1
|
Chi X, Song Y, Wei S, Wang G, Wen Y, Zhang Z, Zhang C, Zhang H, Hu Z, Yu H, Huo B, Shan B, Zhao L. Lian Qiao Wen Dan Decoction attenuates gastric carcinogenesis by alleviating oxidative phosphorylation dysfunction. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 144:156924. [PMID: 40499219 DOI: 10.1016/j.phymed.2025.156924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Revised: 04/30/2025] [Accepted: 05/27/2025] [Indexed: 06/22/2025]
Abstract
BACKGROUND Wen Dan Decoction (WDD) is primarily utilized as an "expectorant" in clinical practice. The combination of WDD and Lian Qiao (LQWDD) exhibits synergistic effects on phlegm resolution, qi regulation, and detoxification. In preliminary clinical practice, we observed that LQWDD effectively alleviates the severity of gastric precancerous lesions. Nevertheless, the underlying mechanism remains elusive. PURPOSE This study was designed to explore the efficacy and potential mechanisms of LQWDD against gastric carcinogenesis. METHODS Retrospective analyses of patients with gastric precancerous lesions and an N-methyl-N9-nitro-N-nitrosoguanidine (MNNG)-induced rat gastric cancer model were conducted to evaluate the efficacy of LQWDD against gastric carcinogenesis. Proteomic analyses were employed to investigate the underlying mechanisms of LQWDD. Moreover, energy metabolites were quantified using metabolomics. Network pharmacology and high-performance liquid chromatography-mass spectrometry were integrated to elucidate the potential active components of LQWDD. Additionally, seahorse analysis and RNA sequencing were used to explore the effects and underlying mechanisms of compound quercetin on oxidative phosphorylation. RESULTS LQWDD effectively impeded gastric carcinogenesis in an MNNG-induced rat gastric cancer model and patients. Proteomic analyses revealed that LQWDD protected the integrity of the oxidative phosphorylation pathway during gastric carcinogenesis, which was significantly suppressed in human gastric intraepithelial neoplasia and MNNG-induced gastric carcinoma. Moreover, LQWDD administration significantly reversed MNNG-induced reductions in the expression of mitochondrial complex I-related proteins (ALDH3A1 and NDUFS4), as well as in ATP, NAD+, and metabolite levels, indicating its protective role against oxidative phosphorylation dysfunction through the modulation of mitochondrial complex I-associated proteins. Furthermore, quercetin and kaempferol were identified as potential active components of LQWDD. These components suppressed the growth of gastric cancer cells and patient-derived organoids while simultaneously increasing intracellular ATP and NAD+ levels. Notably, quercetin exerted its preferential regulatory role in mitochondrial oxidative respiration by activating the AMPK signaling pathway. CONCLUSIONS Our study provides the first systematic evidence that mitochondrial OXPHOS dysfunction plays a pivotal role in gastric carcinogenesis. LQWDD and its potential active ingredient quercetin inhibit gastric carcinogenesis by mitigating oxidative phosphorylation impairment, highlighting on a promising therapeutic prescription for treating with gastric precancerous lesions.
Collapse
Affiliation(s)
- Xiuping Chi
- Research Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, China; Key Laboratory of Tumor Prevention and Precision Diagnosis & Treatment of Hebei Province, Clinical Oncology Research Center, Shijiazhuang, Hebei 050011, China
| | - Yanru Song
- Research Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, China; Key Laboratory of Tumor Prevention and Precision Diagnosis & Treatment of Hebei Province, Clinical Oncology Research Center, Shijiazhuang, Hebei 050011, China
| | - Sisi Wei
- Research Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, China; Key Laboratory of Tumor Prevention and Precision Diagnosis & Treatment of Hebei Province, Clinical Oncology Research Center, Shijiazhuang, Hebei 050011, China
| | - Guangjun Wang
- Key Laboratory of Tumor Prevention and Precision Diagnosis & Treatment of Hebei Province, Clinical Oncology Research Center, Shijiazhuang, Hebei 050011, China
| | - Yang Wen
- Research Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, China; Key Laboratory of Tumor Prevention and Precision Diagnosis & Treatment of Hebei Province, Clinical Oncology Research Center, Shijiazhuang, Hebei 050011, China
| | - Zhe Zhang
- Research Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, China; Key Laboratory of Tumor Prevention and Precision Diagnosis & Treatment of Hebei Province, Clinical Oncology Research Center, Shijiazhuang, Hebei 050011, China
| | - Cong Zhang
- Research Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, China; Key Laboratory of Tumor Prevention and Precision Diagnosis & Treatment of Hebei Province, Clinical Oncology Research Center, Shijiazhuang, Hebei 050011, China
| | - Hongtao Zhang
- Key Laboratory of Tumor Prevention and Precision Diagnosis & Treatment of Hebei Province, Clinical Oncology Research Center, Shijiazhuang, Hebei 050011, China
| | - Zhongdong Hu
- Modern Research Center for Traditional Chinese Medicine, Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Haiyang Yu
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, And State Key Laboratory of Component-Based Chinese Medicine, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Bingjie Huo
- Oncology Department of Interated Chinese and Western Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, China.
| | - Baoen Shan
- Research Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, China; Key Laboratory of Tumor Prevention and Precision Diagnosis & Treatment of Hebei Province, Clinical Oncology Research Center, Shijiazhuang, Hebei 050011, China.
| | - Lianmei Zhao
- Research Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, China; Key Laboratory of Tumor Prevention and Precision Diagnosis & Treatment of Hebei Province, Clinical Oncology Research Center, Shijiazhuang, Hebei 050011, China.
| |
Collapse
|
2
|
Wang R, Cui JF, Lv J, Song JL, Lu YY, Huang XJ, Lin ZK, Zhang SY, Wang SS, Qiu WS. Perillaldehyde synergizes with ferroptosis inducers to promote ferroptotic cell death in gastric cancer. Front Cell Dev Biol 2025; 13:1598520. [PMID: 40530331 PMCID: PMC12170665 DOI: 10.3389/fcell.2025.1598520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2025] [Accepted: 05/22/2025] [Indexed: 06/20/2025] Open
Abstract
Introduction As a traditional medicine and food homologous plant, Perilla frutescens is widely cultivated in China, Japan, and Korea. According to the Compendium of Materia Medica, the leaves, stems, and seeds of perilla can all be used as medicine. Perilla essential oil has been used in traditional Chinese medicine since ancient times. It has been demonstrated that perillaldehyde (PAH), a primary composition of the essential oil extracted from perilla, can inhibit tumor growth through multiple mechanisms. However, the specific mechanisms by which PAH suppresses gastric cancer remain incompletely understood. Methods We performed in vitro experiments using three cell lines (AGS, HGC27, and MFC) to assess the effects of PAH on cell viability, proliferation, and migration of gastric cancer cells. Concurrently, we established a subcutaneous gastric tumor model in BALB/c nude mice for in vivo animal studies. Subsequently, oxidative stress was measured via fluorescence staining techniques (H2DCFDA, DHE, and JC-1). We then evaluated whether PAH induced ferroptosis in gastric cancer cells through FerroOrange staining, quantification of intracellular glutathione (GSH) and lipid peroxidation levels, and Western blotting. Finally, PAH was co-administered with the ferroptosis inhibitor Ferrostatin-1 (Fer-1) or the ferroptosis inducer RSL3, and relevant experiments were re-evaluated. Results In this study, PAH was proven to inhibit the growth of gastric cancer both in vivo and in vitro. It led to a reduction in mitochondrial membrane potential (MMP), an augmentation of the accumulation of reactive oxygen species (ROS), and an elevation of oxidative stress levels. Moreover, PAH decreased intracellular GSH levels while increasing intracellular lipid peroxidation and Fe2+ levels. These effects indicate that PAH induces ferroptosis via inhibiting the system Xc (-)/GSH/GPX4 axis. Furthermore, PAH influenced the expression of proteins related to iron transport and storage and regulated ferroptosis via the P62-Keap1-Nrf2 pathway. When combined with the ferroptosis inducer RSL3, PAH could promote ferroptosis in gastric cancer. Discussion Our research suggests a potential therapeutic strategy in which PAH could be used to synergize with ferroptosis inducers for treating gastric cancer.
Collapse
Affiliation(s)
- Rui Wang
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jin-Feng Cui
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jing Lv
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jia-Lin Song
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yang-Yang Lu
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiao-Juan Huang
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Zhong-Kun Lin
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
- Department of Oncology, Shandong Provincial Third Hospital, Jinan, Shandong, China
| | - Si-Yi Zhang
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Sha-Sha Wang
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Wen-Sheng Qiu
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
3
|
Lu Z, Lyu Z, Dong P, Liu Y, Huang L. N6-methyladenosine RNA modification in stomach carcinoma: Novel insights into mechanisms and implications for diagnosis and treatment. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167793. [PMID: 40088577 DOI: 10.1016/j.bbadis.2025.167793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 02/16/2025] [Accepted: 03/03/2025] [Indexed: 03/17/2025]
Abstract
N6-methyladenosine (m6A) RNA methylation is crucially involved in the genesis and advancement of gastric cancer (GC) by controlling various pathobiological aspects including gene expression, signal transduction, metabolism, cell death, epithelial-mesenchymal transition, angiogenesis, and exosome function. Despite its importance, the exact mechanisms by which m6A modification influences GC biology remain inadequately explored. This review consolidates the latest advances in uncovering the mechanisms and diverse roles of m6A in GC and proposes new research and translational directions. Key regulators (writers, readers, and erasers) of m6A, such as METTL3/14/16 and WTAP, significantly affect cancer progression, anticancer immune response, and treatment outcomes. m6A modification also impacts immune cell infiltration and the tumor microenvironment, highlighting its potential as a diagnostic and prognostic marker. Interactions between m6A methylation and non-coding RNAs offer further novel insights into GC development and therapeutic targets. Targeting m6A regulators could enhance immunotherapy response, overcome treatment resistance, and improve oncological and clinical outcomes. Models based on m6A can precisely predict treatment response and prognosis in GC. Additional investigation is needed to fully understand the mechanisms of m6A methylation and its potential clinical applications and relevance (e.g., as precise markers for early detection, prediction of outcome, and response to therapy and as therapeutic targets) in GC. Future research should focus on in vivo studies, potential clinical trials, and the examination of m6A modification in other types of cancers.
Collapse
Affiliation(s)
- Zhengmao Lu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China
| | - Zhaojie Lyu
- Department of Urology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Peixin Dong
- Department of Obstetrics and Gynecology, Hokkaido University School of Medicine, Hokkaido University, Sapporo, Japan.
| | - Yunmei Liu
- School of Cultural Heritage and Information Management, Shanghai University, Shanghai, China.
| | - Lei Huang
- Department of Gastroenterology, National Clinical Research Center for Digestive Diseases, Shanghai Institute of Pancreatic Diseases, The First Affiliated Hospital of Naval Medical University/Changhai Hospital, Naval Medical University, Shanghai 200433, China; National Key Laboratory of Immunity and Inflammation, Changhai Clinical Research Unit, The First Affiliated Hospital of Naval Medical University/Changhai Hospital, Naval Medical University, Shanghai 200433, China.
| |
Collapse
|
4
|
Jiang L, Lau HCH, Zeng R, Yu J. Diet, Gastric Microbiota, and Metabolites in Gastric Tumorigenesis. RESEARCH (WASHINGTON, D.C.) 2025; 8:0693. [PMID: 40357361 PMCID: PMC12067930 DOI: 10.34133/research.0693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Revised: 04/09/2025] [Accepted: 04/15/2025] [Indexed: 05/15/2025]
Abstract
Gastric cancer (GC) is one of the most common cancers worldwide particularly in Asian populations, and certain diets have been associated with increased risk of GC. Recent advances in microbial profiling technology have facilitated investigations on microbes residing on the gastric mucosa and increasing evidence has revealed the critical roles of non-Helicobacter pylori gastric microbes in gastric tumorigenesis. On the other hand, diets can affect microbial communities, causing compositional and functional shift of the microbiota. In this review, we summarize the influence of various diets including processed meat, salt-preserved food, high-fat diet, and alcohol on the development and progression of GC. We also explore microbial metabolites and host-microbe interactions in gastric tumorigenesis, alongside dietary interventions targeting the microbiota for the prevention and management against GC.
Collapse
Affiliation(s)
- Lanping Jiang
- Institute of Digestive Disease, Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Harry Cheuk-Hay Lau
- Institute of Digestive Disease, Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Ruijie Zeng
- Institute of Digestive Disease, Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Jun Yu
- Institute of Digestive Disease, Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
5
|
Wang L, Jiang W, Li H. Global, regional, and national burden of gastritis and duodenitis from 1990 to 2021 with projections to 2050: a systematic analysis of the Global Burden of Disease Study 2021. Int J Med Sci 2025; 22:2570-2582. [PMID: 40520897 PMCID: PMC12163426 DOI: 10.7150/ijms.109762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Accepted: 04/25/2025] [Indexed: 06/18/2025] Open
Abstract
Background: Gastritis and duodenitis are highly prevalent upper gastrointestinal inflammatory conditions. We estimate the most up-to-date global, regional, and national burden in incidence, prevalence, mortality, and disability-adjusted life-years (DALYs) for gastritis and duodenitis from 1990 to 2021 with projections to 2050. Methods: Population-based data in this study was retrieved from the Global Burden of Diseases, Injuries, and Risk Factors Study 2021 (GBD 2021). We evaluated the temporal trends of age-standardized rates of gastritis and duodenitis prevalence (ASPR), incidence (ASIR), mortality (ASMR), and DALYs (ASDR) across 204 countries and territories, as well as estimated annual percentage changes (EAPC) from 1990 to 2021. Analyses were stratified by sex, age subgroup, socio-demographic index (SDI) at the global, regional, and national level. A Bayesian age-period-cohort model was employed to project ASPR and ASDR of gastritis and duodenitis by sex and age up to 2050. Results: In 2021, 27.20 million (95% UI: 21.85-33.65) individuals globally had gastritis and duodenitis, with an ASIR of 323.24 (95% UI: 261.35-398.64) per 100,000. Globally, the prevalent cases of gastritis and duodenitis in 2021 was higher in females than in males and increased with age, peaking at the fifth decade of life. The number of prevalent cases of gastritis and duodenitis is projected to reach approximately 51.24 million by 2050. In 2021, gastritis and duodenitis caused 2.81 (95% UI: 2.17-3.61) million DALYs worldwide, with an ASDR of 35.64 (95% UI: 27.56-45.77) per 100,000, while the ASMR was 0.54 (95% UI: 0.47-0.62) per 100,000. Despite global decreases in ASPR and ASIR, increasing trends were observed in the low and low-middle SDI regions from 1990 to 2021. The low SDI regions exhibited the highest ASDR of 63.44 (95% UI: 44.08-87.16) per 100,000. Among 21 GBD regions, East Asia exhibited the highest ASPR and ASIR in 2021. The ASPR of gastritis and duodenitis in 2050 is forecast to be 402.14 per 100,000, with an ASDR of approximately 22.09 per 100,000. Conclusion: Despite global reductions in ASPR, ASIR, ASMR, and ASDR between 1990 and 2021, the global burden of gastritis and duodenitis exhibits regional disparities, closely linked to SDI levels. The highest burden of gastritis and duodenitis was observed in East Asia and Sub-Saharan Africa. More potent measures are urgently needed in low SDI regions to forestall the increase of Gastritis and duodenitis burden.
Collapse
Affiliation(s)
- Lufei Wang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Wei Jiang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Hui Li
- Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai 200438, China
- Human Phenome Institute, Zhangjiang Fudan International Innovation Center, Fudan University, Shanghai 200438, China
| |
Collapse
|
6
|
Cho SY, Hwang H, Lee HS, Kwon Y, Khanh Vu N, Baek JG, Jeon M, Bae J, Kwon HC, Kim WK, Kwon J. Chemical constituents from the Korean endemic plant Pseudolysimachion pusanensis inhibit diffuse-type gastric cancer cells. Biomed Pharmacother 2025; 186:118005. [PMID: 40138921 DOI: 10.1016/j.biopha.2025.118005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 03/06/2025] [Accepted: 03/18/2025] [Indexed: 03/29/2025] Open
Abstract
Diffuse-type gastric cancer (GC) is closely associated with genetic abnormalities; however, its exact pathological mechanisms are still not understood. It manifests without symptoms before advanced stages and is often difficult to diagnose using routine imaging tests. Therefore, specific targeted therapies for diffuse GC are currently unavailable. In this study, the extract of Pseudolysimachion pusanensis, which is endemic to Korea, inhibited the proliferation of GC cells, MKN1 and SNU668, while the other extracts of the two endemic Pseudolysimachion species did not show any activity. This led to the molecular networking analysis of Pseudolysimachion species to identify the molecules mostly observed only in P. pusanensis. Thirteen new (1-13) and eight known (14-21) compounds were obtained and structurally characterized. Of these, cucurbitacin derivative compounds 1 and 14-16 showed activity, and particularly, the IC50 value of compound 1 was 0.65 and 0.21 μM. Ki-67 expression analysis, single-cell originated cell proliferation assay, and western blot analysis of apoptotic cell death-related molecules revealed that compound 1 mediated both cytostasis and cellular death via apoptosis. Particularly, this compound exhibited an anti-tumorigenic effect on the invasion of diffuse-type GC cells by perturbing the epithelial-to-mesenchymal transition (EMT) pathway, as demonstrated by invasion assays using both 2D models and in vivo-like 3D spheroid co-culture models, as well as western blot analysis of EMT markers. In addition, some functional groups that may or may not be necessary for the activity of cucurbitacin derivatives were identified, and some clues that the presence of metal ions may affect the activity were provided.
Collapse
Affiliation(s)
- Su-Yeon Cho
- KIST Gangneung Institute of Natural Products, Korea Institute of Science and Technology, Gangneung 25451, Republic of Korea; Natural Product Applied Science, University of Science and Technology (UST), Daejeon 34113, Republic of Korea
| | - Hoseong Hwang
- KIST Gangneung Institute of Natural Products, Korea Institute of Science and Technology, Gangneung 25451, Republic of Korea
| | - Hyeon-Seong Lee
- KIST Gangneung Institute of Natural Products, Korea Institute of Science and Technology, Gangneung 25451, Republic of Korea
| | - Yujin Kwon
- KIST Gangneung Institute of Natural Products, Korea Institute of Science and Technology, Gangneung 25451, Republic of Korea
| | - Ngoc Khanh Vu
- KIST Gangneung Institute of Natural Products, Korea Institute of Science and Technology, Gangneung 25451, Republic of Korea
| | - Jong Gwon Baek
- KIST Gangneung Institute of Natural Products, Korea Institute of Science and Technology, Gangneung 25451, Republic of Korea
| | - Mukyeong Jeon
- KIST Gangneung Institute of Natural Products, Korea Institute of Science and Technology, Gangneung 25451, Republic of Korea
| | - Joonbeom Bae
- Department of Biotechnology, School of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea
| | - Hak Cheol Kwon
- KIST Gangneung Institute of Natural Products, Korea Institute of Science and Technology, Gangneung 25451, Republic of Korea
| | - Won Kyu Kim
- KIST Gangneung Institute of Natural Products, Korea Institute of Science and Technology, Gangneung 25451, Republic of Korea; Natural Product Applied Science, University of Science and Technology (UST), Daejeon 34113, Republic of Korea; Department of Convergence Medicine, Yonsei University Wonju College of Medicine, Wonju 26426, Republic of Korea.
| | - Jaeyoung Kwon
- KIST Gangneung Institute of Natural Products, Korea Institute of Science and Technology, Gangneung 25451, Republic of Korea; Natural Product Applied Science, University of Science and Technology (UST), Daejeon 34113, Republic of Korea; Department of Convergence Medicine, Yonsei University Wonju College of Medicine, Wonju 26426, Republic of Korea.
| |
Collapse
|
7
|
Sundar R, Nakayama I, Markar SR, Shitara K, van Laarhoven HWM, Janjigian YY, Smyth EC. Gastric cancer. Lancet 2025:S0140-6736(25)00052-2. [PMID: 40319897 DOI: 10.1016/s0140-6736(25)00052-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Revised: 11/13/2024] [Accepted: 01/09/2025] [Indexed: 05/07/2025]
Abstract
Gastric cancer remains a major health challenge worldwide, with nearly 1 million new cases annually contributing to more than 650 000 deaths. Epidemiologically, gastric cancer shows substantial geographical variation in incidence, with higher rates in Asia, South America, and eastern Europe, and a rapid increase in early-onset cases among people younger than 50 years. Key risk factors for gastric cancer include Helicobacter pylori infection, diet, obesity, smoking, and genetic predisposition. Early detection through comprehensive diagnostic procedures is crucial for optimising treatment outcomes. Standard treatment approaches for locally advanced gastric cancer include surgical resection, particularly D2 lymphadenectomy, complemented by chemotherapy and radiotherapy. There is increasing implementation of minimally invasive surgical techniques for operable disease and integration of immune checkpoint inhibitors and targeted therapies for advanced stages. Emerging therapies, such as novel targeted treatments and next-generation immunotherapies, show promise in improving survival and quality of life. Future directions in the management of gastric cancer focus on precision medicine, continued advancement in immunotherapy, novel early detection methods, and a multidisciplinary approach to care. These strategies aim to enhance the overall effectiveness of treatment and prognosis worldwide.
