1.Successful pregnancy after Whipple’s procedure for pancreatic neoplasm
Shruthi Dyamappa ; Priyanka P. Yoga ; Vijayan Sharmila
Philippine Journal of Obstetrics and Gynecology 2025;49(1):77-79
Pregnancy after undergoing major gastrointestinal surgeries like the Whipple’s procedure (pancreaticoduodenectomy) for pancreatic neoplasm is rare. This case report describes a 24-year-old woman who conceived and delivered a healthy baby after undergoing a Whipple’s procedure 5 months earlier for a pancreatic tumor. Her pregnancy was managed by a multidisciplinary team, and she delivered at 37 weeks of gestation through cesarean section without any complications. This case highlights the potential for successful pregnancy following a Whipple’s procedure, with proper counseling, coordinated care, and close monitoring during pregnancy.
Pancreatic Neoplasms
;
Pancreaticoduodenectomy
;
Pregnancy
2.Role of noncoding RNA and protein interaction in pancreatic cancer.
Zhang LI ; Tingting ZHANG ; Xiaojuan YANG ; Yong PENG
Chinese Medical Journal 2025;138(9):1019-1036
Noncoding RNAs (ncRNAs) are a class of RNA molecules with little or no protein-coding potential. Emerging evidence indicates that ncRNAs are frequently dysregulated and play pivotal roles in the pathogenesis of pancreatic cancer. Their aberrant expression can arise from chromosomal abnormalities, dysregulated transcriptional control, and epigenetic modifications. ncRNAs function as protein scaffolds or molecular decoys to modulate interactions between proteins and other biomolecules, thereby regulating gene expression and contributing to pancreatic cancer progression. In this review, we summarize the mechanisms underlying ncRNA dysregulation in pancreatic cancer, emphasize the biological significance of ncRNA-protein interactions, and highlight their clinical relevance. A deeper understanding of ncRNA-protein interactions is essential to elucidate molecular mechanisms and advance translational research in pancreatic cancer.
Humans
;
Pancreatic Neoplasms/metabolism*
;
RNA, Untranslated/metabolism*
;
Gene Expression Regulation, Neoplastic/genetics*
3.SMUG1 promoted the progression of pancreatic cancer via AKT signaling pathway through binding with FOXQ1.
Zijian WU ; Wei WANG ; Jie HUA ; Jingyao ZHANG ; Jiang LIU ; Si SHI ; Bo ZHANG ; Xiaohui WANG ; Xianjun YU ; Jin XU
Chinese Medical Journal 2025;138(20):2640-2656
BACKGROUND:
Pancreatic cancer is a lethal malignancy prone to gemcitabine resistance. The single-strand selective monofunctional uracil DNA glycosylase (SMUG1), which is responsible for initiating base excision repair, has been reported to predict the outcomes of different cancer types. However, the function of SMUG1 in pancreatic cancer is still unclear.
METHODS:
Gene and protein expression of SMUG1 as well as survival outcomes were assessed by bioinformatic analysis and verified in a cohort from Fudan University Shanghai Cancer Center. Subsequently, the effect of SMUG1 on proliferation, cell cycle, and migration abilities of SMUG1 cells were detected in vitro . DNA damage repair, apoptosis, and gemcitabine resistance were also tested. RNA sequencing was performed to determine the differentially expressed genes and signaling pathways, followed by quantitative real-time polymerase chain reaction and Western blotting verification. The cancer-promoting effect of forkhead box Q1 (FOXQ1) and SMUG1 on the ubiquitylation of myelocytomatosis oncogene (c-Myc) was also evaluated. Finally, a xenograft model was established to verify the results.
RESULTS:
SMUG1 was highly expressed in pancreatic tumor tissues and cells, which also predicted a poor prognosis. Downregulation of SMUG1 inhibited the proliferation, G1 to S transition, migration, and DNA damage repair ability against gemcitabine in pancreatic cancer cells. SMUG1 exerted its function by binding with FOXQ1 to activate the Protein Kinase B (AKT)/p21 and p27 pathway. Moreover, SMUG1 also stabilized the c-Myc protein via AKT signaling in pancreatic cancer cells.
CONCLUSIONS
SMUG1 promotes proliferation, migration, gemcitabine resistance, and c-Myc protein stability in pancreatic cancer via protein kinase B signaling through binding with FOXQ1. Furthermore, SMUG1 may be a new potential prognostic and gemcitabine resistance predictor in pancreatic ductal adenocarcinoma.
Humans
;
Pancreatic Neoplasms/pathology*
;
Forkhead Transcription Factors/genetics*
;
Signal Transduction/genetics*
;
Animals
;
Cell Line, Tumor
;
Proto-Oncogene Proteins c-akt/metabolism*
;
Cell Proliferation/physiology*
;
Mice
;
Uracil-DNA Glycosidase/genetics*
;
Female
;
Male
;
Gemcitabine
;
Mice, Nude
;
Apoptosis/physiology*
;
Deoxycytidine/analogs & derivatives*
;
Cell Movement/genetics*
4.Expert consensus on laparoscopic and robotic-assisted pancreatoduodenectomy with resection and reconstruction of portal-superior mesenteric vein (2025).
Chinese Journal of Surgery 2025;63(6):461-470
Pancreatoduodenectomy with portal-superior mesenteric vein resection and reconstruction can provide radical surgical opportunities for patients with venous invasion and enable them to benefit from the surgery. With the development of minimally invasive concepts and surgical techniques, laparoscopic and robot-assisted pancreatoduodenectomy with portal-superior mesenteric vein resection and reconstruction is being increasingly widely carried out. This surgical procedure is highly technically demanding, and the perioperative management of patients is complex. However, there is a lack of high-quality and high-level evidence-based clinical studies in this regard. In order to better standardize the clinical application of laparoscopic or robot-assisted pancreatoduodenectomy with portal-superior mesenteric vein resection and reconstruction in China, the Study Group of Minimally Invasive Treatment for Pancreatic Cancer in China Anti-Cancer Association, guided by problems and based on evidence, formed 17 recommendations through full discussions among experts. The recommendations involve the safety, oncological benefits, and perioperative patient management of the minimally invasive approach to pancreatoduodenectomy with portal-superior mesenteric vein resection and reconstruction.
Humans
;
Pancreaticoduodenectomy/methods*
;
Mesenteric Veins/surgery*
;
Laparoscopy
;
Portal Vein/surgery*
;
Robotic Surgical Procedures
;
Pancreatic Neoplasms/surgery*
;
Consensus
5.Effects of p38 phosphorylation on stemness maintenance and chemotherapy drug resistance of PANC-1 cells.
Xueying SHI ; Jinbo YU ; Shihai YANG ; Jin ZHAO
Chinese Journal of Cellular and Molecular Immunology 2025;41(2):116-124
Objective The aim of this study was to investigate the effect of p38 on stem cell maintenance of pancreatic cancer. Methods Human pancreatic cancer cells PANC-1 were treated with different concentrations of 5-fluorouracil(5-FU)(0.5×IC50, IC50, and 2×IC50) for 24 hours, and VX-702 (p38 phosphorylation inhibitor) was added, and the cells were inoculated in 6-well culture dishes with ultra-low adhesion to observe the changes of sphere tumors. The expression levels of cyclin-dependent kinase 2(CDK2), cyclin B1 and D1, Octamer-binding transcription factor 4(OCT4), SRY-box transcription factor 2(SOX2), Nanog and p38 were measured by Western blot. The mRNA expression levels of p38, OCT4, Nanog and SOX2 were tested by RT-PCR. Cell cycle, apoptosis, and the proportion of CD44+CD133+PANC-1 cells were evaluated by flow cytometry. Results The results showed that 5-FU inhibited the formation of tumor spheres in PANC-1 cells, increased CD44+CD133+cell fragments, down-regulated the expression of OCT4, Nanog and SOX2, and inhibited the stemness maintenance of PANC-1 tumor stem cells. Phosphorylation of PANC-1 cells was inhibited by a highly selective p38 MAPK inhibitor, VX-702(p38 mitogen-activated protein kinase inhibitor), which had the same effect as 5-FU treatment. When VX-702 combined with 5-FU was used to treat PANC-1 cells, the therapeutic effect was enhanced. Conclusion p38 inhibitors decreased PANC-1 cell activity and increased cell apoptosis. p38 inhibitors inhibit the stemness maintenance of pancreatic cancer stem cells.
Humans
;
Phosphorylation/drug effects*
;
Cell Line, Tumor
;
p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors*
;
Neoplastic Stem Cells/metabolism*
;
Drug Resistance, Neoplasm/drug effects*
;
Fluorouracil/pharmacology*
;
Pancreatic Neoplasms/pathology*
;
Apoptosis/drug effects*
;
SOXB1 Transcription Factors/genetics*
;
Octamer Transcription Factor-3/genetics*
6.Diagnostic yield and safety of pancreatic cystic lesions: A comparison between EUS-FNA and EUS-FNB.
