1
|
Maturana MJ, Padilla O, Santoro PM, Alarcón MA, Olivares W, Blanco A, Armisen R, Garrido M, Aravena E, Barrientos C, Calvo-Belmar A, Corvalán AH. Methylated Reprimo Cell-Free DNA as a Non-Invasive Biomarker for Gastric Cancer. Int J Mol Sci 2025; 26:3333. [PMID: 40244164 PMCID: PMC11989948 DOI: 10.3390/ijms26073333] [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: 01/23/2025] [Revised: 03/03/2025] [Accepted: 03/08/2025] [Indexed: 04/18/2025] Open
Abstract
Restrictions resulting from the COVID-19 pandemic abruptly reversed the slow decline of the diagnosis and mortality rates of gastric cancer (GC). This scenario highlights the importance of developing cost-effective methods for mass screening and evaluation of treatment response. In this study, we evaluated a non-invasive method based on the circulating methylated cell-free DNA (cfDNA) of Reprimo (RPRM), a tumor suppressor gene associated with the development of GC. Methylated RPRM cfDNA was analyzed in three de-identified cohorts: Cohort 1 comprised 81 participants with GC and 137 healthy donors (HDs); Cohort 2 comprised 27 participants with GC undergoing gastrectomy and/or chemotherapy analyzed at the beginning and after three months of treatment; and Cohort 3 comprised 1105 population-based participants in a secondary prevention program who underwent esophagogastroduodenal (EGD) endoscopy. This cohort includes 180 normal participants, 845 participants with premalignant conditions (692 with chronic atrophic gastritis [AG] and 153 with gastric intestinal metaplasia/low-grade dysplasia [GIM/LGD]), 21 with high-grade dysplasia/early GC [HGD/eGC], and 59 with advanced GC [aGC]). A nested case-control substudy was performed using a combination of methylated RPRM cfDNA and pepsinogens (PG)-I/II ratio. The dense CpG island of the promoter region of the RPRM gene was bisulfite sequenced and analyzed to develop a fluorescence-based real-time PCR assay (MethyLight). This assay allows the determination of the absolute number of copies of methylated RPRM cfDNA. A targeted sequence of PCR amplicon products confirmed the gastric origin of the plasma-isolated samples. In Cohort 1, the mean value of GCs (32,240.00 copies/mL) was higher than that of the HD controls (139.00 copies/mL) (p < 0.0001). After dividing this cohort into training-validation subcohorts, we identified an area under the curve of 0.764 (95% confidence interval (CI) = 0.683-0.845) in the training group. This resulted in a cut-off value of 87.37 copies/mL (sensitivity 70.0% and specificity 80.2%). The validation subcohort predicted a sensitivity of 66.67% and a specificity of 83.33%. In Cohort 2 (monitoring treatment response), RPRM levels significantly decreased in responders (p = 0.0042) compared to non-responders. In Cohort 3 (population-based participants), 18.9% %, 24.1%, 30.7%, 47.0%, and 71.2% of normal, AG, GIM/LGD, HGD/eGC, and aGC participants tested positive for methylated RPRM cfDNA, respectively. Overall sensitivity and specificity in distinguishing normal/premalignant conditions vs. GC were 65.0% (95% CI 53.52% to 75.33%) and 75.9% (95% CI 73.16% to 78.49%), respectively, with an accuracy of 75.11% (95% CI 72.45% to 77.64%). Logistic regression analyses revealed an OR of 1.85 (95% CI 1.11-3.07, p = 0.02) and an odds ratio (OR) of 3.9 (95% CI 1.53-9.93, p = 0.004) for the risk of developing GIM/LGD and HGD/eGC, respectively. The combined methylated RPRM cfDNA and PG-I/II ratio reached a sensitivity of 78.9% (95% CI 54.43% to 93.95%) and specificity of 63.04% (95% CI 52.34% to 72.88%) for detecting HGD/eGC vs. three to six age- and sex-matched participants with premalignant conditions. Our results demonstrate that methylated RPRM cfDNA should be considered a direct biomarker for the non-invasive detection of GC and a predictive biomarker for treatment response.
Collapse
Affiliation(s)
- María José Maturana
- Department of Hematology and Oncology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Portugal 61, Santiago 8330023, Chile; (M.J.M.); (P.M.S.); (M.A.A.); (W.O.); (M.G.)
| | - Oslando Padilla
- School of Public Health, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330023, Chile;
| | - Pablo M. Santoro
- Department of Hematology and Oncology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Portugal 61, Santiago 8330023, Chile; (M.J.M.); (P.M.S.); (M.A.A.); (W.O.); (M.G.)
| | - Maria Alejandra Alarcón
- Department of Hematology and Oncology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Portugal 61, Santiago 8330023, Chile; (M.J.M.); (P.M.S.); (M.A.A.); (W.O.); (M.G.)
| | - Wilda Olivares
- Department of Hematology and Oncology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Portugal 61, Santiago 8330023, Chile; (M.J.M.); (P.M.S.); (M.A.A.); (W.O.); (M.G.)
| | - Alejandro Blanco
- Centro de Genética y Genómica, Instituto de Ciencias e Innovación en Medicina (ICIM), Facultad de Medicina Clínica Alemana Universidad del Desarrollo, Santiago 7550000, Chile; (A.B.); (R.A.)
| | - Ricardo Armisen
- Centro de Genética y Genómica, Instituto de Ciencias e Innovación en Medicina (ICIM), Facultad de Medicina Clínica Alemana Universidad del Desarrollo, Santiago 7550000, Chile; (A.B.); (R.A.)
| | - Marcelo Garrido
- Department of Hematology and Oncology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Portugal 61, Santiago 8330023, Chile; (M.J.M.); (P.M.S.); (M.A.A.); (W.O.); (M.G.)
| | - Edmundo Aravena
- Instituto Chileno Japones de Enfermedades Digestivas, Hospital Clinico San Borja Arriaran, Servicio Salud Metropolitano Central, Santiago, Chile and Fundación Arturo López Pérez, Santiago 8360160, Chile; (E.A.); (C.B.)
| | - Carlos Barrientos
- Instituto Chileno Japones de Enfermedades Digestivas, Hospital Clinico San Borja Arriaran, Servicio Salud Metropolitano Central, Santiago, Chile and Fundación Arturo López Pérez, Santiago 8360160, Chile; (E.A.); (C.B.)
| | - Alfonso Calvo-Belmar
- Hospital Dr. Sotero del Rio, Servicio Salud Metropolitano Sur-Oriente, Santiago 8207257, Chile;
| | - Alejandro H. Corvalán
- Department of Hematology and Oncology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Portugal 61, Santiago 8330023, Chile; (M.J.M.); (P.M.S.); (M.A.A.); (W.O.); (M.G.)
| |
Collapse
|
2
|
Shah SAR, Mumtaz M, Sharif S, Mustafa I, Nayila I. Helicobacter pylori and gastric cancer: current insights and nanoparticle-based interventions. RSC Adv 2025; 15:5558-5570. [PMID: 39967885 PMCID: PMC11834156 DOI: 10.1039/d4ra07886a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 02/07/2025] [Indexed: 02/20/2025] Open
Abstract
Background: H. pylori is recognized as one of the main causes of gastric cancer, and this type of cancer is considered as one of the leading diseases causing cancer deaths all over the world. Knowledge on the interactions between H. pylori and gastric carcinogenesis is important for designing preventive measures. Objective: the objective of this review is to summarize the available literature on H. pylori and gastric cancer, specifically regarding the molecular mechanisms, nanoparticle-based therapy and clinical developments. Methods: the databases including PubMed, Google Scholar and web of science were searched as well as papers from 2010 to 2024 were considered for review. Research literature on H. pylori, gastric cancer, nanoparticles, nanomedicine, and therapeutic interventions was summarized for current findings and possible treatments. Results: the presence of H. pylori in gastric mucosa causes chronic inflammation and several molecular alterations such as DNA alteration, epigenetic changes and activation of oncogenic signaling pathways which causes gastric carcinogenesis. Conventional antibiotic treatments have some issues because of the constantly rising levels of antibiotic resistance. Lipid based nanoformulations, polymeric and metallic nanoparticles have been delivered in treatment of H. pylori to improve drug delivery and alter immunological responses. Conclusion: nanoparticle based interventions have been widely explored as drug delivery systems by improving the treatment strategies against H. pylori induced gastric cancer. Further studies and clinical trials are required to bring these findings into a clinical setting in order to possibly alter the management of H. pylori related gastric malignancies.
