Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
Add more filters

Country/Region as subject
Affiliation country
Publication year range
1.
Pancreatology ; 24(5): 771-778, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38853072

ABSTRACT

BACKGROUND: Pancreatic ductal adenocarcinoma (PDAC) is the digestive malignancy with poor prognosis, and there is still a lack of effective diagnostic biomarkers. OBJECTIVE: We aimed to explore the diagnostic efficiency of DNA methylation in peripheral blood monocytes (PBMCs) in PDAC. METHODS: 850K BeadChips were used to detect genome-wide methylation of PBMCs. For the selected sites, MethylTarget assays was used for further verification. The support vector machine was used to establish the combined panel. RESULTS: A total of 167 PDAC patients and 113 healthy controls were included in this study and were divided into three sets. In the discovery set, we found 4625 differentially methylated positions (DMPs) between cancer group and healthy controls. ZFHX3 (0.16 ± 0.04 vs. 0.18 ± 0.04, P = 0.001), cg01904886 (0.84 ± 0.05 vs. 0.81 ± 0.04, P = 0.02) and NUMBL (0.96 ± 0.005 vs. 0.957 ± 0.005, P = 0.04) were found to be significantly different in training set. The locus with more significant differences, namely ZFHX3, was used for further validation and to establish a combined diagnostic panel with CA19-9. In the validation set, the ROC curve indicated that the AUC value of ZFHX3 was 0.75. The AUC value of the combined model (AUC = 0.92) was higher than that of CA19-9 alone (AUC = 0.88). In patients with normal CA19-9 levels, the ZFHX3 methylation biomarker still maintained good diagnostic efficacy (AUC = 0.71). CONCLUSION: Our study preliminarily suggests that ZFHX3 methylation combined with CA19-9 can improve the detection rate of PDAC. Especially in patients with normal CA19-9, ZFHX3 methylation can maintain stable diagnostic efficacy. The diagnostic value of ZFHX3 methylation still needs to be prospectively validated.


Subject(s)
Biomarkers, Tumor , Carcinoma, Pancreatic Ductal , DNA Methylation , Monocytes , Pancreatic Neoplasms , Humans , Pancreatic Neoplasms/diagnosis , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/blood , Biomarkers, Tumor/blood , Biomarkers, Tumor/genetics , Male , Female , Middle Aged , Monocytes/metabolism , Aged , Carcinoma, Pancreatic Ductal/diagnosis , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/blood , Homeodomain Proteins/genetics , Case-Control Studies
2.
Pancreatology ; 23(2): 204-212, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36710224

ABSTRACT

OBJECTIVES: High-grade gastro-enteropancreatic neuroendocrine neoplasms (GEP-NENs) are a heterogeneous group of rare tumors of two different types: well differentiated neuroendocrine tumors grade 3 (NETs G3) and poorly differentiated neuroendocrine carcinomas (NECs). This study aimed to explore the value of eight common preoperative markers in differentiating NETs G3 from NECs and the prognosis prediction of high-grade GEP-NENs. METHODS: Seventy-two patients diagnosed with high-grade GEP-NENs who underwent surgery at our institution were recruited for this study. Demographic and clinicopathological characteristics, preoperative serum tumor markers, and survival data were collected and analyzed. Kaplan-Meier methods were used to analyze survival rates, and a Cox regression model was used to perform multivariate analyses. RESULTS: Serum carcinoembryonic antigen (CEA) was dramatically higher in NECs than in NETs G3 (P = 0.025). After follow-up, 57 of the 72 patients remained for survival analysis. Elevated serum carbohydrate antigen 19-9 (CA19-9), CEA, cancer antigen 125 and sialic acid (SA) levels indicated poorer survival of high-grade GEP-NEN patients. Only CA19-9 (HR: 6.901, 95% CI: 1.843 to 25.837, P = 0.004) was regarded as an independent risk factor for overall survival. Serum CA19-9 (HR: 4.689, 95% CI: 1.127 to 19.506, P = 0.034) was also regarded as an independent factor for overall survival in NECs. CONCLUSIONS: Serum CEA levels can be used to distinguish NETs G3 from NECs. Preoperative CA19-9, CEA, cancer antigen 125 and SA levels have predictive value in the prognosis of high-grade GEP-NENs. Preoperative CA19-9, neuron-specific enolase, and SA levels can predict the prognosis of NECs.


