Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (45)

Search Parameters:
Keywords = PanINs

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
9 pages, 1785 KB  
Article
Immunohistochemical Demonstration of Tuft Cells in Human Acinar-to-Ductal Metaplasia and Pancreatic Intraepithelial Neoplasia
by Kensuke Nakanishi, Mitsuaki Ishida, Kohei Taniguchi, Kenta Hosomi, Jun Arima, Atsushi Tomioka, Mitsuhiro Asakuma, Yoshiharu Miyamoto, Ko Fujimori, Yoshinobu Hirose and Sang-Woong Lee
Biomedicines 2025, 13(8), 1944; https://doi.org/10.3390/biomedicines13081944 - 8 Aug 2025
Viewed by 540
Abstract
Background/Objectives: Acinar-to-ductal metaplasia (ADM) refers to the dedifferentiation or transdifferentiation of pancreatic acinar cells. Recently, ADM has received considerable attention as a potential precursor of pancreatic tumours. Previous studies in mouse models identified tuft cells, chemosensory epithelial cells, in ADM and pancreatic [...] Read more.
Background/Objectives: Acinar-to-ductal metaplasia (ADM) refers to the dedifferentiation or transdifferentiation of pancreatic acinar cells. Recently, ADM has received considerable attention as a potential precursor of pancreatic tumours. Previous studies in mouse models identified tuft cells, chemosensory epithelial cells, in ADM and pancreatic intraepithelial neoplasia (PanIN), both considered precursor lesions of pancreatic ductal adenocarcinoma (PDAC), but not in PDAC. We examined the presence of tuft cells in human ADM and PanIN. Methods: We analysed tissue samples from 29 patients (16 women, 13 men; median age 74 years) who underwent surgical resection for pancreatic tumours. Immunohistochemical staining for the tuft cell marker, POU2F3, was used to detect tuft cells in ADM and PanIN lesions. Results: ADM was present in all patients. POU2F3-positive tuft cells were observed in 46.4% of ADM lesions (327/705) but not in normal pancreatic acini. The number of POU2F3-positive tuft cells per PanIN area were significantly higher in low-grade PanIN (median, 2 cells; range, 0–20 positive cells) than in high-grade PanIN (median, 0 cell; range 0–4 positive cells) (p = 0.0050). The percentage of POU2F3-positive tuft cells per total cells in low-grade PanIN lesions (median, 1.1%; range 0–2.5%) was also significantly higher than that in high-grade PanIN lesions (median, 0%; range 0–1.1%) (p = 0.0044). Conclusions: Our results suggest that tuft cells emerge in human pancreatic acini during ADM, possibly as part of tissue repair following injury. Full article
(This article belongs to the Section Cell Biology and Pathology)
Show Figures

Figure 1

28 pages, 5449 KB  
Article
The Impact of Peroxiredoxin 3 on Molecular Testing, Diagnosis, and Prognosis in Human Pancreatic Ductal Adenocarcinoma
by Anna Kakehashi, Shugo Suzuki, Yusaku Nishidoi, Atsushi Hagihara, Hiroko Ikenaga, Masayuki Shiota, Guiyu Qiu, Ikue Noura, Yuko Kuwae, Arpamas Vachiraarunwong, Masaki Fujioka, Min Gi, Norifumi Kawada and Hideki Wanibuchi
Cancers 2025, 17(13), 2212; https://doi.org/10.3390/cancers17132212 - 1 Jul 2025
Cited by 1 | Viewed by 751
Abstract
Background/Objective: Pancreatic ductal adenocarcinoma (PDAC) is one of the leading causes of cancer death and tumors with an extremely poor prognosis. In the present study, novel biomarker candidates useful for the early diagnosis and prognosis of human invasive PDAC were investigated. Methods: Biomarker [...] Read more.
Background/Objective: Pancreatic ductal adenocarcinoma (PDAC) is one of the leading causes of cancer death and tumors with an extremely poor prognosis. In the present study, novel biomarker candidates useful for the early diagnosis and prognosis of human invasive PDAC were investigated. Methods: Biomarker candidates were first selected based on the proteomic/bioinformatic and clinico-pathological analyses of 10 and 100 patients with PDAC, respectively, operated at Osaka Metropolitan University Hospital (Exp. 1). Next, the expression and secretion of the target protein and its EV mRNA were investigated in pancreatic cancer cells in vitro and in a Balb/c nude mouse model. In addition, the protein and EV mRNA levels of candidate molecules were measured in the blood serum of 36 PDAC and 10 IPMN patients, and diagnostic significance was assessed (Exp. 2). Results: A significant elevation of peroxiredoxin 3 (PRX3), a mitochondrial matrix protein, was found in PDAC via LC-Ms/Ms analysis. In Exp. 1, PRX3 overexpression was found in PDAC and PanIN lesions and was associated with a tumor infiltrative growth pattern (INFc) and poor overall 1-year patient survival. The prognostic value was significantly improved when PRX3 was combined with serum SPan-1 and DUPAN-2 markers in survival analyses. Furthermore, the PRX3 protein and its extracellular vesicle (EV: exosome and oncosome)-incorporated mRNA were secreted at detectable levels from PANC-1, MIAPaCa-2, and SW1990 cells into the blood of Balb/c nude mice bearing tumors. The overexpression of PRX3 was positively correlated with that of cancer stem cell marker CD44 variant 9 (CD44v9), P-Nrf2, and FOXO3a, as well as the generation of reactive oxygen species. In Exp. 2, a significant increase in PRX3 protein and EV mRNA was detected in the blood serum of PDAC subjects compared to IPMN patients and healthy controls. Significantly higher PRX3 protein levels were found in the IPMN group. The elevation of PRX3 EV mRNA was significantly associated with poor patient survival. Conclusions: These results indicate that PRX3 may become a novel early biomarker for PDAC diagnosis and prognosis. Full article
Show Figures

