1
|
Cannella R, Dioguardi Burgio M, Maino C, Matteini F, Ippolito D, Boraschi P, Zamboni GA, Vernuccio F. Conditions at risk of pancreatic cancer: The radiology perspective. Eur J Radiol 2025; 188:112119. [PMID: 40273500 DOI: 10.1016/j.ejrad.2025.112119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 03/28/2025] [Accepted: 04/15/2025] [Indexed: 04/26/2025]
Abstract
Pancreatic cancer remains one for the most aggressive cancer worldwide, with pancreatic ductal adenocarcinoma being the most common malignant pancreatic lesion, associated with poor prognosis. While surgical resection is the only curative treatment, only a minority of patients is eligible for surgery due to its diagnosis at advanced stages. Therefore, strategies for early detection of pancreatic cancer are needed. This article aims to provide a state-of-the-art review of the most common conditions associated to an increased risk of pancreatic cancer. Conditions linked to risk of pancreatic cancer development include certain pancreato-biliary anatomical variants, intraductal papillary mucinous neoplasms, mucinous cystic neoplasm, and familial pancreatic cancer with specific genetic mutations. Early imaging signs of pancreatic cancer can also be incidentally encountered on CT or MRI performed for other indications and they should be promptly recognized by the radiologists in order to avoid delays in the diagnosis. The features include focal pancreatic atrophy, contour deformity, dilation of the main pancreatic duct (MPD), changes in the caliber of the MPD, abrupt interruption of the MPD, and biliary tree dilation. MRI with the adoption of abbreviated protocols has been increasingly evaluated for the follow-up of cystic lesions. Although screening of the general population is not recommended due to the low incidence and high costs, surveillance with MRI can be considered in selected high-risk individuals.
Collapse
Affiliation(s)
- Roberto Cannella
- Section of Radiology - Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), University of Palermo, Via del Vespro 129, Palermo 90127, Italy.
| | - Marco Dioguardi Burgio
- Université Paris Cité, INSERM, Centre de recherche sur l'inflammation, F-75018 Paris, France; Radiology Department, AP-HP, Hôpital Beaujon, 92110 Clichy, France
| | - Cesare Maino
- Department of Diagnostic Radiology, Fondazione IRCCS San Gerardo dei Tintori, Via Pergolesi 33, 20900 Monza, MB, Italy
| | - Francesco Matteini
- Section of Radiology - Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), University of Palermo, Via del Vespro 129, Palermo 90127, Italy
| | - Davide Ippolito
- Department of Diagnostic Radiology, Fondazione IRCCS San Gerardo dei Tintori, Via Pergolesi 33, 20900 Monza, MB, Italy
| | - Piero Boraschi
- 2nd Unit of Radiology, Department of Radiological Nuclear and Laboratory Medicine - Pisa University Hospital, Via Paradisa 2, 56124 Pisa, Italy
| | - Giulia A Zamboni
- Department of Diagnostics and Public Health, Institute of Radiology, University of Verona, Policlinico GB Rossi, P.Le LA Scuro 10, 37134 Verona, Italy
| | - Federica Vernuccio
- Section of Radiology - Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), University of Palermo, Via del Vespro 129, Palermo 90127, Italy
| |
Collapse
|
2
|
Earl J, Kataki A, Canzian F, Costello E, Campa D, Greenhalf W. Targeting pancreatic cancer screening by identification of pathogenic variants of BRCA2/ BRCA1 in healthy individuals who have no known family history of pancreatic cancer: The arguments for and against. Semin Cancer Biol 2025; 113:1-8. [PMID: 40339999 DOI: 10.1016/j.semcancer.2025.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 04/15/2025] [Accepted: 05/04/2025] [Indexed: 05/10/2025]
Abstract
The majority of patients with pancreatic ductal adenocarcinoma (PDAC) are no longer suitable for treatment with curable intent at the time of diagnosis resulting in a 5-year survival of less than 10 %. Imaging of asymptomatic individuals could identify early cancers, but only with a risk of falsely identifying a benign lesion as malignant. Screening of an unselected population would result in far more such false positives than true early cancers. Selection before screening is therefore essential, but there are very few populations at high enough risk to make screening more beneficial than counterproductive. These populations include carriers of specific mutations in BRCA2, and arguably BRCA1, who have a family history of PDAC. These pathogenic mutations all have a predictable effect in making loss of Homologous Recombination Repair (HRR) likely in a carrier's lifetime. In this review the impact of such loss of HRR function on the likelihood of PDAC development will be discussed. Furthermore, it will be discussed whether the identification of a germline pathogenic mutation is sufficient to justify carrier surveillance for the development of the malignancy, or whether the current practice of screening only those carriers with a close relative diagnosed with PDAC is justifiable, as only a proportion of carriers are at high risk. The review will go beyond this to discuss whether there is an essential need to better define and stratify those at high risk, so that only high-risk carriers are put on surveillance.
Collapse
Affiliation(s)
- Julie Earl
- Biomarkers and Personalized Approach to Cancer (BIOPAC) Group, Ramón y Cajal Health Research Institute (IRYCIS), CIBERONC, Carretera Colmenar Km 9,100, Madrid 28034, Spain
| | - Agapi Kataki
- Department of Propaeudeutic Surgery, Breast Unit, Medical School, National and Kapodistrian University of Athens, Greece
| | - Federico Canzian
- Genomic Epidemiology Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Eithne Costello
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, UK
| | | | - William Greenhalf
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, UK.
| |
Collapse
|
3
|
Guo K, Li S, Wu X, Xiong H. Nanomedicine in the Diagnosis and Treatment of Pancreatic Cancer. Pharmaceutics 2025; 17:449. [PMID: 40284444 PMCID: PMC12030228 DOI: 10.3390/pharmaceutics17040449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2025] [Revised: 03/25/2025] [Accepted: 03/28/2025] [Indexed: 04/29/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an aggressive malignancy with increasing incidence and mortality rates, highlighting the urgent need for early diagnosis and treatment. However, early diagnosis of PDAC is extremely challenging due to the atypical early symptoms or the absence of noticeable symptoms. As a result, many patients are diagnosed with local metastasis, and even patients who are eligible for surgical resection have a high postoperative recurrence rate. Consequently, chemotherapy remains the primary treatment for PDAC. However, the unique biological characteristics of PDAC not only promote tumor progression and metastasis but also often lead to chemoresistance, a significant barrier to successful treatment. Recently, nanomaterials have garnered significant attention as promising materials for diagnosing and treating PDAC, showing great potential in cancer therapy, imaging, and drug delivery. Novel targeted nanomedicines, which encapsulate chemotherapy drugs and gene therapy products, offer significant advantages in overcoming resistance. These nanomedicines not only provide innovative solutions to the limitations of conventional chemotherapy but also improve the selectivity for cancer cells to enhance therapeutic outcomes. Current research is focused on the development of advanced nanomedicines, such as liposomes, nanotubes, and polymer-lipid hybrid systems, aimed at making chemotherapy more effective and longer lasting. This review provides a detailed overview of various nanomedicines utilized in the diagnosis and treatment of PDAC and outlines future directions for their development and key breakthroughs.
Collapse
Affiliation(s)
| | | | - Xinyu Wu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (K.G.); (S.L.)
| | - Huihua Xiong
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (K.G.); (S.L.)
| |
Collapse
|
4
|
Sarac D, Badza Atanasijevic M, Mitrovic Jovanovic M, Kovac J, Lazic L, Jankovic A, Saponjski DJ, Milosevic S, Stosic K, Masulovic D, Radenkovic D, Papic V, Djuric-Stefanovic A. Applicability of Radiomics for Differentiation of Pancreatic Adenocarcinoma from Healthy Tissue of Pancreas by Using Magnetic Resonance Imaging and Machine Learning. Cancers (Basel) 2025; 17:1119. [PMID: 40227615 PMCID: PMC11987955 DOI: 10.3390/cancers17071119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 03/12/2025] [Accepted: 03/20/2025] [Indexed: 04/15/2025] Open
Abstract
BACKGROUND This study analyzed different classifier models for differentiating pancreatic adenocarcinoma from surrounding healthy pancreatic tissue based on radiomic analysis of magnetic resonance (MR) images. METHODS We observed T2W-FS and ADC images obtained by 1.5T-MR of 87 patients with histologically proven pancreatic adenocarcinoma for training and validation purposes and then tested the most accurate predictive models that were obtained on another group of 58 patients. The tumor and surrounding pancreatic tissue were segmented on three consecutive slices, with the largest area of interest (ROI) of tumor marked using MaZda v4.6 software. This resulted in a total of 261 ROIs for each of the observed tissue classes in the training-validation group and 174 ROIs in the testing group. The software extracted a total of 304 radiomic features for each ROI, divided into six categories. The analysis was conducted through six different classifier models with six different feature reduction methods and five-fold subject-wise cross-validation. RESULTS In-depth analysis shows that the best results were obtained with the Random Forest (RF) classifier with feature reduction based on the Mutual Information score (all nine features are from the co-occurrence matrix): an accuracy of 0.94/0.98, sensitivity of 0.94/0.98, specificity of 0.94/0.98, and F1-score of 0.94/0.98 were achieved for the T2W-FS/ADC images from the validation group, retrospectively. In the testing group, an accuracy of 0.69/0.81, sensitivity of 0.86/0.82, specificity of 0.52/0.70, and F1-score of 0.74/0.83 were achieved for the T2W-FS/ADC images, retrospectively. CONCLUSIONS The machine learning approach using radiomics features extracted from T2W-FS and ADC achieved a relatively high sensitivity in the differentiation of pancreatic adenocarcinoma from healthy pancreatic tissue, which could be especially applicable for screening purposes.
Collapse
Affiliation(s)
- Dimitrije Sarac
- Center for Radiology, University Clinical Centre of Serbia, Pasterova No. 2, 11000 Belgrade, Serbia
| | - Milica Badza Atanasijevic
- School of Electrical Engineering, University of Belgrade, Bulevar kralja Aleksandra 73, 11120 Belgrade, Serbia; (M.B.A.)
- Innovation Center of the School of Electrical Engineering in Belgrade, Bulevar kralja Aleksandra 73, 11120 Belgrade, Serbia
| | - Milica Mitrovic Jovanovic
- Center for Radiology, University Clinical Centre of Serbia, Pasterova No. 2, 11000 Belgrade, Serbia
- Department for Radiology, Faculty of Medicine, University of Belgrade, Dr Subotica No. 8, 11000 Belgrade, Serbia
| | - Jelena Kovac
- Center for Radiology, University Clinical Centre of Serbia, Pasterova No. 2, 11000 Belgrade, Serbia
- Department for Radiology, Faculty of Medicine, University of Belgrade, Dr Subotica No. 8, 11000 Belgrade, Serbia
| | - Ljubica Lazic
- Center for Radiology, University Clinical Centre of Serbia, Pasterova No. 2, 11000 Belgrade, Serbia
| | - Aleksandra Jankovic
- Center for Radiology, University Clinical Centre of Serbia, Pasterova No. 2, 11000 Belgrade, Serbia
- Department for Radiology, Faculty of Medicine, University of Belgrade, Dr Subotica No. 8, 11000 Belgrade, Serbia
| | - Dusan J. Saponjski
- Center for Radiology, University Clinical Centre of Serbia, Pasterova No. 2, 11000 Belgrade, Serbia
- Department for Radiology, Faculty of Medicine, University of Belgrade, Dr Subotica No. 8, 11000 Belgrade, Serbia
| | - Stefan Milosevic
- Center for Radiology, University Clinical Centre of Serbia, Pasterova No. 2, 11000 Belgrade, Serbia
| | - Katarina Stosic
- Center for Radiology, University Clinical Centre of Serbia, Pasterova No. 2, 11000 Belgrade, Serbia
| | - Dragan Masulovic
- Center for Radiology, University Clinical Centre of Serbia, Pasterova No. 2, 11000 Belgrade, Serbia
- Department for Radiology, Faculty of Medicine, University of Belgrade, Dr Subotica No. 8, 11000 Belgrade, Serbia
| | - Dejan Radenkovic
- Department for HBP Surgery, Clinic for Digestive Surgery, University Clinical Centre of Serbia, Koste Todorovica Street, No. 6, 11000 Belgrade, Serbia
- Department for Surgery, Faculty of Medicine, University of Belgrade, Dr Subotica No. 8, 11000 Belgrade, Serbia
| | - Veljko Papic
- School of Electrical Engineering, University of Belgrade, Bulevar kralja Aleksandra 73, 11120 Belgrade, Serbia; (M.B.A.)
| | - Aleksandra Djuric-Stefanovic
- Center for Radiology, University Clinical Centre of Serbia, Pasterova No. 2, 11000 Belgrade, Serbia
- Department for Radiology, Faculty of Medicine, University of Belgrade, Dr Subotica No. 8, 11000 Belgrade, Serbia
| |
Collapse
|
5
|
Salas-Escabillas DJ, Hoffman MT, Brender SM, Moore JS, Wen HJ, Benitz S, Davis ET, Long D, Wombwell AM, Chianis ERD, Allen-Petersen BL, Steele NG, Sears RC, Matsumoto I, DelGiorno KE, Crawford HC. Tuft cells transdifferentiate to neural-like progenitor cells in the progression of pancreatic cancer. Dev Cell 2025; 60:837-852.e3. [PMID: 39721583 DOI: 10.1016/j.devcel.2024.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 09/13/2024] [Accepted: 12/02/2024] [Indexed: 12/28/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDA) is partly initiated through the transdifferentiation of acinar cells to metaplasia, which progresses to neoplasia and cancer. Tuft cells (TCs) are chemosensory cells not found in the normal pancreas but arise in cancer precursor lesions and diminish during progression to carcinoma. These metaplastic TCs (mTCs) suppress tumor progression through communication with the tumor microenvironment, but their fate during progression is unknown. To determine the fate of mTCs during PDA progression, we created a dual recombinase lineage trace model, wherein a pancreas-specific FlpO was used to induce tumorigenesis, while a tuft-cell specific Pou2f3CreERT/+ driver was used to induce expression of a tdTomato reporter. We found that mTCs in carcinoma transdifferentiate into neural-like progenitor cells (NRPs), a cell type associated with poor survival in patients. Using conditional knockout and overexpression systems, we found that Myc activity in mTCs is necessary and sufficient to induce this tuft-to-neuroendocrine transition (TNT).
Collapse
Affiliation(s)
- Daniel J Salas-Escabillas
- Cancer Biology, University of Michigan, Ann Arbor, MI, USA; Department of Surgery, Henry Ford Health, Detroit, MI, USA; Department of Pharmacology and Toxicology, Michigan State University, Lansing, MI, USA
| | - Megan T Hoffman
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Sydney M Brender
- Department of Surgery, Henry Ford Health, Detroit, MI, USA; Department of Pharmacology and Toxicology, Michigan State University, Lansing, MI, USA
| | - Jacee S Moore
- Department of Surgery, Henry Ford Health, Detroit, MI, USA; Department of Pharmacology and Toxicology, Michigan State University, Lansing, MI, USA
| | - Hui-Ju Wen
- Department of Surgery, Henry Ford Health, Detroit, MI, USA; Department of Pharmacology and Toxicology, Michigan State University, Lansing, MI, USA
| | - Simone Benitz
- Department of Surgery, Henry Ford Health, Detroit, MI, USA; Department of Pharmacology and Toxicology, Michigan State University, Lansing, MI, USA
| | - Erick T Davis
- Department of Surgery, Henry Ford Health, Detroit, MI, USA; Department of Pharmacology and Toxicology, Michigan State University, Lansing, MI, USA
| | - Daniel Long
- Department of Surgery, Henry Ford Health, Detroit, MI, USA; Department of Pharmacology and Toxicology, Michigan State University, Lansing, MI, USA
| | - Allison M Wombwell
- Department of Surgery, Henry Ford Health, Detroit, MI, USA; Department of Pharmacology and Toxicology, Michigan State University, Lansing, MI, USA
| | - Ella Rose D Chianis
- Department of Biological Sciences, Purdue University, West Lafayette, IN, USA
| | | | - Nina G Steele
- Department of Surgery, Henry Ford Health, Detroit, MI, USA; Department of Pharmacology and Toxicology, Michigan State University, Lansing, MI, USA
| | - Rosalie C Sears
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, OR, USA
| | | | - Kathleen E DelGiorno
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Howard C Crawford
- Department of Surgery, Henry Ford Health, Detroit, MI, USA; Department of Pharmacology and Toxicology, Michigan State University, Lansing, MI, USA.
| |
Collapse
|
6
|
Cheng H, Guo H, Wen C, Sun G, Tang F, Li Y. The dual role of gut microbiota in pancreatic cancer: new insights into onset and treatment. Ther Adv Med Oncol 2025; 17:17588359251324882. [PMID: 40093983 PMCID: PMC11909682 DOI: 10.1177/17588359251324882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Accepted: 02/14/2025] [Indexed: 03/19/2025] Open
Abstract
Pancreatic cancer ranks among the most lethal digestive malignancies, exhibiting a steadily increasing incidence and mortality worldwide. Despite significant advances in cancer research, the 5-year survival rate remains below 10%, predominantly due to delayed diagnosis and limited therapeutic options. Concurrently, the gut microbiota-an integral component of host physiology-has emerged as a crucial player in the pathogenesis of pancreatic cancer. Mounting evidence indicates that alterations in gut microbial composition and function may influence tumor initiation, progression, and response to therapy. This review provides an in-depth examination of the intricate interplay between the gut microbiome and pancreatic cancer, highlighting potential diagnostic biomarkers and exploring microbiome-targeted therapeutic strategies to improve patient outcomes.