Xiaoyu YU ; Mingmei YE ; Yawen NI ; Qianqi LIU ; Pan GONG ; Yuanyuan HUANG ; Xiaoyan WANG ; Li TIAN
Journal of Central South University(Medical Sciences) 2025;50(2):227-236
OBJECTIVES:
In recent years, the incidence and detection rate of pancreatic cystic lesions (PCLs) have increased significantly. Endoscopic ultrasound (EUS) plays an indispensable role in the diagnosis and differential diagnosis of PCLs. However, evidence comparing the diagnostic performance of EUS-guided fine-needle aspiration (EUS-FNA) and fine-needle biopsy (FNB) remains limited. This study aims to compare the diagnostic yield, adequacy of tissue acquisition, and safety between EUS-FNA and EUS-FNB in evaluating PCLs to inform clinical practice.
METHODS:
A retrospective review was conducted on patients with PCLs who underwent either EUS-FNA or EUS-FNB between January 2014 and August 2021. The diagnostic yield, tissue acquisition adequacy, and incidence of adverse events were compared between the 2 groups.
RESULTS:
A total of 90 patients with PCLs were included (52 in the FNA group and 38 in the FNB group). The diagnostic yield was similar between the FNA and FNB groups (94.2% vs 94.7%, P>0.05). The adequacy of tissue acquisition was 71.2% in the FNA group and 81.6% in the FNB group (P>0.05). No statistically significant difference was observed in the incidence of adverse events between the 2 groups (P>0.05).
CONCLUSIONS
Both EUS-FNA and EUS-FNB demonstrate equally high diagnostic yields and tissue adequacy in PCLs, with excellent safety profiles. Both methods are safe and effective diagnostic tools for evaluating PCLs.
Humans
;
Endoscopic Ultrasound-Guided Fine Needle Aspiration/adverse effects*
;
Retrospective Studies
;
Female
;
Male
;
Pancreatic Cyst/diagnostic imaging*
;
Middle Aged
;
Biopsy, Fine-Needle/adverse effects*
;
Aged
;
Pancreatic Neoplasms/diagnosis*
;
Adult
;
Endosonography/methods*
;
Pancreas/pathology*
;
Diagnosis, Differential
7.68Ga-DOTATATE and 18F-FDG PET/CT dual-modality imaging enhances precision of staging and treatment decision for gastroenteropancreatic neuroendocrine neoplasms.
Xiaoxiang ZHANG ; Ying TIAN ; Lilan FU ; Yin ZHANG ; Ye DONG ; Fei XIE ; Li CHEN ; Yanchao HUANG ; Hubing WU ; Jianer TAN
Journal of Southern Medical University 2025;45(6):1212-1219
OBJECTIVES:
To evaluate the value of ⁶⁸Ga-DOTATATE and ¹⁸F-FDG PET/CT imaging in staging and treatment decision for gastroenteropancreatic neuroendocrine neoplasms (GEP-NEN).
METHODS:
This retrospective analysis was conducted in 49 patients with GEP-NEN undergoing 18F-FDG and 68Ga-DOTATATE PET/CT imaging at our hospital from August, 2020 to March, 2023, including 34 newly diagnosed patients and 15 patients with recurrence or metastasis after treatment. GEP-NEN were classified into G1, G2, and G3 neuroendocrine tumors (NET) and neuroendocrine carcinomas (NEC) based on pathological typing. The detection efficiency were classified into 4 patterns based on the number of positive tumor lesions detected by the two tracers: 68Ga-DOTATATE>18F-FDG (A); 68Ga-DOTATATE=18F-FDG (B); 68Ga-DOTATATE<18F-FDG (C); and complementation (D). The value of dual-modality imaging in staging and treatment decision were evaluated by visual analysis.
RESULTS:
In the 49 patients with GEP-NEN, 68Ga-DOTATATE PET/CT was superior to 18F-FDG PET/CT for detecting systemic tumor lesions (P<0.001) and more sensitive for detecting primary/recurrent lesions, lymph node metastasis, liver metastasis, and bone metastasis (P<0.05), while 18F-FDG PET/CT had higher detection rates for lung metastasis and peritoneal metastasis (P<0.05). In terms of the detection efficiency, Pattern A was found in 46.9% (23/49) patients, Pattern B in 38.8% (19/49), Pattern C in 12.2% (6/49), and Pattern D in 2.0% (1/49). The complementary value of ¹⁸F-FDG PET/CT to ⁶⁸Ga-DOTATATE PET/CT was 0% in G1 NET patients (0/13), 8.3% in G2 NET patients (2/24), 50% in G3 NET patients (3/6), and 33.3% in NEC patients (2/6). 12.2% (6/49) of the patients had their staging confirmed or changed due to additional lesions detected by ¹⁸F-FDG PET/CT imaging, resulting subsequently in establishment or adjustment of their treatment plans.
CONCLUSIONS
68Ga-DOTATATE PET/CT imaging should be the primary choice for GEP-NEN patients. Additional ¹⁸F-FDG PET/CT imaging can potentially improve precision of staging and treatment decision-making for G2, G3 and NEC patients but provides virtually no clinical benefits for G1 NET patients.
Humans
;
Positron Emission Tomography Computed Tomography/methods*
;
Neuroendocrine Tumors/therapy*
;
Pancreatic Neoplasms/therapy*
;
Retrospective Studies
;
Organometallic Compounds
;
Stomach Neoplasms/therapy*
;
Neoplasm Staging
;
Fluorodeoxyglucose F18
;
Intestinal Neoplasms/therapy*
;
Female
;
Male
;
Middle Aged
;
Aged
;
Adult
8.Low-intensity pulsed ultrasound and oridonin synergistically induce ferroptosis of pancreatic cancer cells by activating PIEZO1 via the Nrf2/HO-1/GPX4 pathway.
Bihang SUN ; Yujun GUO ; Yulin QI ; Dan YAO ; Wenzhi CHEN ; Nianzhi CHEN
Journal of Southern Medical University 2025;45(10):2160-2170
OBJECTIVES:
To evaluate the inhibitory effect of oridonin against proliferation of pancreatic cancer cells and the mechanism underlying the synergistic effect of low-intensity pulsed ultrasound (LIPUS).
METHODS:
PANC-1 cells treated with different concentrations of oridonin were examined for changes in cell proliferation using CCK-8 assay and in MDA, GSH and ATP levels using flow cytometry. The protein expressions of GPX4, Nrf2 and HO-1 in the treated cells were detected with Western blotting. The effect of Fer-1, a ferroptosis inhibitor, on proliferation of oridonin-treated cells were assessed, and the effects of oridonin combined with LIPUS on PIEZO1 protein expression was evalauted using Western blotting. A C57BL/6J mouse model bearing pancreatic cancer cell xenograft was established and treated with oridonin, LIPUS, or both, and the histological changes in the tumor tissues and tumor cell proliferation were examined with HE staining and immunohistochemistry for Ki67; the changes in GPX4 expression in the tumor tissues were detected using Western blotting and immunofluorescence staining.
RESULTS:
In PANC-1 cells, oridonin treatment significantly inhibited cell proliferation, increased intracellular Fe2+, ROS, and MDA levels, and decreased GSH and ATP levels. Oridonin also resulted in lowered GPX4 and increased HO-1 and Nrf2 protein expression levels in the cells. The combined treatment with LIPUS signficiantly enhanced the inhibitory effect of oridonin on PANC-1 cell viability in vitro and on xenograft growth in the mouse models, resulting also in more obvious reduction of the intensity of Ki67 staining and GPX4 protein expression and more pronounced increase of PIEZO1 protein expression in the tumor tissues in the mouse models.
CONCLUSIONS
LIPUS enhances the effect of oridonin to promote ferroptosis of pancreatic cancer cells by activating PIEZO1 through the Nrf2/HO-1/GPX4 pathway.
Ferroptosis/drug effects*
;
Animals
;
Pancreatic Neoplasms/metabolism*
;
NF-E2-Related Factor 2/metabolism*
;
Humans
;
Cell Line, Tumor
;
Mice
;
Heme Oxygenase-1/metabolism*
;
Diterpenes, Kaurane/pharmacology*
;
Cell Proliferation/drug effects*
;
Mice, Inbred C57BL
;
Phospholipid Hydroperoxide Glutathione Peroxidase
;
Ion Channels/metabolism*
;
Ultrasonic Waves
;
Signal Transduction
9.PDZ-binding kinase as a prognostic biomarker for pancreatic cancer: a pan-cancer analysis and validation in pancreatic adenocarcinoma cells.
Jinguo WANG ; Yang MA ; Zhaoxin LI ; Lifei HE ; Yingze HUANG ; Xiaoming FAN
Journal of Southern Medical University 2025;45(10):2210-2222
OBJECTIVES:
To investigate the prognostic significance of PDZ-binding kinase (PBK) in pan-cancer and its potential as a therapeutic target for pancreatic cancer.
METHODS:
PBK expression levels were investigated in 33 cancer types based on data from TCGA, GEO and CPTAC databases. RT-PCR and Western blotting were employed to examine PBK expression in clinical pancreatic cancer specimens and cell lines. The diagnostic and prognostic value of PBK in pancreatic cancer was evaluated using survival analysis, Cox regression analysis, ROC curve analysis, and clinical correlation studies. Gene enrichment and immune correlation analyses were conducted to explore the potential role of PBK in tumor microenvironment, and its correlation with drug sensitivity was investigated using GDSC and CTRP datasets. In pancreatic cancer BXPC-3 cells, the effects of lentivirus-mediated PBK knockdown on cell proliferation, migration, and invasion were examined using CCK-8, colony formation, and Transwell assays. The interaction between PBK and non-SMC condensin II complex subunit G2 (NCAPG2) was analyzed using co-immunoprecipitation and Western blotting.
RESULTS:
PBK was overexpressed in multiple cancer types, including pancreatic cancer. A high PBK expression was associated with a poor prognosis of the patients and correlated with immune infiltration and alterations in the tumor microenvironment. Elevated PBK expression was positively correlated with the sensitivity to MEK inhibitors (Trametinib) and EGFR inhibitors (Afatinib) but negatively with the sensitivity to Bcl-2 inhibitors (TW37) and niclosamide. In BXPC-3 cells, PBK knockdown significantly suppressed NCAPG2 expression and inhibited cell proliferation, migration, and invasion. Co-immunoprecipitation confirmed a direct binding between PBK and NCAPG2.
CONCLUSIONS
PBK is a key regulator of pancreatic cancer and interacts with NCAPG2 to promote tumor progression, suggesting its value as a potential biomarker and therapeutic target for pancreatic cancer.
Humans
;
Pancreatic Neoplasms/genetics*
;
Prognosis
;
Biomarkers, Tumor/genetics*
;
Cell Line, Tumor
;
Cell Proliferation
;
Adenocarcinoma/metabolism*
;
Tumor Microenvironment
;
Cell Movement
;
Mitogen-Activated Protein Kinase Kinases
10.Evaluation of Fine Needle Biopsy (FNB) for Endoscopic Ultrasound (EUS)-guided tissue acquisition of pancreatic masses to negate the need for rapid on-site evaluation: A randomized control trial
Mark Anthony A. De Lusong ; Nico Nahar I. Pajes
Acta Medica Philippina 2024;58(1):51-56
Background and Objectives:
The benefits of rapid on-site evaluation (ROSE) of endoscopic ultrasound-guided fine needle aspiration (EUS-FNA) of solid masses have not been convincingly shown in large, randomized trials. New equipment using EUS-guided fine needle biopsy (FNB) allows for more material to be acquired that may obviate the need for ROSE. This study aimed to evaluate if EUS-FNB without ROSE was non-inferior to EUS-FNA with ROSE in solid pancreatic masses (SPMs).
Methods:
Patients with SPMs requiring tissue sampling were randomly assigned to undergo either EUS-FNA with ROSE or EUS-FNB without ROSE. The touch-imprint cytology technique was used to perform ROSE. The primary endpoint was diagnostic accuracy and secondary endpoints were specimen quality, complication rates, and procedure time.
Results:
Seventy-eight patients were randomized and analyzed (39 EUS-FNA with ROSE and 39 EUS-FNB without
ROSE). Non-significantly different diagnostic accuracies were noted in both groups (97% with ROSE and 100%
without ROSE, P < 0.371). The bloodiness of histologic samples and complication rates were not significantly different between groups. A significantly shorter mean sampling procedural time was noted for EUS-FNB over EUS-FNA with ROSE (30.4 ± 10.4 vs 35.8 ± 9.8 minutes, P < .02).
Conclusions
EUS-FNB demonstrated equal diagnostic accuracy with shorter procedure times in evaluating SPMs compared to EUS-FNA with ROSE. These new-generation FNB needles may obviate the need for ROSE.
Pancreatic Neoplasms


Result Analysis
Print
Save
E-mail