Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 613
Filtrer
1.
Mol Med Rep ; 30(3)2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38994764

RÉSUMÉ

Pancreatic ductal adenocarcinoma (PDAC) is an extremely aggressive form of cancer with a low survival rate. A successful treatment strategy should not be limited to targeting cancer cells alone, but should adopt a more comprehensive approach, taking into account other influential factors. These include the extracellular matrix (ECM) and immune microenvironment, both of which are integral components of the tumor microenvironment. The present review describes the roles of pancreatic stellate cells, differentiated cancer­associated fibroblasts and the interleukin family, either independently or in combination, in the progression of precursor lesions in pancreatic intraepithelial neoplasia and PDAC. These elements contribute to ECM deposition and immunosuppression in PDAC. Therapeutic strategies that integrate interleukin and/or stromal blockade for PDAC immunomodulation and fibrogenesis have yielded inconsistent results. A deeper comprehension of the intricate interplay between fibrosis, and immune responses could pave the way for more effective treatment targets, by elucidating the mechanisms and causes of ECM fibrosis during PDAC progression.


Sujet(s)
Carcinome du canal pancréatique , Fibrose , Interleukines , Tumeurs du pancréas , Cellules stellaires pancréatiques , Microenvironnement tumoral , Humains , Carcinome du canal pancréatique/immunologie , Carcinome du canal pancréatique/anatomopathologie , Carcinome du canal pancréatique/métabolisme , Cellules stellaires pancréatiques/métabolisme , Cellules stellaires pancréatiques/anatomopathologie , Microenvironnement tumoral/immunologie , Tumeurs du pancréas/immunologie , Tumeurs du pancréas/anatomopathologie , Tumeurs du pancréas/métabolisme , Interleukines/métabolisme , Interleukines/immunologie , Animaux , Matrice extracellulaire/métabolisme , Fibroblastes associés au cancer/métabolisme , Fibroblastes associés au cancer/immunologie , Fibroblastes associés au cancer/anatomopathologie
2.
Nat Commun ; 15(1): 6043, 2024 Jul 18.
Article de Anglais | MEDLINE | ID: mdl-39025845

RÉSUMÉ

Pancreatic ductal adenocarcinoma (PDAC) is a devastating cancer with dismal prognosis due to distant metastasis, even in the early stage. Using RNA sequencing and multiplex immunofluorescence, here we find elevated expression of mixed lineage kinase domain-like pseudo-kinase (MLKL) and enhanced necroptosis pathway in PDAC from early liver metastasis T-stage (T1M1) patients comparing with non-metastatic (T1M0) patients. Mechanistically, MLKL-driven necroptosis recruits macrophages, enhances the tumor CD47 'don't eat me' signal, and induces macrophage extracellular traps (MET) formation for CXCL8 activation. CXCL8 further initiates epithelial-mesenchymal transition (EMT) and upregulates ICAM-1 expression to promote endothelial adhesion. METs also degrades extracellular matrix, that eventually supports PDAC liver metastasis. Meanwhile, targeting necroptosis and CD47 reduces liver metastasis in vivo. Our study thus reveals that necroptosis facilitates PDAC metastasis by evading immune surveillance, and also suggest that CD47 blockade, combined with MLKL inhibitor GW806742X, may be a promising neoadjuvant immunotherapy for overcoming the T1M1 dilemma and reviving the opportunity for radical surgery.


Sujet(s)
Antigènes CD47 , Carcinome du canal pancréatique , Transition épithélio-mésenchymateuse , Pièges extracellulaires , Tumeurs du foie , Macrophages , Nécroptose , Tumeurs du pancréas , Protein kinases , Humains , Tumeurs du pancréas/anatomopathologie , Tumeurs du pancréas/métabolisme , Tumeurs du pancréas/génétique , Tumeurs du pancréas/immunologie , Tumeurs du foie/secondaire , Tumeurs du foie/métabolisme , Animaux , Carcinome du canal pancréatique/anatomopathologie , Carcinome du canal pancréatique/métabolisme , Carcinome du canal pancréatique/immunologie , Carcinome du canal pancréatique/génétique , Souris , Macrophages/métabolisme , Macrophages/immunologie , Lignée cellulaire tumorale , Antigènes CD47/métabolisme , Antigènes CD47/génétique , Protein kinases/métabolisme , Pièges extracellulaires/métabolisme , Molécule-1 d'adhérence intercellulaire/métabolisme , Molécule-1 d'adhérence intercellulaire/génétique , Mâle , Transduction du signal , Femelle , Acrylamides , Sulfonamides
3.
Nat Commun ; 15(1): 6162, 2024 Jul 22.
Article de Anglais | MEDLINE | ID: mdl-39039076

RÉSUMÉ

Senescent cells within tumors and their stroma exert complex pro- and anti-tumorigenic functions. However, the identities and traits of these cells, and the potential for improving cancer therapy through their targeting, remain poorly characterized. Here, we identify a senescent subset within previously-defined cancer-associated fibroblasts (CAFs) in pancreatic ductal adenocarcinomas (PDAC) and in premalignant lesions in mice and humans. Senescent CAFs isolated from mouse and humans expressed elevated levels of immune-regulatory genes. Depletion of senescent CAFs, either genetically or using the Bcl-2 inhibitor ABT-199 (venetoclax), increased the proportion of activated CD8+ T cells in mouse pancreatic carcinomas, whereas induction of CAF senescence had the opposite effect. Combining ABT-199 with an immune checkpoint therapy regimen significantly reduced mouse tumor burden. These results indicate that senescent CAFs in PDAC stroma limit the numbers of activated cytotoxic CD8+ T cells, and suggest that their targeted elimination through senolytic treatment may enhance immunotherapy.


Sujet(s)
Lymphocytes T CD8+ , Fibroblastes associés au cancer , Carcinome du canal pancréatique , Vieillissement de la cellule , Immunothérapie , Tumeurs du pancréas , Sulfonamides , Animaux , Fibroblastes associés au cancer/immunologie , Fibroblastes associés au cancer/métabolisme , Fibroblastes associés au cancer/anatomopathologie , Tumeurs du pancréas/immunologie , Tumeurs du pancréas/thérapie , Tumeurs du pancréas/anatomopathologie , Tumeurs du pancréas/génétique , Lymphocytes T CD8+/immunologie , Souris , Humains , Carcinome du canal pancréatique/immunologie , Carcinome du canal pancréatique/thérapie , Carcinome du canal pancréatique/anatomopathologie , Carcinome du canal pancréatique/génétique , Vieillissement de la cellule/immunologie , Immunothérapie/méthodes , Sulfonamides/pharmacologie , Sulfonamides/usage thérapeutique , Souris de lignée C57BL , Lignée cellulaire tumorale , Activation des lymphocytes/immunologie , Femelle , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Adénocarcinome/immunologie , Adénocarcinome/thérapie , Adénocarcinome/génétique , Adénocarcinome/anatomopathologie , Mâle , Composés hétérocycliques bicycliques
4.
J Extracell Vesicles ; 13(7): e12484, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-39041344

RÉSUMÉ

Pancreatic ductal adenocarcinoma (PDAC) is characterised by immune evasion that contribute to poor prognosis. Cancer-associated fibroblasts (CAFs) play a pivotal role in orchestrating the PDAC tumour microenvironment. We investigated the role of CAF-derived extracellular vesicle (EV)-packaged long non-coding RNAs (lncRNAs) in immune evasion and explored gene therapy using engineered EVs loading small interfering RNAs (siRNAs) as a potential therapeutic strategy. Our findings highlight the significance of EV-packaged lncRNA RP11-161H23.5 from CAF in promoting PDAC immune evasion by downregulating HLA-A expression, a key component of antigen presentation. Mechanistically, RP11-161H23.5 forms a complex with CNOT4, a subunit of the mRNA deadenylase CCR4-NOT complex, enhancing the degradation of HLA-A mRNA by shortening its poly(A) tail. This immune evasion mechanism compromises the anti-tumour immune response. To combat this, we propose an innovative approach utilising engineered EVs as natural and biocompatible nanocarriers for siRNA-based gene therapy and this strategy holds promise for enhancing the effectiveness of immunotherapy in PDAC. Overall, our study sheds light on the critical role of CAF-derived EV-packaged lncRNA RP11-161H23.5/CNOT4/HLA-A axis in PDAC immune evasion and presents a novel avenue for therapeutic intervention.


Sujet(s)
Fibroblastes associés au cancer , Carcinome du canal pancréatique , Vésicules extracellulaires , Antigènes HLA-A , Tumeurs du pancréas , ARN long non codant , Humains , ARN long non codant/génétique , ARN long non codant/métabolisme , Fibroblastes associés au cancer/métabolisme , Fibroblastes associés au cancer/immunologie , Vésicules extracellulaires/métabolisme , Vésicules extracellulaires/immunologie , Tumeurs du pancréas/immunologie , Tumeurs du pancréas/génétique , Tumeurs du pancréas/thérapie , Carcinome du canal pancréatique/immunologie , Carcinome du canal pancréatique/génétique , Carcinome du canal pancréatique/thérapie , Lignée cellulaire tumorale , Antigènes HLA-A/génétique , Antigènes HLA-A/immunologie , Antigènes HLA-A/métabolisme , Échappement immunitaire , Régulation de l'expression des gènes tumoraux , Régulation négative , Petit ARN interférent , Microenvironnement tumoral/immunologie , Animaux , Échappement de la tumeur à la surveillance immunitaire , Souris
5.
Front Immunol ; 15: 1434771, 2024.
Article de Anglais | MEDLINE | ID: mdl-39044834

RÉSUMÉ

The gut microbiome plays a significant role in the pathogenesis of pancreatic ductal adenocarcinoma (PDAC), influencing oncogenesis, immune responses, and treatment outcomes. Studies have identified microbial species like Porphyromonas gingivalis and Fusobacterium nucleatum, that promote PDAC progression through various mechanisms. Additionally, the gut microbiome affects immune cell activation and response to immunotherapy, including immune checkpoint inhibitors and CAR-T therapy. Specific microbes and their metabolites play a significant role in the effectiveness of immune checkpoint inhibitors (ICIs). Alterations in the gut microbiome can either enhance or diminish responses to PD-1/PD-L1 and CTLA-4 blockade therapy. Additionally, bacterial metabolites like trimethylamine N-oxide (TMAO) and lipopolysaccharide (LPS) impact antitumor immunity, offering potential targets to augment immunotherapy responses. Modulating the microbiome through fecal microbiota transplantation, probiotics, prebiotics, dietary changes, and antibiotics shows promise in PDAC treatment, although outcomes are highly variable. Dietary modifications, particularly high-fiber diets and specific fat consumption, influence microbiome composition and impact cancer risk. Combining microbiome-based therapies with existing treatments holds potential for improving PDAC therapy outcomes, but further research is needed to optimize their effectiveness.


Sujet(s)
Carcinome du canal pancréatique , Microbiome gastro-intestinal , Tumeurs du pancréas , Humains , Microbiome gastro-intestinal/immunologie , Carcinome du canal pancréatique/thérapie , Carcinome du canal pancréatique/microbiologie , Carcinome du canal pancréatique/immunologie , Tumeurs du pancréas/microbiologie , Tumeurs du pancréas/thérapie , Tumeurs du pancréas/immunologie , Animaux , Probiotiques/usage thérapeutique , Transplantation de microbiote fécal , Immunothérapie/méthodes , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique
6.
Cells ; 13(13)2024 Jul 04.
Article de Anglais | MEDLINE | ID: mdl-38994997

RÉSUMÉ

Extracellular HSP90α (eHSP90α) is a promoter of tumor development and malignant progression. Patients with malignancies, including pancreatic ductal adenocarcinoma (PDAC), have generally shown 5~10-fold increases in serum/plasma eHSP90α levels. In this study, we developed a humanized antibody HH01 to target eHSP90α and evaluated its anticancer efficacy. HH01, with novel complementarity-determining regions, exhibits high binding affinity toward HSP90α. It recognizes HSP90α epitope sites 235AEEKEDKEEE244 and 251ESEDKPEIED260, with critical amino acid residues E237, E239, D240, K241, E253, and K255. HH01 effectively suppressed eHSP90α-induced invasive and spheroid-forming activities of colorectal cancer and PDAC cell lines by blocking eHSP90α's ligation with the cell-surface receptor CD91. In mouse models, HH01 potently inhibited the tumor growth of PDAC cell grafts/xenografts promoted by endothelial-mesenchymal transition-derived cancer-associated fibroblasts while also reducing serum eHSP90α levels, reflecting its anticancer efficacy. HH01 also modulated tumor immunity by reducing M2 macrophages and reinvigorating immune T-cells. Additionally, HH01 showed low aggregation propensity, high water solubility, and a half-life time of >18 days in mouse blood. It was not cytotoxic to retinal pigmented epithelial cells and showed no obvious toxicity in mouse organs. Our data suggest that targeting eHSP90α with HH01 antibody can be a promising novel strategy for PDAC therapy.


Sujet(s)
Anticorps monoclonaux humanisés , Protéines du choc thermique HSP90 , Tumeurs du pancréas , Humains , Protéines du choc thermique HSP90/métabolisme , Protéines du choc thermique HSP90/antagonistes et inhibiteurs , Animaux , Tumeurs du pancréas/anatomopathologie , Tumeurs du pancréas/métabolisme , Souris , Lignée cellulaire tumorale , Anticorps monoclonaux humanisés/pharmacologie , Anticorps monoclonaux humanisés/usage thérapeutique , Prolifération cellulaire/effets des médicaments et des substances chimiques , Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques , Tests d'activité antitumorale sur modèle de xénogreffe , Adénocarcinome/anatomopathologie , Carcinome du canal pancréatique/anatomopathologie , Carcinome du canal pancréatique/immunologie , Carcinome du canal pancréatique/métabolisme ,
7.
Proc Natl Acad Sci U S A ; 121(29): e2403917121, 2024 Jul 16.
Article de Anglais | MEDLINE | ID: mdl-38980903

RÉSUMÉ

Pancreatic ductal adenocarcinoma (PDA) is a potentially lethal disease lacking effective treatments. Its immunosuppressive tumor microenvironment (TME) allows it to evade host immunosurveillance and limits response to immunotherapy. Here, using the mouse KRT19-deficient (sgKRT19-edited) PDA model, we find that intratumoral accumulation of natural killer T (NKT) cells is required to establish an immunologically active TME. Mechanistically, intratumoral NKT cells facilitate type I interferon (IFN) production to initiate an antitumor adaptive immune response, and orchestrate the intratumoral infiltration of T cells, dendritic cells, natural killer cells, and myeloid-derived suppressor cells. At the molecular level, NKT cells promote the production of type I IFN through the interaction of their CD40L with CD40 on myeloid cells. To evaluate the therapeutic potential of these observations, we find that administration of folinic acid to mice bearing PDA increases NKT cells in the TME and improves their response to anti-PD-1 antibody treatment. In conclusion, NKT cells have an essential role in the immune response to mouse PDA and are potential targets for immunotherapy.


Sujet(s)
Carcinome du canal pancréatique , Cellules T tueuses naturelles , Tumeurs du pancréas , Microenvironnement tumoral , Animaux , Souris , Tumeurs du pancréas/immunologie , Tumeurs du pancréas/anatomopathologie , Tumeurs du pancréas/thérapie , Cellules T tueuses naturelles/immunologie , Microenvironnement tumoral/immunologie , Carcinome du canal pancréatique/immunologie , Carcinome du canal pancréatique/thérapie , Carcinome du canal pancréatique/anatomopathologie , Interféron de type I/immunologie , Interféron de type I/métabolisme , Immunothérapie/méthodes , Souris de lignée C57BL , Leucovorine/administration et posologie , Leucovorine/usage thérapeutique , Humains , Cellules myéloïdes suppressives/immunologie
8.
Proc Natl Acad Sci U S A ; 121(31): e2403002121, 2024 Jul 30.
Article de Anglais | MEDLINE | ID: mdl-39047033

RÉSUMÉ

Pancreatic ductal adenocarcinoma (PDAC) represents a challenge in oncology, with limited treatment options for advanced-stage patients. Chimeric antigen receptor T cell (CAR T) therapy targeting mesothelin (MSLN) shows promise, but challenges such as the hostile immunosuppressive tumor microenvironment (TME) hinder its efficacy. This study explores the synergistic potential of combining proton radiation therapy (RT) with MSLN-targeting CAR T therapy in a syngeneic PDAC model. Proton RT significantly increased MSLN expression in tumor cells and caused a significant increase in CAR T cell infiltration into tumors. The combination therapy reshaped the immunosuppressive TME, promoting antitumorigenic M1 polarized macrophages and reducing myeloid-derived suppressor cells (MDSC). In a flank PDAC model, the combination therapy demonstrated superior attenuation of tumor growth and improved survival compared to individual treatments alone. In an orthotopic PDAC model treated with image-guided proton RT, tumor growth was significantly reduced in the combination group compared to the RT treatment alone. Further, the combination therapy induced an abscopal effect in a dual-flank tumor model, with increased serum interferon-γ levels and enhanced proliferation of extratumoral CAR T cells. In conclusion, combining proton RT with MSLN-targeting CAR T therapy proves effective in modulating the TME, enhancing CAR T cell trafficking, and exerting systemic antitumor effects. Thus, this combinatorial approach could present a promising strategy for improving outcomes in unresectable PDAC.


Sujet(s)
Carcinome du canal pancréatique , Protéines liées au GPI , Immunothérapie adoptive , Mésothéline , Tumeurs du pancréas , Récepteurs chimériques pour l'antigène , Microenvironnement tumoral , Animaux , Tumeurs du pancréas/thérapie , Tumeurs du pancréas/immunologie , Tumeurs du pancréas/radiothérapie , Tumeurs du pancréas/anatomopathologie , Souris , Protéines liées au GPI/métabolisme , Protéines liées au GPI/immunologie , Récepteurs chimériques pour l'antigène/immunologie , Récepteurs chimériques pour l'antigène/métabolisme , Immunothérapie adoptive/méthodes , Microenvironnement tumoral/immunologie , Humains , Carcinome du canal pancréatique/thérapie , Carcinome du canal pancréatique/immunologie , Carcinome du canal pancréatique/radiothérapie , Carcinome du canal pancréatique/anatomopathologie , Lignée cellulaire tumorale , Protonthérapie/méthodes , Association thérapeutique , Lymphocytes T/immunologie , Femelle
9.
Sci Data ; 11(1): 820, 2024 Jul 24.
Article de Anglais | MEDLINE | ID: mdl-39048591

RÉSUMÉ

Tumor organoids are three-dimensional in vitro models which can recapitulate the complex mutational landscape and tissue architecture observed in cancer patients, providing a realistic model for testing novel therapies, including immunotherapies. A significant challenge in organoid research in oncology lies in developing efficient and reliable methods for segmenting organoid images, quantifying organoid growth, regression and response to treatments, as well as predicting the behavior of organoid systems. Up to now, a curated dataset of organoids co-cultured with immune cells is not available. To address this gap, we present a new public dataset, comprising both phase-contrast images of murine and patient-derived tumor organoids of one of the deadliest cancer types, the Pancreatic Ductal Adenocarcinoma, co-cultured with immune cells, and state-of-the-art algorithms for object detection and segmentation. Our dataset, OrganoIDNetData, encompassing 180 images with 33906 organoids, can be a potential common benchmark for different organoids segmentation protocols, moving beyond the current practice of training and testing these algorithms on isolated datasets.


Sujet(s)
Algorithmes , Organoïdes , Tumeurs du pancréas , Tumeurs du pancréas/immunologie , Tumeurs du pancréas/anatomopathologie , Humains , Souris , Animaux , Carcinome du canal pancréatique/immunologie , Carcinome du canal pancréatique/anatomopathologie , Intelligence artificielle
10.
Sci Rep ; 14(1): 17071, 2024 Jul 24.
Article de Anglais | MEDLINE | ID: mdl-39048609

RÉSUMÉ

Pancreatic ductal adenocarcinoma (PDAC) has a high mortality rate; therefore, the development of effective treatments is a priority. The stimulator of interferon genes (STING) pathway enhances tumor immunity by inducing the production of type 1 interferon (IFN) and proinflammatory cytokines and chemokines and promoting the infiltration of cytotoxic T cells. To assess the function of STING on pancreatic tumorigenesis, Ptf1aER-Cre/+ LSL-KrasG12D/+ p53loxP/loxP mice (KPC mice) and Ptf1aER-Cre/+ LSL-KrasG12D/+ p53loxP/loxP/STING-/- mice (KPCS mice) were generated. However, STING deletion did not affect pancreatic tumorigenesis in mice. Because STING is expressed not only in immune cells but also in cancer-associated fibroblasts (CAFs), we evaluated the STING function in PDAC CAFs. A mouse STING agonist 5,6-Dimethyl-9-oxo-9H-xanthene-4-acetic acid (DMXAA) was administered to KPC mice and CAFs from KPC mice and the resulting immune response was evaluated. DMXAA activated STING in PDAC CAFs in KPC mice, promoting cytotoxic T cell infiltration by secreting proinflammatory cytokines and enhancing tumor immunity. We next generated STING-deficient PDAC cells and subcutaneous tumors in which STING was expressed only in CAFs by performing bone marrow transplantation and assessed the antitumor effect of STING-activated CAFs. The administration of DMXAA to subcutaneous tumors expressing STING only in CAFs sustained the antitumor effect of DMXAA. About half of human PDACs lacked STING expression in the cancer stroma, suggesting that STING activation in PDAC CAFs exerts an antitumor effect, and STING agonists can be more effective in tumors with high than in those with low STING expression in the stroma.


Sujet(s)
Fibroblastes associés au cancer , Carcinome du canal pancréatique , Protéines membranaires , Tumeurs du pancréas , Animaux , Protéines membranaires/métabolisme , Protéines membranaires/génétique , Tumeurs du pancréas/immunologie , Tumeurs du pancréas/anatomopathologie , Tumeurs du pancréas/métabolisme , Souris , Fibroblastes associés au cancer/métabolisme , Carcinome du canal pancréatique/immunologie , Carcinome du canal pancréatique/anatomopathologie , Carcinome du canal pancréatique/métabolisme , Carcinome du canal pancréatique/génétique , Carcinome du canal pancréatique/traitement médicamenteux , Humains , Xanthones/pharmacologie , Lignée cellulaire tumorale , Lymphocytes T cytotoxiques/immunologie
11.
Sci Adv ; 10(27): eadm9071, 2024 Jul 05.
Article de Anglais | MEDLINE | ID: mdl-38968363

RÉSUMÉ

Pancreatic ductal adenocarcinoma (PDAC) is the most common form of pancreatic cancer, a disease with dismal overall survival. Advances in treatment are hindered by a lack of preclinical models. Here, we show how a personalized organotypic "avatar" created from resected tissue allows spatial and temporal reporting on a complete in situ tumor microenvironment and mirrors clinical responses. Our perfusion culture method extends tumor slice viability, maintaining stable tumor content, metabolism, stromal composition, and immune cell populations for 12 days. Using multiplexed immunofluorescence and spatial transcriptomics, we identify immune neighborhoods and potential for immunotherapy. We used avatars to assess the impact of a preclinically validated metabolic therapy and show recovery of stromal and immune phenotypes and tumor redifferentiation. To determine clinical relevance, we monitored avatar response to gemcitabine treatment and identify a patient avatar-predictable response from clinical follow-up. Thus, avatars provide valuable information for syngeneic testing of therapeutics and a truly personalized therapeutic assessment platform for patients.


Sujet(s)
Carcinome du canal pancréatique , Tumeurs du pancréas , Médecine de précision , Microenvironnement tumoral , Humains , Médecine de précision/méthodes , Tumeurs du pancréas/immunologie , Tumeurs du pancréas/anatomopathologie , Tumeurs du pancréas/traitement médicamenteux , Tumeurs du pancréas/thérapie , Microenvironnement tumoral/immunologie , Carcinome du canal pancréatique/immunologie , Carcinome du canal pancréatique/thérapie , Carcinome du canal pancréatique/anatomopathologie , , Désoxycytidine/analogues et dérivés , Désoxycytidine/usage thérapeutique , Désoxycytidine/pharmacologie , Lignée cellulaire tumorale , Immunothérapie/méthodes , Animaux ,
12.
World J Gastroenterol ; 30(23): 2927-2930, 2024 Jun 21.
Article de Anglais | MEDLINE | ID: mdl-38946872

RÉSUMÉ

In this editorial, we focus specifically on the mechanisms by which pancreatic inflammation affects pancreatic cancer. Cancer of the pancreas remains one of the deadliest cancer types. The highest incidence and mortality rates of pancreatic cancer are found in developed countries. Trends of pancreatic cancer incidence and mortality vary considerably worldwide. A better understanding of the etiology and identification of the risk factors is essential for the primary prevention of this disease. Pancreatic tumors are characterized by a complex microenvironment that orchestrates metabolic alterations and supports a milieu of interactions among various cell types within this niche. In this editorial, we highlight the foundational studies that have driven our understanding of these processes. In our experimental center, we have carefully studied the mechanisms of that link pancreatic inflammation and pancreatic cancer. We focused on the role of mast cells (MCs). MCs contain pro-angiogenic factors, including tryptase, that are associated with increased angiogenesis in various tumors. In this editorial, we address the role of MCs in angiogenesis in both pancreatic ductal adenocarcinoma tissue and adjacent normal tissue. The assessment includes the density of c-Kit receptor-positive MCs, the density of tryptase-positive MCs, the area of tryptase-positive MCs, and angiogenesis in terms of microvascularization density.


Sujet(s)
Mastocytes , Néovascularisation pathologique , Tumeurs du pancréas , Microenvironnement tumoral , Humains , Tumeurs du pancréas/anatomopathologie , Tumeurs du pancréas/métabolisme , Tumeurs du pancréas/immunologie , Mastocytes/métabolisme , Mastocytes/immunologie , Microenvironnement tumoral/immunologie , Carcinome du canal pancréatique/anatomopathologie , Carcinome du canal pancréatique/immunologie , Carcinome du canal pancréatique/métabolisme , Pancréas/anatomopathologie , Pancréas/immunologie , Pancréas/métabolisme , Animaux , Pancréatite/métabolisme , Pancréatite/anatomopathologie , Pancréatite/immunologie , Facteurs de risque , Médiateurs de l'inflammation/métabolisme , Tryptases/métabolisme , Inflammation/métabolisme
13.
BMC Cancer ; 24(1): 809, 2024 Jul 07.
Article de Anglais | MEDLINE | ID: mdl-38973003

RÉSUMÉ

BACKGROUND: Pancreatic Ductal Adenocarcinoma (PDAC) is an aggressive cancer characterized by an immunosuppressive microenvironment. Patients from specific ethnicities and population groups have poorer prognoses than others. Therefore, a better understanding of the immune landscape in such groups is necessary for disease elucidation, predicting patient outcomes and therapeutic targeting. This study investigated the expression of circulating key immune cell markers in South African PDAC patients of African ancestry. METHODS: Blood samples were obtained from a total of 6 healthy volunteers (HC), 6 Chronic Pancreatitis (CP) and 34 PDAC patients consisting of 22 resectable (RPC), 8 locally advanced (LAPC) and 4 metastatic (MPC). Real-time Quantitative Polymerase Chain reactions (RT-qPCR), Metabolomics, Enzyme-Linked Immunosorbent Assay (ELISA), Reactive Oxygen Species (ROS), and Immunophenotyping assays were conducted. Statistical analysis was conducted in R (v 4.3.2). Additional analysis of single-cell RNA data from 20 patients (16 PDAC and 4 controls) was conducted to interrogate the distribution of T-cell and Natural Killer cell populations. RESULTS: Granulocyte and neutrophil levels were significantly elevated while lymphocytes decreased with PDAC severity. The total percentages of CD3 T-cell subpopulations (helper and double negative T-cells) decreased when compared to HC. Although both NK (p = 0.014) and NKT (p < 0.001) cell levels increased as the disease progressed, their subsets: NK CD56dimCD16- (p = 0.024) and NKTs CD56+ (p = 0.008) cell levels reduced significantly. Of note is the negative association of NK CD56dimCD16- (p < 0.001) cell levels with survival time. The gene expression analyses showed no statistically significant correlation when comparing the PDAC groups with the controls. The inflammatory status of PDAC was assessed by ROS levels of serum which were elevated in CP (p = 0.025), (RPC (p = 0.003) and LAPC (p = 0.008)) while no significant change was observed in MPC, compared to the HC group. ROS was shown to be positively correlated with GlycA (R = 0.45, p = 0.0096). Single-cell analyses showed a significant difference in the ratio of NKT cells per total cell counts in LAPC (p < 0.001) and MPC (p < 0.001) groups compared with HC, confirming observations in our sample group. CONCLUSION: The expression of these immune cell markers observed in this pilot study provides insight into their potential roles in tumour progression in the patient group and suggests their potential utility in the development of immunotherapeutic strategies.


Sujet(s)
Carcinome du canal pancréatique , Évolution de la maladie , Tumeurs du pancréas , Humains , Carcinome du canal pancréatique/immunologie , Carcinome du canal pancréatique/anatomopathologie , Carcinome du canal pancréatique/génétique , Mâle , Femelle , Adulte d'âge moyen , Tumeurs du pancréas/immunologie , Tumeurs du pancréas/anatomopathologie , Tumeurs du pancréas/génétique , République d'Afrique du Sud , Sujet âgé , Adulte , Marqueurs biologiques tumoraux/génétique , Cellules tueuses naturelles/immunologie , Cellules tueuses naturelles/métabolisme , Pancréatite chronique/immunologie , Pancréatite chronique/génétique , Pancréatite chronique/anatomopathologie , Espèces réactives de l'oxygène/métabolisme , Immunophénotypage
14.
Zhongguo Yi Xue Ke Xue Yuan Xue Bao ; 46(3): 354-360, 2024 Jun.
Article de Chinois | MEDLINE | ID: mdl-38953259

RÉSUMÉ

Objective To construct a risk prediction model by integrating the molecular subtypes of pancreatic ductal adenocarcinoma (PDAC) and immune-related genes.Methods With GSE71729 data set (n=145) as the training set,the differentially expressed genes and differential immune-related genes between the squamous and non-squamous subtypes of PDAC were integrated to construct a regulatory network,on the basis of which five immune marker genes regulating the squamous subtype were screened out.An integrated immune score (IIS) model was constructed based on patient survival information and immune marker genes to predict the clinical prognosis of PDAC patients,and its predictive performance was tested with 5 validation sets (n=758).Results PDAC patients were assigned into high risk and low risk groups according to the IIS.In both training and validation sets,the overall survival of patients in the high risk group was shorter than that in the low risk group (both P<0.001).The multivariable Cox regression showed that IIS was an independent prognostic factor for PDAC (HR=2.16,95%CI=1.50-3.10,P<0.001).Conclusion IIS can be used for risk stratification of PDAC patients and may become a potential prognostic marker for PDAC.


Sujet(s)
Carcinome du canal pancréatique , Tumeurs du pancréas , Humains , Carcinome du canal pancréatique/génétique , Carcinome du canal pancréatique/immunologie , Carcinome du canal pancréatique/mortalité , Pronostic , Tumeurs du pancréas/génétique , Tumeurs du pancréas/immunologie , Tumeurs du pancréas/mortalité , Femelle , Mâle , Adulte d'âge moyen , Marqueurs biologiques tumoraux/génétique , Appréciation des risques/méthodes
15.
Sci Rep ; 14(1): 15037, 2024 07 01.
Article de Anglais | MEDLINE | ID: mdl-38951569

RÉSUMÉ

The NK cell is an important component of the tumor microenvironment of pancreatic ductal adenocarcinoma (PDAC), also plays a significant role in PDAC development. This study aimed to explore the relationship between NK cell marker genes and prognosis, immune response of PDAC patients. By scRNA-seq data, we found the proportion of NK cells were significantly downregulated in PDAC and 373 NK cell marker genes were screened out. By TCGA database, we enrolled 7 NK cell marker genes to construct the signature for predicting prognosis in PDAC patients. Cox analysis identified the signature as an independent factor for pancreatic cancer. Subsequently, the predictive power of signature was validated by 6 GEO datasets and had an excellent evaluation. Our analysis of relationship between the signature and patients' immune status revealed that the signature has a strong correlation with immunocyte infiltration, inflammatory reaction, immune checkpoint inhibitors (ICIs) response. The NK cell marker genes are closely related to the prognosis and immune capacity of PDAC patients, and they have potential value as a therapeutic target.


Sujet(s)
Marqueurs biologiques tumoraux , Carcinome du canal pancréatique , Cellules tueuses naturelles , Tumeurs du pancréas , Analyse sur cellule unique , Humains , Carcinome du canal pancréatique/génétique , Carcinome du canal pancréatique/immunologie , Carcinome du canal pancréatique/anatomopathologie , Carcinome du canal pancréatique/mortalité , Cellules tueuses naturelles/immunologie , Pronostic , Tumeurs du pancréas/génétique , Tumeurs du pancréas/immunologie , Tumeurs du pancréas/mortalité , Tumeurs du pancréas/anatomopathologie , Marqueurs biologiques tumoraux/génétique , Analyse sur cellule unique/méthodes , Femelle , Mâle , Régulation de l'expression des gènes tumoraux , Analyse de séquence d'ARN , Microenvironnement tumoral/immunologie , Microenvironnement tumoral/génétique , Adulte d'âge moyen , Sujet âgé , Analyse de profil d'expression de gènes
16.
J Cancer Res Clin Oncol ; 150(6): 300, 2024 Jun 08.
Article de Anglais | MEDLINE | ID: mdl-38850373

RÉSUMÉ

PURPOSE: Tertiary lymphoid structures (TLSs) and CD8 + T-cells are potential prognostic indicators for pancreatic ductal adenocarcinoma (PDAC). We established a novel scoring system for evaluating the risk for PDAC based on TLS- and CD8 + T-cell-related genes. METHODS: We analyzed single-cell sequence data from PDAC patients in the Genome Sequence Archive. Bioinformatics and machine algorithms established and validated a scoring method (T-C score) based on PDAC survival-related genes highly expressed in TLSs and CD8 + T-cells. Patients were stratified into the low- and high-T-C score groups. Differences in survival, pathway enrichment, mutation status, immune cell infiltration, expression of immune checkpoint-associated genes, tumor stemness, and response to antitumor therapy were compared through computer simulation methods. RESULTS: Overall survival differed significantly between the training and validation cohorts' low- and high-T-C score groups. The low-T-C score group correlated with lower tumor mutation burden and lower levels of tumor stemness compared with the high-T-C score group. Patients with lower T-C scores exhibited advantages in immunotherapeutic responses and might be more sensitive to the chemotherapeutic regimen and multi-kinase inhibitors. CONCLUSION: The T-C score could serve as an effective model for predicting the survival and therapeutic responses of patients with PDAC.


Sujet(s)
Lymphocytes T CD8+ , Carcinome du canal pancréatique , Tumeurs du pancréas , Structures lymphoïdes tertiaires , Humains , Carcinome du canal pancréatique/génétique , Carcinome du canal pancréatique/immunologie , Carcinome du canal pancréatique/anatomopathologie , Pronostic , Tumeurs du pancréas/génétique , Tumeurs du pancréas/immunologie , Tumeurs du pancréas/anatomopathologie , Lymphocytes T CD8+/immunologie , Structures lymphoïdes tertiaires/génétique , Structures lymphoïdes tertiaires/immunologie , Structures lymphoïdes tertiaires/anatomopathologie , Génomique/méthodes , Mâle , Femelle , Marqueurs biologiques tumoraux/génétique , Adulte d'âge moyen , Sujet âgé
17.
JCI Insight ; 9(12)2024 May 23.
Article de Anglais | MEDLINE | ID: mdl-38912584

RÉSUMÉ

The regulated glycosylation of the proteome has widespread effects on biological processes that cancer cells can exploit. Expression of N-acetylglucosaminyltransferase V (encoded by Mgat5 or GnT-V), which catalyzes the addition of ß1,6-linked N-acetylglucosamine to form complex N-glycans, has been linked to tumor growth and metastasis across tumor types. Using a panel of murine pancreatic ductal adenocarcinoma (PDAC) clonal cell lines that recapitulate the immune heterogeneity of PDAC, we found that Mgat5 is required for tumor growth in vivo but not in vitro. Loss of Mgat5 results in tumor clearance that is dependent on T cells and dendritic cells, with NK cells playing an early role. Analysis of extrinsic cell death pathways revealed Mgat5-deficient cells have increased sensitivity to cell death mediated by the TNF superfamily, a property that was shared with other non-PDAC Mgat5-deficient cell lines. Finally, Mgat5 knockout in an immunotherapy-resistant PDAC line significantly decreased tumor growth and increased survival upon immune checkpoint blockade. These findings demonstrate a role for N-glycosylation in regulating the sensitivity of cancer cells to T cell killing through classical cell death pathways.


Sujet(s)
Carcinome du canal pancréatique , N-acetylglucosaminyltransferase , Tumeurs du pancréas , Animaux , Humains , Souris , Carcinome du canal pancréatique/immunologie , Carcinome du canal pancréatique/anatomopathologie , Carcinome du canal pancréatique/métabolisme , Carcinome du canal pancréatique/génétique , Lignée cellulaire tumorale , Cellules dendritiques/immunologie , Cellules dendritiques/métabolisme , Glycosylation , Cellules tueuses naturelles/immunologie , Cellules tueuses naturelles/métabolisme , Souris knockout , N-acetylglucosaminyltransferase/métabolisme , N-acetylglucosaminyltransferase/génétique , Tumeurs du pancréas/immunologie , Tumeurs du pancréas/anatomopathologie , Tumeurs du pancréas/métabolisme , Tumeurs du pancréas/génétique , Lymphocytes T/immunologie , Lymphocytes T/métabolisme
18.
Immunobiology ; 229(4): 152822, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38852289

RÉSUMÉ

Pancreatic ductal adenocarcinoma (PDAC) is a highly lethal malignancy that represents a significant challenge in cancer research and clinical management. In this study, we reanalyzed a published single-cell RNA sequencing (scRNA-seq) dataset from PDAC and adjacent tissues to investigate the heterogeneity of tumor and normal tissue, specifically focusing on the regulatory T cells (Tregs) and their interactions with other cells in the tumor microenvironment (TME). Treg cells were identified and clustered into natural Tregs (nTreg) and induced Tregs (iTreg) based on the expression of specific genes. It was found that the number of iTregs was higher in the tumor than in healthy tissues, while the number of n Tregs was higher in healthy tissues. Differential gene expression analysis was performed, and biological process analysis revealed that the Tregs in PDAC were mostly involved in protein targeting and translation pathways. In addition, ligand-receptor pairs between Tregs and other cell types were identified, and the critical communication pathways between Tregs and endothelial and ductal cells were revealed, which could potentially contribute to the immunosuppressive TME of PDAC. These findings provide insights into the role of Tregs in PDAC and their interactions with other cell types in the TME, highlighting potential targets for immunotherapy, such as the inhibitory immune checkpoint receptors CTLA4 and TIGIT, which are known to be expressed on Tregs and have been shown to play a role in suppressing anti-tumor immune responses.


Sujet(s)
Antigène CTLA-4 , Carcinome du canal pancréatique , Régulation de l'expression des gènes tumoraux , Tumeurs du pancréas , Récepteurs immunologiques , Analyse sur cellule unique , Lymphocytes T régulateurs , Microenvironnement tumoral , Humains , Carcinome du canal pancréatique/immunologie , Carcinome du canal pancréatique/génétique , Lymphocytes T régulateurs/immunologie , Antigène CTLA-4/génétique , Récepteurs immunologiques/génétique , Récepteurs immunologiques/métabolisme , Analyse sur cellule unique/méthodes , Tumeurs du pancréas/immunologie , Tumeurs du pancréas/génétique , Microenvironnement tumoral/immunologie , Microenvironnement tumoral/génétique , Analyse de séquence d'ARN , Analyse de profil d'expression de gènes , Transcriptome
19.
Science ; 384(6703): eadh4567, 2024 Jun 28.
Article de Anglais | MEDLINE | ID: mdl-38935717

RÉSUMÉ

Inflammation and tissue damage associated with pancreatitis can precede or occur concurrently with pancreatic ductal adenocarcinoma (PDAC). We demonstrate that in PDAC coupled with pancreatitis (ptPDAC), antigen-presenting type I conventional dendritic cells (cDC1s) are specifically activated. Immune checkpoint blockade therapy (iCBT) leads to cytotoxic CD8+ T cell activation and elimination of ptPDAC with restoration of life span even upon PDAC rechallenge. Using PDAC antigen-loaded cDC1s as a vaccine, immunotherapy-resistant PDAC was rendered sensitive to iCBT with elimination of tumors. cDC1 vaccination coupled with iCBT identified specific CDR3 sequences in the tumor-infiltrating CD8+ T cells with potential therapeutic importance. This study identifies a fundamental difference in the immune microenvironment in PDAC concurrent with, or without, pancreatitis and provides a rationale for combining cDC1 vaccination with iCBT as a potential treatment option.


Sujet(s)
Carcinome du canal pancréatique , Cellules dendritiques , Immunothérapie , Tumeurs du pancréas , Microenvironnement tumoral , Animaux , Souris , Vaccins anticancéreux/immunologie , Vaccins anticancéreux/usage thérapeutique , Carcinome du canal pancréatique/thérapie , Carcinome du canal pancréatique/immunologie , Lymphocytes T CD8+/immunologie , Cellules dendritiques/immunologie , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Immunothérapie/méthodes , Souris de lignée C57BL , Tumeurs du pancréas/thérapie , Tumeurs du pancréas/immunologie , Pancréatite/immunologie , Pancréatite/thérapie , Microenvironnement tumoral/immunologie
20.
Cancer Immunol Immunother ; 73(8): 149, 2024 Jun 04.
Article de Anglais | MEDLINE | ID: mdl-38833018

RÉSUMÉ

Despite the successful application of programmed cell death ligand 1 (PD-L1)-blocking strategies in some types of cancers and well-established prognostic indicators in pancreatic ductal adenocarcinoma (PDAC), the biological and clinical implications of the methylation status of PD-L1/PD-L2 in PDAC remain largely unknown. Therefore, this study aimed to explore the biological role of PD-L1/PD-L2 methylation and its association with clinicopathological features, clinical outcomes, and the immune microenvironment by analyzing the data on PD-L1/PD-L2 methylation and mRNA expression in PDAC cohorts obtained from the Cancer Genome Atlas and International Cancer Genome Consortium. The correlation between PD-L1 promoter methylation and PD-L1 expression and survival was further validated in an independent validation cohort (Peking Union Medical College Hospital [PUMCH] cohort) using pyrosequencing and immunohistochemistry. These results demonstrated that hypomethylation of the PD-L1 promoter was strongly associated with upregulated PD-L1 expression and shorter overall survival in PDAC. Multivariate Cox regression analyses revealed that the PD-L1 promoter methylation was an independent prognostic factor. PD-L1 promoter hypomethylation and high expression were related to aggressive clinical phenotypes. Moreover, both PD-L1 and PD-L2 methylation correlated with immune cell infiltration and the expression of immune checkpoint genes. PD-L1 promoter methylation status was further validated as an independent prognostic biomarker in patients with PDAC using the PUMCH cohort. The prognostic significance of PD-L1 promoter methylation was more discriminative in tumors with perineural/lymphovascular invasion and distant metastasis than in those without perineural/lymphovascular invasion and distant metastasis. In summary, the methylation status of the PD-L1 promoter is a promising biomarker for survival outcomes, immune infiltration, and the potential immune benefits of immunotherapy in PDAC.


Sujet(s)
Antigène CD274 , Carcinome du canal pancréatique , Méthylation de l'ADN , Tumeurs du pancréas , Régions promotrices (génétique) , Humains , Antigène CD274/génétique , Antigène CD274/métabolisme , Carcinome du canal pancréatique/génétique , Carcinome du canal pancréatique/immunologie , Carcinome du canal pancréatique/mortalité , Carcinome du canal pancréatique/anatomopathologie , Régions promotrices (génétique)/génétique , Tumeurs du pancréas/génétique , Tumeurs du pancréas/immunologie , Tumeurs du pancréas/mortalité , Tumeurs du pancréas/anatomopathologie , Femelle , Mâle , Pronostic , Adulte d'âge moyen , Marqueurs biologiques tumoraux/génétique , Microenvironnement tumoral/immunologie , Microenvironnement tumoral/génétique , Sujet âgé , Régulation de l'expression des gènes tumoraux
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE