1
|
Liu SS, Wang JK, Liu MS, Guo DF, Wen Q, Liang YH, Wang T, Zhang KH. ILF2 protein is a promising serum biomarker for early detection of gastric cancer. BMC Cancer 2024; 24:1447. [PMID: 39587551 PMCID: PMC11587746 DOI: 10.1186/s12885-024-13205-6] [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: 08/26/2024] [Accepted: 11/14/2024] [Indexed: 11/27/2024] Open
Abstract
BACKGROUND Our previous small-sample study indicated that serum levels of interleukin enhancer binding factor 2 (ILF2) may have the potential for gastric cancer (GC) detection. The present study was conducted to further validate the diagnostic value of serum ILF2 protein for GC. METHODS Serum specimens and clinical data were collected from patients with GC (n = 99) or benign gastric disease (BGD) (n = 49) and healthy controls (HC) (n = 51). Serum ILF2 levels were measured using enzyme-linked immunosorbent assay. The diagnostic performance of ILF2 was evaluated using the area under the receiver operating characteristic curve (AUC). The independence and synergy of ILF2 in GC diagnosis were analyzed by modeling with conventional blood indicators. RESULTS The median serum ILF2 level was higher in the GC group (227.8ng/mL) than in the BGD group (72.0ng/mL) and the HC group (56.8ng/mL) (p < 0.001), and no significant difference across GC subgroups. The AUCs of ILF2 were 0.915 (95%CI 0.873-0.957) for GC vs. HC, 0.854 (95%CI 0.793-0.915) for GC vs. BGD, 0.885 (95%CI 0.841-0.929) for GC vs. BGD + HC, and 0.888 (95% CI 0.830-0.945) for TNM I stage GC vs. BGD + HC, outperforming conventional blood indicators (corresponding AUCs ranging from 0.641 to 0.782). ILF2 was independent of and synergistic with conventional blood indicators in GC diagnosis, and a simple diagnostic model based on ILF2 and red blood cell count improved the diagnostic performance, with positive rates of approximately 90% in various subgroups of GC. CONCLUSIONS Serum ILF2 protein is a novel and potential serum biomarker for the detection of GC, especially for early GC.
Collapse
Affiliation(s)
- Shao-Song Liu
- Department of Gastroenterology, Jiangxi Provincial Key Laboratory of Digestive Diseases, Jiangxi Clinical Research Center for Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No 17, Yongwai Zheng Street, Nanchang, Jiangxi, 330006, China
| | - Jin-Ke Wang
- Department of Gastroenterology, Jiangxi Provincial Key Laboratory of Digestive Diseases, Jiangxi Clinical Research Center for Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No 17, Yongwai Zheng Street, Nanchang, Jiangxi, 330006, China
| | - Mao-Sheng Liu
- Department of Gastroenterology, Jiangxi Provincial Key Laboratory of Digestive Diseases, Jiangxi Clinical Research Center for Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No 17, Yongwai Zheng Street, Nanchang, Jiangxi, 330006, China
| | - Ding-Fan Guo
- Department of Gastroenterology, Jiangxi Provincial Key Laboratory of Digestive Diseases, Jiangxi Clinical Research Center for Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No 17, Yongwai Zheng Street, Nanchang, Jiangxi, 330006, China
| | - Qi Wen
- Department of Gastroenterology, Jiangxi Provincial Key Laboratory of Digestive Diseases, Jiangxi Clinical Research Center for Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No 17, Yongwai Zheng Street, Nanchang, Jiangxi, 330006, China
| | - Yun-Hui Liang
- Department of Gastroenterology, Jiangxi Provincial Key Laboratory of Digestive Diseases, Jiangxi Clinical Research Center for Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No 17, Yongwai Zheng Street, Nanchang, Jiangxi, 330006, China
| | - Ting Wang
- Department of Gastroenterology, Jiangxi Provincial Key Laboratory of Digestive Diseases, Jiangxi Clinical Research Center for Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No 17, Yongwai Zheng Street, Nanchang, Jiangxi, 330006, China.
| | - Kun-He Zhang
- Department of Gastroenterology, Jiangxi Provincial Key Laboratory of Digestive Diseases, Jiangxi Clinical Research Center for Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No 17, Yongwai Zheng Street, Nanchang, Jiangxi, 330006, China.