Collapse
Affiliation(s)
- Syed Ali Raza Shah
- Institute of Molecular Biology and Biotechnology (IMBB), The University of Lahore Lahore Pakistan
| | - Maria Mumtaz
- Institute of Molecular Biology and Biotechnology (IMBB), The University of Lahore Lahore Pakistan
| | - Sumaira Sharif
- Institute of Molecular Biology and Biotechnology (IMBB), The University of Lahore Lahore Pakistan
| | - Imtiaz Mustafa
- Institute of Molecular Biology and Biotechnology (IMBB), The University of Lahore Lahore Pakistan
| | - Iffat Nayila
- Department of Pharmacy, The University of Lahore Sargodha Campus Sargodha Pakistan
| |
Collapse
|
3
|
Wu HM, Ying XX, Lv LL, Hu JW. Diagnostic implications of neutrophil-to-lymphocyte ratio, platelet-to-lymphocyte ratio, and systemic immune-inflammatory index for gastric carcinoma. World J Gastrointest Surg 2025; 17:100130. [PMID: 39872777 PMCID: PMC11757178 DOI: 10.4240/wjgs.v17.i1.100130] [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: 08/20/2024] [Revised: 09/19/2024] [Accepted: 09/27/2024] [Indexed: 12/27/2024] Open
Abstract
BACKGROUND The diagnosis of gastric carcinoma (GC) is essential for improving clinical outcomes. However, the biomarkers currently used for GC screening are not ideal. AIM To explore the diagnostic implications of the neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio (PLR), and systemic immune-inflammatory index (SII) for GC. METHODS The baseline data of 133 patients with GC and 134 patients with precancerous gastric conditions admitted between January 2022 and December 2023 were retrospectively analyzed. The information on peripheral blood platelet, neutrophil, and lymphocyte counts in each patient was collected, and the NLR, PLR, and SII levels of both groups were calculated. Additionally, multivariate logistic regression analysis was conducted, and the diagnostic implications of NLR, PLR, and SII in differentiating patients with precancerous gastric conditions, compared with those with GC, were analyzed through receiver operating characteristic (ROC) curves. RESULTS The data indicated that NLR, PLR, and SII had abnormally increased levels in the patients with GC. Gender and body mass index were risk factors for the occurrence of GC. ROC data revealed that the areas under the curve of three patients with precancerous gastric conditions, who were differentiated from those with GC, were 0.824, 0.787, and 0.842, respectively. CONCLUSION NLR, PLR, and SII are all abnormally expressed in GC and have diagnostic implications, especially when used as joint indicators, in distinguishing patients with precancerous gastric conditions from those with GC.
Collapse
Affiliation(s)
- Huang-Min Wu
- Department of Gastroenterology, Dongyang People's Hospital, Dongyang 322100, Zhejiang Province, China
| | - Xiao-Xuan Ying
- Department of Gastroenterology, Dongyang People's Hospital, Dongyang 322100, Zhejiang Province, China
| | - Li-Li Lv
- Department of Gastroenterology, Dongyang People's Hospital, Dongyang 322100, Zhejiang Province, China
| | - Jian-Wen Hu
- Department of Gastroenterology, Dongyang People's Hospital, Dongyang 322100, Zhejiang Province, China
| |
Collapse
|
4
|
Han X, Yu W. Value of serum pepsinogen ratio screening for early gastric cancer and precancerous lesions in Youcheng area. World J Gastrointest Surg 2024; 16:3729-3736. [PMID: 39734444 PMCID: PMC11650226 DOI: 10.4240/wjgs.v16.i12.3729] [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: 08/28/2024] [Revised: 10/16/2024] [Accepted: 10/28/2024] [Indexed: 11/27/2024] Open
Abstract
BACKGROUND The 5-year survival rate of patients with advanced gastric cancer remains extremely low (< 15%), whereas the 5-year survival rate of patients with early gastric cancer (EGC) is > 90%. Consequently, strengthening the screening of patients with EGC and precancerous lesions (PCLs) is essential. AIM To identify the value of serum pepsinogen ratio (PGR) screening for EGC and PCLs in the Shengli Oilfield Central Hospital. METHODS We first selected 385 patients with gastric lesions in the Youcheng area, determining benign lesions, PCLs, and EGC in 135, 123, and 127 cases, respectively, based on endoscopy and case diagnosis. The positive rates of pepsinogen I, pepsinogen II and Helicobacter pylori (H. pylori) in the three groups were detected, and the PGR was calculated. Subsequently, we plotted receiver operating characteristic curves to analyze the screening value of PGR and H. pylori-positive rates for PCLs and EGC. RESULTS PGR expression demonstrated a decreasing trend in patients with benign lesions, PCLs, and EGC successively according to the detection results, whereas the H. pylori-positive rate was notably increased in patients with PCLs and EGC compared to those with benign lesions. The area under the curves (AUCs) of PGR, H. pylori, and their combination in differentiating patients with benign lesions from those with PCLs were 0.611, 0.582, and 0.689, respectively; PGR, H. pylori, and their combination had an AUC of 0.618, 0.502, and 0.618 in distinguishing PCL patients from EGC patients, respectively; the AUCs of PGR, H. pylori, and their combination in discriminating patients with benign lesions from those with EGC were 0.708, 0.581, and 0.750, respectively. CONCLUSION PGR has great screening potential for patients with EGC and PCLs in the Youcheng area, and the screening efficiency is further improved by combining the H. pylori-positive rate.
Collapse
Affiliation(s)
- Xue Han
- Department of General Practice, Shengli Oilfield Central Hospital, Dongying 257000, Shandong Province, China
| | - Wei Yu
- Health Management Center, Shengli Oilfield Central Hospital, Dongying 257000, Shandong Province, China
| |
Collapse
|
5
|
Chacko N, Ankri R. Non-invasive early-stage cancer detection: current methods and future perspectives. Clin Exp Med 2024; 25:17. [PMID: 39708168 DOI: 10.1007/s10238-024-01513-x] [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: 09/02/2024] [Accepted: 10/21/2024] [Indexed: 12/23/2024]
Abstract
This review paper explores the realm of non-invasive methods for early cancer detection. Early identification is crucial for effective therapeutic intervention, and non-invasive techniques have emerged as promising tools to enhance diagnostic accuracy and improve patient outcomes. The paper thoroughly examines the advantages, limitations, and prospects of various non-invasive approaches, including blood tests, non-blood-based tests, and diverse imaging modalities. It discusses the biomarkers found in blood for early-stage cancer detection, specifying the types of cancer associated with each biomarker. The non-blood-based tests focus on components in saliva, urine, and breath for cancer detection, alongside current studies and future perspectives on various cancers. Optical imaging methods covered in this review include fluorescence imaging in the near-infrared (NIR) region, bioluminescence imaging, and Raman spectroscopy for early-stage cancer detection. The review also highlights the pros and cons of ultrasound imaging in early-stage cancer detection. Additionally, the clinical implications of using AI for cancer detection, both present and future, are explored. This paper provides valuable insights for researchers and clinicians working in the field of non-invasive early-stage cancer detection.
Collapse
Affiliation(s)
- Neelima Chacko
- Department of Physics, Faculty of Natural Science, Ariel University, 40700, Ariel, Israel
| | - Rinat Ankri
- Department of Physics, Faculty of Natural Science, Ariel University, 40700, Ariel, Israel.
| |
Collapse
|
6
|
Wang H, Ding Y, Zhao S, Li K, Li D. Establishment and validation of a nomogram model for early diagnosis of gastric cancer: a large-scale cohort study. Front Oncol 2024; 14:1463480. [PMID: 39678515 PMCID: PMC11638037 DOI: 10.3389/fonc.2024.1463480] [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: 07/12/2024] [Accepted: 11/12/2024] [Indexed: 12/17/2024] Open
Abstract
Purpose Identifying high-risk populations and diagnosing gastric cancer (GC) early remains challenging. This study aimed to establish and verify a nomogram model for the early diagnosis of GC based on conventional laboratory indicators. Methods We performed a retrospective analysis of the clinical data of 2,770 individuals with first diagnosis of GC and 1,513 patients with benign gastric disease from January 2018 to December 2022. The cases were divided into the training set and validation set randomly, with a ratio of 7:3. Variable screening was performed by least absolute shrinkage and selection operator (LASSO) and logistic regression analysis. A nomogram was constructed in the training set to assist in the early diagnosis of GC. Results There were 4283 patients included in the study, with 2998 patients assigned in the training set and 1285 patients in the validation set. Through LASSO regression and logistic regression analysis, independent variables associated with GC were identified, including CEA, CA199, LYM, HGB, MCH, MCHC, PLT, ALB, TG, HDL, and AFR. The nomogram model was constructed using the above 11 independent indicators. The AUC was 0.803 for the training set and 0.797 for the validation set, indicating that the model showed high clinical diagnostic efficacy. The calibration curves and decision curve analysis (DCA) of the nomogram presented good calibration and clinical application ability. Conclusion Based on the analysis of large sample size, we constructed a nomogram model with 11 routine laboratory indicators, which showed good discrimination ability and calibration.
Collapse
Affiliation(s)
- Haiyu Wang
- School of Public Health, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Yumin Ding
- School of Public Health, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Shujing Zhao
- School of Public Health, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Kaixu Li
- School of Public Health, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Dehong Li
- Department of Clinical Laboratory, Gansu Provincial Hospital, Lanzhou, Gansu, China
| |
Collapse
|
7
|
Leja M. Where are we with gastric cancer screening in Europe in 2024? Gut 2024; 73:2074-2082. [PMID: 39237127 PMCID: PMC11671906 DOI: 10.1136/gutjnl-2024-332705] [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: 04/21/2024] [Accepted: 08/14/2024] [Indexed: 09/07/2024]
Abstract
The absolute number of annual cases of gastric cancer in Europe is rising. The Council of the European Union has recommended implementation of gastric cancer screening for countries or regions with a high gastric cancer incidence and death rates. However, as of 2024 no organised gastric cancer screening programme has been launched in Europe.There are several ways to decrease gastric cancer burden, but the screen and treat strategy for Helicobacter pylori (H. pylori) seems to be the most appropriate for Europe. It has to be noted that increased use of antibiotics would be associated with this strategy.Only organised population-based cancer screening is recommended in the European Union, therefore gastric cancer screening also is expected to fulfil the criteria of an organised screening programme. In this respect, several aspects of screening organisation need to be considered before full implementation of gastric cancer prevention in Europe; the age range of the target group, test types, H. pylori eradication regimens and surveillance strategies are among them. Currently, ongoing projects (GISTAR, EUROHELICAN, TOGAS and EUCanScreen) are expected to provide the missing evidence. Feedback from the decision-makers and the potential target groups, including vulnerable populations, will be important to planning the programme.This paper provides an overview of the recent decisions of the European authorities, the progress towards gastric cancer implementation in Europe and expected challenges. Finally, a potential algorithm for gastric cancer screening in Europe is proposed.
