The present study's objective is to examine, with meticulous detail, the publication patterns related to autophagy in pancreatic cancer (PC) by year, country, institution, journal, citation, and keyword, ultimately forecasting future research foci.
To identify publications, the Web of Science Core Collection was consulted. VOSviewer16.16 was used to scrutinize the contributions from diverse countries/regions, institutes, authors, notable research areas, and prospective future trends. CiteSpace66.R2 programs are a vital component. We further compiled clinical trials concerning PC, with a focus on autophagy.
A comprehensive analysis of autophagy in PC encompassed 1293 research papers, published between 2013 and 2023, which were included in this study. Articles had an average citation count of 3376. China produced the greatest number of publications, the USA coming second, and 50 influential articles were identified via co-citation analysis. Analysis of keyword clusters revealed that metabolic reprogramming, ER stress, mTOR-mediated apoptosis, and extracellular traps were among the most frequently observed groupings. biomarkers tumor Recent co-occurrence cluster analysis highlighted pancreatic stellate cells, autophagy-dependent ferroptosis, autophagy-related pathways, metabolic rewiring, and on-coding RNAs as prominent research areas of interest.
The past few years have witnessed a broader expansion of both research publications and areas of scholarly interest. PC autophagy research has seen notable advancements thanks to the impactful contributions from China and the USA. The current research hotspots not only investigate the modulation, metabolic reprogramming, and ferroptosis of tumor cells themselves, but also explore the tumor microenvironment's role, specifically autophagy in pancreatic stellate cells, and new treatments for targeting this process.
Publications and research interests have, in general, experienced a significant rise in number over the past few years. The United States and China have made significant contributions to research on programmed cell death, particularly in PC cells. Tumor cell modulation, metabolic reprogramming, and ferroptosis are key areas of current research interest, but research is also increasingly focused on tumor microenvironments, such as autophagy in pancreatic stellate cells, and novel treatments aimed at autophagy.
This study explored the clinical significance of a radiomics signature, specifically, its prognostic value in patients with gastric neuroendocrine neoplasms (GNEN).
The study retrospectively examined 182 GNEN patients, all of whom underwent dual-phase enhanced computed tomography. Feature selection and R-signature creation for the arterial, venous, and combined arteriovenous phases were achieved via LASSO-Cox regression analysis. adoptive immunotherapy The performance of the optimal R-signature in predicting overall survival (OS) was examined in the training data set and then verified in a separate validation data set. Clinicopathological factors influencing overall survival (OS) were investigated using univariate and multivariate Cox regression analyses. The performance of a radiomics-clinical nomogram was evaluated, this nomogram consolidates the R-signature with independent clinicopathological risk factors.
Predicting overall survival, the arteriovenous phase combined R-signature showed the most favorable results, outperforming both the independent arterial and venous phase R-signatures in terms of C-index (0.803 vs 0.784, and 0.803 vs 0.756, respectively; P<0.0001). A significant association between the optimal R-signature and OS was observed in both the training and validation cohorts. GNEN patients were classified into high and low prognostic risk groups using the median value of their radiomics scores. Selleck CCS-1477 A novel combined radiomics-clinical nomogram, encompassing an R-signature and independent clinicopathological factors (sex, age, treatment, tumor stage, lymph node involvement, distant metastasis, tumor boundaries, Ki67, and CD56), demonstrated substantially improved prognostic accuracy compared to the clinical nomogram, the R-signature alone, and the traditional TNM system, as indicated by the C-index (0.882 vs 0.861, 0.882 vs 0.803, and 0.882 vs 0.870, respectively; P<0.0001). The calibration curves displayed a substantial consistency between estimated and actual survival, further validated by decision curve analysis as demonstrating the usefulness of the combined radiomics-clinical nomogram in clinical practice.
Patients with GNEN can be stratified into high-risk and low-risk groups based on the R-signature's application. Furthermore, the radiomics-clinical nomogram's predictive power surpassed competing models, potentially assisting clinicians in treatment planning and patient support.
The R-signature offers a potential means of categorizing GNEN patients into high-risk and low-risk groups. In addition, the radiomics-clinical nomogram's predictive capability outperformed alternative models, potentially assisting clinicians in therapeutic decision-making and providing valuable patient guidance.