| |
Collapse
|
2
|
Tang W, Ma X. Application of large-scale and multicohort plasma proteomics data to discover novel causal proteins in gastric cancer. Discov Oncol 2024; 15:570. [PMID: 39422802 PMCID: PMC11489397 DOI: 10.1007/s12672-024-01460-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 10/14/2024] [Indexed: 10/19/2024] Open
Abstract
PURPOSES Gastric cancer (GC) is one of the most common malignant tumors threatening human beings and has a poor prognosis. Therefore, exploring unveiled biomarkers or therapeutic targets for the diagnosis and treatment of GC is crucial. METHODS A total of 5772 protein quantitative trait loci (pQTL) were aggregated from four latest large-scale proteomics cohorts. Two-sample Mendelian randomization (two-sample MR) was utilized to identify the causal effect of blood plasma proteins on GC. Heterogeneity, pleiotropy, and directionality analyses were employed to evaluate proteins identified via two-sample MR. The robustness of results was further validated via colocalization. The drug targets of proteins were evaluated to reveal the compounds that can interfere with these proteins. RESULTS Ten proteins with potential causations in relation to GC were identified: LY6D, SLURP1, MLN, PSCA, THSD1, CFTR, PPM1B, KDM3A, TSC1, and HCG22. Among these proteins, LY6D, SLURP1, and THSD1 were considered as the most reliable biomarkers of GC due to their significant H4 posterior probabilities in colocalization analysis and absence of pleiotropy. Compound 35, nitrosamide, and 0175029-0000 were potential drugs or small molecules targeting LY6D, SLURP1, and THSD1, respectively. CONCLUSION This study identified several plasma proteins as potential biomarkers of GC and provided data support and new insights into the early diagnosis, intervention, and therapeutic targets of GC.
Collapse
Affiliation(s)
- Weihao Tang
- College of Liberal Arts and Sciences, University of Florida, Gainesville, USA
| | - Xiaoke Ma
- School of Computer Science and Technology, Xidian University, Xi'an, China.
| |
Collapse
|
3
|
Millapán T, Gutiérrez Á, Rosas K, Buchegger K, Ili CG, Brebi P. In Silico Insights Reveal Fibronectin 1 as a Theranostic Marker in Gastric Cancer. Int J Mol Sci 2024; 25:11113. [PMID: 39456895 PMCID: PMC11507984 DOI: 10.3390/ijms252011113] [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: 09/28/2024] [Revised: 10/09/2024] [Accepted: 10/10/2024] [Indexed: 10/28/2024] Open
Abstract
Gastric cancer (GC) is a complex and highly variable disease, ranking among the top five cancers diagnosed globally, and a leading cause of cancer-related deaths. Emerging from stomach lining cells amid chronic inflammation, it often advances to preneoplastic stages. Late-stage diagnoses and treatment challenges highlight the critical need for early detection and innovative biomarkers, motivating this study's focus on identifying theranostic markers through gene ontology analysis. By exploring deregulated biological processes, this study aims to uncover insights into cancer progression and associated markers, potentially identifying novel theranostic candidates in GC. Using public data from The Human Protein Atlas, this study pinpointed 299 prognostic genes, delineating 171 with unfavorable prognosis and 128 with favorable prognosis. Functional enrichment and protein-protein interaction analyses, supported by RNAseq results and conducted via Metascape and Cytoscape, highlighted five genes (vWF, FN1, THBS1, PCDH7, and F5) with promising theranostic potential. Notably, FN1 and THBS1 exhibited significant promise, with FN1 showing a 370% expression increase in cancerous tissue, and it is possible that FN1 can also indicate the stratification status in GC. While further validation is essential, these findings provide new insights into molecular alterations in GC and potential avenues for clinical application of theranostic markers.
Collapse
Affiliation(s)
- Tatiana Millapán
- Laboratory of Integrative Biology (LIBi), Centro de Excelencia en Medicina Traslacional (CEMT), Scientific and Technological Bioresource Nucleus (BIOREN), Universidad de La Frontera, Temuco 4810296, Chile; (T.M.)
- Millennium Institute on Immunology and Immunotherapy, Santiago 8331150, Chile
| | - Álvaro Gutiérrez
- Laboratory of Integrative Biology (LIBi), Centro de Excelencia en Medicina Traslacional (CEMT), Scientific and Technological Bioresource Nucleus (BIOREN), Universidad de La Frontera, Temuco 4810296, Chile; (T.M.)
- Millennium Institute on Immunology and Immunotherapy, Santiago 8331150, Chile
- Doctoral Program in Sciences with a Specialization in Applied Cellular and Molecular Biology, Universidad de La Frontera, Temuco 4810296, Chile
| | - Krisnna Rosas
- Laboratory of Integrative Biology (LIBi), Centro de Excelencia en Medicina Traslacional (CEMT), Scientific and Technological Bioresource Nucleus (BIOREN), Universidad de La Frontera, Temuco 4810296, Chile; (T.M.)
