Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 21.342
Filtrer
1.
Support Care Cancer ; 32(8): 501, 2024 Jul 10.
Article de Anglais | MEDLINE | ID: mdl-38985345

RÉSUMÉ

OBJECTIVE: This study aims to use structural equation modeling to explore the pathways and effect sizes of factors influencing the adherence of postoperative patients with digestive tract tumor to oral nutritional supplements, providing a theoretical basis for future nursing intervention measures. METHODS: A total of 300 postoperative patients with digestive tract tumor within 30 days after surgery were conveniently sampled. Surveys were conducted using a General Information Questionnaire, Morisky Medication Adherence Scale, Digestive System Tumor Patient Nutrition Knowledge-Attitude-Behavior Questionnaire, Multidimensional Social Perception Scale, Beliefs about Medical Questionnaire, and General Self-Efficacy Scale. Structural equation modeling was employed to analyze the factors and pathways affecting adherence with oral nutritional supplements. RESULTS: The adherence score of postoperative patients with digestive tract tumor to oral nutritional supplements was 1.61 ± 1.38. The structural equation model had a good fit (χ2/df = 2.685, GFI = 0.930, CFI = 0.913, AGFI = 0.887, IFI = 0.915, and RMSEA = 0.075). Nutrition knowledge, social support, medication beliefs, and self-efficacy were found to be factors influencing adherence with oral nutritional supplements in postoperative patients with digestive tract tumor, with total effects of 0.539, 0.264, 0.215, and 0.180, respectively. Nutrition knowledge indirectly affected adherence through self-efficacy and medication beliefs, while social support indirectly affected adherence through self-efficacy. CONCLUSION: Adherence with oral nutritional supplements in postoperative patients with digestive tract tumor is at a low level. Improving social support, enhancing patients nutrition knowledge, increasing self-efficacy, and strengthening medication beliefs are effective ways to improve patient adherence.


Sujet(s)
Compléments alimentaires , Connaissances, attitudes et pratiques en santé , Adhésion au traitement médicamenteux , Auto-efficacité , Humains , Études transversales , Femelle , Mâle , Adulte d'âge moyen , Adhésion au traitement médicamenteux/statistiques et données numériques , Enquêtes et questionnaires , Sujet âgé , Adulte , Tumeurs de l'appareil digestif/chirurgie , Tumeurs de l'appareil digestif/psychologie , Soutien social , Tumeurs gastro-intestinales/chirurgie , Tumeurs gastro-intestinales/psychologie , Période postopératoire
2.
J Cancer Res Clin Oncol ; 150(7): 331, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38951269

RÉSUMÉ

OBJECTIVE: To conduct a systematic review and meta-analysis of case-control and cohort human studies evaluating metabolite markers identified using high-throughput metabolomics techniques on esophageal cancer (EC), cancer of the gastroesophageal junction (GEJ), and gastric cancer (GC) in blood and tissue. BACKGROUND: Upper gastrointestinal cancers (UGC), predominantly EC, GEJ, and GC, are malignant tumour types with high morbidity and mortality rates. Numerous studies have focused on metabolomic profiling of UGC in recent years. In this systematic review and meta-analysis, we have provided a collective summary of previous findings on metabolites and metabolomic profiling associated with EC, GEJ and GC. METHODS: Following the PRISMA procedure, a systematic search of four databases (Embase, PubMed, MEDLINE, and Web of Science) for molecular epidemiologic studies on the metabolomic profiles of EC, GEJ and GC was conducted and registered at PROSPERO (CRD42023486631). The Newcastle-Ottawa Scale (NOS) was used to benchmark the risk of bias for case-controlled and cohort studies. QUADOMICS, an adaptation of the QUADAS-2 (Quality Assessment of Diagnostic Accuracy) tool, was used to rate diagnostic accuracy studies. Original articles comparing metabolite patterns between patients with and without UGC were included. Two investigators independently completed title and abstract screening, data extraction, and quality evaluation. Meta-analysis was conducted whenever possible. We used a random effects model to investigate the association between metabolite levels and UGC. RESULTS: A total of 66 original studies involving 7267 patients that met the required criteria were included for review. 169 metabolites were differentially distributed in patients with UGC compared to healthy patients among 44 GC, 9 GEJ, and 25 EC studies including metabolites involved in glycolysis, anaerobic respiration, tricarboxylic acid cycle, and lipid metabolism. Phosphatidylcholines, eicosanoids, and adenosine triphosphate were among the most frequently reported lipids and metabolites of cellular respiration, while BCAA, lysine, and asparagine were among the most commonly reported amino acids. Previously identified lipid metabolites included saturated and unsaturated free fatty acids and ketones. However, the key findings across studies have been inconsistent, possibly due to limited sample sizes and the majority being hospital-based case-control analyses lacking an independent replication group. CONCLUSION: Thus far, metabolomic studies have provided new opportunities for screening, etiological factors, and biomarkers for UGC, supporting the potential of applying metabolomic profiling in early cancer diagnosis. According to the results of our meta-analysis especially BCAA and TMAO as well as certain phosphatidylcholines should be implicated into the diagnostic procedure of patients with UGC. We envision that metabolomics will significantly enhance our understanding of the carcinogenesis and progression process of UGC and may eventually facilitate precise oncological and patient-tailored management of UGC.


Sujet(s)
Métabolomique , Humains , Métabolomique/méthodes , Tumeurs de l'oesophage/sang , Tumeurs de l'oesophage/métabolisme , Tumeurs de l'estomac/sang , Tumeurs de l'estomac/métabolisme , Tumeurs de l'estomac/diagnostic , Marqueurs biologiques tumoraux/sang , Marqueurs biologiques tumoraux/métabolisme , Tumeurs gastro-intestinales/sang , Tumeurs gastro-intestinales/métabolisme , Tumeurs gastro-intestinales/diagnostic , Métabolome/physiologie , Études cas-témoins , Jonction oesogastrique/anatomopathologie , Jonction oesogastrique/métabolisme
3.
Cancer Med ; 13(13): e7452, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38953401

RÉSUMÉ

BACKGROUND: Sarcopenic obesity (SO) in patients with gastrointestinal cancer is associated with a poor prognosis. We aimed to investigate the prognostic impact of SO in patients with gastrointestinal cancer, as well as the diagnostic cut-off value of SO in patients with gastrointestinal cancer among Chinese population. METHODS: We conducted a consecutive cohort study. Between January 2017 and January 2019, 289 patients diagnosed with gastrointestinal cancer were included in our study. Skeletal muscle area, total fat area, and subcutaneous fat area were measured by CT scan. All patients were followed up for 5 years. Receiver operating characteristic curves (ROC) were adopted to determine the cut-off values of visceral fat obesity for the prediction of sarcopenia. Based on the cut-off values, patients with sarcopenia combined with visceral fat obesity were divided into the SO group, and the others were divided into the non-sarcopenic obesity (NSO) group. Kaplan-Meier curves and univariate and multivariate Cox proportional hazard models were employed to explore the associations of body composition profiles with 5-year overall survival and disease-specific survival. RESULTS: Obtained from Youden's Index for ROC for the prediction of 5-year survival, skeletal muscle mass index (SMI) ≤40.02 cm2/m2 with VFA ≥ 126.30 cm2 in men and SMI ≤32.05 cm2/m2 with VFA ≥72.42 cm2 in women indicate a risk of poor prognosis in patients diagnosed with gastrointestinal cancer. Patients with SO had poorer 5-year overall survival (OS) than patients with NSO (6.74% vs. 82.84%, p < 0.001), and poorer 5-year DFS (6.74% vs. 81.82%, p < 0.001). In multivariate analysis, we found that the long-term mortality risk was approximately 13-fold higher among patients in the SO group compared to those with no conditions. CONCLUSIONS: Preoperative assessment of SO is useful not only for monitoring nutritional status but also for predicting 5-year OS in gastrointestinal cancer patients.


Sujet(s)
Tumeurs gastro-intestinales , Obésité , Sarcopénie , Humains , Sarcopénie/imagerie diagnostique , Mâle , Femelle , Tumeurs gastro-intestinales/mortalité , Tumeurs gastro-intestinales/complications , Tumeurs gastro-intestinales/anatomopathologie , Pronostic , Adulte d'âge moyen , Obésité/complications , Sujet âgé , Composition corporelle , Courbe ROC , Muscles squelettiques/imagerie diagnostique , Muscles squelettiques/physiopathologie , Muscles squelettiques/anatomopathologie , Estimation de Kaplan-Meier , Graisse intra-abdominale/imagerie diagnostique , Graisse intra-abdominale/physiopathologie
4.
Oncotarget ; 15: 424-438, 2024 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-38953895

RÉSUMÉ

Single-agent TAS102 (trifluridine/tipiracil) and regorafenib are FDA-approved treatments for metastatic colorectal cancer (mCRC). We previously reported that regorafenib combined with a fluoropyrimidine can delay disease progression in clinical case reports of multidrug-resistant mCRC patients. We hypothesized that the combination of TAS102 and regorafenib may be active in CRC and other gastrointestinal (GI) cancers and may in the future provide a treatment option for patients with advanced GI cancer. We investigated the therapeutic effect of TAS102 in combination with regorafenib in preclinical studies employing cell culture, colonosphere assays that enrich for cancer stem cells, and in vivo. TAS102 in combination with regorafenib has synergistic activity against multiple GI cancers in vitro including colorectal and gastric cancer, but not liver cancer cells. TAS102 inhibits colonosphere formation and this effect is potentiated by regorafenib. In vivo anti-tumor effects of TAS102 plus regorafenib appear to be due to anti-proliferative effects, necrosis and angiogenesis inhibition. Growth inhibition by TAS102 plus regorafenib occurs in xenografted tumors regardless of p53, KRAS or BRAF mutations, although more potent tumor suppression was observed with wild-type p53. Regorafenib significantly inhibits TAS102-induced angiogenesis and microvessel density in xenografted tumors, as well inhibits TAS102-induced ERK1/2 activation regardless of RAS or BRAF status in vivo. TAS102 plus regorafenib is a synergistic drug combination in preclinical models of GI cancer, with regorafenib suppressing TAS102-induced increase in microvessel density and p-ERK as contributing mechanisms. The TAS102 plus regorafenib drug combination may be further tested in gastric and other GI cancers.