Collapse
Affiliation(s)
- Mārcis Leja
- Institute of Clinical and Preventive Medicine, University of Latvia, Riga, Latvia
- Department of Gastroenterology, Digestive Diseases Centre GASTRO, Riga, Latvia
- Department of Research, Riga East University Hospital, Riga, Latvia
| |
Collapse
|
8
|
Bazin T, Nozeret K, Julié C, Lamarque D, Touati E. Protein Biomarkers of Gastric Preneoplasia and Cancer Lesions in Blood: A Comprehensive Review. Cancers (Basel) 2024; 16:3019. [PMID: 39272877 PMCID: PMC11394471 DOI: 10.3390/cancers16173019] [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: 07/25/2024] [Revised: 08/20/2024] [Accepted: 08/23/2024] [Indexed: 09/15/2024] Open
Abstract
Gastric cancer (GC) is a major cause of cancer-related mortality worldwide. It is often associated with a bad prognosis because of its asymptomatic phenotype until advanced stages, highlighting the need for its prevention and early detection. GC development is preceded by the emergence of gastric preneoplasia lesions (GPNLs), namely atrophic gastritis (AG), intestinal metaplasia (IM), and dysplasia (DYS). GC is currently diagnosed by endoscopy, which is invasive and costly and has limited effectiveness for the detection of GPNLs. Therefore, the discovery of non-invasive biomarkers in liquid biopsies, such as blood samples, in order to identify the presence of gastric preneoplasia and/or cancer lesions at asymptomatic stages is of paramount interest. This comprehensive review provides an overview of recently identified plasma/serum proteins and their diagnostic performance for the prediction of GPNLs and early cancer lesions. Autoantibodies appear to be promising biomarkers for AG, IM and early gastric cancer detection, along with inflammation and immunity-related proteins and antibodies against H. pylori virulence factors. There is a lack of specific protein biomarkers with which to detect DYS. Despite the need for further investigation and validation, some emerging candidates could pave the way for the development of reliable, non-invasive diagnostic tests for the detection and prevention of GC.
Collapse
Affiliation(s)
- Thomas Bazin
- Department of Gastroenterology and Nutritional Support, Center for Intestinal Failure, Reference Centre of Rare Disease MarDI, Assistance Publique-Hôpitaux de Paris (AP-HP) Beaujon Hospital, University Paris Cité, F-92110 Clichy, France
- Infection & Inflammation, Unité Mixte de Recherche (UMR) 1173, Inserm, Université de Versailles-Saint-Quentin-en-Yvelines (UVSQ)/Université Paris Saclay, F-78180 Montigny-le-Bretonneux, France
| | - Karine Nozeret
- Équipe DMic01-Infection, Génotoxicité et Cancer, Département de Microbiologie, Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche (UMR) 6047, Institut Pasteur, Université Paris Cité, F-75015 Paris, France
| | - Catherine Julié
- Department of Anatomical Pathology, Université Paris Saclay/Université de Versailles-Saint-Quentin-en-Yvelines (UVSQ), Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Ambroise Paré, F-92100 Boulogne-Billancourt, France
| | - Dominique Lamarque
- Infection & Inflammation, Unité Mixte de Recherche (UMR) 1173, Inserm, Université de Versailles-Saint-Quentin-en-Yvelines (UVSQ)/Université Paris Saclay, F-78180 Montigny-le-Bretonneux, France
- Department of Gastroenterology, Université Paris Saclay/Université de Versailles-Saint-Quentin-en-Yvelines (UVSQ), Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Ambroise Paré, F-92100 Boulogne Billancourt, France
| | - Eliette Touati
- Équipe DMic01-Infection, Génotoxicité et Cancer, Département de Microbiologie, Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche (UMR) 6047, Institut Pasteur, Université Paris Cité, F-75015 Paris, France
| |
Collapse
|
9
|
Negahdary M. Role of miRNA-21 in cancer and its application in electrochemical bioanalysis. Bioanalysis 2024; 16:997-1000. [PMID: 38949192 PMCID: PMC11581167 DOI: 10.1080/17576180.2024.2368340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 06/12/2024] [Indexed: 07/02/2024] Open
Affiliation(s)
- Masoud Negahdary
- Department of Biomedical Engineering, Texas A&M University, 600 Discovery Drive, College Station, TX77840-3006, USA
- Center for Remote Health Technologies & Systems, Texas A&M Engineering Experiment Station, 600 Discovery Drive, College Station, TX77840-3006, USA
| |
Collapse
|
10
|
Gu Y, Zhao S. Risk factors for postoperative bleeding following endoscopic submucosal dissection in early gastric cancer: A systematic review and meta-analysis. Medicine (Baltimore) 2024; 103:e37762. [PMID: 38608116 PMCID: PMC11018221 DOI: 10.1097/md.0000000000037762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 03/08/2024] [Indexed: 04/14/2024] Open
Abstract
BACKGROUND Early gastric cancer (EGC) presents a significant challenge in surgical management, particularly concerning postoperative bleeding following endoscopic submucosal dissection. Understanding the risk factors associated with postoperative bleeding is crucial for improving patient outcomes. METHODS Adhering to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines, a systematic review and meta-analysis were conducted across PubMed, Embase, Web of Science, and the Cochrane Library without publication date restrictions. The inclusion criteria encompassed observational studies and randomized controlled trials focusing on EGC patients undergoing endoscopic submucosal dissection and their risk factors for postoperative bleeding. The Newcastle-Ottawa Scale was utilized for quality assessment. The effect size was calculated using random or fixed-effects models based on the observed heterogeneity. We assessed the heterogeneity between studies and conducted a sensitivity analysis. RESULTS In our meta-analysis, 6 studies involving 4868 EGC cases were analyzed. The risk of postoperative bleeding was notably increased with intraoperative ulcer detection (odds ratio: 1.97, 95% confidence interval [CI]: 1.03-3.76, I2 = 61.0%, P = .025) and antithrombotic medication use (odds ratio: 2.02, 95% CI: 1.16-3.51, I2 = 57.2%, P = .039). Lesion resection size showed a significant mean difference (5.16, 95% CI: 2.97-7.98, P < .01), and longer intraoperative procedure time was associated with increased bleeding risk (mean difference: 11.69 minutes, 95% CI: 1.82-26.20, P < .05). Sensitivity analysis affirmed the robustness of these findings, and publication bias assessment indicated no significant bias. CONCLUSIONS In EGC treatment, the risk of post-endoscopic submucosal dissection bleeding is intricately linked to factors like intraoperative ulcer detection, antithrombotic medication use, the extent of lesion resection, and the length of the surgical procedure. These interwoven risk factors necessitate careful consideration and integrated management strategies to enhance patient outcomes and safety in EGC surgeries.
Collapse
Affiliation(s)
- Yuanbo Gu
- Department of Gastroenterology, Jilin Chemical Hospital, Jilin, China
| | - Shuchang Zhao
- Department of Gastroenterology, Jilin Chemical Hospital, Jilin, China
| |
Collapse
|
11
|
Qian S, Xie F, Zhao H, Jiang T, Sang Y, Ye W, Liu Q, Cai D. Detection of chromosomal instability using ultrasensitive chromosomal aneuploidy detection in the diagnosis of precancerous lesions of gastric cancer. Front Genet 2024; 15:1359231. [PMID: 38660675 PMCID: PMC11040259 DOI: 10.3389/fgene.2024.1359231] [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: 12/21/2023] [Accepted: 03/22/2024] [Indexed: 04/26/2024] Open
Abstract
Background The diagnosis of Precancerous Lesions of Gastric Cancer (PLGC) is challenging in clinical practice. We conducted a clinical study by analyzing the information of relevant chromosome copy number variations (CNV) in the TCGA database followed by the UCAD technique to evaluate the value of Chromosomal Instability (CIN) assay in the diagnosis of PLGC. Methods Based on the screening of gastric cancer related data in TCGA database, CNV analysis was performed to explore the information of chromosome CNV related to gastric cancer. Based on the gastroscopic pathology results, 12 specimens of patients with severe atrophy were screened to analyze the paraffin specimens of gastric mucosa by UCAD technology, and to explore the influence of related factors on them. Results The results of CNV in TCGA database suggested that chromosome 7, 8, and 17 amplification was obvious in patients with gastric cancer. UCAD results confirmed that in 12 patients with pathologic diagnosis of severe atrophy, five of them had positive results of CIN, with a positive detection rate of 41.7%, which was mainly manifested in chromosome seven and chromosome eight segments amplification. We also found that intestinalization and HP infection were less associated with CIN. And the sensitivity of CIN measurement results was significantly better than that of tumor indicators. Conclusion The findings suggest that the diagnosis of PLGC can be aided by UCAD detection of CIN, of which Chr7 and 8 may be closely related to PLGC.