- Biotechnology Engineering Program, Universidad de La Frontera, Temuco 4810296, Chile
| | - Kurt Buchegger
- Laboratory of Integrative Biology (LIBi), Centro de Excelencia en Medicina Traslacional (CEMT), Scientific and Technological Bioresource Nucleus (BIOREN), Universidad de La Frontera, Temuco 4810296, Chile; (T.M.)
- Millennium Institute on Immunology and Immunotherapy, Santiago 8331150, Chile
- BMRC, Biomedical Research Consortium, Santiago 8331150, Chile
- Department of Basic Sciences, Faculty of Medicine, Universidad de La Frontera, Temuco 4810296, Chile
| | - Carmen Gloria Ili
- Laboratory of Integrative Biology (LIBi), Centro de Excelencia en Medicina Traslacional (CEMT), Scientific and Technological Bioresource Nucleus (BIOREN), Universidad de La Frontera, Temuco 4810296, Chile; (T.M.)
- Millennium Institute on Immunology and Immunotherapy, Santiago 8331150, Chile
- BMRC, Biomedical Research Consortium, Santiago 8331150, Chile
| | - Priscilla Brebi
- Laboratory of Integrative Biology (LIBi), Centro de Excelencia en Medicina Traslacional (CEMT), Scientific and Technological Bioresource Nucleus (BIOREN), Universidad de La Frontera, Temuco 4810296, Chile; (T.M.)
- Millennium Institute on Immunology and Immunotherapy, Santiago 8331150, Chile
- BMRC, Biomedical Research Consortium, Santiago 8331150, Chile
| |
Collapse
|
4
|
Tong X, Du J, Jiang Q, Wu Q, Zhao S, Chen S. Lenvatinib acts on platelet‑derived growth factor receptor β to suppress the malignant behaviors of gastric cancer cells. Oncol Lett 2024; 28:483. [PMID: 39170883 PMCID: PMC11338234 DOI: 10.3892/ol.2024.14616] [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: 03/29/2024] [Accepted: 07/22/2024] [Indexed: 08/23/2024] Open
Abstract
Given the limited treatment options and high mortality rates associated with gastric cancer, there is a need to explore novel therapeutic options. The present study aimed to investigate the efficacy of lenvatinib, a multi-target tyrosine kinase inhibitor, in mitigating the progress of gastric cancer in vitro. Comprehensive analyses were conducted to assess the impact of lenvatinib on gastric cancer cells, focusing on the inhibition of viability, suppression of proliferation, induction of apoptosis and reduction of metastatic potential. The effects of lenvatinib on these activities were determined using 5-ethynyl-2'-deoxyuridine staining, colony formation assay, flow cytometry, western blotting, scratch assay and Transwell assay. In addition, bioinformatics analyses were employed to identify key regulatory targets of lenvatinib, with particular attention given to platelet-derived growth factor receptor β (PDGFRB). In addition, the effects of PDGFRB overexpression on the regulation of lenvatinib were explored. Lenvatinib demonstrated significant inhibitory effects on the viability, proliferation and metastatic capabilities of MKN45 and HGC27 gastric cancer cell lines. Bioinformatics analyses identified PDGFRB as a crucial target of lenvatinib, with its downregulation showing promise in enhancing overall survival rates of patients with gastric cancer. By contrast, PDGFRB overexpression reversed the effects of lenvatinib on cells. The present findings underscore the potential of lenvatinib as a promising therapeutic option in the treatment of gastric cancer. By elucidating its mechanism of action and identifying PDGFRB as a primary target, the present study may aid further clinical advancements.
