Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Mol Biotechnol ; 2024 Mar 25.
Article in English | MEDLINE | ID: mdl-38526683

ABSTRACT

Pancreatic adenocarcinoma (PAAD) is a fatal disease with poor survival. Increasing evidence show that hypoxia-induced exosomes are associated with cancer progression. Here, we aimed to investigate the function of hsa_circ_0007678 (circR3HCC1L) and hypoxic PAAD cell-derived exosomal circR3HCC1L in PAAD progression. Through the exoRBase 2.0 database, we screened for a circular RNA circR3HCC1L related to PAAD. Changes of circR3HCC1L in PAAD samples and cells were analyzed with real-time quantitative polymerase chain reaction (RT-qPCR). Cell proliferation, migration, invasion were analyzed by colony formation, cell counting, and transwell assays. Measurements of glucose uptake and lactate production were done using corresponding kits. Several protein levels were detected by western blotting. The regulation mechanism of circR3HCC1L was verified by dual-luciferase reporter, RNA immunoprecipitation, and RNA pull-down assays. Exosomes were separated by differential ultracentrifugation. Animal experiments were used to verify the function of hypoxia-derived exosomal circR3HCC1L. CircR3HCC1L was upregulated in PAAD samples and hypoxic PAAD cells. Knockdown of circR3HCC1L decreased hypoxia-driven PAAD cell proliferation, migration, invasion, and glycolysis. Hypoxic PAAD cell-derived exosomes had higher levels of circR3HCC1L, hypoxic PAAD cell-derived exosomal circR3HCC1L promoted normoxic cancer cell malignant transformation and glycolysis in vitro and xenograft tumor growth in mouse models in vivo. Mechanistically, circR3HCC1L regulated pyruvate kinase M2 (PKM2) expression via sponging miR-873-5p. Also, PKM2 overexpression or miR-873-5p silencing offset circR3HCC1L knockdown-mediated effects on hypoxia-challenged PAAD cell malignant transformation and glycolysis. Hypoxic PAAD cell-derived exosomal circR3HCC1L facilitated PAAD progression through the miR-873-5p/PKM2 axis, highlighting the contribution of hypoxic PAAD cell-derived exosomal circR3HCC1L in PAAD.

2.
Medicine (Baltimore) ; 102(34): e34932, 2023 Aug 25.
Article in English | MEDLINE | ID: mdl-37653818

ABSTRACT

BACKGROUND: Hepatocellular carcinoma (HCC) is an exceedingly prevalent malignancy with an exceptionally poor prognosis. Targeted therapy is an effective treatment option for patients with advanced HCC. However, there have been no bibliometric analyses of targeted therapies for HCC. METHODS: This study aimed to assess the current status and future directions of targeted therapy for HCC to provide future scholars with clearer research contents and popular themes. Methods: Literature on targeted therapy for HCC from 2008 to 2022 was obtained from the Web of Science (WoS) and assessed using bibliometric methodology. Additionally, the VOS viewer was applied in the visualization study to conduct bibliographic coupling, co-authorship, co-citation, and co-occurrence analyses of publications. RESULTS: A total of 10,779 papers were subsequently selected. Over the past 15 years, there has been a progressive increase in the number of publications on an annualized basis. China released the most publications in the field, whereas the United States had the highest H-index. Cancers published the most papers. Fudan University had the greatest sway in this area. Studies could be divided into 5 clusters: "Gene and expression research," "Mechanism study," "Nanoparticle study," "Targeted drug research," and "Clinical study." CONCLUSIONS: In the upcoming years, more papers on targeted therapy for HCC are expected to be released, demonstrating the potential for this topic to flourish. Particularly, "Clinical study" is the following trendy topic in this field. Other research subfields may likewise exhibit a continuous tendency towards balanced development.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Humans , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Authorship , Bibliometrics , China
3.
J Cancer Res Clin Oncol ; 149(13): 11857-11871, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37410139