Sujet(s)
Association médicamenteuse , Synergie des médicaments , Tumeurs gastro-intestinales , Mutation , Cellules souches tumorales , Néovascularisation pathologique , Phénylurées , Protéines proto-oncogènes B-raf , Protéines proto-oncogènes p21(ras) , Pyridines , Pyrrolidines , Facteur de transcription STAT-3 , Thymine , Trifluorothymidine , Uracile , Tests d'activité antitumorale sur modèle de xénogreffe , Humains , Trifluorothymidine/pharmacologie , Phénylurées/pharmacologie , Animaux , Pyridines/pharmacologie , Protéines proto-oncogènes B-raf/génétique , Protéines proto-oncogènes B-raf/métabolisme , Protéines proto-oncogènes p21(ras)/génétique , Protéines proto-oncogènes p21(ras)/métabolisme , Néovascularisation pathologique/traitement médicamenteux , Néovascularisation pathologique/génétique , Néovascularisation pathologique/métabolisme , Cellules souches tumorales/effets des médicaments et des substances chimiques , Cellules souches tumorales/métabolisme , Cellules souches tumorales/anatomopathologie , Tumeurs gastro-intestinales/traitement médicamenteux , Tumeurs gastro-intestinales/génétique , Tumeurs gastro-intestinales/anatomopathologie , Tumeurs gastro-intestinales/métabolisme , Uracile/pharmacologie , Uracile/analogues et dérivés , Souris , Facteur de transcription STAT-3/métabolisme , Facteur de transcription STAT-3/génétique , Thymine/pharmacologie , Lignée cellulaire tumorale , Pyrrolidines/pharmacologie , Pyrrolidines/usage thérapeutique , Protocoles de polychimiothérapie antinéoplasique/pharmacologie , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Transduction du signal/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques ,
5.
Med Sci Monit ; 30: e943551, 2024 Jul 03.
Article de Anglais | MEDLINE | ID: mdl-38959178

RÉSUMÉ

BACKGROUND Gastrointestinal diffuse large B-cell lymphoma (GI-DLBCL) is the most common histological subtype of extra-nodal DLBCL, but the risk factors, prognostic biomarkers, histopathological classifications, and treatment strategies have not had significant progress. Emerging evidence shows that cystatin SN (CST1) is involved in tumor progression in several cancer types, but its role in GI-DLBCL has not been revealed. MATERIAL AND METHODS We established a cohort consisting of 84 patients with GI-DLBCL who underwent surgical resection. The expression of CST1 in the cohort was investigated by immunohistochemistry, which divided the patients into subgroups with low or high expression of CST1. Moreover, the CST1 expression in GI-DLBCL tissues or adjacent GI tissues were compared with RT-qPCR. The correlation between CST1 expression and clinicopathological factors was analyzed with the chi-square test. The prognostic significance of CST1 was estimated by univariate and multivariate analysis, and statistical significance was analyzed with the log-rank test. RESULTS CST1 was aberrantly upregulated in GI-DLBCL tissues compared with in non-tumor GI tissues. High expression of CST1 indicated poor prognosis of GI-DLBCL (P=0.012), and CST1 can be regarded as an independent prognostic biomarker of GI-DLBCL (hazard ratio=3.07). In our study, serum lactate dehydrogenase (P=0.002), performance status (P=0.003), Lugano stage (P=0.002), and International Prognostic Index (P=0.001) were also prognostic factors of GI-DLBCL. CONCLUSIONS CST1 is an independent prognostic biomarker of GI-DLBCL, indicating unfavorable prognosis. Our results suggested that CST1 detection can be a promising method to stratify high-risk patients and guide individual treatment.


Sujet(s)
Marqueurs biologiques tumoraux , Tumeurs gastro-intestinales , Lymphome B diffus à grandes cellules , Humains , Mâle , Femelle , Lymphome B diffus à grandes cellules/génétique , Lymphome B diffus à grandes cellules/métabolisme , Lymphome B diffus à grandes cellules/anatomopathologie , Marqueurs biologiques tumoraux/métabolisme , Marqueurs biologiques tumoraux/génétique , Pronostic , Adulte d'âge moyen , Tumeurs gastro-intestinales/métabolisme , Tumeurs gastro-intestinales/anatomopathologie , Tumeurs gastro-intestinales/génétique , Sujet âgé , Adulte , Cystatines salivaires/métabolisme , Cystatines salivaires/génétique , Immunohistochimie , Études de cohortes
6.
BMC Cancer ; 24(1): 828, 2024 Jul 11.
Article de Anglais | MEDLINE | ID: mdl-38992597

RÉSUMÉ

BACKGROUND: Most gastrointestinal stromal tumors (GISTs) harbor c-KIT or PDGFRA mutations. Administration of tyrosine kinase inhibitors (TKIs) has significantly improved the survival of patients with GISTs. We aimed to evaluate the clinical outcome of advanced or recurrent GIST patients in Taiwan. METHODS: Patients diagnosed between 2010 and 2020 were enrolled. The collected data included baseline characteristics, treatment pattern, treatment outcome, genetic aberrations and survival status. Progression-free survival (PFS) and overall survival (OS) were analyzed and plotted with the Kaplan-Meier method. Cox regression analysis was used to analyze the prognostic factors of survival. RESULTS: A total of 224 patients with advanced or recurrent GISTs treated with TKIs were enrolled. All patients received imatinib treatment. Ninety-three and 42 patients received sunitinib and regorafenib treatment, respectively. The 48-month PFS and OS rates for patients treated with imatinib were 50.5% and 79.5%, respectively. c-KIT exon 9 and PDGFRA mutations were prognostic factors for a poor PFS and PDGFRA mutation was a prognostic factor for a poor OS in patients treated with imatinib in multivariate Cox regression analysis. The median PFS of patients who received sunitinib treatment was 12.76 months (95% confidence interval (CI), 11.01-14.52). Patients with c-KIT exon 9 mutations had a longer PFS than those with other genetic aberrations. The median PFS of patients treated with regorafenib was 7.14 months (95% CI, 3.39-10.89). CONCLUSIONS: We present real-world clinical outcomes for advanced GIST patients treated with TKIs and identify mutational status as an independent prognostic factor for patient survival.