Collapse
Affiliation(s)
- Raghav Sundar
- Department of Medicine, Section of Medical Oncology, Yale School of Medicine, New Haven, CT, USA; Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Department of Haematology-Oncology, National University Cancer Institute, Singapore
| | - Izuma Nakayama
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Sheraz R Markar
- Surgical Intervention Trials Unit, Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Kohei Shitara
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Hanneke W M van Laarhoven
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, Netherlands; Department of Medical Oncology, Amsterdam UMC Location University of Amsterdam, Amsterdam, Netherlands
| | - Yelena Y Janjigian
- Department of Medicine, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY, USA
| | - Elizabeth C Smyth
- Oxford NIHR Biomedical Research Centre, Churchill Hospital, Oxford, UK.
| |
Collapse
|
8
|
Wizenty J, Sigal M. Helicobacter pylori, microbiota and gastric cancer - principles of microorganism-driven carcinogenesis. Nat Rev Gastroenterol Hepatol 2025; 22:296-313. [PMID: 40011753 DOI: 10.1038/s41575-025-01042-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/16/2025] [Indexed: 02/28/2025]
Abstract
The demonstration that Helicobacter pylori is a pathogenic bacterium with marked carcinogenic potential has paved the way for new preventive approaches for gastric cancer. Although decades of research have uncovered complex interactions of H. pylori with epithelial cells, current insights have refined our view on H. pylori-associated carcinogenesis. Specifically, the cell-type-specific effects on gastric stem and progenitor cells deep in gastric glands provide a new view on the ability of the bacteria to colonize long-term, manipulate host responses and promote gastric pathology. Furthermore, new, large-scale epidemiological data have shed light on factors that determine why only a subset of carriers progress to gastric cancer. Currently, technological advances have brought yet another revelation: H. pylori is far from the only microorganism able to colonize the stomach. Instead, the stomach is colonized by a diverse gastric microbiota, and there is emerging evidence for the occurrence and pathological effect of dysbiosis resulting from an aberrant interplay between H. pylori and the gastric mucosa. With the weight of this evidence mounting, here we consider how the lessons learned from H. pylori research inform and synergize with this emerging field to bring a more comprehensive understanding of the role of microbes in gastric carcinogenesis.
Collapse
Affiliation(s)
- Jonas Wizenty
- Department of Hepatology and Gastroenterology, Charité - Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Biomedical Innovation Academy and BIH Charité Clinician Scientist Program, Berlin, Germany
| | - Michael Sigal
- Department of Hepatology and Gastroenterology, Charité - Universitätsmedizin Berlin, Berlin, Germany.
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.
| |
Collapse
|
9
|
Liu P, Zhou S, Zhou Z, Jin Z, Chen W, Li Z, Xu J, Chen F, Li Y, Wen Y, Zhang S, Zhang C, Li B, Zhao J, Chen H. Discovery and antitumor evaluation of a mitochondria-targeting ruthenium complex for effective cancer therapy. Cancer Lett 2025; 616:217582. [PMID: 40021041 DOI: 10.1016/j.canlet.2025.217582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 02/19/2025] [Accepted: 02/20/2025] [Indexed: 03/03/2025]
Abstract
Ruthenium-based metallodrugs have garnered attention as a promising alternative for anticancer therapy, aiming to overcome chemoresistance and severe side effects linked to platinum-based drugs. However, ruthenium complexes tested in clinical trials to date have yielded unsatisfactory results. This study synthesized a positively charged ruthenium complex (Ru-2) that effectively penetrated cancer cells and exhibited superior cytotoxicity to cisplatin in vitro against cancer cell lines and organoids. Ru-2 selectively targeted mitochondria, disrupting their function by depolarizing mitochondrial membrane potential, elevating reactive oxygen species production, and impairing both oxidative phosphorylation and the tricarboxylic acid cycle. Furthermore, Ru-2 triggered endoplasmic reticulum (ER) stress and apoptosis. Integrative transcriptomic and proteomic analyses, performed using RNA sequencing and mass spectrometry, identified key molecular changes in cancer cells treated with Ru-2. For enhanced in vivo application, we developed a transferrin-based nanomedicine formulation, TF/Ru-2, incorporating Ru-2 into transferrin. In vivo studies demonstrated that both Ru-2 and TF/Ru-2 exhibited superior antitumor efficacy and improved biosafety compared to cisplatin. This study presents a novel ruthenium complex and a transferrin-based drug delivery platform with significant potential for future cancer therapies.
Collapse
Affiliation(s)
- Peng Liu
- Digestive Diseases Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, Guangdong, China
| | - Shangbo Zhou
- Digestive Diseases Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, Guangdong, China
| | - Zhijun Zhou
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Zihan Jin
- Digestive Diseases Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, Guangdong, China
| | - Wei Chen
- Digestive Diseases Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, Guangdong, China
| | - Zihang Li
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, Guangdong, China
| | - Jiaqi Xu
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, Guangdong, China
| | - Feng Chen
- Digestive Diseases Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, Guangdong, China
| | - You Li
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, Guangdong, China
| | - Yingfei Wen
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, Guangdong, China
| | - Shiqiang Zhang
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, Guangdong, China
| | - Changhua Zhang
- Digestive Diseases Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, Guangdong, China; Clinical Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, Guangdong, China
| | - Binbin Li
- Digestive Diseases Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, Guangdong, China.
| | - Jing Zhao
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, Guangdong, China.
| | - Hengxing Chen
- Digestive Diseases Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, Guangdong, China; Clinical Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, Guangdong, China.
| |
Collapse
|
10
|
Jiang W, Shi J, Zhu Y, Yin L, Song Y, Zhang J, Lin X, Zhong J, Lu Y, Ma Y. A novel prognostic model based on migrasome-related LncRNAs for gastric cancer. Sci Rep 2025; 15:14572. [PMID: 40281132 PMCID: PMC12032148 DOI: 10.1038/s41598-025-99781-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Accepted: 04/22/2025] [Indexed: 04/29/2025] Open
Abstract
Gastric cancer (GC) represents a substantial public health challenge, characterized by elevated morbidity and mortality rates. Migrasomes, a newly discovered type of extracellular vesicle, have been highlighted as important contributors to cancer progression, though their specific role in GC remains unclear. To address this issue, we developed the first prognostic model utilizing migrasome-related long non-coding RNAs (MRLs). This model aims to deepen the understanding of GC pathogenesis and improve patient outcomes. Clinical and transcriptional data for 407 GC patients from TCGA were classified as training and testing sets. Through Pearson correlation analysis, 537 MRLs were recognized, and LASSO and Cox regression analyses further refined the list to four key lncRNAs (AC012055.1, LINC01150, AC053503.4, AC107021.2) for constructing the prognostic model. Kaplan-Meier survival analysis indicated a significantly poorer prognosis for the high-risk group. PCA confirmed the model's robustness, and univariate and multivariate analyses validated it as an independent predictor of clinical outcomes. The ROC curve and C-index evaluations further affirmed the model's predictive power. We developed a nomogram combining the MRLs signature with clinical parameters to enhance prognostic accuracy. GO, KEGG and GSEA were performed on migrasome-related genes associated with GC. Furthermore, high-risk patients exhibited increased immune cell infiltration and reduced tumor mutation burden, both associated with poorer outcomes. Additionally, twenty-nine potential therapeutic agents were identified. This novel MRLs-based model provides crucial insights into GC biology and represents a valuable tool for improving patient management and therapeutic strategies.
Collapse
Affiliation(s)
- Wenhao Jiang
- Department of Medical Genetics, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Cheng Du, 610041, China
| | - Jiaying Shi
- Department of Medical Genetics, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Cheng Du, 610041, China
| | - Yingchuan Zhu
- Department of Medical Genetics, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Cheng Du, 610041, China
| | - Lan Yin
- Department of Medical Genetics, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Cheng Du, 610041, China
| | - Yue Song
- Department of Medical Genetics, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Cheng Du, 610041, China
| | - Jingfei Zhang
- Department of Medical Genetics, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Cheng Du, 610041, China
| | - Xinyu Lin
- Department of Medical Genetics, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Cheng Du, 610041, China
| | - Jiaxiu Zhong
- Department of Medical Genetics, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Cheng Du, 610041, China
| | - Yilu Lu
- Department of Medical Genetics, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Cheng Du, 610041, China
| | - Yongxin Ma
- Department of Medical Genetics, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Cheng Du, 610041, China.
| |
Collapse
|
11
|
Ma X, Wang Y, Kong L, Zhu W, Zhou X, Li J, Zhao W, Mao X, Tan G. FZHWT alleviates chronic atrophic gastritis by inhibiting inflammatory pathways and promoting mucosal repair. Int Immunopharmacol 2025; 153:114473. [PMID: 40127620 DOI: 10.1016/j.intimp.2025.114473] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2025] [Revised: 03/04/2025] [Accepted: 03/10/2025] [Indexed: 03/26/2025]
Abstract
BACKGROUND Chronic atrophic gastritis (CAG) is a prevalent chronic digestive disorder that, through sustained inflammation, can lead to severe mucosal damage and even gastric cancer. Current treatments offer limited efficacy, whereas Fu-Zheng-Huo-Wei Decoction (FZHWT), a traditional Chinese medicine (TCM) formulation, shows promising potential in treating CAG. PURPOSE This study aims to identify the key active components of FZHWT and evaluate its therapeutic effects on CAG. METHODS UPLC-MS/MS was used to identify the bioactive compounds in FZHWT. A CAG rat model was established to assess its therapeutic effects, and transcriptome sequencing was conducted to identify key targets and mechanisms. A CAG cell model was used for validation of the transcriptomic findings, and histological techniques and molecular biology methods were employed for further validation. RESULTS A total of 1362 chemical components were identified in FZHWT, of which 25 are bioavailable compounds. Differential metabolite analysis revealed four key active ingredients: Nicotiflorin, Stachydrine, 5-O-p-Coumaroylquinic acid, and N-(4-oxopentyl)-acetamide. In the CAG rat model, FZHWT significantly reduced inflammation and gastric mucosal damage. Transcriptome sequencing highlighted Sema5a as a key target and revealed the involvement of several inflammatory signaling pathways. In the CAG cell model, FZHWT alleviated CAG by inhibiting inflammation and promoting gastric mucosal repair. CONCLUSIONS FZHWT demonstrates significant therapeutic potential in treating CAG by modulating inflammatory pathways and promoting mucosal repair. This study provides new insights into the treatment of CAG and supports the modernization of multi-component TCM formulas.
Collapse
Affiliation(s)
- Xuehui Ma
- Department of Preclinical, Wuxi Affiliated Hospital of Nanjing University of Chinese Medicine, Wuxi 214200, China
| | - Yongli Wang
- Department of Preclinical, Wuxi Affiliated Hospital of Nanjing University of Chinese Medicine, Wuxi 214200, China
| | - Lingjing Kong
- Department of Preclinical, Wuxi Affiliated Hospital of Nanjing University of Chinese Medicine, Wuxi 214200, China
| | - Wen Zhu
- Department of Preclinical, Wuxi Affiliated Hospital of Nanjing University of Chinese Medicine, Wuxi 214200, China
| | - Xin Zhou
- Department of Preclinical, Wuxi Affiliated Hospital of Nanjing University of Chinese Medicine, Wuxi 214200, China
| | - Jian Li
- Department of Preclinical, Wuxi Affiliated Hospital of Nanjing University of Chinese Medicine, Wuxi 214200, China
| | - Wei Zhao
- Department of Preclinical, Wuxi Affiliated Hospital of Nanjing University of Chinese Medicine, Wuxi 214200, China
| | - Xiaoyun Mao
- Department of Preclinical, Wuxi Affiliated Hospital of Nanjing University of Chinese Medicine, Wuxi 214200, China
| | - Guangxing Tan
- Department of Preclinical, Wuxi Affiliated Hospital of Nanjing University of Chinese Medicine, Wuxi 214200, China.
| |
Collapse
|
12
|
Ożga K, Stepuch P, Maciejewski R, Sadok I. Promising Gastric Cancer Biomarkers-Focus on Tryptophan Metabolism via the Kynurenine Pathway. Int J Mol Sci 2025; 26:3706. [PMID: 40332338 PMCID: PMC12027761 DOI: 10.3390/ijms26083706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Revised: 04/08/2025] [Accepted: 04/10/2025] [Indexed: 05/08/2025] Open
Abstract
Currently, gastric cancer treatment remains an enormous challenge and requires a multidisciplinary approach. Globally, the incidence and prevalence of gastric cancer vary, with the highest rates found in East Asia, Central Europe, and Eastern Europe. Early diagnosis is critical for successful surgical removal of gastric cancer, but the disease often develops asymptomatically. Therefore, many cases are diagnosed at an advanced stage, resulting in poor survival. Metastatic gastric cancer also has a poor prognosis. Therefore, it is urgent to identify reliable molecular disease markers and develop an effective medical treatment for advanced stages of the disease. This review summarizes potential prognostic or predictive markers of gastric cancer. Furthermore, the role of tryptophan metabolites from the kynurenine pathway as prognostic, predictive, and diagnostic factors of gastric cancer is discussed, as this metabolic pathway is associated with tumor immune resistance.
Collapse
Affiliation(s)
- Kinga Ożga
- Department of Biomedicine and Environmental Research, Institute of Biological Sciences, Faculty of Medicine, Collegium Medicum, The John Paul II Catholic University of Lublin, Konstantynów 1J, 20-708 Lublin, Poland;
| | - Paweł Stepuch
- II Department of Oncological Surgery with Subdivision of Minimal Invasive Surgery, Center of Oncology of the Lublin Region St. Jana z Dukli, Jaczewskiego 7, 20-090 Lublin, Poland;
| | - Ryszard Maciejewski
- Faculty of Medicine, Collegium Medicum, The John Paul II Catholic University of Lublin, Konstantynów 1H, 20-708 Lublin, Poland;
| | - Ilona Sadok
- Department of Biomedical and Analytical Chemistry, Institute of Biological Sciences, Faculty of Medicine, Collegium Medicum, The John Paul II Catholic University of Lublin, Konstantynów 1J, 20-708 Lublin, Poland
| |
Collapse
|
13
|
Li D, Morgan DR, Corral JE, Montgomery EA, Riquelme A, Shah SC. Gastric Cancer Screening in the United States: A Review of Current Evidence, Challenges, and Future Perspectives. Am J Gastroenterol 2025; 120:765-777. [PMID: 40072512 DOI: 10.14309/ajg.0000000000003301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Accepted: 12/18/2024] [Indexed: 03/14/2025]
Abstract
Gastric cancer remains a leading cause of cancer-related mortality worldwide. In the United States, gastric cancer incidence and mortality are substantially higher among non-White racial and ethnic groups and new immigrants from high-incidence countries. This is in large part related to the higher prevalence of Helicobacter pylori -associated gastric premalignant changes in these populations. Apart from primary prevention, early detection of gastric cancer is the principal strategy to reduce gastric cancer mortality and improve survival. Extensive evidence in Asian countries has demonstrated the benefits of endoscopic screening in detecting early-stage gastric cancer and reducing gastric cancer-related mortality. By contrast, direct, high-quality US-based data, such as from large clinical trials or observational studies, on important outcomes of gastric cancer screening are still lacking. In this review, we evaluate and summarize the latest global evidence on the epidemiology and predisposing factors of gastric cancer as well as the efficacy, benefits vs. risks, and cost-effectiveness of gastric cancer screening. We further discuss the critical knowledge gaps and challenges in promoting gastric cancer screening in the United States. Dedicated research is urgently needed to enrich the US-based data on gastric cancer primary and secondary prevention to inform clinical practice and reduce gastric cancer-related morbidity and mortality in a cost and resource efficient manner.
Collapse
Affiliation(s)
- Dan Li
- Department of Gastroenterology, Kaiser Permanente Medical Center, Santa Clara, California, USA
- Kaiser Permanente Northern California Division of Research, Oakland, California, USA
| | - Douglas R Morgan
- Division of Gastroenterology, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Juan E Corral
- Division of Gastroenterology, Prisma Health, Greenville, South Carolina, USA
| | - Elizabeth A Montgomery
- Department of Pathology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Arnoldo Riquelme
- Department of Gastroenterology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Center for Control and Prevention of Cancer (CECAN), Santiago, Chile
| | - Shailja C Shah
- Division of Gastroenterology, University of California, San Diego, La Jolla, California, USA
- Gastroenterology Section, Jennifer Moreno Department of Veterans Affairs Medical Center, La Jolla, California, USA
| |
Collapse
|
14
|
Cheng M, Yu Y, Watanabe T, Yoshimoto Y, Kaji S, Yube Y, Kaneda M, Orita H, Mine S, Wu YY, Fukunaga T. Evaluation of three lymph node staging systems for prognostic prediction in gastric cancer: A systematic review and meta-analysis. World J Gastrointest Oncol 2025; 17:98103. [PMID: 40092941 PMCID: PMC11866223 DOI: 10.4251/wjgo.v17.i3.98103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 11/08/2024] [Accepted: 12/25/2024] [Indexed: 02/14/2025] Open
Abstract
BACKGROUND Lymph node status is a critical prognostic factor in gastric cancer (GC), but stage migration may occur in pathological lymph nodes (pN) staging. To address this, alternative staging systems such as the positive lymph node ratio (LNR) and log odds of positive lymph nodes (LODDS) were introduced. AIM To assess the prognostic accuracy and stratification efficacy of three nodal staging systems in GC. METHODS A systematic review identified 12 studies, from which hazard ratios (HRs) for overall survival (OS) were summarized. Sensitivity analyses, subgroup analyses, publication bias assessments, and quality evaluations were conducted. To enhance comparability, data from studies with identical cutoff values for pN, LNR, and LODDS were pooled. Homogeneous stratification was then applied to generate Kaplan-Meier (KM) survival curves, assessing the stratification efficacy of three staging systems. RESULTS The HRs and 95% confidence intervals for pN, LNR, and LODDS were 2.16 (1.72-2.73), 2.05 (1.65-2.55), and 3.15 (2.15-4.37), respectively, confirming all three as independent prognostic risk factors for OS. Comparative analysis of HRs demonstrated that LODDS had superior prognostic predictive power over LNR and pN. KM curves for pN (N0, N1, N2, N3a, N3b), LNR (0.1/0.2/0.5), and LODDS (-1.5/-1.0/-0.5/0) revealed significant differences (P < 0.001) among all prognostic stratifications. Mean differences and standard deviations in 60-month relative survival were 27.93% ± 0.29%, 41.70% ± 0.30%, and 26.60% ± 0.28% for pN, LNR, and LODDS, respectively. CONCLUSION All three staging systems are independent prognostic factors for OS. LODDS demonstrated the highest specificity, making it especially useful for predicting outcomes, while pN was the most effective in homogeneous stratification, offering better patient differentiation. These findings highlight the complementary roles of LODDS and pN in enhancing prognostic accuracy and stratification.
Collapse
Affiliation(s)
- Ming Cheng
- Department of Upper Gastroenterological Surgery, Juntendo University School of Medicine, Tokyo 113-8431, Japan
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Soochow University, Suzhou 215004, Jiangsu Province, China
| | - Yang Yu
- Department of Upper Gastroenterological Surgery, Juntendo University School of Medicine, Tokyo 113-8431, Japan
- Department of Gastrointestinal Surgery, Peking University Cancer Hospital, Beijing 100142, China
| | - Takehiro Watanabe
- Department of Upper Gastroenterological Surgery, Juntendo University School of Medicine, Tokyo 113-8431, Japan
| | - Yutaro Yoshimoto
- Department of Upper Gastroenterological Surgery, Juntendo University School of Medicine, Tokyo 113-8431, Japan
| | - Sanae Kaji
- Department of Upper Gastroenterological Surgery, Juntendo University School of Medicine, Tokyo 113-8431, Japan
| | - Yukinori Yube
- Department of Upper Gastroenterological Surgery, Juntendo University School of Medicine, Tokyo 113-8431, Japan
| | - Munehisa Kaneda
- Department of Upper Gastroenterological Surgery, Juntendo University School of Medicine, Tokyo 113-8431, Japan
| | - Hajime Orita
- Department of Upper Gastroenterological Surgery, Juntendo University School of Medicine, Tokyo 113-8431, Japan
| | - Shinji Mine
- Department of Upper Gastroenterological Surgery, Juntendo University School of Medicine, Tokyo 113-8431, Japan
| | - You-Yong Wu
- Department of Upper Gastroenterological Surgery, Juntendo University School of Medicine, Tokyo 113-8431, Japan
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Soochow University, Suzhou 215004, Jiangsu Province, China
| | - Tetsu Fukunaga
- Department of Upper Gastroenterological Surgery, Juntendo University School of Medicine, Tokyo 113-8431, Japan
| |
Collapse
|
15
|
Padmanaban D, Rajkumar P, Vijayakumar M, Jayaprakash V, Sekar N, Valayapathi R, Azhagudurai A, Chinnasamy A. Exploring sulphated polysaccharides from marine bivalves: Unveiling potent anti-gastric cancer activity through cell cycle arrest and apoptosis. JOURNAL OF ASIAN NATURAL PRODUCTS RESEARCH 2025:1-22. [PMID: 39985784 DOI: 10.1080/10286020.2025.2468319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 02/11/2025] [Accepted: 02/13/2025] [Indexed: 02/24/2025]
Abstract
This study investigates the extraction of partially purified sulphated polysaccharides from marine bivalves through enzymatic digestion followed by sequential deproteinization and evaluates their anti-gastric cancer activity. Chemical analysis confirmed high uronic acid and sulfate content, with FT-IR validating characteristic functional groups. Donax variabilis polysaccharide demonstrated significant, dose-dependent anti-proliferative effects against AGS cells. Mechanistic studies revealed morphological changes, ROS generation, LDH release, and mitochondrial membrane disruption, leading to apoptosis. Flow cytometry confirmed G0/G1 phase arrest. These findings suggest that D. variabilis sulphated polysaccharides exhibit potent anti-gastric cancer properties, likely through oxidative stress and mitochondrial dysfunction, warranting further investigation for therapeutic applications.
Collapse
Affiliation(s)
- Deepakrajasekar Padmanaban
- Department of Zoology, University of Madras, Guindy Campus, Chennai, Tamil Nadu600 025, India
- Crustacean Culture Division, ICAR-CIBA, 75, Santhome High Road, M. R. C. Nagar, R. A. Puram, Chennai, Tamil Nadu600 028, India
| | - Pavithra Rajkumar
- Department of Zoology, University of Madras, Guindy Campus, Chennai, Tamil Nadu600 025, India
- Department of Biological Sciences, SRM AP University, Mangalagiri Mandal, Guntur District, Andhra Pradesh522502, India
| | - Mahalakshmi Vijayakumar
- Department of Zoology, University of Madras, Guindy Campus, Chennai, Tamil Nadu600 025, India
| | - Vennila Jayaprakash
- Department of Zoology, University of Madras, Guindy Campus, Chennai, Tamil Nadu600 025, India
| | - Niranjni Sekar
- Department of Zoology, University of Madras, Guindy Campus, Chennai, Tamil Nadu600 025, India
| | - Rajasekar Valayapathi
- Department of Zoology, University of Madras, Guindy Campus, Chennai, Tamil Nadu600 025, India
| | - Aarthi Azhagudurai
- Department of Zoology, University of Madras, Guindy Campus, Chennai, Tamil Nadu600 025, India
| | - Arulvasu Chinnasamy
- Department of Zoology, University of Madras, Guindy Campus, Chennai, Tamil Nadu600 025, India
| |
Collapse
|
16
|
Lin L, Huang T, Li L, Lin Y, Chen F, Zheng Z, Zhou J, Wang Y, You W, Duan Y, An Y, He S, Ye W. Single-cell profiling reveals a reduced epithelial defense system, decreased immune responses and the immune regulatory roles of different fibroblast subpopulations in chronic atrophic gastritis. J Transl Med 2025; 23:159. [PMID: 39905493 PMCID: PMC11796052 DOI: 10.1186/s12967-025-06150-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 01/18/2025] [Indexed: 02/06/2025] Open
Abstract
PURPOSE To identify key cellular changes and molecular events in atrophic mucosa, we aimed to elucidate the molecular mechanisms driving the occurrence of chronic atrophic gastritis (CAG). METHODS We used single-cell RNA sequencing (scRNA-seq) to characterize changes in the epithelial state and tissue microenvironment associated with CAG. The molecular changes were identified by comparing differentially expressed genes (DEGs) between the two mucosa states. Gene Ontology (GO) pathway enrichment analysis was used to explore the potential functional changes in each cell subtype in atrophic mucosa. Gene set score analysis was conducted to compare the functional roles of different fibroblast subtypes and functional changes in cell subtypes between the CAG and control groups. Metabolic analysis was performed to compare the metabolic activity of C1Q+ macrophages under different conditions. NichNet analysis was used to analyze the regulatory relationships between CCL11+APOE+ fibroblasts and C1Q+ macrophages and between CCL11+APOE+ fibroblasts and CD8+ effector T cells. Transcription factor (TF) analysis was performed to determine the transcription status of different T-cell subtypes in atrophic and normal mucosa. RESULTS We generated a single-cell transcriptomic atlas from 3 CAG biopsy samples and paired adjacent normal tissues. Our analysis revealed that chief cells and parietal cells exhibited a loss of detoxification ability and that surface mucous cells displayed a reduced antimicrobial defense ability in CAG lesions. The mucous neck cells in CAG lesions showed upregulation of genes related to cell cycle transition, which may lead to aberrant DNA replication. Additionally, cells with the T exhaustion phenotype infiltrated under CAG condition. C1Q+ macrophages exhibited reduced phagocytosis, downregulated expression of pattern recognition receptors and decreased metabolic activity. NichNet analysis revealed that a subpopulation of CXCL11+APOE+ fibroblasts regulated the inflammatory response in the pathogenesis of atrophic gastritis. APSN+CXCL11+APOE+ fibroblasts were found to be associated with gastric cancer (GC) development. CONCLUSIONS The main goal of this study was to comprehensively elucidate the cellular changes in CAG lesions. We observed an immune decline in the mucosal microenvironment during the development of CAG, including a reduced immune response of C1Q+ macrophages, reduced cytotoxicity of T cells, and increased infiltration of exhausted T cells. Specifically, we demonstrated that different epithelial subtypes aberrantly express genes related to susceptibility to external bacterial infection and aberrant cell cycle progression. Our study provides new insights into the functions of epithelial changes and immune alterations during the development of CAG.
Collapse
Affiliation(s)
- Lin Lin
- Institute of Population Medicine, School of Public Health, Fujian Medical University, 1 Xuefu North Road, Fuzhou, 350122, China
| | - Tingxuan Huang
- Department of Gastroenterology, Fujian Medical University Union Hospital, Fuzhou, 350001, China
- Fujian Clinical Research Center for Digestive System Tumors and Upper Gastrointestinal Diseases, Fuzhou, 350001, China
| | - Lizhi Li
- Department of Pediatric Surgery, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001, China
| | - Yang Lin
- Department of Pediatric Surgery, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001, China
| | - Feng Chen
- Department of Pediatric Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| | - Ziyi Zheng
- Department of Pediatric Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| | - Jie Zhou
- Department of Pediatric Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| | - Yizhe Wang
- Institute of Population Medicine, School of Public Health, Fujian Medical University, 1 Xuefu North Road, Fuzhou, 350122, China
| | - Weihao You
- Institute of Population Medicine, School of Public Health, Fujian Medical University, 1 Xuefu North Road, Fuzhou, 350122, China
| | - Yujie Duan
- Institute of Population Medicine, School of Public Health, Fujian Medical University, 1 Xuefu North Road, Fuzhou, 350122, China
| | - Yawen An
- Institute of Population Medicine, School of Public Health, Fujian Medical University, 1 Xuefu North Road, Fuzhou, 350122, China
| | - Shiwei He
- Institute of Population Medicine, School of Public Health, Fujian Medical University, 1 Xuefu North Road, Fuzhou, 350122, China.
| | - Weimin Ye
- Institute of Population Medicine, School of Public Health, Fujian Medical University, 1 Xuefu North Road, Fuzhou, 350122, China.
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350122, China.
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, 17177, Sweden.
| |
Collapse
|
17
|
Li X, Tang B, Yujie O, Xu C, Yuan S. Single-cell RNA Sequencing Analysis Reveals Cancer-associated Fibroblast Signature for Prediction of Clinical Outcomes and Immunotherapy in Gastric Cancer. J Immunother 2025; 48:63-77. [PMID: 39206772 DOI: 10.1097/cji.0000000000000539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 07/12/2024] [Indexed: 09/04/2024]
Abstract
Gastric cancer (GC) is a significant worldwide health concern and is a leading cause of cancer-related mortality. Immunotherapy has arisen as a promising strategy to stimulate the patient's immune system in combating cancer cells. Nevertheless, the effectiveness of immunotherapy in individuals with gastric cancer (GC) is not yet optimal. Thus, it is crucial to discover biomarkers capable appof predicting the advantages of immunotherapy for tailored treatment. The tumor microenvironment (TME) and its constituents, including cancer-associated fibroblasts (CAFs), exert a substantial influence on immune responses and treatment outcomes. In this investigation, we utilized single-cell RNA sequencing to profile CAFs in GC and established a scoring method, referred to as the CAF score (CAFS), for the prediction of patient prognosis and response to immunotherapy. Through our analysis, we successfully identified distinct subgroups within CAFs based on CAF score (CAFS), namely CAFS-high and CAFS-low subgroups. Notably, we noted that individuals within the CAFS-high subgroup experienced a lessF favorable prognosis and displayed diminished responsiveness to immunotherapy in contrast to the CAFS low subgroup. Furthermore, we analyzed the mutation and immune characteristics of these subgroups, identifying differentially mutated genes and immune cell compositions. We established that CAFS could forecast treatment advantages in patients with gastric cancer, both for chemotherapy and immunotherapy. Its efficacy was additionally confirmed in contrast to other biomarkers, including Tumor Immune Dysfunction and Exclusion (TIDE) and Immunophenotypic Score (IPS). These findings emphasize the clinical relevance and potential utility of CAFS in guiding personalized treatment strategies for gastric cancer.
Collapse
Affiliation(s)
- Xiaoxiao Li
- Shandong University Cancer Center
- Center for GI Cancer Diagnosis and Treatment, The Affiliated Hospital of Qingdao University, Qingdao
| | - Bo Tang
- Department of Oncology, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China
| | - Ouyang Yujie
- Acupuncture and Massage College, Chengdu University of Traditional Chinese Medicine, Chengdu
| | - Chuan Xu
- Department of Oncology, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China
| | - Shuanghu Yuan
- Shandong University Cancer Center
- Department of Radiology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| |
Collapse
|
18
|
He L, Zhou J, Ding D, Jiang Y, Yang R, Li Z. MiR-99a-3p downregulates TRIM21 to promote gastric cancer development. Mol Cell Biochem 2025; 480:1001-1012. [PMID: 38720056 PMCID: PMC11835897 DOI: 10.1007/s11010-024-05005-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 04/03/2024] [Indexed: 02/19/2025]
Abstract
Gastric cancer (GC) stands as one of the most formidable malignancies worldwide. It is well-established that miRNAs play a crucial role in the initiation and progression of various human cancers. Among these, miR-99a-3p has been implicated in the pathogenesis of GC. In the context of our study, we embarked on the comprehensive examination of miR-99a-3p expression in GC cells. Additionally, we sought to establish a correlation between miR-99a-3p expression levels and the overall survival (OS) of GC patients, and our findings hinted at its potential role in predicting an unfavorable prognosis. To further investigate the functional implications of miR-99a-3p in GC, we conducted a series of cell-based experiments after successfully knocking down miR-99a-3p. These investigations uncovered a substantial inhibition of cellular events associated with tumor progression. Moreover, employing TargetScan, we identified Tripartite motif-containing protein 21 (TRIM21) as a putative target with a binding site for miR-99a-3p. Subsequent dual-luciferase reporter gene assay confirmed the direct interaction between miR-99a-3p and TRIM21. Western blot analysis validated the alteration in TRIM21 expression levels, revealing an upregulation upon miR-99a-3p knockdown. Building on these molecular findings, we extended our investigations to human GC tissues, where we observed a downregulation of TRIM21, which, notably, correlated with shorter overall survival. Lastly, to further solidify our conclusions, we conducted a series of in vitro and in vivo rescue experiments, collectively suggesting that miR-99a-3p promoted the progression of GC cells through the downregulation of TRIM21. In summary, our study comprehensively explored the role of miR-99a-3p in GC, revealing its association with unfavorable patient outcomes, functional implications in tumor progression, and a direct regulatory relationship with TRIM21. These findings collectively underscore the significance of miR-99a-3p in the pathogenesis of GC and present a potential therapeutic avenue for further investigation.
Collapse
Affiliation(s)
- Ling He
- Department of Gastroenterology, Affiliated Hospital of Jiangxi University of TCM, No.445, Bayi Road, Nanchang, 330006, Jiangxi, China
| | - Jiaoli Zhou
- Jiangxi University of TCM, No.56, Yangming Road, Nanchang, 330006, Jiangxi, China
| | - Doukun Ding
- Jiangxi University of TCM, No.56, Yangming Road, Nanchang, 330006, Jiangxi, China
| | - Yongjing Jiang
- Jiangxi University of TCM, No.56, Yangming Road, Nanchang, 330006, Jiangxi, China
| | - Rui Yang
- Jiangxi University of TCM, No.56, Yangming Road, Nanchang, 330006, Jiangxi, China
| | - Zhiming Li
- Department of Oncology, Affiliated Hospital of Jiangxi University of TCM, No.445, Bayi Road, Nanchang, 330006, Jiangxi, China.
| |
Collapse
|
19
|
Xie F, Chen B, Lyu Y, Yu P, Fang C, Leung KT, Wang S, Xu D, Yu J, Lo KW, To KF, Kang W. Deciphering the Differences Between Epstein-Barr Virus-Associated and Negative Gastric Cancer in the Prospect of CDKN2A Genomic Alterations and Lymphoid Infiltration. Cancer Med 2025; 14:e70409. [PMID: 39844467 PMCID: PMC11754542 DOI: 10.1002/cam4.70409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 10/24/2024] [Accepted: 10/27/2024] [Indexed: 01/24/2025] Open
Abstract
BACKGROUND Gastric cancer (GC) is a major health concern worldwide. One important contributing factor is the presence of the Epstein-Barr virus (EBV). However, the molecular pattern of how EBV participates in the malignant transition process remains unclear. METHODS GC samples were stained by immunohistochemistry, fluorescent and EBV-encoded small RNA in situ hybridization to identify CD8 expression, CDKN2A genomic alteration, and EBV existence. Functional potentials of EBV infection were predicted by bioinformatic enrichment analysis. RESULTS CDKN2A genestayed intact in all EBV-associated GC cases. Meanwhile, CDKN2A deletion (8.43% cases) was exclusive to EBV-negative GC cases. Furthermore, EBV infection was positively correlated with CD8+T cell infiltration, and both of them predicted better prognosis. CONCLUSION This study highlighted the comprehensive impact of EBV infection in GC formation and proposed a thought-provoking observation for further investigation into the roles of CDKN2A and EBV infection in gastric tumorigenesis.