Collapse
Affiliation(s)
- Suting Qian
- Hangzhou Hospital of TCM Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Feifei Xie
- Hangzhou Hospital of TCM Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Haoyu Zhao
- Hangzhou Hospital of TCM Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Ting Jiang
- Hangzhou Hospital of TCM Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Yi Sang
- Hangzhou Hospital of TCM Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Wei Ye
- Hangzhou Hospital of TCM Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Qingsheng Liu
- Hangzhou Hospital of TCM Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Danli Cai
- Intensive Care Unit, The First Affiliated hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
| |
Collapse
|
12
|
Jiang T, Xu L, Qu X, Li R, Cheng Y, He H. Hsa_circ_0014606 Derived from Exosomes Promotes Gastric Carcinoma Tumorigenesis and Proliferation by Sponging miR-514b-3p to Upregulate HNRNPC. Dig Dis Sci 2024; 69:811-820. [PMID: 38217675 DOI: 10.1007/s10620-023-08254-z] [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: 10/25/2023] [Accepted: 12/14/2023] [Indexed: 01/15/2024]
Abstract
Gastric cancer is a common malignant tumor, and due to its insidious onset and limited screening methods, most patients are diagnosed with advanced disease and have a poor prognosis. The circRNA in exosomes has an essential role in cancer diagnosis and treatment. However, the part of hsa_circ_0014606 within exosomes in gastric cancer progression is unclear. Firstly, we extracted exosomes from the serum of gastric cancer patients and healthy individuals by ultracentrifugation and analyzed the expression of hsa_circ_0014606 in both exosomes; then knocked down hsa_circ_0014606 in vivo and in vitro, respectively, to observe its effect on the physiological function of gastric cancer cells; finally, we used bioinformatics to screen hsa_circ_0014606 targeting miRNAs and mRNAs, and experiments were performed to verify the interrelationship between the three. The results showed that the level of hsa_circ_0014606 in the serum exosomes of gastric cancer patients was significantly higher than that of the healthy population. The knockdown of hsa_circ_0014606 slowed the proliferation of gastric cancer cells, significantly reduced migration and invasion ability, accelerated apoptosis, and reduced tumor size in mice. In addition, the expression of hsa_circ_0014606 was negatively correlated with the expression of miR-514b-3p and positively correlated with the expression of heterogeneous nuclear ribonucleoprotein C (HNRNPC). In conclusion, hsa_circ_0014606 exerted a pro-cancer effect indirectly through miR-514b-3p targeting gene HNRNPC, and this study provides a new potential target for treating gastric cancer.
Collapse
Affiliation(s)
- Tao Jiang
- Department of Otolaryngology, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning, China
| | - Lingling Xu
- Department of Medical Oncology, The Second Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Xiaona Qu
- Department of Medical Oncology, The Second Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Rui Li
- Department of Otolaryngology, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning, China
| | - Ye Cheng
- Department of Medical Oncology, The Second Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Hongmei He
- Department of Medical Oncology, The Second Hospital of Dalian Medical University, Dalian, Liaoning, China.
| |
Collapse
|
13
|
Xiang T, Wei Z, Ye C, Liu G. Prognostic impact and immunotherapeutic implications of NETosis-related gene signature in gastric cancer patients. J Cell Mol Med 2024; 28:e18087. [PMID: 38146607 PMCID: PMC10902305 DOI: 10.1111/jcmm.18087] [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: 10/04/2023] [Revised: 12/01/2023] [Accepted: 12/11/2023] [Indexed: 12/27/2023] Open
Abstract
The role of NETosis and its related molecules remains unclear in gastric cancer. The data used in this study was directly downloaded from the Cancer Genome Atlas (TCGA) database. All analysis and plots are completed in R software using diverse R packages. In our study, we collected the list of NETosis-related genes from previous publications. Based on the list and expression profile of gastric cancer patients from the TCGA database, we identified the NETosis-related genes significantly correlated with patients survival. Then, CLEC6A, BST1 and TLR7 were identified through LASSO regression and multivariate Cox regression analysis for prognosis model construction. This prognosis model showed great predictive efficiency in both training and validation cohorts. We noticed that the high-risk patients might have a worse survival performance. Next, we explored the biological enrichment difference between high- and low-risk patients and found that many carcinogenic pathways were upregulated in the high-risk patients. Meanwhile, we investigated the genomic instability, mutation burden and immune microenvironment difference between high- and low-risk patients. Moreover, we noticed that low-risk patients were more sensitive to immunotherapy (85.95% vs. 56.22%). High-risk patients were more sensitive to some small molecules compounds like camptothecin_1003, cisplatin_1005, cytarabine_1006, nutlin-3a (-)_1047, gemcitabine_1190, WZ4003_1614, selumetinib_1736 and mitoxantrone_1810. In summary, our study comprehensively explored the role of NETosis-related genes in gastric cancer, which can provide direction for relevant studies.
Collapse
Affiliation(s)
- Tian Xiang
- Department of Clinical Laboratory CenterCentral Hospital of Enshi Tujia and Miao Autonomous PrefectureEnshiChina
| | | | - Chen Ye
- Hubei University of MedicineShiyanChina
| | - Gao Liu
- Department of Gastrointestinal SurgeryCentral Hospital of Enshi Tujia and Miao Autonomous PrefectureEnshiChina
| |
Collapse
|
14
|
Pei B, Zhao G, Geng Z, Wang Y, Wang M, Wang X, Xiong S, Zheng M. Identifying potential DNA methylation markers for the detection of esophageal cancer in plasma. Front Genet 2023; 14:1222617. [PMID: 37867599 PMCID: PMC10586502 DOI: 10.3389/fgene.2023.1222617] [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: 05/14/2023] [Accepted: 09/21/2023] [Indexed: 10/24/2023] Open
Abstract
Background: Esophageal cancer (EC) is a leading cause of cancer-related deaths in China, with the 5-year survival rate reaching less than 30%, because most cases were diagnosed and treated at the advanced stage. However, there is still a lack of low-cost, efficient, and accurate non-invasive methods for the early detection of EC at present. Methods: A total of 48 EC plasma and 101 control plasma samples were collected in a training cohort from 1 January 2021 to 31 December 2021, and seven cancer-related DNA methylation markers (ELMO1, ZNF582, FAM19A4, PAX1, C13orf18, JAM3 and TERT) were tested in these samples to select potential markers. In total, 20 EC, 10 gastric cancer (GC), 10 colorectal cancer (CRC), and 20 control plasma samples were collected in a validation cohort to evaluate the two-gene panel. Results: ZNF582, FAM19A4, JAM3, or TERT methylation in plasma was shown to significantly distinguish EC and control subjects (p < 0.05), and the combination of ZNF582 and FAM19A4 methylation was the two-gene panel that exhibited the best performance for the detection of EC with 60.4% sensitivity (95% CI: 45.3%-73.9%) and 83.2% specificity (95% CI: 74.1%-89.6%) in the training cohort. The performance of this two-gene panel showed no significant difference between different age and gender groups. When the two-gene panel was combined with CEA, the sensitivity for EC detection was further improved to 71.1%. In the validation cohort, the sensitivity of the two-gene panel for detecting EC, GC, and CRC was 60.0%, 30.0%, and 30.0%, respectively, with a specificity of 90.0%. Conclusion: The identified methylation marker panel provided a potential non-invasive strategy for EC detection, but further validation should be performed in more clinical centers.
Collapse
Affiliation(s)
- Bing Pei
- The Suqian Clinical College of Xuzhou Medical University, Suqian, China
- Department of Clinical Laboratory, The Affiliated Suqian First People’s Hospital of Nanjing Medical University, Suqian, China
| | - Guodong Zhao
- Zhejiang University Kunshan Biotechnology Laboratory, Zhejiang University Kunshan Innovation Institute, Kunshan, China
- Department of R&D, Suzhou VersaBio Technologies Co Ltd., Kunshan, China
| | - Zhixin Geng
- Department of Clinical Laboratory, The Affiliated Suqian First People’s Hospital of Nanjing Medical University, Suqian, China
| | - Yue Wang
- Department of R&D, Suzhou VersaBio Technologies Co Ltd., Kunshan, China
| | - Menglin Wang
- Department of Clinical Laboratory, The Affiliated Suqian First People’s Hospital of Nanjing Medical University, Suqian, China
| | - Xiaomei Wang
- Department of R&D, Suzhou VersaBio Technologies Co Ltd., Kunshan, China
| | - Shangmin Xiong
- Zhejiang University Kunshan Biotechnology Laboratory, Zhejiang University Kunshan Innovation Institute, Kunshan, China
- Department of R&D, Suzhou VersaBio Technologies Co Ltd., Kunshan, China
| | - Minxue Zheng
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, China
| |
Collapse
|
15
|
Qin Y, Geng JX, Huang B. Clinical value of serum pepsinogen in the diagnosis and treatment of gastric diseases. World J Gastrointest Oncol 2023; 15:1174-1181. [PMID: 37546552 PMCID: PMC10401465 DOI: 10.4251/wjgo.v15.i7.1174] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 05/28/2023] [Accepted: 06/14/2023] [Indexed: 07/12/2023] Open
Abstract
Pepsinogen, secreted from the gastric mucosa, is the precursor of pepsin. It is categorized as pepsinogen 1 and pepsinogen 2 based on its immunogenicity. The pepsinogen content that can enter the blood circulation through the capillaries of the gastric mucosa is approximately 1% and remains stable all the time. The pepsinogen content in serum will change with the pathological changes of gastric mucosa. Therefore, the level of pepsinogen in serum can play a role in serologic biopsy to reflect the function and morphology of different regions of gastric mucosa and serve as an indicator of gastric disease. This study conducts relevant research on serum pepsinogen 1, pepsinogen 2, and the ratio of pepsinogen 1 to pepsinogen 2, and reviews their important value in clinical diagnosis of Helicobacter pylori infection, gastric ulcer, and even gastric carcinoma, providing ideas for other researchers.