Sujet(s)
Tumeurs stromales gastro-intestinales , Mutation , Récidive tumorale locale , Inhibiteurs de protéines kinases , Protéines proto-oncogènes c-kit , Récepteur au PDGF alpha , Enregistrements , Humains , Tumeurs stromales gastro-intestinales/traitement médicamenteux , Tumeurs stromales gastro-intestinales/mortalité , Tumeurs stromales gastro-intestinales/génétique , Tumeurs stromales gastro-intestinales/anatomopathologie , Femelle , Mâle , Taïwan/épidémiologie , Adulte d'âge moyen , Inhibiteurs de protéines kinases/usage thérapeutique , Sujet âgé , Protéines proto-oncogènes c-kit/génétique , Adulte , Récepteur au PDGF alpha/génétique , Récidive tumorale locale/traitement médicamenteux , Récidive tumorale locale/génétique , Sunitinib/usage thérapeutique , Mésilate d'imatinib/usage thérapeutique , Pronostic , Sujet âgé de 80 ans ou plus , Tumeurs gastro-intestinales/traitement médicamenteux , Tumeurs gastro-intestinales/mortalité , Tumeurs gastro-intestinales/génétique , Tumeurs gastro-intestinales/anatomopathologie , Phénylurées/usage thérapeutique , Pyridines/usage thérapeutique , Taux de survie , Survie sans progression , Estimation de Kaplan-Meier
8.
Front Immunol ; 15: 1428529, 2024.
Article de Anglais | MEDLINE | ID: mdl-38994371

RÉSUMÉ

Background: Immune checkpoint inhibitors (ICIs) have revolutionized gastrointestinal cancer treatment, yet the absence of reliable biomarkers hampers precise patient response prediction. Methods: We developed and validated a genomic mutation signature (GMS) employing a novel artificial intelligence network to forecast the prognosis of gastrointestinal cancer patients undergoing ICIs therapy. Subsequently, we explored the underlying immune landscapes across different subtypes using multiomics data. Finally, UMI-77 was pinpointed through the analysis of drug sensitization data from the Genomics of Drug Sensitivity in Cancer (GDSC) database. The sensitivity of UMI-77 to the AGS and MKN45 cell lines was evaluated using the cell counting kit-8 (CCK8) assay and the plate clone formation assay. Results: Using the artificial intelligence network, we developed the GMS that independently predicts the prognosis of gastrointestinal cancer patients. The GMS demonstrated consistent performance across three public cohorts and exhibited high sensitivity and specificity for 6, 12, and 24-month overall survival (OS) in receiver operating characteristic (ROC) curve analysis. It outperformed conventional clinical and molecular features. Low-risk samples showed a higher presence of cytolytic immune cells and enhanced immunogenic potential compared to high-risk samples. Additionally, we identified the small molecule compound UMI-77. The half-maximal inhibitory concentration (IC50) of UMI-77 was inversely related to the GMS. Notably, the AGS cell line, classified as high-risk, displayed greater sensitivity to UMI-77, whereas the MKN45 cell line, classified as low-risk, showed less sensitivity. Conclusion: The GMS developed here can reliably predict survival benefit for gastrointestinal cancer patients on ICIs therapy.


Sujet(s)
Tumeurs gastro-intestinales , Immunothérapie , Mutation , Humains , Tumeurs gastro-intestinales/génétique , Tumeurs gastro-intestinales/immunologie , Tumeurs gastro-intestinales/traitement médicamenteux , Tumeurs gastro-intestinales/thérapie , Pronostic , Lignée cellulaire tumorale , Immunothérapie/méthodes , Marqueurs biologiques tumoraux/génétique , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Intelligence artificielle , Mâle , Femelle
9.
Front Immunol ; 15: 1375730, 2024.
Article de Anglais | MEDLINE | ID: mdl-39007138

RÉSUMÉ

Solid gastrointestinal tumors often respond poorly to immunotherapy for the complex tumor microenvironment (TME), which is exacerbated by immune system alterations. Immunosenescence is the process of increased diversification of immune genes due to aging and other factors, leading to a decrease in the recognition function of the immune system. This process involves immune organs, immune cells, and the senescence-associated secretory phenotype (SASP). The most fundamental change is DNA damage, resulting in TME remodeling. The main manifestations are worsening inflammation, increased immunosuppressive SASP production, decreased immune cell antitumor activity, and the accumulation of tumor-associated fibroblasts and myeloid-derived suppressor cells, making antitumor therapy less effective. Senotherapy strategies to remove senescent cells and block key senescence processes can have synergistic effects with other treatments. This review focuses on immunoenescence and its impact on the solid TME. We characterize the immunosenescent TME and discuss future directions for antitumor therapies targeting senescence.