Collapse
Affiliation(s)
- Huijuan Cheng
- School of Life Sciences, Lanzhou University, Lanzhou, Gansu, P.R. China
- Gansu Provincial Key Laboratory of Environmental Oncology, Lanzhou University Second Hospital, Lanzhou, Gansu, P.R. China
| | - Hongkai Guo
- The Second School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, P.R. China
| | - Chengming Wen
- The Second School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, P.R. China
| | - Guodong Sun
- The Second School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, P.R. China
- Department of Medical Affairs, Lanzhou University First Hospital, Lanzhou, Gansu, P.R. China
| | - Futian Tang
- Gansu Provincial Key Laboratory of Environmental Oncology, Lanzhou University Second Hospital, Lanzhou, Gansu, P.R. China
| | - Yumin Li
- Gansu Provincial Key Laboratory of Environmental Oncology, Lanzhou University Second Hospital, No. 82, Cuiyingmen, Chengguan, Lanzhou, Gansu 730000, P.R. China
| |
Collapse
|
7
|
Chen J, Zhang X, Zhang G, Zhu F, Liu W. Serum-derived exosomal miR-7977 combined with miR-451a as a potential biomarker for pancreatic ductal adenocarcinoma. BMC Cancer 2025; 25:295. [PMID: 39972247 PMCID: PMC11837301 DOI: 10.1186/s12885-025-13659-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 02/05/2025] [Indexed: 02/21/2025] Open
Abstract
OBJECTIVES To explore the potential of serum exosomal miRNAs as novel biomarkers for pancreatic ductal adenocarcinoma (PDAC). METHODS Serum exosomal miRNAs were screened and verified by microarray analysis and quantitative real-time PCR (qRT-PCR) in patients with PDAC and healthy controls. The correlation between the clinical characteristics of PDAC and candidate exosomal miRNAs was analyzed, and the diagnostic performance of the candidate biomarkers was evaluated. RESULTS Serum exosomal miR-7977 and miR-451a were significantly upregulated in PDAC patients compared with healthy controls, and the levels of miR-7977 and miR-451a in serum exosomes were closely associated with the clinical stage and metastasis of PDAC patients. The area under curve (AUC) values of serum exosomal miR-7977 and miR-451a for PDAC were 0.825 and 0.804 in the training set and 0.796 and 0.830 in the validation set, respectively. A biomarker panel consisting of these two miRNAs resulted in a diagnostic power with an AUC of 0.901 in the training set and 0.918 in the validation set. CONCLUSIONS Serum exosomal miR-7977 and miR-451a might be diagnostic biomarkers for PDAC. These two miRNAs, when combined, exhibit optimal diagnostic performance.
Collapse
Affiliation(s)
- Jia Chen
- Department of Laboratory Medicine, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.
| | - Xue Zhang
- Department of Pathology, Affiliated Hospital of Chengdu University, Chengdu, 610081, China
| | - Guanyi Zhang
- Department of Laboratory Medicine, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Fan Zhu
- Department of Vascular Surgery, Fuwai Yunnan Cardiovascular Disease Hospital, Affiliated Cardiovascular Hospital of Kunming Medical University, Kunming, 650000, China.
- Department of Vascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, 410011, China.
| | - Weiwei Liu
- Department of Laboratory Medicine, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.
| |
Collapse
|
8
|
R S, J V, F P, DA B, A L, E G, A F, C T, L B, B E. 3D modeling to predict vascular involvement in resectable pancreatic adenocarcinoma. Heliyon 2025; 11:e41473. [PMID: 39850404 PMCID: PMC11754173 DOI: 10.1016/j.heliyon.2024.e41473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 11/27/2024] [Accepted: 12/23/2024] [Indexed: 01/25/2025] Open
Abstract
Background Current management of patients with borderline resectable pancreatic adenocarcinoma (BR-PDAC) depends on the degree of involvement of the major arterial and venous structures. The aim of this study was to evaluate 3D segmentation and printing to predict tumor size and vascular involvement of BR-PDAC to improve pre-operative planning of vascular resection and better select patients for neoadjuvant therapy. Methods We retrospectively evaluated 16 patients with BR-PDAC near vascular structures who underwent pancreatoduodenectomy (PD) with or without vascular resection between 2015 and 2021. The pre-operative computed tomography (CT) images were processed by segmentation with 3D reconstruction and printed as 3D models. Two radiologists specialized in pancreatic imaging and two pancreatic surgeons blindly and independently analyzed the pre-operative CT scans and 3D models using a defined checklist. Their evaluations were compared to the pre-operative 2D-CT reports utilized for patient management. A positive delta was defined by the 3D analysis resulting in greater accuracy in predicting vascular involvement as proven intraoperatively or histopathologically. Results Fourteen PD, one total pancreatectomy, and one exploratory laparotomy were performed. Ten patients had a positive delta concerning vascular involvement of the superior mesenteric or portal vein. Tumor extension was also more accurately evaluated by 3D modeling than by 2D-CT (p < 0.05). Conclusions Our pilot study demonstrates that 3D segmentation can provide additional information for choosing the best treatment strategy and surgical plain in patients with BR-PDAC. Especially for upcoming mini-invasive techniques like laparoscopic and robotic resections, better pre-operative planning is essential to allow safety and prevent vascular injury.
Collapse
Affiliation(s)
- Sguinzi R
- Department of General Surgery, Fribourg Cantonal Hospital, 1700, Fribourg, Switzerland
| | - Vidal J
- Department of Radiology, Fribourg Cantonal Hospital, 1700, Fribourg, Switzerland
| | - Poroes F
- Department of Radiology, Fribourg Cantonal Hospital, 1700, Fribourg, Switzerland
| | - Bartolucci DA
- Department of Radiology, Fribourg Cantonal Hospital, 1700, Fribourg, Switzerland
| | - Litchinko A
- Department of General Surgery, University Hospital of Geneva, 1205, Switzerland
| | - Gossin E
- University of Fribourg, Faculty of Science and Medicine - Section of Medicine, 1700, Fribourg, Switzerland
| | - Fingerhut A
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, PR China
| | - Toso C
- Department of General Surgery, University Hospital of Geneva, 1205, Switzerland
| | - Buhler L
- Department of General Surgery, Fribourg Cantonal Hospital, 1700, Fribourg, Switzerland
| | - Egger B
- Department of General Surgery, Fribourg Cantonal Hospital, 1700, Fribourg, Switzerland
- University of Fribourg, Faculty of Science and Medicine - Section of Medicine, 1700, Fribourg, Switzerland
| |
Collapse
|
9
|
Zhan T, Betge J, Schulte N, Dreikhausen L, Hirth M, Li M, Weidner P, Leipertz A, Teufel A, Ebert MP. Digestive cancers: mechanisms, therapeutics and management. Signal Transduct Target Ther 2025; 10:24. [PMID: 39809756 PMCID: PMC11733248 DOI: 10.1038/s41392-024-02097-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 10/20/2024] [Accepted: 11/29/2024] [Indexed: 01/16/2025] Open
Abstract
Cancers of the digestive system are major contributors to global cancer-associated morbidity and mortality, accounting for 35% of annual cases of cancer deaths. The etiologies, molecular features, and therapeutic management of these cancer entities are highly heterogeneous and complex. Over the last decade, genomic and functional studies have provided unprecedented insights into the biology of digestive cancers, identifying genetic drivers of tumor progression and key interaction points of tumor cells with the immune system. This knowledge is continuously translated into novel treatment concepts and targets, which are dynamically reshaping the therapeutic landscape of these tumors. In this review, we provide a concise overview of the etiology and molecular pathology of the six most common cancers of the digestive system, including esophageal, gastric, biliary tract, pancreatic, hepatocellular, and colorectal cancers. We comprehensively describe the current stage-dependent pharmacological management of these malignancies, including chemo-, targeted, and immunotherapy. For each cancer entity, we provide an overview of recent therapeutic advancements and research progress. Finally, we describe how novel insights into tumor heterogeneity and immune evasion deepen our understanding of therapy resistance and provide an outlook on innovative therapeutic strategies that will shape the future management of digestive cancers, including CAR-T cell therapy, novel antibody-drug conjugates and targeted therapies.
Collapse
Affiliation(s)
- Tianzuo Zhan
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- DKFZ Hector Cancer Institute at University Medical Center Mannheim, Mannheim, Germany
- Mannheim Cancer Center, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Molecular Medicine Partnership Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Johannes Betge
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- DKFZ Hector Cancer Institute at University Medical Center Mannheim, Mannheim, Germany
- Mannheim Cancer Center, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Junior Clinical Cooperation Unit Translational Gastrointestinal Oncology and Preclinical Models, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Nadine Schulte
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Mannheim Cancer Center, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Lena Dreikhausen
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Molecular Medicine Partnership Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Michael Hirth
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Moying Li
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Philip Weidner
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Antonia Leipertz
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Andreas Teufel
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Matthias P Ebert
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.
- DKFZ Hector Cancer Institute at University Medical Center Mannheim, Mannheim, Germany.
- Mannheim Cancer Center, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.
- Molecular Medicine Partnership Unit, European Molecular Biology Laboratory, Heidelberg, Germany.
| |
Collapse
|
10
|
Konikoff T, Loebl N, Benson AA, Green O, Sandler H, Gingold-Belfer R, Levi Z, Perl L, Dotan I, Shamah S. Enhancing detection of various pancreatic lesions on endoscopic ultrasound through artificial intelligence: a basis for computer-aided detection systems. J Gastroenterol Hepatol 2025; 40:235-240. [PMID: 39538430 DOI: 10.1111/jgh.16814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 10/24/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND AND AIM Endoscopic ultrasound (EUS) is the most sensitive method for evaluation of pancreatic lesions but is limited by significant operator dependency. Artificial intelligence (AI), in the form of computer-aided detection (CADe) systems, has shown potential in increasing accuracy and bridging operator dependency in several endoscopic domains. However, the complexity of integrating AI into EUS is far more challenging. This aims to develop and test the basis for a CADe system for real-time detection and segmentation of all pancreatic lesions. METHODS In this single-center study EUS studies of pancreatic findings were included. Lesions were outlined by two expert (>5 years performing EUS) endoscopists, and the two leading types of models were benchmarked. The models' performance was evaluated through per-pixel intersection over union (IoU). RESULTS A total of 1497 EUS images from 165 patients were evaluated. The dataset included malignancies, neuroendocrine tumors, benign cysts, chronic and acute pancreatitis, normal fatty pancreas, and benign lesions. The best model demonstrated detection and segmentation on the test set with a mean IoU of 0.73, achieving a PPV, NPV, total accuracy, and ROC of 0.82, 0.96, 0.95, and 0.95, respectively. The algorithm is adaptable for real-time processing. CONCLUSIONS We developed and tested deep learning models for real-time detection and segmentation of pancreatic lesions during EUS with promising results. This constitutes the basis for a CADe system for EUS, which could be valuable in future detection and evaluation of pancreatic lesions. Further studies for validation and generalization are underway.
Collapse
Affiliation(s)
- Tom Konikoff
- Division of Gastroenterology, Rabin Medical Center, Petach-Tikva, Israel
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Nadav Loebl
- Rabin Medical Center Innovation Lab, Rabin Medical Center, Petah Tikva, Israel
| | - Ariel A Benson
- Hadassah Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Institute of Gastroenterology and Liver Diseases, Jerusalem, Israel
| | - Orr Green
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Hunter Sandler
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Rachel Gingold-Belfer
- Division of Gastroenterology, Rabin Medical Center, Petach-Tikva, Israel
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Zohar Levi
- Division of Gastroenterology, Rabin Medical Center, Petach-Tikva, Israel
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Leor Perl
- Rabin Medical Center Innovation Lab, Rabin Medical Center, Petah Tikva, Israel
| | - Iris Dotan
- Division of Gastroenterology, Rabin Medical Center, Petach-Tikva, Israel
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Steven Shamah
- Division of Gastroenterology, Rabin Medical Center, Petach-Tikva, Israel
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
11
|
Palencia-Campos A, Ruiz-Cañas L, Abal-Sanisidro M, López-Gil JC, Batres-Ramos S, Saraiva SM, Yagüe B, Navarro D, Alcalá S, Rubiolo JA, Bidan N, Sánchez L, Mura S, Hermann PC, de la Fuente M, Sainz B. Reprogramming tumor-associated macrophages with lipid nanosystems reduces PDAC tumor burden and liver metastasis. J Nanobiotechnology 2024; 22:795. [PMID: 39719597 DOI: 10.1186/s12951-024-03010-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 11/12/2024] [Indexed: 12/26/2024] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) requires innovative therapeutic strategies to counteract its progression and metastatic potential. Since the majority of patients are diagnosed with advanced metastatic disease, treatment strategies targeting not only the primary tumor but also metastatic lesions are needed. Tumor-Associated Macrophages (TAMs) have emerged as central players, significantly influencing PDAC progression and metastasis. Our objective was to validate an innovative therapeutic strategy involving the reprogramming of TAMs using lipid nanosystems to prevent the formation of a pro-metastatic microenvironment in the liver. RESULTS In vitro results demonstrate that M2-polarized macrophages lose their M2-phenotype following treatment with lipid nanoemulsions composed of vitamin E and sphingomyelin (VitE:SM), transitioning to an M0/M1 state. Specifically, VitE:SM nanoemulsion treatment decreased the expression of macrophage M2 markers such as Arg1 and Egr2, while M1 markers such as Cd86, Il-1b and Il-12b increased. Additionally, the TGF-βR1 inhibitor Galunisertib (LY2157299) was loaded into VitE:SM nanoemulsions and delivered to C57BL/6 mice orthotopically injected with KPC PDAC tumor cells. Treated mice showed diminished primary tumor growth and reduced TAM infiltration in the liver. Moreover, we observed a decrease in liver metastasis with the nanoemulsion treatment in an intrasplenic model of PDAC liver metastasis. Finally, we validated the translatability of our VitE:SM nanosystem therapy in a human cell-based 3D co-culture model in vivo, underscoring the pivotal role of macrophages in the nanosystem's therapeutic effect in the context of human PDAC metastasis. CONCLUSIONS The demonstrated effectiveness and safety of our nanosystem therapy highlights a promising therapeutic approach for PDAC, showcasing its potential in reprogramming TAMs and mitigating the occurrence of liver metastasis.
Collapse
Affiliation(s)
- Adrián Palencia-Campos
- Cancer Stem Cells and Fibroinflammatory Microenvironment Group, Instituto de Investigaciones Biomédicas (IIBm) Sols-Morreale CSIC-UAM, 28029, Madrid, Spain
- Biomarkers and Personalized Approach to Cancer Group (BIOPAC), Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Area 3 Cancer, 28049, Madrid, Spain
| | - Laura Ruiz-Cañas
- Cancer Stem Cells and Fibroinflammatory Microenvironment Group, Instituto de Investigaciones Biomédicas (IIBm) Sols-Morreale CSIC-UAM, 28029, Madrid, Spain
- Biomarkers and Personalized Approach to Cancer Group (BIOPAC), Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Area 3 Cancer, 28049, Madrid, Spain
- Biobanco Hospital Universitario Ramón y Cajal, IRYCIS, Madrid, Spain
| | - Marcelina Abal-Sanisidro
- Nano-Oncology and Translational Therapeutics Group, IDIS, Complexo Hospitalario Universitario de Santiago de Compostela, 15706, Santiago de Compostela, Spain
- University of Santiago de Compostela (USC), 15782, Santiago de Compostela, Spain
- Centro de Investigación Biomédica en Red, CIBERONC, ISCIII, Área Cáncer, Madrid, Spain
| | - Juan Carlos López-Gil
- Cancer Stem Cells and Fibroinflammatory Microenvironment Group, Instituto de Investigaciones Biomédicas (IIBm) Sols-Morreale CSIC-UAM, 28029, Madrid, Spain
- Biomarkers and Personalized Approach to Cancer Group (BIOPAC), Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Area 3 Cancer, 28049, Madrid, Spain
- Department of Biochemistry, Autónoma University of Madrid (UAM), 28029, Madrid, Spain
| | - Sandra Batres-Ramos
- Cancer Stem Cells and Fibroinflammatory Microenvironment Group, Instituto de Investigaciones Biomédicas (IIBm) Sols-Morreale CSIC-UAM, 28029, Madrid, Spain
- Biomarkers and Personalized Approach to Cancer Group (BIOPAC), Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Area 3 Cancer, 28049, Madrid, Spain
| | - Sofia Mendes Saraiva
- Nano-Oncology and Translational Therapeutics Group, IDIS, Complexo Hospitalario Universitario de Santiago de Compostela, 15706, Santiago de Compostela, Spain
- Centro de Investigación Biomédica en Red, CIBERONC, ISCIII, Área Cáncer, Madrid, Spain
| | - Balbino Yagüe
- Cancer Stem Cells and Fibroinflammatory Microenvironment Group, Instituto de Investigaciones Biomédicas (IIBm) Sols-Morreale CSIC-UAM, 28029, Madrid, Spain
- Biomarkers and Personalized Approach to Cancer Group (BIOPAC), Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Area 3 Cancer, 28049, Madrid, Spain
| | - Diego Navarro
- Cancer Stem Cells and Fibroinflammatory Microenvironment Group, Instituto de Investigaciones Biomédicas (IIBm) Sols-Morreale CSIC-UAM, 28029, Madrid, Spain
- Biomarkers and Personalized Approach to Cancer Group (BIOPAC), Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Area 3 Cancer, 28049, Madrid, Spain
- Department of Biochemistry, Autónoma University of Madrid (UAM), 28029, Madrid, Spain
| | - Sonia Alcalá
- Cancer Stem Cells and Fibroinflammatory Microenvironment Group, Instituto de Investigaciones Biomédicas (IIBm) Sols-Morreale CSIC-UAM, 28029, Madrid, Spain
- Biomarkers and Personalized Approach to Cancer Group (BIOPAC), Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Area 3 Cancer, 28049, Madrid, Spain
| | - Juan A Rubiolo
- Department of Zoology, Genetics and Physical Anthropology, Faculty of Veterinary, University of Santiago de Compostela (USC), Lugo, Spain
- Laboratorio Mixto de Biotecnología Acuática, Facultad de Ciencias Bioquímicas y Farmacéuticas, UNR, 2000, Rosario, Argentina
| | - Nadège Bidan
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, 91400, Orsay, France
| | - Laura Sánchez
- Department of Zoology, Genetics and Physical Anthropology, Faculty of Veterinary, University of Santiago de Compostela (USC), Lugo, Spain
| | - Simona Mura
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, 91400, Orsay, France
| | | | - María de la Fuente
- Nano-Oncology and Translational Therapeutics Group, IDIS, Complexo Hospitalario Universitario de Santiago de Compostela, 15706, Santiago de Compostela, Spain
- Centro de Investigación Biomédica en Red, CIBERONC, ISCIII, Área Cáncer, Madrid, Spain
- DIVERSA Technologies S.L, Edificio Emprendia, Campus Sur, 15782, Santiago de Compostela, Spain
| | - Bruno Sainz
- Cancer Stem Cells and Fibroinflammatory Microenvironment Group, Instituto de Investigaciones Biomédicas (IIBm) Sols-Morreale CSIC-UAM, 28029, Madrid, Spain.
- Biomarkers and Personalized Approach to Cancer Group (BIOPAC), Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Area 3 Cancer, 28049, Madrid, Spain.
- Centro de Investigación Biomédica en Red, CIBERONC, ISCIII, Área Cáncer, Madrid, Spain.
| |
Collapse
|
12
|
Tanaka H, Mise Y, Takahashi A, Kawano F, Takeda Y, Imamura H, Ichida H, Yoshioka R, Saiura A. Analyzing the high frequency of false-positive carcinoembryonic antigen elevations in postoperative pancreatic ductal adenocarcinoma. Langenbecks Arch Surg 2024; 410:6. [PMID: 39672933 DOI: 10.1007/s00423-024-03577-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 12/09/2024] [Indexed: 12/15/2024]
Abstract
PURPOSE The dynamics of postoperative carcinoembryonic antigen (CEA) in pancreatic ductal adenocarcinoma (PDAC) patients have not been well assessed. This study investigated the correlation between postoperative CEA elevations and tumor recurrence. METHODS Medical records were retrospectively analyzed for 84 patients who received curative resection for PDAC from January 2019 to December 2020. Postoperative CEA levels were monitored for a minimum of 12 months. False-positive CEA elevation was defined as a CEA level exceeding 5 ng/mL without evidence of recurrence in imaging studies. RESULTS Of the examined patients, 59 (70%) exhibited CEA > 5 ng/mL within the observation period. The sensitivity and specificity of elevated CEA levels for detecting recurrence were 84% and 41%, respectively. CEA elevations without tumor recurrence were observed in 27 patients, indicating a false-positive rate of 59%. More than half of these patients demonstrated peak CEA levels between 5 and 10 ng/mL, while only true-positive patients exhibited CEA levels exceeding 40.0 ng/mL. CONCLUSION CEA may rise in more than half of postoperative PDAC patients without recurrence. CEA alone is not a robust postoperative marker.
Collapse
Affiliation(s)
- Haruka Tanaka
- Department of Hepatobiliary-Pancreatic Surgery, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Yoshihiro Mise
- Department of Hepatobiliary-Pancreatic Surgery, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan.
| | - Atsushi Takahashi
- Department of Hepatobiliary-Pancreatic Surgery, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Fumihiro Kawano
- Department of Hepatobiliary-Pancreatic Surgery, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Yoshinori Takeda
- Department of Hepatobiliary-Pancreatic Surgery, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Hiroshi Imamura
- Department of Hepatobiliary-Pancreatic Surgery, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Hirofumi Ichida
- Department of Hepatobiliary-Pancreatic Surgery, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Ryuji Yoshioka
- Department of Hepatobiliary-Pancreatic Surgery, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Akio Saiura
- Department of Hepatobiliary-Pancreatic Surgery, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| |
Collapse
|
13
|
Xi Y, Zhao Z, Wang F, Zhang D, Guo Y. IRTIDP: A simple integrated real-time isolation and detection platform for small extracellular vesicles Glypican-1 in pancreatic cancer patients. Talanta 2024; 280:126766. [PMID: 39191106 DOI: 10.1016/j.talanta.2024.126766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 08/12/2024] [Accepted: 08/23/2024] [Indexed: 08/29/2024]
Abstract
Glypican-1 (GPC-1) protein-positive small extracellular vesicles (GPC-1+-sEV) have been proposed as potential biomarkers for early diagnosis of pancreatic cancer. In this study, we present an integrated real-time isolation and detection platform (IRTIDP) to capture and analyze GPC-1+-sEV directly from sera of pancreatic cancer patients. First, CD63 antibody-modified metal-organic framework (MOF) materials were utilized to enrich sEVs with a capture efficiency of 93.93 %. Second, a SERS probe was constructed by Raman reporter 4-MBA and GPC-1 antibody modified SERS active silver nanoparticles (AgNPs), which formed a sandwich complex structure of "MOFs@GPC-1+-sEV@AgNPs-4-MBA" with MOFs-enriched sEVs. The IRTSDP can complete the capture and detection process within 35 min, with a detection limit for 1 GPC-1+-sEV/μL, and linear range between 105∼109 GPC-1+-sEV/mL. Furthermore, this approach has been applied to quantify serum sEV GPC-1 in clinical pancreatic cancer patients. Based on the SERS intensity analysis, pancreatic cancer patients can be distinguished from pancreatic cystadenoma patients and healthy individuals effectively using this innovative platform that provides highly specific and sensitive means for early diagnosis of pancreatic cancer as well as other tumor types.
Collapse
Affiliation(s)
- Yuge Xi
- Nanobiosensing and Microfluidic Point-of-Care Testing, Key Laboratory of Luzhou, Department of Clinical Laboratory, The Affiliated Traditional Chinese Medicine Hospital,Southwest Medical University, Luzhou, 646000, PR China; Department of Laboratory Medicine, The People's Hospital of Chongging Liangjiang New Area, No. 199 Ren Xing Road, Yubei, Chongqing, 401121, PR China
| | - Zijun Zhao
- Nanobiosensing and Microfluidic Point-of-Care Testing, Key Laboratory of Luzhou, Department of Clinical Laboratory, The Affiliated Traditional Chinese Medicine Hospital,Southwest Medical University, Luzhou, 646000, PR China
| | - Fen Wang
- Nanobiosensing and Microfluidic Point-of-Care Testing, Key Laboratory of Luzhou, Department of Clinical Laboratory, The Affiliated Traditional Chinese Medicine Hospital,Southwest Medical University, Luzhou, 646000, PR China
| | - Dan Zhang
- Nanobiosensing and Microfluidic Point-of-Care Testing, Key Laboratory of Luzhou, Department of Clinical Laboratory, The Affiliated Traditional Chinese Medicine Hospital,Southwest Medical University, Luzhou, 646000, PR China
| | - Yongcan Guo
- Nanobiosensing and Microfluidic Point-of-Care Testing, Key Laboratory of Luzhou, Department of Clinical Laboratory, The Affiliated Traditional Chinese Medicine Hospital,Southwest Medical University, Luzhou, 646000, PR China.
| |
Collapse
|
14
|
Xu X, Zhang X, Deng X, Sheng F, Cao G, Fu D, Guan M. Quantitative detection of miR-25 for early diagnosis, postoperative assessment and TNM staging of pancreatic cancer. HUMAN GENE 2024; 42:201350. [DOI: 10.1016/j.humgen.2024.201350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2025]
|
15
|
Biswas S, Afrose S, Mita MA, Hasan MR, Shimu MSS, Zaman S, Saleh MA. Next-Generation Sequencing: An Advanced Diagnostic Tool for Detection of Pancreatic Disease/Disorder. JGH Open 2024; 8:e70061. [PMID: 39605899 PMCID: PMC11599877 DOI: 10.1002/jgh3.70061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 11/05/2024] [Accepted: 11/11/2024] [Indexed: 11/29/2024]
Abstract
The pancreas is involved in digestion and glucose regulation in the human body. Given the recognized link between chronic pancreatitis and pancreatic cancer, addressing pancreatic disorders and pancreatic cancer is particularly challenging. This review aims to highlight the limitations of traditional methods in diagnosing pancreatic disorders and cancer and explore several next-generation sequencing (NGS) approaches as a promising alternative. There are distinct clinical symptoms that are shared by a number of clinical phenotypes of pancreatic illness induced by particular genetic mutations. Traditional diagnostic methods encompass computed tomography, magnetic resonance imaging, contrast-enhanced Doppler ultrasound, endoscopic ultrasound, endoscopic retrograde cholangiopancreatography, transabdominal ultrasound, laparoscopy, and positron emission tomography have a prognostic ability of only 5% or less and a 5-year survival rate. Genetic sequencing can be employed as an alternative to conventional diagnostic techniques. Sanger sequencing and NGS are currently largely operated genome analysis, with no exception for pancreatic disease diagnosis. The NGS methods can sequence millions to billions of short DNA fragments, enabling enormous sample screening in a short amount of time with low-abundance detection, like in 0.1%-1% mutation prevalence declining approximate cost. Whole-genome sequencing, whole-exome sequencing, RNA sequencing, and single-cell NGS are a few NGS methods utilized to diagnose pancreatic disease. For both research and clinical applications, the NGS techniques can provide a precise diagnosis of pancreatic disorders in a short amount of time at a reasonable expenditure.
Collapse
Affiliation(s)
- Suvro Biswas
- Miocrobiology Laboratory, Department of Genetic Engineering and BiotechnologyUniversity of RajshahiBangladesh
| | - Shamima Afrose
- Department of Genetic Engineering and BiotechnologyUniversity of RajshahiRajshahiBangladesh
| | - Mohasana Akter Mita
- Department of Genetic Engineering and BiotechnologyUniversity of RajshahiRajshahiBangladesh
| | - Md. Robiul Hasan
- Department of Genetic Engineering and BiotechnologyUniversity of RajshahiRajshahiBangladesh
| | | | - Shahriar Zaman
- Miocrobiology Laboratory, Department of Genetic Engineering and BiotechnologyUniversity of RajshahiBangladesh
| | - Md. Abu Saleh
- Miocrobiology Laboratory, Department of Genetic Engineering and BiotechnologyUniversity of RajshahiBangladesh
| |
Collapse
|
16
|
Guo M, Sheng W, Yuan X, Wang X. Neutrophils as promising therapeutic targets in pancreatic cancer liver metastasis. Int Immunopharmacol 2024; 140:112888. [PMID: 39133956 DOI: 10.1016/j.intimp.2024.112888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 08/01/2024] [Accepted: 08/04/2024] [Indexed: 09/01/2024]
Abstract
Pancreatic cancer is characterized by an extremely poor prognosis and presents significant treatment challenges. Liver metastasis is the leading cause of death in patients with pancreatic cancer. Recent studies have highlighted the significant impact of neutrophils on tumor occurrence and progression, as well as their crucial role in the pancreatic cancer tumor microenvironment. Neutrophil infiltration plays a critical role in the progression and prognosis of pancreatic cancer. Neutrophils contribute to pancreatic cancer liver metastasis through various mechanisms, including angiogenesis, immune suppression, immune evasion, and epithelial-mesenchymal transition (EMT). Therefore, targeting neutrophils holds promise as an important therapeutic strategy for inhibiting pancreatic cancer liver metastasis. This article provides a summary of research findings on the involvement of neutrophils in pancreatic cancer liver metastasis and analyzes their potential as therapeutic targets. This research may provide new insights for the treatment of pancreatic cancer and improve the prognosis of patients with this disease.
Collapse
Affiliation(s)
- Minjie Guo
- Department of Thoracic Oncology, Cancer Institute of Jiangsu University, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Wanying Sheng
- Department of Thoracic Oncology, Cancer Institute of Jiangsu University, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Xiao Yuan
- Cancer Institute of Jiangsu University, Zhenjiang, China.
| | - Xu Wang
- Department of Thoracic Oncology, Cancer Institute of Jiangsu University, Affiliated Hospital of Jiangsu University, Zhenjiang, China.
| |
Collapse
|
17
|
Yuan Y, Yang J, Sun Q, Huang Y. Precomputed low-frequency lighting in cinematic volume rendering. PLoS One 2024; 19:e0312339. [PMID: 39432508 PMCID: PMC11493242 DOI: 10.1371/journal.pone.0312339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 10/04/2024] [Indexed: 10/23/2024] Open
Abstract
Cinematic Rendering (CR) employs physical models such as ray tracing and global illumination to simulate real-world light phenomena, producing high-quality images with rich details. In the medical field, CR can significantly aid doctors in accurate diagnosis and preoperative planning. However, doctors require efficient real-time rendering when using CR, which presents a challenge due to the substantial computing resources demanded by CR's ray tracing and global illumination models. Precomputed lighting can enhance the efficiency of real-time rendering by freezing certain scene variables. Typically, precomputed methods freeze geometry and materials. However, since the physical rendering of medical images relies on volume data rendering of transfer functions, the CR algorithm cannot utilize precomputed methods directly. To improve the rendering efficiency of the CR algorithm, we propose a precomputed low-frequency lighting method. By simulating the lighting pattern of shadowless surgical lamps, we adopt a spherical distribution of multiple light sources, with each source capable of illuminating the entire volume of data. Under the influence of these large-area multi-light sources, the precomputed lighting adheres to physical principles, resulting in shadow-free and uniformly distributed illumination. We integrated this precomputed method into the ray-casting algorithm, creating an accelerated CR algorithm that achieves more than twice the rendering efficiency of traditional CR rendering.
Collapse
Affiliation(s)
- Yuliang Yuan
- School of Computer Science and Engineeing, Northeastern University, Shenyang, Liaoning, P. R. China
| | - Jinzhu Yang
- School of Computer Science and Engineeing, Northeastern University, Shenyang, Liaoning, P. R. China
| | - Qi Sun
- School of Computer Science and Engineeing, Northeastern University, Shenyang, Liaoning, P. R. China
| | - Yan Huang
- School of Computer Science and Engineeing, Northeastern University, Shenyang, Liaoning, P. R. China
| |
Collapse
|
18
|
Lee YN, Sung MK, Hwang DW, Park Y, Kwak BJ, Lee W, Song KB, Lee JH, Yoo C, Kim KP, Chang HM, Ryoo BY, Kim SC. Clinical Outcomes of Surgery after Neoadjuvant Chemotherapy in Locally Advanced Pancreatic Ductal Adenocarcinoma. Cancer Res Treat 2024; 56:1240-1251. [PMID: 38901824 PMCID: PMC11491246 DOI: 10.4143/crt.2023.977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 06/10/2024] [Indexed: 06/22/2024] Open
Abstract
PURPOSE Clinical outcomes of surgery after neoadjuvant chemotherapy have not been investigated for locally advanced pancreatic cancer (LAPC), despite well-established outcomes in borderline resectable pancreatic cancer (BRPC). This study aimed to investigate the clinical outcomes of patients with LAPC who underwent curative resection following neoadjuvant chemotherapy. MATERIALS AND METHODS We retrospectively reviewed the records of patients diagnosed with pancreatic adenocarcinoma between January 2017 and December 2020. RESULTS Among 1,358 patients, 260 underwent surgery following neoadjuvant chemotherapy. Among 356 LAPC patients, 98 (27.5%) and 147 (35.1%) of 418 BRPC patients underwent surgery after neoadjuvant chemotherapy. Compared to resectable pancreatic cancer (resectable PC) with upfront surgery, both LAPC and BRPC exhibited higher rates of venous resection (28.6% vs. 49.0% vs. 4.0%), arterial resection (30.6% vs. 6.8% vs. 0.5%) and greater estimated blood loss (260.5 vs. 213.1 vs. 70.4 mL). However, hospital stay, readmission rates, and postoperative pancreatic fistula rates (grade B or C) did not differ significantly between LAPC, BRPC, and resectable PC. Overall and relapse-free survival did not differ significantly between LAPC and BRPC patients. The median overall survival was 37.3 months for LAPC and 37.0 months for BRPC. The median relapse-free survival was 22.7 months for LAPC and 26.0 months for BRPC. CONCLUSION Overall survival time and postoperative complications in LAPC patients who underwent curative resection following neoadjuvant chemotherapy showed similar results to those of BRPC patients. Further research is needed to identify specific sub-populations of LAPC patients who benefit most from conversion surgery and to minimize postoperative complications.
Collapse
Affiliation(s)
- Yoo Na Lee
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Min Kyu Sung
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Dae Wook Hwang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Yejong Park
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Bong Jun Kwak
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Woohyung Lee
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Ki Byung Song
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jae Hoon Lee
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Changhoon Yoo
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Kyu-Pyo Kim
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Heung-Moon Chang
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Baek-Yeol Ryoo
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Song Cheol Kim
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Brain Korea 21 Project, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| |
Collapse
|
19
|
Jain V, Sakhuja P, Agarwal AK, Sirdeshmukh R, Siraj F, Gautam P. Lymph Node Metastasis in Gastrointestinal Carcinomas: A View from a Proteomics Perspective. Curr Oncol 2024; 31:4455-4475. [PMID: 39195316 PMCID: PMC11352871 DOI: 10.3390/curroncol31080333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 05/14/2024] [Accepted: 05/23/2024] [Indexed: 08/29/2024] Open
Abstract
Lymph node metastasis (LNM) is one of the major prognostic factors in human gastrointestinal carcinomas (GICs). The lymph node-positive patients have poorer survival than node-negative patients. LNM is directly associated with the recurrence and poor survival of patients with GICs. The early detection of LNM in patients and designing effective therapies to suppress LNM may significantly impact the survival of these patients. The rapid progress made in proteomic technologies could be successfully applied to identify molecular targets for cancers at high-throughput levels. LC-MS/MS analysis enables the identification of proteins involved in LN metastasis, which can be utilized for diagnostic and therapeutic applications. This review summarizes the studies on LN metastasis in GICs using proteomic approaches to date.
Collapse
Affiliation(s)
- Vaishali Jain
- Indian Council of Medical Research, National Institute of Pathology, New Delhi 110029, India
- Faculty of Health Sciences, Manipal Academy of Higher Education (MAHE), Manipal 576104, India
| | - Puja Sakhuja
- Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi 110002, India
| | - Anil Kumar Agarwal
- Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi 110002, India
| | - Ravi Sirdeshmukh
- Faculty of Health Sciences, Manipal Academy of Higher Education (MAHE), Manipal 576104, India
- Institute of Bioinformatics, International Tech Park, Bangalore 560066, India
| | - Fouzia Siraj
- Indian Council of Medical Research, National Institute of Pathology, New Delhi 110029, India
| | - Poonam Gautam
- Indian Council of Medical Research, National Institute of Pathology, New Delhi 110029, India
| |
Collapse
|
20
|
Sabogal JC, Conde Monroy D, Rey Chaves CE, Ayala D, González J. Delayed gastric emptying after pancreatoduodenectomy: an analysis of risk factors. Updates Surg 2024; 76:1247-1255. [PMID: 38598061 PMCID: PMC11341576 DOI: 10.1007/s13304-024-01795-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 02/25/2024] [Indexed: 04/11/2024]
Abstract
BACKGROUND Delayed gastric emptying (DGE) is a frequent complication after pancreatoduodenectomy. Preoperative factors are limited and controversial. This study aims to identify associated factors related to this complication in the Colombian population. METHODS A retrospective review of a prospectively collected database was conducted. All patients over 18 years of age who underwent pancreaticoduodenectomy were included. Associations with DGE syndrome were evaluated with logistic regression analysis, Odds ratio, and b-coefficient were provided when appropriate. RESULTS 205 patients were included. Male patients constituted 54.15% (n = 111). 53 patients (25.85%) were diagnosed with DGE syndrome. Smoking habit (OR 17.58 p 0.00 95% CI 7.62-40.51), hydromorphone use > 0.6 mg/daily (OR 11.04 p 0.03 95% CI 1.26-96.66), bilirubin levels > 6 mg/dL (OR 2.51 p 0.02 95% CI 1.12-5.61), and pancreatic fistula type B (OR 2.72 p 0.02 CI 1.74-10.00). DISCUSSION Smoking history, opioid use (hydromorphone > 0.6 mg/Daily), type B pancreatic fistula, and bilirubin levels > 6 mg/dL should be considered as risk factors for DGE.
Collapse
Affiliation(s)
- Juan Carlos Sabogal
- Hepatobiliary and Pancreatic Surgery Department, Hospital Universitario Mayor, Méderi, Bogotá, Colombia
- School of Medicine, Universidad del Rosario, Bogotá, Colombia
| | - Danny Conde Monroy
- Hepatobiliary and Pancreatic Surgery Department, Hospital Universitario Mayor, Méderi, Bogotá, Colombia
- School of Medicine, Universidad del Rosario, Bogotá, Colombia
| | - Carlos Eduardo Rey Chaves
- Estudiante de Posgrado Cirugía General, Facultad de Medicina, Pontificia Universidad Javeriana, Carrera 6A #51A-48, 111711, Bogotá D.C, Colombia.
| | - Daniela Ayala
- Hepatobiliary and Pancreatic Surgery Department, Hospital Universitario Mayor, Méderi, Bogotá, Colombia
| | - Juliana González
- Hepatobiliary and Pancreatic Surgery Department, Hospital Universitario Mayor, Méderi, Bogotá, Colombia
| |
Collapse
|
21
|
Bilreiro C, Andrade L, Santiago I, Marques RM, Matos C. Imaging of pancreatic ductal adenocarcinoma - An update for all stages of patient management. Eur J Radiol Open 2024; 12:100553. [PMID: 38357385 PMCID: PMC10864763 DOI: 10.1016/j.ejro.2024.100553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 02/02/2024] [Accepted: 02/03/2024] [Indexed: 02/16/2024] Open
Abstract
Background Pancreatic ductal adenocarcinoma (PDAC) is a common and lethal cancer. From diagnosis to disease staging, response to neoadjuvant therapy assessment and patient surveillance after resection, imaging plays a central role, guiding the multidisciplinary team in decision-planning. Review aims and findings This review discusses the most up-to-date imaging recommendations, typical and atypical findings, and issues related to each step of patient management. Example cases for each relevant condition are presented, and a structured report for disease staging is suggested. Conclusion Despite current issues in PDAC imaging at different stages of patient management, the radiologist is essential in the multidisciplinary team, as the conveyor of relevant imaging findings crucial for patient care.
Collapse
Affiliation(s)
- Carlos Bilreiro
- Radiology Department, Champalimaud Foundation, Lisbon, Portugal
- Champalimaud Research, Champalimaud Foundation, Lisbon, Portugal
- Nova Medical School, Lisbon, Portugal
| | - Luísa Andrade
- Radiology Department, Champalimaud Foundation, Lisbon, Portugal
| | - Inês Santiago
- Radiology Department, Champalimaud Foundation, Lisbon, Portugal
| | - Rui Mateus Marques
- Nova Medical School, Lisbon, Portugal
- Radiology Department, Hospital de S. José, Lisbon, Portugal
| | - Celso Matos
- Radiology Department, Champalimaud Foundation, Lisbon, Portugal
- Champalimaud Research, Champalimaud Foundation, Lisbon, Portugal
| |
Collapse
|
22
|
Qu J, Xiao X, Wei X, Qian X. A causality-inspired generalized model for automated pancreatic cancer diagnosis. Med Image Anal 2024; 94:103154. [PMID: 38552527 DOI: 10.1016/j.media.2024.103154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 02/29/2024] [Accepted: 03/20/2024] [Indexed: 04/16/2024]
Abstract
Pancreatic cancer (PC) is a severely malignant cancer variant with high mortality. Since PC has no obvious symptoms, most PC patients are belatedly diagnosed at advanced disease stages. Recently, artificial intelligence (AI) approaches have demonstrated promising prospects for early diagnosis of pancreatic cancer. However, certain non-causal factors (such as intensity and texture appearance variations, also called confounders) tend to induce spurious correlation with PC diagnosis. This undermines the generalization performance and the clinical applicability of the AI-based PC diagnosis approaches. Therefore, we propose a causal intervention based automated method for pancreatic cancer diagnosis with contrast-enhanced computerized tomography (CT) images, where a confounding effects reduction scheme is developed for alleviating spurious correlations to achieve unbiased learning, thereby improving the generalization performance. Specifically, a continuous image generation strategy was developed to simulate wide variations of intensity differences caused by imaging heterogeneities, where Monte Carlo sampling is added to further enhance the continuity of simulated images. Then, to enhance the pancreatic texture variability, a texture diversification method was introduced in conjunction with gradient-based data augmentation. Finally, a causal intervention strategy was proposed to alleviate the adverse confounding effects by decoupling the causal and non-causal factors and combining them randomly. Extensive experiments showed remarkable diagnosis performance on a cross-validation dataset. Also, promising generalization performance with an average accuracy of 0.87 was attained on three independent test sets of a total of 782 subjects. Therefore, the proposed method shows high clinical feasibility and applicability for pancreatic cancer diagnosis.
Collapse
Affiliation(s)
- Jiaqi Qu
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, PR China
| | - Xiang Xiao
- Department of Medical Imaging, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, PR China
| | - Xunbin Wei
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, PR China; Peking University Cancer Hospital & Institute, Beijing, 100142, PR China; Biomedical Engineering Department, Peking University, Beijing, 100081, PR China; Institute of Medical Technology, Peking University Health Science Center, Beijing, 100191, PR China; International Cancer Institute, Peking University, Beijing 100191, PR China.
| | - Xiaohua Qian
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, PR China.
| |
Collapse
|
23
|
Huang YP, Yeh CA, Ma YS, Chen PY, Lai KC, Lien JC, Hsieh WT. PW06 suppresses cancer cell metastasis in human pancreatic carcinoma MIA PaCa-2 cells via the inhibitions of p-Akt/mTOR/NF-κB and MMP2/MMP9 signaling pathways in vitro. ENVIRONMENTAL TOXICOLOGY 2024; 39:2768-2781. [PMID: 38264921 DOI: 10.1002/tox.24143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 12/14/2023] [Accepted: 01/06/2024] [Indexed: 01/25/2024]
Abstract
PW06 [(E)-3-(9-ethyl-9H-carbazol-3-yl)-1-(2,5-dimethoxyphenyl) prop-2-en-1-one], a kind of the carbazole derivative containing chalcone moiety, induced cell apoptosis in human pancreatic carcinoma in vitro. There is no investigation to show that PW06 inhibits cancer cell metastasis in human pancreatic carcinoma in vitro. Herein, PW06 (0.1-0.8 μM) significantly exists in the antimetastatic activities of human pancreatic carcinoma MIA PaCa-2 cells in vitro. Wound healing assay shows PW06 at 0.2 μM suppressed cell mobility by 7.45 and 16.55% at 6 and 24 hours of treatments. PW06 at 0.1 and 0.2 μM reduced cell mobility by 14.72 and 21.8% for 48 hours of treatment. Transwell chamber assay indicated PW06 (0.1-0.2 μM) suppressed the cell migration (decreased 26.67-35.42%) and invasion (decreased 48.51-68.66%). Atomic force microscopy assay shows PW06 (0.2 μM) significantly changed the shape of cell morphology. The gelatin zymography assay indicates PW06 decreased MMP2's and MMP9's activities at 48 hours of treatment. Western blotting assay further confirms PW06 reduced levels of MMP2 and MMP9 and increased protein expressions of EGFR, SOS1, and Ras. PW06 also increased the p-JNK, p-ERK, and p-p38. PW06 increased the expression of PI3K, PTEN, Akt, GSK3α/β, and E-cadherin. Nevertheless, results also show PW06 decreased p-Akt, mTOR, NF-κB, p-GSK3β, β-catenin, Snail, N-cadherin, and vimentin in MIA PaCa-2 cells. The confocal laser microscopy examination shows PW06 increased E-cadherin but decreased vimentin in MIA PaCa-2 cells. Together, our findings strongly suggest that PW06 inhibited the p-Akt/mTOR/NF-κB/MMPs pathways, increased E-cadherin, and decreased N-cadherin/vimentin, suppressing the migration and invasion in MIA PaCa-2 cells in vitro.
Collapse
Affiliation(s)
- Yi-Ping Huang
- Department of Physiology, School of Medicine, China Medical University, Taichung, Taiwan
| | - Chun-An Yeh
- Department of Physiology, School of Medicine, China Medical University, Taichung, Taiwan
| | - Yi-Shih Ma
- School of Chinese Medicine for Post-Baccalaureate, College of Medicine, I-Shou University, Kaohsiung, Taiwan
- Department of Chinese Medicine, E-Da Cancer Hospital, Kaohsiung, Taiwan
| | - Po-Yuan Chen
- Department of Biological Science and Technology, College of Life Science, China Medical University, Taichung, Taiwan
| | - Kuang-Chi Lai
- Department of Medical Laboratory Science and Biotechnology, College of Medical Technology, Chung Hwa University of Medical Technology, Tainan, Taiwan
- Department of Surgery, School of Medicine, China Medical University, Taichung, Taiwan
| | - Jin-Cherng Lien
- School of Pharmacy, China Medical University, Taichung, Taiwan
| | - Wen-Tsong Hsieh
- Chinese Medicine Research Center, China Medical University, Taichung, Taiwan
- Department of Pharmacology, China Medical University, Taichung, Taiwan
| |
Collapse
|
24
|
Salas-Escabillas DJ, Hoffman MT, Moore JS, Brender SM, Wen HJ, Benitz S, Davis ET, Long D, Wombwell AM, Steele NG, Sears RC, Matsumoto I, DelGiorno KE, Crawford HC. Tuft cells transdifferentiate to neural-like progenitor cells in the progression of pancreatic cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.12.579982. [PMID: 38405804 PMCID: PMC10888969 DOI: 10.1101/2024.02.12.579982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDA) is partly initiated through the transdifferentiation of acinar cells to metaplastic ducts that act as precursors of neoplasia and cancer. Tuft cells are solitary chemosensory cells not found in the normal pancreas but arise in metaplasia and neoplasia, diminishing as neoplastic lesions progress to carcinoma. Metaplastic tuft cells (mTCs) function to suppress tumor progression through communication with the tumor microenvironment, but their fate during progression is unknown. To determine the fate of mTCs during PDA progression, we have created a lineage tracing model that uses a tamoxifen-inducible tuft-cell specific Pou2f3CreERT/+ driver to induce transgene expression, including the lineage tracer tdTomato or the oncogene Myc. mTC lineage trace models of pancreatic neoplasia and carcinoma were used to follow mTC fate. We found that mTCs, in the carcinoma model, transdifferentiate into neural-like progenitor cells (NRPs), a cell type associated with poor survival in PDA patients. Using conditional knock-out and overexpression systems, we found that Myc activity in mTCs is necessary and sufficient to induce this Tuft-to-Neuroendocrine-Transition (TNT).
Collapse
Affiliation(s)
- Daniel J. Salas-Escabillas
- Cancer Biology, University of Michigan, Ann Arbor, MI
- Department of Surgery, Henry Ford Health, Detroit, MI
| | - Megan T. Hoffman
- Department of Immunology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | | | | | - Hui-Ju Wen
- Department of Surgery, Henry Ford Health, Detroit, MI
| | - Simone Benitz
- Department of Surgery, Henry Ford Health, Detroit, MI
| | | | - Dan Long
- Department of Surgery, Henry Ford Health, Detroit, MI
| | | | | | - Rosalie C. Sears
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, OR
| | | | - Kathleen E. DelGiorno
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN
| | - Howard C. Crawford
- Department of Surgery, Henry Ford Health, Detroit, MI
- Cancer Biology Program, Wayne State University, Detroit, MI
| |
Collapse
|
25
|
Jiang W, Shi X, Zhang X, Li Z, Yue J. Feasibility and safety of contrast-enhanced magnetic resonance-guided adaptive radiotherapy for upper abdominal tumors: A preliminary exploration. Phys Imaging Radiat Oncol 2024; 30:100582. [PMID: 38765880 PMCID: PMC11099332 DOI: 10.1016/j.phro.2024.100582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 04/22/2024] [Accepted: 04/23/2024] [Indexed: 05/22/2024] Open
Abstract
This study investigates the use of contrast-enhanced magnetic resonance (MR) in MR-guided adaptive radiotherapy (MRgART) for upper abdominal tumors. Contrast-enhanced T1-weighted MR (cT1w MR) using half doses of gadoterate was used to guide daily adaptive radiotherapy for tumors poorly visualized without contrast. The use of gadoterate was found to be feasible and safe in 5-fraction MRgART and could improve the contrast-to-noise ratio of MR images. And the use of cT1w MR could reduce the interobserver variation of adaptive tumor delineation compared to plain T1w MR (4.41 vs. 6.58, p < 0.001) and T2w MR (4.41 vs. 7.42, p < 0.001).
Collapse
Affiliation(s)
- Wenheng Jiang
- Department of Graduate, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Xihua Shi
- Department of Radiation Physics, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Xiang Zhang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Zhenjiang Li
- Department of Radiation Physics, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Jinbo Yue
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| |
Collapse
|
26
|
Liu K, Li Q, Wang X, Fu C, Sun H, Chen C, Zeng M. Feasibility of deep learning-reconstructed thin-slice single-breath-hold HASTE for detecting pancreatic lesions: A comparison with two conventional T2-weighted imaging sequences. RESEARCH IN DIAGNOSTIC AND INTERVENTIONAL IMAGING 2024; 9:100038. [PMID: 39076579 PMCID: PMC11265199 DOI: 10.1016/j.redii.2023.100038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Accepted: 12/26/2023] [Indexed: 07/31/2024]
Abstract
Objective The objective of this study was to evaluate the clinical feasibility of deep learning reconstruction-accelerated thin-slice single-breath-hold half-Fourier single-shot turbo spin echo imaging (HASTEDL) for detecting pancreatic lesions, in comparison with two conventional T2-weighted imaging sequences: compressed-sensing HASTE (HASTECS) and BLADE. Methods From March 2022 to January 2023, a total of 63 patients with suspected pancreatic-related disease underwent the HASTEDL, HASTECS, and BLADE sequences were enrolled in this retrospectively study. The acquisition time, the pancreatic lesion conspicuity (LCP), respiratory motion artifact (RMA), main pancreatic duct conspicuity (MPDC), overall image quality (OIQ), signal-to-noise ratio (SNR), and contrast-noise-ratio (CNR) of the pancreatic lesions were compared among the three sequences by two readers. Results The acquisition time of both HASTEDL and HASTECS was 16 s, which was significantly shorter than that of 102 s for BLADE. In terms of qualitative parameters, Reader 1 and Reader 2 assigned significantly higher scores to the LCP, RMA, MPDC, and OIQ for HASTEDL compared to HASTECS and BLADE sequences; As for the quantitative parameters, the SNR values of the pancreatic head, body, tail, and lesions, the CNR of the pancreatic lesion measured by the two readers were also significantly higher for HASTEDL than for HASTECS and BLADE sequences. Conclusions Compared to conventional T2WI sequences (HASTECS and BLADE), deep-learning reconstructed HASTE enables thin slice and single-breath-hold acquisition with clinical acceptable image quality for detection of pancreatic lesions.
Collapse
Affiliation(s)
- Kai Liu
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Medical Imaging, Shanghai 200032, China
| | - Qing Li
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Medical Imaging, Shanghai 200032, China
| | - Xingxing Wang
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Caixia Fu
- Siemens (Shenzhen) Magnetic Resonance Ltd., Shenzhen, China
| | - Haitao Sun
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Medical Imaging, Shanghai 200032, China
| | - Caizhong Chen
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Medical Imaging, Shanghai 200032, China
| | - Mengsu Zeng
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Medical Imaging, Shanghai 200032, China
| |
Collapse
|
27
|
Al-Adham ISI, Jaber N, Ali Agha ASA, Al-Remawi M, Al-Akayleh F, Al-Muhtaseb N, Collier PJ. Sporadic regional re-emergent cholera: a 19th century problem in the 21st century. J Appl Microbiol 2024; 135:lxae055. [PMID: 38449342 DOI: 10.1093/jambio/lxae055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 02/24/2024] [Accepted: 03/04/2024] [Indexed: 03/08/2024]
Abstract
Cholera, caused by Vibrio cholerae, is a severe diarrheal disease that necessitates prompt diagnosis and effective treatment. This review comprehensively examines various diagnostic methods, from traditional microscopy and culture to advanced nucleic acid testing like polymerase spiral reaction and rapid diagnostic tests, highlighting their advantages and limitations. Additionally, we explore evolving treatment strategies, with a focus on the challenges posed by antibiotic resistance due to the activation of the SOS response pathway in V. cholerae. We discuss promising alternative treatments, including low-pressure plasma sterilization, bacteriophages, and selenium nanoparticles. The paper emphasizes the importance of multidisciplinary approaches combining novel diagnostics and treatments in managing and preventing cholera, a persistent global health challenge. The current re-emergent 7th pandemic of cholera commenced in 1961 and shows no signs of abeyance. This is probably due to the changing genetic profile of V. cholerae concerning bacterial pathogenic toxins. Given this factor, we argue that the disease is effectively re-emergent, particularly in Eastern Mediterranean countries such as Lebanon, Syria, etc. This review considers the history of the current pandemic, the genetics of the causal agent, and current treatment regimes. In conclusion, cholera remains a significant global health challenge that requires prompt diagnosis and effective treatment. Understanding the history, genetics, and current treatments is crucial in effectively addressing this persistent and re-emergent disease.
Collapse
Affiliation(s)
- Ibrahim S I Al-Adham
- Faculty of Pharmacy and Medical Sciences, University of Petra, Amman 961343, Jordan
| | - Nisrein Jaber
- Faculty of Pharmacy, Al Zaytoonah University of Jordan, Amman 11733, Jordan
| | - Ahmed S A Ali Agha
- Faculty of Pharmacy and Medical Sciences, University of Petra, Amman 961343, Jordan
| | - Mayyas Al-Remawi
- Faculty of Pharmacy and Medical Sciences, University of Petra, Amman 961343, Jordan
| | - Faisal Al-Akayleh
- Faculty of Pharmacy and Medical Sciences, University of Petra, Amman 961343, Jordan
| | - Najah Al-Muhtaseb
- Faculty of Pharmacy and Medical Sciences, University of Petra, Amman 961343, Jordan
| | - Phillip J Collier
- Faculty of Pharmacy and Medical Sciences, University of Petra, Amman 961343, Jordan
| |
Collapse
|
28
|
Dang C, Bian Q, Wang F, Wang H, Liang Z. Machine learning identifies SLC6A14 as a novel biomarker promoting the proliferation and metastasis of pancreatic cancer via Wnt/β-catenin signaling. Sci Rep 2024; 14:2116. [PMID: 38267509 PMCID: PMC10808089 DOI: 10.1038/s41598-024-52646-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 01/22/2024] [Indexed: 01/26/2024] Open
Abstract
Pancreatic cancer (PC) has the poorest prognosis compared to other common cancers because of its aggressive nature, late detection, and resistance to systemic treatment. In this study, we aimed to identify novel biomarkers for PC patients and further explored their function in PC progression. We analyzed GSE62452 and GSE28735 datasets, identifying 35 differentially expressed genes (DEGs) between PC specimens and non-tumors. Based on 35 DEGs, we performed machine learning and identified eight diagnostic genes involved in PC progression. Then, we further screened three critical genes (CTSE, LAMC2 and SLC6A14) using three GEO datasets. A new diagnostic model was developed based on them and showed a strong predictive ability in screen PC specimens from non-tumor specimens in GEO, TCGA datasets and our cohorts. Then, clinical assays based on TCGA datasets indicated that the expression of LAMC2 and SLC6A14 was associated with advanced clinical stage and poor prognosis. The expressions of LAMC2 and SLC6A14, as well as the abundances of a variety of immune cells, exhibited a significant positive association with one another. Functionally, we confirmed that SLC6A14 was highly expressed in PC and its knockdown suppressed the proliferation, migration, invasion and EMT signal via regulating Wnt/β-catenin signaling pathway. Overall, our findings developed a novel diagnostic model for PC patients. SLC6A14 may promote PC progression via modulating Wnt/β-catenin signaling. This work offered a novel and encouraging new perspective that holds potential for further illuminating the clinicopathological relevance of PC as well as its molecular etiology.
Collapse
Affiliation(s)
- Cunshu Dang
- Department of Hepatobiliary Gastrointestinal Surgery, Tianjin Fourth Central Hospital, No.1 Zhongshan Road, Tianjin, China.
| | - Quan Bian
- Department of Plastic and Reconstructive Surgery, Tianjin Nankai Hospital, Tianjin, China
| | - Fengbiao Wang
- Department of Hepatobiliary Gastrointestinal Surgery, Tianjin Fourth Central Hospital, No.1 Zhongshan Road, Tianjin, China
| | - Han Wang
- Department of Otorhinolaryngology-Head and Neck Surgery, Tianjin Fourth Central Hospital, Tianjin, China
| | - Zhipeng Liang
- Department of Hepatobiliary Gastrointestinal Surgery, Tianjin Fourth Central Hospital, No.1 Zhongshan Road, Tianjin, China
| |
Collapse
|
29
|
Sharath NS, Misra R, Ghosh J. Application of hydrogel-based drug delivery system for pancreatic cancer. RECENT ADVANCES IN NANOCARRIERS FOR PANCREATIC CANCER THERAPY 2024:73-93. [DOI: 10.1016/b978-0-443-19142-8.00011-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
30
|
Tharrun Daniel Paul L, Munuswamy-Ramanujam G, Kumar RCS, Ramachandran V, Gnanasampanthapandian D, Palaniyandi K. Recent advancement in molecular markers of pancreatic cancer. BIOMARKERS IN CANCER DETECTION AND MONITORING OF THERAPEUTICS 2024:121-149. [DOI: 10.1016/b978-0-323-95114-2.00025-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
31
|
Berbís MÁ, Godino FP, Rodríguez-Comas J, Nava E, García-Figueiras R, Baleato-González S, Luna A. Radiomics in CT and MR imaging of the liver and pancreas: tools with potential for clinical application. Abdom Radiol (NY) 2024; 49:322-340. [PMID: 37889265 DOI: 10.1007/s00261-023-04071-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 09/15/2023] [Accepted: 09/19/2023] [Indexed: 10/28/2023]
Abstract
Radiomics allows the extraction of quantitative imaging features from clinical magnetic resonance imaging (MRI) and computerized tomography (CT) studies. The advantages of radiomics have primarily been exploited in oncological applications, including better characterization and staging of oncological lesions and prediction of patient outcomes and treatment response. The potential introduction of radiomics in the clinical setting requires the establishment of a standardized radiomics pipeline and a quality assurance program. Radiomics and texture analysis of the liver have improved the differentiation of hypervascular lesions such as adenomas, focal nodular hyperplasia, and hepatocellular carcinoma (HCC) during the arterial phase, and in the pretreatment determination of HCC prognostic factors (e.g., tumor grade, microvascular invasion, Ki-67 proliferation index). Radiomics of pancreatic CT and MR images has enhanced pancreatic ductal adenocarcinoma detection and its differentiation from pancreatic neuroendocrine tumors, mass-forming chronic pancreatitis, or autoimmune pancreatitis. Radiomics can further help to better characterize incidental pancreatic cystic lesions, accurately discriminating benign from malignant intrapancreatic mucinous neoplasms. Nonetheless, despite their encouraging results and exciting potential, these tools have yet to be implemented in the clinical setting. This non-systematic review will describe the essential steps in the implementation of the radiomics and feature extraction workflow from liver and pancreas CT and MRI studies for their potential clinical application. A succinct overview of reported radiomics applications in the liver and pancreas and the challenges and limitations of their implementation in the clinical setting is also discussed, concluding with a brief exploration of the future perspectives of radiomics in the gastroenterology field.
Collapse
Affiliation(s)
- M Álvaro Berbís
- Department of Radiology, HT Médica, San Juan de Dios Hospital, 14960, Córdoba, Spain.
- Department of Radiology, HT Médica, San Juan de Dios Hospital, Av. del Brillante, 106, 14012, Córdoba, Spain.
| | | | | | - Enrique Nava
- Department of Communications Engineering, University of Málaga, 29016, Málaga, Spain
| | - Roberto García-Figueiras
- Abdominal Imaging Section, University Clinical Hospital of Santiago, 15706, Santiago de Compostela, A Coruña, Spain
| | - Sandra Baleato-González
- Abdominal Imaging Section, University Clinical Hospital of Santiago, 15706, Santiago de Compostela, A Coruña, Spain
| | - Antonio Luna
- Department of Radiology, HT Médica, Clínica las Nieves, 23007, Jaén, Spain
| |
Collapse
|
32
|
Zhang Z, Tian H, Xu Z, Bian Y, Wu J. Application of a pyramid pooling Unet model with integrated attention mechanism and Inception module in pancreatic tumor segmentation. J Appl Clin Med Phys 2023; 24:e14204. [PMID: 37937804 PMCID: PMC10691628 DOI: 10.1002/acm2.14204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 09/26/2023] [Accepted: 10/28/2023] [Indexed: 11/09/2023] Open
Abstract
BACKGROUND The segmentation and recognition of pancreatic tumors are crucial tasks in the diagnosis and treatment of pancreatic diseases. However, due to the relatively small proportion of the pancreas in the abdomen and significant shape and size variations, pancreatic tumor segmentation poses considerable challenges. PURPOSE To construct a network model that combines a pyramid pooling module with Inception architecture and SE attention mechanism (PIS-Unet), and observe its effectiveness in pancreatic tumor images segmentation, thereby providing supportive recommendations for clinical practitioners. MATERIALS AND METHODS A total of 303 patients with histologically confirmed pancreatic cystic neoplasm (PCN), including serous cystic neoplasm (SCN) and mucinous cystic neoplasm (MCN), from Shanghai Changhai Hospital between March 2011 and November 2021 were included. A total of 1792 T2-weighted imaging (T2WI) slices were used to build a CNN model. The model employed a pyramid pooling Inception module with a fused attention mechanism. The attention mechanism enhanced the network's focus on local features, while the Inception module and pyramid pooling allowed the network to extract features at different scales and improve the utilization efficiency of global information, thereby effectively enhancing network performance. RESULTS Using three-fold cross-validation, the model constructed by us achieved a dice score of 85.49 ± 2.02 for SCN images segmentation, and a dice score of 87.90 ± 4.19 for MCN images segmentation. CONCLUSION This study demonstrates that using deep learning networks for the segmentation of PCNs yields favorable results. Applying this network as an aid to physicians in PCN diagnosis shows potential for clinical applications.
Collapse
Affiliation(s)
- Zhiwei Zhang
- School of Health Science and EngineeringUniversity of Shanghai for Science and TechnologyShanghaiChina
| | - Hui Tian
- School of Health Science and EngineeringUniversity of Shanghai for Science and TechnologyShanghaiChina
| | - Zhenshun Xu
- School of Health Science and EngineeringUniversity of Shanghai for Science and TechnologyShanghaiChina
| | - Yun Bian
- Department of RadiologyChanghai HospitalThe Navy Military Medical UniversityShanghaiChina
| | - Jie Wu
- School of Health Science and EngineeringUniversity of Shanghai for Science and TechnologyShanghaiChina
| |
Collapse
|
33
|
Vahabi M, Comandatore A, Centra C, Blandino G, Morelli L, Giovannetti E. Thinking small to win big? A critical review on the potential application of extracellular vesicles for biomarker discovery and new therapeutic approaches in pancreatic cancer. Semin Cancer Biol 2023; 97:50-67. [PMID: 37956937 DOI: 10.1016/j.semcancer.2023.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 09/29/2023] [Accepted: 11/07/2023] [Indexed: 11/20/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an extremely deadly form of cancer, with limited progress in 5-year survival rates despite significant research efforts. The main challenges in treating PDAC include difficulties in early detection, and resistance to current therapeutic approaches due to aggressive molecular and microenvironment features. These challenges emphasize the importance of identifying clinically validated biomarkers for early detection and clinical management. Extracellular vesicles (EVs), particularly exosomes, have emerged as crucial mediators of intercellular communication by transporting molecular cargo. Recent research has unveiled their role in initiation, metastasis, and chemoresistance of PDAC. Consequently, utilizing EVs in liquid biopsies holds promise for the identification of biomarkers for early detection, prognosis, and monitoring of drug efficacy. However, numerous limitations, including challenges in isolation and characterization of homogeneous EVs populations, as well as the absence of standardized protocols, can affect the reliability of studies involving EVs as biomarkers, underscoring the necessity for a prudent approach. EVs have also garnered considerable attention as a promising drug delivery system and novel therapy for tumors. The loading of biomolecules or chemical drugs into exosomes and their subsequent delivery to target cells can effectively impede tumor progression. Nevertheless, there are obstacles that must be overcome to ensure the accuracy and efficacy of therapies relying on EVs for the treatment of tumors. In this review, we examine both recent advancements and remaining obstacles, exploring the potential of utilizing EVs in biomarker discovery as well as for the development of drug delivery vehicles.
Collapse
Affiliation(s)
- Mahrou Vahabi
- Department of Medical Oncology, Amsterdam UMC, VU University, Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Annalisa Comandatore
- Department of Medical Oncology, Amsterdam UMC, VU University, Cancer Center Amsterdam, Amsterdam, Netherlands; General Surgery Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Chiara Centra
- Department of Medical Oncology, Amsterdam UMC, VU University, Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Giovanni Blandino
- IRCCS Regina Elena National Cancer Institute, Oncogenomic and Epigenetic Laboratory, Rome, Italy
| | - Luca Morelli
- General Surgery Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Elisa Giovannetti
- Department of Medical Oncology, Amsterdam UMC, VU University, Cancer Center Amsterdam, Amsterdam, Netherlands; Fondazione Pisana per la Scienza, Pisa, Italy.
| |
Collapse
|
34
|
Duncan ZN, Summerlin D, West JT, Packard AT, Morgan DE, Galgano SJ. PET/MRI for evaluation of patients with pancreatic cancer. Abdom Radiol (NY) 2023; 48:3601-3609. [PMID: 37191756 DOI: 10.1007/s00261-023-03943-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 04/25/2023] [Accepted: 04/26/2023] [Indexed: 05/17/2023]
Abstract
Pancreatic cancers are the third leading cause of cancer-related death in the USA and outcomes remain poor despite improvements in imaging and treatment paradigms. Currently, computed tomography (CT) and magnetic resonance imaging (MRI) are frequently utilized for staging and restaging of these malignancies, but positron emission tomography (PET)/CT can play a role in troubleshooting and improve whole-body staging. PET/MRI is a novel imaging modality that allows for simultaneous acquisition of PET and MRI images, leading to improved image quality and potential increased sensitivity. Early studies suggest that PET/MRI may play a larger role in pancreatic cancer imaging in future. This manuscript will briefly discuss current imaging approaches to pancreatic cancer and outline existing evidence and published data supporting the use of PET/MRI for pancreatic cancers.
Collapse
Affiliation(s)
- Zoey N Duncan
- University of Alabama at Birmingham School of Medicine, Birmingham, AL, USA
| | - David Summerlin
- Department of Radiology, University of Alabama at Birmingham Medical Center, Birmingham, AL, USA
| | - Janelle T West
- Department of Radiology, University of Alabama at Birmingham Medical Center, Birmingham, AL, USA
| | | | - Desiree E Morgan
- Department of Radiology, University of Alabama at Birmingham Medical Center, Birmingham, AL, USA
| | - Samuel J Galgano
- Department of Radiology, University of Alabama at Birmingham Medical Center, Birmingham, AL, USA.
| |
Collapse
|
35
|
Jackson ME, Stroud AJ, Somerset AE, Beal EW. Resection of a Pancreatic Ductal Adenocarcinoma With Pancreaticoduodenectomy in a Patient With a Prior Roux-en-Y Gastric Bypass and Retained Gastric Remnant. Am Surg 2023; 89:6400-6402. [PMID: 37767990 DOI: 10.1177/00031348231204903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/29/2023]
Affiliation(s)
| | - Alyssa J Stroud
- Department of Surgery, Wayne State University, Detroit, MI, USA
| | - Amy E Somerset
- Department of Surgery, Wayne State University, Detroit, MI, USA
| | - Eliza W Beal
- Department of Surgery, Wayne State University, Detroit, MI, USA
- Department of Oncology, Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
| |
Collapse
|
36
|
Wang S, Zhang Y, Xu Y, Yang P, Liu C, Gong H, Lei J. Progress in the application of dual-energy CT in pancreatic diseases. Eur J Radiol 2023; 168:111090. [PMID: 37742372 DOI: 10.1016/j.ejrad.2023.111090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 08/19/2023] [Accepted: 09/06/2023] [Indexed: 09/26/2023]
Abstract
Pancreatic diseases are difficult to diagnose due to their insidious onset and complex pathophysiological developmental characteristics. In recent years, dual-energy computed tomography (DECT) imaging technology has rapidly advanced. DECT can quantitatively extract and analyze medical imaging features and establish a correlation between these features and clinical results. This feature enables the adoption of more modern and accurate clinical diagnosis and treatment strategies for patients with pancreatic diseases so as to achieve the goal of non-invasive, low-cost, and personalized treatment. The purpose of this review is to elaborate on the application of DECT for the diagnosis, biological characterization, and prediction of the survival of patients with pancreatic diseases (including pancreatitis, pancreatic cancer, pancreatic cystic tumor, pancreatic neuroendocrine tumor, and pancreatic injury) and to summarize its current limitations and future research prospects.
Collapse
Affiliation(s)
- Sha Wang
- The First Clinical Medical College of Lanzhou University, Lanzhou 730000, China
| | - Yanli Zhang
- The First Clinical Medical College of Lanzhou University, Lanzhou 730000, China; Department of Radiology, The First Hospital of Lanzhou University, Lanzhou 730000, China; Radiological Clinical Medicine Research Center of Gansu Province, Lanzhou 730000, China
| | - Yongsheng Xu
- The First Clinical Medical College of Lanzhou University, Lanzhou 730000, China; Department of Radiology, The First Hospital of Lanzhou University, Lanzhou 730000, China; Radiological Clinical Medicine Research Center of Gansu Province, Lanzhou 730000, China
| | - Pengcheng Yang
- Department of Radiology, The First Hospital of Lanzhou University, Lanzhou 730000, China
| | - Chuncui Liu
- The First Clinical Medical College of Lanzhou University, Lanzhou 730000, China
| | - Hengxin Gong
- The First Clinical Medical College of Lanzhou University, Lanzhou 730000, China
| | - Junqiang Lei
- The First Clinical Medical College of Lanzhou University, Lanzhou 730000, China; Department of Radiology, The First Hospital of Lanzhou University, Lanzhou 730000, China; Radiological Clinical Medicine Research Center of Gansu Province, Lanzhou 730000, China.
| |
Collapse
|
37
|
Kim MJ, Kim HS, Kang HW, Lee DE, Hong WC, Kim JH, Kim M, Cheong JH, Kim HJ, Park JS. SLC38A5 Modulates Ferroptosis to Overcome Gemcitabine Resistance in Pancreatic Cancer. Cells 2023; 12:2509. [PMID: 37887353 PMCID: PMC10605569 DOI: 10.3390/cells12202509] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 10/20/2023] [Accepted: 10/22/2023] [Indexed: 10/28/2023] Open
Abstract
Pancreatic cancer is characterized by a poor prognosis, with its five-year survival rate lower than that of any other cancer type. Gemcitabine, a standard treatment for pancreatic cancer, often has poor outcomes for patients as a result of chemoresistance. Therefore, novel therapeutic targets must be identified to overcome gemcitabine resistance. Here, we found that SLC38A5, a glutamine transporter, is more highly overexpressed in gemcitabine-resistant patients than in gemcitabine-sensitive patients. Furthermore, the deletion of SLC38A5 decreased the proliferation and migration of gemcitabine-resistant PDAC cells. We also found that the inhibition of SLC38A5 triggered the ferroptosis signaling pathway via RNA sequencing. Also, silencing SLC38A5 induced mitochondrial dysfunction and reduced glutamine uptake and glutathione (GSH) levels, and downregulated the expressions of GSH-related genes NRF2 and GPX4. The blockade of glutamine uptake negatively modulated the mTOR-SREBP1-SCD1 signaling pathway. Therefore, suppression of SLC38A5 triggers ferroptosis via two pathways that regulate lipid ROS levels. Similarly, we observed that knockdown of SLC38A5 restored gemcitabine sensitivity by hindering tumor growth and metastasis in the orthotopic mouse model. Altogether, our results demonstrate that SLC38A5 could be a novel target to overcome gemcitabine resistance in PDAC therapy.
Collapse
Affiliation(s)
- Myeong Jin Kim
- Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06229, Republic of Korea; (M.J.K.); (H.S.K.); (H.W.K.); (D.E.L.); (W.C.H.); (J.H.K.); (M.K.)
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University, Seoul 03722, Republic of Korea;
| | - Hyung Sun Kim
- Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06229, Republic of Korea; (M.J.K.); (H.S.K.); (H.W.K.); (D.E.L.); (W.C.H.); (J.H.K.); (M.K.)
| | - Hyeon Woong Kang
- Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06229, Republic of Korea; (M.J.K.); (H.S.K.); (H.W.K.); (D.E.L.); (W.C.H.); (J.H.K.); (M.K.)
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University, Seoul 03722, Republic of Korea;
| | - Da Eun Lee
- Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06229, Republic of Korea; (M.J.K.); (H.S.K.); (H.W.K.); (D.E.L.); (W.C.H.); (J.H.K.); (M.K.)
| | - Woosol Chris Hong
- Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06229, Republic of Korea; (M.J.K.); (H.S.K.); (H.W.K.); (D.E.L.); (W.C.H.); (J.H.K.); (M.K.)
- Yonsei University College of Medicine, Seoul 06229, Republic of Korea
| | - Ju Hyun Kim
- Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06229, Republic of Korea; (M.J.K.); (H.S.K.); (H.W.K.); (D.E.L.); (W.C.H.); (J.H.K.); (M.K.)
- Yonsei University College of Medicine, Seoul 06229, Republic of Korea
| | - Minsoo Kim
- Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06229, Republic of Korea; (M.J.K.); (H.S.K.); (H.W.K.); (D.E.L.); (W.C.H.); (J.H.K.); (M.K.)
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University, Seoul 03722, Republic of Korea;
| | - Jae-Ho Cheong
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University, Seoul 03722, Republic of Korea;
- Department of Surgery, Yonsei University College of Medicine, Seoul 06229, Republic of Korea
| | - Hyo Jung Kim
- Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06229, Republic of Korea; (M.J.K.); (H.S.K.); (H.W.K.); (D.E.L.); (W.C.H.); (J.H.K.); (M.K.)
| | - Joon Seong Park
- Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06229, Republic of Korea; (M.J.K.); (H.S.K.); (H.W.K.); (D.E.L.); (W.C.H.); (J.H.K.); (M.K.)
| |
Collapse
|
38
|
Sansaloni-Pastor S, Lange N. Unleashing the potential of 5-Aminolevulinic acid: Unveiling a promising target for cancer diagnosis and treatment beyond photodynamic therapy. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY. B, BIOLOGY 2023; 247:112771. [PMID: 37647818 DOI: 10.1016/j.jphotobiol.2023.112771] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 08/09/2023] [Accepted: 08/21/2023] [Indexed: 09/01/2023]
Abstract
The therapeutic properties of 5-aminolevulinic acid (5-ALA) have been extensively studied for cancer detection and treatment using photodynamic therapy (PDT). When administered externally, 5-ALA is converted to protoporphyrin IX (PpIX) in cancer cells, which generates reactive oxygen species (ROS) upon exposure to light. This process enables targeted cell death induction and cancer detection. Given the highly conserved nature of heme biosynthesis over billions of years, we hypothesized that natural mechanisms might exist to prevent excessive accumulation of PpIX or heme resulting from 5-ALA overload. Therefore, we anticipated alterations in protein expression profiles upon exogenous administration of 5-ALA. To understand cellular responses to 5-ALA, we investigated protein expression changes and identified OR1B1 as a promising target in bladder, prostate, lung, and cervical cancer cells. OR1B1 expression was observed only with 5-ALA and ferrous chloride, highlighting the central role of heme in this discovery. Immunofluorescence and electron microscopy confirmed OR1B1's sub-cellular localization. These findings suggest that 5-ALA transformation in cancer cells and OR1B1 expression have potential for enhancing cancer detection and developing alternative treatments, including immunotherapy. This approach overcomes the limitations of PDT and opens new avenues for effective and targeted cancer interventions.
Collapse
Affiliation(s)
- Sara Sansaloni-Pastor
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Rue Michel-Servet 1, 1211 Geneva, Switzerland; School of Pharmaceutical Sciences, University of Geneva, Rue Michel-Servet 1, 1211 Geneva, Switzerland
| | - Norbert Lange
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Rue Michel-Servet 1, 1211 Geneva, Switzerland; School of Pharmaceutical Sciences, University of Geneva, Rue Michel-Servet 1, 1211 Geneva, Switzerland.
| |
Collapse
|
39
|
Takahashi K, Takeda Y, Ono Y, Isomoto H, Mizukami Y. Current status of molecular diagnostic approaches using liquid biopsy. J Gastroenterol 2023; 58:834-847. [PMID: 37470859 PMCID: PMC10423147 DOI: 10.1007/s00535-023-02024-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 07/08/2023] [Indexed: 07/21/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive and lethal cancers, and developing an efficient and reliable approach for its early-stage diagnosis is urgently needed. Precancerous lesions of PDAC, such as pancreatic intraepithelial neoplasia (PanIN) and intraductal papillary mucinous neoplasms (IPMN), arise through multiple steps of driver gene alterations in KRAS, TP53, CDKN2A, SMAD4, or GNAS. Hallmark mutations play a role in tumor initiation and progression, and their detection in bodily fluids is crucial for diagnosis. Recently, liquid biopsy has gained attention as an approach to complement pathological diagnosis, and in addition to mutation signatures in cell-free DNA, cell-free RNA, and extracellular vesicles have been investigated as potential diagnostic and prognostic markers. Integrating such molecular information to revise the diagnostic criteria for pancreatic cancer can enable a better understanding of the pathogenesis underlying inter-patient heterogeneity, such as sensitivity to chemotherapy and disease outcomes. This review discusses the current diagnostic approaches and clinical applications of genetic analysis in pancreatic cancer and diagnostic attempts by liquid biopsy and molecular analyses using pancreatic juice, duodenal fluid, and blood samples. Emerging knowledge in the rapidly advancing liquid biopsy field is promising for molecular profiling and diagnosing pancreatic diseases with significant diversity.
Collapse
Affiliation(s)
- Kenji Takahashi
- Division of Metabolism and Biosystemic Science, Gastroenterology, and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, 2-1 Midorigaoka Higashi, Asahikawa, Hokkaido, 078-8510, Japan.
| | - Yohei Takeda
- Division of Medicine and Clinical Science, Department of Multidisciplinary Internal Medicine, Faculty of Medicine, Tottori University, Tottori, Japan
| | - Yusuke Ono
- Division of Metabolism and Biosystemic Science, Gastroenterology, and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, 2-1 Midorigaoka Higashi, Asahikawa, Hokkaido, 078-8510, Japan
- Institute of Biomedical Research, Sapporo Higashi Tokushukai Hospital, Sapporo, Japan
| | - Hajime Isomoto
- Division of Medicine and Clinical Science, Department of Multidisciplinary Internal Medicine, Faculty of Medicine, Tottori University, Tottori, Japan
| | - Yusuke Mizukami
- Division of Metabolism and Biosystemic Science, Gastroenterology, and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, 2-1 Midorigaoka Higashi, Asahikawa, Hokkaido, 078-8510, Japan
- Institute of Biomedical Research, Sapporo Higashi Tokushukai Hospital, Sapporo, Japan
| |
Collapse
|
40
|
Chen L, Fu H, He H, Lou K, Li Q, Ye J, Feng G, Yu C. Automated Synthesis and Preclinical Evaluation of Optimized Integrin α6-Targeted Positron Emission Tomography Imaging of Pancreatic Cancer. Mol Pharm 2023; 20:4277-4284. [PMID: 37463487 DOI: 10.1021/acs.molpharmaceut.3c00321] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/20/2023]
Abstract
Integrin α6 has been considered a promising biomarker, is overexpressed in many tumors, and plays a vital role in tumor formation, recurrence, and metastasis. In this study, we identified a novel high-affinity integrin α6-targeted peptide named RD2 (Arg-Trp-Tyr-Asp-PEG4)2-Lys-Lys and developed a 18F-radiolabeled peptide tracer ([18F]-AlF-NOTA-RD2) and evaluated its potential application in positron emission tomography (PET) imaging of pancreatic cancer. [18F]-AlF-NOTA-RD2 was produced using GMP (Good Manufacturing Practice of Medical Products)-compliant automatic radiosynthesis on a single GE FASTLab2 cassette-type synthesis module. The stability of [18F]-AlF-NOTA-RD2 was analyzed in phosphate-buffered saline (PBS) and fetal bovine serum (FBS). The cell uptake assay of the tracer was assessed using PANC-1 cells. In addition, small-animal PET imaging and biodistribution studies of [18F]-AlF-NOTA-RD2 were performed in pancreatic cancer subcutaneous tumor-bearing mice. The PET tracer [18F]-AlF-NOTA-RD2 was obtained with a radiochemical yield of 23.7 ± 4.7%, radiochemical purity of >99%, and molar activity of 165.7 ± 59.1 GBq/μmol. [18F]-AlF-NOTA-RD2 exhibited good in vitro stability in PBS and FBS. LogP octanol water value for the tracer was -2.28 ± 0.05 (n = 3). The binding affinity of RD2 to the integrin α6 protein (Kd = 0.13 ± 3.65 μM, n = 3) was significantly higher than that of the RWY (CRWYDENAC) (Kd = 6.97 ± 1.44 μM, n = 3). Small-animal PET imaging and biodistribution also revealed that [18F]-AlF-NOTA-RD2 displayed rapid and good tumor uptake and lower liver background uptake in PANC-1 tumor-bearing mice. [18F]-AlF-NOTA-RD2 showed significant radioactivity accumulation in tumors and was successfully blocked by NOTA-RD2. Compared with [18F]-FDG, [18F]-AlF-NOTA-RD2 PET imaging and biodistribution studies in PANC-1 xenograft tumor-bearing mice confirmed a good tumor-to-muscle ratio (8.69 ± 2.03 vs 1.41 ± 0.23, respectively) at 0.5 h and (2.99 ± 3.02 vs 1.43 ± 0.17, respectively) at 1 h post injection. Autoradiography of human pancreatic cancer tumor tissues further confirmed high accumulation of [18F]-AlF-NOTA-RD2. In summary, we developed an optimized integrin α6-targeted imaging tracer and obtained high radioactivity products with a cassette-type synthesis module; moreover, the tracer exhibited good binding affinity with integrin α6 and good target specificity for PANC-1 cells in xenograft pancreatic tumor-bearing mice, demonstrating its promising application as a noninvasive PET radiotracer of integrin α6 expression in pancreatic cancer.
Collapse
Affiliation(s)
- Liping Chen
- Department of Nuclear Medicine, Affiliated Hospital of Jiangnan University, 1000 Hefeng Road, Binhu District, Wuxi 214062, P.R. China
| | - Haitian Fu
- Department of Nuclear Medicine, Affiliated Hospital of Jiangnan University, 1000 Hefeng Road, Binhu District, Wuxi 214062, P.R. China
| | - Huihui He
- Department of Nuclear Medicine, Affiliated Hospital of Jiangnan University, 1000 Hefeng Road, Binhu District, Wuxi 214062, P.R. China
| | - Kequan Lou
- Department of Nuclear Medicine, Affiliated Hospital of Jiangnan University, 1000 Hefeng Road, Binhu District, Wuxi 214062, P.R. China
| | - Qingbo Li
- Department of Nuclear Medicine, Affiliated Hospital of Jiangnan University, 1000 Hefeng Road, Binhu District, Wuxi 214062, P.R. China
| | - Jiacong Ye
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou 510060, P.R. China
| | - Guokai Feng
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou 510060, P.R. China
| | - Chunjing Yu
- Department of Nuclear Medicine, Affiliated Hospital of Jiangnan University, 1000 Hefeng Road, Binhu District, Wuxi 214062, P.R. China
| |
Collapse
|
41
|
Kim SY, Jo MJ, Yoon MS, Jin CE, Shin YB, Lee JM, Shin HJ, Oh JG, Cho JM, Kim H, Park H, Choi YW, Park CW, Kim JS, Shin DH. Gemcitabine and rapamycin-loaded mixed polymeric thermogel for metastatic pancreatic cancer therapy. J Control Release 2023; 360:796-809. [PMID: 37437850 DOI: 10.1016/j.jconrel.2023.07.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 06/12/2023] [Accepted: 07/07/2023] [Indexed: 07/14/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the 4th leading cause of cancer-related death and has a poor 5-year overall survival. The superior therapeutic benefits of combination or co-administration of drugs as intraperitoneal chemotherapy have increased interest in developing strategies to deliver chemotherapeutic agents to patients safely. In this study, we prepared a gel comprising the thermosensitive poly(lactide-co-glycolide)-b-poly(ethylene glycol)-b-poly(lactide-co-glycolide) (PLGA-PEG-PLGA) polymer and gemcitabine (GEM), which is currently used as the primary chemotherapy for PDAC and rapamycin (RAPA), a mammalian TOR (mTOR) inhibitor, to deliver the drug through intraperitoneal injection. We performed in vitro cytotoxicity experiments to verify the synergistic effects of the two drugs at different molar ratios and characterized the physicochemical properties of the GEM, RAPA, and GEM/RAPA-loaded thermosensitive PLGA-PEG-PLGA gels, hereafter referred to as (g(G), g(R), and g(GR)), respectively. The g(GR) comprising PLGA-PEG-PLGA polymer (25% w/v) and GEM and RAPA at a molar ratio of 11:1 showed synergism and was optimized. An in vitro cytotoxicity assay was performed by treating Panc-1-luc2 tumor spheroids with g(G), g(R), or g(GR). The g(GR) treatment group showed a 2.75-fold higher inhibition rate than the non-treated (NT) and vehicle-treated groups. Furthermore, in vivo drug release assay in mice by intraperitoneal injection of g(G), g(R), or g(GR) showed a more rapid release rate of GEM than RAPA, similar to the in vitro release pattern. The drugs in the gel were released faster in vivo than in vitro and degraded in 48 h. In addition, g(GR) showed the highest anti-tumor efficacy with no toxicity to mice. These results provide evidence for the safety and efficacy of g(GR) for intraperitoneal drug delivery. This study will assist in developing and clinically administering topical anti-cancer formulations.
Collapse
Affiliation(s)
- Seo Yeon Kim
- College of Pharmacy, Chungbuk National University, Cheongju 28160, Republic of Korea
| | - Min Jeong Jo
- College of Pharmacy, Chungbuk National University, Cheongju 28160, Republic of Korea
| | - Moon Sup Yoon
- College of Pharmacy, Chungbuk National University, Cheongju 28160, Republic of Korea
| | - Chae Eun Jin
- College of Pharmacy, Chungbuk National University, Cheongju 28160, Republic of Korea
| | - Yu Been Shin
- College of Pharmacy, Chungbuk National University, Cheongju 28160, Republic of Korea
| | - Jae Min Lee
- College of Pharmacy, Chungbuk National University, Cheongju 28160, Republic of Korea
| | - Hee Ji Shin
- College of Pharmacy, Chungbuk National University, Cheongju 28160, Republic of Korea
| | - Joon Gyo Oh
- R&D Center, Huons Co., Ltd., Ansan, 15588, Republic of Korea
| | - Jae Min Cho
- R&D Center, Huons Co., Ltd., Ansan, 15588, Republic of Korea
| | - Hyunjun Kim
- R&D Center, Huons Co., Ltd., Ansan, 15588, Republic of Korea
| | - Hyunjin Park
- R&D Center, Huons Co., Ltd., Ansan, 15588, Republic of Korea
| | - Yong-Won Choi
- R&D Center, Huons Co., Ltd., Ansan, 15588, Republic of Korea
| | - Chun-Woong Park
- College of Pharmacy, Chungbuk National University, Cheongju 28160, Republic of Korea
| | - Jin-Seok Kim
- Drug Information Research Institute (DIRI), College of Pharmacy, Sookmyung Women's University, Seoul 04310, Republic of Korea
| | - Dae Hwan Shin
- College of Pharmacy, Chungbuk National University, Cheongju 28160, Republic of Korea.
| |
Collapse
|
42
|
Choi M, Park M, Lee SH, Lee MJ, Paik Y, Jang SI, Lee DK, Lee S, Kang CM. Development of a metabolite calculator for diagnosis of pancreatic cancer. Cancer Med 2023; 12:15933-15944. [PMID: 37350558 PMCID: PMC10469663 DOI: 10.1002/cam4.6233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 04/22/2023] [Accepted: 06/01/2023] [Indexed: 06/24/2023] Open
Abstract
BACKGROUND Carbohydrate antigen (CA) 19-9 is a known pancreatic cancer (PC) biomarker, but is not commonly used for general screening due to its low sensitivity and specificity. This study aimed to develop a serum metabolites-based diagnostic calculator for detecting PC with high accuracy. METHODS A targeted quantitative approach of direct flow injection-tandem mass spectrometry combined with liquid chromatography-tandem mass spectrometry was employed for metabolomic analysis of serum samples using an Absolute IDQ™ p180 kit. Integrated metabolomic analysis was performed on 241 pooled or individual serum samples collected from healthy donors and patients from nine disease groups, including chronic pancreatitis, PC, other cancers, and benign diseases. Orthogonal partial least squares discriminant analysis (OPLS-DA) based on characteristics of 116 serum metabolites distinguished patients with PC from those with other diseases. Sparse partial least squares discriminant analysis (SPLS-DA) was also performed, incorporating simultaneous dimension reduction and variable selection. Predictive performance between discrimination models was compared using a 2-by-2 contingency table of predicted probabilities obtained from the models and actual diagnoses. RESULTS Predictive values obtained through OPLS-DA for accuracy, sensitivity, specificity, balanced accuracy, and area under the receiver operating characteristic curve (AUC) were 0.9825, 0.9916, 0.9870, 0.9866, and 0.9870, respectively. The number of metabolite candidates was narrowed to 76 for SPLS-DA. The SPLS-DA-obtained predictive values for accuracy, sensitivity, specificity, balanced accuracy, and AUC were 0.9773, 0.9649, 0.9832, 0.9741, and 0.9741, respectively. CONCLUSIONS We successfully developed a 76 metabolome-based diagnostic panel for detecting PC that demonstrated high diagnostic performance in differentiating PC from other diseases.
Collapse
Affiliation(s)
- Munseok Choi
- Department of Surgery, Yongin Severance HospitalYonsei University College of MedicineYongin‐siSouth Korea
| | - Minsu Park
- Department of Information and StatisticsChungnam National UniversityDaejeonSouth Korea
| | - Sung Hwan Lee
- Department of Surgery, CHA Bundang Medical CenterCHA UniversitySouth Korea
| | - Min Jung Lee
- Yonsei Proteome Research Center and Department of Integrated OMICS for Biomedical Science and Department of Biochemistry, College of Life Science and BiotechnologyYonsei UniversitySeoulSouth Korea
| | - Young‐Ki Paik
- Yonsei Proteome Research Center and Department of Integrated OMICS for Biomedical Science and Department of Biochemistry, College of Life Science and BiotechnologyYonsei UniversitySeoulSouth Korea
| | - Sung Il Jang
- Department of Internal Medicine, Gangnam Severance HospitalYonsei University College of MedicineSeoulSouth Korea
| | - Dong Ki Lee
- Department of Internal Medicine, Gangnam Severance HospitalYonsei University College of MedicineSeoulSouth Korea
| | - Sang‐Guk Lee
- Department of Laboratory Medicine, Severance HospitalYonsei University College of MedicineSeoulSouth Korea
| | - Chang Moo Kang
- Department of Surgery, Severance HospitalYonsei University College of MedicineSeoulSouth Korea
| |
Collapse
|
43
|
Singh M, Anvekar P, Baraskar B, Pallipamu N, Gadam S, Cherukuri ASS, Damani DN, Kulkarni K, Arunachalam SP. Prospective of Pancreatic Cancer Diagnosis Using Cardiac Sensing. J Imaging 2023; 9:149. [PMID: 37623681 PMCID: PMC10455647 DOI: 10.3390/jimaging9080149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 07/14/2023] [Accepted: 07/17/2023] [Indexed: 08/26/2023] Open
Abstract
Pancreatic carcinoma (Ca Pancreas) is the third leading cause of cancer-related deaths in the world. The malignancies of the pancreas can be diagnosed with the help of various imaging modalities. An endoscopic ultrasound with a tissue biopsy is so far considered to be the gold standard in terms of the detection of Ca Pancreas, especially for lesions <2 mm. However, other methods, like computed tomography (CT), ultrasound, and magnetic resonance imaging (MRI), are also conventionally used. Moreover, newer techniques, like proteomics, radiomics, metabolomics, and artificial intelligence (AI), are slowly being introduced for diagnosing pancreatic cancer. Regardless, it is still a challenge to diagnose pancreatic carcinoma non-invasively at an early stage due to its delayed presentation. Similarly, this also makes it difficult to demonstrate an association between Ca Pancreas and other vital organs of the body, such as the heart. A number of studies have proven a correlation between the heart and pancreatic cancer. The tumor of the pancreas affects the heart at the physiological, as well as the molecular, level. An overexpression of the SMAD4 gene; a disruption in biomolecules, such as IGF, MAPK, and ApoE; and increased CA19-9 markers are a few of the many factors that are noted to affect cardiovascular systems with pancreatic malignancies. A comprehensive review of this correlation will aid researchers in conducting studies to help establish a definite relation between the two organs and discover ways to use it for the early detection of Ca Pancreas.
Collapse
Affiliation(s)
- Mansunderbir Singh
- Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA; (M.S.); (B.B.); (N.P.)
| | - Priyanka Anvekar
- Department of Medicine, Division of Infectious Diseases, Mayo Clinic, Rochester, MN 55905, USA;
| | - Bhavana Baraskar
- Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA; (M.S.); (B.B.); (N.P.)
| | - Namratha Pallipamu
- Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA; (M.S.); (B.B.); (N.P.)
| | - Srikanth Gadam
- Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA; (M.S.); (B.B.); (N.P.)
| | - Akhila Sai Sree Cherukuri
- GIH Artificial Intelligence Laboratory (GAIL), Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA
- Microwave Engineering and Imaging Laboratory (MEIL), Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Devanshi N. Damani
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55905, USA;
- Department of Internal Medicine, Texas Tech University Health Science Center, El Paso, TX 79995, USA
| | - Kanchan Kulkarni
- Centre de Recherche Cardio-Thoracique de Bordeaux, University of Bordeaux, INSERM, U1045, 33000 Bordeaux, France;
- IHU Liryc, Heart Rhythm Disease Institute, Fondation Bordeaux Université, 33600 Bordeaux, France
| | - Shivaram P. Arunachalam
- Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA; (M.S.); (B.B.); (N.P.)
- GIH Artificial Intelligence Laboratory (GAIL), Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA
- Microwave Engineering and Imaging Laboratory (MEIL), Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA
- Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
44
|
Weng H, Feng W, Li F, Huang D, Lin L, Wang Z. Transcription factor ETV1-induced lncRNA MAFG-AS1 promotes migration, invasion, and epithelial-mesenchymal transition of pancreatic cancer cells by recruiting IGF2BP2 to stabilize ETV1 expression. Growth Factors 2023:1-13. [PMID: 37428861 DOI: 10.1080/08977194.2023.2227272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 04/28/2023] [Indexed: 07/12/2023]
Abstract
We investigated the mechanism of ETS-translocation variant 1 (ETV1)/lncRNA-MAFG-AS1 in pancreatic cancer (PC). MAFG-AS1 and ETV1 levels in PC cell lines and HPNE cells were determined using reverse transcription quantitative polymerase chain reaction (RT-qPCR) and Western blotting (WB). After transfection with sh-MAFG-AS1, PC cell invasion, migration, proliferation, and epithelial-mesenchymal transition (EMT)-related proteins were measured by 5-ethynyl-2'-deoxyuridine (EdU), Transwell assay, and WB. The binding between ETV1 and MAFG-AS1 was studied using dual-luciferase assay and chromatin immunoprecipitation. The interactions between MAFG-AS1, IGF2BP2, and ETV1 were tested. Combined experiments were further performed using sh-MAFG-AS1 and pcDNA-ETV1 simultaneously. ETV1/MAFG-AS1 was highly expressed in PC cells. Blocking MAFG-AS1 inhibited the malignant behaviors of PC cells. ETV1 induced MAFG-AS1 transcription in PC cells. MAFG-AS1 stabilized ETV1 mRNA by recruiting IGF2BP2. ETV1 overexpression partially antagonized the suppression of silencing MAFG-AS1 on PC cells. ETV1-induced MAFG-AS1 stabilized the ETV1 expression by recruiting IGF2BP2 and promoted PC cell migration, invasion, proliferation, and EMT.
Collapse
Affiliation(s)
- Hanqin Weng
- Department of Hepatobiliary Surgery, Dongguan People's Hospital, Dongguan, China
| | - Weijian Feng
- Department of Hepatobiliary Surgery, Dongguan People's Hospital, Dongguan, China
| | - Fengling Li
- Department of Anesthesiology, Dongguan People's Hospital, Dongguan, China
| | - Dong Huang
- Department of Hepatobiliary Surgery, Dongguan People's Hospital, Dongguan, China
| | - Liangyi Lin
- Department of Hepatobiliary Surgery, Dongguan People's Hospital, Dongguan, China
| | - Zaiguo Wang
- Department of Hepatobiliary Surgery, Dongguan People's Hospital, Dongguan, China
| |
Collapse
|
45
|
Viriyasaranon T, Chun JW, Koh YH, Cho JH, Jung MK, Kim SH, Kim HJ, Lee WJ, Choi JH, Woo SM. Annotation-Efficient Deep Learning Model for Pancreatic Cancer Diagnosis and Classification Using CT Images: A Retrospective Diagnostic Study. Cancers (Basel) 2023; 15:3392. [PMID: 37444502 DOI: 10.3390/cancers15133392] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 06/26/2023] [Accepted: 06/26/2023] [Indexed: 07/15/2023] Open
Abstract
The aim of this study was to develop a novel deep learning (DL) model without requiring large-annotated training datasets for detecting pancreatic cancer (PC) using computed tomography (CT) images. This retrospective diagnostic study was conducted using CT images collected from 2004 and 2019 from 4287 patients diagnosed with PC. We proposed a self-supervised learning algorithm (pseudo-lesion segmentation (PS)) for PC classification, which was trained with and without PS and validated on randomly divided training and validation sets. We further performed cross-racial external validation using open-access CT images from 361 patients. For internal validation, the accuracy and sensitivity for PC classification were 94.3% (92.8-95.4%) and 92.5% (90.0-94.4%), and 95.7% (94.5-96.7%) and 99.3 (98.4-99.7%) for the convolutional neural network (CNN) and transformer-based DL models (both with PS), respectively. Implementing PS on a small-sized training dataset (randomly sampled 10%) increased accuracy by 20.5% and sensitivity by 37.0%. For external validation, the accuracy and sensitivity were 82.5% (78.3-86.1%) and 81.7% (77.3-85.4%) and 87.8% (84.0-90.8%) and 86.5% (82.3-89.8%) for the CNN and transformer-based DL models (both with PS), respectively. PS self-supervised learning can increase DL-based PC classification performance, reliability, and robustness of the model for unseen, and even small, datasets. The proposed DL model is potentially useful for PC diagnosis.
Collapse
Affiliation(s)
- Thanaporn Viriyasaranon
- Graduate Program in System Health Science and Engineering, Division of Mechanical and Biomedical Engineering, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Jung Won Chun
- Center for Liver and Pancreatobiliary Cancer, National Cancer Center, Goyang 10408, Republic of Korea
| | - Young Hwan Koh
- Center for Liver and Pancreatobiliary Cancer, National Cancer Center, Goyang 10408, Republic of Korea
| | - Jae Hee Cho
- Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Min Kyu Jung
- Department of Internal Medicine, Kyungpook National University Hospital, Daegu 41944, Republic of Korea
| | - Seong-Hun Kim
- Department of Internal Medicine, Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju 54907, Republic of Korea
| | - Hyo Jung Kim
- Department of Gastroenterology, Korea University Guro Hospital, Seoul 10408, Republic of Korea
| | - Woo Jin Lee
- Center for Liver and Pancreatobiliary Cancer, National Cancer Center, Goyang 10408, Republic of Korea
| | - Jang-Hwan Choi
- Graduate Program in System Health Science and Engineering, Division of Mechanical and Biomedical Engineering, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Sang Myung Woo
- Center for Liver and Pancreatobiliary Cancer, National Cancer Center, Goyang 10408, Republic of Korea
| |
Collapse
|
46
|
Zhao Z, Liu L, Chen H, Li S, Guo Y, Hou X, Yang J. Thymoquinone affects the gemcitabine sensitivity of pancreatic cancer by regulating collagen via hypoxia inducible factor-1α. Front Pharmacol 2023; 14:1138265. [PMID: 37324458 PMCID: PMC10264578 DOI: 10.3389/fphar.2023.1138265] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 05/23/2023] [Indexed: 06/17/2023] Open
Abstract
Objective: To clarify the potential therapeutic effects of thymoquinone (TQ) on pancreatic cancer and its gemcitabine (GEM) sensitivity. Methods: The expression levels of hypoxia inducible factor-1α (HIF-1α), collagens (COL1A1, COL3A1, and COL5A1), and transforming growth factor-β1 (TGFβ1) in pancreatic cancer and para-carcinoma tissues were compared using immunohistochemical methods, and their relationships with TNM staging were analyzed. The effects of TQ on apoptosis, migration, invasion, and GEM sensitivity of pancreatic cancer cells were assessed using in vitro and in vivo experiments. Western blot and immunohistochemistry were used to detect the expression levels of HIF-1α, extracellular matrix (ECM) production pathway-related proteins, and TGFβ/Smad signaling pathway-related proteins. Results: The expression levels of HIF-1α, COL1A1, COL3A1, COL5A1, and TGFβ1 in pancreatic cancer tissues were significantly higher than those in para-carcinoma tissues and correlated with TNM staging (p < 0.05). TQ and GEM administration inhibited the migration and invasion of the human pancreatic cancer cell line PANC-1 and promoted the apoptosis of PANC-1 cells. The combination of TQ and GEM was more effective than GEM alone. Western blot analysis showed that the expression levels of HIF-1α, ECM production pathway-related proteins, and TGFβ/Smad signaling pathway-related proteins were significantly decreased when TQ was used to treat PANC-1 cells (p < 0.05), and the expression levels of these proteins in the TQ + GEM group were significantly more decreased than those in the GEM group. Overexpression or knockdown of HIF-1α in PANC-1 cells showed the same effects as those induced by TQ administration. In vivo experiments showed that in PANC-1 tumor-bearing mice, tumor volume and tumor weight in mice treated with GEM and TQ were significantly lower than those in control or GEM-treated mice, whereas cell apoptosis was significantly increased (p < 0.05). Western blot and immunohistochemistry results showed that the levels of HIF-1α, ECM production pathway-related proteins, and TGFβ/Smad signaling pathway-related proteins in the GEM + TQ treatment group were further decreased compared to the control group or the GEM treatment group (p < 0.05). Conclusion: In pancreatic cancer cells, TQ can promote apoptosis, inhibit migration, invasion, and metastasis, and enhance the sensitivity to GEM. The underlying mechanism may involve the regulation of ECM production through the TGFβ/Smad pathway, in which HIF-1α plays a key role.
Collapse
Affiliation(s)
- Zhanxue Zhao
- Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China
- Department of General Surgery, Qinghai Provincial People’s Hospital, Xining, Qinghai, China
| | - Linxun Liu
- Department of General Surgery, Qinghai Provincial People’s Hospital, Xining, Qinghai, China
| | - Hekai Chen
- Department of General Surgery, Peking University BinHai Hospital, Tianjin, China
| | - Shuai Li
- Department of Clinical Pharmacy, Affiliated Hospital of Qinghai University, Xining, Qinghai, China
| | - Yan Guo
- Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China
- Department of Pathology, Qinghai Provincial People’s Hospital, Xining, Qinghai, China
| | - Xiaofan Hou
- Graduate School, Qinghai University, Xining, Qinghai, China
| | - Jinyu Yang
- Department of General Surgery, Qinghai Provincial People’s Hospital, Xining, Qinghai, China
| |
Collapse
|
47
|
Palma AM, Bushnell GG, Wicha MS, Gogna R. Tumor microenvironment interactions with cancer stem cells in pancreatic ductal adenocarcinoma. Adv Cancer Res 2023; 159:343-372. [PMID: 37268400 PMCID: PMC11218813 DOI: 10.1016/bs.acr.2023.02.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the most common type of pancreatic cancer in the United States. Additionally, the low survival rate makes PDAC the third-leading cause of cancer-related mortality in the United States, and it is projected that by 2030, it will become the second-leading cause of cancer mortality. Several biological factors contribute to PDAC aggressiveness, and their understanding will narrow the gap from biology to clinical care of PDAC, leading to earlier diagnoses and the development of better treatment options. In this review, we describe the origins of PDAC highlighting the role of cancer stem cells (CSC). CSC, also known as tumor initiating cells, which exhibit a unique metabolism that allows them to maintain a highly plastic, quiescent, immune- and therapy-evasive state. However, CSCs can exit quiescence during proliferation and differentiation, with the capacity to form tumors while constituting a small population in tumor tissues. Tumorigenesis depends on the interactions between CSCs and other cellular and non-cellular components in the microenvironment. These interactions are fundamental to support CSC stemness and are maintained throughout tumor development and metastasis. PDAC is characterized by a massive desmoplastic reaction, which result from the deposition of high amounts of extracellular matrix components by stromal cells. Here we review how this generates a favorable environment for tumor growth by protecting tumor cells from immune responses and chemotherapy and inducing tumor cell proliferation and migration, leading to metastasis formation ultimately leading to death. We emphasize the interactions between CSCs and the tumor microenvironment leading to metastasis formation and posit that better understanding and targeting of these interactions will improve patient outcomes.
Collapse
Affiliation(s)
| | - Grace G Bushnell
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States
| | - Max S Wicha
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States.
| | - Rajan Gogna
- VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States.
| |
Collapse
|
48
|
Chen Y, Lin X, Zou X, Qian Y, Liu Y, Wang R, Wang X, Yu X, Liu C, Cheng H. A novel immune checkpoint score system for prognostic evaluation in pancreatic adenocarcinoma. BMC Gastroenterol 2023; 23:113. [PMID: 37024802 PMCID: PMC10080823 DOI: 10.1186/s12876-023-02748-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 03/28/2023] [Indexed: 04/08/2023] Open
Abstract
BACKGROUND Pancreatic adenocarcinoma (PAAD) remains a lethal malignancy making the detection of novel prognostic biomarkers urgent. Limited studies have investigated the predictive capability of immune checkpoints in PAAD. METHOD Gene expression data and correlative clinical information of PAAD cohort were obtained from public databases, including TCGA, ICGC, GTEX and GEO databases. Risk factors were screened and used to establish a risk score model through LASSO and Cox regression analyses. The prognostic ability of the risk score model was demonstrated. The association between risk score with immune cells infiltration, immune checkpoint genes expression, immunogenic cell death, somatic mutations and signaling pathways enrichment were analysed. scRNA-seq data were collected to confirmed the immune checkpoints expression in PAAD samples. The prognosis prediction ability of OX40/TNFRSF4 was identified. The mRNA and protein expression of OX40 in our clinical specimens were examined by RT-PCR and IHC method and its prognosis ability was verified. RESULTS First of all, the difference of immune microenvironment between pancreatic cancer and adjacent tissues was shown. A risk score system based on three immune checkpoints (OX40, TNFSF14 and KIR3DL1) was established. The risk score model was an independent prognostic factor and performed well regarding overall survival (OS) predictions among PAAD patients. A nomogram was established to facilitate the risk model application in clinical prognosis. Immune cells including naive B cells, CD8+ T cells and Tregs were negatively correlated with the risk score. The risk score was associated with expression of immune checkpoint genes, immunogenic cell death related genes and somatic mutations. Glycolysis processes, IL-2-STAT5, IL-6-STAT3, and mTORC1 signaling pathways were enriched in the high-risk score group. Furthermore, scRNA-seq data confirmed that TNFRSF4, TNFSF14 and KIR3DL1 were expressed on immune cells in PAAD samples. We then identified OX40 as an independent prognosis-related gene, and a higher OX40 expression was associated with increased survival rate and immune environment change. In 84 PAAD clinical specimens collected from our center, we confirmed that higher OX40 mRNA expression levels were related to a good prognosis. The protein expression of OX40 on tumor-infiltrating immune cells (TIICs), endothelial cells and tumor cells was verified in PAAD tissues by immunohistochemistry (IHC) method. CONCLUSIONS Overall, our findings strongly suggested that the three-immune checkpoints score system might be useful in the prognosis and design of personalized treatments for PAAD patients. Finally, we identified OX40 as an independent potential biomarker for PAAD prognosis prediction.
Collapse
Affiliation(s)
- Yusheng Chen
- Department of Pancreatic Surgery, Shanghai Cancer Center, Fudan University, 270 DongAn Road, Xuhui, Shanghai, 200032, People's Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Xuan Lin
- Department of Pancreatic Surgery, Shanghai Cancer Center, Fudan University, 270 DongAn Road, Xuhui, Shanghai, 200032, People's Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Xuan Zou
- Department of Pancreatic Surgery, Shanghai Cancer Center, Fudan University, 270 DongAn Road, Xuhui, Shanghai, 200032, People's Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Yunzhen Qian
- Department of Pancreatic Surgery, Shanghai Cancer Center, Fudan University, 270 DongAn Road, Xuhui, Shanghai, 200032, People's Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Yu Liu
- Department of Pancreatic Surgery, Shanghai Cancer Center, Fudan University, 270 DongAn Road, Xuhui, Shanghai, 200032, People's Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Ruijie Wang
- Department of Pancreatic Surgery, Shanghai Cancer Center, Fudan University, 270 DongAn Road, Xuhui, Shanghai, 200032, People's Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Xu Wang
- Department of Pancreatic Surgery, Shanghai Cancer Center, Fudan University, 270 DongAn Road, Xuhui, Shanghai, 200032, People's Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Xianjun Yu
- Department of Pancreatic Surgery, Shanghai Cancer Center, Fudan University, 270 DongAn Road, Xuhui, Shanghai, 200032, People's Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Chen Liu
- Department of Pancreatic Surgery, Shanghai Cancer Center, Fudan University, 270 DongAn Road, Xuhui, Shanghai, 200032, People's Republic of China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China.
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China.
| | - He Cheng
- Department of Pancreatic Surgery, Shanghai Cancer Center, Fudan University, 270 DongAn Road, Xuhui, Shanghai, 200032, People's Republic of China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China.
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
49
|
Hegagi M, James P, Hsu A, Tanuseputro P. Home Care Use and Out-of-Hospital Death in Pancreatic Cancer Patients: A Retrospective Cohort Study. J Palliat Care 2023; 38:175-183. [PMID: 35179424 DOI: 10.1177/08258597221075088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Objective: This study aimed to determine the factors associated with usage of home care, including palliative home care, in patients with pancreatic cancer in Ontario. In addition, this study attempted to investigate factors associated with early-onset palliative home care as well as the impact of home care services on survival and out-of-hospital death. Methods: The Ontario Cancer Registry (OCR) was used to identify and capture basic patient/cancer characteristics of index cases of pancreatic cancer diagnosed between April first, 2010 and March 31st, 2016. Patients that received home care were identified using the Home Care Database (HCD) and stratified into general, transition-to-palliative, and early-onset palliative home care. Logistic regressions were used to describe determinants of home care use and determinants of out-of-hospital death. Results: A total of 6888 pancreatic cancer patients met eligibility criteria for this study. A high proportion of patients (83.7%) received home care, including palliative home care (56.8%). In general, older patients (OR = 3.07) and those with more advanced malignancy (OR = 4.98) for stage 4 versus stage 1) had greater odds of receiving palliative home care. Patients receiving home care (P < .01) and those residing in rural regions (P < .01) had greater odds of out-of-hospital death. Conclusion: A large proportion of patients with pancreatic cancer are directed to home care and those that do are more likely to die outside of hospital. Age and stage at diagnosis are significant predictors of home care use. Differences exist in the healthcare experience of patients depending on if they receive home care services and the type of home care.
Collapse
Affiliation(s)
- Mehdi Hegagi
- Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Paul James
- Department of Medicine, University Health Network, University of Toronto, Toronto, Ontario, Canada
- Institute for Clinical Evaluation Sciences, Cancer Research Program, Toronto, Ontario, Canada
| | - Amy Hsu
- Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
- Bruyère Research Institute, Ottawa, Ontario, Canada
- Department of Family Medicine, University of Ottawa, Ottawa, Canada
| | - Peter Tanuseputro
- Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
- Bruyère Research Institute, Ottawa, Ontario, Canada
- ICES, Population Health and Primary Care, Ottawa, Ontario, Canada
| |
Collapse
|
50
|
Zhang J, Li J, Xiong Y, Li R. Circ_0000284 upregulates RHPN2 to facilitate pancreatic cancer proliferation, metastasis, and angiogenesis through sponging miR-1179. J Biochem Mol Toxicol 2023; 37:e23274. [PMID: 36536496 DOI: 10.1002/jbt.23274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 08/15/2022] [Accepted: 12/02/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND Circular RNA (circRNA) has been confirmed to be a key regulator for pancreatic cancer (PC) progression, but the role of circ_0000284 in PC development remains unclear. METHODS Quantitative real-time PCR was used to measure the expression of circ_0000284, microRNA (miR)-1179, and rhophilin 2 (RHPN2). PC cell proliferation, metastasis, angiogenesis, and apoptosis were assessed by EdU assay, transwell assay, tube formation assay, and flow cytometry. Relative protein expression was determined by western blot analysis. The interaction between miR-1179 and circ_0000284 or RHPN2 was confirmed by dual-luciferase reporter assay and RNA pull-down assay. RESULTS Circ_0000284 was significantly upregulated in PC tissues and cells, and its knockdown inhibited PC cell proliferation, migration, invasion, and angiogenesis while promoting apoptosis. MiR-1179 was downregulated in PC tissues and cells, and it could be sponged by circ_0000284. Moreover, the miR-1179 inhibitor reversed the regulation of circ_0000284 knockdown on PC cell progression. The highly expressed RHPN2 was found in PC tissues and cells, and it could be targeted by miR-1179. Also, circ_0000284 sponged miR-1179 to regulate RHPN2 expression. Overexpressed RHPN2 could reverse the regulation of circ_0000284 knockdown on PC cell progression. In addition, interference of circ_0000284 was discovered to repress PC tumor growth by regulating miR-1179/RHPN2.RHPN2. CONCLUSION To sum up, our data confirmed that circ_0000284 facilitated PC malignant progression depending on the regulation of miR-1179/RHPN2 axis, suggesting that circ_0000284 might be a potential target for PC treatment.
Collapse
Affiliation(s)
- Jian Zhang
- Department of Geratic Gastroenterological Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jiangwei Li
- Department of Geratic Gastroenterological Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yongqiang Xiong
- Department of Geratic Gastroenterological Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Ren Li
- Department of Geratic Gastroenterological Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|