Collapse
Affiliation(s)
- Fuda Xie
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Sir Y.K. Pao Cancer Center, Prince of Wales HospitalThe Chinese University of Hong KongHong KongSARChina
- Institute of Digestive Disease, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health ScienceThe Chinese University of Hong KongHong KongSARChina
- CUHK‐Shenzhen Research InstituteShenzhenChina
| | - Bonan Chen
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Sir Y.K. Pao Cancer Center, Prince of Wales HospitalThe Chinese University of Hong KongHong KongSARChina
- Institute of Digestive Disease, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health ScienceThe Chinese University of Hong KongHong KongSARChina
- CUHK‐Shenzhen Research InstituteShenzhenChina
| | - Yang Lyu
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Sir Y.K. Pao Cancer Center, Prince of Wales HospitalThe Chinese University of Hong KongHong KongSARChina
| | - Peiyao Yu
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Sir Y.K. Pao Cancer Center, Prince of Wales HospitalThe Chinese University of Hong KongHong KongSARChina
| | - Canbin Fang
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Sir Y.K. Pao Cancer Center, Prince of Wales HospitalThe Chinese University of Hong KongHong KongSARChina
| | - Kam Tong Leung
- Department of PediatricsThe Chinese University of Hong KongHong KongSARChina
| | - Shouyu Wang
- Department of Hepatobiliary SurgeryThe Affiliated Drum Tower Hospital of Nanjing University Medical SchoolNanjingChina
| | - Dazhi Xu
- Department of Gastric Surgery, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
| | - Jun Yu
- Institute of Digestive Disease, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health ScienceThe Chinese University of Hong KongHong KongSARChina
- Department of Medicine and TherapeuticsThe Chinese University of Hong KongHong KongSARChina
| | - Kwok Wai Lo
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Sir Y.K. Pao Cancer Center, Prince of Wales HospitalThe Chinese University of Hong KongHong KongSARChina
| | - Ka Fai To
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Sir Y.K. Pao Cancer Center, Prince of Wales HospitalThe Chinese University of Hong KongHong KongSARChina
| | - Wei Kang
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Sir Y.K. Pao Cancer Center, Prince of Wales HospitalThe Chinese University of Hong KongHong KongSARChina
- Institute of Digestive Disease, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health ScienceThe Chinese University of Hong KongHong KongSARChina
- CUHK‐Shenzhen Research InstituteShenzhenChina
| |
Collapse
|
20
|
Laurindo LF, Pomini KT, de Lima EP, Laurindo LF, Rodrigues VD, da Silva Camarinha Oliveira J, Araújo AC, Guiguer EL, Rici REG, Maria DA, de Alvares Goulart R, Direito R, Barbalho SM. Isoorientin: Unveiling the hidden flavonoid's promise in combating cancer development and progression - A comprehensive review. Life Sci 2025; 360:123280. [PMID: 39608447 DOI: 10.1016/j.lfs.2024.123280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 11/10/2024] [Accepted: 11/25/2024] [Indexed: 11/30/2024]
Abstract
Cancer remains one of the leading causes of mortality worldwide, characterized by uncontrolled cell growth and the ability of tumors to invade surrounding tissues and spread to distant organs. Despite significant advancements in early detection, diagnosis, and treatment, many cancers still present substantial challenges due to their heterogeneity, resistance to conventional therapies, and severe side effects of existing treatments. Consequently, there is an ongoing need for novel therapeutic agents to selectively target cancer cells, enhance the efficacy of current treatments, and minimize adverse effects. Isoorientin (ISO) is a naturally occurring flavonoid known for its anticancer properties. ISO has demonstrated the ability to influence several critical processes in cancer progression, such as cell proliferation, apoptosis, and metastasis. Due to the absence of clinical trials, we included only in vitro studies, reviewing 13 investigations. These studies covered diverse cancer types, including lung, brain, oral, liver, pancreatic, and gastric cancers, and assessed various outcomes related to cell viability, apoptosis, migration, and molecular pathway modulation. By synthesizing data from these investigations, our review seeks to provide a thorough understanding of ISO's anticancer effects, its mechanisms of action, and its potential as a therapeutic agent.
Collapse
Affiliation(s)
- Lucas Fornari Laurindo
- Department of Biochemistry and Pharmacology, School of Medicine, Faculdade de Medicina de Marília (FAMEMA), Marília 17519-030, São Paulo, Brazil; Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, São Paulo, Brazil; Department of Administration, Associate Degree in Hospital Management, Universidade de Marília (UNIMAR), Marília 17525-902, São Paulo, Brazil.
| | - Karina Torres Pomini
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, São Paulo, Brazil; Postgraduate Program in Structural and Functional Interactions in Rehabilitation, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, São Paulo, Brazil
| | - Enzo Pereira de Lima
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, São Paulo, Brazil
| | - Lívia Fornari Laurindo
- Department of Biochemistry and Pharmacology, School of Medicine, Faculdade de Medicina de São José do Rio Preto (FAMERP), São José do Rio Preto 15090-000, São Paulo, Brazil
| | - Victória Dogani Rodrigues
- Department of Biochemistry and Pharmacology, School of Medicine, Faculdade de Medicina de Marília (FAMEMA), Marília 17519-030, São Paulo, Brazil
| | - Jéssica da Silva Camarinha Oliveira
- Department of Biochemistry and Pharmacology, School of Medicine, Faculdade de Medicina de Marília (FAMEMA), Marília 17519-030, São Paulo, Brazil
| | - Adriano Cressoni Araújo
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, São Paulo, Brazil; Postgraduate Program in Structural and Functional Interactions in Rehabilitation, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, São Paulo, Brazil
| | - Elen Landgraf Guiguer
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, São Paulo, Brazil; Postgraduate Program in Structural and Functional Interactions in Rehabilitation, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, São Paulo, Brazil; Department of Biochemistry and Nutrition, School of Food and Technology of Marília (FATEC), Marília 17500-000, São Paulo, Brazil
| | - Rose Eli Grassi Rici
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, São Paulo, Brazil; Graduate Program in Anatomy of Domestic and Wild Animals, College of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo 05508-220, São Paulo, Brazil
| | - Durvanei Augusto Maria
- Development and innovation Laboratory, Butantan Institute, São Paulo 05585-000, São Paulo, Brazil
| | - Ricardo de Alvares Goulart
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, São Paulo, Brazil; Postgraduate Program in Structural and Functional Interactions in Rehabilitation, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, São Paulo, Brazil
| | - Rosa Direito
- Laboratory of Systems Integration Pharmacology, Clinical and Regulatory Science, Research Institute for Medicines, Universidade de Lisboa (iMed.ULisboa), Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal.
| | - Sandra Maria Barbalho
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, São Paulo, Brazil; Postgraduate Program in Structural and Functional Interactions in Rehabilitation, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, São Paulo, Brazil; Department of Biochemistry and Nutrition, School of Food and Technology of Marília (FATEC), Marília 17500-000, São Paulo, Brazil; UNIMAR Charity Hospital, Universidade de Marília (UNIMAR), Marília 17525-902, São Paulo, Brazil
| |
Collapse
|
21
|
Shin JH, Shin SH. A Comprehensive Review of Naringenin, a Promising Phytochemical with Therapeutic Potential. J Microbiol Biotechnol 2024; 34:2425-2438. [PMID: 39572023 PMCID: PMC11733549 DOI: 10.4014/jmb.2410.10006] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 11/01/2024] [Accepted: 11/12/2024] [Indexed: 12/31/2024]
Abstract
Disorders, including cancer, metabolic disorders, and neurodegenerative diseases, can threaten human health; therefore, disease prevention is essential. Naringenin, a phytochemical with low toxicity, has been used in various disease prevention studies. This study aimed to comprehensively review the effects of naringenin on human health. First, we introduced the general characteristics of naringenin and its pharmacokinetic features when absorbed in the body. Next, we summarized the inhibitory effects of naringenin on colorectal, gastric, lung, breast, ovarian, cervical, prostate, bladder, liver, pancreatic, and skin cancers in preclinical studies. Lastly, we investigated the inhibitory effects of naringenin on metabolic disorders, including diabetes, obesity, hyperlipidemia, hypertension, cardiac toxicity, hypertrophy, steatosis, liver disease, and arteriosclerosis, as well as on neurodegenerative diseases, including Alzheimer's disease and Parkinson's disease. In conclusion, naringenin may serve as a significant natural compound that benefits human health.
Collapse
Affiliation(s)
- Jun Hong Shin
- Department of Food and Nutrition, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Seung Ho Shin
- Department of Food and Nutrition, Gyeongsang National University, Jinju 52828, Republic of Korea
- Department of Bio & Medical Bigdata (BK4 Program), Gyeongsang National University, Jinju 52828, Republic of Korea
| |
Collapse
|
22
|
Liu Z, Xu H, You W, Pan K, Li W. Helicobacter pylori eradication for primary prevention of gastric cancer: progresses and challenges. JOURNAL OF THE NATIONAL CANCER CENTER 2024; 4:299-310. [PMID: 39735441 PMCID: PMC11674435 DOI: 10.1016/j.jncc.2024.06.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 06/20/2024] [Accepted: 06/27/2024] [Indexed: 12/31/2024] Open
Abstract
Gastric cancer remains a significant global health challenge, causing a substantial number of cancer-related deaths, particularly in China. While the exact causes of gastric cancer are still being investigated, Helicobacter pylori (H. pylori) infection has been identified as the primary risk factor, which triggers chronic inflammation and a multistage progression of gastric lesions that may lead to carcinogenesis over a long latency time. Since the 1990s, numerous efforts have focused on assessing the effectiveness of H. pylori eradication in preventing new cases of gastric cancer among both the general population and patients who have undergone early-stage cancer treatment. This body of work, including several community-based interventions and meta-analyses, has shown a reduction in both the incidence of and mortality from gastric cancer following H. pylori treatment, alongside a decreased risk of metachronous gastric cancer. In this review, we seek to consolidate current knowledge on the effects of H. pylori treatment on gastric cancer prevention, its systemic consequences, cost-effectiveness, and the influence of antibiotic resistance and host characteristics on treatment outcomes. We further discuss the potential for precision primary prevention of H. pylori treatment and comment on the efficient implementation of test-and-treat policies and allocation of health resources towards minimizing the burden of gastric cancer globally.
Collapse
Affiliation(s)
- Zongchao Liu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Cancer Epidemiology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Hengmin Xu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Cancer Epidemiology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Weicheng You
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Cancer Epidemiology, Peking University Cancer Hospital and Institute, Beijing, China
| | - Kaifeng Pan
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Cancer Epidemiology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Wenqing Li
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Cancer Epidemiology, Peking University Cancer Hospital & Institute, Beijing, China
| |
Collapse
|
23
|
Liu J, Li M, Chen G, Yang J, Jiang Y, Li F, Hua H. Jianwei Xiaoyan granule ameliorates chronic atrophic gastritis by regulating HIF-1α-VEGF pathway. JOURNAL OF ETHNOPHARMACOLOGY 2024; 334:118591. [PMID: 39025161 DOI: 10.1016/j.jep.2024.118591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 06/19/2024] [Accepted: 07/15/2024] [Indexed: 07/20/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Jianwei Xiaoyan Granule (JWXYG) is the traditional Chinese medicine preparation in Jiangyin Hospital Affiliated to Nanjing University of Chinese Medicine, which has been widely used in clinical treatment of chronic atrophic gastritis (CAG). However, the material basis and potential mechanism of JWXYG in the treatment of CAG are not clear. PURPOSE To explore the material basis and potential mechanism of JWXYG in the treatment of CAG. METHODS In this study, the components of JWXYG were analyzed by HPLC-Q-TOF-MS/MS. Then, the CAG model in rats established by a composite modeling method and MC cell model induced by MNNG were used to explore the improvement effect of JWXYG on CAG. Finally, the potential mechanism of JWXYG in the treatment of CAG was preliminarily predicted based on network pharmacology and validated experimentally. RESULTS Thirty-one components of JWXYG were analyzed through HPLC-Q-TOF-MS/MS, such as albiflorin, paeoniflorin, lobetyolin firstly. Research results in vivo showed that the gastric mucosa became thinner, intestinal metaplasia appeared, the number of glands was reduced, the serum levels of PG I and PG II increased and the contents of G17 and IL-6 reduced in CAG model rats. After 4 weeks of JWXYG (2.70 g/kg) administration, these conditions were significantly improved. In addition, cell viability, migration, and invasion of MNNG-induced MC cells was inhibited by JWXYG treatment (800 μg/mL). Furthermore, the results of network pharmacology indicated that HIF-1 and VEGF signaling pathways might play important roles in the therapeutic process. Then the results of Western blot, immunohistochemistry and immunofluorescence confirmed that with JWXYG treatment, the increased expression of HIF-1α, VEGF and VEGFR2 in gastric issue of CAG rats were restrained. Eventually, potential components of JWXYG in the treatment of CAG were predicted through molecular docking to elucidate the material basis. CONCLUSION JWXYG could inhibit angiogenesis by regulating HIF-1α-VEGF pathway to exert therapeutic effects on CAG. Our study explored the potential mechanisms and material basis of JWXYG in the treatment of CAG and provides experimental data for the clinical rational application of JWXYG.
Collapse
Affiliation(s)
- Jia Liu
- Jiangyin Hospital Affiliated to Nanjing University of Chinese Medicine, Jiangyin, 214400, Jiangsu Province, China.
| | - Mengyu Li
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Research Center for Traceability and Standardization of TCMs, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China.
| | - Guobao Chen
- Jiangyin Hospital Affiliated to Nanjing University of Chinese Medicine, Jiangyin, 214400, Jiangsu Province, China.
| | - Junhui Yang
- Jiangyin Hospital Affiliated to Nanjing University of Chinese Medicine, Jiangyin, 214400, Jiangsu Province, China.
| | - Ying Jiang
- Jiangyin Hospital Affiliated to Nanjing University of Chinese Medicine, Jiangyin, 214400, Jiangsu Province, China.
| | - Fang Li
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Research Center for Traceability and Standardization of TCMs, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China.
| | - Haibing Hua
- Jiangyin Hospital Affiliated to Nanjing University of Chinese Medicine, Jiangyin, 214400, Jiangsu Province, China.
| |
Collapse
|
24
|
Si YT, Xiong XS, Wang JT, Yuan Q, Li YT, Tang JW, Li YN, Zhang XY, Li ZK, Lai JX, Umar Z, Yang WX, Li F, Wang L, Gu B. Identification of chronic non-atrophic gastritis and intestinal metaplasia stages in the Correa's cascade through machine learning analyses of SERS spectral signature of non-invasively-collected human gastric fluid samples. Biosens Bioelectron 2024; 262:116530. [PMID: 38943854 DOI: 10.1016/j.bios.2024.116530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 06/13/2024] [Accepted: 06/15/2024] [Indexed: 07/01/2024]
Abstract
The progression of gastric cancer involves a complex multi-stage process, with gastroscopy and biopsy being the standard procedures for diagnosing gastric diseases. This study introduces an innovative non-invasive approach to differentiate gastric disease stage using gastric fluid samples through machine-learning-assisted surface-enhanced Raman spectroscopy (SERS). This method effectively identifies different stages of gastric lesions. The XGBoost algorithm demonstrates the highest accuracy of 96.88% and 91.67%, respectively, in distinguishing chronic non-atrophic gastritis from intestinal metaplasia and different subtypes of gastritis (mild, moderate, and severe). Through blinded testing validation, the model can achieve more than 80% accuracy. These findings offer new possibilities for rapid, cost-effective, and minimally invasive diagnosis of gastric diseases.
Collapse
Affiliation(s)
- Yu-Ting Si
- Medical Technology School, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Xue-Song Xiong
- Huai'an Hospital Affiliated to Yangzhou University (The Fifth People's Hospital of Huai'an), Huai'an, Jiangsu Province, China
| | - Jin-Ting Wang
- Huai'an Hospital Affiliated to Yangzhou University (The Fifth People's Hospital of Huai'an), Huai'an, Jiangsu Province, China
| | - Quan Yuan
- School of Medical Informatics and Engineering, Xuzhou Medical University, Xuzhou, Jiangsu Province, China; Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, China
| | - Yu-Ting Li
- Huai'an Hospital Affiliated to Yangzhou University (The Fifth People's Hospital of Huai'an), Huai'an, Jiangsu Province, China
| | - Jia-Wei Tang
- Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, China; Division of Microbiology and Immunology, School of Biomedical Sciences, The University of Western Australia, Crawley, Western Australia, Australia
| | - Yong-Nian Li
- Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, China
| | - Xin-Yu Zhang
- Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, China
| | - Zheng-Kang Li
- Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, China
| | - Jin-Xin Lai
- Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, China
| | - Zeeshan Umar
- Marshall Laboratory of Biomedical Engineering, School of Medicine, Shenzhen University, Guangdong Province, China
| | - Wei-Xuan Yang
- Huai'an Hospital Affiliated to Yangzhou University (The Fifth People's Hospital of Huai'an), Huai'an, Jiangsu Province, China
| | - Fen Li
- Huai'an Hospital Affiliated to Yangzhou University (The Fifth People's Hospital of Huai'an), Huai'an, Jiangsu Province, China.
| | - Liang Wang
- School of Medical Informatics and Engineering, Xuzhou Medical University, Xuzhou, Jiangsu Province, China; Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, China; Division of Microbiology and Immunology, School of Biomedical Sciences, The University of Western Australia, Crawley, Western Australia, Australia; The Center for Precision Health, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, 6027, Australia.
| | - Bing Gu
- Medical Technology School, Xuzhou Medical University, Xuzhou, Jiangsu Province, China; Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, China.
| |
Collapse
|
25
|
Dong X, Cheng T, Zhang L, Song L, Shi C. CircTSN promotes the proliferation and metastasis of gastric cancer through the miR-1825/SLC38A2 signaling axis. Discov Oncol 2024; 15:533. [PMID: 39379756 PMCID: PMC11461732 DOI: 10.1007/s12672-024-01407-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Accepted: 10/01/2024] [Indexed: 10/10/2024] Open
Abstract
BACKGROUND Comprehensive treatment of gastric cancer (GC) is progressing, but the rapid proliferation and metastasis of GC remains a cause of high recurrence and mortality rates. In this study we investigated GC-associated circRNA tending to yield more insight into the mechanisms of gastric cancer development. METHODS We detected the expression levels of circTSN in GC tissues and cell lines using qRT-PCR. The circular structure of circTSN was confirmed by Sanger sequencing, agarose gel electrophoresis and RNase R. A series of cell functional experiments were employed to investigate the implication of circTSN aberrant expression on the proliferation and metastasis of GC cells. The predicted binding domain between circTSN and miR-1825 was analyzed by luciferase reporter gene analysis. Meanwhile, subcutaneous tumor xenografts in nude mice were used to validate the role of circTSN in vivo. RESULTS It was found that RNA levels of circTSN were significantly elevated in GC tissues and cell lines, which was also confirmed to contain a closed-loop structure. CCK8, clone formation, EdU, transwell and in vivo experiments indicated that the highly expressed circTSN was involved in the proliferation and metastasis process of GC. In addition, circTSN modulates the expression of SLC38A2 by sequence-specific binding to miR-1825. CONCLUSION This study identified that circTSN, which is highly expressed in GC, was able to contribute to the proliferation and metastasis of GC cell through miR-1825/SLC38A2 axis and this might provide a new candidate target for the precision treatment of GC.
Collapse
Affiliation(s)
- Xuqiang Dong
- Department of Gastrointestinal Surgery, Yixing People's Hospital, Wuxi, Jiangsu, China
| | - Tianyu Cheng
- Department of Gastrointestinal Surgery, Yixing People's Hospital, Wuxi, Jiangsu, China
| | - Lijun Zhang
- Department of Gastrointestinal Surgery, Yixing People's Hospital, Wuxi, Jiangsu, China
| | - Liqun Song
- Department of Operating Room, Yixing People's Hospital, Wuxi, Jiangsu, China.
| | - Chao Shi
- Department of Gastrointestinal Surgery, Yixing People's Hospital, Wuxi, Jiangsu, China.
| |
Collapse
|
26
|
Lordick F, Rha SY, Muro K, Yong WP, Lordick Obermannová R. Systemic Therapy of Gastric Cancer-State of the Art and Future Perspectives. Cancers (Basel) 2024; 16:3337. [PMID: 39409957 PMCID: PMC11475804 DOI: 10.3390/cancers16193337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 09/27/2024] [Accepted: 09/27/2024] [Indexed: 10/20/2024] Open
Abstract
Background: The prognosis of patients diagnosed with locally advanced and metastatic gastric and esophago-gastric junction cancer is critical. The optimal choice of systemic therapy is essential to optimize survival outcomes. Methods: A comprehensive literature review via PubMed and analysis of major oncology congresses (European Society for Medical Oncology and American Society of Clinical Oncology websites) were conducted to ascertain the current status and latest developments in the systemic treatment of patients with localized or advanced gastric and esophago-gastric junction adenocarcinoma. Results: While neoadjuvant and perioperative chemotherapy for localized tumor stages is the preferred approach in the Western Hemisphere, adjuvant chemotherapy remains the preferred course of action in East Asia. The administration of chemotherapy, typically in the form of combinations comprising platinum and fluoropyrimidine compounds in combination with docetaxel, represents a standard of care. Investigations are underway into the potential of immunotherapy and other biologically targeted agents in the perioperative setting. To select the most appropriate therapy for advanced gastric cancer, including adenocarcinoma of the esophago-gastric junction, it is essential to determine biomarkers such as HER2 expression, PD-L1 combined positive score (CPS) (combined positive score), Claudin 18.2, and microsatellite instability (MSI). In the present clinical context, the standard first-line therapy is a combination of fluoropyrimidine and a platinum derivative. The selection of chemotherapy in combination with antibodies is contingent upon the specific biomarker under consideration. Conclusions: This article reviews the current state of the art based on recent clinical trial results and provides an outlook on the future of systemic therapy.