Figure 1

35 pages, 5451 KB  
Review
Innate Immunity and Platelets: Unveiling Their Role in Chronic Pancreatitis and Pancreatic Cancer
by Juliane Blümke, Moritz Schameitat, Atul Verma, Celina Limbecker, Elise Arlt, Sonja M. Kessler, Heike Kielstein, Sebastian Krug, Ivonne Bazwinsky-Wutschke and Monika Haemmerle
Cancers 2025, 17(10), 1689; https://doi.org/10.3390/cancers17101689 - 17 May 2025
Cited by 1 | Viewed by 2352
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive and lethal forms of cancer, characterized by a highly desmoplastic tumor microenvironment. One main risk factor is chronic pancreatitis (CP). Progression of CP to PDAC is greatly influenced by persistent inflammation promoting genomic [...] Read more.
Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive and lethal forms of cancer, characterized by a highly desmoplastic tumor microenvironment. One main risk factor is chronic pancreatitis (CP). Progression of CP to PDAC is greatly influenced by persistent inflammation promoting genomic instability, acinar–ductal metaplasia, and pancreatic intraepithelial neoplasia (PanIN) formation. Components of the extracellular matrix, including immune cells, can modulate this progression phase. This includes cells of the innate immune system, such as natural killer (NK) cells, macrophages, dendritic cells, mast cells, neutrophils, and myeloid-derived suppressor cells (MDSCs), either promoting or inhibiting tumor growth. On one hand, innate immune cells can trigger inflammatory responses that support tumor progression by releasing cytokines and growth factors, fostering tumor cell proliferation, invasion, and metastasis. On the other hand, they can also activate immune surveillance mechanisms, which can limit tumor development. For example, NK cells are cytotoxic innate lymphoid cells that are able to kill tumor cells, and active dendritic cells are crucial for a functioning anti-tumor immune response. In contrast, mast cells and MDSCs rather support a pro-tumorigenic tumor microenvironment that is additionally sustained by platelets. Once thought to play a role in hemostasis only, platelets are now recognized as key players in inflammation and cancer progression. By releasing cytokines, growth factors, and pro-angiogenic mediators, platelets help shape an immunosuppressive microenvironment that promotes fibrotic remodeling, tumor initiation, progression, metastasis, and immune evasion. Neutrophils and macrophages exist in different functional subtypes that can both act pro- and anti-tumorigenic. Understanding the complex interactions between innate immune cells, platelets, and early precursor lesions, as well as PDAC cells, is crucial for developing new therapeutic approaches that can harness the immune and potentially also the coagulation system to target and eliminate tumors, offering hope for improved patient outcomes. Full article
(This article belongs to the Special Issue Management of Pancreatic Cancer)
Show Figures