Collapse
Affiliation(s)
- Yuan Qin
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, Zhejiang Province, China
| | - Jia-Xin Geng
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, Zhejiang Province, China
| | - Biao Huang
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, Zhejiang Province, China
| |
Collapse
|
16
|
Škapars R, Gašenko E, Broza YY, Sīviņš A, Poļaka I, Bogdanova I, Pčolkins A, Veliks V, Folkmanis V, Lesčinska A, Liepniece-Karele I, Haick H, Rumba-Rozenfelde I, Leja M. Breath Volatile Organic Compounds in Surveillance of Gastric Cancer Patients following Radical Surgical Management. Diagnostics (Basel) 2023; 13:diagnostics13101670. [PMID: 37238155 DOI: 10.3390/diagnostics13101670] [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: 03/24/2023] [Revised: 04/20/2023] [Accepted: 05/07/2023] [Indexed: 05/28/2023] Open
Abstract
As of today, there is a lack of a perfect non-invasive test for the surveillance of patients for potential relapse following curative treatment. Breath volatile organic compounds (VOCs) have been demonstrated to be an accurate diagnostic tool for gastric cancer (GC) detection; here, we aimed to prove the yield of the markers in surveillance, i.e., following curative surgical management. Patients were sampled in regular intervals before and within 3 years following curative surgery for GC; gas chromatography-mass spectrometry (GC-MS) and nanosensor technologies were used for the VOC assessment. GC-MS measurements revealed a single VOC (14b-Pregnane) that significantly decreased at 12 months, and three VOCs (Isochiapin B, Dotriacontane, Threitol, 2-O-octyl-) that decreased at 18 months following surgery. The nanomaterial-based sensors S9 and S14 revealed changes in the breath VOC content 9 months after surgery. Our study results confirm the cancer origin of the particular VOCs, as well as suggest the value of breath VOC testing for cancer patient surveillance, either during the treatment phase or thereafter, for potential relapse.
Collapse
Affiliation(s)
- Roberts Škapars
- Institute of Clinical and Preventive Medicine, Faculty of Medicine, University of Latvia, LV-1586 Riga, Latvia
- Department of Abdominal and Soft Tissue Surgery, Oncology Center of Latvia, Riga East University Hospital, LV-1038 Riga, Latvia
| | - Evita Gašenko
- Institute of Clinical and Preventive Medicine, Faculty of Medicine, University of Latvia, LV-1586 Riga, Latvia
- Department of Abdominal and Soft Tissue Surgery, Oncology Center of Latvia, Riga East University Hospital, LV-1038 Riga, Latvia
| | - Yoav Y Broza
- Department of Chemical Engineering and Russel Berrie Nanotechnology Institute, Technion-Israel Institute of Technology, Haifa 3200003, Israel
| | - Armands Sīviņš
- Institute of Clinical and Preventive Medicine, Faculty of Medicine, University of Latvia, LV-1586 Riga, Latvia
- Department of Abdominal and Soft Tissue Surgery, Oncology Center of Latvia, Riga East University Hospital, LV-1038 Riga, Latvia
| | - Inese Poļaka
- Institute of Clinical and Preventive Medicine, Faculty of Medicine, University of Latvia, LV-1586 Riga, Latvia
| | - Inga Bogdanova
- Institute of Clinical and Preventive Medicine, Faculty of Medicine, University of Latvia, LV-1586 Riga, Latvia
- Department of Abdominal and Soft Tissue Surgery, Oncology Center of Latvia, Riga East University Hospital, LV-1038 Riga, Latvia
| | - Andrejs Pčolkins
- Institute of Clinical and Preventive Medicine, Faculty of Medicine, University of Latvia, LV-1586 Riga, Latvia
- Department of Abdominal and Soft Tissue Surgery, Oncology Center of Latvia, Riga East University Hospital, LV-1038 Riga, Latvia
| | - Viktors Veliks
- Institute of Clinical and Preventive Medicine, Faculty of Medicine, University of Latvia, LV-1586 Riga, Latvia
| | - Valdis Folkmanis
- Institute of Clinical and Preventive Medicine, Faculty of Medicine, University of Latvia, LV-1586 Riga, Latvia
| | - Anna Lesčinska
- Institute of Clinical and Preventive Medicine, Faculty of Medicine, University of Latvia, LV-1586 Riga, Latvia
- Department of Abdominal and Soft Tissue Surgery, Oncology Center of Latvia, Riga East University Hospital, LV-1038 Riga, Latvia
| | - Inta Liepniece-Karele
- Institute of Clinical and Preventive Medicine, Faculty of Medicine, University of Latvia, LV-1586 Riga, Latvia
- Department of Abdominal and Soft Tissue Surgery, Oncology Center of Latvia, Riga East University Hospital, LV-1038 Riga, Latvia
| | - Hossam Haick
- Department of Chemical Engineering and Russel Berrie Nanotechnology Institute, Technion-Israel Institute of Technology, Haifa 3200003, Israel
| | - Ingrīda Rumba-Rozenfelde
- Institute of Clinical and Preventive Medicine, Faculty of Medicine, University of Latvia, LV-1586 Riga, Latvia
| | - Mārcis Leja
- Institute of Clinical and Preventive Medicine, Faculty of Medicine, University of Latvia, LV-1586 Riga, Latvia
- Department of Abdominal and Soft Tissue Surgery, Oncology Center of Latvia, Riga East University Hospital, LV-1038 Riga, Latvia
| |
Collapse
|
17
|
Alrushaid N, Khan FA, Al-Suhaimi EA, Elaissari A. Nanotechnology in Cancer Diagnosis and Treatment. Pharmaceutics 2023; 15:1025. [PMID: 36986885 PMCID: PMC10052895 DOI: 10.3390/pharmaceutics15031025] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 03/07/2023] [Accepted: 03/10/2023] [Indexed: 03/30/2023] Open
Abstract
Traditional cancer diagnosis has been aided by the application of nanoparticles (NPs), which have made the process easier and faster. NPs possess exceptional properties such as a larger surface area, higher volume proportion, and better targeting capabilities. Additionally, their low toxic effect on healthy cells enhances their bioavailability and t-half by allowing them to functionally penetrate the fenestration of epithelium and tissues. These particles have attracted attention in multidisciplinary areas, making them the most promising materials in many biomedical applications, especially in the treatment and diagnosis of various diseases. Today, many drugs are presented or coated with nanoparticles for the direct targeting of tumors or diseased organs without harming normal tissues/cells. Many types of nanoparticles, such as metallic, magnetic, polymeric, metal oxide, quantum dots, graphene, fullerene, liposomes, carbon nanotubes, and dendrimers, have potential applications in cancer treatment and diagnosis. In many studies, nanoparticles have been reported to show intrinsic anticancer activity due to their antioxidant action and cause an inhibitory effect on the growth of tumors. Moreover, nanoparticles can facilitate the controlled release of drugs and increase drug release efficiency with fewer side effects. Nanomaterials such as microbubbles are used as molecular imaging agents for ultrasound imaging. This review discusses the various types of nanoparticles that are commonly used in cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Noor Alrushaid
- Department of Stem Cell Biology, Institute for Research and Medical Consultations, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia
- Univ. Lyon, University Claude Bernard Lyon-1, CNRS, ISA-UMR 5280, 69622 Lyon, France
| | - Firdos Alam Khan
- Department of Stem Cell Biology, Institute for Research and Medical Consultations, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia
| | - Ebtesam Abdullah Al-Suhaimi
- Biology Department, College of Science, Institute of Research & Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia
| | - Abdelhamid Elaissari
- Univ. Lyon, University Claude Bernard Lyon-1, CNRS, ISA-UMR 5280, 69622 Lyon, France
| |
Collapse
|
18
|
Rao Bommi J, Kummari S, Lakavath K, Sukumaran RA, Panicker LR, Marty JL, Yugender Goud K. Recent Trends in Biosensing and Diagnostic Methods for Novel Cancer Biomarkers. BIOSENSORS 2023; 13:398. [PMID: 36979610 PMCID: PMC10046866 DOI: 10.3390/bios13030398] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 03/13/2023] [Accepted: 03/15/2023] [Indexed: 06/18/2023]
Abstract
Cancer is one of the major public health issues in the world. It has become the second leading cause of death, with approximately 75% of cancer deaths transpiring in low- or middle-income countries. It causes a heavy global economic cost estimated at more than a trillion dollars per year. The most common cancers are breast, colon, rectum, prostate, and lung cancers. Many of these cancers can be treated effectively and cured if detected at the primary stage. Nowadays, around 50% of cancers are detected at late stages, leading to serious health complications and death. Early diagnosis of cancer diseases substantially increases the efficient treatment and high chances of survival. Biosensors are one of the potential screening methodologies useful in the early screening of cancer biomarkers. This review summarizes the recent findings about novel cancer biomarkers and their advantages over traditional biomarkers, and novel biosensing and diagnostic methods for them; thus, this review may be helpful in the early recognition and monitoring of treatment response of various human cancers.
Collapse
Affiliation(s)
| | - Shekher Kummari
- Department of Chemistry, Indian Institute of Technology Palakkad, Palakkad 678 557, Kerala, India
| | - Kavitha Lakavath
- Department of Chemistry, Indian Institute of Technology Palakkad, Palakkad 678 557, Kerala, India
| | - Reshmi A. Sukumaran
- Department of Chemistry, Indian Institute of Technology Palakkad, Palakkad 678 557, Kerala, India
| | - Lakshmi R. Panicker
- Department of Chemistry, Indian Institute of Technology Palakkad, Palakkad 678 557, Kerala, India
| | - Jean Louis Marty
- Université de Perpignan Via Domitia, 52 Avenue Paul Alduy, 66860 Perpignan, France
| | - Kotagiri Yugender Goud
- Department of Chemistry, Indian Institute of Technology Palakkad, Palakkad 678 557, Kerala, India
| |
Collapse
|
19
|
Gašenko E, Bogdanova I, Sjomina O, Aleksandraviča I, Kiršners A, Ancāns G, Rudzīte D, Vangravs R, Sīviņš A, Škapars R, Tzivian L, Polaka I, Folkmanis V, Leja M. Assessing the utility of pepsinogens and gastrin-17 in gastric cancer detection. Eur J Cancer Prev 2023:00008469-990000000-00046. [PMID: 36912185 DOI: 10.1097/cej.0000000000000791] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/14/2023]
Abstract
OBJECTIVES The aim of the study was to determine the proportion of gastric cancer patients with decreased levels of pepsinogen and gastrin-17 in plasma, with the goal of providing indirect evidence of the sensitivity of these biomarkers when applied in a cancer screening setting. METHODS The levels of pepsinogens I and II, gastrin-17, and Helicobacter pylori immunoglobulin antibodies in plasma samples of gastric cancer patients were evaluated using the GastroPanel test system (Biohit Oyj, Helsinki, Finland). A decreased level of the pepsinogen I/II ratio was defined as less than three, while a decrease in gastrin-17 was defined as less than 1 pmol/L. Univariate analysis using non-parametric tests was used to investigate differences between normal and low concentrations of biomarkers. RESULTS In total, 481 plasma samples from patients (59.9% male) with a median age of 64 years (ranging from 27 to 88 years) were analyzed. Out of the 400 cases of gastric cancer (83.2% of the total), 182 were categorized as the intestinal type, 141 as the diffuse type, 60 as the mixed type, and 17 as indeterminate according to the Lauren classification system. The H. pylori immunoglobulin test was positive in 74.0% of the patients. Pepsinogen I/II ratio was decreased in 32.4% (36.8% of the intestinal type); gastrin-17 in 12.3% (10.1% of the antral region) of all cases. CONCLUSION The majority of gastric cancer patients had normal levels of pepsinogen and gastrin-17, suggesting that these biomarkers have limited application as screening tools in the Caucasian population.
Collapse
Affiliation(s)
| | | | - Olga Sjomina
- Department of Internal Medicine, Riga East University Hospital
| | - Ilona Aleksandraviča
- Faculty of Medicine, Institute of Clinical and Preventive Medicine, University of Latvia, Riga
| | - Arnis Kiršners
- Faculty of Medicine, Institute of Clinical and Preventive Medicine, University of Latvia, Riga
| | - Guntis Ancāns
- Department of Surgery, Jēkabpils Regional Hospital, Jēkabpils
| | | | - Reinis Vangravs
- Faculty of Medicine, Institute of Clinical and Preventive Medicine, University of Latvia, Riga
| | - Armands Sīviņš
- Department of Abdominal and Soft Tissue Surgery, Clinic of Oncological Surgery
| | - Roberts Škapars
- Department of Abdominal and Soft Tissue Surgery, Clinic of Oncological Surgery
| | - Lilian Tzivian
- Faculty of Medicine, Institute of Clinical and Preventive Medicine, University of Latvia, Riga
| | - Inese Polaka
- Faculty of Medicine, Institute of Clinical and Preventive Medicine, University of Latvia, Riga
| | - Valdis Folkmanis
- Faculty of Medicine, Institute of Clinical and Preventive Medicine, University of Latvia, Riga
| | - Mārcis Leja
- Department of Research, Riga East University Hospital, Riga, Latvia
| |
Collapse
|
20
|
Poniewierska-Baran A, Warias P, Zgutka K. Sirtuins (SIRTs) As a Novel Target in Gastric Cancer. Int J Mol Sci 2022; 23:ijms232315119. [PMID: 36499440 PMCID: PMC9737976 DOI: 10.3390/ijms232315119] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 11/25/2022] [Accepted: 11/26/2022] [Indexed: 12/03/2022] Open
Abstract
Gastric cancer is a major health burden worldwide. Among all neoplasms, gastric cancer is the fifth most common and the third most deadly type of cancer. It is known that sirtuins (SIRTs), are NAD+-dependent histone deacetylases regulating important metabolic pathways. High expression of SIRTs in the human body can regulate metabolic processes; they prevent inflammation but also resist cell death and aging processes. The seven members of this family enzymes can also play a fundamental role in process of carcinogenesis by influencing cell viability, apoptosis and metastasis. This review collects and discusses the role of all seven sirtuins (SIRT1-SIRT7) in the pathogenesis of gastric cancer (GC).
Collapse
Affiliation(s)
- Agata Poniewierska-Baran
- Institute of Biology, University of Szczecin, Felczaka 3c, 71-412 Szczecin, Poland
- Correspondence:
| | - Paulina Warias
- Department of Physiology, Pomeranian Medical University in Szczecin, Powstancow Wielkopolskich 72, 70-111 Szczecin, Poland
| | - Katarzyna Zgutka
- Department of Physiology in Health Sciences, Faculty of Health Sciences, Pomeranian Medical University, Szczecin, Żołnierska 54, 70-210 Szczecin, Poland
| |
Collapse
|
21
|
Lorraine-Francis H, Newberry E, Aziz I. Diagnostic yield of upper gastrointestinal endoscopy in patients attending a UK centre with symptoms compatible with Rome IV functional dyspepsia. Frontline Gastroenterol 2022; 14:306-311. [PMID: 37409327 PMCID: PMC11138181 DOI: 10.1136/flgastro-2022-102268] [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: 07/16/2022] [Accepted: 11/11/2022] [Indexed: 11/22/2022] Open
Abstract
Background Dyspeptic symptoms are common and mainly due to functional dyspepsia (FD). The Rome IV criteria mandate a normal upper gastrointestinal (UGI) endoscopy before diagnosing FD. However, endoscopies are costly, resource-intensive procedures that generate substantial waste. Hence, simpler means of diagnosing FD are desirable. Objectives To determine what proportion of UGI endoscopies are represented by patients with symptoms compatible with Rome IV FD, and the diagnostic yield in this cohort stratified according to alarm features. Methods Adult patients attending a UK centre for outpatient UGI endoscopy completed a preprocedure questionnaire on demographics, medical history, alarm features, mood, somatisation and GI symptoms. Alarm features were defined as age ≥55 years, dysphagia, anaemia, unintentional weight loss, UGI bleed or a family history of UGI cancer. Clinically significant endoscopic findings were cancers, Barrett's oesophagus, erosive oesophagitis, peptic ulcers or strictures. Results Of 387 patients attending for an outpatient non-surveillance diagnostic UGI endoscopy, 221 had symptoms compatible with FD whereas 166 did not. Approximately 80% in both groups had alarm features, with a similar prevalence of clinically significant endoscopic findings at ~10%. UGI endoscopy was normal in a cohort of 9% (n=35) with symptoms compatible with FD and no alarm features, while benign peptic ulcer was noted in two of 29 cases without FD symptoms and no alarm features. Conclusion 1-in-10 UGI endoscopies are performed in patients with symptoms compatible with FD and no alarm features, in whom there is no diagnostic yield. We recommend such patients receive a positive diagnosis of FD without endoscopy.