ABSTRACT

INTRODUCTION: Surgery represents a primary therapeutic approach for borderline resectable and locally advanced pancreatic cancer (BR/LAPC). However, BR/LAPC lesions exhibit high heterogeneity and not all BR/LAPC patients who undergo surgery can derive beneficial outcomes. The present study aims to employ machine learning (ML) algorithms to identify those who would obtain benefits from the primary tumor surgery. METHODS: We retrieved clinical data of patients with BR/LAPC from the Surveillance, Epidemiology, and End Results (SEER) database and classified them into surgery and non-surgery groups based on primary tumor surgery status. To eliminate confounding factors, propensity score matching (PSM) was employed. We hypothesized that patients who underwent surgery and had a longer median cancer-specific survival (CSS) than those who did not undergo surgery would certainly benefit from surgical intervention. Clinical and pathological features were utilized to construct six ML models, and model effectiveness was compared through measures such as the area under curve (AUC), calibration plots, and decision curve analysis (DCA). We selected the best-performing algorithm (i.e., XGBoost) to predict postoperative benefits. The SHapley Additive exPlanations (SHAP) approach was used to interpret the XGBoost model. Additionally, data from 53 Chinese patients prospectively collected was used for external validation of the model. RESULTS: According to the results of the tenfold cross-validation in the training cohort, the XGBoost model yielded the best performance (AUC = 0.823, 95%CI 0.707-0.938). The internal (74.3% accuracy) and external (84.3% accuracy) validation demonstrated the generalizability of the model. The SHAP analysis provided explanations independent of the model, highlighting important factors related to postoperative survival benefits in BR/LAPC, with age, chemotherapy, and radiation therapy being the top three important factors. CONCLUSION: By integrating of ML algorithms and clinical data, we have established a highly efficient model to facilitate clinical decision-making and assist clinicians in selecting the population that would benefit from surgery.


Subject(s)
Pancreatic Neoplasms , Humans , Pancreatic Neoplasms/pathology , Machine Learning , Pancreatic Neoplasms
4.
J Laparoendosc Adv Surg Tech A ; 33(9): 890-896, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37074117

ABSTRACT

Background: In pancreatic cancer surgery, tumor violation of blood vessels is often considered a contraindication to surgery, especially laparoscopic surgery. We have completed 17 cases of major venous repair or reconstruction during laparoscopic pancreatic surgery, and we believe that this surgical method may be safe and feasible based on the skilled laparoscopic techniques. Materials and Methods: Between January 2014 and March 2022, a prospective cohort of 17 patients underwent major venous repair or reconstruction in our department. Among them, 15 cases underwent laparoscopic pancreaticoduodenectomy, 1 case underwent laparoscopic distal pancreatectomy, and 1 case underwent laparoscopic central pancreatectomy. In all of these cases, the pancreatic tumor invaded either portal veins (PV) or superior mesenteric veins. Given these clinical situations, 13 cases accepted laparoscopic venous resection and reconstruction, and 4 cases underwent venous repair. Results: Ten of 17 patients (58.8%) were male. The mean age was 67.1 (range 57-81). All patients' operations were successfully completed without transit to open. The average blocking time of venous resection and reconstruction was 30.1 (range 15-41) minutes and the average time of venous wedge resection and stitching was 24.0 (range 18-30) minutes. After surgeries, there were no complications such as PV stenosis, bleeding, thrombosis, and liver failure. Thirteen patients died within 2 years because of the tumor recurrence, and 4 patients are currently followed by outpatient visits, with no obvious signs of tumor recurrence. Conclusion: Studies have shown that the reconstruction or repair of the major veins under laparoscopic surgery is safe and effective. We recommended that surgeons need to have the basics of open surgery in case laparoscopic surgery cannot be continued, and have proficient laparoscopic surgery techniques combined with extensive training to achieve a learning curve for vascular anastomosis. Clinical Trial Registration number: KY2021SL152-01.


Subject(s)
Laparoscopy , Pancreatic Neoplasms , Aged , Female , Humans , Male , Laparoscopy/methods , Neoplasm Recurrence, Local/surgery , Pancreatectomy/methods , Pancreatic Neoplasms/surgery , Pancreaticoduodenectomy/methods , Portal Vein/surgery , Prospective Studies , Retrospective Studies , Treatment Outcome , Middle Aged , Aged, 80 and over
5.
Front Oncol ; 13: 1087700, 2023.
Article in English | MEDLINE | ID: mdl-36776324