Sujet(s)
Tumeurs gastro-intestinales , Immunosénescence , Microenvironnement tumoral , Humains , Tumeurs gastro-intestinales/immunologie , Tumeurs gastro-intestinales/thérapie , Microenvironnement tumoral/immunologie , Immunosénescence/immunologie , Animaux , Immunothérapie/méthodes , Phénotype sécrétoire associé à la sénescence/immunologie , Vieillissement de la cellule/immunologie
10.
Nutrients ; 16(13)2024 Jun 23.
Article de Anglais | MEDLINE | ID: mdl-38999742

RÉSUMÉ

BACKGROUND: Ultra-Processed Foods (UPFs) are increasingly consumed worldwide, even in regions with strong dietary traditions like the Mediterranean and can play a crucial role in the development of chronic diseases, including cancer. This population-based prospective cohort study investigates the association between UPF consumption and gastrointestinal cancers and other causes of mortality in Southern Italy. METHODS: Data were collected from 4870 participants in the MICOL and NUTRIHEP cohorts. The EPIC questionnaire was used to elicit information on food and drink consumption and UPFs were categorized by degree of processing according to the NOVA classification. Cox proportional hazards regression and competing risk models were employed for statistical analysis. RESULTS: UPF consumption was positively associated with all-cause mortality: participants in the 3rd UFP quartile, as compared to the lowest, had a 27% higher risk of death (SHR 1.27 95% CI, 1.03; 1.57), while in the highest quartile as compared to the lowest, the risk was 34% higher (SHR 1.34 95% CI, 1.00; 1.79). Higher UPFs intake was also correlated with an increased gastrointestinal cancers mortality risk, especially the 2nd (SHR 1.65, 95% CI: 1.01; 2.71) and 4th quartile (SHR 3.14 95% CI: 1.56; 6.32), with a dose-dependent effect. For the other cancers, a SHR 1.61 (95% CI 1.03; 2.54) was observed for the 3rd quartile. CONCLUSIONS: Our results reinforce the link between UPF consumption and cancer risk, emphasizing the urgent need for interventions targeting dietary patterns.


Sujet(s)
Aliments de restauration rapide , Tumeurs gastro-intestinales , Humains , Italie/épidémiologie , Femelle , Mâle , Adulte d'âge moyen , Tumeurs gastro-intestinales/mortalité , Tumeurs gastro-intestinales/étiologie , Tumeurs gastro-intestinales/épidémiologie , Facteurs de risque , Études prospectives , Aliments de restauration rapide/effets indésirables , Aliments de restauration rapide/statistiques et données numériques , Sujet âgé , Adulte , Manipulation des aliments , Modèles des risques proportionnels , Régime alimentaire/effets indésirables , Régime alimentaire/statistiques et données numériques , Enquêtes et questionnaires , Aliments transformés
11.
Eur J Oncol Nurs ; 71: 102649, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38954929

RÉSUMÉ

PURPOSE: Physical activity (PA) is beneficial but difficult to maintain during chemotherapy. This pilot RCT explored the feasibility of the MI-Walk intervention-an 8-week motivational enhancement therapy- and home-based brisk walking intervention-among gastrointestinal (GI) cancer survivors receiving chemotherapy. METHODS: Sixty stage II-IV GI cancer survivors were recruited from 5 sites at their second infusion visit. Participants were randomized to receive PA education alone or the MI-Walk intervention: motivational enhancement therapy consisting of 3 motivational interviewing and self-efficacy-enhancing counseling sessions, a Fitbit Charge 2, exercise diaries, telephone follow-up, scripted motivational email messages, and optional weekly walking groups. RESULTS: The enrollment and completion rates were 62% and 90%, respectively. The MI-Walk participants (n = 29; mean age = 56.79, SD = 11.72; 97% white; 79% male) reported a baseline moderate-vigorous PA duration of 250.93 (SD = 636.52) min/wk. The mean MI-Walk Intervention acceptability score was 50.32 (SD = 12.02) on a scale of 14-70. Mean Fitbit and counseling helpfulness scores on a 5-point scale were 3.67 (SD = 1.43) and 3.44 (SD = 1.36), respectively. Participants' Fitbit moderate-vigorous PA 8-week averages ranged from 0 to 716.88 min/wk; 64% of participants adhered to ≥127 min/wk. Several characteristics (e.g., age, comorbidity, PA level, employment status, BMI, education level, gender, symptoms) were associated with enrollment, attrition, and intervention acceptability and adherence (p < 0.05). CONCLUSION: Enrollment and retention were adequate. The Fitbit and counseling were the most helpful. Acceptability and adherence varied based on participant characteristics; therefore, intervention tailoring and further research among cancer survivors less physically active at baseline and most in need of complex exercise intervention are needed. CLINICALTRIALS: gov NCT03515356.


Sujet(s)
Survivants du cancer , Exercice physique , Études de faisabilité , Tumeurs gastro-intestinales , Humains , Femelle , Mâle , Adulte d'âge moyen , Survivants du cancer/psychologie , Survivants du cancer/statistiques et données numériques , Tumeurs gastro-intestinales/traitement médicamenteux , Sujet âgé , Projets pilotes , Traitement par les exercices physiques/méthodes , Entretien motivationnel/méthodes , Antinéoplasiques , Adulte , Marche à pied
13.
PLoS One ; 19(7): e0304359, 2024.
Article de Anglais | MEDLINE | ID: mdl-39018292

RÉSUMÉ

To explore the application efficacy and significance of deep learning in anesthesia management for gastrointestinal tumors (GITs) surgery, 80 elderly patients with GITs who underwent surgical intervention at our institution between January and September 2021 were enrolled. According to the preoperative anesthesia management methodology, patients were rolled into a control (Ctrl) group (using 10 mg dexamethasone 1-2 hours before surgery) and an experimental (Exp) group (using a deep learning-based anesthesia monitoring system on the basis of the Ctrl group), with 40 cases in each group. A comprehensive comparative analysis was performed between the two cohorts, encompassing postoperative cognitive evaluations, Montreal Cognitive Assessment (MoCA) scores, gastrointestinal functionality, serum biomarkers (including interleukin (IL)-6, C-reactive protein (CRP), and cortisol levels), length of hospitalization, incidence of complications, and other pertinent metrics. The findings demonstrated that anesthesia monitoring facilitated by deep learning algorithms effectively assessed the anesthesia state of patients. Compared to the Ctrl group, patients in the Exp group showed significant differences in cognitive assessments (word recall, number connection, number coding) (P<0.05). Additionally, the Exp group exhibited a notably increased MoCA score (25.3±2.4), significantly shorter time to first flatus postoperatively (35.8±13.7 hours), markedly reduced postoperative pain scores, significantly shortened time to tolerate a liquid diet postoperatively (19.6±5.2 hours), accelerated recovery of serum-related indicators, and a significantly decreased mean length of hospital stay (11.4±3.2 days) compared to the Ctrl group. In summary, administering dexamethasone under the anesthesia management of GITs surgery based on gradient boosting decision tree (GBDT) and pharmacokinetics pharmacodynamics (PKPD) models can promote patient recovery, reduce the incidence of postoperative cognitive impairment (POCD), and improve patient prognosis.


Sujet(s)
Apprentissage profond , Dexaméthasone , Tumeurs gastro-intestinales , Humains , Dexaméthasone/administration et posologie , Dexaméthasone/usage thérapeutique , Femelle , Mâle , Sujet âgé , Tumeurs gastro-intestinales/chirurgie , Sujet âgé de 80 ans ou plus , Anesthésie/méthodes , Complications postopératoires/prévention et contrôle , Durée du séjour , Cognition/effets des médicaments et des substances chimiques
14.
Sci Rep ; 14(1): 16689, 2024 Jul 19.
Article de Anglais | MEDLINE | ID: mdl-39030227

RÉSUMÉ

Associations between psychiatric disorders and digestive tract cancers have been proposed. However, the causal link between these factors remains unclear. This study pioneers Mendelian randomization (MR) analysis to explore the genetic link between psychiatric disorders and digestive tract cancers risk. We analysed data on six psychiatric disorders [schizophrenia, bipolar disorder, major depressive disorder (MDD), attention deficit hyperactivity disorder, autism spectrum disorder, and panic disorder (PD)] and digestive tract cancers [esophagus cancer (EC), gastric cancer (GC), and colorectal cancer (CRC)] from genome-wide association studies databases. Using instrumental variables identified from significant single nucleotide polymorphism associations, we employed the inverse variance weighted (IVW) method alongside the weighted median (WM) method and MR-Egger regression. The results revealed no causal link between psychiatric disorders and the risk of EC or GC. Psychiatric disorders were not identified as risk factors for CRC. Notably, PD demonstrated a lower CRC risk (OR = 0.79, 95% CI 0.66-0.93, P = 0.01). This MR analysis underscores the lack of a causal association between psychiatric disorders and digestive tract cancers risk while suggesting a potential protective effect of PD against CRC.


Sujet(s)
Étude d'association pangénomique , Analyse de randomisation mendélienne , Troubles mentaux , Polymorphisme de nucléotide simple , Humains , Troubles mentaux/génétique , Troubles mentaux/épidémiologie , Facteurs de risque , Prédisposition génétique à une maladie , Tumeurs gastro-intestinales/génétique , Tumeurs gastro-intestinales/épidémiologie , Tumeurs de l'appareil digestif/génétique , Tumeurs de l'appareil digestif/épidémiologie , Tumeurs de l'appareil digestif/étiologie , Trouble dépressif majeur/génétique , Trouble dépressif majeur/épidémiologie
15.
Front Cell Infect Microbiol ; 14: 1392129, 2024.
Article de Anglais | MEDLINE | ID: mdl-39035354

RÉSUMÉ

Helicobacter pylori (H. pylori) is a harmful bacterium that is difficult to conveniently diagnose and effectively eradicate. Chronic H. pylori infection increases the risk of gastrointestinal diseases, even cancers. Despite the known findings, more underlying mechanisms are to be deeply explored to facilitate the development of novel prevention and treatment strategies of H. pylori infection. Long noncoding RNAs (lncRNAs) are RNAs with more than 200 nucleotides. They may be implicated in cell proliferation, inflammation and many other signaling pathways of gastrointestinal cancer progression. The dynamic expression of lncRNAs indicates their potential to be diagnostic or prognostic biomarkers. In this paper, we comprehensively summarize the processes of H. pylori infection and the treatment methods, review the known findings of lncRNA classification and functional mechanisms, elucidate the roles of lncRNAs in H. pylori-related gastrointestinal cancer, and discuss the clinical perspectives of lncRNAs.


Sujet(s)
Tumeurs gastro-intestinales , Infections à Helicobacter , Helicobacter pylori , ARN long non codant , ARN long non codant/génétique , ARN long non codant/métabolisme , Humains , Helicobacter pylori/génétique , Helicobacter pylori/pathogénicité , Tumeurs gastro-intestinales/microbiologie , Tumeurs gastro-intestinales/génétique , Infections à Helicobacter/microbiologie , Infections à Helicobacter/complications , Transduction du signal
16.
BMC Cancer ; 24(1): 870, 2024 Jul 19.
Article de Anglais | MEDLINE | ID: mdl-39030476

RÉSUMÉ

BACKGROUND: Population-based cancer registries (PBCRs) are the primary source of information for cancer surveillance and monitoring. Currently, there are 30 active PBCRs in Brazil. The objective of this study was to analyze the data quality of five gastrointestinal cancers (esophagus, stomach, colorectal, liver, and pancreas) according to the criteria of comparability, validity, completeness, and timeliness in Brazilian cancer registries. METHODS: This study included data from Brazilian PBCRs with more than ten years of historical data starting in the year 2000, regardless of the type of defined geographical coverage (state, metropolitan region, or capital), totaling 16 registries. Brazilian PBCRs were evaluated based on four international data quality criteria: comparability, validity (accuracy), completeness, and timeliness. All cancer cases were analyzed, except for nonmelanoma skin cancer cases (C44) and five gastrointestinal tumors (esophageal cancer, stomach cancer, colorectal cancer, liver cancer, and pancreatic cancer) per cancer registry and sex, according to the available period. RESULTS: The 16 Brazilian PBCRs represent 17% of the population (36 million inhabitants in 2021) according to data from 2000 to 2018. There was a variation in the incidence in the historical series ranging from 12 to 19 years. The proportion of morphologically verified (MV%) cases varied from 74.3% (Manaus) to 94.8% (Aracaju), and the proportion of incidentally reported death certificate only (DCO%) cases varied from 3.0% (São Paulo) to 23.9% (Espírito Santo). High-lethality malignant neoplasms, such as liver and pancreas, had DCO percentages greater than 30% in most cancer registries. The sixteen registries have more than a 48-month delay in data release compared to the 2022 calendar year. CONCLUSION: The studied Brazilian cancer registries met international comparability criteria; however, half of the registries showed indices below the expected levels for validity and completeness criteria for high-lethality tumors such as liver and pancreas tumors, in addition to a long delay in data availability and disclosure. Significant efforts are necessary to ensure the operational and stability of the PBCR in Brazil, which continues to be a tool for monitoring cancer incidence and assessing national cancer control policies.


Sujet(s)
Exactitude des données , Tumeurs gastro-intestinales , Enregistrements , Humains , Enregistrements/statistiques et données numériques , Brésil/épidémiologie , Tumeurs gastro-intestinales/épidémiologie , Mâle , Femelle , Incidence , Tumeurs du pancréas/épidémiologie , Surveillance de la population
17.
BMJ Open ; 14(7): e084827, 2024 Jul 20.
Article de Anglais | MEDLINE | ID: mdl-39032931

RÉSUMÉ

OBJECTIVE: This study aimed to compare the effects of patient-controlled intravenous analgesia (PCIA) with and without low-basal infusion on postoperative hypoxaemia. DESIGN: A randomised parallel-group non-inferiority trial. SETTING: The trial was conducted at a grade-A tertiary hospital from December 2021 to August 2022. PARTICIPANTS: 160 adults undergoing gastrointestinal tumour surgery and receiving postoperative PCIA. INTERVENTIONS: Participants randomly received a low-basal (0.1 mg/hour of hydromorphone) or no-basal infusion PCIA for postoperative 48 hours. PRIMARY AND SECONDARY OUTCOME MEASURES: Primary outcome was area under curve (AUC) per hour for hypoxaemia, defined as pulse oxygen saturation (SpO2) <95%. Secondary outcomes included: AUC per hour at SpO2<90% and <85%, hydromorphone consumption, ambulation time and analgesic outcomes up to 48 hours after surgery. RESULTS: Among 160 randomised patients, 159 completed the trial. An intention-to-treat analysis showed that AUC per hour (SpO2<95%) was greater in the low-basal infusion group compared with the no-basal infusion group, with a median difference of 0.097 (95% CI 0.001 to 0.245). Non-inferiority (margin: ratio of means (ROM) of 1.25) was not confirmed since the ROM between the two groups was 2.146 (95% CI 2.138 to 2.155). Hydromorphone consumption was higher in the low-basal group than in the no-basal group (median: 5.2 mg versus 1.6 mg, p<0.001). Meanwhile, there were no differences in the AUC values at the other two hypoxaemia thresholds, in ambulation time, or pain scores between the groups. CONCLUSIONS: Among the patients receiving hydromorphone PCIA after gastrointestinal tumour resection, low-basal infusion was inferior to no-basal infusion PCIA for postoperative hypoxaemia at SpO2<95% up to 48 hours after surgery. TRIAL REGISTRATION NUMBER: ChiCTR2100054317.