Collapse
Affiliation(s)
- Xiaoyi Tong
- Graduate School, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310000, P.R. China
- Department of Pharmacy, Lanxi People's Hospital, Jinhua, Zhejiang 321100, P.R. China
| | - Jun Du
- Department of Nursing, Lanxi People's Hospital, Jinhua, Zhejiang 321100, P.R. China
| | - Qiaoling Jiang
- Department of Clinical Laboratory, Lanxi People's Hospital, Jinhua, Zhejiang 321100, P.R. China
| | - Qiaoli Wu
- Department of Pharmacy, Lanxi People's Hospital, Jinhua, Zhejiang 321100, P.R. China
| | - Shuxia Zhao
- Department of Pharmacy, Lanxi People's Hospital, Jinhua, Zhejiang 321100, P.R. China
| | - Shuhang Chen
- Department of Gastroenterology, Lanxi People's Hospital, Jinhua, Zhejiang 321100, P.R. China
| |
Collapse
|
5
|
Bintintan V, Burz C, Pintea I, Muntean A, Deleanu D, Lupan I, Samasca G. The Importance of Extracellular Vesicle Screening in Gastric Cancer: A 2024 Update. Cancers (Basel) 2024; 16:2574. [PMID: 39061213 PMCID: PMC11274824 DOI: 10.3390/cancers16142574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 07/14/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
Extracellular vesicles, or EVs, are membrane-bound nanocompartments produced by tumor cells. EVs carry proteins and nucleic acids from host cells to target cells, where they can transfer lipids, proteomes, and genetic material to change the function of target cells. EVs serve as reservoirs for mobile cellular signals. The collection of EVs using less invasive processes has piqued the interest of many researchers. Exosomes carry substances that can suppress the immune system. If the results of exosome screening are negative, immunotherapy will be beneficial for GC patients. In this study, we provide an update on EVs and GC based on ongoing review papers and clinical trials.
Collapse
Affiliation(s)
- Vasile Bintintan
- Department of Surgery 1, Iuliu Hatieganu University of Medicine and Pharmacy, 400006 Cluj-Napoca, Romania;
| | - Claudia Burz
- Institute of Oncology “Prof. Ion Chiricuta”, 400015 Cluj-Napoca, Romania;
- Department of Immunology, Iuliu Hatieganu University of Medicine and Pharmacy, 400006 Cluj-Napoca, Romania; (I.P.); (A.M.); (D.D.)
| | - Irena Pintea
- Department of Immunology, Iuliu Hatieganu University of Medicine and Pharmacy, 400006 Cluj-Napoca, Romania; (I.P.); (A.M.); (D.D.)
| | - Adriana Muntean
- Department of Immunology, Iuliu Hatieganu University of Medicine and Pharmacy, 400006 Cluj-Napoca, Romania; (I.P.); (A.M.); (D.D.)
| | - Diana Deleanu
- Department of Immunology, Iuliu Hatieganu University of Medicine and Pharmacy, 400006 Cluj-Napoca, Romania; (I.P.); (A.M.); (D.D.)
| | - Iulia Lupan
- Department of Molecular Biology, Babes-Bolyai University, 400084 Cluj-Napoca, Romania;
| | - Gabriel Samasca
- Department of Immunology, Iuliu Hatieganu University of Medicine and Pharmacy, 400006 Cluj-Napoca, Romania; (I.P.); (A.M.); (D.D.)
| |
Collapse
|
6
|
Sasajima N, Sumazaki M, Oshima Y, Ito M, Yajima S, Takizawa H, Wang H, Li SY, Zhang BS, Yoshida Y, Hiwasa T, Shimada H. Stage-Specific Alteration and Prognostic Relationship of Serum Fumarate Hydratase Autoantibodies in Gastric Cancer. Int J Mol Sci 2024; 25:5470. [PMID: 38791507 PMCID: PMC11121488 DOI: 10.3390/ijms25105470] [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: 05/09/2024] [Accepted: 05/13/2024] [Indexed: 05/26/2024] Open
Abstract
The relationship between energy production and cancer is attracting attention. This study aimed to investigate the clinicopathological significance of fumarate hydratase (FH), a tricarboxylic acid cycle enzyme, in gastric cancer using autoantibodies as biomarkers. The study analyzed 116 patients who underwent gastric cancer surgery and 96 healthy controls. Preoperative serum FH autoantibody (s-FH-Ab) titers were analyzed using an immunosorbent assay with an amplified luminescent proximity homogeneous assay. Receiver operating characteristic analysis was used to determine the cutoff s-FH-Ab titer. Clinicopathological factors and prognosis were compared between the high and low s-FH-Ab groups. The s-FH-Ab levels were significantly higher in the gastric cancer group than in the control group (p = 0.01). Levels were elevated even in patients with stage I gastric cancer compared with healthy controls (p = 0.02). A low s-FH-Ab level was significantly associated with distant metastasis (p = 0.01), peritoneal dissemination (p < 0.05), and poor overall survival (p < 0.01). Multivariate analysis revealed that low s-FH-Ab levels were an independent risk factor for poor prognosis (p < 0.01). Therefore, s-FH-Ab levels may be a useful biomarker for early diagnosis and the prediction of prognosis in patients with gastric cancer.