ABSTRACT

Objective: Cancer of the pancreas is a life-threatening condition and has a high distant metastasis (DM) rate of over 50% at diagnosis. Therefore, this study aimed to determine whether patterns of distant metastases correlated with prognosis in pancreatic ductal adenocarcinoma (PDAC) with metastatic spread, and build a novel nomogram capable of predicting the 6, 12, 18-month survival rate with high accuracy. Methods: We analyzed data from the Surveillance, Epidemiology, and End Results (SEER) database for cases of PDAC with DM. Kaplan-Meier analysis, log-rank tests and Cox-regression proportional hazards model were used to assess the impact of site and number of DM on the cancer-specific survival (CSS) and over survival (OS). A total of 2709 patients with DM were randomly assigned to the training group and validation group in a 7:3 ratio. A nomogram was constructed by the dependent risk factors which were determined by multivariate Cox-regression analysis. An assessment of the discrimination and ability of the prediction model was made by measuring AUC, C-index, calibration curve and decision curve analysis (DCA). In addition, we collected 98 patients with distant metastases at the time of initial diagnosis from Ningbo University Affiliated LiHuili Hospital to verify the efficacy of the prediction model. Results: There was a highest incidence of liver metastases from pancreatic cancer (2387,74.36%), followed by lung (625,19.47%), bone (190,5.92%), and brain (8,0.25%). The prognosis of liver metastases differed from that of lung metastases, and the presence of multiple organ metastases was associated with poorer prognosis. According to univariate and multivariate Cox-regression analyses, seven factors (i.e., diagnosis age, tumor location, grade of tumor differentiation, T-stage, receipt of surgery, receipt of chemotherapy status, presence of multiple organ metastases) were included in our nomogram model. In internal and external validation, the ROC curves, C-index, calibration curves and DCA were calculated, which confirmed that this nomogram can precisely predict prognosis of PDAC with DM. Conclusion: Metastatic PDAC patients with liver metastases tended to have a worse prognosis than those with lung metastases. The number of DM had significant effect on the overall survival rate of metastatic PDAC. This study had a high prediction accuracy, which was helpful clinicians to analyze the prognosis of PDAC with DM and implement individualized diagnosis and treatment.

6.
Clinics (Sao Paulo) ; 77: 100047, 2022.
Article in English | MEDLINE | ID: mdl-35662010

ABSTRACT

OBJECTIVES: Emerging evidence has demonstrated that LINC01857 exerts a pivotal function in many cancers. However, its function in Pancreatic Ductal Adenocarcinoma (PDAC) still remains unclear. This study was designed to investigate the regulatory character of LINC01857 in PDAC. METHODS: Bioinformatic tools and databases were used to seek potential miRNAs and mRNAs. Gene expression was evaluated by Reverse Transcription quantitative real-time Polymerase Chain Reaction (RT-qPCR), and western blot was used for protein level detection. A subcellular fraction assay was done to ascertain the location of LINC01857 in PANC-1 and BxPC-3 human pancreatic cancer cells. CCK-8, EdU, wound healing and Transwell assays were performed to inquire into the influence of LINC01857, and SPARC -related Modular Calcium-binding protein-2 (SMOC2) on cell viability, proliferation, migration, and invasion, respectively. The interaction between LINC01857 and its downstream genes was explored by RNA immunoprecipitation and luciferase reporter assays. RESULTS: LINC01857 levels were significantly elevated in PDAC. Knockdown of LINC01857 significantly restrained the proliferation, migration, invasion, and Epithelial-Mesenchymal Transition (EMT) process of PDAC cells. MiR-19a-3p was a downstream target of LINC01857, and miR-19a-3p levels were significantly decreased in PDAC cells. In addition, SMOC2 expression had a negative correlation with that of miR-19a-3p, and SMOC2 was a downstream target of miR-19a-3p. Furthermore, SMOC2 upregulation partially abolished the inhibitive influence of LINC01857 downregulation on cell proliferation, migration, invasion, and the EMT process. CONCLUSION: LINC01857 promotes malignant phenotypes of PDAC cells via upregulation of SMOC2 by interacting with miR-19a-3p.


Subject(s)
Adenocarcinoma , Carcinoma, Pancreatic Ductal , MicroRNAs , Pancreatic Neoplasms , RNA, Long Noncoding , Adenocarcinoma/genetics , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Carcinoma, Pancreatic Ductal/metabolism , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , MicroRNAs/genetics , Pancreatic Neoplasms/pathology , RNA, Long Noncoding/genetics , Pancreatic Neoplasms
7.
Bioengineered ; 13(3): 6978-6995, 2022 03.
Article in English | MEDLINE | ID: mdl-35311623

ABSTRACT

Studies over the past decades have implicated lncRNAs in promoting the development, migration and invasion of gastric cancer (GC). However, the role and mechanism of lncRNA MBNL1-AS1 in GC promotion are poorly understood. In this research, qRT-PCR showed that MBNL1-AS1 was down-regulated in GC tissues and cells. Cell experiments and the animal study demonstrated that MBNL1-AS1 knockdown accelerated GC cell proliferation, migration, and invasion, thus restraining cell apoptosis. Meanwhile, overexpression of MBNL1-AS1 repressed GC cell promotion. Bioinformatics analysis confirmed that MBNL1-AS1 binds to miR-424-5p via negative modulation. Rescue experiments showed that decreased miR-424-5p level inhibited GC cell promotion by silencing MBNL1-AS1. Furthermore, Smad7 was identified as a target of miR-424-5p that could reverse the promotion of GC cell growth mediated by miR-424-5p. Western blot results proved that MBNL1-AS1 affected TGF-ß/SMAD pathways by regulating the miR-424-5p/Smad7 axis. Collectively, MBNL1-AS1 restrained GC growth via the miR-424-5p/Smad7 axis and thus could be a promising target for GC therapy. These findings illustrate that lncRNA MBNL1-AS1, as a tumor suppressor gene, participates in GC progression by regulating miR-424-5p/Smad7 axis, thus activating TGF-ß/EMT pathways. The evidence may provide a potential marker for GC patients.


Subject(s)
MicroRNAs , RNA, Long Noncoding , Stomach Neoplasms , Animals , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Humans , MicroRNAs/genetics , RNA, Long Noncoding/genetics , RNA-Binding Proteins/genetics , Smad7 Protein/genetics , Stomach Neoplasms/genetics , Transforming Growth Factor beta/genetics
8.
Clinics ; 77: 100047, 2022. tab, graf
Article in English | LILACS-Express | LILACS | ID: biblio-1384617

ABSTRACT

Abstract Objectives Emerging evidence has demonstrated that LINC01857 exerts a pivotal function in many cancers. However, its function in Pancreatic Ductal Adenocarcinoma (PDAC) still remains unclear. This study was designed to investigate the regulatory character of LINC01857 in PDAC. Methods Bioinformatic tools and databases were used to seek potential miRNAs and mRNAs. Gene expression was evaluated by Reverse Transcription quantitative real-time Polymerase Chain Reaction (RT-qPCR), and western blot was used for protein level detection. A subcellular fraction assay was done to ascertain the location of LINC01857 in PANC-1 and BxPC-3 human pancreatic cancer cells. CCK-8, EdU, wound healing and Transwell assays were performed to inquire into the influence of LINC01857, and SPARC -related Modular Calcium-binding protein-2 (SMOC2) on cell viability, proliferation, migration, and invasion, respectively. The interaction between LINC01857 and its downstream genes was explored by RNA immunoprecipitation and luciferase reporter assays. Results LINC01857 levels were significantly elevated in PDAC. Knockdown of LINC01857 significantly restrained the proliferation, migration, invasion, and Epithelial-Mesenchymal Transition (EMT) process of PDAC cells. MiR-19a-3p was a downstream target of LINC01857, and miR-19a-3p levels were significantly decreased in PDAC cells. In addition, SMOC2 expression had a negative correlation with that of miR-19a-3p, and SMOC2 was a downstream target of miR-19a-3p. Furthermore, SMOC2 upregulation partially abolished the inhibitive influence of LINC01857 downregulation on cell proliferation, migration, invasion, and the EMT process. Conclusion LINC01857 promotes malignant phenotypes of PDAC cells via upregulation of SMOC2 by interacting with miR-19a-3p. HIGHLIGHTS LINC01857 is upregulated in PAAD and promotes malignant cellular behaviors. LINC01857 interacts with miR-19a-3p to regulate SMOC2 expression. LINC01857 promotes malignant cellular phenotypes by upregulating SMOC2.