Subject(s)
Carcinoma, Neuroendocrine , Intestinal Neoplasms , Neuroendocrine Tumors , Pancreatic Neoplasms , Stomach Neoplasms , Humans , Prognosis , Biomarkers, Tumor , CA-19-9 Antigen , CA-125 Antigen , Carcinoembryonic Antigen , Pancreatic Neoplasms/pathology , Neuroendocrine Tumors/pathology
3.
Biochem Biophys Res Commun ; 632: 1-9, 2022 12 03.
Article in English | MEDLINE | ID: mdl-36191371

ABSTRACT

OBJECTIVE: This study aimed to determine the role of COL17A1 in tumor progression and predict the prognosis of pancreatic cancer (PC). METHODS: RNA-seq data from The Cancer Genome Atlas and Genotype-Tissue Expression were analyzed using bioinformatics methods. "Limma" package was used to screen differentially expressed genes (DEGs). Prognostic-associated data were further analyzed using univariate Cox regression and verified using the GSE28375 and GSE62452 datasets. Protein-protein interaction (PPI) network analysis was integrated to screen for hub genes. In vitro quantitative real-time PCR (qPCR) and western blotting were used to detect gene expression. The functional attributes of PC cells were verified by wound healing assays, migration and invasion assays, Cell Counting Kit 8 (CCK8), and 5-ethynyl-2'-deoxyuridine (EdU) assay. RESULTS: On analyzing PC data, 4637 DEGs were identified. Of these, 2399 genes were upregulated and 2238 were downregulated. Through PPI network analysis, we identified that COL17A1 expression was highly correlated with poor prognosis of patients with PC. Functional attribute assays in the in vitro study showed that COL17A1 knockdown inhibited PC cell proliferation, migration, and invasion. CONCLUSIONS: According to our results, COL17A1 promotes PC cell proliferation, migration, and invasion mediated by the epithelial-mesenchymal transition (EMT) pathway. Thus, COL17A1 could be used as a prognostic marker in PC.


Subject(s)
Pancreatic Neoplasms , Humans , Cell Movement/genetics , Cell Line, Tumor , Pancreatic Neoplasms/diagnosis , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Epithelial-Mesenchymal Transition/genetics , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , Pancreatic Neoplasms
4.
Ann Surg Oncol ; 27(10): 4017-4025, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32144621

ABSTRACT

BACKGROUND: Many inflammatory markers can be used for the prognostication of pancreatic cancer, but which combination of inflammatory factors may be the best remains unclear. This study focused on the potential feasibility of the newly discovered C-reactive protein (CRP)/lymphocyte ratio (CLR) as a prognostic biomarker for patients with pancreatic cancer. METHODS: The study enrolled 997 patients with pancreatic cancer. Six combinations of inflammatory markers, namely, the neutrophil/lymphocyte ratio (NLR), the platelet/lymphocyte ratio (PLR), the CRP/albumin ratio (CAR), the neutrophil/albumin ratio (NAR), the platelet/albumin ratio (PAR), and CLR, were examined to determine which combination offers the highest accuracy for predicting poor survival by receiver operating characteristic curve analysis. The prognostic value of the CLR was analyzed by uni- and multivariate analyses. RESULTS: The newly developed CLR was more accurate than the NLR, PLR, CAR, NAR, and PAR in predicting survival. The optimal cutoff value for the CLR was calculated to be 1.8 for survival. A CLR higher than 1.8 was associated with poor survival in both the univariate (hazard ratio [HR] 2.00; P < 0.001) and multivariate (HR 1.73; P < 0.001) analyses. In addition, a CLR higher than 1.8 was an independent risk factor for patients with stage 2 (HR 1.85; P = 0.001), stage 3 (HR 1.83; P = 0.001), or stage 4 (HR 1.70; P < 0.001) disease. CONCLUSIONS: Pretreatment CLR can be considered a feasible biomarker for the prognostic prediction of pancreatic cancer. An elevated CLR was an independent risk factor for poor survival, with a cutoff value of 1.8.