Collapse
Affiliation(s)
- Natsuko Sasajima
- Department of Gastroenterological Surgery, Toho University School of Medicine, Tokyo 143-8541, Japan; (N.S.); (Y.O.); (S.Y.)
| | - Makoto Sumazaki
- Department of Clinical Oncology, Toho University Graduate School of Medicine, Tokyo 143-8541, Japan; (M.S.); (M.I.); (T.H.)
| | - Yoko Oshima
- Department of Gastroenterological Surgery, Toho University School of Medicine, Tokyo 143-8541, Japan; (N.S.); (Y.O.); (S.Y.)
| | - Masaaki Ito
- Department of Clinical Oncology, Toho University Graduate School of Medicine, Tokyo 143-8541, Japan; (M.S.); (M.I.); (T.H.)
| | - Satoshi Yajima
- Department of Gastroenterological Surgery, Toho University School of Medicine, Tokyo 143-8541, Japan; (N.S.); (Y.O.); (S.Y.)
| | - Hirotaka Takizawa
- Port Square Kashiwado Clinic, Kashiwado Memorial Foundation, Chiba 260-0025, Japan;
| | - Hao Wang
- Department of Neurological Surgery, Chiba University Graduate School of Medicine, Chiba 260-8670, Japan; (H.W.); (S.-Y.L.); (B.-S.Z.); (Y.Y.)
| | - Shu-Yang Li
- Department of Neurological Surgery, Chiba University Graduate School of Medicine, Chiba 260-8670, Japan; (H.W.); (S.-Y.L.); (B.-S.Z.); (Y.Y.)
| | - Bo-Shi Zhang
- Department of Neurological Surgery, Chiba University Graduate School of Medicine, Chiba 260-8670, Japan; (H.W.); (S.-Y.L.); (B.-S.Z.); (Y.Y.)
| | - Yoichi Yoshida
- Department of Neurological Surgery, Chiba University Graduate School of Medicine, Chiba 260-8670, Japan; (H.W.); (S.-Y.L.); (B.-S.Z.); (Y.Y.)
| | - Takaki Hiwasa
- Department of Clinical Oncology, Toho University Graduate School of Medicine, Tokyo 143-8541, Japan; (M.S.); (M.I.); (T.H.)
- Department of Neurological Surgery, Chiba University Graduate School of Medicine, Chiba 260-8670, Japan; (H.W.); (S.-Y.L.); (B.-S.Z.); (Y.Y.)
| | - Hideaki Shimada
- Department of Gastroenterological Surgery, Toho University School of Medicine, Tokyo 143-8541, Japan; (N.S.); (Y.O.); (S.Y.)
- Department of Clinical Oncology, Toho University Graduate School of Medicine, Tokyo 143-8541, Japan; (M.S.); (M.I.); (T.H.)
| |
Collapse
|
7
|
Burz C, Pop V, Silaghi C, Lupan I, Samasca G. Prognosis and Treatment of Gastric Cancer: A 2024 Update. Cancers (Basel) 2024; 16:1708. [PMID: 38730659 PMCID: PMC11083929 DOI: 10.3390/cancers16091708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 04/23/2024] [Accepted: 04/26/2024] [Indexed: 05/13/2024] Open
Abstract
Due to the high death rate associated with gastric cancer, a great deal of research has been conducted on this disease. The goal of this paper was to start a trimestral review of 2024 for the year that had just started. The scientific literature from 1 January 2024 was chosen with consideration of the the guidelines of the European Society of Medical Oncology (ESMO), which are updated with new findings but not systematically reviewed annually. We used the search term "gastric cancer" to find the most current publications in the PubMed database related to the prognosis and treatment of gastric cancer. As previously said, the only articles that satisfied the inclusion criteria were those from 2024. Articles with case reports were eliminated since they had nothing to do with our research. The treatment of gastric cancer is the focus of the majority of articles from 2024. The primary research axes include surgery and immunonutrition, immunotherapy and Helicobacter pylori, and therapeutic targets. Patients with GC may experience less psychological, social, and financial hardship if the recently identified markers discovered in circulation are better assessed and validated. This could be achieved by either including the markers in an artificial intelligence-based diagnostic score or by using them in conjunction with traditional diagnostic methods. Due to the rising death rate associated with GC, funding for research into diagnosis, prognosis, therapy, and therapeutic targets is essential.
Collapse
Affiliation(s)
- Claudia Burz
- Institute of Oncology “Prof. Dr. Ion Chiricuta”, 400015 Cluj-Napoca, Romania; (C.B.); (V.P.)