Collapse
Affiliation(s)
- Florian Lordick
- Department of Medicine (Oncology, Gastroenterology, Hepatology, Pulmonology), University of Leipzig Medical Center, Cancer Center Central Germany, 04103 Leipzig, Germany
| | - Sun Young Rha
- Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Kei Muro
- Department of Clinical Oncology, Aichi Cancer Center Hospital, Nagoya 464-8681, Japan
| | - Wei Peng Yong
- Department of Haematology-Oncology, National University Cancer Institute, Singapore 119074, Singapore
| | - Radka Lordick Obermannová
- Department of Comprehensive Cancer Care, Masaryk Memorial Cancer Institute, Faculty of Medicine, Masaryk University, 656 53 Brno, Czech Republic
| |
Collapse
|
27
|
Basirinia G, Ali M, Comelli A, Sperandeo A, Piana S, Alongi P, Longo C, Di Raimondo D, Tuttolomondo A, Benfante V. Theranostic Approaches for Gastric Cancer: An Overview of In Vitro and In Vivo Investigations. Cancers (Basel) 2024; 16:3323. [PMID: 39409942 PMCID: PMC11476023 DOI: 10.3390/cancers16193323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 09/24/2024] [Accepted: 09/26/2024] [Indexed: 10/20/2024] Open
Abstract
Gastric cancer (GC) is the second most common cause of cancer-related death worldwide and a serious public health concern. This high death rate is mostly caused by late-stage diagnoses, which lead to poor treatment outcomes. Radiation immunotherapy and targeted therapies are becoming increasingly popular in GC treatment, in addition to surgery and systemic chemotherapy. In this review, we have focused on both in vitro and in vivo research, which presents a summary of recent developments in targeted therapies for gastric cancer. We explore targeted therapy approaches, including integrin receptors, HER2, Claudin 18, and glutathione-responsive systems. For instance, therapies targeting the integrin receptors such as the αvβ3 and αvβ5 integrins have shown promise in enhancing diagnostic precision and treatment efficacy. Furthermore, nanotechnology provides novel approaches to targeted drug delivery and imaging. These include glutathione-responsive nanoplatforms and cyclic RGD peptide-conjugated nanoparticles. These novel strategies seek to reduce systemic toxicity while increasing specificity and efficacy. To sum up, the review addresses the significance of personalized medicine and advancements in gastric cancer-targeted therapies. It explores potential methods for enhancing gastric cancer prognosis and treatment in the future.
Collapse
Affiliation(s)
- Ghazal Basirinia
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Molecular and Clinical Medicine, University of Palermo, 90127 Palermo, Italy; (D.D.R.); (A.T.)
- Ri.MED Foundation, Via Bandiera 11, 90133 Palermo, Italy; (G.B.); (M.A.)
| | - Muhammad Ali
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Molecular and Clinical Medicine, University of Palermo, 90127 Palermo, Italy; (D.D.R.); (A.T.)
- Ri.MED Foundation, Via Bandiera 11, 90133 Palermo, Italy; (G.B.); (M.A.)
| | - Albert Comelli
- Ri.MED Foundation, Via Bandiera 11, 90133 Palermo, Italy; (G.B.); (M.A.)
- NBFC—National Biodiversity Future Center, 90133 Palermo, Italy
| | - Alessandro Sperandeo
- Pharmaceutical Factory, La Maddalena S.P.A., Via San Lorenzo Colli, 312/d, 90146 Palermo, Italy; (A.S.); (S.P.)
| | - Sebastiano Piana
- Pharmaceutical Factory, La Maddalena S.P.A., Via San Lorenzo Colli, 312/d, 90146 Palermo, Italy; (A.S.); (S.P.)
| | - Pierpaolo Alongi
- Nuclear Medicine Unit, A.R.N.A.S. Civico Di Cristina e Benfratelli Hospitals, P.zza N. Leotta 4, 90127 Palermo, Italy; (P.A.); (C.L.)
- Advanced Diagnostic Imaging-INNOVA Project, Department of Radiological Sciences, A.R.N.A.S. Civico Di Cristina e Benfratelli Hospitals, P.zza N. Leotta 4, 90127 Palermo, Italy
| | - Costanza Longo
- Nuclear Medicine Unit, A.R.N.A.S. Civico Di Cristina e Benfratelli Hospitals, P.zza N. Leotta 4, 90127 Palermo, Italy; (P.A.); (C.L.)
| | - Domenico Di Raimondo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Molecular and Clinical Medicine, University of Palermo, 90127 Palermo, Italy; (D.D.R.); (A.T.)
| | - Antonino Tuttolomondo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Molecular and Clinical Medicine, University of Palermo, 90127 Palermo, Italy; (D.D.R.); (A.T.)
| | - Viviana Benfante
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Molecular and Clinical Medicine, University of Palermo, 90127 Palermo, Italy; (D.D.R.); (A.T.)
- Advanced Diagnostic Imaging-INNOVA Project, Department of Radiological Sciences, A.R.N.A.S. Civico Di Cristina e Benfratelli Hospitals, P.zza N. Leotta 4, 90127 Palermo, Italy
| |
Collapse
|
28
|
Wang Y, Guo Z, Yang Z, Deng Q, Huang Y, Chen Y. Long intergenic noncoding RNA for IGF2BP2 stability suppresses gastric cancer cell apoptosis by inhibiting the maturation of microRNA-34a. Open Med (Wars) 2024; 19:20240992. [PMID: 39381424 PMCID: PMC11459274 DOI: 10.1515/med-2024-0992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 05/05/2024] [Accepted: 06/09/2024] [Indexed: 10/10/2024] Open
Abstract
The oncogenic role of long intergenic noncoding RNA for IGF2BP2 stability (LINRIS) has been reported in colorectal cancer. This research aimed to study its potential involvement in gastric cancer (GC). In this study, paired GC and non-tumor tissues were obtained from 64 GC patients, and the levels of LINRIS, mature microRNA-34a (miR-34a), and miR-34a precursor in these tissues were measured with RT-qPCR. Linear regression was used to analyze their correlations. The role of LINRIS overexpression and siRNA silencing in regulating the maturation of miR-34a was analyzed by RT-qPCR. Cell apoptosis was studied with flow cytometry. It was observed that LINRIS was overexpressed in GC and showed a negative correlation with mature miR-34a, but not miR-34a precursor. In GC cells, LINRIS siRNA silencing upregulated mature miR-34a level, but not miR-34a precursor level. LINRIS overexpression downregulated miR-34a level. Cell apoptosis analysis showed that LINRIS siRNA silencing and miR-34a overexpression promoted GC cell apoptosis and suppressed cell migration and invasion, while LINRIS overexpression suppressed cell apoptosis and enhanced cell migration and invasion. In addition, the effect of LINRIS overexpression was reversed by miR-34a overexpression. Therefore, LINRIS siRNA silencing in GC may promote cell apoptosis by promoting miR-34a maturation.
Collapse
Affiliation(s)
- Yao Wang
- Department of Gastrointestinal Surgery, Zhongshan City People’s Hospital, Zhongshan, Guangdong, 528400, P.R. China
| | - Zhigang Guo
- Department of Gastrointestinal Surgery, Zhongshan City People’s Hospital, Zhongshan, Guangdong, 528400, P.R. China
| | - Zhifeng Yang
- Department of Gastrointestinal Surgery, Zhongshan City People’s Hospital, Zhongshan, Guangdong, 528400, P.R. China
| | - Qingyan Deng
- Department of Gastrointestinal Surgery, Zhongshan City People’s Hospital, Zhongshan, Guangdong, 528400, P.R. China
| | - Yueming Huang
- Department of Gastrointestinal Surgery, Zhongshan City People’s Hospital, Zhongshan, Guangdong, 528400, P.R. China
| | - Yanhong Chen
- Department of Hospital Infection Management, Zhongshan City People’s Hospital, No. 2 Sunwen East Road, Shiqi District, Zhongshan, Guangdong, 528400, P.R. China
| |
Collapse
|
29
|
Li F, Wang Y, Ping X, Yin JC, Wang F, Zhang X, Li X, Zhai J, Shen L. Molecular evolution of intestinal-type early gastric cancer according to Correa cascade. J Biomed Res 2024; 38:1-16. [PMID: 39314047 DOI: 10.7555/jbr.38.20240118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/25/2024] Open
Abstract
Early screening is crucial for the prevention of intestinal-type gastric cancer. The objective of the current study was to ascertain molecular evolution of intestinal-type gastric cancer according to the Correa cascade for the precise gastric cancer screening. We collected sequential lesions of the Correa cascade in the formalin-fixed and paraffin-embedded endoscopic submucosal dissection-resected specimens from 14 Chinese patients by microdissection, and subsequently determined the profiles of somatic aberrations during gastric carcinogenesis using the whole exome sequencing, identifying multiple variants at different Correa stages. The results showed that TP53, PCLO, and PRKDC were the most frequently mutated genes in the early gastric cancer (EGC). A high frequency of TP53 alterations was found in low-grade intraepithelial neoplasia (LGIN), which further increased in high-grade intraepithelial neoplasia (HGIN) and EGC. Intestinal metaplasia (IM) had no significant correlation with EGC in terms of mutational spectra, whereas both LGIN and HGIN showed higher genomic similarities to EGC, compared with IM. Based on Jaccard similarity coefficients, three evolutionary models were further constructed, and most patients showed linear progression from LGIN to HGIN, ultimately resulting in EGC. The ECM-receptor interaction pathway was revealed to be involved in the linear evolution. Additionally, the retrospective validation study of 39 patients diagnosed with LGIN indicated that PRKDC mutations, in addition to TP53 mutations, may drive LGIN progression to HGIN or EGC. In conclusion, the current study unveils the genomic evolution across the Correa cascade of intestinal-type gastric cancer, elucidates the underlying molecular mechanisms of gastric carcinogenesis, and provides some evidence for potential personalized gastric cancer surveillance.
Collapse
Affiliation(s)
- Fangyuan Li
- Digestive Endoscopy Center, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210029, China
| | - Yaohui Wang
- Department of Pathology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210029, China
| | - Xiaochun Ping
- Department of General Surgery, the First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Jiani C Yin
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., Nanjing, Jiangsu 210061, China
| | - Fufeng Wang
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., Nanjing, Jiangsu 210061, China
| | - Xian Zhang
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., Nanjing, Jiangsu 210061, China
| | - Xiang Li
- Department of Surgical Oncology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210029, China
| | - Jing Zhai
- Department of Surgical Oncology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210029, China
| | - Lizong Shen
- Department of General Surgery, the First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- Department of Surgical Oncology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210029, China
| |
Collapse
|
30
|
Gan S, Li C, Hou R, Tian G, Zhao Y, Ren D, Zhou W, Zhao F, Lv K, Yang J. Dynamic changes of the immune microenvironment in the development of gastric cancer caused by inflammation. MOLECULAR THERAPY. ONCOLOGY 2024; 32:200849. [PMID: 39228396 PMCID: PMC11369508 DOI: 10.1016/j.omton.2024.200849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 06/21/2024] [Accepted: 07/16/2024] [Indexed: 09/05/2024]
Abstract
Precancerous lesions typically precede gastric cancer (GC), but the molecular mechanisms underlying the transition from these lesions to GC remain unclear. Therefore, it is urgent to understand this transition from precancerous lesions to GC, which is crucial for the early diagnosis and treatment of GC. In this study, we merged multiple single-cell RNA sequencing datasets to investigate the molecular changes in distinct cell types associated with the progression of GC. First, we observed an increasing abundance of immune cells and a decrease in non-immune cells from non-atrophic gastritis to GC. Five immune cell types were significantly enriched in GC compared to precancerous lesions. Moreover, we found that the interleukin (IL)-17 signaling pathway and Th17 cell differentiation were significantly up-regulated in immune cell subsets during GC progression. Some genes in these processes were predominantly expressed at the GC stage, highlighting their potential as diagnostic markers. Furthermore, we validated our findings using bulk RNA sequencing data from The Cancer Genome Atlas and confirmed consistent immune cell changes during GC progression. Our study provides insights into the immune infiltration and signaling pathways involved in the development of GC, contributing to the development of early diagnosis and targeted treatment strategies for this malignancy.
Collapse
Affiliation(s)
- Siyuan Gan
- School of Mathematical Sciences, Ocean University of China, Qingdao, China
| | - Changfu Li
- Department of Digestive Internal Medicine, Daqing Longnan Hospital, The Fifth Affiliated Hospital of Qiqihar Medical College, Daqing 163000, China
| | - Rui Hou
- Geneis Beijing Co., Ltd, Beijing 100102, China
- Qingdao Geneis Institute of Big Data Mining and Precision Medicine, Qingdao 266000, China
| | - Geng Tian
- Geneis Beijing Co., Ltd, Beijing 100102, China
- Qingdao Geneis Institute of Big Data Mining and Precision Medicine, Qingdao 266000, China
| | - Yuan Zhao
- School of Electrical and Information Engineering, Anhui University of Technology, Ma'anshan, China
| | - Dan Ren
- Department of Pathology, Daqing Longnan Hospital, The Fifth Affiliated Hospital of Qiqihar Medical College, Daqing 163000, China
| | - Wenjing Zhou
- Department of Oncology, Hiser Medical Center of Qingdao, No. 4, Renmin Road, Shibei District, Qingdao, China
| | - Fei Zhao
- School of Mathematical Sciences, Ocean University of China, Qingdao, China
| | - Kebo Lv
- School of Mathematical Sciences, Ocean University of China, Qingdao, China
| | - Jialiang Yang
- Geneis Beijing Co., Ltd, Beijing 100102, China
- Qingdao Geneis Institute of Big Data Mining and Precision Medicine, Qingdao 266000, China
| |
Collapse
|
31
|
Lu H, Xu Z, Shao L, Li P, Xia Y. High infiltration of immune cells with lower immune activity mediated the heterogeneity of gastric adenocarcinoma and promoted metastasis. Heliyon 2024; 10:e37092. [PMID: 39319155 PMCID: PMC11419928 DOI: 10.1016/j.heliyon.2024.e37092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 07/21/2024] [Accepted: 08/27/2024] [Indexed: 09/26/2024] Open
Abstract
Background Gastric adenocarcinoma (GA) is a heterogeneous malignancy with high invasion and metastasis. We aimed to explore the metastatic characteristics of GA using single-cell RNA-sequencing (scRNA-seq) analysis. Methods The scRNA-seq dataset was downloaded from the GEO database and the "Seurat" package was used to perform the scRNA-seq analysis. The CellMarker2.0 database provided gene markers. Subsequently, differentially expressed genes (DEGs) were identified using the FindMarkers function and subjected to enrichment analysis with the "ClusterProlifer". "GseaVis" package was used for visualizing the gene levels. Finally, the SCENIC analysis was performed for identifying key regulons. The expression level and functionality of the key genes were verified by quantitative real-time PCR (qRT-PCR), wound healing and transwell assays. Results A total of 7697 cells were divided into 8 cell subsets, in which the Cytotoxic NK/T cells, Myeloid cells and Myofibroblasts had higher proportion in the metastatic tissues. Further screening of DEGs and enrichment analysis revealed that in the metastatic tissues, NK cells, monocytes and inflammatory fibroblasts with low immune levels contributed to GA metastasis. In addition, this study identified a series of key immune-related regulons that mediated the lower immune activity of immune cells. Further in vitro experiment verified that CXCL8 was a key factor mediating the proliferation and migration of GA cells. Conclusion The scRNA-seq analysis showed that high infiltration of immune cells with lower immune activity mediated heterogeneity to contribute to GA metastasis.
Collapse
Affiliation(s)
- Hongpeng Lu
- Department of Gastroenterology, The First Affiliated Hospital of Ningbo University, Ningbo, 315010, China
| | - Zhihui Xu
- Department of Gastroenterology, Ninghai County Second Hospital, Ningbo, 315600, China
| | - Lihong Shao
- Department of Gastroenterology, The First Affiliated Hospital of Ningbo University, Ningbo, 315010, China
| | - Peifei Li
- Department of Gastroenterology, The First Affiliated Hospital of Ningbo University, Ningbo, 315010, China
| | - Yonghong Xia
- Department of Gastroenterology, Ninghai County Second Hospital, Ningbo, 315600, China
| |
Collapse
|
32
|
Xu J, Yu B, Wang F, Yang J. Single-cell RNA sequencing to map tumor heterogeneity in gastric carcinogenesis paving roads to individualized therapy. Cancer Immunol Immunother 2024; 73:233. [PMID: 39271545 PMCID: PMC11399521 DOI: 10.1007/s00262-024-03820-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 08/27/2024] [Indexed: 09/15/2024]
Abstract
Gastric cancer (GC) is a highly heterogeneous disease with a complex tumor microenvironment (TME) that encompasses multiple cell types including cancer cells, immune cells, stromal cells, and so on. Cancer-associated cells could remodel the TME and influence the progression of GC and therapeutic response. Single-cell RNA sequencing (scRNA-seq), as an emerging technology, has provided unprecedented insights into the complicated biological composition and characteristics of TME at the molecular, cellular, and immunological resolutions, offering a new idea for GC studies. In this review, we discuss the novel findings from scRNA-seq datasets revealing the origin and evolution of GC, and scRNA-seq is a powerful tool for investigating transcriptional dynamics and intratumor heterogeneity (ITH) in GC. Meanwhile, we demonstrate that the vital immune cells within TME, including T cells, B cells, macrophages, and stromal cells, play an important role in the disease progression. Additionally, we also overview that how scRNA-seq facilitates our understanding about the effects on individualized therapy of GC patients. Spatial transcriptomes (ST) have been designed to determine spatial distribution and capture local intercellular communication networks, enabling a further understanding of the relationship between the spatial background of a particular cell and its functions. In summary, scRNA-seq and other single-cell technologies provide a valuable perspective for molecular and pathological disease characteristics and hold promise for advancing basic research and clinical practice in GC.
Collapse
Affiliation(s)
- Jiao Xu
- Precision Medicine Center, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277 West Yanta Road., Xi'an, 710061, Shaanxi, People's Republic of China
| | - Bixin Yu
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277 West Yanta Road., Xi'an, 710061, Shaanxi, People's Republic of China
| | - Fan Wang
- Phase I Clinical Trial Research Center, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, People's Republic of China.
| | - Jin Yang
- Precision Medicine Center, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277 West Yanta Road., Xi'an, 710061, Shaanxi, People's Republic of China.
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277 West Yanta Road., Xi'an, 710061, Shaanxi, People's Republic of China.
- Phase I Clinical Trial Research Center, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, People's Republic of China.