Collapse
Affiliation(s)
- Hannah Lorraine-Francis
- Academic Department of Gastroenterology, Sheffield Teaching Hospitals, Sheffield, UK
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Ellen Newberry
- Academic Department of Gastroenterology, Sheffield Teaching Hospitals, Sheffield, UK
| | - Imran Aziz
- Academic Department of Gastroenterology, Sheffield Teaching Hospitals, Sheffield, UK
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| |
Collapse
|
22
|
Haque E, Esmail A, Muhsen I, Salah H, Abdelrahim M. Recent Trends and Advancements in the Diagnosis and Management of Gastric Cancer. Cancers (Basel) 2022; 14:5615. [PMID: 36428707 PMCID: PMC9688354 DOI: 10.3390/cancers14225615] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 11/10/2022] [Accepted: 11/10/2022] [Indexed: 11/17/2022] Open
Abstract
Gastric cancer is an enigmatic malignancy that has recently been shown to be increasing in incidence globally. There has been recent progress in emerging technologies for the diagnosis and treatment of the disease. Improvements in non-invasive diagnostic techniques with serological tests and biomarkers have led to decreased use of invasive procedures such as endoscopy. A multidisciplinary approach is used to treat gastric cancer, with recent significant advancements in systemic therapies used in combination with cytotoxic chemotherapies. New therapeutic targets have been identified and clinical trials are taking place to assess their efficacy and safety. In this review, we provide an overview of the current and emerging treatment strategies and diagnostic techniques for gastric cancer.
Collapse
Affiliation(s)
- Emaan Haque
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
| | - Abdullah Esmail
- Section of GI Oncology, Houston Methodist Neal Cancer Center, Houston, TX 77030, USA
| | - Ibrahim Muhsen
- Section of Hematology and Oncology, Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Haneen Salah
- Department of Pathology, Houston Methodist Hospital, Houston, TX 77030, USA
| | - Maen Abdelrahim
- Section of GI Oncology, Houston Methodist Neal Cancer Center, Houston, TX 77030, USA
- Cockrell Center for Advanced Therapeutic Phase I Program, Houston Methodist Research Institute, Houston, TX 77030, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY 10021, USA
| |
Collapse
|
23
|
High Expression of COL10A1 Is an Independent Predictive Poor Prognostic Biomarker and Associated with Immune Infiltration in Advanced Gastric Cancer Microenvironment. JOURNAL OF ONCOLOGY 2022; 2022:1463316. [PMID: 36276283 PMCID: PMC9584694 DOI: 10.1155/2022/1463316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 09/22/2022] [Accepted: 09/26/2022] [Indexed: 11/17/2022]
Abstract
Medical technology has become more and more sophisticated recently, which, however, fails to contribute to a better prognosis for patients suffering advanced gastric cancer (GC). Hence, new biomarkers specific to GC diagnosis and prognosis shall be identified urgently. This study screened differentially expressed genes (DEGs) between 375 GC samples and 32 paracancer tissue samples from TCGA datasets. The expression of Collagen type X alpha 1 (COL10A1) in GC was analyzed. The chi-square test assisted in analyzing the relevance of COL10A1 to the clinicopathologic characteristics. The Kaplan-Meier method helped to assess the survival curves and log-rank tests assisted in the examination of the differences. The Cox proportional hazard regression model served for analyzing the risk factors for GC. Then, we developed a nomogram that contained the COL10A1 expression and clinical information. Finally, how COL10A1 expression was associated with the immune infiltration was also evaluated. In this study, 7179 upregulated and 3771 downregulated genes were identified. Among them, COL10A1 expression was distinctly increased in GC specimens compared with nontumor specimens. High COL10A1 expression exhibited an obvious relation to tumor T and pathologic stage. ROC assays confirmed the diagnostic value of COL10A1 expression in screening GC samples from normal samples. Survival data displayed that patients with high COL10A1 expression exhibited a shorter OS and DSS than those with low COL10A1 expression. We obtained a predictive nomogram, which could better predict the COL10A1 expression by virtue of discrimination and calibration. The prognostic value of COL10A1 expression was further confirmed in GSE84426 datasets. Immune assays revealed that COL10A1 expression was associated with tumor-filtrating immune cells, like CD8 T cells, cytotoxic cells, DC, eosinophils, iDC, macrophages, mast cells, NK CD56dim cells, NK cells, pDC, T helper cells, Tem, Th1 cells, Th17 cells, and Treg. Overall, we firstly proved that COL10A1 may be a novel and valuable prognostic and diagnostic factor for GC patients. In addition, COL10A1 has potential to be an immune indicator in GC.
Collapse
|
24
|
Li Z, Song M, Han S, Jin C, Yang J. The prognostic role of circulating tumor cells in gastric cancer: A meta-analysis. Front Oncol 2022; 12:963091. [PMID: 36313657 PMCID: PMC9610107 DOI: 10.3389/fonc.2022.963091] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 08/29/2022] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVE We conducted a meta-analysis to evaluate the relationship between circulating tumor cells (CTC) and the prognosis of patients with gastric cancer. MATERIALS AND METHODS The cohort studies reporting on the relationship between CTC and prognosis of gastric cancer were collected from Pubmed, Cochrane, Embase, CNKI, WanFang Data, and VIP databases. The two researchers independently screened the literature, extracted the data, and evaluated the bias risk of the included literature. The data were analyzed by Revman software (Review Manager version 5.4). RESULT A total of 14 retrospective cohort studies with 1053 patients were included. The results showed that the overall survival time (OS) and progression-free survival time (PFS) of CTC-positive patients were shorter compared to CTC-negative patients. Taking into consideration the critical value of CTC positive patients, country of origin, sample size, treatment mode, and study time, the subgroup analysis showed that CTC-positive was related to the shortening of OS in patients with gastric cancer. Based on the subgroup analysis of the factors such as CTC positive critical value < 2.8, sample size ≥ 75, mixed therapy, longer study duration, country, and immunofluorescence detection of CTC, it was found that OS in CTC positive group was shorter than that in CTC-negative group (all P<0.05), while the critical value of positive CTC ≥ 2.8, sample size ≥ 75, choice of treatment only for operation or non-operation, short study time and molecular detection of CTC were not associated with OS (all P>0.05). In addition, CTC-positive patients had a more advanced TNM staging, poorer tumor differentiation, and earlier distant metastasis. CONCLUSION CTC can be used as a prognostic indicator of gastric cancer. Gastric cancer patients with positive CTC may have a poorer prognosis compared to those with CTC-negative tumors. SYSTEMATIC REVIEW REGISTRATION https://www.crd.york.ac.uk/PROSPERO/, identifier CRD42022323155.
Collapse
Affiliation(s)
- Zuxi Li
- The First Clinical School of Gansu University of Chinese Medicine, Lanzhou, China
- General Surgery Clinical Medical Center, Gansu Provincial Hospital, Lanzhou, China
- Gansu Provincial Key Laboratory of Molecular Diagnosis and Precision Therapy of Surgical Tumors, Gansu Provincial Hospital, Lanzhou, China
| | - Meijuan Song
- The First Clinical School of Gansu University of Chinese Medicine, Lanzhou, China
- General Surgery Clinical Medical Center, Gansu Provincial Hospital, Lanzhou, China
- Gansu Provincial Key Laboratory of Molecular Diagnosis and Precision Therapy of Surgical Tumors, Gansu Provincial Hospital, Lanzhou, China
| | - Shangjun Han
- The First Clinical School of Gansu University of Chinese Medicine, Lanzhou, China
- General Surgery Clinical Medical Center, Gansu Provincial Hospital, Lanzhou, China
- Gansu Provincial Key Laboratory of Molecular Diagnosis and Precision Therapy of Surgical Tumors, Gansu Provincial Hospital, Lanzhou, China
| | - Chuanwei Jin
- The First Clinical School of Gansu University of Chinese Medicine, Lanzhou, China
- General Surgery Clinical Medical Center, Gansu Provincial Hospital, Lanzhou, China
- Gansu Provincial Key Laboratory of Molecular Diagnosis and Precision Therapy of Surgical Tumors, Gansu Provincial Hospital, Lanzhou, China
| | - Jing Yang
- The First Clinical School of Gansu University of Chinese Medicine, Lanzhou, China
- General Surgery Clinical Medical Center, Gansu Provincial Hospital, Lanzhou, China
- Gansu Provincial Key Laboratory of Molecular Diagnosis and Precision Therapy of Surgical Tumors, Gansu Provincial Hospital, Lanzhou, China
| |
Collapse
|
25
|
Comparing Different Anesthesia Methods on Anesthetic Effect and Postoperative Pain in Patients with Early Gastric Cancer during Endoscopic Submucosal Dissection. JOURNAL OF ONCOLOGY 2022; 2022:7299360. [PMID: 36072967 PMCID: PMC9444400 DOI: 10.1155/2022/7299360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 07/18/2022] [Accepted: 08/06/2022] [Indexed: 11/17/2022]
Abstract
Endoscopic submucosal dissection (ESD) is a minimally invasive technique to completely peel the pathological mucosa from the submucosa under endoscopy, which has been often utilized to treat early gastric cancer. During the operation, anesthesia is required to reduce the discomfort due to the complexity, high risk, and longtime operation of ESD. In this study, we compared different anesthesia methods on anesthetic effect and postoperative pain in patients (≥65 years old) with early gastric cancer during ESD. For this purpose, 60 patients with early gastric cancer who were more than 65 years old were selected from January 2019 to December 2021, where 30 patients treated with simple intravenous general anesthesia were divided into the simple group and 30 patients treated with intravenous combined inhalation general anesthesia were regarded as the composite group. The hemodynamic index, wake-up time, postoperative pain intensity, operation time, and the incidence of adverse reactions were compared between the two groups. For the hemodynamic index before incision, after incision, and at the end of the operation, the mean arterial pressure (MAP) in the composite group was higher than that in the simple group (P < 0.05) and the heart rate (HR) was lower than that in the simple group (P < 0.05). After the ESD operation, the wake-up time and visual analogue scale (VAS) in the composite group were lower than those in the simple group (P < 0.05). In addition, the ESD operation time and incidence of adverse reactions in the composite group was significantly lower than that in the simple group (P < 0.05). These results showed that intravenous combined inhalation general anesthesia had a good anesthetic effect, stable hemodynamics during ESD operation, and slight postoperative pain.
Collapse
|
26
|
Molecular and Circulating Biomarkers of Gastric Cancer. Int J Mol Sci 2022; 23:ijms23147588. [PMID: 35886934 PMCID: PMC9322632 DOI: 10.3390/ijms23147588] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 07/07/2022] [Accepted: 07/07/2022] [Indexed: 01/27/2023] Open
Abstract
Gastric cancer (GC)—a common tumor that affects humans worldwide—is highly malignant with a poor prognosis. GC is frequently not diagnosed until a relatively advanced stage. Early detection and efficient monitoring of tumor dynamics are prerequisites for reducing disease burden and mortality. Minimally invasive methods are needed to establish a diagnosis or monitoring the response to treatment of gastric cancer. Blood-based biomarker assays for the detection of early-stage GC could be of great relevance both for the risk group or for population-wide based screening programs, The currently used tumor marker assays for detecting GC are simple and rapid, but their use is limited by their low sensitivity and specificity. In recent years, several markers have been identified and tested for their clinical relevance in the management of gastric cancer. Here we review the available literature on plasma classical tumor markers, circulating free microRNAs (cfmiRNAs), circulating cell-free DNA (cfDNA), circulating tumor cells (CTCs), autoantibodies against tumor associated antigens (TAAs), and circulating extracellular vesicles (EVs) for diagnosis and monitoring of gastric cancer. This review summarizes the present status and approaches for these biomarkers, which could be potentially used for early diagnosis and accurate prediction of therapeutic approaches. We also discuss the future perspective and challenges in the search for new biomarkers of gastric cancer.
Collapse
|
27
|
Ramirez-Garrastacho M, Bajo-Santos C, Line A, Martens-Uzunova ES, de la Fuente JM, Moros M, Soekmadji C, Tasken KA, Llorente A. Extracellular vesicles as a source of prostate cancer biomarkers in liquid biopsies: a decade of research. Br J Cancer 2022; 126:331-350. [PMID: 34811504 PMCID: PMC8810769 DOI: 10.1038/s41416-021-01610-8] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 10/19/2021] [Accepted: 10/21/2021] [Indexed: 01/02/2023] Open
Abstract
Prostate cancer is a global cancer burden and considerable effort has been made through the years to identify biomarkers for the disease. Approximately a decade ago, the potential of analysing extracellular vesicles in liquid biopsies started to be envisaged. This was the beginning of a new exciting area of research investigating the rich molecular treasure found in extracellular vesicles to identify biomarkers for a variety of diseases. Vesicles released from prostate cancer cells and cells of the tumour microenvironment carry molecular information about the disease that can be analysed in several biological fluids. Numerous studies document the interest of researchers in this field of research. However, methodological issues such as the isolation of vesicles have been challenging. Remarkably, novel technologies, including those based on nanotechnology, show promise for the further development and clinical use of extracellular vesicles as liquid biomarkers. Development of biomarkers is a long and complicated process, and there are still not many biomarkers based on extracellular vesicles in clinical use. However, the knowledge acquired during the last decade constitutes a solid basis for the future development of liquid biopsy tests for prostate cancer. These are urgently needed to bring prostate cancer treatment to the next level in precision medicine.
Collapse
Affiliation(s)
- Manuel Ramirez-Garrastacho
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | | | - Aija Line
- Latvian Biomedical Research and Study Centre, Riga, Latvia
| | - Elena S Martens-Uzunova
- Erasmus MC Cancer Institute, University Medical Center Rotterdam, Department of Urology, Laboratory of Experimental Urology, Erasmus MC, Rotterdam, The Netherlands
| | - Jesus Martinez de la Fuente
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-Universidad de Zaragoza, Zaragoza, Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
| | - Maria Moros
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-Universidad de Zaragoza, Zaragoza, Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
| | - Carolina Soekmadji
- Department of Cell and Molecular Biology, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- School of Biomedical Sciences, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Kristin Austlid Tasken
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Alicia Llorente
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway.
- Department for Mechanical, Electronics and Chemical Engineering, Oslo Metropolitan University, Oslo, Norway.
| |
Collapse
|
28
|
Jiang S, Gao H, He J, Shi J, Tong Y, Wu J. Machine learning: A non-invasive prediction method for gastric cancer based on a survey of lifestyle behaviors. Front Artif Intell 2022; 5:956385. [PMID: 36052291 PMCID: PMC9424643 DOI: 10.3389/frai.2022.956385] [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: 05/30/2022] [Accepted: 07/11/2022] [Indexed: 02/05/2023] Open
Abstract
Gastric cancer remains an enormous threat to human health. It is extremely significant to make a clear diagnosis and timely treatment of gastrointestinal tumors. The traditional diagnosis method (endoscope, surgery, and pathological tissue extraction) of gastric cancer is usually invasive, expensive, and time-consuming. The machine learning method is fast and low-cost, which breaks through the limitations of the traditional methods as we can apply the machine learning method to diagnose gastric cancer. This work aims to construct a cheap, non-invasive, rapid, and high-precision gastric cancer diagnostic model using personal behavioral lifestyles and non-invasive characteristics. A retrospective study was implemented on 3,630 participants. The developed models (extreme gradient boosting, decision tree, random forest, and logistic regression) were evaluated by cross-validation and the generalization ability in our test set. We found that the model developed using fingerprints based on the extreme gradient boosting (XGBoost) algorithm produced better results compared with the other models. The overall accuracy of which test set was 85.7%, AUC was 89.6%, sensitivity 78.7%, specificity 76.9%, and positive predictive values 73.8%, verifying that the proposed model has significant medical value and good application prospects.
Collapse
Affiliation(s)
- Siqing Jiang
- Department of Public Health, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Real-Doctor Artificial Intelligence Research Center, Zhejiang University, Hangzhou, China
| | - Haojun Gao
- Department of Public Health, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Real-Doctor Artificial Intelligence Research Center, Zhejiang University, Hangzhou, China
| | - Jiajin He
- Department of Public Health, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Jiaqi Shi
- Department of Public Health, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Real-Doctor Artificial Intelligence Research Center, Zhejiang University, Hangzhou, China
| | - Yuling Tong
- Department of General Practice/Health Management Center, School of Medicine, The Second Affiliated Hospital of Zhejiang University, Hangzhou, China
- Yuling Tong
| | - Jian Wu
- Department of Public Health, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Real-Doctor Artificial Intelligence Research Center, Zhejiang University, Hangzhou, China
- *Correspondence: Jian Wu
| |
Collapse
|
29
|
Advances in the Aetiology & Endoscopic Detection and Management of Early Gastric Cancer. Cancers (Basel) 2021; 13:cancers13246242. [PMID: 34944861 PMCID: PMC8699285 DOI: 10.3390/cancers13246242] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 12/06/2021] [Accepted: 12/10/2021] [Indexed: 12/24/2022] Open
Abstract
Simple Summary Gastric adenocarcinoma has remained a highly lethal disease. Awareness and recognition of preneoplastic conditions (including gastric atrophy and intestinal metaplasia) using high-resolution white-light endoscopy as well as chromoendoscopy is therefore essential. Helicobacter pylori, a class I carcinogen, remains the main contributor to the development of sporadic distal gastric neoplasia. Management of early gastric neoplasia with endoscopic resections should be in line with standard indications. A multidisciplinary approach to any case of an early gastric neoplasia is imperative. Hereditary forms of gastric cancer require a tailored approach and individua-lized surveillance. Abstract The mortality rates of gastric carcinoma remain high, despite the progress in research and development in disease mechanisms and treatment. Therefore, recognition of gastric precancerous lesions and early neoplasia is crucial. Two subtypes of sporadic gastric cancer have been recognized: cardia subtype and non-cardia (distal) subtype, the latter being more frequent and largely associated with infection of Helicobacter pylori, a class I carcinogen. Helicobacter pylori initiates the widely accepted Correa cascade, describing a stepwise progression through precursor lesions from chronic inflammation to gastric atrophy, gastric intestinal metaplasia and neoplasia. Our knowledge on He-licobacter pylori is still limited, and multiple questions in the context of its contribution to the pathogenesis of gastric neoplasia are yet to be answered. Awareness and recognition of gastric atrophy and intestinal metaplasia on high-definition white-light endoscopy, image-enhanced endoscopy and magnification endoscopy, in combination with histology from the biopsies taken accurately according to the protocol, are crucial to guiding the management. Standard indications for endoscopic resections (endoscopic mucosal resection and endoscopic submucosal dissection) of gastric dysplasia and intestinal type of gastric carcinoma have been recommended by multiple societies. Endoscopic evaluation and surveillance should be offered to individuals with an inherited predisposition to gastric carcinoma.
Collapse
|