9.
Signal Transduct Target Ther ; 6(1): 55, 2021 02 10.
Article in English | MEDLINE | ID: mdl-33563892

ABSTRACT

Neuroendocrine carcinoma (NEC) of the gallbladder (GB-NEC) is a rare but extremely malignant subtype of gallbladder cancer (GBC). The genetic and molecular signatures of GB-NEC are poorly understood; thus, molecular targeting is currently unavailable. In the present study, we applied whole-exome sequencing (WES) technology to detect gene mutations and predicted somatic single-nucleotide variants (SNVs) in 15 cases of GB-NEC and 22 cases of general GBC. In 15 GB-NECs, the C > T mutation was predominant among the 6 types of SNVs. TP53 showed the highest mutation frequency (73%, 11/15). Compared with neuroendocrine carcinomas of other organs, significantly mutated genes (SMGs) in GB-NECs were more similar to those in pulmonary large-cell neuroendocrine carcinomas (LCNECs), with driver roles for TP53 and RB1. In the COSMIC database of cancer-related genes, 211 genes were mutated. Strikingly, RB1 (4/15, 27%) and NAB2 (3/15, 20%) mutations were found specifically in GB-NECs; in contrast, mutations in 29 genes, including ERBB2 and ERBB3, were identified exclusively in GBC. Mutations in RB1 and NAB2 were significantly related to downregulation of the RB1 and NAB2 proteins, respectively, according to immunohistochemical (IHC) data (p values = 0.0453 and 0.0303). Clinically actionable genes indicated 23 mutated genes, including ALK, BRCA1, and BRCA2. In addition, potential somatic SNVs predicted by ISOWN and SomVarIUS constituted 6 primary COSMIC mutation signatures (1, 3, 30, 6, 7, and 13) in GB-NEC. Genes carrying somatic SNVs were enriched mainly in oncogenic signaling pathways involving the Notch, WNT, Hippo, and RTK-RAS pathways. In summary, we have systematically identified the mutation landscape of GB-NEC, and these findings may provide mechanistic insights into the specific pathogenesis of this deadly disease.


Subject(s)
Anaplastic Lymphoma Kinase/genetics , Carcinoma, Neuroendocrine/genetics , Gallbladder Neoplasms/genetics , Retinoblastoma Binding Proteins/genetics , Tumor Suppressor Protein p53/genetics , Ubiquitin-Protein Ligases/genetics , Aged , Aged, 80 and over , BRCA1 Protein/genetics , BRCA2 Protein/genetics , Biomarkers, Tumor/genetics , Exome/genetics , Female , Gallbladder Neoplasms/pathology , Genome, Human/genetics , Humans , Male , Middle Aged , Mutation/genetics , Signal Transduction/genetics , Exome Sequencing
10.
Biochem Cell Biol ; 98(4): 474-483, 2020 08.
Article in English | MEDLINE | ID: mdl-32114773

ABSTRACT

Long non-coding RNA (lncRNA) is known to be involved in a variety of diseases. However, the role of Gm4419 in hepatic ischemia-reperfusion (I/R) injury remains unknown. To study this, we first established a rat model of hepatic I/R, and a BRL-3A cell model of hypoxia-reoxygenation (H/R) for in vivo and in vitro studies. Staining with hematoxylin and eosin and hepatic injury scores were used to evaluate the degree of hepatic I/R injury. Cell apoptosis was assessed via staining with Edu, and with annexin V-FITC-propidium iodide assays. The interactions between Gm4419 and miR-455, as well as miR-455 and SOX6 were evaluated via luciferase reporter activity assays and RNA immunoprecipitation assays. In vivo, we found that Gm4419 was up-regulated in the rats subjected to I/R. Moreover, knockdown of Gm4419 alleviated the I/R-induced liver damage in the rats. In vitro, knockdown of Gm4419 alleviated H/R-induced apoptosis in BRL-3A cells. Interestingly, we found that miR-455 is a target of Gm4419, and Gm4419 regulates the expression of miR-455 via sponging. Furthermore, SOX6 was proven to be the target of miR-455. Finally, rescue experiments confirmed that knockdown of Gm4419 inhibits apoptosis by regulating miR-455 and SOX6 in H/R-treated BRL-3A cells. Therefore, our findings show that the lncRNA Gm4419 accelerates hepatic I/R injury by targeting the miR-455-SOX6 axis, which suggests a novel therapeutic target for hepatic I/R injury.


Subject(s)
Liver Diseases/genetics , MicroRNAs/genetics , RNA, Long Noncoding/genetics , Reperfusion Injury/genetics , SOXD Transcription Factors/metabolism , Animals , Apoptosis/physiology , Cell Line , Disease Models, Animal , Liver Diseases/metabolism , Liver Diseases/pathology , Male , MicroRNAs/metabolism , RNA, Long Noncoding/metabolism , Rats , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , SOXD Transcription Factors/genetics , Signal Transduction
11.
Gene ; 687: 1-8, 2019 Mar 01.
Article in English | MEDLINE | ID: mdl-30399427