- Department of Immunology, Iuliu Hatieganu University of Medicine and Pharmacy, 400162 Cluj-Napoca, Romania
| | - Vlad Pop
- Institute of Oncology “Prof. Dr. Ion Chiricuta”, 400015 Cluj-Napoca, Romania; (C.B.); (V.P.)
| | - Ciprian Silaghi
- Department of Biochemistry, Iuliu Hatieganu University of Medicine and Pharmacy, 400338 Cluj-Napoca, Romania;
| | - Iulia Lupan
- Institute for Interdisciplinary Research in Bio-Nano-Sciences, 400271 Cluj-Napoca, Romania;
- Department of Molecular Biology, Babes-Bolyai University, 400084 Cluj-Napoca, Romania
| | - Gabriel Samasca
- Department of Immunology, Iuliu Hatieganu University of Medicine and Pharmacy, 400162 Cluj-Napoca, Romania
| |
Collapse
|
8
|
Fasano R, Serratì S, Rafaschieri T, Longo V, Di Fonte R, Porcelli L, Azzariti A. Small-Cell Lung Cancer: Is Liquid Biopsy a New Tool Able to Predict the Efficacy of Immunotherapy? Biomolecules 2024; 14:396. [PMID: 38672414 PMCID: PMC11048475 DOI: 10.3390/biom14040396] [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: 02/07/2024] [Revised: 03/18/2024] [Accepted: 03/22/2024] [Indexed: 04/28/2024] Open
Abstract
Small-cell lung cancer (SCLC) cases represent approximately 15% of all lung cancer cases, remaining a recalcitrant malignancy with poor survival and few treatment options. In the last few years, the addition of immunotherapy to chemotherapy improved clinical outcomes compared to chemotherapy alone, resulting in the current standard of care for SCLC. However, the advantage of immunotherapy only applies to a few SCLC patients, and predictive biomarkers selection are lacking for SCLC. In particular, due to some features of SCLC, such as high heterogeneity, elevated cell plasticity, and low-quality tissue samples, SCLC biopsies cannot be used as biomarkers. Therefore, the characterization of the tumor and, subsequently, the selection of an appropriate therapeutic combination may benefit greatly from liquid biopsy. Soluble factors, circulating tumor DNA (ctDNA), circulating tumor cells (CTCs), and extracellular vesicles (EVs) are now useful tools in the characterization of SCLC. This review summarizes the most recent data on biomarkers detectable with liquid biopsy, emphasizing their role in supporting tumor detection and their potential role in SCLC treatment choice.
Collapse
Affiliation(s)
- Rossella Fasano
- Laboratory of Experimental Pharmacology, IRCCS Istituto Tumori Giovanni Paolo II, V.Le O. Flacco, 65, 70124 Bari, Italy; (R.F.); (T.R.); (R.D.F.); (L.P.); (A.A.)
| | - Simona Serratì
- Laboratory of Experimental Pharmacology, IRCCS Istituto Tumori Giovanni Paolo II, V.Le O. Flacco, 65, 70124 Bari, Italy; (R.F.); (T.R.); (R.D.F.); (L.P.); (A.A.)
| | - Tania Rafaschieri
- Laboratory of Experimental Pharmacology, IRCCS Istituto Tumori Giovanni Paolo II, V.Le O. Flacco, 65, 70124 Bari, Italy; (R.F.); (T.R.); (R.D.F.); (L.P.); (A.A.)
| | - Vito Longo
- Medical Thoracic Oncology Unit, IRCCS Istituto Tumori Giovanni Paolo II, 70124 Bari, Italy;
| | - Roberta Di Fonte
- Laboratory of Experimental Pharmacology, IRCCS Istituto Tumori Giovanni Paolo II, V.Le O. Flacco, 65, 70124 Bari, Italy; (R.F.); (T.R.); (R.D.F.); (L.P.); (A.A.)
| | - Letizia Porcelli
- Laboratory of Experimental Pharmacology, IRCCS Istituto Tumori Giovanni Paolo II, V.Le O. Flacco, 65, 70124 Bari, Italy; (R.F.); (T.R.); (R.D.F.); (L.P.); (A.A.)
| | - Amalia Azzariti
- Laboratory of Experimental Pharmacology, IRCCS Istituto Tumori Giovanni Paolo II, V.Le O. Flacco, 65, 70124 Bari, Italy; (R.F.); (T.R.); (R.D.F.); (L.P.); (A.A.)
| |
Collapse
|
9
|
Nakajima D, Konno R, Miyashita Y, Ishikawa M, Ohara O, Kawashima Y. Proteome Analysis of Serum Purified Using Solanum tuberosum and Lycopersicon esculentum Lectins. Int J Mol Sci 2024; 25:1315. [PMID: 38279312 PMCID: PMC10816257 DOI: 10.3390/ijms25021315] [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: 12/27/2023] [Revised: 01/17/2024] [Accepted: 01/19/2024] [Indexed: 01/28/2024] Open
Abstract
Serum and plasma exhibit a broad dynamic range of protein concentrations, posing challenges for proteome analysis. Various technologies have been developed to reduce this complexity, including high-abundance depletion methods utilizing antibody columns, extracellular vesicle enrichment techniques, and trace protein enrichment using nanobead cocktails. Here, we employed lectins to address this, thereby extending the scope of biomarker discovery in serum or plasma using a novel approach. We enriched serum proteins using 37 different lectins and subjected them to LC-MS/MS analysis with data-independent acquisition. Solanum tuberosum lectin (STL) and Lycopersicon esculentum lectin (LEL) enabled the detection of more serum proteins than the other lectins. STL and LEL bind to N-acetylglucosamine oligomers, emphasizing the significance of capturing these oligomer-binding proteins when analyzing serum trace proteins. Combining STL and LEL proved more effective than using them separately, allowing us to identify over 3000 proteins from serum through single-shot proteome analysis. We applied the STL/LEL trace-protein enrichment method to the sera of systemic lupus erythematosus model mice. This revealed differences in >1300 proteins between the systemic lupus erythematosus model and control mouse sera, underscoring the utility of this method for biomarker discovery.
Collapse
Affiliation(s)
- Daisuke Nakajima
- Department of Applied Genomics, Kazusa DNA Research Institute, 2-5-23 Kazusa Kamatari, Kisarazu 292-0818, Chiba, Japan; (D.N.); (R.K.); (Y.M.); (M.I.); (O.O.)
| | - Ryo Konno
- Department of Applied Genomics, Kazusa DNA Research Institute, 2-5-23 Kazusa Kamatari, Kisarazu 292-0818, Chiba, Japan; (D.N.); (R.K.); (Y.M.); (M.I.); (O.O.)
| | - Yasuomi Miyashita
- Department of Applied Genomics, Kazusa DNA Research Institute, 2-5-23 Kazusa Kamatari, Kisarazu 292-0818, Chiba, Japan; (D.N.); (R.K.); (Y.M.); (M.I.); (O.O.)
- Department of Developmental Biology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Chiba, Japan
| | - Masaki Ishikawa
- Department of Applied Genomics, Kazusa DNA Research Institute, 2-5-23 Kazusa Kamatari, Kisarazu 292-0818, Chiba, Japan; (D.N.); (R.K.); (Y.M.); (M.I.); (O.O.)
| | - Osamu Ohara
- Department of Applied Genomics, Kazusa DNA Research Institute, 2-5-23 Kazusa Kamatari, Kisarazu 292-0818, Chiba, Japan; (D.N.); (R.K.); (Y.M.); (M.I.); (O.O.)
| | - Yusuke Kawashima
- Department of Applied Genomics, Kazusa DNA Research Institute, 2-5-23 Kazusa Kamatari, Kisarazu 292-0818, Chiba, Japan; (D.N.); (R.K.); (Y.M.); (M.I.); (O.O.)
| |
Collapse
|
10
|
Ke X, Cai X, Bian B, Shen Y, Zhou Y, Liu W, Wang X, Shen L, Yang J. Predicting early gastric cancer risk using machine learning: A population-based retrospective study. Digit Health 2024; 10:20552076241240905. [PMID: 38559579 PMCID: PMC10979538 DOI: 10.1177/20552076241240905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 03/05/2024] [Indexed: 04/04/2024] Open
Abstract
Background Early detection and treatment are crucial for reducing gastrointestinal tumour-related mortality. The diagnostic efficiency of the most commonly used diagnostic markers for gastric cancer (GC) is not very high. A single laboratory test cannot meet the requirements of early screening, and machine learning methods are needed to aid the early diagnosis of GC by combining multiple indicators. Methods Based on the XGBoost algorithm, a new model was developed to distinguish between GC and precancerous lesions in newly admitted patients between 2018 and 2023 using multiple laboratory tests. We evaluated the ability of the prediction score derived from this model to predict early GC. In addition, we investigated the efficacy of the model in correctly screening for GC given negative protein tumour marker results. Results The XHGC20 model constructed using the XGBoost algorithm could distinguish GC from precancerous disease well (area under the receiver operating characteristic curve [AUC] = 0.901), with a sensitivity, specificity and cut-off value of 0.830, 0.806 and 0.265, respectively. The prediction score was very effective in the diagnosis of early GC. When the cut-off value was 0.27, and the AUC was 0.888, the sensitivity and specificity were 0.797 and 0.807, respectively. The model was also effective at evaluating GC given negative conventional markers (AUC = 0.970), with the sensitivity and specificity of 0.941 and 0.906, respectively, which helped to reduce the rate of missed diagnoses. Conclusions The XHGC20 model established by the XGBoost algorithm integrates information from 20 clinical laboratory tests and can aid in the early screening of GC, providing a useful new method for auxiliary laboratory diagnosis.
Collapse
Affiliation(s)
- Xing Ke
- Department of Clinical Laboratory, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Faculty of Medical Laboratory Science, College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Artificial Intelligence Medicine, Shanghai Academy of Experimental Medicine, Shanghai, China
- Department of Pathology, Ruijin Hospital and College of Basic Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xinyu Cai
- Department of Clinical Laboratory, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bingxian Bian
- Department of Clinical Laboratory, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuanheng Shen
- Department of Clinical Laboratory, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yunlan Zhou
- Department of Clinical Laboratory, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei Liu
- Department of Research Collaboration, R&D Center, Beijing Deepwise & League of PHD Technology Co., Ltd, Beijing, China
| | - Xu Wang
- Department of Pathology, Ruijin Hospital and College of Basic Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lisong Shen
- Department of Clinical Laboratory, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Faculty of Medical Laboratory Science, College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Artificial Intelligence Medicine, Shanghai Academy of Experimental Medicine, Shanghai, China
| | - Junyao Yang
- Department of Clinical Laboratory, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Faculty of Medical Laboratory Science, College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Artificial Intelligence Medicine, Shanghai Academy of Experimental Medicine, Shanghai, China
| |
Collapse
|
11
|
Ji C, Cai H, Jin X, Yin K, Zhao D, Feng Z, Liu L. Identification of Immune Infiltrating Cell-Related Biomarkers in Early Gastric Cancer Progression. Technol Cancer Res Treat 2024; 23:15330338241262724. [PMID: 38860335 PMCID: PMC11168250 DOI: 10.1177/15330338241262724] [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: 12/24/2023] [Revised: 04/25/2024] [Accepted: 05/28/2024] [Indexed: 06/12/2024] Open
Abstract
OBJECTIVES Gastric cancer (GC) is one of the most prevalent malignancies worldwide, and early detection is crucial for improving patient survival rates. We aimed to identify immune infiltrating cell-related biomarkers in early gastric cancer (EGC) progression. METHODS The GSE55696 and GSE130823 datasets with low-grade intraepithelial neoplasia (LGIN), high-grade intraepithelial neoplasia (HGIN), and EGC samples were downloaded from the Gene Expression Omnibus database to perform an observational study. Immune infiltration analysis was performed by single sample gene set enrichment analysis and Estimation of STromal and Immune cells in MAlignant Tumor tissues using Expression data. Weighted gene co-expression network analysis was used to explore the co-expression modules and genes, and further enrichment analysis was performed on these genes. A protein-protein interaction (PPI) network of these genes was constructed to identify biomarkers associated with EGC progression. Screened hub genes were validated by the rank sum test and reverse transcription quantitative polymerase chain reaction. RESULTS Immune scores were significantly elevated in EGC samples compared to LGIN and HGIN samples. The green-yellow module exhibited the strongest correlation with both immune score and disease progression. The 87 genes within this module were associated with the chemokine signaling pathways, the PI3K-Akt signaling pathways, leukocyte transendothelial migration, and Ras signaling pathways. Through PPI network analysis, the hub genes identified were protein tyrosine phosphatase receptor-type C (PTPRC), pleckstrin, CD53, CD48, lymphocyte cytosolic protein 1 (LCP1), hematopoietic cell-specific Lyn substrate 1, IKAROS Family Zinc Finger 1, Bruton tyrosine kinase, and Vav guanine nucleotide exchange factor 1. Notably, CD48, LCP1, and PTPRC showed high expression levels in EGC samples, with the remaining hub genes demonstrating a similar expression trend. CONCLUSION This study identified 9 immune cell-related biomarkers that may be actively involved in the progression of EGC and serve as potential targets for GC diagnosis and treatment.
Collapse
Affiliation(s)
- Chenguang Ji
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, P.R. China
| | - Hongmei Cai
- Deparment of Oncology, Hebei Chest Hospital, Shijiazhuang, Hebei, P.R. China
| | - Xiaoxu Jin
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, P.R. China
| | - Kaige Yin
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, P.R. China
| | - Dongqiang Zhao
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, P.R. China
| | - Zhijie Feng
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, P.R. China
| | - Li Liu
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, P.R. China
| |
Collapse
|