Figure 1

15 pages, 1839 KB  
Article
Molecular Pathway and Immune Profile Analysis of IPMN-Derived Versus PanIN-Derived Pancreatic Ductal Adenocarcinomas
by Margaret A. Park, Kristyn Gumpper-Fedus, Somashekar G. Krishna, Maria C. Genilo-Delgado, Stephen Brantley, Phil A. Hart, Mary E. Dillhoff, Maria F. Gomez, Toni L. Basinski, Shaffer R. Mok, Anjuli K. Luthra, Jason B. Fleming, Amir Mohammadi, Barbara A. Centeno, Kun Jiang, Aleksandra Karolak, Daniel Jeong, Dung-Tsa Chen, Paul A. Stewart, Jamie K. Teer, Zobeida Cruz-Monserrate and Jennifer B. Permuthadd Show full author list remove Hide full author list
Int. J. Mol. Sci. 2024, 25(23), 13164; https://doi.org/10.3390/ijms252313164 - 7 Dec 2024
Cited by 1 | Viewed by 2304
Abstract
Intraductal papillary mucinous neoplasms (IPMN) are commonly detected pancreatic cysts that may transform into pancreatic ductal adenocarcinoma (PDAC). Predicting which IPMNs will progress to PDAC remains a clinical challenge. Moreover, identifying those clinically evident IPMNs for which a surveillance approach is best is [...] Read more.
Intraductal papillary mucinous neoplasms (IPMN) are commonly detected pancreatic cysts that may transform into pancreatic ductal adenocarcinoma (PDAC). Predicting which IPMNs will progress to PDAC remains a clinical challenge. Moreover, identifying those clinically evident IPMNs for which a surveillance approach is best is a dire clinical need. Therefore, we aimed to identify molecular signatures that distinguished between PDAC with and without clinical evidence of an IPMN to identify novel molecular pathways related to IPMN-derived PDAC that could help guide biomarker development. Data from the Oncology Research Information Exchange Network (ORIEN) multi-institute sequencing project were utilized to analyze 66 PDAC cases from Moffitt Cancer Center and The Ohio State University Wexner Medical Center, for which tumor whole transcriptome sequencing datasets were generated. Cases were classified based on whether a tumor had originated from an IPMN (n = 16) or presumably through the pancreatic intraepithelial neoplasia (PanIN) pathway (n = 50). We then performed differential expression and pathway analysis using Gene-Set Enrichment Analysis (GSEA) and Pathway Analysis with Down-weighted Genes (PADOG) algorithms. We also analyzed immune profiles using the Tumor-Immune Microenvironment Deconvolution web portal for Bulk Transcriptomics (TIMEx). Both GSEA and TIMEx indicate that PanIN-derived PDAC tumors enrich inflammatory pathways (complement, hedgehog signaling, coagulation, inflammatory response, apical surface, IL-2/STAT5, IL-6/STAT3, EMT, KRAS signaling, apical junction, IFN-gamma, allograft rejection) and are comparatively richer in almost all immune cell types than those from IPMN-derived PDAC. IPMN-derived tumors were enriched for metabolic and energy-generating pathways (oxidative phosphorylation, unfolded protein response, pancreas beta cells, adipogenesis, fatty acid metabolism, protein secretion), and the most significantly upregulated genes (padj < 0.001) included mucin 2 (MUC2) and gastrokine-2 (GKN2). Further, the metabolic-linked gene signature enriched in the IPMN-derived samples is associated with a cluster of early-stage and long-survival (top 4th quartile) PDAC cases from The Cancer Genome Atlas (TCGA) expression database. Our data suggest that IPMN-derived and PanIN-derived PDACs differ in the expression of immune profiles and metabolic pathways. These initial findings warrant validation and follow-up to develop biomarker-based strategies for early PDAC detection and treatment. Full article
(This article belongs to the Section Molecular Biology)
Show Figures

Figure 1

22 pages, 2651 KB  
Article
Retrospective Investigator-Initiated Trial on Tocopherol Acetate Vaginal Administration in Pre-and Postmenopausal Women
by Noemi Venditti, Giulio Petronio Petronio, Antonio Guarnieri, Laura Pietrangelo, Angela Spicciato, Alessio Colalillo, Giovanna Paola Sabusco, Dionisio Franco Barattini, Aldo Di Franco, Stefano Papini, Francesco Cosentino and Roberto Di Marco
Diseases 2024, 12(10), 237; https://doi.org/10.3390/diseases12100237 - 2 Oct 2024
Viewed by 1916
Abstract
Background: Menopause, a natural phase in a woman’s life, often adversely affects physical, mental, sexual, and emotional well-being due to low estrogen levels. This study examines the impact of vaginal ovules with tocopherol acetate (Filme Gyno-V® ovules, manufactured by Panin Srl and [...] Read more.
Background: Menopause, a natural phase in a woman’s life, often adversely affects physical, mental, sexual, and emotional well-being due to low estrogen levels. This study examines the impact of vaginal ovules with tocopherol acetate (Filme Gyno-V® ovules, manufactured by Panin Srl and distributed by Hulka Srl, Italy), 500 mg per ovule, on vaginal health in pre- and menopausal women. Methods: Fifty women aged 50–70 were divided into menopausal (28) and premenopausal (22) cohorts and treated with the ovules for two weeks, with assessments before and after treatment. Results: The findings showed that distressing symptoms of vaginal atrophy, such as dryness, itching, and pain during intercourse, were resolved post-treatment. A molecular analysis revealed a reduction in Escherichia coli in both cohorts and an increase in three species of Lactobacillus in premenopausal patients. Conclusions: This study concludes that Filme Gyno-V ovules may benefit vaginal health by alleviating atrophy symptoms and promoting healthy vaginal microbiota. Full article
Show Figures

Figure 1

15 pages, 3923 KB  
Article
CCK Receptor Inhibition Reduces Pancreatic Tumor Fibrosis and Promotes Nanoparticle Delivery
by Thomas Abraham, Michael Armold, Christopher McGovern, John F. Harms, Matthew C. Darok, Christopher Gigliotti, Bernadette Adair, Jennifer L. Gray, Deborah F. Kelly, James H. Adair and Gail L. Matters
Biomedicines 2024, 12(5), 1024; https://doi.org/10.3390/biomedicines12051024 - 7 May 2024
Cited by 1 | Viewed by 2497
Abstract
The poor prognosis for pancreatic ductal adenocarcinoma (PDAC) patients is due in part to the highly fibrotic nature of the tumors that impedes delivery of therapeutics, including nanoparticles (NPs). Our prior studies demonstrated that proglumide, a cholecystokinin receptor (CCKR) antagonist, reduced fibrosis pervading [...] Read more.
The poor prognosis for pancreatic ductal adenocarcinoma (PDAC) patients is due in part to the highly fibrotic nature of the tumors that impedes delivery of therapeutics, including nanoparticles (NPs). Our prior studies demonstrated that proglumide, a cholecystokinin receptor (CCKR) antagonist, reduced fibrosis pervading PanIN lesions in mice. Here, we further detail how the reduced fibrosis elicited by proglumide achieves the normalization of the desmoplastic tumor microenvironment (TME) and improves nanoparticle uptake. One week following the orthotopic injection of PDAC cells, mice were randomized to normal or proglumide-treated water for 3–6 weeks. Tumors were analyzed ex vivo for fibrosis, vascularity, stellate cell activation, vascular patency, and nanoparticle distribution. The histological staining and three-dimensional imaging of tumors each indicated a reduction in stromal collagen in proglumide-treated mice. Proglumide treatment increased tumor vascularity and decreased the activation of cancer-associated fibroblasts (CAFs). Additionally, PANC-1 cells with the shRNA-mediated knockdown of the CCK2 receptor showed an even greater reduction in collagen, indicating the CCK2 receptors on tumor cells contribute to the desmoplastic TME. Proglumide-mediated reduction in fibrosis also led to functional changes in the TME as evidenced by the enhanced intra-tumoral distribution of small (<12 nm) Rhodamine-loaded nanoparticles. The documented in vivo, tumor cell-intrinsic anti-fibrotic effects of CCK2R blockade in both an immunocompetent syngeneic murine PDAC model as well as a human PDAC xenograft model demonstrates that CCK2R antagonists, such as proglumide, can improve the delivery of nano-encapsulated therapeutics or imaging agents to pancreatic tumors. Full article
(This article belongs to the Special Issue Early Diagnosis and Targeted Therapy of Pancreatic Cancer)
Show Figures

Figure 1

19 pages, 3969 KB  
Article
Cytokine CCL9 Mediates Oncogenic KRAS-Induced Pancreatic Acinar-to-Ductal Metaplasia by Promoting Reactive Oxygen Species and Metalloproteinases
by Geou-Yarh Liou, Crystal J. Byrd, Peter Storz and Justin K. Messex
Int. J. Mol. Sci. 2024, 25(9), 4726; https://doi.org/10.3390/ijms25094726 - 26 Apr 2024
Cited by 5 | Viewed by 2470
Abstract
Pancreatic ductal adenocarcinoma (PDAC) can originate from acinar-to-ductal metaplasia (ADM). Pancreatic acini harboring oncogenic Kras mutations are transdifferentiated to a duct-like phenotype that further progresses to become pancreatic intraepithelial neoplasia (PanIN) lesions, giving rise to PDAC. Although ADM formation is frequently observed in [...] Read more.
Pancreatic ductal adenocarcinoma (PDAC) can originate from acinar-to-ductal metaplasia (ADM). Pancreatic acini harboring oncogenic Kras mutations are transdifferentiated to a duct-like phenotype that further progresses to become pancreatic intraepithelial neoplasia (PanIN) lesions, giving rise to PDAC. Although ADM formation is frequently observed in KrasG12D transgenic mouse models of PDAC, the exact mechanisms of how oncogenic KrasG12D regulates this process remain an enigma. Herein, we revealed a new downstream target of oncogenic Kras, cytokine CCL9, during ADM formation. Higher levels of CCL9 and its receptors, CCR1 and CCR3, were detected in ADM regions of the pancreas in p48cre:KrasG12D mice and human PDAC patients. Knockdown of CCL9 in KrasG12D-expressed pancreatic acini reduced KrasG12D-induced ADM in a 3D organoid culture system. Moreover, exogenously added recombinant CCL9 and overexpression of CCL9 in primary pancreatic acini induced pancreatic ADM. We also showed that, functioning as a downstream target of KrasG12D, CCL9 promoted pancreatic ADM through upregulation of the intracellular levels of reactive oxygen species (ROS) and metalloproteinases (MMPs), including MMP14, MMP3 and MMP2. Blockade of MMPs via its generic inhibitor GM6001 or knockdown of specific MMP such as MMP14 and MMP3 decreased CCL9-induced pancreatic ADM. In p48cre:KrasG12D transgenic mice, blockade of CCL9 through its specific neutralizing antibody attenuated pancreatic ADM structures and PanIN lesion formation. Furthermore, it also diminished infiltrating macrophages and expression of MMP14, MMP3 and MMP2 in the ADM areas. Altogether, our results provide novel mechanistic insight into how oncogenic Kras enhances pancreatic ADM through its new downstream target molecule, CCL9, to initiate PDAC. Full article
Show Figures

Figure 1

23 pages, 7386 KB  
Article
Upregulated Matrisomal Proteins and Extracellular Matrix Mechanosignaling Underlie Obesity-Associated Promotion of Pancreatic Ductal Adenocarcinoma
by Richard T. Waldron, Aurelia Lugea, Hui-Hua Chang, Hsin-Yuan Su, Crystal Quiros, Michael S. Lewis, Mingtian Che, V. Krishnan Ramanujan, Enrique Rozengurt, Guido Eibl and Stephen J. Pandol
Cancers 2024, 16(8), 1593; https://doi.org/10.3390/cancers16081593 - 21 Apr 2024
Cited by 1 | Viewed by 3676
Abstract
Diet-induced obesity (DIO) promotes pancreatic ductal adenocarcinoma (PDAC) in mice expressing KRasG12D in the pancreas (KC mice), but the precise mechanisms remain unclear. Here, we performed multiplex quantitative proteomic and phosphoproteomic analysis by liquid chromatography–tandem mass spectrometry and further bioinformatic and spatial analysis [...] Read more.
Diet-induced obesity (DIO) promotes pancreatic ductal adenocarcinoma (PDAC) in mice expressing KRasG12D in the pancreas (KC mice), but the precise mechanisms remain unclear. Here, we performed multiplex quantitative proteomic and phosphoproteomic analysis by liquid chromatography–tandem mass spectrometry and further bioinformatic and spatial analysis of pancreas tissues from control-fed versus DIO KC mice after 3, 6, and 9 months. Normal pancreatic parenchyma and associated proteins were steadily eliminated and the novel proteins, phosphoproteins, and signaling pathways associated with PDAC tumorigenesis increased until 6 months, when most males exhibited cancer, but females did not. Differentially expressed proteins and phosphoproteins induced by DIO revealed the crucial functional role of matrisomal proteins, which implies the roles of upstream regulation by TGFβ, extracellular matrix-receptor signaling to downstream PI3K-Akt-mTOR-, MAPK-, and Yap/Taz activation, and crucial effects in the tumor microenvironment such as metabolic alterations and signaling crosstalk between immune cells, cancer-associated fibroblasts (CAFs), and tumor cells. Staining tissues from KC mice localized the expression of several prognostic PDAC biomarkers and elucidated tumorigenic features, such as robust macrophage infiltration, acinar–ductal metaplasia, mucinous PanIN, distinct nonmucinous atypical flat lesions (AFLs) surrounded by smooth muscle actin-positive CAFs, invasive tumors with epithelial–mesenchymal transition arising close to AFLs, and expanding deserted areas by 9 months. We next used Nanostring GeoMX to characterize the early spatial distribution of specific immune cell subtypes in distinct normal, stromal, and PanIN areas. Taken together, these data richly contextualize DIO promotion of Kras-driven PDAC tumorigenesis and provide many novel insights into the signaling pathways and processes involved. Full article
(This article belongs to the Collection Recent Advances in Pancreatic Ductal Adenocarcinoma)
Show Figures

Figure 1

17 pages, 3055 KB  
Article
A Comparison of Spatial and Phenotypic Immune Profiles of Pancreatic Ductal Adenocarcinoma and Its Precursor Lesions
by Thomas Enzler, Jiaqi Shi, Jake McGue, Brian D. Griffith, Lei Sun, Vaibhav Sahai, Hari Nathan and Timothy L. Frankel
Int. J. Mol. Sci. 2024, 25(5), 2953; https://doi.org/10.3390/ijms25052953 - 3 Mar 2024
Cited by 3 | Viewed by 2535
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a devastating disease with a 5-year survival rate of 12.5%. PDAC predominantly arises from non-cystic pancreatic intraepithelial neoplasia (PanIN) and cystic intraductal papillary mucinous neoplasm (IPMN). We used multiplex immunofluorescence and computational imaging technology to characterize, map, and [...] Read more.
Pancreatic ductal adenocarcinoma (PDAC) is a devastating disease with a 5-year survival rate of 12.5%. PDAC predominantly arises from non-cystic pancreatic intraepithelial neoplasia (PanIN) and cystic intraductal papillary mucinous neoplasm (IPMN). We used multiplex immunofluorescence and computational imaging technology to characterize, map, and compare the immune microenvironments (IMEs) of PDAC and its precursor lesions. We demonstrate that the IME of IPMN was abundantly infiltrated with CD8+ T cells and PD-L1-positive antigen-presenting cells (APCs), whereas the IME of PanIN contained fewer CD8+ T cells and fewer PD-L1-positive APCs but elevated numbers of immunosuppressive regulatory T cells (Tregs). Thus, immunosuppression in IPMN and PanIN seems to be mediated by different mechanisms. While immunosuppression in IPMN is facilitated by PD-L1 expression on APCs, Tregs seem to play a key role in PanIN. Our findings suggest potential immunotherapeutic interventions for high-risk precursor lesions, namely, targeting PD-1/PD-L1 in IPMN and CTLA-4-positive Tregs in PanIN to restore immunosurveillance and prevent progression to cancer. Tregs accumulate with malignant transformation, as observed in PDAC, and to a lesser extent in IPMN-associated PDAC (IAPA). High numbers of Tregs in the microenvironment of PDAC went along with a markedly decreased interaction between CD8+ T cells and cancerous epithelial cells (ECs), highlighting the importance of Tregs as key players in immunosuppression in PDAC. We found evidence that a defect in antigen presentation, further aggravated by PD-L1 expression on APC, may contribute to immunosuppression in IAPA, suggesting a role for PD-L1/PD-1 immune checkpoint inhibitors in the treatment of IAPA. Full article
Show Figures

Figure 1

24 pages, 11647 KB  
Article
Krüppel-like Factor 5 Plays an Important Role in the Pathogenesis of Chronic Pancreatitis
by Maryam Alavi, Ana Mejia-Bautista, Meiyi Tang, Jela Bandovic, Avi Z. Rosenberg and Agnieszka B. Bialkowska
Cancers 2023, 15(22), 5427; https://doi.org/10.3390/cancers15225427 - 15 Nov 2023
Cited by 1 | Viewed by 2168
Abstract
Chronic pancreatitis results in the formation of pancreatic intraepithelial neoplasia (PanIN) and poses a risk of developing pancreatic cancer. Our previous study demonstrated that Krüppel-like factor 5 (KLF5) is necessary for forming acinar-to-ductal metaplasia (ADM) in acute pancreatitis. Here, we investigated the role [...] Read more.
Chronic pancreatitis results in the formation of pancreatic intraepithelial neoplasia (PanIN) and poses a risk of developing pancreatic cancer. Our previous study demonstrated that Krüppel-like factor 5 (KLF5) is necessary for forming acinar-to-ductal metaplasia (ADM) in acute pancreatitis. Here, we investigated the role of KLF5 in response to chronic injury in the pancreas. Human tissues originating from chronic pancreatitis patients showed increased levels of epithelial KLF5. An inducible genetic model combining the deletion of Klf5 and the activation of KrasG12D mutant expression in pancreatic acinar cells together with chemically induced chronic pancreatitis was used. The chronic injury resulted in increased levels of KLF5 in both control and KrasG12D mutant mice. Furthermore, it led to numerous ADM and PanIN lesions and extensive fibrosis in the KRAS mutant mice. In contrast, pancreata with Klf5 loss (with or without KrasG12D) failed to develop ADM, PanIN, or significant fibrosis. Furthermore, the deletion of Klf5 reduced the expression level of cytokines and fibrotic components such as Il1b, Il6, Tnf, Tgfb1, Timp1, and Mmp9. Notably, using ChIP-PCR, we showed that KLF5 binds directly to the promoters of Il1b, Il6, and Tgfb1 genes. In summary, the inactivation of Klf5 inhibits ADM and PanIN formation and the development of pancreatic fibrosis. Full article
(This article belongs to the Section Molecular Cancer Biology)
Show Figures

Figure 1

15 pages, 8929 KB  
Article
The Expression of Two Distinct Sets of Glycolytic Enzymes Reveals Differential Effects of Glycolytic Reprogramming on Pancreatic Ductal Tumorigenesis in Mice
by Yannan Zhang, Fangfang Zheng, Fan Wang, Xingqian Liu, Cong Xiang, Shiyu Fu, Kun Shen and Geng Liu
Biomedicines 2023, 11(11), 2962; https://doi.org/10.3390/biomedicines11112962 - 2 Nov 2023
Cited by 2 | Viewed by 1978
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is associated with enhanced aerobic glycolysis through elevated glucose uptake and the upregulated expression of genes encoding rate-limiting glycolytic enzymes. However, the direct impact of altered glycolytic pathways on pancreatic tumor progression has not been thoroughly investigated. Here, we [...] Read more.
Pancreatic ductal adenocarcinoma (PDAC) is associated with enhanced aerobic glycolysis through elevated glucose uptake and the upregulated expression of genes encoding rate-limiting glycolytic enzymes. However, the direct impact of altered glycolytic pathways on pancreatic tumor progression has not been thoroughly investigated. Here, we utilized two strains of BAC transgenic mice with pancreatic expression of two distinct sets of glycolytic genes each arranged in a polycistronic fashion (PFKFB3-HK2-GLUT1 and LDHA-PDK1, respectively) to investigate the role of altered glycolysis on the development of pancreatic ductal tumor development in the Pdx1-Cre; LSL-KrasG12D mice. The overexpression of the two sets of glycolytic genes exhibited no significant effects on tumor development in the 4–5-month-old mice (the PanIN2 lesions stage). In the 9–10-month-old mice, the overexpression of PFKFB3-HK2-GLUT1 significantly accelerated PanIN3 progression, exhibiting elevated levels of ductal cell marker CK19 and tumor fibrosis. Surprisingly, the overexpression of LDHA-PDK1 significantly attenuated the progression of PanIN3 in the 9–10-month-old mice with significantly downregulated levels of CK19 and fibrosis. Therefore, distinct set of glycolytic enzymes that are involved in different glycolytic routes exhibited contrasting effects on pancreatic ductal tumor development depending on the tumor stages, providing novel insights into the complexity of the glycolytic pathway in the perspective of PDAC development and therapy. Full article
(This article belongs to the Section Cancer Biology and Oncology)
Show Figures

Figure 1

15 pages, 1665 KB  
Review
Cholesterol Metabolism in Pancreatic Cancer
by Artur Rebelo, Jörg Kleeff and Yoshiaki Sunami
Cancers 2023, 15(21), 5177; https://doi.org/10.3390/cancers15215177 - 27 Oct 2023
Cited by 13 | Viewed by 4687
Abstract
Pancreatic cancer’s substantial impact on cancer-related mortality, responsible for 8% of cancer deaths and ranking fourth in the US, persists despite advancements, with a five-year relative survival rate of only 11%. Forecasts predict a 70% surge in new cases and a 72% increase [...] Read more.
Pancreatic cancer’s substantial impact on cancer-related mortality, responsible for 8% of cancer deaths and ranking fourth in the US, persists despite advancements, with a five-year relative survival rate of only 11%. Forecasts predict a 70% surge in new cases and a 72% increase in global pancreatic cancer-related deaths by 2040. This review explores the intrinsic metabolic reprogramming of pancreatic cancer, focusing on the mevalonate pathway, including cholesterol biosynthesis, transportation, targeting strategies, and clinical studies. The mevalonate pathway, central to cellular metabolism, significantly shapes pancreatic cancer progression. Acetyl coenzyme A (Acetyl-CoA) serves a dual role in fatty acid and cholesterol biosynthesis, fueling acinar-to-ductal metaplasia (ADM) and pancreatic intraepithelial neoplasia (PanIN) development. Enzymes, including acetoacetyl-CoA thiolase, 3-hydroxy-3methylglutaryl-CoA (HMG-CoA) synthase, and HMG-CoA reductase, are key enzymes in pancreatic cancer. Inhibiting HMG-CoA reductase, e.g., by using statins, shows promise in delaying PanIN progression and impeding pancreatic cancer. Dysregulation of cholesterol modification, uptake, and transport significantly impacts tumor progression, with Sterol O-acyltransferase 1 (SOAT1) driving cholesterol ester (CE) accumulation and disrupted low-density lipoprotein receptor (LDLR) expression contributing to cancer recurrence. Apolipoprotein E (ApoE) expression in tumor stroma influences immune suppression. Clinical trials targeting cholesterol metabolism, including statins and SOAT1 inhibitors, exhibit potential anti-tumor effects, and combination therapies enhance efficacy. This review provides insights into cholesterol metabolism’s convergence with pancreatic cancer, shedding light on therapeutic avenues and ongoing clinical investigations. Full article
(This article belongs to the Section Molecular Cancer Biology)
Show Figures

Figure 1

21 pages, 1520 KB  
Review
Acinar-to-Ductal Metaplasia (ADM): On the Road to Pancreatic Intraepithelial Neoplasia (PanIN) and Pancreatic Cancer
by Louis Marstrand-Daucé, Diane Lorenzo, Anaïs Chassac, Pascal Nicole, Anne Couvelard and Cécile Haumaitre
Int. J. Mol. Sci. 2023, 24(12), 9946; https://doi.org/10.3390/ijms24129946 - 9 Jun 2023
Cited by 37 | Viewed by 12605
Abstract
Adult pancreatic acinar cells show high plasticity allowing them to change in their differentiation commitment. Pancreatic acinar-to-ductal metaplasia (ADM) is a cellular process in which the differentiated pancreatic acinar cells transform into duct-like cells. This process can occur as a result of cellular [...] Read more.
Adult pancreatic acinar cells show high plasticity allowing them to change in their differentiation commitment. Pancreatic acinar-to-ductal metaplasia (ADM) is a cellular process in which the differentiated pancreatic acinar cells transform into duct-like cells. This process can occur as a result of cellular injury or inflammation in the pancreas. While ADM is a reversible process allowing pancreatic acinar regeneration, persistent inflammation or injury can lead to the development of pancreatic intraepithelial neoplasia (PanIN), which is a common precancerous lesion that precedes pancreatic ductal adenocarcinoma (PDAC). Several factors can contribute to the development of ADM and PanIN, including environmental factors such as obesity, chronic inflammation and genetic mutations. ADM is driven by extrinsic and intrinsic signaling. Here, we review the current knowledge on the cellular and molecular biology of ADM. Understanding the cellular and molecular mechanisms underlying ADM is critical for the development of new therapeutic strategies for pancreatitis and PDAC. Identifying the intermediate states and key molecules that regulate ADM initiation, maintenance and progression may help the development of novel preventive strategies for PDAC. Full article
(This article belongs to the Special Issue Digestive Inflammation and New Therapeutical Targets)
Show Figures

Figure 1

16 pages, 554 KB  
Review
KRAS-Dependency in Pancreatic Ductal Adenocarcinoma: Mechanisms of Escaping in Resistance to KRAS Inhibitors and Perspectives of Therapy
by Enrico Gurreri, Giannicola Genovese, Luigi Perelli, Antonio Agostini, Geny Piro, Carmine Carbone and Giampaolo Tortora
Int. J. Mol. Sci. 2023, 24(11), 9313; https://doi.org/10.3390/ijms24119313 - 26 May 2023
Cited by 15 | Viewed by 5443
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is still one of the deadliest cancers in oncology because of its increasing incidence and poor survival rate. More than 90% of PDAC patients are KRAS mutated (KRASmu), with KRASG12D and KRASG12V being the most common mutations. Despite this [...] Read more.
Pancreatic ductal adenocarcinoma (PDAC) is still one of the deadliest cancers in oncology because of its increasing incidence and poor survival rate. More than 90% of PDAC patients are KRAS mutated (KRASmu), with KRASG12D and KRASG12V being the most common mutations. Despite this critical role, its characteristics have made direct targeting of the RAS protein extremely difficult. KRAS regulates development, cell growth, epigenetically dysregulated differentiation, and survival in PDAC through activation of key downstream pathways, such as MAPK-ERK and PI3K-AKT-mammalian target of rapamycin (mTOR) signaling, in a KRAS-dependent manner. KRASmu induces the occurrence of acinar-to-ductal metaplasia (ADM) and pancreatic intraepithelial neoplasia (PanIN) and leads to an immunosuppressive tumor microenvironment (TME). In this context, the oncogenic mutation of KRAS induces an epigenetic program that leads to the initiation of PDAC. Several studies have identified multiple direct and indirect inhibitors of KRAS signaling. Therefore, KRAS dependency is so essential in KRASmu PDAC that cancer cells have secured several compensatory escape mechanisms to counteract the efficacy of KRAS inhibitors, such as activation of MEK/ERK signaling or YAP1 upregulation. This review will provide insights into KRAS dependency in PDAC and analyze recent data on inhibitors of KRAS signaling, focusing on how cancer cells establish compensatory escape mechanisms. Full article
(This article belongs to the Special Issue Cancer Prevention with Molecular Target Therapies 4.0)
Show Figures

Figure 1

15 pages, 1033 KB  
Review
Artificial Intelligence in the Diagnosis and Treatment of Pancreatic Cystic Lesions and Adenocarcinoma
by Joanna Jiang, Wei-Lun Chao, Stacey Culp and Somashekar G. Krishna
Cancers 2023, 15(9), 2410; https://doi.org/10.3390/cancers15092410 - 22 Apr 2023
Cited by 26 | Viewed by 7198
Abstract
Pancreatic cancer is projected to become the second leading cause of cancer-related mortality in the United States by 2030. This is in part due to the paucity of reliable screening and diagnostic options for early detection. Amongst known pre-malignant pancreatic lesions, pancreatic intraepithelial [...] Read more.
Pancreatic cancer is projected to become the second leading cause of cancer-related mortality in the United States by 2030. This is in part due to the paucity of reliable screening and diagnostic options for early detection. Amongst known pre-malignant pancreatic lesions, pancreatic intraepithelial neoplasia (PanIN) and intraductal papillary mucinous neoplasms (IPMNs) are the most prevalent. The current standard of care for the diagnosis and classification of pancreatic cystic lesions (PCLs) involves cross-sectional imaging studies and endoscopic ultrasound (EUS) and, when indicated, EUS-guided fine needle aspiration and cyst fluid analysis. However, this is suboptimal for the identification and risk stratification of PCLs, with accuracy of only 65–75% for detecting mucinous PCLs. Artificial intelligence (AI) is a promising tool that has been applied to improve accuracy in screening for solid tumors, including breast, lung, cervical, and colon cancer. More recently, it has shown promise in diagnosing pancreatic cancer by identifying high-risk populations, risk-stratifying premalignant lesions, and predicting the progression of IPMNs to adenocarcinoma. This review summarizes the available literature on artificial intelligence in the screening and prognostication of precancerous lesions in the pancreas, and streamlining the diagnosis of pancreatic cancer. Full article
Show Figures

Figure 1

Back to TopTop