ABSTRACT

MEG2 was recently found to have important functions in human cancers. However, the expression status and biological functions of MEG2 in hepatocellular carcinoma (HCC) remain unknown. In this study, we demonstrated that MEG2 expression was reduced in HCC tissues and cell lines using qRT-PCR, western blot and immunohistochemical staining. Decreased MEG2 expression predicted unfavorable clinical features and decreased overall survival and disease-free survival of HCC patients. In vitro functional assays showed that overexpression of MEG2 inhibited the cell viability, migration and invasion of HCCLM3 cells while MEG2 knockdown promoted these biological functions of Hep3B cells. Subcutaneous injection model and tail vein injection model showed that forced expression of MEG2 in HCCLM3 decreased the growth and lung metastasis of HCCLM3 cells in nude mice. Mechanically, MEG2 inhibited the EMT and AKT phosphorylation of HCC cells. The promoting effects of MEG2 knockdown on EMT, cell viability, proliferation, migration and invasion of Hep3B cells was blocked by AKT phosphorylation inhibition. In all, this study demonstrates that MEG2 inhibits the growth and metastasis of hepatocellular carcinoma by inhibiting AKT pathway.


Subject(s)
Carcinoma, Hepatocellular/pathology , Cell Movement , Cell Proliferation , Liver Neoplasms/pathology , Lung Neoplasms/secondary , Protein Tyrosine Phosphatases, Non-Receptor/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Female , Follow-Up Studies , Gene Expression Regulation, Neoplastic , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lymphatic Metastasis , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Neoplasm Invasiveness , Prognosis , Protein Tyrosine Phosphatases, Non-Receptor/genetics , Proto-Oncogene Proteins c-akt/genetics , Survival Rate , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
12.
ANZ J Surg ; 87(7-8): E26-E31, 2017 Jul.
Article in English | MEDLINE | ID: mdl-25880020

ABSTRACT

BACKGROUND: Risk factors for bile leakage after hemihepatectomy are unknown. METHODS: A prospectively maintained database review identified patients undergoing hemihepatectomy between 1 January 2009 and 30 September 2014. Patients were divided into B/C and non-B/C bile leakage groups. Risk factors for bile leakage were predicted and assessments of their impact on patients were made. RESULTS: Bile leakage occurred in 91 of the 297 patients (30.6%); 64 cases were classified as grade B bile leakage (21.5%) and three cases as grade C bile leakage (1.0%). Multivariate analysis confirmed that elevated preoperative alanine transaminase (ALT), positive bile culture during surgery, hilar bile duct plasty, bilioenteric anastomosis and laparoscopic surgery were risk factors for B/C grade bile leakage (P < 0.05). Percutaneous transhepatic biliary drainage (PTBD) and endoscopic nasobiliary drainage (ENBD) were protective factors for B/C grade bile leakage (P < 0.05). PTBD, ENBD and Kehr's T-tube drainage could reduce the drainage volume and duration of drainage after bile leakage (P < 0.05). The incidence of wound infection, abdominal infection, major complications and the Clavien classification system score in the B/C bile leakage group were higher than those in the non-B/C bile leakage group (P < 0.05). Patients in the B/C bile leakage group also required prolonged hospitalization (P < 0.05). The mortality of two groups was similar (P > 0.05). CONCLUSION: Patient with elevated preoperative ALT, positive bile cultures during surgery, hilar bile duct plasty, bilioenteric anastomosis and laparoscopic surgery are more likely to complicate bile leakage. We should use biliary drainage such as preoperative PTBD, ENBD or intraoperative Kehr's T-tube drainage to reduce and treat bile leakage in patients with high risk of bile leakage.


Subject(s)
Anastomotic Leak/therapy , Bile , Hepatectomy/methods , Anastomotic Leak/epidemiology , Female , Humans , Male , Middle Aged , Prospective Studies , Risk Factors , Treatment Outcome
13.
Hepatobiliary Surg Nutr ; 3(6): 421-2, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25568867

ABSTRACT

Laparoscopic pancreaticoduodenectomy (LPD) is one of the most complex operations in general surgery. With the development and maturation of surgical technology, more and more of such surgeries have been reported each year. Five LPDs have been performed in our department in the past year. We have achieved very satisfying clinical results with very few complications. The average operation takes 6.5 hours, which is significantly shorter compared to prior operations. In addition, the average hospitalization time was significantly shortened. Here we present a case report on one of the LPDs.

SELECTION OF CITATIONS
SEARCH DETAIL
...