The prognosis for colorectal cancer (CRC) patients presenting with a BRAF mutation is generally very poor. A pressing need exists to pinpoint prognostic factors associated with BRAF-mutant colorectal cancers. RNF43, part of the ENF ubiquitin ligase family, is involved in the Wnt signaling cascade. In a variety of human cancers, the presence of RNF43 mutations is frequently observed. An insufficient number of studies have investigated RNF43's part in the etiology of CRC. The objective of this study was to investigate how RNF43 mutations affect molecular characteristics and the long-term outcome in BRAF-mutated colorectal cancers.
A retrospective examination of 261 samples from CRC patients with the BRAF mutation was performed. Matched peripheral blood samples and tumor tissue were subjected to targeted sequencing using a 1021-gene panel, focusing on cancer-related genes. Survival of patients was then assessed, considering the molecular characteristics that may have impacted it. From the cBioPortal dataset, 358 CRC patients carrying a BRAF mutation were selected for further validation.
This study was spurred by a compelling case of a CRC patient, whose remission reached 70% and whose progression-free survival extended to 13 months, in the context of BRAF V600E and RNF43 co-mutation. Genomic research indicated that RNF43 mutations played a role in altering the genomic characteristics of patients with a BRAF mutation, specifically affecting microsatellite instability (MSI), tumor mutation burden (TMB), and the prevalence of common gene mutations. Survival analysis in BRAF-mutant colorectal cancer (CRC) demonstrated that RNF43 mutations are a predictive biomarker for a more favorable outcome in terms of progression-free survival (PFS) and overall survival (OS).
The collective impact of RNF43 mutations on genomic characteristics was found to be linked to improved clinical outcomes in BRAF-mutant colorectal cancer patients.
In our collective analysis, RNF43 mutations were linked to favorable genomic characteristics, ultimately improving clinical outcomes for BRAF-mutant CRC patients.
The grim reality of colorectal cancer is the annual death toll of hundreds of thousands worldwide, an unfortunately projected rise in incidence anticipated within the next two decades. In the context of metastasis, the availability of cytotoxic therapies is constrained, resulting in a minimal enhancement of survival outcomes for patients. Consequently, the investigation has transitioned to recognizing the mutation patterns within colorectal cancers and the design of therapeutic interventions specifically targeting them. Based on actionable molecular alterations and genetic profiles, this review examines up-to-date systemic treatment strategies for metastatic colorectal cancer.
A study was undertaken to analyze the correlation between the creatinine/cystatin C ratio and progression-free survival (PFS) and overall survival (OS) in colorectal cancer (CRC) patients who received surgical care.
In a retrospective study, 975 colorectal cancer (CRC) patients who underwent surgical resection between January 2012 and 2015 were examined. A three-sample curve, designed to be restricted, was employed to demonstrate the non-linear correlation between the creatinine-cystatin C ratio and PFS/OS. Using the Kaplan-Meier method in conjunction with a Cox regression model, researchers investigated the relationship between the creatinine-cystatin C ratio and the survival of colorectal cancer (CRC) patients. Prognostic variables demonstrating a p-value of 0.05 in multivariate statistical models were incorporated into the construction of prognostic nomograms. A receiver operating characteristic curve analysis was conducted to compare the comparative efficacy of prognostic nomograms and the traditional pathological stage.
Colorectal cancer (CRC) patients exhibiting a negative correlation between creatinine/cystatin C ratio and adverse progression-free survival (PFS) were observed. Individuals exhibiting a low creatinine/cystatin C ratio demonstrated significantly reduced progression-free survival (PFS) and overall survival (OS) compared to those with a high ratio. PFS was observed to be 508% versus 639% (p = 0.0002), while OS was 525% versus 689% (p < 0.0001). Analysis of various factors in CRC patients demonstrated that a low creatinine/cystatin C ratio was associated with an increased risk of diminished progression-free survival (hazard ratio [HR] = 1.286, 95% confidence interval [CI] = 1.007–1.642, p = 0.0044) and a shorter overall survival time (hazard ratio [HR] = 1.410, 95% confidence interval [CI] = 1.087–1.829, p = 0.0010). Nomograms utilizing the creatinine/cystatin C ratio display predictive strength, supported by a concordance index surpassing 0.7, facilitating the prediction of the 1-5 year prognosis.
The creatinine/cystatin C ratio might serve as a useful prognostic indicator for predicting progression-free survival and overall survival in colorectal cancer patients, contributing to pathological staging and, alongside tumor markers, facilitating in-depth prognostic stratification in this patient population.