Subject(s)
Pancreatic Neoplasms , C-Reactive Protein , Humans , Lymphocytes , Neutrophils , Prognosis , Retrospective Studies
5.
Pancreatology ; 20(4): 716-721, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32249060

ABSTRACT

BACKGROUND/OBJECTIVES: Mounting evidence has suggested that acute pancreatitis (AP) is a risk factor for pancreatic ductal adenocarcinoma (PDAC), but its role in survival in PDAC patients was rarely investigated. The objective was to investigate the association of a history of AP with survival among PDAC patients who underwent surgical resection. METHODS: A retrospective cohort study comprising 632 patients who were diagnosed with resectable PDAC was conducted. Survival was evaluated by history of AP prior to a diagnosis of PDAC using Kaplan-Meier methods and log-rank tests. Multivariate analyses for mortality were estimated using the Cox proportional hazards model. Propensity score matching methods were used to balance the difference of clinical characteristics between patients with and without AP history. RESULTS: The log-rank tests showed that patients with a history of AP had a worse overall survival than those without a history of AP (p = 0.006). The multivariable-adjusted hazard ratio (HR) for mortality comparing participants with AP to those without AP was 1.808 (95% CI: 1.241-2.632, p = 0.002). Patients with a recent history of AP (<2 years), rather than patients with a remote history of AP (≥2 years), were found to have significantly worse survival (p = 0.014) than those without a history of AP. After adjusted for PSM, history of AP remained an independent survival predictor of PDAC following surgical resection. CONCLUSIONS: Our findings indicate that a history of AP, especially a recent history of AP, is associated with poor survival among patients with resectable pancreatic ductal adenocarcinoma.


Subject(s)
Carcinoma, Pancreatic Ductal/pathology , Pancreatic Neoplasms/pathology , Pancreatitis/pathology , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Female , Humans , Male , Middle Aged , Pancreatic Neoplasms/drug therapy , Risk Factors , Survival Analysis
6.
Cancer Control ; 27(1): 1073274820915947, 2020.
Article in English | MEDLINE | ID: mdl-32268796

ABSTRACT

Lymphatic metastasis is a major determinant of the outcome of resected pancreatic cancer. Gemcitabine-based adjuvant chemotherapy can improve the outcome of resected pancreatic cancer. However, the efficacy of gemcitabine against pancreatic cancer stratified by nodal involvement is unclear. In this study, patients who had undergone curative resection of pancreatic adenocarcinoma (612 cases) were included. The efficacy of adjuvant gemcitabine-based regimen, stratified by nodal status (negative, positive) or N substage (N0, no nodal involvement; N1, 1-3-node involvement; N2, ≥4-node involvement), was examined. Both the node-negative (hazard ratio [HR] = 0.62, 95% confidence interval [CI], 0.44-0.87, P = .006) and node-positive subgroups (HR = 0.45, 95% CI, 0.33-0.62, P < .001) benefited from gemcitabine-based adjuvant chemotherapy. Patients with N0 (ie, the node-negative subgroup) or N1 (HR = 0.36, 95% CI, 0.25-0.52, P < .001) disease benefited from gemcitabine-based chemotherapy. However, patients with N2 tumors (HR = 0.95, 95% CI, 0.50-1.78, P = .867) had poor response to gemcitabine-based treatment. Therefore, we postulate that resected pancreatic cancer with N2 node involvement is refractory to gemcitabine-based adjuvant chemotherapy. A more intensive adjuvant regimen may be required for N2 subgroup patients.


Subject(s)
Chemotherapy, Adjuvant/methods , Deoxycytidine/analogs & derivatives , Lymph Nodes/drug effects , Lymphatic Metastasis/pathology , Pancreatic Neoplasms/drug therapy , Deoxycytidine/pharmacology , Deoxycytidine/therapeutic use , Drug Resistance, Neoplasm , Female , Humans , Lymph Nodes/pathology , Male , Middle Aged , Gemcitabine , Pancreatic Neoplasms
7.
Acta Biochim Biophys Sin (Shanghai) ; 52(3): 241-250, 2020 Mar 18.
Article in English | MEDLINE | ID: mdl-32072180

ABSTRACT

Pancreatic ductal adenocarcinoma is one of the deadliest malignant tumors, and many genes play important roles in its development. The hepatocyte nuclear factor-1a (HNF-1a) gene encodes HNF-1a, which is a transcriptional activator. HNF-1a regulates the tissue-specific expression of multiple genes, especially in pancreatic islet cells and in the liver. However, the role of the HNF-1a gene in the development of pancreatic cancer is still unclear. Here, we used immunohistochemical staining and real-time PCR to analyze HNF-1a expression in pancreatic cancer tissue. Stable cell lines with HNF-1a knockdown or overexpression were established to analyze the role of HNF-1a in pancreatic cancer cell proliferation and apoptosis by colony formation assay and flow cytometry. We also analyzed the L-type pyruvate kinase (PKLR) promoter sequence to identify the regulatory effect of HNF-1a on PKLR transcription and confirmed the HNF-1a binding site in the PKLR promoter via a chromatin immunoprecipitation assay. HNF-1a was found to be overexpressed in pancreatic cancer and promoted proliferation while inhibiting apoptosis in pancreatic cancer cells. PKLR was identified as the downstream target gene of HNF-1a and binding of HNF-1a at two sites in PKLR (-1931/-1926 and -966/-961) regulated PKLR transcription. In conclusion, HNF-1a is overexpressed in pancreatic cancer, and the transcription factor HNF-1a can promote pancreatic cancer growth and apoptosis resistance via its target gene PKLR.


Subject(s)
Hepatocyte Nuclear Factor 1-alpha/metabolism , Pancreatic Neoplasms/metabolism , Pyruvate Kinase/metabolism , Apoptosis/drug effects , Binding Sites , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Line , Cell Line, Tumor , Cell Proliferation/drug effects , Hepatocyte Nuclear Factor 1-alpha/biosynthesis , Hepatocyte Nuclear Factor 1-alpha/genetics , Humans , Immunohistochemistry/methods , Nuclear Proteins/metabolism , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Promoter Regions, Genetic , Trans-Activators/metabolism , Transcription Factors/metabolism , Transcriptional Activation
8.
Mol Cancer ; 18(1): 100, 2019 05 23.
Article in English | MEDLINE | ID: mdl-31122251

ABSTRACT

Apoptosis resistance is to a large extent a major obstacle leading to chemotherapy failure during cancer treatment. Bypassing the apoptotic pathway to induce cancer cell death is considered to be a promising approach to overcoming this problem. Necroptosis is a regulated necrotic cell death modality in a caspase-independent fashion and is mainly mediated by Receptor-Interacting Protein 1 (RIP1), RIP3, and Mixed Lineage Kinase Domain-Like (MLKL). Necroptosis serves as an alternative mode of programmed cell death overcoming apoptosis resistance and may trigger and amplify antitumor immunity in cancer therapy.The role of necroptosis in cancer is complicated. The expression of key regulators of the necroptotic pathway is generally downregulated in cancer cells, suggesting that cancer cells may also evade necroptosis to survive; however, in certain types of cancer, the expression level of key mediators is elevated. Necroptosis can elicit strong adaptive immune responses that may defend against tumor progression; however, the recruited inflammatory response may also promote tumorigenesis and cancer metastasis, and necroptosis may generate an immunosuppressive tumor microenvironment. Necroptosis also reportedly promotes oncogenesis and cancer metastasis despite evidence demonstrating its antimetastatic role in cancer. In addition, necroptotic microenvironments can direct lineage commitment to determine cancer subtype development in liver cancer. A plethora of compounds and drugs targeting necroptosis exhibit potential antitumor efficacy, but their clinical feasibility must be validated.Better knowledge of the necroptotic pathway mechanism and its physiological and pathological functions is urgently required to solve the remaining mysteries surrounding the role of necroptosis in cancer. In this review, we briefly introduce the molecular mechanism and characteristics of necroptosis, the interplay between necroptosis and other cell death mechanisms, crosstalk of necroptosis and metabolic signaling and detection methods. We also summarize the intricate role of necroptosis in tumor progression, cancer metastasis, prognosis of cancer patients, cancer immunity regulation, cancer subtype determination and cancer therapeutics.


Subject(s)
Gene Regulatory Networks , Necroptosis , Neoplasms/metabolism , Disease Progression , Gene Expression Regulation, Neoplastic , Humans , Neoplasms/immunology , Protein Kinases/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Tumor Microenvironment
9.
Ann Surg Oncol ; 26(6): 1649-1656, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30924017

ABSTRACT

BACKGROUND: Metastatic lesion to the pancreas accounts for approximately 2% of pancreatic neoplasms. There is no prospective, randomized or case-controlled study evaluating the role of pancreatic metastasectomy. METHODS: The PubMed, EMBASE, and Cochrane Library electronic databases were searched for studies published between January 1, 2001 and December 31, 2017. Studies with five or more patients who received pancreatic metastasectomy and data from our institution (29 patients) were included. The Kaplan-Meier method was used for survival analysis. RESULTS: A total of 414 patients from 20 institutions who underwent pancreatic resections were included. Of the reported 31 kinds of primary neoplasms, renal-cell carcinoma (RCC) comprised the most (54.3%). At the time of diagnosis, although 40.3% patients were asymptomatic, abdominal pain (34.8%) and jaundice (20.6%) were relatively common. As for surgical type, pancreatoduodenectomy, total pancreatectomy, distal pancreatectomy, and enucleation took up 37.9%, 11.4%, 43.5%, and 7.2% respectively. The mortality and morbidity rates were 1.4% and 48.3% respectively. Patients with symptoms at the time of diagnosis had significantly shorter survival compared with asymptomatic patients (p = 0.017). Those with RCC as primary tumor had significantly longer survival compared with non-RCC patients (p < 0.001). Positive margin also predicts worse prognosis (p = 0.035). CONCLUSIONS: Pancreatic metastasectomy is safe and associated with acceptable short- and intermediate-term results. In the conditions of RCC as the primary tumor, being asymptomatic, or negative resection margin, a better prognosis after resection can be achieved.