Sujet(s)
Analgésie autocontrôlée , Analgésiques morphiniques , Hydromorphone , Hypoxie , Douleur postopératoire , Humains , Hydromorphone/administration et posologie , Mâle , Femelle , Adulte d'âge moyen , Hypoxie/prévention et contrôle , Hypoxie/étiologie , Analgésie autocontrôlée/méthodes , Analgésiques morphiniques/administration et posologie , Analgésiques morphiniques/usage thérapeutique , Douleur postopératoire/traitement médicamenteux , Douleur postopératoire/prévention et contrôle , Sujet âgé , Perfusions veineuses , Tumeurs gastro-intestinales/chirurgie , Tumeurs gastro-intestinales/complications , Adulte
18.
Curr Oncol ; 31(6): 3329-3341, 2024 Jun 13.
Article de Anglais | MEDLINE | ID: mdl-38920736

RÉSUMÉ

Early integrated palliative care (EIPC) for patients with advanced cancers requires the involvement of family doctors (FDs) and oncologists. We compared attitudes between patients and their providers regarding the delivery of EIPC. Patients with newly diagnosed incurable gastrointestinal (GI) cancer at a tertiary cancer centre in Ontario, Canada, were surveyed using a study-specific instrument regarding the importance of and preferences for accessing support across eight domains of palliative care. Physicians within the circle of care completed a parallel survey for each patient. The concordance between patient and physician responses was analyzed. A total of 66 patients were surveyed (median age 69, 35% female). All had an oncologist, 12% had a specialist palliative care provider (SPC), and 97% had an FD, but only 41% listed the FD as part of the care team. In total, 95 providers responded (oncologist = 68, FD = 21, SPC = 6; response rate 92%; 1-3 physician responses per patient). Disease management and physical concerns were most important to patients. Patients preferred to access care in these domains from oncologists or SPCs. For all other domains, most patients attributed primary responsibility to self or family rather than any healthcare provider. Thus, concordance was poor between patient and physician responses. Across most domains of palliative care, we found low agreement between cancer patients and their physicians regarding responsibilities for care, with FDs appearing to have limited involvement at this stage.


Sujet(s)
Tumeurs gastro-intestinales , Soins palliatifs , Humains , Soins palliatifs/méthodes , Femelle , Mâle , Tumeurs gastro-intestinales/thérapie , Tumeurs gastro-intestinales/psychologie , Sujet âgé , Adulte d'âge moyen , Études prospectives , Enquêtes et questionnaires , Préférence des patients , Attitude du personnel soignant , Sujet âgé de 80 ans ou plus , Adulte , Ontario
19.
Curr Oncol ; 31(6): 3513-3528, 2024 Jun 17.
Article de Anglais | MEDLINE | ID: mdl-38920742

RÉSUMÉ

In controlled phase II trials, major prognostic factors need to be well balanced between arms. The main procedures used are SPBR (Stratified Permuted Block Randomization) and minimization. First, we provide a systematic review of the treatment allocation procedure used in gastrointestinal oncology controlled phase II trials published in 2019. Second, we performed simulations using data from six phase II studies to measure the impacts of imbalances and bias on the efficacy estimations. From the 40 articles analyzed, all mentioned randomization in both the title and abstract, the median number of patients included was 109, and 77.5% were multicenter. Of the 27 studies that reported at least one stratification variable, 10 included the center as a stratification variable, 10 used minimization, 9 used SBR, and 8 were unspecified. In real data studies, the imbalance increased with the number of centers. The total and marginal imbalances were higher with SBR than with minimization, and the difference increased with the number of centers. The efficiency estimates per arm were close to the original trial estimate in both procedures. Minimization is often used in cases of numerous centers and guarantees better similarity between arms for stratification variables for total and marginal imbalances in phase II trials.


Sujet(s)
Essais cliniques de phase II comme sujet , Humains , Essais cliniques de phase II comme sujet/méthodes , Pronostic , Essais contrôlés randomisés comme sujet , Tumeurs gastro-intestinales/traitement médicamenteux , Plan de recherche , Tumeurs de l'appareil digestif/traitement médicamenteux
20.
PLoS One ; 19(6): e0305994, 2024.
Article de Anglais | MEDLINE | ID: mdl-38924054

RÉSUMÉ

BACKGROUND: The association between red meat, fish, and processed meat consumption and the risk of developing gastrointestinal (GI) cancers remains inconclusive despite several investigations. Therefore, we conducted a systematic review and meta-analysis of observational studies to update the existing scientific evidence. METHODS: We searched PubMed, Web of Science, and Scopus databases until May 20, 2023. We analyzed observational studies that examined the associations between red and processed meat and fish consumption and GI cancers. We assessed between-study heterogeneity using the χ2 and τ2 tests, as well as I2 statistics. We explored the likelihood of publication bias using Begg's and Egger's tests and trim-and-fill analysis. We reported the overall effect sizes as odds ratios (ORs) with a 95% confidence interval (CI) using a random-effects model. RESULTS: Of the 21,004 studies identified, 95 studies involving 5,794,219 participants were included in the meta-analysis. The consumption of high levels of red meat, as compared to low levels, was found to significantly increase the risk of developing esophageal, pancreatic, liver, colon, rectal, and colorectal cancers. Similarly, the consumption of high levels of processed meat, as compared to low levels, significantly increased the risk of pancreatic, colon, rectal, and colorectal cancers. In contrast, the consumption of high levels of fish, as compared to low levels, significantly reduced the risk of colon, rectal, and colorectal cancers. CONCLUSIONS: This meta-analysis provides updated evidence on the association between red meat, processed meat, and fish consumption and the risk of developing five major types of GI cancers.


Sujet(s)
Tumeurs gastro-intestinales , Études observationnelles comme sujet , Viande rouge , Humains , Tumeurs gastro-intestinales/étiologie , Tumeurs gastro-intestinales/épidémiologie , Viande rouge/effets indésirables , Animaux , Poissons , Produits carnés/effets indésirables , Facteurs de risque , Viande/effets indésirables , Produits de la mer , Régime alimentaire/effets indésirables
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE