqiang Wang
- Department of General Surgery, Lianyungang Municipal Oriental Hospital, Lianyungang, Jiangsu 222042, People's Republic of China
| | - Hongbo Zhu
- Department of General Surgery, Lianyungang Municipal Oriental Hospital, Lianyungang, Jiangsu 222042, People's Republic of China
| |
Collapse
|
231
|
Attia H, Smyth E. Evolving therapies in advanced oesophago-gastric cancers and the increasing role of immunotherapy. Expert Rev Anticancer Ther 2021; 21:535-546. [PMID: 33349073 DOI: 10.1080/14737140.2021.1866548] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
INTRODUCTION Esophagogastric cancers remain a considerable health burden and among the top causes of global cancer-related deaths. Chemotherapy remains the cornerstone of treatment for patients with advanced disease. Doublet platinum/fluoropyrimidine therapy is established as first-line treatment with the option of adding a taxane in selected patients. Irinotecan, taxanes, and ramucirumab are approved as second-line treatments. Results from the trials KEYNOTE-059, ATTRACTION-2, and TAGS have established the use of immune checkpoint inhibitors and trifluridine/tipiracil as a third-line treatment. High PD-L1 expression, microsatellite instability, tumor mutational burden, and Epstein-Barr virus status may also be used to enrich for responses to immunotherapy. AREAS COVERED In this review, we discuss the outcome of recent trials in the later lines of therapy for esophagogastric cancer and place these in the context of current treatment paradigms. We also discuss the biology of esophagogastric cancers and how this might inform the development of new treatments. Finally, we comment on promising new drugs in development. EXPERT OPINION Recent advances in the treatment of chemo-refractory esophagogastric cancer add to the improving survival of patients with this disease. Further research is needed to improve patient selection to therapies and the earlier incorporation of these agents in the treatment journey.
Collapse
Affiliation(s)
- Hossameldin Attia
- Cambridge University Hospitals NHS Foundation Trust, Addenbrooke's Hospital, Cambridge, UK
| | - Elizabeth Smyth
- Cambridge University Hospitals NHS Foundation Trust, Addenbrooke's Hospital, Cambridge, UK
| |
Collapse
|
232
|
Berger Y, Giurcanu M, Vining CC, Schuitevoerder D, Posner MC, Roggin KK, Polite BN, Liao CY, Eng OS, Catenacci DVT, Turaga KK. Cytoreductive Surgery for Selected Patients Whose Metastatic Gastric Cancer was Treated with Systemic Chemotherapy. Ann Surg Oncol 2021; 28:4433-4443. [PMID: 33420565 DOI: 10.1245/s10434-020-09475-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 12/01/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND The authors hypothesized that cytoreductive surgery (CRS, comprising gastrectomy combined with metastasectomy) in addition to systemic chemotherapy (SC) is associated with a better survival than chemotherapy alone for patients with metastatic gastric adenocarcinoma (MGA). METHODS Patients with MGA who received SC between 2004 and 2016 were identified using the National Cancer Database (NCDB). Nearest-neighbor 1:1 propensity score-matching was used to create comparable groups. Overall survival (OS) was compared between subgroups using Kaplan-Meier analyses. Immortal bias analysis was performed among those who survived longer than 90 days. RESULTS The study identified 29,728 chemotherapy-treated patients, who were divided into the following four subgroups: no surgery (NS, n = 25,690), metastasectomy alone (n = 1170), gastrectomy alone (n = 2248), and CRS (n = 620) with median OS periods of 8.6, 10.9, 14.8, and 16.3 months, respectively (p < 0.001). Compared with the patients who underwent NS, the patients who had CRS were younger (58.9 ± 13.4 vs 62.0 ± 13.1 years), had a lower proportion of disease involving multiple sites (4.6% vs 19.1%), and were more likely to be clinically occult (cM0 stage: 59.2% vs 8.3%) (p < 0.001 for all). The median OS for the propensity-matched patients who underwent CRS (n = 615) was longer than for those with NS (16.4 vs 9.3 months; p < 0.001), including in those with clinical M1 stage (n = 210). In the Cox regression model using the matched data, the hazard ratio for CRS versus NS was 0.56 (95% confidence interval [CI], 0.49-0.63). In the immortal-matched cohort, the corresponding median OS was 17.0 versus 9.5 months (p < 0.001). CONCLUSIONS In addition to SC, CRS may be associated with an OS benefit for a selected group of MGA patients meriting further prospective investigation.
Collapse
Affiliation(s)
- Yaniv Berger
- Department of Surgery, University of Chicago Medical Center, Chicago, IL, USA
| | - Mihai Giurcanu
- Department of Public Health Sciences, University of Chicago Medical Center, Chicago, IL, USA
| | - Charles C Vining
- Department of Surgery, University of Chicago Medical Center, Chicago, IL, USA
| | | | - Mitchell C Posner
- Department of Surgery, University of Chicago Medical Center, Chicago, IL, USA
| | - Kevin K Roggin
- Department of Surgery, University of Chicago Medical Center, Chicago, IL, USA
| | - Blase N Polite
- Department of Medicine, University of Chicago Medical Center, Chicago, IL, USA
| | - Chih-Yi Liao
- Department of Medicine, University of Chicago Medical Center, Chicago, IL, USA
| | - Oliver S Eng
- Department of Surgery, University of Chicago Medical Center, Chicago, IL, USA
| | | | - Kiran K Turaga
- Department of Surgery, University of Chicago Medical Center, Chicago, IL, USA.
| |
Collapse
|
233
|
Xu B, Li S, Fang Y, Zou Y, Song D, Zhang S, Cai Y. Proprotein Convertase Subtilisin/Kexin Type 9 Promotes Gastric Cancer Metastasis and Suppresses Apoptosis by Facilitating MAPK Signaling Pathway Through HSP70 Up-Regulation. Front Oncol 2021; 10:609663. [PMID: 33489919 PMCID: PMC7817950 DOI: 10.3389/fonc.2020.609663] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 11/12/2020] [Indexed: 01/24/2023] Open
Abstract
Objective To examine the effect of proprotein convertase subtilisin/kexin type 9 (PCSK9) on gastric cancer (GC) progression and prognosis, and to explore the underlying mechanism. Methods PCSK9 expression levels in human GC tissues were determined by quantitative real-time PCR, western blotting, and immunohistochemical assay. PCSK9 serum levels were detected by enzyme-linked immunosorbent assay. The relationships of PCSK9 and GC progression and survival were analyzed using the Chi-square test, Kaplan-Meier analysis, and Cox proportional hazards model. The effect of PCSK9 on cell invasion, migration, and apoptosis were determined in human GC cell lines and mouse xenograft model separately using PCSK9 knockdown and overexpression strategies. The PCSK9 interacting molecules, screened by co-immunoprecipitation combined with LC-MS/MS, were identified by immunofluorescence localization and western blotting. Additionally, the mitogen-activated protein kinase (MAPK) pathway was assessed by western blotting. Results PCSK9 mRNA and protein levels were significantly elevated in GC tissues compared with the paired normal tissues at our medical center (P < 0.001). Notably, the up-regulation of PCSK9 expression in GC tissues was related to tumor progression and poor survival. GC patients had higher serum levels of PCSK9 than the age-matched healthy controls (P < 0.001); PCSK9 promoted invasive and migratory ability and inhibited apoptosis in GC cells with no apparent affection in cell proliferation. The silencing of PCSK9 reversed these effects, suppressing tumor metastasis in vitro and in vivo. Furthermore, PCSK9 maintained these functions through up-regulating heat shock protein 70 (HSP70), ultimately facilitating the mitogen-activated protein kinase (MAPK) pathway. Conclusion Collectively, our data revealed that high PCSK9 expression levels in GC tissue were correlated with GC progression and poor prognosis and that PCSK9 could promote GC metastasis and suppress apoptosis by facilitating MAPK signaling pathway through HSP70 up-regulation. PCSK9 may represent a novel potential therapeutic target in GC.
Collapse
Affiliation(s)
- Beili Xu
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Shuyu Li
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yong Fang
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yanting Zou
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Dongqiang Song
- Department of Hepatic Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Shuncai Zhang
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yu Cai
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
234
|
The Prevalence of Blastocystis Infection in Pediatric Patients with Malignancy: A Single-Center Study in Ahvaz, Iran. ARCHIVES OF PEDIATRIC INFECTIOUS DISEASES 2021. [DOI: 10.5812/pedinfect.104068] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background: Blastocystis spp. is known as one of the few intestinal parasites, prevalent in more than 5% and 30 - 60% of the population in industrialized and developing countries, respectively. In this respect, immunocompromised individuals, such as patients undergoing chemotherapy or those with malignancies, are at risk of the clinical symptoms of Blastocystis infection; however, the given condition is often self-limiting in healthy individuals. Objectives: The current study aimed at evaluating the prevalence of Blastocystis infection in children with malignancies receiving chemo drugs. Methods: The current descriptive, cross-sectional study was conducted on 52 stool specimens collected from patients with cancer admitted to the Oncology Ward of Shahid Baqaei 2 Hospital, Ahvaz, Iran, for six months. A standardized questionnaire was filled out for all cases. Each specimen was also prepared using direct smear, the Lugol iodine staining, and the formalin-ether condensation method. Results: Blastocystis spp. was detected in 21.1% of the cases among them, 11.5% demonstrated gastrointestinal symptoms; therefore, a significant relationship was observed between Blastocystis infection and gastrointestinal symptoms. Conclusion: Patients undergoing chemotherapy should be screened for opportunistic parasitic infections such as Blastocystis to avoid potentially life-threatening outcomes. Besides, further studies are required to identify the subtypes of Blastocystis.
Collapse
|
235
|
Ishak NS, Abdul Rahman H, Lee SHF, Lu SK, Naing L. Incidence, Survival and Prognostic Factors of Oesophagogastric Cancer. J Gastrointest Cancer 2021; 53:130-143. [PMID: 33392958 DOI: 10.1007/s12029-020-00559-w] [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] [Accepted: 11/25/2020] [Indexed: 11/26/2022]
Abstract
INTRODUCTION Oesophagogastric cancer is one of the leading causes of cancer death worldwide due to its aggressive nature. Despite the high mortality rate, there is limited information regarding this cancer in Brunei. AIM To estimate the incidence and survival duration of oesophagogastric cancer patients, to identify prognostic factors of oesophagogastric cancer and associated factors for late-stage oesophagogastric cancer detection. METHODS A retrospective study on all oesophagogastric cancer patients registered in the population-based national cancer registry in Brunei from January 2010 to December 2018. Kaplan-Meier and Cox proportional hazard regression survival analyses and multiple logistic regression were applied. RESULTS Sixty-eight oesophagogastric cancer patients' data were retrieved from the registry. The incidence was 2.75 cases per 100,000 adults per year. Median survival time was 1.18 years (95% CI: 0.77, 1.80) and the 3-year survival rate was 26.3%. Age (61-70 years) (adjusted HR = 0.38; 95% CI: 0.17, 0.89; p = 0.025) and those who have undergone chemotherapy (adj. HR = 0.40; 95% CI: 0.18, 0.90; p = 0.026) have a significantly lower mortality risk. Obesity (adj. HR = 11.94; 95% CI: 1.94, 73.36; p = 0.007), and stage 4 (advanced stage) cancer (adj. HR=4.11; 95% CI: 1.97, 8.58; p< 0.001) have a significantly higher mortality risk. Females have 3-time odds (adj. OR = 3.05; 95% CI: 1.09, 9.02; p = 0.038) of presenting with stage 4 cancer. Smokers have 13-time odds (Adj. OR=12.99; 95% CI: 1.92, 262.0; p = 0.025) of presenting with stage 4 cancer. CONCLUSION Prognosis of oesophagogastric cancer remains poor. Addressing late detection and improve endoscopic surveillance and awareness of symptoms may help improve prognosis and mortality.
Collapse
Affiliation(s)
- Nurul Syuhada Ishak
- PAPRSB Institute of Health Sciences, Universiti Brunei Darussalam, Jalan Tungku Link, Gadong, BE1410, Brunei Darussalam
| | - Hanif Abdul Rahman
- PAPRSB Institute of Health Sciences, Universiti Brunei Darussalam, Jalan Tungku Link, Gadong, BE1410, Brunei Darussalam.
| | - Shirley H F Lee
- PAPRSB Institute of Health Sciences, Universiti Brunei Darussalam, Jalan Tungku Link, Gadong, BE1410, Brunei Darussalam
| | - Shir Kiong Lu
- The Brunei Cancer Centre, Pantai Jerudong Specialist Centre, Bandar Seri Begawan, Brunei
| | - Lin Naing
- PAPRSB Institute of Health Sciences, Universiti Brunei Darussalam, Jalan Tungku Link, Gadong, BE1410, Brunei Darussalam
| |
Collapse
|
236
|
Wang H, Lu Y, Wang M, Wu Y, Wang X, Li Y. Roles of E3 ubiquitin ligases in gastric cancer carcinogenesis and their effects on cisplatin resistance. J Mol Med (Berl) 2021; 99:193-212. [PMID: 33392633 DOI: 10.1007/s00109-020-02015-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 11/15/2020] [Accepted: 11/20/2020] [Indexed: 12/28/2022]
Abstract
Although gastric cancer (GC) is one of the most common cancers with high incidence and mortality rates, its pathogenesis is still not elucidated. GC carcinogenesis is complicated and involved in the activation of oncoproteins and inactivation of tumor suppressors. The ubiquitin-proteasome system (UPS) is crucial for protein degradation and regulation of physiological and pathological processes. E3 ubiquitin ligases are pivotal enzymes in UPS, containing various subfamily proteins. Previous studies report that some E3 ligases, including SKP2, CUL1, and MDM2, act as oncoproteins in GC carcinogenesis. On the other hand, FBXW7, FBXL5, FBXO31, RNF43, and RNF180 exert as tumor suppressors in GC carcinogenesis. Moreover, E3 ligases modulate cell growth, cell apoptosis, and cell cycle; thus, it is complicated to confer cisplatin resistance/sensitivity in GC cells. The intrinsic and acquired cisplatin resistance limits its clinical application against GC. In this review, we explore oncogenic and tumor suppressive roles of E3 ligases in GC carcinogenesis and focus on the effects of E3 ligases on cisplatin resistance in GC cells, which will provide novel therapeutic targets for GC therapy, especially for cisplatin-resistant patients.
Collapse
Affiliation(s)
- Huizhen Wang
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Yida Lu
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Mingliang Wang
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Youliang Wu
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Xiaodong Wang
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Yongxiang Li
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China.
| |
Collapse
|
237
|
External validity of docetaxel triplet trials in advanced gastric cancer: are there patients who still benefit? Gastric Cancer 2021; 24:445-456. [PMID: 32970266 PMCID: PMC7902567 DOI: 10.1007/s10120-020-01116-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Accepted: 08/23/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND The purpose of our study was to develop an online calculator to estimate the effect of docetaxel triplets (DPF) in first line of advanced gastric cancer (AGC), and to assess the external validity of docetaxel trials in individual patients. METHODS The study includes patients with HER2(-) AGC treated with platin and fluoropyrimidine (PF) or with DPF in first line. Treatment effect and interactions were assessed using Bayesian accelerated failure time models. RESULT The series comprises 1376 patients; 238 treated with DPF and 1138 with PF between 2008 and 2019. DPF was associated with increased progression-free survival (PFS) and overall survival (OS) with time ratio (TR) 1.27 (95% credible interval [CrI], 1.15-1.40), and TR 1.19 (95% CrI, 1.09-1.27), respectively. Serious adverse events were more common with DPF, particularly hematological effects (32% vs 22%). Younger participants received greater DPF dose density without achieving greater disease control, while severe toxicity was likewise higher. DPF yielded superior OS in Lauren intestinal (TR 1.27, 95% CrI, 1.08-1.11) vs diffuse subtype (TR 1.17, 95% CrI, 1.09-1.24) and the probability of increasing OS > 15% was 90% vs 67% in each subtype, respectively. The effect dwindles over time, which can be attributed to pathological changes and clinical practice changes. CONCLUSION Our study confirms the effect of DPF is highly dependent on several clinical-pathological variables, with discreet and gradually declining benefit over platinum doublets in later years, at the expense of increased toxicity. These results may help to underpin the idea that external validity of AGC trials should be revised regularly.
Collapse
|
238
|
Sissung TM, Cordes L, Peer CJ, Gandhy S, Redman J, Strauss J, Figg WD. Case report: severe toxicity in an African-American patient receiving FOLFOX carrying uncommon allelic variants in DPYD. Pharmacogenomics 2021; 22:81-85. [PMID: 33305610 PMCID: PMC7831885 DOI: 10.2217/pgs-2020-0120] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 10/26/2020] [Indexed: 12/25/2022] Open
Abstract
Cancers of the colon are commonly treated with fluoropyrimidines, which often cause severe toxicities in patients with certain variants in DPYD. Y186C (rs115232898) and a variant in the 3' untranslated region (rs12132152) are uncommon alleles previously observed in African-Americans. An African-American female underwent 5-fluorouracil-based therapy (400 mg/m2 bolus, 1200 mg/m2/day over 46 h). The patient experienced severe pancytopenia after the first cycle. After 5-fluorouracil (5-FU) dose reduction (600 mg/m2/day), the steady-state 5-FU plasma concentration became 474 ng/ml (range 301-619 ng/ml) and increased following a subsequence dose increase (800 mg/m2/day; 1248 ng/ml). After a 1000 mg/m2/day dose resulted in myelosuppression, 5-FU was again de-escalated for the remaining cycles (600 mg/m2). The observed complications are likely a function of uncommon genetic variants that affect DPYD metabolism.
Collapse
Affiliation(s)
- Tristan M Sissung
- Clinical Pharmacology Program, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Lisa Cordes
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Cody J Peer
- Clinical Pharmacology Program, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Shruti Gandhy
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Jason Redman
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Julius Strauss
- Laboratory of Tumor Immunology & Biology, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - William D Figg
- Clinical Pharmacology Program, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| |
Collapse
|
239
|
Cai L, Ouyang G, Wang X, Li Z, Shen Y. Radiotherapy Combined with Chemotherapy for Regional Lymph Node Recurrence in Gastric Cancer. Cancer Manag Res 2021; 12:13339-13346. [PMID: 33380839 PMCID: PMC7769069 DOI: 10.2147/cmar.s280225] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 11/23/2020] [Indexed: 01/26/2023] Open
Abstract
Purpose Regional lymph node recurrence (RLNR) in gastric cancer is uncommon. We investigated the effects of radiotherapy combined with chemotherapy against limited RLNR and analyzed the regularity of regional lymph node recurrence and metastasis. Patients and Methods This retrospective study included 34 gastric cancer patients with limited RLNR after D2 lymphadenectomy between January 2012 and May 2018. All patients received systemic chemotherapy and local radiotherapy with median dose of 52.5 Gy (30–66 Gy in fractions of 1.8–3.0 Gy daily, five times weekly). All sites of recurrent and metastatic lymph nodes were collected and analyzed. Results The median follow-up was 19 months (range 7–60 months). After treatment, complete response and partial response were observed in 32.4% and 55.9% of patients, respectively. The median overall survival (OS) and progression-free survival (PFS) were 18 months and 13 months. On multivariate analysis, age (≤60 vs >60) was associated with a significantly better OS (p = 0.025) and radiation dose (<54 Gy vs ≥54 Gy) was considered as an independent prognostic factor for PFS (p = 0.000). During radiotherapy, three patients developed grade 3 gastrointestinal toxicity, and no deaths were related to the treatments. The most commonly metastatic lymph nodes were the No. 4, No. 3, No. 6, No. 5, No. 7, No. 9, and No. 8 nodes; the recurrent lymph nodes were mainly located in the No. 16b, No. 16a, No. 9, No. 14, No. 7, No. 13, and No. 8 nodes. Conclusion The selected gastric cancer patients with limited RLNR may benefit from radiotherapy combined with chemotherapy. High-dose radiotherapy (≥54 Gy) lead to better PFS and tend to extend OS. The major lymph node recurrence sites were in the gastric vascular region (especially No. 16a/b nodes).
Collapse
Affiliation(s)
- Liang Cai
- Lung Cancer Center, West China School of Medicine, West China Hospital of Sichuan University, Chengdu, People's Republic of China
| | - Ganlu Ouyang
- Department of Abdominal Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Xin Wang
- Department of Abdominal Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Zhiping Li
- Department of Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Yali Shen
- Department of Abdominal Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| |
Collapse
|
240
|
Zhou KI, Peterson B, Serritella A, Thomas J, Reizine N, Moya S, Tan C, Wang Y, Catenacci DVT. Spatial and Temporal Heterogeneity of PD-L1 Expression and Tumor Mutational Burden in Gastroesophageal Adenocarcinoma at Baseline Diagnosis and after Chemotherapy. Clin Cancer Res 2020; 26:6453-6463. [PMID: 32820017 PMCID: PMC7744325 DOI: 10.1158/1078-0432.ccr-20-2085] [Citation(s) in RCA: 100] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 07/15/2020] [Accepted: 08/17/2020] [Indexed: 12/12/2022]
Abstract
PURPOSE Intrapatient heterogeneity of programmed death ligand 1 (PD-L1) expression and tumor mutational burden (TMB) in gastroesophageal adenocarcinoma (GEA) could influence their roles as predictive biomarkers for response to immune checkpoint inhibitors (ICI). In this retrospective analysis, we evaluated the spatiotemporal heterogeneity and prognostic relevance of PD-L1 expression and TMB in GEA. EXPERIMENTAL DESIGN A cohort of 211 patients with stage II-IV GEA was retrospectively reviewed for a total of 407 tumor samples with PD-L1 expression data and 319 tumor samples with TMB data. PD-L1 status was defined as positive if combined positive score (CPS) ≥1 using the 22C3 pharmDx assay. TMB levels were categorized as low, intermediate, or high (≤5, 5-15, or >15 mutations/Mb), or using a single threshold (<10 or ≥10 mutation/Mb), determined by next-generation sequencing using a targeted gene panel. RESULTS Of 407 tumors, 56% were PD-L1 negative and 44% PD-L1 positive. Of 319 tumors, 50% were TMB-low, 45% TMB-intermediate, and 5% TMB-high; 86% had <10 and 14% ≥10 mutations/Mb. TMB level was significantly associated with MSI-status. PD-L1 expression and TMB exhibited marked spatial heterogeneity between baseline primary and metastatic tumors (61% and 69% concordance), and temporal heterogeneity between tumors before and after chemotherapy (57%-63% and 73%-75% concordance). PD-L1 expression and TMB were not significantly associated with overall survival. CONCLUSIONS PD-L1 expression and TMB exhibit marked spatial and temporal heterogeneity in GEA. This heterogeneity should be considered when obtaining tumor samples for molecular testing and when deciding whether ICI therapy is appropriate.See related commentary by Klempner et al., p. 6401.
Collapse
Affiliation(s)
- Katherine I Zhou
- Medical Scientist Training Program, University of Chicago, Chicago, Illinois
| | - Bryan Peterson
- Section of Hematology/Oncology, Department of Medicine, University of Chicago Medical Center and Biological Sciences, Chicago, Illinois
| | - Anthony Serritella
- Department of Medicine, University of Chicago Medical Center, Chicago, Illinois
| | - Joseph Thomas
- Department of Medicine, University of Chicago Medical Center, Chicago, Illinois
| | - Natalie Reizine
- Section of Hematology/Oncology, Department of Medicine, University of Chicago Medical Center and Biological Sciences, Chicago, Illinois
| | - Stephanie Moya
- Section of Hematology/Oncology, Department of Medicine, University of Chicago Medical Center and Biological Sciences, Chicago, Illinois
| | - Carol Tan
- Section of Hematology/Oncology, Department of Medicine, University of Chicago Medical Center and Biological Sciences, Chicago, Illinois
| | - Yan Wang
- Section of Hematology/Oncology, Department of Medicine, University of Chicago Medical Center and Biological Sciences, Chicago, Illinois
| | - Daniel V T Catenacci
- Section of Hematology/Oncology, Department of Medicine, University of Chicago Medical Center and Biological Sciences, Chicago, Illinois.
| |
Collapse
|
241
|
Identification of Core Prognosis-Related Candidate Genes in Chinese Gastric Cancer Population Based on Integrated Bioinformatics. BIOMED RESEARCH INTERNATIONAL 2020; 2020:8859826. [PMID: 33381592 PMCID: PMC7748906 DOI: 10.1155/2020/8859826] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 11/18/2020] [Accepted: 11/24/2020] [Indexed: 12/29/2022]
Abstract
Background Gastric cancer (GC) is one of the leading causes of cancer-related mortality worldwide. There are great geographical differences in the incidence of GC, and somatic mutation rates of driver genes are also different. The present study is aimed at screening core prognosis-related candidate genes in Chinese gastric cancer population based on integrated bioinformatics for the early diagnosis and prognosis of GC. Methods In the present study, the differentially expressed genes (DEGs) in GC were identified using four microarray datasets from the Gene Expression Omnibus (GEO) database. The samples of these datasets were all from China. Functional enrichment analysis of DEGs was conducted to evaluate the underlying molecular mechanisms involved in GC. Protein-protein interaction (PPI) network and cytoHubba were performed to determine hub genes associated with GC. Gene Expression Profiling Interactive Analysis (GEPIA) and Human Protein Atlas (HPA) were performed to validate the hub genes. Results A total of 240 DEGs were obtained through the RRA method, including 80 upregulated genes and 160 downregulated genes. Upregulated genes were mainly enriched in extracellular matrix organization, extracellular matrix, and extracellular matrix structural constituent. The downregulated genes were mainly enriched in digestion, extracellular space, and oxidoreductase activity. The KEGG pathway enrichment analysis showed that the upregulated genes were mainly associated with ECM-receptor interaction, focal adhesion, and PI3K-Akt signaling pathway. And downregulated genes were mainly associated with the metabolism of xenobiotics by cytochrome P450, metabolic pathways, and gastric acid secretion. The transcriptional and translational expression levels of the genes including COL1A1, COL5A2, COL12A1, and VCAN were higher in GC tissues than normal tissues. Conclusion A total of four genes including COL1A1, COL5A2, COL12A1, and VCAN were considered potential GC biomarkers in the Chinese population. And ECM-receptor interaction, focal adhesion, and PI3K-Akt signaling pathway were revealed to be important mechanisms of GC. Our findings provide novel insights into the occurrence and progression of GC in the Chinese population.
Collapse
|
242
|
Salapa J, Bushman A, Lowe K, Irudayaraj J. Nano drug delivery systems in upper gastrointestinal cancer therapy. NANO CONVERGENCE 2020; 7:38. [PMID: 33301056 PMCID: PMC7728832 DOI: 10.1186/s40580-020-00247-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 11/23/2020] [Indexed: 05/02/2023]
Abstract
Upper gastrointestinal (GI) carcinomas are characterized as one of the deadliest cancer types with the highest recurrence rates. Their treatment is challenging due to late diagnosis, early metastasis formation, resistance to systemic therapy and complicated surgeries performed in poorly accessible locations. Current cancer medication face deficiencies such as high toxicity and systemic side-effects due to the non-specific distribution of the drug agent. Nanomedicine has the potential to offer sophisticated therapeutic possibilities through adjusted delivery systems. This review aims to provide an overview of novel approaches and perspectives on nanoparticle (NP) drug delivery systems for gastrointestinal carcinomas. Present regimen for the treatment of upper GI carcinomas are described prior to detailing various NP drug delivery formulations and their current and potential role in GI cancer theranostics with a specific emphasis on targeted nanodelivery systems. To date, only a handful of NP systems have met the standard of care requirements for GI carcinoma patients. However, an increasing number of studies provide evidence supporting NP-based diagnostic and therapeutic tools. Future development and strategic use of NP-based drug formulations will be a hallmark in the treatment of various cancers. This article seeks to highlight the exciting potential of novel NPs for targeted cancer therapy in GI carcinomas and thus provide motivation for further research in this field.
Collapse
Affiliation(s)
- Julia Salapa
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801 USA
- Department of Physics, Technical University of Vienna, Karlsplatz 13, 1040 Vienna, Austria
| | - Allison Bushman
- Department of Mechanical Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801 USA
| | - Kevin Lowe
- Carle Foundation Hospital South, Urbana, IL 61801 USA
- Carle-Illinois College of Medicine, Urbana, IL 61801 USA
| | - Joseph Irudayaraj
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801 USA
- Carle-Illinois College of Medicine, Urbana, IL 61801 USA
- Cancer Center at Illinois, Urbana, IL 61801 USA
- Biomedical Research Facility, 3rd Floor Mills Breast Cancer Institute, Carle Foundation Hospital South, Urbana, IL 61801 USA
| |
Collapse
|
243
|
Kou Y, Tong B, Wu W, Liao X, Zhao M. Berberine Improves Chemo-Sensitivity to Cisplatin by Enhancing Cell Apoptosis and Repressing PI3K/AKT/mTOR Signaling Pathway in Gastric Cancer. Front Pharmacol 2020; 11:616251. [PMID: 33362566 PMCID: PMC7756080 DOI: 10.3389/fphar.2020.616251] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Accepted: 11/11/2020] [Indexed: 12/16/2022] Open
Abstract
Gastric cancer is one of the most common malignancies ranks as the second leading cause of cancer-related mortality in the world. Cisplatin (DDP) is commonly used for gastric cancer treatment, whereas recurrence and metastasis are common because of intrinsic and acquired DDP-resistance. The aim of this study is to examine the effects of berberine on the DDP-resistance in gastric cancer and explore the underling mechanisms. In this study, we established the DDP-resistant gastric cancer cells, where the IC50 values of DDP in the BGC-823/DDP and SGC-7901/DDP were significantly higher than that in the corresponding parental cells. Berberine could concentration-dependently inhibited the cell viability of BGC-823 and SGC-7901 cells; while the inhibitory effects of berberine on the cell viability were largely attenuated in the DDP-resistant cells. Berberine pre-treatment significantly sensitized BGC-823/DDP and SGC-7901/DDP cells to DDP. Furthermore, berberine treatment concentration-dependently down-regulated the multidrug resistance-associated protein 1 and multi-drug resistance-1 protein levels in the BGC-823/DDP and SGC7901/DDP cells. Interestingly, the cell apoptosis of BGC-823/DDP and SGC-7901/DDP cells was significantly enhanced by co-treatment with berberine and DDP. The results from animals also showed that berberine treatment sensitized SGC-7901/DDP cells to DDP in vivo. Mechanistically, berberine significantly suppressed the PI3K/AKT/mTOR in the BGC-823/DDP and SGC-7901/DDP cells treated with DDP. In conclusion, we observed that berberine sensitizes gastric cancer cells to DDP. Further mechanistic findings suggested that berberine-mediated DDP-sensitivity may be associated with reduced expression of drug transporters (multi-drug resistance-1 and multidrug resistance-associated protein 1), enhanced apoptosis and repressed PI3K/AKT/mTOR signaling.
Collapse
Affiliation(s)
- Yingying Kou
- GCP Office, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Bending Tong
- Department of Pharmacy, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Weiqing Wu
- Department of Health Management, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical College of Jinan University, Shenzhen, China
| | - Xiangqing Liao
- Department of Health Management, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical College of Jinan University, Shenzhen, China
| | - Min Zhao
- Department of Pharmacy, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
244
|
Sun T, Li K, Zhu K, Yan R, Dang C, Yuan D. SNHG6 Interacted with miR-325-3p to Regulate Cisplatin Resistance of Gastric Cancer by Targeting GITR. Onco Targets Ther 2020; 13:12181-12193. [PMID: 33268996 PMCID: PMC7701159 DOI: 10.2147/ott.s262896] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 11/11/2020] [Indexed: 01/23/2023] Open
Abstract
Background Cisplatin resistance results in the failure of platinum-based chemotherapy and relapse of gastric cancer. We aimed to investigate the potential regulating role of SNHG6/miR-325-3p/GITR in reversing cisplatin resistance. Patients and Methods A total of 137 gastric cancer patients were recruited. qRT-PCR and ELISA were used to test the expression of target genes. CCK-8 and caspase 3/7 kit were used to test the cell viability and apoptosis rate. Dual luciferase reporter gene and RNA-pull down assay were used to investigate the potential interaction between target genes. Results SNHG6 and GITR were up regulated in gastric cancer; however, miR-325-3p was down-regulated. Besides, SNHG6, miR-325-3p and GITR expression were associated with gastric cancer prognosis. Then, we found that GITR and SNHG6 promoted proliferation and inhibited apoptosis of MKN45 and MKN45 cisplatin resistance cell line; however, miR-325-3p inhibited proliferation and promoted apoptosis of these cell lines. Furthermore, SNHG6 might bind to miR-325-3p to regulate its expression, and miR-325-3p directly interacted with the 3`UTR of GITR. Conclusion SNHG6 binds to miR-325-3p, which directly interacted with GITR to regulate cisplatin resistance of gastric cancer.
Collapse
Affiliation(s)
- Tuanhe Sun
- Department of Surgical Oncology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Kang Li
- Department of Surgical Oncology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Kun Zhu
- Department of Surgical Oncology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Rong Yan
- Department of Surgical Oncology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Chengxue Dang
- Department of Surgical Oncology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Dawei Yuan
- Department of Surgical Oncology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, People's Republic of China
| |
Collapse
|
245
|
Xu J, Wang Z, Huang Y, Wang Y, Xiang L, He X. A spirostanol saponin isolated from Tupistra chinensis Baker simultaneously induces apoptosis and autophagy by regulating the JNK pathway in human gastric cancer cells. Steroids 2020; 164:108737. [PMID: 33002483 DOI: 10.1016/j.steroids.2020.108737] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 09/15/2020] [Accepted: 09/21/2020] [Indexed: 12/23/2022]
Abstract
T-17, a bioactive spirostanol saponin extracted from Tupistra chinensis Baker, was previously reported with anti-inflammatory and cytotoxic activities. However, the mechanism underlying of its anti-proliferation activity remains to be elucidated. In this study, we investigated the anti-gastric cancer cell growth activity of T-17 in terms of cell viability, colony formation, cell cycle, induction of apoptosis/autophagy, and JNK pathway. T-17 showed dose-dependent cytotoxicity in SGC-7901 and AGS cell lines, it induced caspase-mediated apoptosis as well as G0/G1 phase arrest and modulation of cyclinE2 and p21 expression. In addition, T-17 promoted the cancer cell autophagy as evidenced with increased expression of Beclin-1 and decreased p62 in western blot and formation of GFP-LC3 puncta. Furthermore, T-17-induced autophagy decreased gastric cancer cell apoptosis as assessed by pharmacological autophagy inhibitors and ATG5 siRNA usage. Importantly, the activation of JNK pathway was simultaneously involved in T-17-induced apoptosis and autophagy. Taken together, the results suggest that T-17 is a promising cytotoxic agent for therapeutic treatment of human gastric adenocarcinoma, which provides a good foundation for further research and development of Tupistra chinensis Baker.
Collapse
Affiliation(s)
- Jingwen Xu
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Engineering Research Center for Lead Compounds & Drug Discovery, Guangzhou 510006, China
| | - Zhe Wang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Yuying Huang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Yihai Wang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Engineering Research Center for Lead Compounds & Drug Discovery, Guangzhou 510006, China
| | - Limin Xiang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Xiangjiu He
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Engineering Research Center for Lead Compounds & Drug Discovery, Guangzhou 510006, China.
| |
Collapse
|
246
|
Li Y, Liu H, Cui Y, Chen H, Cui X, Shao J, Su F, He X. miR-424-3p Contributes to the Malignant Progression and Chemoresistance of Gastric Cancer. Onco Targets Ther 2020; 13:12201-12211. [PMID: 33273826 PMCID: PMC7705957 DOI: 10.2147/ott.s280717] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 11/09/2020] [Indexed: 01/10/2023] Open
Abstract
Background Gastric cancer (GC) is one of the most common and lethal malignancies worldwide. Therefore, a better understanding of the mechanism of its malignant progression and chemoresistance will be helpful for the treatment of patients with GC. Methods The gene expression profiles downloaded from GEO database and the TargetScan Human were used to identify the key regulation model based on miRNA by bioinformatics analyses. The regulation of miRNA to target was clarified by luciferase assay, qPCR, and Western blotting. Then, the in vitro and in vivo experiments were further conducted by overexpression or knockdown of miRNA and/or target to examine the regulation effects and clarify the mechanism. Results In the present study, miR-424-3p was identified to be differentially expressed among normal gastric, GC, and chemoresistant GC tissues. Target analysis results indicated that ABCC2, a chemoresistance-related gene, was a regulated target of miR-424-3p. The in vitro and in vivo experiment results further demonstrated that miR-424-3p relied on ABCC2-induced chemoresistance to promote GC proliferation and metastasis. Conclusion Overall, this study revealed that miR-424-3p contributed to the malignant progression and chemoresistance of GC. Thus, miR-424-3p could be a potential target for the treatment of GC.
Collapse
Affiliation(s)
- Yongyuan Li
- Department of General Surgery, The Fifth Central Hospital, Tianjin 300450, People's Republic of China.,Department of General Surgery, Tianjin Medical University General Hospital, Tianjin 300052, People's Republic of China
| | - Hongjie Liu
- Department of Radiology, The Fifth Central Hospital, Tianjin 300450, People's Republic of China
| | - Yu Cui
- Department of General Surgery, The Fifth Central Hospital, Tianjin 300450, People's Republic of China
| | - Hekai Chen
- Department of General Surgery, The Fifth Central Hospital, Tianjin 300450, People's Republic of China
| | - Xuejun Cui
- Department of General Surgery, The Fifth Central Hospital, Tianjin 300450, People's Republic of China
| | - Jianping Shao
- Department of General Surgery, The Fifth Central Hospital, Tianjin 300450, People's Republic of China
| | - Feng Su
- Department of General Surgery, The Fifth Central Hospital, Tianjin 300450, People's Republic of China
| | - Xianghui He
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin 300052, People's Republic of China
| |
Collapse
|
247
|
Wu CE, Xue WW, Zhuang YW, Ding DW, Zhou JY, Liu SL, Wang RP, Shu P. A clinical study on the efficacy of Yiqi Huayu Jiedu decoction for reducing the risk of postoperative recurrence and metastasis of gastric cancer: Protocol for a multicenter, randomized, double-blind, placebo-controlled trial. Medicine (Baltimore) 2020; 99:e23417. [PMID: 33235121 PMCID: PMC7710168 DOI: 10.1097/md.0000000000023417] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [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 Gastric cancer (GC) is one of the top 10 malignant tumors worldwide and poses a great threat to human life and health, the prevention and treatment of which has become the focus and difficulty of medical research. With its unique advantages, traditional Chinese medicine (TCM) is widely used in the prevention and treatment of postoperative recurrence and metastasis of GC as well as the improvement of patients' quality of life. The aim of this study is to elucidate the curative effect and the underlying mechanism of Yiqi Huayu Jiedu (YQHYJD) decoction. METHODS/DESIGN This is a prospective, multicenter, randomized controlled trial continuing 3 years. Two hundred ninety-eight eligible patients will be randomly divided into 2 groups, the chemotherapy combined with placebo and the chemotherapy combined with YQHYJD group at a ratio of 1:1. All patients will receive the treatment for 6 months and follow up for 3 years. The primary outcomes are disease-free survival, and 1-year, 2-year, 3-year progression-free survival rate, while the secondary outcomes are tumor makers, TCM syndrome score, quality of life score, overall chemotherapy completion rate, intestinal flora diversity test, immune function (T, B lymphocyte subsets and NK cells) test. The Security index includes blood, urine and stool routine, electrocardiogram, liver function (ALT), and renal function (BUN, Scr). All of these outcomes will be analyzed at the end of the trial. DISCUSSION This research will provide the valuable evidence for the efficacy and safety of Yiqi Huayu Jiedu decoction in postoperative GC. Furthermore, it will be helpful to form a higher level of evidence-based medical basis for TCM in the treatment of GC recurrence and metastasis. TRIAL REGISTRATION ChiCTR2000039038.
Collapse
Affiliation(s)
- Cun-En Wu
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine
| | - Wei-Wei Xue
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine
| | - Yu-Wen Zhuang
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine
- Traditional Chinese Medicine Department, Jinling Hospital, School of Medicine, Nanjing University
| | - Da-Wei Ding
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine
| | - Jin-Yong Zhou
- Central Laboratory, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, China
| | - Shen-Lin Liu
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine
| | - Rui-Ping Wang
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine
| | - Peng Shu
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine
| |
Collapse
|
248
|
Catenacci DVT, Moya S, Lomnicki S, Chase LM, Peterson BF, Reizine N, Alpert L, Setia N, Xiao SY, Hart J, Siddiqui UD, Hogarth DK, Eng OS, Turaga K, Roggin K, Posner MC, Chang P, Narula S, Rampurwala M, Ji Y, Karrison T, Liao CY, Polite BN, Kindler HL. Personalized Antibodies for Gastroesophageal Adenocarcinoma (PANGEA): A Phase II Study Evaluating an Individualized Treatment Strategy for Metastatic Disease. Cancer Discov 2020; 11:308-325. [PMID: 33234578 DOI: 10.1158/2159-8290.cd-20-1408] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 11/01/2020] [Accepted: 11/18/2020] [Indexed: 02/07/2023]
Abstract
The one-year and median overall survival (mOS) rates of advanced gastroesophageal adenocarcinomas (GEA) are ∼50% and <12 months, respectively. Baseline spatial and temporal molecular heterogeneity of targetable alterations may be a cause of failure of targeted/immunooncologic therapies. This heterogeneity, coupled with infrequent incidence of some biomarkers, has resulted in stalled therapeutic progress. We hypothesized that a personalized treatment strategy, applied at first diagnosis then serially over up to three treatment lines using monoclonal antibodies combined with optimally sequenced chemotherapy, could contend with these hurdles. This was tested using a novel clinical expansion-platform type II design with a survival primary endpoint. Of 68 patients by intention-to-treat, the one-year survival rate was 66% and mOS was 15.7 months, meeting the primary efficacy endpoint (one-sided P = 0.0024). First-line response rate (74%), disease control rate (99%), and median progression-free survival (8.2 months) were superior to historical controls. The PANGEA strategy led to improved outcomes warranting a larger randomized study. SIGNIFICANCE: This study highlights excellent outcomes achieved by individually optimizing chemotherapy, biomarker profiling, and matching of targeted therapies at baseline and over time for GEA. Testing a predefined treatment strategy resulted in improved outcomes versus historical controls. Therapeutic resistance observed in correlative analyses suggests that dual targeted inhibition may be beneficial.This article is highlighted in the In This Issue feature, p. 211.
Collapse
Affiliation(s)
- Daniel V T Catenacci
- The University of Chicago, Section of Hematology/Oncology, Department of Medicine, Chicago, Illinois.
| | - Stephanie Moya
- The University of Chicago, Section of Hematology/Oncology, Department of Medicine, Chicago, Illinois
| | - Samantha Lomnicki
- The University of Chicago, Section of Hematology/Oncology, Department of Medicine, Chicago, Illinois
| | - Leah M Chase
- The University of Chicago, Section of Hematology/Oncology, Department of Medicine, Chicago, Illinois
| | - Bryan F Peterson
- The University of Chicago, Section of Hematology/Oncology, Department of Medicine, Chicago, Illinois
| | - Natalie Reizine
- The University of Chicago, Section of Hematology/Oncology, Department of Medicine, Chicago, Illinois
| | - Lindsay Alpert
- The University of Chicago, Department of Pathology, Chicago, Illinois
| | - Namrata Setia
- The University of Chicago, Department of Pathology, Chicago, Illinois
| | - Shu-Yuan Xiao
- The University of Chicago, Department of Pathology, Chicago, Illinois
| | - John Hart
- The University of Chicago, Department of Pathology, Chicago, Illinois
| | - Uzma D Siddiqui
- The University of Chicago, Department of Medicine, Center for Endoscopic Research and Therapeutics (CERT), Chicago, Illinois
| | - D Kyle Hogarth
- The University of Chicago, Department of Medicine, Section of Pulmonology, Chicago, Illinois
| | - Oliver S Eng
- The University of Chicago, Department of Surgery, Chicago, Illinois
| | - Kiran Turaga
- The University of Chicago, Department of Surgery, Chicago, Illinois
| | - Kevin Roggin
- The University of Chicago, Department of Surgery, Chicago, Illinois
| | | | - Paul Chang
- The University of Chicago, Department of Radiology, Chicago, Illinois
| | | | | | - Yuan Ji
- The University of Chicago, Department of Public Health Sciences, Chicago, Illinois
| | - Theodore Karrison
- The University of Chicago, Department of Public Health Sciences, Chicago, Illinois
| | - Chih-Yi Liao
- The University of Chicago, Section of Hematology/Oncology, Department of Medicine, Chicago, Illinois
| | - Blase N Polite
- The University of Chicago, Section of Hematology/Oncology, Department of Medicine, Chicago, Illinois
| | - Hedy L Kindler
- The University of Chicago, Section of Hematology/Oncology, Department of Medicine, Chicago, Illinois
| |
Collapse
|
249
|
El Rami FE, Barsoumian HB, Khneizer GW. Hereditary diffuse gastric cancer therapeutic roadmap: current and novel approaches in a nutshell. Ther Adv Med Oncol 2020; 12:1758835920967238. [PMID: 33193828 PMCID: PMC7607792 DOI: 10.1177/1758835920967238] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 09/23/2020] [Indexed: 12/24/2022] Open
Abstract
Hereditary diffuse gastric cancer (HDGC) is a rare malignancy characterized by autosomal dominant inheritance of pathological variants of the CDH1 gene encoding E-cadherin, which is involved in cell–cell adhesion, maintenance of epithelial architecture, tumor suppression, and regulation of intracellular signaling pathways. Late-stage recognition of HDGC is typically associated with a poor clinical outcome due to its metastatic potential and risk of lobular breast cancer (LBC) development. The American College of Gastroenterology issued guidelines to evaluate HDGC, test for CDH1 genetic variants, and recommend prophylactic gastrectomy for carriers of CDH1 mutations. If surgery is not pursued, endoscopy is a surveillance alternative, although it carries a limited ability to detect malignant foci. As part of clinical research efforts, novel endoscopy advances are currently studied, and a center of excellence for HDGC was created for a comprehensive multidisciplinary team approach. Within this review, we cover current conventional treatment modalities such as gastrectomy and chemotherapy, as the mainstay treatments, in addition to Pembrolizumab, an immune checkpoint inhibitor, as the last therapeutic resort. We also shed light on novel and promising approaches with emphasis on immunotherapy to treat HDGC. We further break down the therapeutic paradigms to utilize molecular tools, antibodies against checkpoint inhibitors, TGF-β and tyrosine kinase inhibitors, cell-based adoptive therapies, and oncolytic viral therapies. We aim to expand the understanding on how to modulate the tumor microenvironment to tip the balance towards an anti-tumor phenotype, prevent metastasis of the primary disease, and potentially alter the therapeutic landscape for HDGC.
Collapse
Affiliation(s)
- Fadi E El Rami
- Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Gebran W Khneizer
- Department of Internal Medicine, Indiana University Hospital, 550 N University Blvd, Suite 1501, Indianapolis, IN 46202, USA
| |
Collapse
|
250
|
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: 149] [Impact Index Per Article: 37.3] [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
|