Subject(s)
Metastasectomy/mortality , Pancreatic Neoplasms/surgery , Humans , Neoplasm Metastasis , Pancreatic Neoplasms/pathology , Prognosis , Survival Rate
10.
World J Surg Oncol ; 17(1): 137, 2019 Aug 06.
Article in English | MEDLINE | ID: mdl-31387646

ABSTRACT

BACKGROUND: Both the 7th and 8th editions of the American Joint Committee on Cancer (AJCC) staging systems have been introduced for pancreatic adenocarcinoma. However, the applicability of these classifications for invasive intraductal papillary mucinous neoplasms (IPMN) has not been systematically examined. METHODS: Patients with invasive IPMN were retrieved from a cohort of 18 geographical sites (1973-2014 varying) in the Surveillance, Epidemiology, and End Results (SEER) cancer registry. The 7th and 8th editions of the AJCC staging were compared. Survival rates and multivariate analyses were computed. RESULTS: In total, 1216 patients with resected invasive IPMN were included. A major difference between the 7th and 8th systems is the definition of stage IIA (7th, beyond the pancreas without involvement of major arteries; 8th, maximum tumor diameter > 4 cm). The hazard ratio (HR) of stage IIA disease (versus stage IA, HR = 2.33, P < 0.001) was higher than that of stage IB disease (HR = 1.48, P = 0.087) by the 7th edition classification, whereas the HR of stage IIA disease (HR = 1.26, P = 0.232) was even lower than that of stage IB disease (HR = 1.48, P = 0.040) by the 8th edition classification. In addition, for the 8th edition staging system, tumor size was not a predictor of survival in patients with resectable tumor > 2 cm (size > 4 cm versus > 2 ≤ 4 cm, HR = 0.91, P = 0.420). CONCLUSIONS: The AJCC 7th edition staging classification is more applicable than the 8th edition classification for invasive IPMN.


Subject(s)
Adenocarcinoma, Mucinous/pathology , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Papillary/pathology , Neoplasm Staging/standards , Pancreatic Intraductal Neoplasms/pathology , Adenocarcinoma, Mucinous/classification , Adenocarcinoma, Mucinous/surgery , Adolescent , Adult , Aged , Aged, 80 and over , Carcinoma, Pancreatic Ductal/classification , Carcinoma, Pancreatic Ductal/surgery , Carcinoma, Papillary/classification , Carcinoma, Papillary/surgery , Cohort Studies , Female , Follow-Up Studies , Humans , Male , Middle Aged , Neoplasm Invasiveness , Pancreatic Intraductal Neoplasms/classification , Pancreatic Intraductal Neoplasms/surgery , SEER Program , Survival Rate , Young Adult
11.
Cancer Immunol Immunother ; 67(12): 1815-1823, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30167864

ABSTRACT

INTRODUCTION: Natural killer cells (NK) are often believed to play a positive role in the antitumor immune response. However, this is not the case for patients with advanced pancreatic cancer. This study was performed to determine the unique subtype of "educated" NK cells and their prognostic value in patients with advanced pancreatic cancer. METHODS: We divided 378 eligible patients into a derivation cohort (September 2010 to December 2014, n = 239) and a validation cohort (January 2015 to April 2016, n = 139). Flow cytometry was performed to analyze NK cells. Enzyme-linked immunosorbent assay was used to detect interleukin-2 (IL-2), interferon gamma (IFN-γ) and tumor necrosis factor alpha (TNF-α) production. The Kaplan-Meier method and the Cox proportional hazards model were used. RESULTS: Survival analysis showed that a high density of NK cells accompanied by a high neutrophil-to-lymphocyte ratio was associated with reduced overall survival in both the derivation and validation cohorts. Multivariable analysis also showed that high NK infiltration (HR 1.45, 95% CI 1.17 to 1.79, p = 0.001) was an independent prognostic factor. In these patients, high NK infiltration was associated with reduced levels of IL-2, IFN-γ and TNF-α, although only IFN-γ reached statistical significance, which accounted for this unique phenomenon. DISCUSSION: Natural killer cells in patients with advanced pancreatic cancer are a unique subtype with anergic features. A high density of NKs predicts poor survival in these patients, possibly because an active inflammatory response and reduced secretion of IL-2, IFN-γ and TNF-α inhibit NK activation.