- Cancer Center, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, People's Republic of China.
| |
Collapse
|
33
|
Wang X, Liu Y, Zhao Q, Wang X, Chen X, Hou L, Tian S, Peng ZM, Han XJ, Wang T, Zhang Z, Tou FF, Huang S, Rao J, Chen L, Zheng Z. PILRB potentiates the PI3K/AKT signaling pathway and reprograms cholesterol metabolism to drive gastric tumorigenesis and metastasis. Cell Death Dis 2024; 15:642. [PMID: 39227585 PMCID: PMC11372125 DOI: 10.1038/s41419-024-07026-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 08/09/2024] [Accepted: 08/22/2024] [Indexed: 09/05/2024]
Abstract
Paired immunoglobin-like type 2 receptor beta (PILRB) mainly plays a crucial role in regulating innate immunity, but whether PILRB is involved in cancer is poorly understood. Here, we report that PILRB potentiates the PI3K/AKT pathway to drive gastric tumorigenesis by binding and stabilizing IRS4, which could hyperactivate the PI3K/AKT pathway. Firstly, the levels of PILRB are upregulated in human gastric cancer (GC) specimens and associated with poor prognosis in patients with GC. In addition, our data show that PILRB promotes cell proliferation, colony formation, cell migration and invasion in GC cells in vitro and in vivo. Mechanistically, PILRB recruits the deubiquitination enzymes OTUB1 to IRS4 and relieves K48-linked ubiquitination of IRS4, protecting IRS4 protein from proteasomal-mediated degradation and subsequent activation of the PI3K/AKT pathway. Importantly, the levels of PILRB are positively correlated with IRS4 in GC specimens. Meanwhile, we also found that PILRB reprogrammed cholesterol metabolism by altering ABCA1 and SCARB1 expression levels, and PILRB-expression confers GC cell resistance to statin treatment. Taken together, our findings illustrate that the oncogenic role of PILRB in gastric tumorigenesis, providing new insights into the regulation of PI3K/AKT signaling in GC and establishing PILRB as a biomarker for simvastatin therapy resistance in GC.
Collapse
Affiliation(s)
- Xing Wang
- Centre for Medical Research and Translation, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, 330006, China
| | - Yuanyuan Liu
- Department of Otolaryngology: Head and Neck Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Qiuyan Zhao
- Department of Gastroenterology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xin Wang
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Xinyi Chen
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Chaoyang, China
| | - Li Hou
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Chaoyang, China
| | - Shaodan Tian
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Chaoyang, China
| | - Zi-Mei Peng
- Institute of Clinical Medicine, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, 330006, China
| | - Xiao-Jian Han
- Institute of Geriatrics, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, 330006, PR China
| | - Tao Wang
- Institute of Geriatrics, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, 330006, PR China
| | - Zhen Zhang
- Institute of Clinical Medicine, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, 330006, China
| | - Fang-Fang Tou
- Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, 330006, China
| | - Shan Huang
- Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, 330006, China.
| | - Jun Rao
- Jiangxi Cancer Hospital, The Second Affiliated Hospital of Nanchang Medical College, Jiangxi Clinical Research Center for Cancer, Nanchang, Jiangxi, PR China.
| | - Lixiao Chen
- Department of Otolaryngology: Head and Neck Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China.
| | - Zhi Zheng
- Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, 330006, China.
| |
Collapse
|
34
|
Yoon JH, Byun HJ, Kim SY, Jung DH, Lee SK. Exosomal LINC00853 promotes progression of gastric cancer via the MAP17/PDZK1/AKT signaling pathway. Noncoding RNA Res 2024; 9:876-886. [PMID: 38586313 PMCID: PMC10997811 DOI: 10.1016/j.ncrna.2024.03.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 03/26/2024] [Accepted: 03/26/2024] [Indexed: 04/09/2024] Open
Abstract
Although rare, there is ongoing research into biomarkers that predict the onset and recurrence of gastric cancer, particularly focusing on substances found in exosomes. Long non-coding RNAs (lncRNAs) have garnered attention for their potential in diagnosing gastric cancer. This study investigates the role of lncRNAs in gastric cancer, focusing on their presence in exosomes as potential biomarkers for the disease's onset and recurrence. We utilized the ArrayStar Human LncRNA array 2.0 to analyze lncRNA expression in tissues from early-stage gastric cancer patients. Our analysis highlighted LINC00853, which was significantly upregulated in cancer tissues and implicated in promoting epithelial-mesenchymal transition via the MAP17/PDZK1/AKT pathway. Functional studies on AGS and MKN74 gastric cancer cell lines demonstrated that LINC00853 facilitates cell proliferation, invasion, and migration. Additionally, RNA immunoprecipitation and electrophoretic mobility shift assays confirmed LINC00853 interaction with MAP17. Importantly, LINC00853 was also detected in exosomes from both patient samples and cell lines, and its downregulation led to decreased tumorigenicity in AGS cells. These findings suggest that both cellular and exosomal LINC00853 contribute to gastric cancer pathogenesis and may serve as valuable biomarkers for the disease.
Collapse
Affiliation(s)
| | | | - Seo Yeon Kim
- Department of Internal Medicine, Yonsei Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, South Korea
| | - Da Hyun Jung
- Department of Internal Medicine, Yonsei Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, South Korea
| | - Sang Kil Lee
- Department of Internal Medicine, Yonsei Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, South Korea
| |
Collapse
|
35
|
Chen Y, Wang W. Exploring the Influence of T Cell Marker Gene Expression on the Pathobiology and Clinical Prognostic Outcomes in Intestinal-Type Gastric Carcinoma. J Gastrointest Cancer 2024; 55:1410-1424. [PMID: 39136893 DOI: 10.1007/s12029-024-01104-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/06/2024] [Indexed: 08/27/2024]
Abstract
BACKGROUND Gastric cancer (GC) poses a significant global health challenge. This study is aimed at elucidating the role of the immune system, particularly T cells and their subtypes, in the pathogenesis and progression of intestinal-type gastric carcinoma (GC), and at evaluating the predictive utility of a T cell marker gene-based risk score for overall survival. METHODS We performed an extensive analysis using single-cell RNA sequencing data to map the diversity of immune cells and identify specific T cell marker genes within GC. Pseudotime trajectory analysis was employed to observe the expression patterns of tumor-related pathways and transcription factors (TFs) at various disease stages. We developed a risk score using data from The Cancer Genome Atlas (TCGA) as a training set and validated it with the GSE15459 dataset. RESULTS Our analysis revealed distinct patterns of T cell marker gene expression associated with different stages of GC. The risk score, based on these markers, successfully stratified patients into high-risk and low-risk groups with significantly different overall survival prospects. High-risk patients exhibited poorer survival outcomes compared to low-risk patients (p < 0.05). Additionally, the risk score was capable of identifying patients across a spectrum from chronic atrophic gastritis to early GC. CONCLUSION The findings enhance the understanding of the tumor immune microenvironment in GC and propose new immunotherapeutic targets. The T cell marker gene-based risk score offers a potential tool for gastroenterologists to tailor treatment plans more precisely according to the cancer's severity.
Collapse
Affiliation(s)
- Yixuan Chen
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, 100 Huaihai Avenue, Hefei, 230012, Anhui, China
- School of Life Sciences, Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui, China
- Anhui Public Health Clinical Center, Hefei, 230022, China
| | - Wenbin Wang
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, 100 Huaihai Avenue, Hefei, 230012, Anhui, China.
- Anhui Public Health Clinical Center, Hefei, 230022, China.
| |
Collapse
|
36
|
Wada Y, Nishi M, Yoshikawa K, Takasu C, Tokunaga T, Nakao T, Kashihara H, Yoshimoto T, Shimada M. Circulating Exosomal MicroRNA Signature Predicts Peritoneal Metastasis in Patients with Advanced Gastric Cancer. Ann Surg Oncol 2024; 31:5997-6006. [PMID: 38951411 DOI: 10.1245/s10434-024-15592-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 05/26/2024] [Indexed: 07/03/2024]
Abstract
BACKGROUND Despite a radical operation, about half of gastric cancer (GC) patients with advanced GC experience peritoneal metastasis (PM), and the patients with PM have a poor prognosis. However, because staging laparoscopy was a highly invasive procedure for patients, identification of PM using a liquid biopsy can be useful for patients with GC. METHODS This study analyzed two genome-wide miRNA expression profiling datasets (GSE164174 and TCGA). The study prioritized biomarkers in pretreatment plasma specimens from clinical training and validation cohorts of patients with GC. The authors developed an integrated exosomal miRNA panel and established a risk-stratification model, which was combined with the miRNA panel and currently used tumor markers (CEA, CA19-9, CA125, and CA72-4 levels). RESULTS The comprehensive discovery effort identified a four-miRNA panel that robustly predicted the metastasis with excellent accuracy in the TCGA dataset (area under the curve [AUC] 0.86). A circulating exosomal miRNA panel was established successfully with remarkable diagnostic accuracy in the clinical training (AUC 0.85) and validation (AUC 0.86) cohorts. Moreover, the predictive accuracy of the panel was significantly superior to that of conventional clinical factors (P < 0.01), and the risk-stratification model was dramatically superior to the panel and currently used clinical factors for predicting PM (AUC 0.94; univariate: odds ratio [OR] 77.00 [P < 0.01]; multivariate OR 57.71 [P = 0.01]). CONCLUSIONS The novel risk-stratification model for predicting PM has potential for clinical translation as a liquid biopsy assay for patients with GC. The study findings highlight the potential clinical impact of the model for improved selection and management of patients with GC.
Collapse
Affiliation(s)
- Yuma Wada
- Department of Surgery, Tokushima University, Tokushima, Japan
| | - Masaaki Nishi
- Department of Surgery, Tokushima University, Tokushima, Japan.
| | - Kozo Yoshikawa
- Department of Surgery, Tokushima University, Tokushima, Japan
| | - Chie Takasu
- Department of Surgery, Tokushima University, Tokushima, Japan
| | - Takuya Tokunaga
- Department of Surgery, Tokushima University, Tokushima, Japan
| | - Toshihiro Nakao
- Department of Surgery, Tokushima University, Tokushima, Japan
| | | | | | - Mitsuo Shimada
- Department of Surgery, Tokushima University, Tokushima, Japan
| |
Collapse
|
37
|
Zhao Q, Yu H, Shi M, Wang X, Fan Z, Wang Z. Tumor microenvironment characteristics of lipid metabolism reprogramming related to ferroptosis and EndMT influencing prognosis in gastric cancer. Int Immunopharmacol 2024; 137:112433. [PMID: 38870879 DOI: 10.1016/j.intimp.2024.112433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 06/05/2024] [Accepted: 06/05/2024] [Indexed: 06/15/2024]
Abstract
BACKGROUND Gastric cancer (GC) is a refractory malignant tumor with high tumor heterogeneity, a low rate of early diagnosis, and poor patient prognosis. Lipid metabolism reprogramming plays a critical role in tumorigenesis and progression, but its prognostic role and regulatory mechanism in GC are rarely studied. Thus, the identification of signatures related to lipid metabolism is necessary and may present a new avenue for improving the overall prognosis of GC. METHODS Lipid metabolism-associated genes (LMAGs) with differential expression in tumor and tumor-adjacent tissue were acquired to identify lipid metabolism-associated subtypes. The differentially expressed genes (DEGs) between the two clusters were then utilized for prognostic analysis and signature construction. Additionally, pathway enrichment analysis and immune cell infiltration analysis were employed to identify the characteristics of the prognostic model. Further analyses were conducted at the single-cell level to better understand the model's prognostic mechanism. Finally, the prediction of immunotherapy response was used to suggest potential treatments. RESULTS Two lipid metabolism-associated subtypes were identified and 9 prognosis-related genes from the DEGs between the two clusters were collected for the construction of the prognostic model named lipid metabolism-associated signature (LMAS). Then we found the low LMAS patients with favorable prognoses were more sensitive to ferroptosis in the Cancer Genome Atlas of Stomach Adenocarcinoma (TCGA-STAD). Meanwhile, the tumor cells exhibiting high levels of lipid peroxidation and accumulation of reactive oxygen species (ROS) in single-cell levels were primarily enriched in the low LMAS group, which was more likely to induce ferroptosis. In addition, endothelial cells and cancer-associated fibroblasts (CAFs) facilitated tumor angiogenesis, proliferation, invasion, and metastasis through endothelial-mesenchymal transition (EndMT), affecting the prognosis of the patients with high LMAS scores. Moreover, CD1C- CD141- dendritic cells (DCs) also secreted pro-tumorigenic cytokines to regulate the function of endothelial cells and CAFs. Finally, the patients with low LMAS scores might have better efficacy in immunotherapy. CONCLUSIONS A LMAS was constructed to guide GC prognosis and therapy. Meanwhile, a novel anti-tumor effect was found in lipid metabolism reprogramming of GC which improved patients' prognosis by regulating the sensitivity of tumor cells to ferroptosis. Moreover, EndMT may have a negative impact on GC prognosis.
Collapse
Affiliation(s)
- Qian Zhao
- Inner Mongolia Key Laboratory of Disease-Related Biomarkers, The Second Affiliated Hospital, Baotou Medical College, Baotou 014030, China; School of Basic Medicine, Baotou Medical College, Baotou 014040, China
| | - Hui Yu
- Translational Medicine Center, Baotou Medical College, Baotou 014040, China
| | - Mengqi Shi
- School of Basic Medicine, Baotou Medical College, Baotou 014040, China
| | - Xujie Wang
- School of Basic Medicine, Baotou Medical College, Baotou 014040, China
| | - Zixu Fan
- Inner Mongolia Key Laboratory of Disease-Related Biomarkers, The Second Affiliated Hospital, Baotou Medical College, Baotou 014030, China
| | - Zhanli Wang
- Inner Mongolia Key Laboratory of Disease-Related Biomarkers, The Second Affiliated Hospital, Baotou Medical College, Baotou 014030, China.
| |
Collapse
|
38
|
Marashi A, Hasany S, Moghimi S, Kiani R, Mehran Asl S, Dareghlou YA, Lorestani P, Varmazyar S, Jafari F, Ataeian S, Naghavi K, Sajjadi SM, Haratian N, Alinezhad A, Azhdarimoghaddam A, Sadat Rafiei SK, Anar MA. Targeting gut-microbiota for gastric cancer treatment: a systematic review. Front Med (Lausanne) 2024; 11:1412709. [PMID: 39170038 PMCID: PMC11337614 DOI: 10.3389/fmed.2024.1412709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 07/17/2024] [Indexed: 08/23/2024] Open
Abstract
Background Preclinical research has identified the mechanisms via which bacteria influence cancer treatment outcomes. Clinical studies have demonstrated the potential to modify the microbiome in cancer treatment. Herein, we systematically analyze how gut microorganisms interact with chemotherapy and immune checkpoint inhibitors, specifically focusing on how gut bacteria affect the pharmacokinetics and pharmacodynamics of cancer treatment. Method This study searched Web of Science, Scopus, and PubMed until August 2023. Studies were screened by their title and abstract using the Rayyan intelligent tool for systematic reviews. Quality assessment of studies was done using the JBI critical appraisal tool. Result Alterations in the gut microbiome are associated with gastric cancer and precancerous lesions. These alterations include reduced microbial alpha diversity, increased bacterial overgrowth, and decreased richness and evenness of gastric bacteria. Helicobacter pylori infection is associated with reduced richness and evenness of gastric bacteria, while eradication only partially restores microbial diversity. The gut microbiome also affects the response to cancer treatments, with higher abundances of Lactobacillus associated with better response to anti-PD-1/PD-L1 immunotherapy and more prolonged progression-free survival. Antibiotic-induced gut microbiota dysbiosis can reduce the anti-tumor efficacy of 5-Fluorouracil treatment, while probiotics did not significantly enhance it. A probiotic combination containing Bifidobacterium infantis, Lactobacillus acidophilus, Enterococcus faecalis, and Bacillus cereus can reduce inflammation, enhance immunity, and restore a healthier gut microbial balance in gastric cancer patients after partial gastrectomy. Conclusion Probiotics and targeted interventions to modulate the gut microbiome have shown promising results in cancer prevention and treatment efficacy.Systematic review registration: https://osf.io/6vcjp.
Collapse
Affiliation(s)
- Amir Marashi
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Saina Hasany
- Student Research Committee, Islamic Azad University Tehran Medical Sciences, Tehran, Iran
| | - Sadra Moghimi
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Kiani
- Student Research Committee, Islamic Azad University Tehran Medical Sciences, Mashhad, Iran
| | - Sina Mehran Asl
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Student Research Committee, Islamic Azad University Tehran Medical Sciences, Tehran, Iran
| | | | - Parsa Lorestani
- School of Medicine, Shahroud Azad University of Medical Sciences, Shahroud, Iran
| | - Shirin Varmazyar
- Student Research Committee, School of Medicine, Alborz University of Medical Sciences, Alborz, Iran
| | - Fatemeh Jafari
- School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Shakiba Ataeian
- School of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Kiana Naghavi
- Student Research Committee, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Negar Haratian
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Arman Alinezhad
- Student Research Committee, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | | | - Mahsa Asadi Anar
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
39
|
Wang R, Zhao Y, Zhou L, Lin F, Wan M, Gan A, Wu B, Yan T, Jia Y. Costunolide ameliorates MNNG-induced chronic atrophic gastritis through inhibiting oxidative stress and DNA damage via activation of Nrf2. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 130:155581. [PMID: 38810553 DOI: 10.1016/j.phymed.2024.155581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 01/27/2024] [Accepted: 04/01/2024] [Indexed: 05/31/2024]
Abstract
BACKGROUND Chronic atrophic gastritis (CAG) is a chronic digestive disease. Modern research has revealed substantial evidence indicating that the progression of CAG is closely linked to the occurrence of oxidative stress-induced DNA damage and apoptosis in the gastric mucosa. Additionally, research has indicated that Costunolide (COS), the primary active compound found in Aucklandiae Radix, a traditional herb, exhibits antioxidant properties. Nevertheless, the therapeutic potential of COS in treating CAG and its molecular targets have not yet been determined. PURPOSE The objective of this research was to explore the potential gastric mucosal protective effects and mechanisms of COS against N-Methyl-N´-nitro-N-nitrosoguanidine (MNNG)-induced CAG. METHODS Firstly, the MNNG-induced rat CAG model was established in vivo. Occurrence of CAG was detected through macroscopic examination of the stomachs and H&E staining. Additionally, we assessed oxidative stress, DNA damage, and apoptosis using biochemical detection, Western blot, immunohistochemistry and immunofluorescence. Then, an in vitro model was developed to induce MNNG-induced damage in GES-1 cells, and the occurrence of cell damage was determined by Hoechst 33,342 staining and flow cytometry. Finally, the key targets of COS for the treatment of CAG were identified through molecular docking, cellular thermal shift assay (CETSA), and inhibitor ML385. RESULTS In vivo studies demonstrated that COS promotes the expression of Nrf2 in gastric tissues. This led to an increased expression of SOD, GSH, HO-1, while reducing the production of MDA. Furthermore, COS inhibited DNA damage and apoptosis by suppressing the expression of γH2AX and PARP1 in gastric tissues. In vitro studies showed that COS effectively reversed apoptosis induced by MNNG in GES-1 cells. Additionally, COS interacted with Nrf2 to promote its expression. Furthermore, the expression levels of SOD, GSH, and HO-1 were augmented, while the generation of ROS and MDA was diminished. CONCLUSIONS Our results indicate that COS exhibits therapeutic effects on CAG through the promotion of Nrf2 expression and inhibition of oxidative stress and DNA damage. Therefore, COS has the potential to provide new drugs for the treatment of CAG.
Collapse
Affiliation(s)
- Ruixuan Wang
- School of Functional Food and Wine, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang, Liaoning Province, 110016, China
| | - Youdong Zhao
- School of Functional Food and Wine, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang, Liaoning Province, 110016, China
| | - Lei Zhou
- School of Functional Food and Wine, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang, Liaoning Province, 110016, China
| | - Fei Lin
- School of Functional Food and Wine, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang, Liaoning Province, 110016, China
| | - Meiqi Wan
- School of Functional Food and Wine, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang, Liaoning Province, 110016, China
| | - Anna Gan
- School of Functional Food and Wine, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang, Liaoning Province, 110016, China
| | - Bo Wu
- School of Functional Food and Wine, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang, Liaoning Province, 110016, China
| | - Tingxu Yan
- School of Functional Food and Wine, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang, Liaoning Province, 110016, China.
| | - Ying Jia
- School of Functional Food and Wine, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang, Liaoning Province, 110016, China.
| |
Collapse
|
40
|
Ma S, Xu Y, Qin X, Tao M, Gu X, Shen L, Chen Y, Zheng M, Qin S, Wu G, Ju S. RUNX1, FUS, and ELAVL1-induced circPTPN22 promote gastric cancer cell proliferation, migration, and invasion through miR-6788-5p/PAK1 axis-mediated autophagy. Cell Mol Biol Lett 2024; 29:95. [PMID: 38956466 PMCID: PMC11218243 DOI: 10.1186/s11658-024-00610-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 06/14/2024] [Indexed: 07/04/2024] Open
Abstract
BACKGROUND An increasing number of studies have demonstrated the association of circular RNAs (circRNAs) with the pathological processes of various diseases and their involvement in the onset and progression of multiple cancers. Nevertheless, the functional roles and underlying mechanisms of circRNAs in the autophagy regulation of gastric cancer (GC) have not been fully elucidated. METHODS We used transmission electron microscopy and the mRFP-GFP-LC3 dual fluorescent autophagy indicator to investigate autophagy regulation. The cell counting kit-8 assay, colony formation assay, 5-ethynyl-2'-deoxyuridine incorporation assay, Transwell assay, and Western blot assay were conducted to confirm circPTPN22's influence on GC progression. Dual luciferase reporter assays validated the binding between circPTPN22 and miR-6788-5p, as well as miR-6788-5p and p21-activated kinase-1 (PAK1). Functional rescue experiments assessed whether circPTPN22 modulates PAK1 expression by competitively binding miR-6788-5p, affecting autophagy and other biological processes in GC cells. We investigated the impact of circPTPN22 on in vivo GC tumors using a nude mouse xenograft model. Bioinformatics tools predicted upstream regulatory transcription factors and binding proteins of circPTPN22, while chromatin immunoprecipitation and ribonucleoprotein immunoprecipitation assays confirmed the binding status. RESULTS Upregulation of circPTPN22 in GC has been shown to inhibit autophagy and promote cell proliferation, migration, and invasion. Mechanistically, circPTPN22 directly binds to miR-6788-5p, subsequently regulating the expression of PAK1, which activates protein kinase B (Akt) and extracellular signal-regulated kinase (Erk) phosphorylation. This modulation ultimately affects autophagy levels in GC cells. Additionally, runt-related transcription factor 1 (RUNX1) negatively regulates circPTPN22 expression, while RNA-binding proteins such as FUS (fused in sarcoma) and ELAVL1 (recombinant ELAV-like protein 1) positively regulate its expression. Inhibition of the autophagy pathway can increase FUS expression, further upregulating circPTPN22 in GC cells, thereby exacerbating the progression of GC. CONCLUSION Under the regulation of the transcription factor RUNX1 and RNA-binding proteins FUS and ELAVL1, circPTPN22 activates the phosphorylation of Akt and Erk through the miR-6788-5p/PAK1 axis, thereby modulating autophagy in GC cells. Inhibition of autophagy increases FUS, which in turn upregulates circPTPN22, forming a positive feedback loop that ultimately accelerates the progression of GC.
Collapse
Affiliation(s)
- Shuo Ma
- Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, Xisi Road, NO.20, Nantong, 226001, Jiangsu, China
- Center of Clinical Laboratory Medicine, Zhongda Hospital, Medical School of Southeast University, Nanjing, 210009, Jiangsu, China
- Diagnostics Department, Medical School of Southeast University, Nanjing, 210009, Jiangsu, China
| | - Yanhua Xu
- Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, Xisi Road, NO.20, Nantong, 226001, Jiangsu, China
- Department of Laboratory Medicine, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, 225000, Jiangsu, China
| | - Xinyue Qin
- Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, Xisi Road, NO.20, Nantong, 226001, Jiangsu, China
| | - Mei Tao
- Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, Xisi Road, NO.20, Nantong, 226001, Jiangsu, China
| | - Xinliang Gu
- Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, Xisi Road, NO.20, Nantong, 226001, Jiangsu, China
| | - Lei Shen
- Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, Xisi Road, NO.20, Nantong, 226001, Jiangsu, China
| | - Yinhao Chen
- Department of Integrated Oncology, Center for Integrated Oncology (CIO), University Hospital Bonn, Bonn, Germany
| | - Ming Zheng
- Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, Xisi Road, NO.20, Nantong, 226001, Jiangsu, China
| | - Shiyi Qin
- Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, Xisi Road, NO.20, Nantong, 226001, Jiangsu, China
| | - Guoqiu Wu
- Center of Clinical Laboratory Medicine, Zhongda Hospital, Medical School of Southeast University, Nanjing, 210009, Jiangsu, China.
- Diagnostics Department, Medical School of Southeast University, Nanjing, 210009, Jiangsu, China.
| | - Shaoqing Ju
- Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, Xisi Road, NO.20, Nantong, 226001, Jiangsu, China.
| |
Collapse
|
41
|
Ishizu K, Hayashi T, Ogawa R, Nishino M, Sakon R, Wada T, Otsuki S, Yamagata Y, Katai H, Matsui Y, Yoshikawa T. Characteristics of Metachronous Remnant Gastric Cancer After Proximal Gastrectomy: A Retrospective Analysis. J Gastric Cancer 2024; 24:280-290. [PMID: 38960887 PMCID: PMC11224721 DOI: 10.5230/jgc.2024.24.e21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 04/03/2024] [Accepted: 04/17/2024] [Indexed: 07/05/2024] Open
Abstract
PURPOSE Despite annual endoscopy, patients with metachronous remnant gastric cancer (MRGC) following proximal gastrectomy (PG) are at times ineligible for endoscopic resection (ER). This study aimed to clarify the clinical risk factors for ER inapplicability. MATERIALS AND METHODS We reviewed the records of 203 patients who underwent PG for cT1 gastric cancer between 2006 and 2015. The remnant stomach was categorized as a pseudofornix, corpus, or antrum. RESULTS Thirty-two MRGCs were identified in the 29 patients. Twenty MRGCs were classified as ER (ER group, 62.5%), whereas 12 were not (non-ER group, 37.5%). MRGCs were located in the pseudo-fornix in 1, corpus in 5, and antrum in 14 in the ER group, and in the pseudo-fornix in 6, corpus in 4, and antrum in 2 in the non-ER group (P=0.019). Multivariate analysis revealed that the pseudo-fornix was an independent risk factor for non-ER (P=0.014). In the non-ER group, MRGCs at the pseudo-fornix (n=6) had more frequent undifferentiated-type histology (4/6 vs. 0/6), deeper (≥pT1b2; 6/6 vs. 2/6) and nodal metastasis (3/6 vs. 0/6) than non-pseudo-fornix lesions (n=6). We examined the visibility of the region developing MRGC on an annual follow-up endoscopy one year before MRGC detection. In seven lesions at the pseudofornix, visibility was only secured in two (28.6%) because of food residues. Of the 25 lesions in the non-pseudo-fornix, visibility was secured in 21 lesions (84%; P=0.010). CONCLUSIONS Endoscopic visibility increases the chances of ER applicability. Special preparation is required to ensure the complete clearance of food residues in the pseudo-fornix.
Collapse
Affiliation(s)
- Kenichi Ishizu
- Department of Gastric Surgery, National Cancer Center Hospital, Tokyo, Japan
- Cancer Medicine, Cooperative Graduate School, Jikei University Graduate School of Medicine, Tokyo, Japan
| | - Tsutomu Hayashi
- Department of Gastric Surgery, National Cancer Center Hospital, Tokyo, Japan
| | - Rei Ogawa
- Department of Gastric Surgery, National Cancer Center Hospital, Tokyo, Japan
| | - Masashi Nishino
- Department of Gastric Surgery, National Cancer Center Hospital, Tokyo, Japan
| | - Ryota Sakon
- Department of Gastric Surgery, National Cancer Center Hospital, Tokyo, Japan
| | - Takeyuki Wada
- Department of Gastric Surgery, National Cancer Center Hospital, Tokyo, Japan
| | - Sho Otsuki
- Department of Gastric Surgery, National Cancer Center Hospital, Tokyo, Japan
| | - Yukinori Yamagata
- Department of Gastric Surgery, National Cancer Center Hospital, Tokyo, Japan
| | - Hitoshi Katai
- Department of Gastric Surgery, National Cancer Center Hospital, Tokyo, Japan
- Department of Gastrointestinal Surgery, Tachikawa Hospital, Tokyo, Japan
| | - Yoshiyuki Matsui
- Cancer Medicine, Cooperative Graduate School, Jikei University Graduate School of Medicine, Tokyo, Japan
| | - Takaki Yoshikawa
- Department of Gastric Surgery, National Cancer Center Hospital, Tokyo, Japan.
| |
Collapse
|
42
|
Yen H, Chen P, Huang RY, Jeng J, Lai I. Clinicopathological features and cancer transcriptomic profiling of poorly cohesive gastric carcinoma subtypes. J Pathol Clin Res 2024; 10:e12387. [PMID: 38860888 PMCID: PMC11165978 DOI: 10.1002/2056-4538.12387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 05/16/2024] [Accepted: 05/20/2024] [Indexed: 06/12/2024]
Abstract
Gastric poorly cohesive carcinoma (PCC) manifests with a diffuse pattern and diverse tumor cell morphologies, often indicating a more unfavorable prognosis. Recent consensus has reclassified PCC based on the proportion of signet-ring cells (SRCs) in tumors for research purposes. The two most distinct subtypes, poorly cohesive carcinoma not otherwise specified (PCC-NOS) and signet-ring cell carcinoma (SRCC), are characterized by less than 10% and more than 90% SRCs, respectively. However, research comparing the clinicopathological and transcriptomic differences between these subtypes remains limited. In this study, we conducted a comparative analysis of clinicopathological features in 55 advanced-stage PCCs, consisting of 43 PCC-NOS and 12 SRCC cases. Subsequently, 12 PCC-NOS and 5 SRCC cases were randomly selected for initial cancer-related gene expression profiling and pathway enrichment analysis using the GeoMx digital spatial profiler, followed by validation in a separate validation group comprising 16 PCC-NOS and 6 SRCC cases. These transcriptomic findings were then correlated with tumor morphology and clinicopathological data. PCC-NOS cases exhibited larger tumor size, a higher prevalence of pathological N3 disease, and a worse 1-year progression-free survival rate compared to SRCC cases. Clustering of PCC-NOS and SRCC was successfully achieved using the GeoMx Cancer Transcriptome Atlas. Among all studied genes, only MMP7 showed differential expression, with its overexpression significantly associated with the PCC-NOS subtype, increased perineural invasion, and earlier disease progression. Pathway analysis revealed significantly enriched pathways in PCC-NOS related to vesicle-mediated transport, adaptive immune systems, oncogenic signaling, and extracellular matrix organization, while SRCC displayed significant enrichment in pathways associated with respiratory electron transport and the cell cycle. In conclusion, this study compares and correlates clinicopathological features and transcriptomic data between PCC-NOS and SRCC at advanced stages, employing the latest consensus classification and a novel platform for analysis.
Collapse
Affiliation(s)
- Hung‐Hsuan Yen
- Department of SurgeryNational Taiwan University Hospital Hsin‐Chu BranchHsinchuTaiwan
- Department of SurgeryNational Taiwan University HospitalTaipeiTaiwan
| | - Pin‐Yu Chen
- Graduate Institute of Anatomy and Cell Biology, College of MedicineNational Taiwan UniversityTaipeiTaiwan
| | - Ruby Yun‐Ju Huang
- School of Medicine, College of MedicineNational Taiwan UniversityTaipeiTaiwan
| | - Jung‐Ming Jeng
- Department of PathologyNational Taiwan University HospitalTaipeiTaiwan
| | - I‐Rue Lai
- Department of SurgeryNational Taiwan University HospitalTaipeiTaiwan
- Graduate Institute of Anatomy and Cell Biology, College of MedicineNational Taiwan UniversityTaipeiTaiwan
| |
Collapse
|
43
|
Yasuda T, Wang YA. Gastric cancer immunosuppressive microenvironment heterogeneity: implications for therapy development. Trends Cancer 2024; 10:627-642. [PMID: 38600020 PMCID: PMC11292672 DOI: 10.1016/j.trecan.2024.03.008] [Citation(s) in RCA: 35] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 03/15/2024] [Accepted: 03/18/2024] [Indexed: 04/12/2024]
Abstract
Although immunotherapy has revolutionized solid tumor treatment, durable responses in gastric cancer (GC) remain limited. The heterogeneous tumor microenvironment (TME) facilitates immune evasion, contributing to resistance to conventional and immune therapies. Recent studies have highlighted how specific TME components in GC acquire immune escape capabilities through cancer-specific factors. Understanding the underlying molecular mechanisms and targeting the immunosuppressive TME will enhance immunotherapy efficacy and patient outcomes. This review summarizes recent advances in GC TME research and explores the role of the immune-suppressive system as a context-specific determinant. We also provide insights into potential treatments beyond checkpoint inhibition.
Collapse
Affiliation(s)
- Tadahito Yasuda
- Brown Center for Immunotherapy, Department of Medicine, Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - Y Alan Wang
- Brown Center for Immunotherapy, Department of Medicine, Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| |
Collapse
|
44
|
Sun S, Li L, Xu M, Wei Y, Shi F, Liu S. Epstein-Barr virus positive gastric cancer: the pathological basis of CT findings and radiomics models prediction. Abdom Radiol (NY) 2024; 49:1779-1791. [PMID: 38656367 DOI: 10.1007/s00261-024-04306-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 03/17/2024] [Accepted: 03/20/2024] [Indexed: 04/26/2024]
Abstract
PURPOSE To analyze the clinicopathologic information and CT imaging features of Epstein-Barr virus (EBV)-positive gastric cancer (GC) and establish CT-based radiomics models to predict the EBV status of GC. METHODS This retrospective study included 144 GC cases, including 48 EBV-positive cases. Pathological and immunohistochemical information was collected. CT enlarged LN and morphological characteristics were also assessed. Radiomics models were constructed to predict the EBV status, including decision tree (DT), logistic regression (LR), random forest (RF), and support vector machine (SVM). RESULTS T stage, Lauren classification, histological differentiation, nerve invasion, VEGFR2, E-cadherin, PD-L1, and Ki67 differed significantly between the EBV-positive and -negative groups (p = 0.015, 0.030, 0.006, 0.022, 0.028, 0.030, < 0.001, and < 0.001, respectively). CT enlarged LN and large ulceration differed significantly between the two groups (p = 0.019 and 0.043, respectively). The number of patients in the training and validation cohorts was 100 (with 33 EBV-positive cases) and 44 (with 15 EBV-positive cases). In the training cohort, the radiomics models using DT, LR, RF, and SVM yielded areas under the curve (AUCs) of 0.905, 0.771, 0.836, and 0.886, respectively. In the validation cohort, the diagnostic efficacy of radiomics models using the four classifiers were 0.737, 0.722, 0.751, and 0.713, respectively. CONCLUSION A significantly higher proportion of CT enlarged LN and a significantly lower proportion of large ulceration were found in EBV-positive GC. The prediction efficiency of radiomics models with different classifiers to predict EBV status in GC was good.
Collapse
Affiliation(s)
- Shuangshuang Sun
- Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, No. 321 Zhongshan Road, Nanjing, 210008, Jiangsu, China
| | - Lin Li
- Department of Pathology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Mengying Xu
- Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, No. 321 Zhongshan Road, Nanjing, 210008, Jiangsu, China
| | - Ying Wei
- Department of Research and Development, Shanghai United Imaging Intelligence Co., Ltd, Shanghai, 200000, China
| | - Feng Shi
- Department of Research and Development, Shanghai United Imaging Intelligence Co., Ltd, Shanghai, 200000, China
| | - Song Liu
- Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, No. 321 Zhongshan Road, Nanjing, 210008, Jiangsu, China.
| |
Collapse
|
45
|
Vilarinho T, Pádua D, Pereira B, Mesquita P, Almeida R. MISP Is Overexpressed in Intestinal Metaplasia and Gastric Cancer. Curr Oncol 2024; 31:2769-2779. [PMID: 38785491 PMCID: PMC11120023 DOI: 10.3390/curroncol31050210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 04/29/2024] [Accepted: 05/10/2024] [Indexed: 05/25/2024] Open
Abstract
Gastric cancer is the fifth most common cancer and the fourth cause of global cancer mortality. The identification of new biomarkers and drug targets is crucial to allow the better prognosis and treatment of patients. The mitotic spindle positioning (MISP) protein has the function of correcting mitotic spindle positioning and centrosome clustering and has been implicated in the cytokinesis and migration of cancer cells. The goal of this work was to evaluate the expression and clinical relevance of MISP in gastric cancer. MISP expression was evaluated by immunohistochemistry in a single hospital series (n = 286) of gastric adenocarcinomas and compared with normal gastric mucosa and intestinal metaplasia, a preneoplastic lesion. MISP was detected on the membrane in 83% of the cases, being overexpressed in gastric cancer compared to normal gastric mucosa (n = 10). Its expression was negatively associated with diffuse and poorly cohesive types. On the other hand, it was strongly expressed in intestinal metaplasia where it was associated with MUC2 and CDX2 expression. Furthermore, when we silenced MISP in vitro, a significant decrease in the viability of gastric carcinoma cells was observed. In conclusion, MISP is overexpressed in gastric cancer, being associated with an intestinal phenotype in gastric carcinogenesis and having a role in cellular proliferation.
Collapse
Affiliation(s)
- Tomás Vilarinho
- i3S—Institute for Research and Innovation in Health, University of Porto, 4200-135 Porto, Portugal; (T.V.); (D.P.); (B.P.); (P.M.)
| | - Diana Pádua
- i3S—Institute for Research and Innovation in Health, University of Porto, 4200-135 Porto, Portugal; (T.V.); (D.P.); (B.P.); (P.M.)
- IPATIMUP—Institute of Molecular Pathology and Immunology, University of Porto, 4200-465 Porto, Portugal
| | - Bruno Pereira
- i3S—Institute for Research and Innovation in Health, University of Porto, 4200-135 Porto, Portugal; (T.V.); (D.P.); (B.P.); (P.M.)
- IPATIMUP—Institute of Molecular Pathology and Immunology, University of Porto, 4200-465 Porto, Portugal
| | - Patrícia Mesquita
- i3S—Institute for Research and Innovation in Health, University of Porto, 4200-135 Porto, Portugal; (T.V.); (D.P.); (B.P.); (P.M.)
- IPATIMUP—Institute of Molecular Pathology and Immunology, University of Porto, 4200-465 Porto, Portugal
| | - Raquel Almeida
- i3S—Institute for Research and Innovation in Health, University of Porto, 4200-135 Porto, Portugal; (T.V.); (D.P.); (B.P.); (P.M.)
- IPATIMUP—Institute of Molecular Pathology and Immunology, University of Porto, 4200-465 Porto, Portugal
- Biology Department, Faculty of Sciences, University of Porto, 4169-007 Porto, Portugal
| |
Collapse
|
46
|
Zhao Y, Liu Z, Deng K, Qu H, Zhang Q, Zhou P, Yang M, Yang X, Wang H, Li R, Xia J. Identification of TAP1 as a T-cell related therapeutic target in gastric cancer by mediating oxalipliatin-related synergistic enhancement of immunotherapy. Int Immunopharmacol 2024; 132:111998. [PMID: 38593510 DOI: 10.1016/j.intimp.2024.111998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Revised: 11/30/2023] [Accepted: 03/31/2024] [Indexed: 04/11/2024]
Abstract
BACKGROUND Given the intricate molecular complexities and heterogeneity inherent in T-cell immunotherapy of gastric cancer (GC), elucidative T-cell-related biomarkers were imperative needed for facilitating the prediction of GC patient prognosis and identify potential synergistic therapeutic targets. METHODS We conducted COX regression analysis in TISIDB, TCGA-STAD, and GEO databases to identify 19 GC T-cell-mediated sensitivity tumor killing (TTK) genes (key GCTTKs). Based on key GCTTKs, we constructed two TTK patterns and analyzed their metabolic pathways, mutation features, clinical data distribution, immune cell infiltration, and prognosis. LASSO regression was performed to develop a T-cell-mediated GC Prognosis (TGCP) model. We validated the TGCP model in GC patients. TAP1 was further selected for investigation of its biological functions and molecular mechanisms. We assessed the potential of TAP1 as a promising therapeutic target for GC using Patient-derived organoids (PDOs)-derived xenografts (PDOXs) models of GC. RESULTS The TTK patterns display notable disparities. The TGCP model showcases its proficiency in predicting immune response efficacy, effectively distinguishes immunotherapy difference GC patients. Our findings find further confirmation in PDOX models, affirming TAP1 can enhance immunotherapy facilitated by PDL1 inhibitors. Furthermore, Oxaliplatin, by promoting TAP1 expression, augments PDL1 expression, thereby enhancing the efficacy of immunotherapy. CONCLUSIONS We constructed a TGCP model, which demonstrates satisfactory predictive accuracy. Out of 9 prognostic genes, TAP1 was validated as a synergistic target for Oxaliplatin and PDL1 inhibitors, offering a genetic-level explanation for the synergy observed in GC treatment involving Oxaliplatin in combination with PDL1 inhibitors.
Collapse
Affiliation(s)
- Yupeng Zhao
- Department of General Surgery, The Affiliated Wuxi No. 2 People's Hospital of Nanjing Medical University, Wuxi, PR China; Department of General Surgery, Jiangnan University Medical Center, Wuxi, PR China
| | - Ziyuan Liu
- Department of General Surgery, The Affiliated Wuxi No. 2 People's Hospital of Nanjing Medical University, Wuxi, PR China; Department of General Surgery, Jiangnan University Medical Center, Wuxi, PR China
| | - Kaiyuan Deng
- Department of General Surgery, The Affiliated Wuxi No. 2 People's Hospital of Nanjing Medical University, Wuxi, PR China; Department of General Surgery, Jiangnan University Medical Center, Wuxi, PR China
| | - Huiheng Qu
- Department of General Surgery, The Affiliated Wuxi No. 2 People's Hospital of Nanjing Medical University, Wuxi, PR China; Department of General Surgery, Jiangnan University Medical Center, Wuxi, PR China
| | - Qing Zhang
- Affiliated WuXi Clinical College of Nantong University, Wuxi, PR China
| | - Peng Zhou
- Department of General Surgery, The Affiliated Wuxi No. 2 People's Hospital of Nanjing Medical University, Wuxi, PR China; Department of General Surgery, Jiangnan University Medical Center, Wuxi, PR China
| | - Mengqi Yang
- Department of General Surgery, The Affiliated Wuxi No. 2 People's Hospital of Nanjing Medical University, Wuxi, PR China
| | - Xiao Yang
- Department of General Surgery, Jiangnan University Medical Center, Wuxi, PR China
| | - Hao Wang
- Department of General Surgery, The Affiliated Wuxi No. 2 People's Hospital of Nanjing Medical University, Wuxi, PR China; Department of General Surgery, Jiangnan University Medical Center, Wuxi, PR China
| | - Ranran Li
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China.
| | - Jiazeng Xia
- Department of General Surgery, The Affiliated Wuxi No. 2 People's Hospital of Nanjing Medical University, Wuxi, PR China; Department of General Surgery, Jiangnan University Medical Center, Wuxi, PR China; Affiliated WuXi Clinical College of Nantong University, Wuxi, PR China.
| |
Collapse
|
47
|
Echeverría-Garcés G, Ramos-Medina MJ, Vargas R, Cabrera-Andrade A, Altamirano-Colina A, Freire MP, Montalvo-Guerrero J, Rivera-Orellana S, Echeverría-Espinoza P, Quiñones LA, López-Cortés A. Gastric cancer actionable genomic alterations across diverse populations worldwide and pharmacogenomics strategies based on precision oncology. Front Pharmacol 2024; 15:1373007. [PMID: 38756376 PMCID: PMC11096557 DOI: 10.3389/fphar.2024.1373007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 04/10/2024] [Indexed: 05/18/2024] Open
Abstract
Introduction: Gastric cancer is one of the most prevalent types of cancer worldwide. The World Health Organization (WHO), the International Agency for Research on Cancer (IARC), and the Global Cancer Statistics (GLOBOCAN) reported an age standardized global incidence rate of 9.2 per 100,000 individuals for gastric cancer in 2022, with a mortality rate of 6.1. Despite considerable progress in precision oncology through the efforts of international consortia, understanding the genomic features and their influence on the effectiveness of anti-cancer treatments across diverse ethnic groups remains essential. Methods: Our study aimed to address this need by conducting integrated in silico analyses to identify actionable genomic alterations in gastric cancer driver genes, assess their impact using deleteriousness scores, and determine allele frequencies across nine global populations: European Finnish, European non-Finnish, Latino, East Asian, South Asian, African, Middle Eastern, Ashkenazi Jewish, and Amish. Furthermore, our goal was to prioritize targeted therapeutic strategies based on pharmacogenomics clinical guidelines, in silico drug prescriptions, and clinical trial data. Results: Our comprehensive analysis examined 275,634 variants within 60 gastric cancer driver genes from 730,947 exome sequences and 76,215 whole-genome sequences from unrelated individuals, identifying 13,542 annotated and predicted oncogenic variants. We prioritized the most prevalent and deleterious oncogenic variants for subsequent pharmacogenomics testing. Additionally, we discovered actionable genomic alterations in the ARID1A, ATM, BCOR, ERBB2, ERBB3, CDKN2A, KIT, PIK3CA, PTEN, NTRK3, TP53, and CDKN2A genes that could enhance the efficacy of anti-cancer therapies, as suggested by in silico drug prescription analyses, reviews of current pharmacogenomics clinical guidelines, and evaluations of phase III and IV clinical trials targeting gastric cancer driver proteins. Discussion: These findings underline the urgency of consolidating efforts to devise effective prevention measures, invest in genomic profiling for underrepresented populations, and ensure the inclusion of ethnic minorities in future clinical trials and cancer research in developed countries.
Collapse
Affiliation(s)
- Gabriela Echeverría-Garcés
- Centro de Referencia Nacional de Genómica, Secuenciación y Bioinformática, Instituto Nacional de Investigación en Salud Pública “Leopoldo Izquieta Pérez”, Quito, Ecuador
- Latin American Network for the Implementation and Validation of Clinical Pharmacogenomics Guidelines (RELIVAF-CYTED), Santiago, Chile
| | - María José Ramos-Medina
- German Cancer Research Center (DKFZ), Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Rodrigo Vargas
- Latin American Network for the Implementation and Validation of Clinical Pharmacogenomics Guidelines (RELIVAF-CYTED), Santiago, Chile
- Department of Molecular Biology, Galileo University, Guatemala City, Guatemala
| | - Alejandro Cabrera-Andrade
- Escuela de Enfermería, Facultad de Ciencias de La Salud, Universidad de Las Américas, Quito, Ecuador
- Grupo de Bio-Quimioinformática, Universidad de Las Américas, Quito, Ecuador
| | | | - María Paula Freire
- Cancer Research Group (CRG), Faculty of Medicine, Universidad de Las Américas, Quito, Ecuador
| | | | | | | | - Luis A. Quiñones
- Latin American Network for the Implementation and Validation of Clinical Pharmacogenomics Guidelines (RELIVAF-CYTED), Santiago, Chile
- Laboratory of Chemical Carcinogenesis and Pharmacogenetics, Department of Basic-Clinical Oncology (DOBC), Faculty of Medicine, University of Chile, Santiago, Chile
- Department of Pharmaceutical Sciences and Technology, Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santiago, Chile
| | - Andrés López-Cortés
- Cancer Research Group (CRG), Faculty of Medicine, Universidad de Las Américas, Quito, Ecuador
| |
Collapse
|
48
|
Di Giorgio C, Morretta E, Lupia A, Bellini R, Massa C, Urbani G, Bordoni M, Marchianò S, Lachi G, Rapacciuolo P, Finamore C, Sepe V, Chiara Monti M, Moraca F, Natalizi N, Graziosi L, Distrutti E, Biagioli M, Catalanotti B, Donini A, Zampella A, Fiorucci S. Bile acids serve as endogenous antagonists of the Leukemia inhibitory factor (LIF) receptor in oncogenesis. Biochem Pharmacol 2024; 223:116134. [PMID: 38494064 DOI: 10.1016/j.bcp.2024.116134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 03/11/2024] [Accepted: 03/14/2024] [Indexed: 03/19/2024]
Abstract
The leukemia inhibitory factor (LIF) is member of interleukin (IL)-6 family of cytokines involved immune regulation, morphogenesis and oncogenesis. In cancer tissues, LIF binds a heterodimeric receptor (LIFR), formed by a LIFRβ subunit and glycoprotein(gp)130, promoting epithelial mesenchymal transition and cell growth. Bile acids are cholesterol metabolites generated at the interface of host metabolism and the intestinal microbiota. Here we demonstrated that bile acids serve as endogenous antagonist to LIFR in oncogenesis. The tissue characterization of bile acids content in non-cancer and cancer biopsy pairs from gastric adenocarcinomas (GC) demonstrated that bile acids accumulate within cancer tissues, with glyco-deoxycholic acid (GDCA) functioning as negative regulator of LIFR expression. In patient-derived organoids (hPDOs) from GC patients, GDCA reverses LIF-induced stemness and proliferation. In summary, we have identified the secondary bile acids as the first endogenous antagonist to LIFR supporting a development of bile acid-based therapies in LIF-mediated oncogenesis.
Collapse
Affiliation(s)
| | - Elva Morretta
- University of Salerno, Department of Pharmacy, Salerno, Italy
| | - Antonio Lupia
- University of Cagliari, Department of Life and Environmental Sciences, Cagliari, Italy; Net4Science srl, University "Magna Græcia", Campus Salvatore Venuta, Viale Europa, Catanzaro 88100, Italy
| | - Rachele Bellini
- University of Perugia, Department of Medicine and Surgery, Perugia, Italy
| | - Carmen Massa
- University of Perugia, Department of Medicine and Surgery, Perugia, Italy
| | - Ginevra Urbani
- University of Perugia, Department of Medicine and Surgery, Perugia, Italy
| | - Martina Bordoni
- University of Perugia, Department of Medicine and Surgery, Perugia, Italy
| | - Silvia Marchianò
- University of Perugia, Department of Medicine and Surgery, Perugia, Italy
| | - Ginevra Lachi
- University of Perugia, Department of Medicine and Surgery, Perugia, Italy
| | | | - Claudia Finamore
- University of Naples Federico II, Department of Pharmacy, Naples, Italy
| | - Valentina Sepe
- University of Naples Federico II, Department of Pharmacy, Naples, Italy
| | | | - Federica Moraca
- Net4Science srl, University "Magna Græcia", Campus Salvatore Venuta, Viale Europa, Catanzaro 88100, Italy; University of Naples Federico II, Department of Pharmacy, Naples, Italy
| | | | | | | | - Michele Biagioli
- University of Perugia, Department of Medicine and Surgery, Perugia, Italy
| | - Bruno Catalanotti
- University of Naples Federico II, Department of Pharmacy, Naples, Italy
| | - Annibale Donini
- University of Perugia, Department of Medicine and Surgery, Perugia, Italy
| | - Angela Zampella
- University of Naples Federico II, Department of Pharmacy, Naples, Italy
| | - Stefano Fiorucci
- University of Perugia, Department of Medicine and Surgery, Perugia, Italy.
| |
Collapse
|
49
|
Won Y, Jang B, Lee SH, Reyzer ML, Presentation KS, Kim H, Caldwell B, Zhang C, Lee HS, Lee C, Trinh VQ, Tan MCB, Kim K, Caprioli RM, Choi E. Oncogenic Fatty Acid Metabolism Rewires Energy Supply Chain in Gastric Carcinogenesis. Gastroenterology 2024; 166:772-786.e14. [PMID: 38272100 PMCID: PMC11040571 DOI: 10.1053/j.gastro.2024.01.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 01/12/2024] [Accepted: 01/16/2024] [Indexed: 01/27/2024]
Abstract
BACKGROUND & AIMS Gastric carcinogenesis develops within a sequential carcinogenic cascade from precancerous metaplasia to dysplasia and adenocarcinoma, and oncogenic gene activation can drive the process. Metabolic reprogramming is considered a key mechanism for cancer cell growth and proliferation. However, how metabolic changes contribute to the progression of metaplasia to dysplasia remains unclear. We have examined metabolic dynamics during gastric carcinogenesis using a novel mouse model that induces Kras activation in zymogen-secreting chief cells. METHODS We generated a Gif-rtTA;TetO-Cre;KrasG12D (GCK) mouse model that continuously induces active Kras expression in chief cells after doxycycline treatment. Histologic examination and imaging mass spectrometry were performed in the GCK mouse stomachs at 2 to 14 weeks after doxycycline treatment. Mouse and human gastric organoids were used for metabolic enzyme inhibitor treatment. The GCK mice were treated with a stearoyl- coenzyme A desaturase (SCD) inhibitor to inhibit the fatty acid desaturation. Tissue microarrays were used to assess the SCD expression in human gastrointestinal cancers. RESULTS The GCK mice developed metaplasia and high-grade dysplasia within 4 months. Metabolic reprogramming from glycolysis to fatty acid metabolism occurred during metaplasia progression to dysplasia. Altered fatty acid desaturation through SCD produces a novel eicosenoic acid, which fuels dysplastic cell hyperproliferation and survival. The SCD inhibitor killed both mouse and human dysplastic organoids and selectively targeted dysplastic cells in vivo. SCD was up-regulated during carcinogenesis in human gastrointestinal cancers. CONCLUSIONS Active Kras expression only in gastric chief cells drives the full spectrum of gastric carcinogenesis. Also, oncogenic metabolic rewiring is an essential adaptation for high-energy demand in dysplastic cells.
Collapse
Affiliation(s)
- Yoonkyung Won
- Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee; Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Bogun Jang
- Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee; Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, Tennessee; Department of Pathology, Jeju National University College of Medicine and Jeju National University Hospital, Jeju, Republic of Korea
| | - Su-Hyung Lee
- Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee; Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Michelle L Reyzer
- Mass Spectrometry Research Center, Vanderbilt University, Nashville, Tennessee
| | - Kimberly S Presentation
- Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee; Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Hyesung Kim
- Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee; Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, Tennessee; Department of Pathology, Jeju National University College of Medicine, Jeju, Republic of Korea
| | - Brianna Caldwell
- Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee; Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Changqing Zhang
- Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee; Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Hye Seung Lee
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea; Department of Pathology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Cheol Lee
- Department of Pathology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Vincent Q Trinh
- The Digital Histology and Advanced Pathology Research, The Institute for Research in Immunology and Cancer (IRIC) of the Université de Montréal, Montréal, Québec, Canada
| | - Marcus C B Tan
- Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee; Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, Tennessee; Vanderbilt Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Kwangho Kim
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee; Department of Chemistry, Vanderbilt University, Nashville, Tennessee
| | - Richard M Caprioli
- Mass Spectrometry Research Center, Vanderbilt University, Nashville, Tennessee
| | - Eunyoung Choi
- Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee; Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, Tennessee; Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee.
| |
Collapse
|
50
|
Wang Q, Zhang Q, Zhu J, Li L, Zeng R, Ding H, Li Z, Feng T, Hao R, Zhang G. Nomogram for predicting overall survival after curative gastrectomy using inflammatory, nutritional and pathological factors. Clin Transl Oncol 2024; 26:1001-1011. [PMID: 37996667 DOI: 10.1007/s12094-023-03340-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 10/20/2023] [Indexed: 11/25/2023]
Abstract
PURPOSE To establish a nomogram for predicting the overall survival (OS) in patients with gastric cancer (GC) based on inflammatory, nutritional and pathological factors. METHODS GC patients underwent curative gastrectomy from January 2012 to June 2017 in our hospital were included, and were classified into training set and validation set with a ratio of 7:3. Then variables associated with OS were analyzed using univariate and multivariate Cox regression analysis. Nomograms predicting OS were built using variables from multivariable Cox models. Finally, Kaplan-Meier curve and Log-rank test were also conducted to analyze the 1-yr, 3-yr and 5-yr OS to validate the efficiency of risk stratification of the nomogram. RESULTS A total of 366 GC patients were included. After univariate and multivariate Cox regression analysis, age (HR = 1.52, 95% CI = 1.01-2.30, P = 0.044), CA50 (HR = 1.90, 95% CI = 1.12-3.21, P = 0.017), PNI (HR = 1.65, 95% CI = 1.13-2.39, P = 0.009), SII (HR = 1.46, 95% CI = 1.03-2.08, P = 0.036), T stage (HR = 2.26, 95% CI = 1.01-5.05, P = 0.048; HR = 7.24, 95% CI = 3.64-14.40, P < 0.001) were independent influencing factors on the survival time of GC patients. Five factors including CEA, prognostic nutritional index (PNI), systemic immune-inflammation index (SII), ln (tumor size), T stage, and N stage were identified and entered the nomogram, which showed good discrimination and calibration in both sets. On internal validation, 1-yr, 3-yr and 5-yr nomogram demonstrated a good discrimination with an area under the ROC curve (AUC) of 0.77, 0.84 and 0.86, respectively. The AUC for 1-yr, 3-yr and 5-yr nomogram in validation set was 0.77, 0.79 and 0.81, respectively. The OS in low risk group of training cohort and validation cohort was significantly higher than that of intermediate risk group and high risk group, respectively. CONCLUSIONS We established a nomogram based on PNI, SII and pathological factors for predicting OS in GC patients. In addition, its efficiency was validated by validation set and stratified analysis.
Collapse
Affiliation(s)
- Qi Wang
- Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, 250100, China
| | - Qiang Zhang
- Department of General Surgery, Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao, 266000, China
| | - Jiankang Zhu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Shandong First Medical University, No. 16766 Jingshi Road, Jinan, 250100, China
| | - Linchuan Li
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Shandong First Medical University, No. 16766 Jingshi Road, Jinan, 250100, China
| | - Runzhi Zeng
- Department of General Surgery, Shandong Provincial Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250014, China
| | - Huanxin Ding
- Department of General Surgery, Shandong Provincial Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250014, China
| | - Zhenmin Li
- Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, 250100, China
| | - Tianyi Feng
- Department of General Surgery, Shandong Provincial Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250014, China
| | - Ruiqi Hao
- Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, 250100, China
| | - Guangyong Zhang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Shandong First Medical University, No. 16766 Jingshi Road, Jinan, 250100, China.
| |
Collapse
|