Subject(s)
Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/mortality , Cell Communication/immunology , Clonal Anergy/immunology , Killer Cells, Natural/immunology , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/mortality , Adult , Aged , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cytokines/metabolism , Female , Humans , Kaplan-Meier Estimate , Killer Cells, Natural/metabolism , Killer Cells, Natural/pathology , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Male , Middle Aged , Neoplasm Staging , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Prognosis , Proportional Hazards Models , Tumor Burden
12.
Pancreatology ; 18(8): 971-976, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30131287

ABSTRACT

BACKGROUND: Carbohydrate antigen 19-9 (CA19-9) is the best-validated biomarker for pancreatic cancer. The National Comprehensive Cancer Network (NCCN) guideline asserts that "CA19-9 will be undetectable in Lewis antigen-negative individuals". However, reports of CA19-9 secretion and its significance in Lewis (-) patients with pancreatic cancer have been inconsistent. This study was to examine serum CA19-9 levels in patients with pancreatic cancer according to Lewis status. METHODS: Patients with pancreatic cancer (1482 cases) were retrieved from a prospectively maintained database. Patients with benign pancreatic disease (210 cases) and normal subjects (315 cases) were used as controls. Lewis genotypes were examined by fucosyltransferase 3 (FUT3) sequencing. RESULTS: In patients with pancreatic cancer, 8.4% of subjects were Lewis (-), but only 41.9% of Lewis (-) subjects had CA19-9 values ≤ 2 U/mL. CA19-9 was even elevated (>37 U/mL) in 27.4% of Lewis (-) patients. The area under the receiver operating characteristic (ROC) curve for CA19-9 as a diagnostic biomarker was 0.842 in Lewis (-) patients with pancreatic cancer, which is closing to that of CA19-9 applied in all of patients with pancreatic cancer (0.898). Lewis (-) status was an independent prognostic factor for shorter survival in a multivariable analysis (hazard ratio (HR), 1.30, 95% confidence interval (CI), 1.03-1.64; P = 0.028). CONCLUSIONS: Not all Lewis (-) patients with pancreatic cancer are non-secretors of CA19-9. Contrary to general understanding, CA19-9 can retain its utility as a biomarker in these patients in spite of Lewis (-) genotype.


Subject(s)
Biomarkers, Tumor/blood , CA-19-9 Antigen/blood , Pancreatic Neoplasms/blood , Aged , Biomarkers, Tumor/genetics , CA-19-9 Antigen/genetics , Female , Fucosyltransferases/analysis , Fucosyltransferases/genetics , Genotype , Humans , Male , Middle Aged , Pancreatic Diseases/diagnosis , Pancreatic Neoplasms/diagnosis , Pancreatic Neoplasms/genetics , Survival Analysis
14.
Cancer Lett ; 587: 216680, 2024 Apr 10.
Article in English | MEDLINE | ID: mdl-38346584

ABSTRACT

Metabolic reprogramming is a typical hallmark of cancer. Enhanced glycolysis in tumor cells leads to the accumulation of lactate, which is traditionally considered metabolic waste. With the development of high-resolution liquid chromatography coupled with tandem mass spectrometry (LC-MS/MS), the lactate-derived, lysine lactylation(Kla), has been identified. Kla can alter the spatial configuration of chromatin and regulate the expression of corresponding genes. Metabolic reprogramming and epigenetic remodeling have been extensively linked. Accumulating studies have subsequently expanded the framework on the key roles of this protein translational modification (PTM) in tumors and have provided a new concept of cancer-specific regulation by Kla.


Subject(s)
Lysine , Neoplasms , Humans , Chromatography, Liquid , Tandem Mass Spectrometry , Lactic Acid , Neoplasms/genetics
15.
Front Endocrinol (Lausanne) ; 15: 1281622, 2024.
Article in English | MEDLINE | ID: mdl-38524630

ABSTRACT

Background: CXC chemokine receptor 4 (CXCR4) is associated with the progression and metastasis of numerous malignant tumors. However, its relationship with Gastroenteropancreatic Neuroendocrine Neoplasms Grade 3 (GEP-NENs G3) is unclear. The aim of this study was to characterize the expression of CXCR4 in GEP-NENS and to explore the clinical and prognostic value of CXCR4. Methods: This study retrospectively collected clinical and pathological data from patients with GEP-NENs who receiving surgery in Qilu Hospital of Shandong University from January 2013 to April 2021, and obtained the overall survival of the patients based on follow-up. Immunohistochemistry (IHC) was performed on pathological paraffin sections to observe CXCR4 staining. Groups were made according to pathological findings. Kaplan-Meier (K-M) curve was used to evaluate prognosis. SPSS 26.0 was used for statistical analysis. Results: 100 GEP-NENs G3 patients were enrolled in this study. There was a significant difference in primary sites (P=0.002), Ki-67 index (P<0.001), and Carcinoembryonic Antigen (CEA) elevation (P=0.008) between neuroendocrine tumor (NET) G3 and neuroendocrine carcinoma (NEC). CXCR4 was highly expressed only in tumors, low or no expressed in adjacent tissues (P<0.001). The expression level of CXCR4 in NEC was significantly higher than that in NET G3 (P=0.038). The K-M curves showed that there was no significant difference in overall survival between patients with high CXCR4 expression and patients with low CXCR4 expression, either in GEP-NEN G3 or NEC (P=0.920, P=0.842. respectively). Conclusion: Differential expression of CXCR4 was found between tumor and adjacent tissues and between NET G3 and NEC. Our results demonstrated that CXCR4 can be served as a new IHC diagnostic indicator in the diagnosis and differential diagnosis of GEP-NENs G3. Further studies with multi-center, large sample size and longer follow-up are needed to confirm the correlation between CXCR4 expression level and prognosis.


Subject(s)
Carcinoma, Neuroendocrine , Intestinal Neoplasms , Neuroendocrine Tumors , Pancreatic Neoplasms , Stomach Neoplasms , Humans , Receptors, CXCR4 , Retrospective Studies , Intestinal Neoplasms/pathology , Stomach Neoplasms/pathology , Pancreatic Neoplasms/pathology , Neuroendocrine Tumors/pathology , Carcinoma, Neuroendocrine/pathology
16.
Cancer Lett ; 567: 216267, 2023 07 28.
Article in English | MEDLINE | ID: mdl-37315709

ABSTRACT

Effector, memory and exhaustion are three phenotypes of CD8+ T cell. In tumor microenvironment (TME), metabolism dysfunction of the three should take the blame for immune escape. Against background of CD8+ T cell in normal development, multiple determinants in TME, including nutrition competition, PD-1 signals and other cancer - CD8+ T cell interactions, cause metabolism reprograming, including failure in energy metabolism and other abnormal lipid metabolism. Further, incompatibility among metabolism patterns of three phenotypes results in unresponsiveness of immune checkpoint blockade (ICB). Therefore, combination of ICB and drugs aiming at abnormal lipid metabolism provides promising direction to improve cancer therapy. This review focuses on lipid metabolism of CD8+ T cell and aims to provide innovative therapy strategy to cure cancer.


Subject(s)
Lipid Metabolism , Neoplasms , Humans , Neoplasms/genetics , Neoplasms/therapy , CD8-Positive T-Lymphocytes , Energy Metabolism , Nutritional Status , Tumor Microenvironment
17.
Biochim Biophys Acta Mol Basis Dis ; 1869(1): 166583, 2023 01 01.
Article in English | MEDLINE | ID: mdl-36270476

ABSTRACT

DNA methylation profiles are in dynamic equilibrium via the initiation of methylation, maintenance of methylation and demethylation, which control gene expression and chromosome stability. Changes in DNA methylation patterns play important roles in carcinogenesis and primarily manifests as hypomethylation of the entire genome and the hypermethylation of individual loci. These changes may be reflected in blood-based DNA, which provides a non-invasive means for cancer monitoring. Previous blood-based DNA detection objects primarily included circulating tumor DNA/cell-free DNA (ctDNA/cfDNA), circulating tumor cells (CTCs) and exosomes. Researchers gradually found that methylation changes in peripheral blood mononuclear cells (PBMCs) also reflected the presence of tumors. Blood-based DNA methylation is widely used in early diagnosis, prognosis prediction, dynamic monitoring after treatment and other fields of clinical research on cancer. The reversible methylation of genes also makes them important therapeutic targets. The present paper summarizes the changes in DNA methylation in cancer based on existing research and focuses on the characteristics of the detection objects of blood-based DNA, including ctDNA/cfDNA, CTCs, exosomes and PBMCs, and their application in clinical research.


Subject(s)
Cell-Free Nucleic Acids , Circulating Tumor DNA , Neoplasms , Humans , DNA Methylation , Leukocytes, Mononuclear , Biomarkers, Tumor/genetics , Circulating Tumor DNA/genetics , Neoplasms/genetics
18.
Biochim Biophys Acta Rev Cancer ; 1878(6): 189014, 2023 11.
Article in English | MEDLINE | ID: mdl-37918451

ABSTRACT

Microbes are widely present in various organs of the human body and play important roles in numerous physiological and pathological processes. Nevertheless, owing to multiple limiting factors, such as contamination and low biomass, the current understanding of the intratumoral microbiome is limited. The intratumoral microbiome exerts tumor-promoting or tumor-suppressive effects by engaging in metabolic reactions within the body, regulating signaling cancer-related pathways, and impacting both host cells function and immune system. It is important to emphasize that intratumoral microbes exhibit substantial heterogeneity in terms of composition and abundance across various tumor types, thereby potentially influencing diverse aspects of tumorigenesis, progression, and metastasis. These findings suggest that intratumoral microbiome have great potential as diagnostic and prognostic biomarkers. By manipulating the intratumoral microbes to employ cancer therapy, the efficacy of chemotherapy or immunotherapy can be enhanced while minimizing adverse effects. In this review, we comprehensively describe the composition and function of the intratumoral microbiome in various human solid tumors. Combining recent advancements in research, we discuss the origins, mechanisms, and prospects of the clinical applications of intratumoral microbiome.


Subject(s)
Microbiota , Neoplasms , Humans , Neoplasms/therapy , Carcinogenesis , Immunotherapy , Signal Transduction
19.
Environ Sci Pollut Res Int ; 30(9): 24479-24493, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36342609

ABSTRACT

Optimized fertilizer and tillage management can be an effective strategy for high ecological efficiencies as well as crop yields. The objective of this study was to assess the impact of diverse management practices on carbon footprint, and ecosystem services in a wheat-maize cropping system. An in situ field experiment field was conducted from 2018 to 2020 on the North China Plain, and six treatments were established: deep tillage (DT), shallow tillage (ST), no tillage (NT), deep tillage + adding organic fertilizer (DTF), shallow tillage + adding organic fertilizer (STF), and no tillage + adding organic fertilizer (NTF). The results showed that adding organic fertilizer and the deeper tillage depth caused higher direct CO2 and N2O emission fluxes. DTF treatment significantly increased carbon footprint either per-unit area (CFa) or per-unit net income (CFe). Compared with DT treatment, STF treatment had higher CFa but lower CFe by increasing net income through boosted crop yields. Besides, the highest ecosystem service values (ESV) were present in STF treatment during both 2 years (42,017.13 CNY ha-1 and 43,352.03 CNY ha-1). In conclusion, STF treatment was an optimal management practice to trade-off grain yields and ecological efficiencies in a wheat-maize cropping system. Furthermore, this study highlights that adding organic fertilizer could be an efficient option toward sustainable farmland utilization with high soil carbon sequestration capacity and high ESV.


Subject(s)
Agriculture , Triticum , Agriculture/methods , Zea mays , Ecosystem , Fertilizers , Soil , Edible Grain , Fertilization , China
20.
Cancer Lett ; 575: 216403, 2023 Oct 28.
Article in English | MEDLINE | ID: mdl-37741433

ABSTRACT

Obesity is an essential risk factor for pancreatic cancer (PC). Macrophage-induced inflammation plays a pivotal role in obesity-associated carcinogenesis and disease progression; however, the underlying molecular mechanisms remain unclear. In this study, we found that fatty acid-binding protein 4 (FABP4) overexpressed in serum of obese patients and was associated with poor overall survival. In vivo and in vitro experiments have revealed that FABP4 induces macrophage-related inflammation to promote cancer cell migration, invasion and metastasis under obese conditions. Mechanistically, FABP4 participates in transferring saturated fatty acid to induce macrophages pyroptosis in a caspase-1/GSDMD-dependent manner and mediates NOD-like receptor thermal protein domain associated protein 3 (NLRP3)/IL-1ß axis in macrophages, which further regulates epithelial-mesenchymal transition signals to promote the migration, invasion, and metastasis of PC cells. Our results suggest that FABP4 in macrophages is a crucial regulator of the NLRP3/IL-1ß axis to promote the progression of PC under obese conditions, which could act as a promising molecular target for treating of PC patients with obesity.

SELECTION OF CITATIONS
SEARCH DETAIL