1
|
Ma Y, Dumesny C, Dong L, Ang CS, Nikfarjam M, He H. Knockout of p21-Activated Kinase 4 Stimulates MHC I Expression of Pancreatic Cancer Cells via an Autophagy-Independent Pathway. Cancers (Basel) 2025; 17:511. [PMID: 39941877 PMCID: PMC11817421 DOI: 10.3390/cancers17030511] [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: 12/21/2024] [Revised: 01/26/2025] [Accepted: 02/01/2025] [Indexed: 02/16/2025] Open
Abstract
BACKGROUND/OBJECTIVES Pancreatic ductal adenocarcinoma (PDA) is one of the most malignant solid cancers. KRAS mutation accounts for over 90% of cases. p21-activated kinases (PAKs) act downstream of KRAS and are involved in tumorigenesis. The inhibition of PAK4 suppresses PDA by stimulating the tumor infiltration of cytotoxic T cells. The major histocompatibility complex class I (MHC I) is a key in presenting antigens to cytotoxic T cells. MHC I degradation via autophagy promotes the immune evasion of pancreatic cancer. We investigated the effect of PAK4 inhibition on MHC I expression and autophagy. METHODS In this study, using proteomic analysis, fluorescence-activated cell sorting (FACS), and immunoblotting, we examined the effect of PAK4 knockout (KO) in human PDA cells on the expression of MHC I and autophagy to identify the mechanism involved in the stimulation of cytotoxic T cells by PAK4 inhibition. RESULTS We found that PAK4 KO increased MHC I expression in two human PDA cell lines: MiaPaCa-2 and PANC-1. PAK4 KO also increased cancer cell autophagy. However, the inhibition of autophagy by chloroquine (CQ) did not affect the effect of PAK4 KO on apoptosis and cell death. More importantly, the inhibition of autophagy by CQ did not alter the expression of MHC I stimulated by PAK4 KO, indicating that PAK4 KO stimulated MHC I expression via an autophagy-independent pathway. CONCLUSIONS We identified a role of PAK4 in MHC I expression by PDA cells, which is independent of autophagy.
Collapse
Affiliation(s)
- Yi Ma
- Department of Surgery, Austin Precinct, University of Melbourne, Heidelberg, VIC 3084, Australia; (Y.M.); (C.D.); (L.D.); (M.N.)
- Department of General Surgery, Monash Health, Clayton, VIC 3806, Australia
| | - Chelsea Dumesny
- Department of Surgery, Austin Precinct, University of Melbourne, Heidelberg, VIC 3084, Australia; (Y.M.); (C.D.); (L.D.); (M.N.)
| | - Li Dong
- Department of Surgery, Austin Precinct, University of Melbourne, Heidelberg, VIC 3084, Australia; (Y.M.); (C.D.); (L.D.); (M.N.)
| | - Ching-Seng Ang
- Mass Spectrometry and Proteomics Facility, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC 3052, Australia;
| | - Mehrdad Nikfarjam
- Department of Surgery, Austin Precinct, University of Melbourne, Heidelberg, VIC 3084, Australia; (Y.M.); (C.D.); (L.D.); (M.N.)
- Department of Hepato-Pancreato-Biliary Surgery, Austin Health, Heidelberg, VIC 3084, Australia
| | - Hong He
- Department of Surgery, Austin Precinct, University of Melbourne, Heidelberg, VIC 3084, Australia; (Y.M.); (C.D.); (L.D.); (M.N.)
| |
Collapse
|
2
|
Xu J, Pham MD, Corbo V, Ponz-Sarvise M, Oni T, Öhlund D, Hwang CI. Advancing pancreatic cancer research and therapeutics: the transformative role of organoid technology. Exp Mol Med 2025; 57:50-58. [PMID: 39814914 PMCID: PMC11799150 DOI: 10.1038/s12276-024-01378-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] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 10/11/2024] [Accepted: 10/14/2024] [Indexed: 01/18/2025] Open
Abstract
Research on pancreatic cancer has transformed with the advent of organoid technology, providing a better platform that closely mimics cancer biology in vivo. This review highlights the critical advancements facilitated by pancreatic organoid models in understanding disease progression, evaluating therapeutic responses, and identifying biomarkers. These three-dimensional cultures enable the proper recapitulation of the cellular architecture and genetic makeup of the original tumors, providing insights into the complex molecular and cellular dynamics at various stages of pancreatic ductal adenocarcinoma (PDAC). We explore the applications of pancreatic organoids in dissecting the tumor microenvironment (TME); elucidating cancer progression, metastasis, and drug resistance mechanisms; and personalizing therapeutic strategies. By overcoming the limitations of traditional 2D cultures and animal models, the use of pancreatic organoids has significantly accelerated translational research, which is promising for improving diagnostic and therapeutic approaches in clinical settings, ultimately aiming to improve the outcomes of patients with pancreatic cancer.
Collapse
Affiliation(s)
- Jihao Xu
- Department of Microbiology and Molecular Genetics, University of California, Davis, Davis, CA, 95616, USA
| | - Minh Duc Pham
- Department of Microbiology and Molecular Genetics, University of California, Davis, Davis, CA, 95616, USA
| | - Vincenzo Corbo
- Department of Engineering for Innovation Medicine, University of Verona, Verona, Italy
| | - Mariano Ponz-Sarvise
- Department of Medical Oncology and Program in Solid Tumors, Cima-Universidad de Navarra, Cancer Center Clinica Universidad de Navarra (CCUN), Pamplona, Pamplona, Spain
| | - Tobiloba Oni
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
| | - Daniel Öhlund
- Umeå University, Department of Diagnostics and Intervention, and Wallenberg Centre for Molecular Medicine at Umeå University, Umeå, Sweden
| | - Chang-Il Hwang
- Department of Microbiology and Molecular Genetics, University of California, Davis, Davis, CA, 95616, USA.
- University of California Davis Comprehensive Cancer Center, Sacramento, CA, 95817, USA.
| |
Collapse
|
3
|
Pian LL, Song MH, Wang TF, Qi L, Peng TL, Xie KP. Identification and analysis of pancreatic intraepithelial neoplasia: opportunities and challenges. Front Endocrinol (Lausanne) 2025; 15:1401829. [PMID: 39839479 PMCID: PMC11746065 DOI: 10.3389/fendo.2024.1401829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 12/17/2024] [Indexed: 01/23/2025] Open
Abstract
Pancreatic intraepithelial neoplasia (PanIN) is the most common precursor lesion of pancreatic ductal adenocarcinoma (PDAC), which has poor prognosis with a short median overall survival of 6-12 months and a low 5-year survival rate of approximately 3%. It is crucial to remove PanIN lesions to prevent the development of invasive PDAC, as PDAC spreads rapidly outside the pancreas. This review aims to provide the latest knowledge on PanIN risk, pathology, cellular origin, genetic susceptibility, and diagnosis, while identifying research gaps that require further investigation in this understudied area of precancerous lesions. PanINs are classified into PanIN 1, PanIN 2, and PanIN 3, with PanIN 3 having the highest likelihood of developing into invasive PDAC. Differentiating between PanIN 2 and PanIN 3 is clinically significant. Genetic alterations found in PDAC are also present in PanIN and increase with the grade of PanIN. Imaging methods alone are insufficient for distinguishing PanIN, necessitating the use of genetic and molecular tests for identification. In addition, metabolomics technologies and miRNAs are playing an increasingly important role in the field of cancer diagnosis, offering more possibilities for efficient identification of PanIN. Although detecting and stratifying the risk of PanIN poses challenges, the combined utilization of imaging, genetics, and metabolomics holds promise for improving patient survival in this field.
Collapse
Affiliation(s)
- Ling-ling Pian
- School of Medicine, The South China University of Technology, Guangzhou, Guangdong, China
- Division of Gastroenterology, Institute of Digestive Disease, Affiliated Qingyuan Hospital, The Sixth Clinical Medical School, Guangzhou Medical University, Qingyuan People’s Hospital, Qingyuan, Guangdong, China
| | - Mei-hui Song
- Division of Gastroenterology, Institute of Digestive Disease, Affiliated Qingyuan Hospital, The Sixth Clinical Medical School, Guangzhou Medical University, Qingyuan People’s Hospital, Qingyuan, Guangdong, China
| | - Teng-fei Wang
- Division of Gastroenterology, Institute of Digestive Disease, Affiliated Qingyuan Hospital, The Sixth Clinical Medical School, Guangzhou Medical University, Qingyuan People’s Hospital, Qingyuan, Guangdong, China
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong, China
| | - Ling Qi
- Division of Gastroenterology, Institute of Digestive Disease, Affiliated Qingyuan Hospital, The Sixth Clinical Medical School, Guangzhou Medical University, Qingyuan People’s Hospital, Qingyuan, Guangdong, China
| | - Tie-li Peng
- Division of Gastroenterology, Institute of Digestive Disease, Affiliated Qingyuan Hospital, The Sixth Clinical Medical School, Guangzhou Medical University, Qingyuan People’s Hospital, Qingyuan, Guangdong, China
| | - Ke-ping Xie
- School of Medicine, The South China University of Technology, Guangzhou, Guangdong, China
| |
Collapse
|
4
|
Kane LE, Mellotte GS, Mylod E, Dowling P, Marcone S, Scaife C, Kenny EM, Henry M, Meleady P, Ridgway PF, MacCarthy F, Conlon KC, Ryan BM, Maher SG. Multi-omic biomarker panel in pancreatic cyst fluid and serum predicts patients at a high risk of pancreatic cancer development. Sci Rep 2025; 15:129. [PMID: 39747972 PMCID: PMC11696309 DOI: 10.1038/s41598-024-83742-4] [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: 10/07/2024] [Accepted: 12/17/2024] [Indexed: 01/04/2025] Open
Abstract
Integration of multi-omic data for the purposes of biomarker discovery can provide novel and robust panels across multiple biological compartments. Appropriate analytical methods are key to ensuring accurate and meaningful outputs in the multi-omic setting. Here, we extensively profile the proteome and transcriptome of patient pancreatic cyst fluid (PCF) (n = 32) and serum (n = 68), before integrating matched omic and biofluid data, to identify biomarkers of pancreatic cancer risk. Differential expression analysis, feature reduction, multi-omic data integration, unsupervised hierarchical clustering, principal component analysis, spearman correlations and leave-one-out cross-validation were performed using RStudio and CombiROC software. An 11-feature multi-omic panel in PCF [PIGR, S100A8, REG1A, LGALS3, TCN1, LCN2, PRSS8, MUC6, SNORA66, miR-216a-5p, miR-216b-5p] generated an AUC = 0.806. A 13-feature multi-omic panel in serum [SHROOM3, IGHV3-72, IGJ, IGHA1, PPBP, APOD, SFN, IGHG1, miR-197-5p, miR-6741-5p, miR-3180, miR-3180-3p, miR-6782-5p] produced an AUC = 0.824. Integration of the strongest performing biomarkers generated a 10-feature cross-biofluid multi-omic panel [S100A8, LGALS3, SNORA66, miR-216b-5p, IGHV3-72, IGJ, IGHA1, PPBP, miR-3180, miR-3180-3p] with an AUC = 0.970. Multi-omic profiling provides an abundance of potential biomarkers. Integration of data from different omic compartments, and across biofluids, produced a biomarker panel that performs with high accuracy, showing promise for the risk stratification of patients with pancreatic cystic lesions.
Collapse
Affiliation(s)
- Laura E Kane
- Department of Surgery, Trinity St. James's Cancer Institute, Trinity Translational Medicine Institute, Trinity College Dublin, St. James's Hospital, Dublin 8, Ireland
| | - Gregory S Mellotte
- Department of Gastroenterology, Tallaght University Hospital, Dublin 24, Ireland
| | - Eimear Mylod
- Department of Surgery, Trinity St. James's Cancer Institute, Trinity Translational Medicine Institute, Trinity College Dublin, St. James's Hospital, Dublin 8, Ireland
| | - Paul Dowling
- Department of Biology, Maynooth University, Maynooth, Ireland
- Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Ireland
| | - Simone Marcone
- Department of Surgery, Trinity St. James's Cancer Institute, Trinity Translational Medicine Institute, Trinity College Dublin, St. James's Hospital, Dublin 8, Ireland
| | - Caitriona Scaife
- Mass Spectrometry Facility, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin 4, Ireland
| | - Elaine M Kenny
- ELDA Biotech, Newhall, M7 Business Park, Co. Kildare, Ireland
| | - Michael Henry
- National Institute for Cellular Biotechnology, Dublin City University, Dublin 9, Ireland
| | - Paula Meleady
- National Institute for Cellular Biotechnology, Dublin City University, Dublin 9, Ireland
| | - Paul F Ridgway
- Department of Surgery, Centre for Pancreatico-Biliary Diseases, Trinity College Dublin, St. James's Hospital, Dublin 8, Ireland
| | - Finbar MacCarthy
- Department of Clinical Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, St. James's Hospital, Dublin 8, Ireland
| | - Kevin C Conlon
- Department of Surgery, School of Medicine, Trinity College Dublin, Dublin 2, Ireland
| | - Barbara M Ryan
- Department of Gastroenterology, Tallaght University Hospital, Dublin 24, Ireland
| | - Stephen G Maher
- Department of Surgery, Trinity St. James's Cancer Institute, Trinity Translational Medicine Institute, Trinity College Dublin, St. James's Hospital, Dublin 8, Ireland.
| |
Collapse
|
5
|
Torres C, Mancinelli G, Chen JWE, Cordoba-Chacon J, Pins D, Saeed S, McKinney R, Castellanos K, Orsi G, Singhal M, Patel A, Acebedo J, Coleman A, Heneche J, Yalagala PCR, Subbaiah PV, Leal C, Grimaldo S, Ortuno FM, Bishehsari F, Grippo PJ. Cell Membrane Fatty Acids and PIPs Modulate the Etiology of Pancreatic Cancer by Regulating AKT. Nutrients 2024; 17:150. [PMID: 39796583 PMCID: PMC11722924 DOI: 10.3390/nu17010150] [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] [Revised: 12/17/2024] [Accepted: 12/20/2024] [Indexed: 01/13/2025] Open
Abstract
Background: Pancreatic ductal adenocarcinoma (PDAC) is one of the worst solid malignancies in regard to outcomes and metabolic dysfunction leading to cachexia. It is alarming that PDAC incidence rates continue to increase and warrant the need for innovative approaches to combat this disease. Due to its relatively slow progression (10-20 years), prevention strategies represent an effective means to improve outcomes. One of the risk factors for many cancers and for pancreatic cancer in particular is diet. Hence, our objective is to understand how a diet rich in ω3 and ω6 polyunsaturated fatty acids affects the progression of this disease. Methods: We investigated polyunsaturated fatty acid (PUFA) effects on disease progression employing both in vitro (PDAC cell lines) and in vivo (EL-Kras and KC mice) approaches. Also, we gathered data from the National Health and Nutrition Examination Survey (NHANES) and the National Cancer Institute (NCI) from 1999 to 2017 for a retrospective observational study. Results: The consumption of PUFAs in a patient population correlates with increased PDAC incidence, particularly when the ω3 intake increases to a lesser extent than ω6. Our data demonstrate dietary PUFAs can be incorporated into plasma membrane lipids affecting PI3K/AKT signaling and support the emergence of membrane-targeted therapies. Moreover, we show that the phospholipid composition of a lipid nanoparticle (LNP) can impact the cell membrane integrity and, ultimately, cell viability after administration of these LNPs. Conclusions: Cancer prevention is impactful particularly for those with very poor prognosis, including pancreatic cancer. Our results point to the importance of dietary intervention in this disease when detected early and the potential to improve the antiproliferative effect of drug efficacy when combined with these regimens in later stages of pancreatic cancer.
Collapse
Affiliation(s)
- Carolina Torres
- Department of Biochemistry and Molecular Biology III and Immunology, Faculty of Medicine, University of Granada, 18071 Granada, Spain
- Instituto de Investigacion Biosanitaria ibs.GRANADA, 18012 Granada, Spain
| | - Georgina Mancinelli
- Department of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA; (G.M.); (S.S.); (R.M.); (A.P.)
| | - Jee-Wei Emily Chen
- Department of Materials Science & Engineering, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA; (J.-W.E.C.)
| | - Jose Cordoba-Chacon
- Department of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA; (G.M.); (S.S.); (R.M.); (A.P.)
| | - Danielle Pins
- Department of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA; (G.M.); (S.S.); (R.M.); (A.P.)
| | - Sara Saeed
- Department of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA; (G.M.); (S.S.); (R.M.); (A.P.)
| | - Ronald McKinney
- Department of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA; (G.M.); (S.S.); (R.M.); (A.P.)
| | - Karla Castellanos
- Department of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA; (G.M.); (S.S.); (R.M.); (A.P.)
| | | | - Megha Singhal
- Department of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA; (G.M.); (S.S.); (R.M.); (A.P.)
| | - Akshar Patel
- Department of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA; (G.M.); (S.S.); (R.M.); (A.P.)
| | - Jose Acebedo
- Department of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA; (G.M.); (S.S.); (R.M.); (A.P.)
| | - Adonis Coleman
- Department of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA; (G.M.); (S.S.); (R.M.); (A.P.)
| | - Jorge Heneche
- Department of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA; (G.M.); (S.S.); (R.M.); (A.P.)
| | - Poorna Chandra Rao Yalagala
- Department of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA; (G.M.); (S.S.); (R.M.); (A.P.)
| | - Papasani V. Subbaiah
- Department of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA; (G.M.); (S.S.); (R.M.); (A.P.)
| | - Cecilia Leal
- Department of Materials Science & Engineering, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA; (J.-W.E.C.)
| | - Sam Grimaldo
- Department of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA; (G.M.); (S.S.); (R.M.); (A.P.)
| | - Francisco M. Ortuno
- Department of Computer Architecture and Computer Technology, University of Granada, 18071 Granada, Spain
| | - Faraz Bishehsari
- Department of Medicine, Rush University Medical Center, Chicago, IL 60612, USA
| | - Paul J. Grippo
- Department of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA; (G.M.); (S.S.); (R.M.); (A.P.)
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Illinois Chicago, 840 S. Wood Street, CSB 708, Chicago, IL 60612, USA
| |
Collapse
|
6
|
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
|
7
|
Valenti G, Laise P, Takahashi R, Wu F, Ruan T, Vasciaveo A, Jiang Z, Sunagawa M, Middelhoff M, Nienhüser H, Fu N, Malagola E, Hayakawa Y, Iuga AC, Califano A, Wang TC. Regulatory network analysis of Dclk1 gene expression reveals a tuft cell-ILC2 axis that inhibits pancreatic tumor progression. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.30.610508. [PMID: 39257805 PMCID: PMC11383664 DOI: 10.1101/2024.08.30.610508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
Dclk1 expression defines a rare population of cells in the normal pancreas whose frequency is increased at early stages of pancreatic tumorigenesis. The identity and the precise roles of Dclk1 expressing cells in pancreas have been matter of debate, although evidence suggests their involvement in a number of key functions, including regeneration and neoplasia. We employed a recently developed Dclk1 reporter mouse model and single cell RNAseq analysis to define Dclk1 expressing cells in normal pancreas and pancreatic neoplasia. In normal pancreas, Dclk1 epithelial expression identifies subsets of ductal, islet and acinar cells. In pancreatic neoplasia, Dclk1 expression identifies five epithelial cell populations, among which acinar-to-ductal metaplasia (ADM)-like cells and tuft-like cells are predominant. These two cell populations play opposing roles in pancreatic neoplasia, with Dclk1+ ADM-like cells sustaining tumor growth while Dclk1+ tuft-like cells restraining tumor progression. The differentiation of Kras mutant acinar cells into Dclk1+ tuft-like cells requires the activation of the transcription factor SPIB and is further supported by a cellular paracrine loop involving cancer group 2 innate lymphoid cells (ILC2) and cancer activated fibroblasts (CAFs) that provide IL13 and IL33, respectively. In turn, Dclk1+ tuft-like cells release angiotensinogen that plays protective roles against pancreatic neoplasia. Overall, our study provides novel insights on the biology of Dclk1+ cells in normal pancreas and unveils a protective axis against pancreatic neoplasia, involving CAFs, ILC2 and Dclk1+ tuft-like cells, which ultimately results in angiotensinogen release.
Collapse
Affiliation(s)
- Giovanni Valenti
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University, New York, New York, USA
- These authors contributed equally
| | - Pasquale Laise
- Department of Systems Biology, Columbia University, New York, New York, USA
- DarwinHealth Inc., New York, New York, USA
- These authors contributed equally
| | - Ryota Takahashi
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University, New York, New York, USA
| | - Feijing Wu
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University, New York, New York, USA
| | - Tuo Ruan
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University, New York, New York, USA
| | | | - Zhengyu Jiang
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University, New York, New York, USA
| | - Masaki Sunagawa
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University, New York, New York, USA
| | - Moritz Middelhoff
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, TU Munich, Germany
| | - Henrik Nienhüser
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University, New York, New York, USA
| | - Na Fu
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University, New York, New York, USA
| | - Ermanno Malagola
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University, New York, New York, USA
| | - Yoku Hayakawa
- Graduate School of Medicine, Department of Gastroenterology, The University of Tokyo, Tokyo, Japan
| | - Alina C. Iuga
- Department of Pathology and Cell Biology, Columbia University, New York, New York, USA
| | - Andrea Califano
- Department of Systems Biology, Columbia University, New York, New York, USA
| | - Timothy C. Wang
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University, New York, New York, USA
- Lead Contact
| |
Collapse
|
8
|
Ma Y, He X, Di Y, Li W, Sun L, Zhang X, Xu L, Bai Z, Li Z, Cai L, Sun H, Corpe C, Wang J. Circular RNA LIPH promotes pancreatic cancer glycolysis and progression through sponge miR-769-3p and interaction with GOLM1. Clin Transl Med 2024; 14:e70003. [PMID: 39167076 PMCID: PMC11337905 DOI: 10.1002/ctm2.70003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 08/01/2024] [Accepted: 08/07/2024] [Indexed: 08/23/2024] Open
Affiliation(s)
- Yan Ma
- Central LaboratoryZhongshan Hospital (Xiamen), Fudan UniversityXiamenFujianChina
- Shanghai Public Health Clinical CenterFudan UniversityShanghaiChina
| | - Xiaomeng He
- Shanghai Public Health Clinical CenterFudan UniversityShanghaiChina
| | - Yang Di
- Department of Pancreatic Surgery, Pancreatic Disease InstituteHuashan Hospital, Shanghai Medical College, Fudan UniversityShanghaiChina
| | - Wenyang Li
- Department of Physiology and PathophysiologyHexi University School of MedicineZhangyeGansuChina
| | - Lixiang Sun
- Central LaboratoryZhongshan Hospital (Xiamen), Fudan UniversityXiamenFujianChina
| | - Xin Zhang
- Central LaboratoryZhongshan Hospital (Xiamen), Fudan UniversityXiamenFujianChina
| | - Li Xu
- Shanghai Public Health Clinical CenterFudan UniversityShanghaiChina
| | - Zhihui Bai
- Central LaboratoryZhongshan Hospital (Xiamen), Fudan UniversityXiamenFujianChina
| | - Zehuan Li
- Department of General SurgeryZhongshan Hospital, Fudan UniversityShanghaiChina
| | - Lijun Cai
- Central LaboratoryZhongshan Hospital (Xiamen), Fudan UniversityXiamenFujianChina
| | - Huaqin Sun
- Central LaboratoryZhongshan Hospital (Xiamen), Fudan UniversityXiamenFujianChina
| | | | - Jin Wang
- Central LaboratoryZhongshan Hospital (Xiamen), Fudan UniversityXiamenFujianChina
- Shanghai Public Health Clinical CenterFudan UniversityShanghaiChina
| |
Collapse
|
9
|
Mustafa M, Abbas K, Alam M, Habib S, Zulfareen, Hasan GM, Islam S, Shamsi A, Hassan I. Investigating underlying molecular mechanisms, signaling pathways, emerging therapeutic approaches in pancreatic cancer. Front Oncol 2024; 14:1427802. [PMID: 39087024 PMCID: PMC11288929 DOI: 10.3389/fonc.2024.1427802] [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: 05/04/2024] [Accepted: 07/01/2024] [Indexed: 08/02/2024] Open
Abstract
Pancreatic adenocarcinoma, a clinically challenging malignancy constitutes a significant contributor to cancer-related mortality, characterized by an inherently poor prognosis. This review aims to provide a comprehensive understanding of pancreatic adenocarcinoma by examining its multifaceted etiologies, including genetic mutations and environmental factors. The review explains the complex molecular mechanisms underlying its pathogenesis and summarizes current therapeutic strategies, including surgery, chemotherapy, and emerging modalities such as immunotherapy. Critical molecular pathways driving pancreatic cancer development, including KRAS, Notch, and Hedgehog, are discussed. Current therapeutic strategies, including surgery, chemotherapy, and radiation, are discussed, with an emphasis on their limitations, particularly in terms of postoperative relapse. Promising research areas, including liquid biopsies, personalized medicine, and gene editing, are explored, demonstrating the significant potential for enhancing diagnosis and treatment. While immunotherapy presents promising prospects, it faces challenges related to immune evasion mechanisms. Emerging research directions, encompassing liquid biopsies, personalized medicine, CRISPR/Cas9 genome editing, and computational intelligence applications, hold promise for refining diagnostic approaches and therapeutic interventions. By integrating insights from genetic, molecular, and clinical research, innovative strategies that improve patient outcomes can be developed. Ongoing research in these emerging fields holds significant promise for advancing the diagnosis and treatment of this formidable malignancy.
Collapse
Affiliation(s)
- Mohd Mustafa
- Department of Biochemistry, J.N. Medical College, Faculty of Medicine, Aligarh Muslim University, Aligarh, India
| | - Kashif Abbas
- Department of Zoology, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, India
| | - Mudassir Alam
- Department of Zoology, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, India
| | - Safia Habib
- Department of Biochemistry, J.N. Medical College, Faculty of Medicine, Aligarh Muslim University, Aligarh, India
| | - Zulfareen
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| | - Gulam Mustafa Hasan
- Department of Basic Medical Science, College of Medicine, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Sidra Islam
- Department of Inflammation & Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Anas Shamsi
- Center of Medical and Bio-Allied Health Sciences Research (CMBHSR), Ajman University, Ajman, United Arab Emirates
| | - Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| |
Collapse
|
10
|
Ma Y, Dumesny C, Dong L, Ang CS, Asadi K, Zhan Y, Nikfarjam M, He H. Inhibition of P21-activated kinases 1 and 4 synergistically suppresses the growth of pancreatic cancer by stimulating anti-tumour immunity. Cell Commun Signal 2024; 22:287. [PMID: 38797819 PMCID: PMC11129409 DOI: 10.1186/s12964-024-01670-2] [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: 02/21/2024] [Accepted: 05/19/2024] [Indexed: 05/29/2024] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDA) is one of the most lethal types of cancer, and KRAS oncogene occurs in over 90% of cases. P21-activated kinases (PAK), containing six members (PAK1 to 6), function downstream of KRAS. PAK1 and PAK4 play important roles in carcinogenesis, but their combinational effect remains unknown. In this study, we have determined the effect of dual inhibition of PAK1 and PAK4 in PDA progression using knockout (KO) cancer cell lines. METHODS Murine wild-type (WT) and PAK1KO pancreatic cancer cell lines were isolated from PAK1+/+ and PAK1-/- KPC (LSL-KrasG12D/+; LSL-Trp53 R172H/+; Pdx-1-Cre) mice. KPC PAK4KO and KPC PAK1&4 KO cell lines were generated from KPC WT and KPC PAK1KO cell lines respectively using the CRISPR-CAS9 gene knockout technique. PAK WT and KO cell lines were used in mouse models of pancreatic tumours. Cells and tumour tissue were also used in flow cytometry and proteomic studies. A human PDA tissue microarray was stained by immunohistochemistry. RESULTS Double knock out of PAK1 and PAK4 caused complete regression of tumour in a syngeneic mouse model. PAK4KO inhibited tumour growth by stimulating a rapid increase of cytotoxic CD8+ T cell infiltration. PAK1KO synergistically with PAK4KO increased cytotoxic CD8+ T cell infiltration and stimulated a sustained infiltration of CD8+ T cells at a later phase to overcome the immune evasion in the PAK4KO tumour. The human PDA tissue microarray study showed the important role of PAK1 and PAK4 in intra-tumoral T-cell function. CONCLUSION Our results demonstrated that dual inhibition of PAK1 and PAK4 synergistically suppressed PDA progression by stimulating cytotoxic CD8 + T cell response.
Collapse
Affiliation(s)
- Yi Ma
- Department of Surgery, Austin Precinct, University of Melbourne, Level 8, Lance Townsend Building, Austin Hospital, 145 Studley Road, Heidelberg, VIC, Australia
- Department of General Surgery, Monash Health, Clayton, VIC, Australia
| | - Chelsea Dumesny
- Department of Surgery, Austin Precinct, University of Melbourne, Level 8, Lance Townsend Building, Austin Hospital, 145 Studley Road, Heidelberg, VIC, Australia
| | - Li Dong
- Department of Surgery, Austin Precinct, University of Melbourne, Level 8, Lance Townsend Building, Austin Hospital, 145 Studley Road, Heidelberg, VIC, Australia
| | - Ching-Seng Ang
- Mass Spectrometry and Proteomics Facility, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC, Australia
| | - Khashayar Asadi
- Department of Anatomical Pathology, Austin Health, Heidelberg, VIC, Australia
| | - Yifan Zhan
- Drug Discovery, Shanghai Huaota Biopharm, Shanghai, China
| | - Mehrdad Nikfarjam
- Department of Surgery, Austin Precinct, University of Melbourne, Level 8, Lance Townsend Building, Austin Hospital, 145 Studley Road, Heidelberg, VIC, Australia
- Department of Hepato-Pancreato-Biliary Surgery, Austin Health, Heidelberg, VIC, Australia
| | - Hong He
- Department of Surgery, Austin Precinct, University of Melbourne, Level 8, Lance Townsend Building, Austin Hospital, 145 Studley Road, Heidelberg, VIC, Australia.
| |
Collapse
|
11
|
Vendramini-Costa DB, Francescone R, Franco-Barraza J, Luong T, Graves M, de Aquino AM, Steele N, Gardiner JC, Dos Santos SAA, Ogier C, Malloy E, Borghaei L, Martinez E, Zhigarev DI, Tan Y, Lee H, Zhou Y, Cai KQ, Klein-Szanto AJ, Wang H, Andrake M, Dunbrack RL, Campbell K, Cukierman E. Netrin G1 Ligand is a new stromal immunomodulator that promotes pancreatic cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.15.594354. [PMID: 38798370 PMCID: PMC11118300 DOI: 10.1101/2024.05.15.594354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Understanding pancreatic cancer biology is fundamental for identifying new targets and for developing more effective therapies. In particular, the contribution of the stromal microenvironment to pancreatic cancer tumorigenesis requires further exploration. Here, we report the stromal roles of the synaptic protein Netrin G1 Ligand (NGL-1) in pancreatic cancer, uncovering its pro-tumor functions in cancer-associated fibroblasts and in immune cells. We observed that the stromal expression of NGL-1 inversely correlated with patients' overall survival. Moreover, germline knockout (KO) mice for NGL-1 presented decreased tumor burden, with a microenvironment that is less supportive of tumor growth. Of note, tumors from NGL-1 KO mice produced less immunosuppressive cytokines and displayed an increased percentage of CD8 + T cells than those from control mice, while preserving the physical structure of the tumor microenvironment. These effects were shown to be mediated by NGL-1 in both immune cells and in the local stroma, in a TGF-β-dependent manner. While myeloid cells lacking NGL-1 decreased the production of immunosuppressive cytokines, NGL-1 KO T cells showed increased proliferation rates and overall polyfunctionality compared to control T cells. CAFs lacking NGL-1 were less immunosuppressive than controls, with overall decreased production of pro-tumor cytokines and compromised ability to inhibit CD8 + T cells activation. Mechanistically, these CAFs downregulated components of the TGF-β pathway, AP-1 and NFAT transcription factor families, resulting in a less tumor-supportive phenotype. Finally, targeting NGL-1 genetically or using a functionally antagonistic small peptide phenocopied the effects of chemotherapy, while modulating the immunosuppressive tumor microenvironment (TME), rather than eliminating it. We propose NGL-1 as a new local stroma and immunomodulatory molecule, with pro-tumor roles in pancreatic cancer. Statement of Significance Here we uncovered the pro-tumor roles of the synaptic protein NGL-1 in the tumor microenvironment of pancreatic cancer, defining a new target that simultaneously modulates tumor cell, fibroblast, and immune cell functions. This study reports a new pathway where NGL-1 controls TGF-β, AP-1 transcription factor members and NFAT1, modulating the immunosuppressive microenvironment in pancreatic cancer. Our findings highlight NGL-1 as a new stromal immunomodulator in pancreatic cancer.
Collapse
|
12
|
Tsesmelis M, Büttner UFG, Gerstenlauer M, Manfras U, Tsesmelis K, Du Z, Sperb N, Weissinger SE, Möller P, Barth TFE, Maier HJ, Chan LK, Wirth T. NEMO/NF-κB signaling functions as a double-edged sword in PanIN formation versus progression to pancreatic cancer. Mol Cancer 2024; 23:103. [PMID: 38755681 PMCID: PMC11097402 DOI: 10.1186/s12943-024-01989-x] [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: 12/08/2023] [Accepted: 03/31/2024] [Indexed: 05/18/2024] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is marked by a dismal survival rate, lacking effective therapeutics due to its aggressive growth, late-stage diagnosis, and chemotherapy resistance. Despite debates on NF-κB targeting for PDAC treatment, no successful approach has emerged. METHODS To elucidate the role of NF-κB, we ablated NF-κB essential modulator (NEMO), critical for conventional NF-κB signaling, in the pancreata of mice that develop precancerous lesions (KC mouse model). Secretagogue-induced pancreatitis by cerulein injections was utilized to promote inflammation and accelerate PDAC development. RESULTS NEMO deletion reduced fibrosis and inflammation in young KC mice, resulting in fewer pancreatic intraepithelial neoplasias (PanINs) at later stages. Paradoxically, however, NEMO deletion accelerated the progression of these fewer PanINs to PDAC and reduced median lifespan. Further, analysis of tissue microarrays from human PDAC sections highlighted the correlation between reduced NEMO expression in neoplastic cells and poorer prognosis, supporting our observation in mice. Mechanistically, NEMO deletion impeded oncogene-induced senescence (OIS), which is normally active in low-grade PanINs. This blockage resulted in fewer senescence-associated secretory phenotype (SASP) factors, reducing inflammation. However, blocked OIS fostered replication stress and DNA damage accumulation which accelerated PanIN progression to PDAC. Finally, treatment with the DNA damage-inducing reagent etoposide resulted in elevated cell death in NEMO-ablated PDAC cells compared to their NEMO-competent counterparts, indicative of a synthetic lethality paradigm. CONCLUSIONS NEMO exhibited both oncogenic and tumor-suppressive properties during PDAC development. Caution is suggested in therapeutic interventions targeting NF-κB, which may be detrimental during PanIN progression but beneficial post-PDAC development.
Collapse
Affiliation(s)
- Miltiadis Tsesmelis
- Institute of Physiological Chemistry, University of Ulm, Meyerhofstrasse, 89081, Ulm, Baden-Württemberg, Germany
| | - Ulrike F G Büttner
- Institute of Physiological Chemistry, University of Ulm, Meyerhofstrasse, 89081, Ulm, Baden-Württemberg, Germany
| | - Melanie Gerstenlauer
- Institute of Physiological Chemistry, University of Ulm, Meyerhofstrasse, 89081, Ulm, Baden-Württemberg, Germany
| | - Uta Manfras
- Institute of Physiological Chemistry, University of Ulm, Meyerhofstrasse, 89081, Ulm, Baden-Württemberg, Germany
| | - Konstantinos Tsesmelis
- Institute of Physiological Chemistry, University of Ulm, Meyerhofstrasse, 89081, Ulm, Baden-Württemberg, Germany
| | - Ziwei Du
- Institute of Physiological Chemistry, University of Ulm, Meyerhofstrasse, 89081, Ulm, Baden-Württemberg, Germany
| | - Nadine Sperb
- Institute of Physiological Chemistry, University of Ulm, Meyerhofstrasse, 89081, Ulm, Baden-Württemberg, Germany
| | | | - Peter Möller
- Institute of Pathology, University of Ulm, 89081, Ulm, Baden-Württemberg, Germany
| | - Thomas F E Barth
- Institute of Pathology, University of Ulm, 89081, Ulm, Baden-Württemberg, Germany
| | - Harald J Maier
- Institute of Physiological Chemistry, University of Ulm, Meyerhofstrasse, 89081, Ulm, Baden-Württemberg, Germany
- Novartis Pharma, 4056, Basel, AG, Switzerland
| | - Lap Kwan Chan
- Institute of Physiological Chemistry, University of Ulm, Meyerhofstrasse, 89081, Ulm, Baden-Württemberg, Germany.
- Department of Pathology and Molecular Pathology, University Hospital of Zurich, 8091, Zurich, Switzerland.
- Institute of Molecular Cancer Research, University of Zurich, 8057, Zurich, Switzerland.
| | - Thomas Wirth
- Institute of Physiological Chemistry, University of Ulm, Meyerhofstrasse, 89081, Ulm, Baden-Württemberg, Germany.
| |
Collapse
|
13
|
Rao J, Sinn M, Pelzer U, Riess H, Oettle H, Demir IE, Friess H, Jäger C, Steiger K, Muckenhuber A. KRT81 and HNF1A expression in pancreatic ductal adenocarcinoma: investigation of predictive and prognostic value of immunohistochemistry-based subtyping. J Pathol Clin Res 2024; 10:e12377. [PMID: 38750616 PMCID: PMC11096282 DOI: 10.1002/2056-4538.12377] [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: 12/18/2023] [Revised: 03/19/2024] [Accepted: 04/29/2024] [Indexed: 05/18/2024]
Abstract
Even after decades of research, pancreatic ductal adenocarcinoma (PDAC) remains a highly lethal disease and responses to conventional treatments remain mostly poor. Subclassification of PDAC into distinct biological subtypes has been proposed by various groups to further improve patient outcome and reduce unnecessary side effects. Recently, an immunohistochemistry (IHC)-based subtyping method using cytokeratin-81 (KRT81) and hepatocyte nuclear factor 1A (HNF1A) could recapitulate some of the previously established molecular subtyping methods, while providing significant prognostic and, to a limited degree, also predictive information. We refined the KRT81/HNF1A subtyping method to classify PDAC into three distinct biological subtypes. The prognostic value of the IHC-based method was investigated in two primary resected cohorts, which include 269 and 286 patients, respectively. In the second cohort, we also assessed the predictive effect for response to erlotinib + gemcitabine. In both PDAC cohorts, the new HNF1A-positive subtype was associated with the best survival, the KRT81-positive subtype with the worst, and the double-negative with an intermediate survival (p < 0.001 and p < 0.001, respectively) in univariate and multivariate analyses. In the second cohort (CONKO-005), the IHC-based subtype was additionally found to have a potential predictive value for the erlotinib-based treatment effect. The revised IHC-based subtyping using KRT81 and HNF1A has prognostic significance for PDAC patients and may be of value in predicting treatment response to specific therapeutic agents.
Collapse
Affiliation(s)
- Jia Rao
- Institute of PathologyTechnical University of MunichMunichGermany
| | - Marianne Sinn
- Department of Haematology, Oncology and Tumour Immunology, CONKO‐Study‐GroupCharité – University Medicine BerlinBerlinGermany
- Department of Internal Medicine IIUniversity Medical Center of Hamburg‐EppendorfHamburgGermany
| | - Uwe Pelzer
- Department of Haematology, Oncology and Tumour Immunology, CONKO‐Study‐GroupCharité – University Medicine BerlinBerlinGermany
| | - Hanno Riess
- Department of Haematology, Oncology and Tumour Immunology, CONKO‐Study‐GroupCharité – University Medicine BerlinBerlinGermany
| | - Helmut Oettle
- Department of Haematology, Oncology and Tumour Immunology, CONKO‐Study‐GroupCharité – University Medicine BerlinBerlinGermany
| | - Ihsan E Demir
- Department of Surgery, Klinikum rechts der Isar, School of MedicineTechnical University of MunichMunichGermany
- Else Kröner Clinician Scientist Professor for Translational Pancreatic SurgeryMunichGermany
| | - Helmut Friess
- Department of Surgery, Klinikum rechts der Isar, School of MedicineTechnical University of MunichMunichGermany
| | - Carsten Jäger
- Department of Surgery, Klinikum rechts der Isar, School of MedicineTechnical University of MunichMunichGermany
| | - Katja Steiger
- Institute of PathologyTechnical University of MunichMunichGermany
| | | |
Collapse
|
14
|
Lee DU, Han BS, Jung KH, Hong SS. Tumor Stroma as a Therapeutic Target for Pancreatic Ductal Adenocarcinoma. Biomol Ther (Seoul) 2024; 32:281-290. [PMID: 38590092 PMCID: PMC11063484 DOI: 10.4062/biomolther.2024.029] [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: 02/20/2024] [Revised: 03/20/2024] [Accepted: 03/21/2024] [Indexed: 04/10/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has a poor prognosis owing to its desmoplastic stroma. Therefore, therapeutic strategies targeting this tumor stroma should be developed. In this study, we describe the heterogeneity of cancer-associated fibroblasts (CAFs) and their diverse roles in the progression, immune evasion, and resistance to treatment of PDAC. We subclassified the spatial distribution and functional activity of CAFs to highlight their effects on prognosis and drug delivery. Extracellular matrix components such as collagen and hyaluronan are described for their roles in tumor behavior and treatment outcomes, implying their potential as therapeutic targets. We also discussed the roles of extracellular matrix (ECM) including matrix metalloproteinases and tissue inhibitors in PDAC progression. Finally, we explored the role of the adaptive and innate immune systems in shaping the PDAC microenvironment and potential therapeutic strategies, with a focus on immune cell subsets, cytokines, and immunosuppressive mechanisms. These insights provide a comprehensive understanding of PDAC and pave the way for the development of prognostic markers and therapeutic interventions.
Collapse
Affiliation(s)
- Dae Ui Lee
- Department of Medicine, College of Medicine, Inha University, Incheon 22332, Republic of Korea
| | - Beom Seok Han
- Program in Biomedical Science & Engineering, The Graduate School, Inha University, Incheon 22212, Republic of Korea
| | - Kyung Hee Jung
- Department of Medicine, College of Medicine, Inha University, Incheon 22332, Republic of Korea
| | - Soon-Sun Hong
- Department of Medicine, College of Medicine, Inha University, Incheon 22332, Republic of Korea
- Program in Biomedical Science & Engineering, The Graduate School, Inha University, Incheon 22212, Republic of Korea
| |
Collapse
|
15
|
Dutta D, Ray P, De A, Ghosh A, Hazra RS, Ghosh P, Banerjee S, Diaz FJ, Upadhyay SP, Quadir M, Banerjee SK. pH-responsive targeted nanoparticles release ERK-inhibitor in the hypoxic zone and sensitize free gemcitabine in mutant K-Ras-addicted pancreatic cancer cells and mouse model. PLoS One 2024; 19:e0297749. [PMID: 38687749 PMCID: PMC11060587 DOI: 10.1371/journal.pone.0297749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Accepted: 01/12/2024] [Indexed: 05/02/2024] Open
Abstract
Therapeutic options for managing Pancreatic ductal adenocarcinoma (PDAC), one of the deadliest types of aggressive malignancies, are limited and disappointing. Therefore, despite suboptimal clinical effects, gemcitabine (GEM) remains the first-line chemotherapeutic drug in the clinic for PDAC treatment. The therapeutic limitations of GEM are primarily due to poor bioavailability and the development of chemoresistance resulting from the addiction of mutant-K-RAS/AKT/ERK signaling-mediated desmoplastic barriers with a hypoxic microenvironment. Several new therapeutic approaches, including nanoparticle-assisted drug delivery, are being investigated by us and others. This study used pH-responsive nanoparticles encapsulated ERK inhibitor (SCH772984) and surface functionalized with tumor-penetrating peptide, iRGD, to target PDAC tumors. We used a small molecule, SCH772984, to target ERK1 and ERK2 in PDAC and other cancer cells. This nanocarrier efficiently released ERKi in hypoxic and low-pH environments. We also found that the free-GEM, which is functionally weak when combined with nanoencapsulated ERKi, led to significant synergistic treatment outcomes in vitro and in vivo. In particular, the combination approaches significantly enhanced the GEM effect in PDAC growth inhibition and prolonged survival of the animals in a genetically engineered KPC (LSL-KrasG12D/+/LSL-Trp53R172H/+/Pdx-1-Cre) pancreatic cancer mouse model, which is not observed in a single therapy. Mechanistically, we anticipate that the GEM efficacy was increased as ERKi blocks desmoplasia by impairing the production of desmoplastic regulatory factors in PDAC cells and KPC mouse tumors. Therefore, 2nd generation ERKi (SCH 772984)-iRGD-pHNPs are vital for the cellular response to GEM and denote a promising therapeutic target in PDAC with mutant K-RAS.
Collapse
Affiliation(s)
- Debasmita Dutta
- Department of Coatings and Polymeric Materials, North Dakota State University, Fargo, ND, United States of America
| | - Priyanka Ray
- Department of Coatings and Polymeric Materials, North Dakota State University, Fargo, ND, United States of America
| | - Archana De
- Cancer Research Unit, VA Medical Center, Kansas City, MO, United States of America
| | - Arnab Ghosh
- Cancer Research Unit, VA Medical Center, Kansas City, MO, United States of America
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, United States of America
| | - Raj Shankar Hazra
- Department of Coatings and Polymeric Materials, North Dakota State University, Fargo, ND, United States of America
| | - Pratyusha Ghosh
- Department of Coatings and Polymeric Materials, North Dakota State University, Fargo, ND, United States of America
- Cancer Research Unit, VA Medical Center, Kansas City, MO, United States of America
| | - Snigdha Banerjee
- Cancer Research Unit, VA Medical Center, Kansas City, MO, United States of America
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, United States of America
| | - Francisco J. Diaz
- Department of Biostatistics & Data Science, University of Kansas Medical Center, Kansas City, KS, United States of America
| | - Sunil P. Upadhyay
- Cancer Research Unit, VA Medical Center, Kansas City, MO, United States of America
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, United States of America
| | - Mohiuddin Quadir
- Department of Coatings and Polymeric Materials, North Dakota State University, Fargo, ND, United States of America
| | - Sushanta K. Banerjee
- Cancer Research Unit, VA Medical Center, Kansas City, MO, United States of America
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, United States of America
| |
Collapse
|
16
|
Gu M, Liu Y, Xin P, Guo W, Zhao Z, Yang X, Ma R, Jiao T, Zheng W. Fundamental insights and molecular interactions in pancreatic cancer: Pathways to therapeutic approaches. Cancer Lett 2024; 588:216738. [PMID: 38401887 DOI: 10.1016/j.canlet.2024.216738] [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: 01/08/2024] [Revised: 02/08/2024] [Accepted: 02/18/2024] [Indexed: 02/26/2024]
Abstract
The gastrointestinal tract can be affected by a number of diseases that pancreatic cancer (PC) is a malignant manifestation of them. The prognosis of PC patients is unfavorable and because of their diagnosis at advanced stage, the treatment of this tumor is problematic. Owing to low survival rate, there is much interest towards understanding the molecular profile of PC in an attempt in developing more effective therapeutics. The conventional therapeutics for PC include surgery, chemotherapy and radiotherapy as well as emerging immunotherapy. However, PC is still incurable and more effort should be performed. The molecular landscape of PC is an underlying factor involved in increase in progression of tumor cells. In the presence review, the newest advances in understanding the molecular and biological events in PC are discussed. The dysregulation of molecular pathways including AMPK, MAPK, STAT3, Wnt/β-catenin and non-coding RNA transcripts has been suggested as a factor in development of tumorigenesis in PC. Moreover, cell death mechanisms such as apoptosis, autophagy, ferroptosis and necroptosis demonstrate abnormal levels. The EMT and glycolysis in PC cells enhance to ensure their metastasis and proliferation. Furthermore, such abnormal changes have been used to develop corresponding pharmacological and nanotechnological therapeutics for PC.
Collapse
Affiliation(s)
- Ming Gu
- Department of Breast Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, 110001, China
| | - Yang Liu
- Department of Urology, The First Hospital of China Medical University, Shenyang, Liaoning, 110001, China
| | - Peng Xin
- Department of Urology, The First Hospital of China Medical University, Shenyang, Liaoning, 110001, China
| | - Wei Guo
- Department of Pancreatic-Biliary Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, 110001, China
| | - Zimo Zhao
- Department of Pancreatic-Biliary Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, 110001, China
| | - Xu Yang
- Department of Pancreatic-Biliary Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, 110001, China
| | - Ruiyang Ma
- Department of Otorhinolaryngology, The First Hospital of China Medical University, Shenyang, Liaoning, 110001, China.
| | - Taiwei Jiao
- Department of Gastroenterology and Endoscopy, The First Hospital of China Medical University, Shenyang, Liaoning, 110001, China.
| | - Wenhui Zheng
- Department of Anesthesiology, The Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110001, China.
| |
Collapse
|
17
|
Alseud K, Ostlund T, Durymanov M, Reineke J, Halaweish F. Synthesis and biological activity of 11-Oxygenated and heterocyclic estrone analogs in pancreatic cancer monolayers and 3D spheroids. Bioorg Med Chem 2024; 103:117678. [PMID: 38489997 DOI: 10.1016/j.bmc.2024.117678] [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: 01/25/2024] [Revised: 03/01/2024] [Accepted: 03/04/2024] [Indexed: 03/17/2024]
Abstract
Pancreatic Ductal Adenocarcinoma (PDAC), representing over 90 % of pancreatic cancer diagnoses, is an aggressive disease with survivability among the worst of all cancers due to its difficulty in detection and its high metastatic properties. Current therapies for PDAC show limited success at extending life expectancies, primarily due to cancer resistance and lack of patient-specific targeted therapies. This work highlights the design and evaluation of estrone-derived analogs with both heterocyclic side-chain functionality and 11-oxygenated functionality for use in pancreatic cancer. First-round heterocyclic analogs show preliminary promise in AsPC-1 and Panc-1 cell lines, with IC50 values as low as 10.16 ± 0.83 µM. Their success, coupled with design choices from other studies, led to the synthesis of novel 11-hydroxyl and 11-keto estrone analogs that show potent in-vitro toxicity against various pancreatic cancer models. The three most cytotoxic analogs, KA1, KA2, and KA9 demonstrated low micromolar activities in both MTT and CellTiter assays in three pancreatic cancer cell lines: AsPC-1, Panc-1, and BxPC-3, as well as in a co-culture of Panc-1 and pancreatic stellate cells. IC50 values for KA9 (4.17 ± 0.90, 5.28 ± 1.87, and 5.70 ± 0.65 µM respectively) shows consistency in all cell lines tested. KA9 is also able to cause an increase in caspases 3 and 7 activity, key markers for apoptosis, at non-cytotoxic concentrations. Additional work was performed by generating 3D pancreatic cancer spheroids to better modulate the pancreatic tumor microenvironment, and KA9 continued to show the best IC50 values (21.0 and 24.3 µM) in both cell types tested. KA9 was also able to prevent the growth of spheroids whereas the standard chemotherapy, Gemcitabine, could not, suggesting that it may be a potent analog for future development of treatments. Molecular dynamic simulations were also performed to confirm biological findings and uncovered that KA9's preferential binding location is in the active site pocket of key proteins involved in cytotoxicity.
Collapse
Affiliation(s)
- Khaled Alseud
- Department of Chemistry and Biochemistry, South Dakota State University, Brookings, SD 57006, USA; Department of Pharmaceutical Chemistry, College of Pharmacy, King Khalid University, Abha 61421, Saudi Arabia
| | - Trevor Ostlund
- Department of Chemistry and Biochemistry, South Dakota State University, Brookings, SD 57006, USA
| | - Mikhail Durymanov
- Department of Pharmaceutical Sciences, South Dakota State University, Brookings, SD 57006, USA
| | - Joshua Reineke
- Department of Pharmaceutical Sciences, South Dakota State University, Brookings, SD 57006, USA
| | - Fathi Halaweish
- Department of Chemistry and Biochemistry, South Dakota State University, Brookings, SD 57006, USA.
| |
Collapse
|
18
|
Giri SS, Tripathi AS, Erkekoğlu P, Zaki MEA. Molecular pathway of pancreatic cancer-associated neuropathic pain. J Biochem Mol Toxicol 2024; 38:e23638. [PMID: 38613466 DOI: 10.1002/jbt.23638] [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: 06/09/2023] [Revised: 11/29/2023] [Accepted: 12/21/2023] [Indexed: 04/15/2024]
Abstract
The pancreas is a heterocrine gland that has both exocrine and endocrine parts. Most pancreatic cancer begins in the cells that line the ducts of the pancreas and is called pancreatic ductal adenocarcinoma (PDAC). PDAC is the most encountered pancreatic cancer type. One of the most important characteristic features of PDAC is neuropathy which is primarily due to perineural invasion (PNI). PNI develops tumor microenvironment which includes overexpression of fibroblasts cells, macrophages, as well as angiogenesis which can be responsible for neuropathy pain. In tumor microenvironment inactive fibroblasts are converted into an active form that is cancer-associated fibroblasts (CAFs). Neurotrophins they also increase the level of Substance P, calcitonin gene-related peptide which is also involved in pain. Matrix metalloproteases are the zinc-associated proteases enzymes which activates proinflammatory interleukin-1β into its activated form and are responsible for release and activation of Substance P which is responsible for neuropathic pain by transmitting pain signal via dorsal root ganglion. All the molecules and their role in being responsible for neuropathic pain are described below.
Collapse
Affiliation(s)
| | - Alok Shiomurti Tripathi
- Department of Pharmacology, Era College of Pharmacy, Era University, Lucknow, Uttar Pradesh, India
| | - Pınar Erkekoğlu
- Department of Pharmaceutical Toxicology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Magdi E A Zaki
- Department of Chemistry, Faculty of Science, Imam Mohammad lbn Saud Islamic University, Riyadh, Saudi Arabia
| |
Collapse
|
19
|
Stefanoudakis D, Frountzas M, Schizas D, Michalopoulos NV, Drakaki A, Toutouzas KG. Significance of TP53, CDKN2A, SMAD4 and KRAS in Pancreatic Cancer. Curr Issues Mol Biol 2024; 46:2827-2844. [PMID: 38666907 PMCID: PMC11049225 DOI: 10.3390/cimb46040177] [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: 02/28/2024] [Revised: 03/16/2024] [Accepted: 03/21/2024] [Indexed: 04/28/2024] Open
Abstract
The present review demonstrates the major tumor suppressor genes, including TP53, CDKN2A and SMAD4, associated with pancreatic cancer. Each gene's role, prevalence and impact on tumor development and progression are analyzed, focusing on the intricate molecular landscape of pancreatic cancer. In addition, this review underscores the prognostic significance of specific mutations, such as loss of TP53, and explores some potential targeted therapies tailored to these molecular signatures. The findings highlight the importance of genomic analyses for risk assessment, early detection and the design of personalized treatment approaches in pancreatic cancer. Overall, this review provides a comprehensive analysis of the molecular intricacies of pancreatic tumors, paving the way for more effective and tailored therapeutic interventions.
Collapse
Affiliation(s)
- Dimitrios Stefanoudakis
- First Propaedeutic Department of Surgery, Hippocration General Hospital, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece; (D.S.); (N.V.M.)
| | - Maximos Frountzas
- First Propaedeutic Department of Surgery, Hippocration General Hospital, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece; (D.S.); (N.V.M.)
| | - Dimitrios Schizas
- First Department of Surgery, Laikon General Hospital, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Nikolaos V. Michalopoulos
- First Propaedeutic Department of Surgery, Hippocration General Hospital, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece; (D.S.); (N.V.M.)
| | - Alexandra Drakaki
- Division of Hematology and Oncology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Konstantinos G. Toutouzas
- First Propaedeutic Department of Surgery, Hippocration General Hospital, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece; (D.S.); (N.V.M.)
| |
Collapse
|
20
|
Grygoryev D, Ekstrom T, Manalo E, Link JM, Alshaikh A, Keith D, Allen-Petersen BL, Sheppard B, Morgan T, Soufi A, Sears RC, Kim J. Sendai virus is robust and consistent in delivering genes into human pancreatic cancer cells. Heliyon 2024; 10:e27221. [PMID: 38463758 PMCID: PMC10923719 DOI: 10.1016/j.heliyon.2024.e27221] [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: 07/02/2023] [Revised: 02/22/2024] [Accepted: 02/26/2024] [Indexed: 03/12/2024] Open
Abstract
Background Pancreatic ductal adenocarcinoma (PDAC) is a highly intratumorally heterogeneous disease that includes several subtypes and is highly plastic. Effective gene delivery to all PDAC cells is essential for modulating gene expression and identifying potential gene-based therapeutic targets in PDAC. Most current gene delivery systems for pancreatic cells are optimized for islet or acinar cells. Lentiviral vectors are the current main gene delivery vectors for PDAC, but their transduction efficiencies vary depending on pancreatic cell type, and are especially poor for the classical subtype of PDAC cells from both primary tumors and cell lines. Methods We systemically compare transduction efficiencies of glycoprotein G of vesicular stomatitis virus (VSV-G)-pseudotyped lentiviral and Sendai viral vectors in human normal pancreatic ductal and PDAC cells. Results We find that the Sendai viral vector gives the most robust gene delivery efficiency regardless of PDAC cell type. Therefore, we propose using Sendai viral vectors to transduce ectopic genes into PDAC cells.
Collapse
Affiliation(s)
- Dmytro Grygoryev
- Cancer Early Detection Advanced Research Center at Knight Cancer Institute, Oregon Health & Science University School of Medicine, USA
| | - Taelor Ekstrom
- Cancer Early Detection Advanced Research Center at Knight Cancer Institute, Oregon Health & Science University School of Medicine, USA
| | - Elise Manalo
- Cancer Early Detection Advanced Research Center at Knight Cancer Institute, Oregon Health & Science University School of Medicine, USA
| | - Jason M. Link
- Department of Molecular and Medical Genetics, Oregon Health & Science University School of Medicine, USA
- Brenden-Colson Center for Pancreatic Care, Oregon Health & Science University School of Medicine, USA
| | - Amani Alshaikh
- The University of Edinburgh, Centre for Regenerative Medicine, Institute of Regeneration and Repair, Institute of Stem Cell Research, Edinburgh, UK
- King Abdulaziz City for Science and Technology, Health Sector (KACST), Riyadh, Saudi Arabia
| | - Dove Keith
- Brenden-Colson Center for Pancreatic Care, Oregon Health & Science University School of Medicine, USA
| | - Brittany L. Allen-Petersen
- Department of Molecular and Medical Genetics, Oregon Health & Science University School of Medicine, USA
- Brenden-Colson Center for Pancreatic Care, Oregon Health & Science University School of Medicine, USA
| | - Brett Sheppard
- Brenden-Colson Center for Pancreatic Care, Oregon Health & Science University School of Medicine, USA
- Department of Surgery, Oregon Health & Science University School of Medicine, USA
| | - Terry Morgan
- Cancer Early Detection Advanced Research Center at Knight Cancer Institute, Oregon Health & Science University School of Medicine, USA
- Department of Pathology, Oregon Health & Science University School of Medicine, USA
- Cancer Biology Research Program, Knight Cancer Institute, Oregon Health & Science University School of Medicine, Portland, OR, 97201, USA
| | - Abdenour Soufi
- The University of Edinburgh, Centre for Regenerative Medicine, Institute of Regeneration and Repair, Institute of Stem Cell Research, Edinburgh, UK
| | - Rosalie C. Sears
- Department of Molecular and Medical Genetics, Oregon Health & Science University School of Medicine, USA
- Brenden-Colson Center for Pancreatic Care, Oregon Health & Science University School of Medicine, USA
- Cancer Biology Research Program, Knight Cancer Institute, Oregon Health & Science University School of Medicine, Portland, OR, 97201, USA
| | - Jungsun Kim
- Cancer Early Detection Advanced Research Center at Knight Cancer Institute, Oregon Health & Science University School of Medicine, USA
- Department of Molecular and Medical Genetics, Oregon Health & Science University School of Medicine, USA
- Cancer Biology Research Program, Knight Cancer Institute, Oregon Health & Science University School of Medicine, Portland, OR, 97201, USA
| |
Collapse
|
21
|
Tindall RR, Bailey-Lundberg JM, Cao Y, Ko TC. The TGF-β superfamily as potential therapeutic targets in pancreatic cancer. Front Oncol 2024; 14:1362247. [PMID: 38500662 PMCID: PMC10944957 DOI: 10.3389/fonc.2024.1362247] [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: 12/28/2023] [Accepted: 02/15/2024] [Indexed: 03/20/2024] Open
Abstract
The transforming growth factor (TGF)-β superfamily has important physiologic roles and is dysregulated in many pathologic processes, including pancreatic cancer. Pancreatic cancer is one of the most lethal cancer diagnoses, and current therapies are largely ineffective due to tumor resistance and late-stage diagnosis with poor prognosis. Recent efforts are focused on the potential of immunotherapies in improving therapeutic results for patients with pancreatic cancer, among which TGF-β has been identified as a promising target. This review focuses on the role of TGF-β in the diseased pancreas and pancreatic cancer. It also aims to summarize the current status of therapies targeting the TGF-β superfamily and postulate potential future directions in targeting the TGF-β signaling pathways.
Collapse
Affiliation(s)
- Rachel R. Tindall
- McGovern Medical School, Department of Surgery, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Jennifer M. Bailey-Lundberg
- McGovern Medical School, Department of Anesthesiology, Critical Care, and Pain Medicine, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Yanna Cao
- McGovern Medical School, Department of Surgery, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Tien C. Ko
- McGovern Medical School, Department of Surgery, The University of Texas Health Science Center at Houston, Houston, TX, United States
| |
Collapse
|
22
|
Jiménez DJ, Javed A, Rubio-Tomás T, Seye-Loum N, Barceló C. Clinical and Preclinical Targeting of Oncogenic Pathways in PDAC: Targeted Therapeutic Approaches for the Deadliest Cancer. Int J Mol Sci 2024; 25:2860. [PMID: 38474109 DOI: 10.3390/ijms25052860] [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: 12/21/2023] [Revised: 01/22/2024] [Accepted: 01/30/2024] [Indexed: 03/14/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the leading causes of cancer-related death worldwide. It is commonly diagnosed in advanced stages and therapeutic interventions are typically constrained to systemic chemotherapy, which yields only modest clinical outcomes. In this review, we examine recent developments in targeted therapy tailored to address distinct molecular pathway alteration required for PDAC. Our review delineates the principal signaling pathways and molecular mechanisms implicated in the initiation and progression of PDAC. Subsequently, we provide an overview of prevailing guidelines, ongoing investigations, and prospective research trajectories related to targeted therapeutic interventions, drawing insights from randomized clinical trials and other pertinent studies. This review focus on a comprehensive examination of preclinical and clinical data substantiating the efficacy of these therapeutic modalities, emphasizing the potential of combinatorial regimens and novel therapies to enhance the quality of life for individuals afflicted with PDAC. Lastly, the review delves into the contemporary application and ongoing research endeavors concerning targeted therapy for PDAC. This synthesis serves to bridge the molecular elucidation of PDAC with its clinical implications, the evolution of innovative therapeutic strategies, and the changing landscape of treatment approaches.
Collapse
Affiliation(s)
- Diego J Jiménez
- Translational Pancreatic Cancer Oncogenesis Group, Health Research Institute of the Balearic Islands (IdISBa), Hospital Universitari Son Espases, 07120 Palma de Mallorca, Spain
| | - Aadil Javed
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Teresa Rubio-Tomás
- School of Medicine, University of Crete, 70013 Herakleion, Crete, Greece
| | - Ndioba Seye-Loum
- Translational Pancreatic Cancer Oncogenesis Group, Health Research Institute of the Balearic Islands (IdISBa), Hospital Universitari Son Espases, 07120 Palma de Mallorca, Spain
| | - Carles Barceló
- Translational Pancreatic Cancer Oncogenesis Group, Health Research Institute of the Balearic Islands (IdISBa), Hospital Universitari Son Espases, 07120 Palma de Mallorca, Spain
| |
Collapse
|
23
|
Yousef A, Yousef M, Chowdhury S, Abdilleh K, Knafl M, Edelkamp P, Alfaro-Munoz K, Chacko R, Peterson J, Smaglo BG, Wolff RA, Pant S, Lee MS, Willis J, Overman M, Doss S, Matrisian L, Hurd MW, Snyder R, Katz MHG, Wang H, Maitra A, Shen JP, Zhao D. Impact of KRAS mutations and co-mutations on clinical outcomes in pancreatic ductal adenocarcinoma. NPJ Precis Oncol 2024; 8:27. [PMID: 38310130 PMCID: PMC10838312 DOI: 10.1038/s41698-024-00505-0] [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: 07/22/2023] [Accepted: 01/05/2024] [Indexed: 02/05/2024] Open
Abstract
The relevance of KRAS mutation alleles to clinical outcome remains inconclusive in pancreatic adenocarcinoma (PDAC). We conducted a retrospective study of 803 patients with PDAC (42% with metastatic disease) at MD Anderson Cancer Center. Overall survival (OS) analysis demonstrated that KRAS mutation status and subtypes were prognostic (p < 0.001). Relative to patients with KRAS wildtype tumors (median OS 38 months), patients with KRASG12R had a similar OS (median 34 months), while patients with KRASQ61 and KRASG12D mutated tumors had shorter OS (median 20 months [HR: 1.9, 95% CI 1.2-3.0, p = 0.006] and 22 months [HR: 1.7, 95% CI 1.3-2.3, p < 0.001], respectively). There was enrichment of KRASG12D mutation in metastatic tumors (34% vs 24%, OR: 1.7, 95% CI 1.2-2.4, p = 0.001) and enrichment of KRASG12R in well and moderately differentiated tumors (14% vs 9%, OR: 1.7, 95% CI 1.05-2.99, p = 0.04). Similar findings were observed in the external validation cohort (PanCAN's Know Your Tumor® dataset, n = 408).
Collapse
Affiliation(s)
- Abdelrahman Yousef
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mahmoud Yousef
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Saikat Chowdhury
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kawther Abdilleh
- Pancreatic Cancer Action Network, Manhattan Beach, Los Angeles, CA, USA
| | - Mark Knafl
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Paul Edelkamp
- Department of Data Engineering & Analytics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kristin Alfaro-Munoz
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ray Chacko
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jennifer Peterson
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Brandon G Smaglo
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Robert A Wolff
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Shubham Pant
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Michael S Lee
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jason Willis
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Michael Overman
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sudheer Doss
- Pancreatic Cancer Action Network, Manhattan Beach, Los Angeles, CA, USA
| | - Lynn Matrisian
- Pancreatic Cancer Action Network, Manhattan Beach, Los Angeles, CA, USA
| | - Mark W Hurd
- Sheikh Ahmed Center for Pancreatic Cancer Research, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Rebecca Snyder
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Matthew H G Katz
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Huamin Wang
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Anirban Maitra
- Sheikh Ahmed Center for Pancreatic Cancer Research, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - John Paul Shen
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Dan Zhao
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
24
|
Xu J, Roe J, Lee E, Tonelli C, Ji KY, Younis OW, Somervile TD, Yao M, Milazzo JP, Tiriac H, Kolarzyk AM, Lee E, Grem JL, Lazenby AJ, Grunkemeyer JA, Hollingsworth MA, Grandgenett PM, Borowsky AD, Park Y, Vakoc CR, Tuveson DA, Hwang C. Engrailed-1 Promotes Pancreatic Cancer Metastasis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2308537. [PMID: 38110836 PMCID: PMC10853725 DOI: 10.1002/advs.202308537] [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: 11/08/2023] [Indexed: 12/20/2023]
Abstract
Engrailed-1 (EN1) is a critical homeodomain transcription factor (TF) required for neuronal survival, and EN1 expression has been shown to promote aggressive forms of triple negative breast cancer. Here, it is reported that EN1 is aberrantly expressed in a subset of pancreatic ductal adenocarcinoma (PDA) patients with poor outcomes. EN1 predominantly repressed its target genes through direct binding to gene enhancers and promoters, implicating roles in the activation of MAPK pathways and the acquisition of mesenchymal cell properties. Gain- and loss-of-function experiments demonstrated that EN1 promoted PDA transformation and metastasis in vitro and in vivo. The findings nominate the targeting of EN1 and downstream pathways in aggressive PDA.
Collapse
Affiliation(s)
- Jihao Xu
- Department of Microbiology and Molecular GeneticsUniversity of California DavisDavisCA95616USA
- Comprehensive Cancer CenterUniversity of California DavisSacramentoCA95817USA
| | - Jae‐Seok Roe
- Department of BiochemistryYonsei UniversitySeoul03722South Korea
- Cold Spring Harbor LaboratoryCold Spring HarborNY11724USA
| | - EunJung Lee
- Department of Microbiology and Molecular GeneticsUniversity of California DavisDavisCA95616USA
- Cold Spring Harbor LaboratoryCold Spring HarborNY11724USA
- Lustgarten Foundation Pancreatic Cancer Research LaboratoryCold Spring HarborNY11724USA
| | - Claudia Tonelli
- Cold Spring Harbor LaboratoryCold Spring HarborNY11724USA
- Lustgarten Foundation Pancreatic Cancer Research LaboratoryCold Spring HarborNY11724USA
| | - Keely Y. Ji
- Department of Microbiology and Molecular GeneticsUniversity of California DavisDavisCA95616USA
| | - Omar W. Younis
- Department of Microbiology and Molecular GeneticsUniversity of California DavisDavisCA95616USA
| | | | - Melissa Yao
- Cold Spring Harbor LaboratoryCold Spring HarborNY11724USA
- Lustgarten Foundation Pancreatic Cancer Research LaboratoryCold Spring HarborNY11724USA
| | | | - Herve Tiriac
- Cold Spring Harbor LaboratoryCold Spring HarborNY11724USA
- Lustgarten Foundation Pancreatic Cancer Research LaboratoryCold Spring HarborNY11724USA
| | - Anna M. Kolarzyk
- Nancy E. and Peter C. Meinig School of Biomedical EngineeringCornell UniversityIthacaNY14853USA
| | - Esak Lee
- Nancy E. and Peter C. Meinig School of Biomedical EngineeringCornell UniversityIthacaNY14853USA
| | - Jean L. Grem
- Department of MedicineUniversity of Nebraska Medical CenterOmahaNE68198USA
| | - Audrey J. Lazenby
- Department of MedicineUniversity of Nebraska Medical CenterOmahaNE68198USA
| | | | | | | | - Alexander D. Borowsky
- Department of PathologySchool of MedicineUniversity of California DavisSacramentoCA95817USA
| | - Youngkyu Park
- Cold Spring Harbor LaboratoryCold Spring HarborNY11724USA
- Lustgarten Foundation Pancreatic Cancer Research LaboratoryCold Spring HarborNY11724USA
| | | | - David A. Tuveson
- Cold Spring Harbor LaboratoryCold Spring HarborNY11724USA
- Lustgarten Foundation Pancreatic Cancer Research LaboratoryCold Spring HarborNY11724USA
| | - Chang‐Il Hwang
- Department of Microbiology and Molecular GeneticsUniversity of California DavisDavisCA95616USA
- Comprehensive Cancer CenterUniversity of California DavisSacramentoCA95817USA
| |
Collapse
|
25
|
Ramírez-Maldonado E, López Gordo S, Major Branco RP, Pavel MC, Estalella L, Llàcer-Millán E, Guerrero MA, López-Gordo E, Memba R, Jorba R. Clinical Application of Liquid Biopsy in Pancreatic Cancer: A Narrative Review. Int J Mol Sci 2024; 25:1640. [PMID: 38338919 PMCID: PMC10855073 DOI: 10.3390/ijms25031640] [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: 12/07/2023] [Revised: 01/21/2024] [Accepted: 01/24/2024] [Indexed: 02/12/2024] Open
Abstract
Pancreatic ductal adenocarcinoma contributes significantly to global cancer-related deaths, featuring only a 10% survival rate over five years. The quest for novel tumor markers is critical to facilitate early diagnosis and tailor treatment strategies for this disease, which is key to improving patient outcomes. In pancreatic ductal adenocarcinoma, these markers have been demonstrated to play a crucial role in early identification, continuous monitoring, and prediction of its prognosis and have led to better patient outcomes. Nowadays, biopsy specimens serve to ascertain diagnosis and determine tumor type. However, liquid biopsies present distinct advantages over conventional biopsy techniques. They offer a noninvasive, easily administered procedure, delivering insights into the tumor's status and facilitating real-time monitoring. Liquid biopsies encompass a variety of elements, such as circulating tumor cells, circulating tumor DNA, extracellular vesicles, microRNAs, circulating RNA, tumor platelets, and tumor endothelial cells. This review aims to provide an overview of the clinical applications of liquid biopsy as a technique in the management of pancreatic cancer.
Collapse
Affiliation(s)
- Elena Ramírez-Maldonado
- HBP Unit, General Surgery Department, Joan XXIII University Hospital, 43005 Tarragona, Spain; (M.-C.P.); (L.E.); (M.A.G.); (R.M.); (R.J.)
- Medicine and Surgery Department, Rovira i Virgili University, 43204 Reus, Spain
| | - Sandra López Gordo
- General Surgery Department, Maresme Health Consortium, 08304 Mataro, Spain;
| | | | - Mihai-Calin Pavel
- HBP Unit, General Surgery Department, Joan XXIII University Hospital, 43005 Tarragona, Spain; (M.-C.P.); (L.E.); (M.A.G.); (R.M.); (R.J.)
- Medicine and Surgery Department, Rovira i Virgili University, 43204 Reus, Spain
| | - Laia Estalella
- HBP Unit, General Surgery Department, Joan XXIII University Hospital, 43005 Tarragona, Spain; (M.-C.P.); (L.E.); (M.A.G.); (R.M.); (R.J.)
- Medicine and Surgery Department, Rovira i Virgili University, 43204 Reus, Spain
| | - Erik Llàcer-Millán
- HBP Unit, General Surgery Department, Joan XXIII University Hospital, 43005 Tarragona, Spain; (M.-C.P.); (L.E.); (M.A.G.); (R.M.); (R.J.)
- Medicine and Surgery Department, Rovira i Virgili University, 43204 Reus, Spain
| | - María Alejandra Guerrero
- HBP Unit, General Surgery Department, Joan XXIII University Hospital, 43005 Tarragona, Spain; (M.-C.P.); (L.E.); (M.A.G.); (R.M.); (R.J.)
- Medicine and Surgery Department, Rovira i Virgili University, 43204 Reus, Spain
| | | | - Robert Memba
- HBP Unit, General Surgery Department, Joan XXIII University Hospital, 43005 Tarragona, Spain; (M.-C.P.); (L.E.); (M.A.G.); (R.M.); (R.J.)
- Medicine and Surgery Department, Rovira i Virgili University, 43204 Reus, Spain
| | - Rosa Jorba
- HBP Unit, General Surgery Department, Joan XXIII University Hospital, 43005 Tarragona, Spain; (M.-C.P.); (L.E.); (M.A.G.); (R.M.); (R.J.)
- Medicine and Surgery Department, Rovira i Virgili University, 43204 Reus, Spain
| |
Collapse
|
26
|
Oketch DJA, Giulietti M, Piva F. Copy Number Variations in Pancreatic Cancer: From Biological Significance to Clinical Utility. Int J Mol Sci 2023; 25:391. [PMID: 38203561 PMCID: PMC10779192 DOI: 10.3390/ijms25010391] [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: 11/24/2023] [Revised: 12/20/2023] [Accepted: 12/24/2023] [Indexed: 01/12/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the most common type of pancreatic cancer, characterized by high tumor heterogeneity and a poor prognosis. Inter- and intra-tumoral heterogeneity in PDAC is a major obstacle to effective PDAC treatment; therefore, it is highly desirable to explore the tumor heterogeneity and underlying mechanisms for the improvement of PDAC prognosis. Gene copy number variations (CNVs) are increasingly recognized as a common and heritable source of inter-individual variation in genomic sequence. In this review, we outline the origin, main characteristics, and pathological aspects of CNVs. We then describe the occurrence of CNVs in PDAC, including those that have been clearly shown to have a pathogenic role, and further highlight some key examples of their involvement in tumor development and progression. The ability to efficiently identify and analyze CNVs in tumor samples is important to support translational research and foster precision oncology, as copy number variants can be utilized to guide clinical decisions. We provide insights into understanding the CNV landscapes and the role of both somatic and germline CNVs in PDAC, which could lead to significant advances in diagnosis, prognosis, and treatment. Although there has been significant progress in this field, understanding the full contribution of CNVs to the genetic basis of PDAC will require further research, with more accurate CNV assays such as single-cell techniques and larger cohorts than have been performed to date.
Collapse
Affiliation(s)
| | - Matteo Giulietti
- Department of Specialistic Clinical and Odontostomatological Sciences, Polytechnic University of Marche, 60131 Ancona, Italy
| | - Francesco Piva
- Department of Specialistic Clinical and Odontostomatological Sciences, Polytechnic University of Marche, 60131 Ancona, Italy
| |
Collapse
|
27
|
Ansardamavandi A, Nikfarjam M, He H. PAK in Pancreatic Cancer-Associated Vasculature: Implications for Therapeutic Response. Cells 2023; 12:2692. [PMID: 38067120 PMCID: PMC10705971 DOI: 10.3390/cells12232692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 11/20/2023] [Accepted: 11/22/2023] [Indexed: 12/18/2023] Open
Abstract
Angiogenesis has been associated with numbers of solid tumours. Anti-angiogenesis drugs starve tumours of nutrients and oxygen but also make it difficult for a chemo reagent to distribute into a tumour, leading to aggressive tumour growth. Anti-angiogenesis drugs do not appear to improve the overall survival rate of pancreatic cancer. Vessel normalisation is merging as one of the new approaches for halting tumour progression by facilitating the tumour infiltration of immune cells and the delivery of chemo reagents. Targeting p21-activated kinases (PAKs) in cancer has been shown to inhibit cancer cell growth and improve the efficacy of chemotherapy. Inhibition of PAK enhances anti-tumour immunity and stimulates the efficacy of immune checkpoint blockades. Inhibition of PAK also improves Car-T immunotherapy by reprogramming the vascular microenvironment. This review summarizes current research on PAK's role in tumour vasculature and therapeutical response, with a focus on pancreatic cancer.
Collapse
Affiliation(s)
- Arian Ansardamavandi
- Department of Surgery, Austin Precinct, The University of Melbourne, 145 Studley Rd, Heidelberg, VIC 3084, Australia; (A.A.); (M.N.)
| | - Mehrdad Nikfarjam
- Department of Surgery, Austin Precinct, The University of Melbourne, 145 Studley Rd, Heidelberg, VIC 3084, Australia; (A.A.); (M.N.)
- Department of Hepatopancreatic-Biliary Surgery, Austin Health, 145 Studley Rd, Heidelberg, VIC 3084, Australia
| | - Hong He
- Department of Surgery, Austin Precinct, The University of Melbourne, 145 Studley Rd, Heidelberg, VIC 3084, Australia; (A.A.); (M.N.)
| |
Collapse
|
28
|
Vassilenko V, Moura PC, Raposo M. Diagnosis of Carcinogenic Pathologies through Breath Biomarkers: Present and Future Trends. Biomedicines 2023; 11:3029. [PMID: 38002028 PMCID: PMC10669878 DOI: 10.3390/biomedicines11113029] [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: 10/04/2023] [Revised: 11/08/2023] [Accepted: 11/09/2023] [Indexed: 11/26/2023] Open
Abstract
The assessment of volatile breath biomarkers has been targeted with a lot of interest by the scientific and medical communities during the past decades due to their suitability for an accurate, painless, non-invasive, and rapid diagnosis of health states and pathological conditions. This paper reviews the most relevant bibliographic sources aiming to gather the most pertinent volatile organic compounds (VOCs) already identified as putative cancer biomarkers. Here, a total of 265 VOCs and the respective bibliographic sources are addressed regarding their scientifically proven suitability to diagnose a total of six carcinogenic diseases, namely lung, breast, gastric, colorectal, prostate, and squamous cell (oesophageal and laryngeal) cancers. In addition, future trends in the identification of five other forms of cancer, such as bladder, liver, ovarian, pancreatic, and thyroid cancer, through perspective volatile breath biomarkers are equally presented and discussed. All the results already achieved in the detection, identification, and quantification of endogenous metabolites produced by all kinds of normal and abnormal processes in the human body denote a promising and auspicious future for this alternative diagnostic tool, whose future passes by the development and employment of newer and more accurate collection and analysis techniques, and the certification for utilisation in real clinical scenarios.
Collapse
Affiliation(s)
- Valentina Vassilenko
- Laboratory for Instrumentation, Biomedical Engineering and Radiation Physics (LIBPhys-UNL), Department of Physics, NOVA School of Science and Technology, NOVA University of Lisbon, Campus FCT-UNL, 2829-516 Caparica, Portugal;
| | - Pedro Catalão Moura
- Laboratory for Instrumentation, Biomedical Engineering and Radiation Physics (LIBPhys-UNL), Department of Physics, NOVA School of Science and Technology, NOVA University of Lisbon, Campus FCT-UNL, 2829-516 Caparica, Portugal;
| | | |
Collapse
|
29
|
Shields MA, Metropulos AE, Spaulding C, Hirose T, Ohno S, Pham TN, Munshi HG. BET inhibition rescues ciliogenesis and ameliorates pancreatitis-driven phenotypic changes in mice with Par3 loss. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.14.557654. [PMID: 37745543 PMCID: PMC10515915 DOI: 10.1101/2023.09.14.557654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
The apical-basal polarity of pancreatic acinar cells is essential for maintaining tissue architecture. However, the mechanisms by which polarity proteins regulate acinar pancreas tissue homeostasis are poorly understood. Here, we evaluate the role of Par3 in acinar pancreas injury and homeostasis. While Par3 loss in the mouse pancreas disrupts tight junctions, Par3 loss is dispensable for pancreatogenesis. However, with aging, Par3 loss results in low-grade inflammation, acinar degeneration, and pancreatic lipomatosis. Par3 loss also exacerbates pancreatitis-induced acinar cell loss, resulting in pronounced pancreatic lipomatosis and failure to regenerate. Moreover, Par3 loss in mice harboring mutant Kras causes extensive pancreatic intraepithelial neoplastic (PanIN) lesions and large pancreatic cysts. We also show that Par3 loss restricts injury-induced primary ciliogenesis. Significantly, targeting BET proteins enhances primary ciliogenesis during pancreatitis-induced injury and, in mice with Par3 loss, limits pancreatitis-induced acinar loss and facilitates acinar cell regeneration. Combined, this study demonstrates how Par3 restrains pancreatitis- and Kras-induced changes in the pancreas and identifies a potential role for BET inhibitors to attenuate pancreas injury and facilitate pancreas tissue regeneration.
Collapse
Affiliation(s)
- Mario A. Shields
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- The Robert H. Lurie Comprehensive Cancer Center, Chicago, IL, USA
| | - Anastasia E. Metropulos
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Jesse Brown VA Medical Center, Chicago, IL, USA
| | - Christina Spaulding
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Jesse Brown VA Medical Center, Chicago, IL, USA
| | - Tomonori Hirose
- Department of Molecular Biology, Yokohama City University School of Medicine, Yokohama, Japan
- Department of Cell Biology, Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Shigeo Ohno
- Department of Molecular Biology, Yokohama City University School of Medicine, Yokohama, Japan
| | - Thao N.D. Pham
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- The Robert H. Lurie Comprehensive Cancer Center, Chicago, IL, USA
| | - Hidayatullah G. Munshi
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- The Robert H. Lurie Comprehensive Cancer Center, Chicago, IL, USA
- Jesse Brown VA Medical Center, Chicago, IL, USA
| |
Collapse
|
30
|
Gao H, Yin J, Guan X, Zhang S, Peng S, Liu X, Xing F. CMTM6 as a potential therapy target is associated with immunological tumor microenvironment and can promote migration and invasion in pancreatic adenocarcinoma. Funct Integr Genomics 2023; 23:306. [PMID: 37726578 PMCID: PMC10509136 DOI: 10.1007/s10142-023-01235-5] [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: 04/12/2023] [Revised: 08/31/2023] [Accepted: 09/13/2023] [Indexed: 09/21/2023]
Abstract
CMTM6 has been connected to the development of several malignancies. However, it is still unknown what function CMTM6 serves in pancreatic adenocarcinoma (PAAD). We obtained RNA sequencing information of PAAD from public datasets and predicted statistical significance of CMTM6 survival in accordance with Kaplan-Meier curves. Gene set enrichment assessment (GSEA) was employed to analyze changes in pathways. Then, we systematically investigated the association involving CMTM6 and the immunological traits within the tumor microenvironment (TME) of PAAD, including immune pathways, immunomodulators, immune infiltrating cells, inflammatory activities, and immunotherapy response prediction. To demonstrate the biologically malignant properties of CMTM6 expression, the Cell Counting Kit-8, transwell experiments, colony formation, and wound healing were utilized. Upregulated CMTM6 expression was revealed within PAAD tissues, which was associated with more frequent somatic mutations and worse survival outcomes. Specifically, CMTM6 expression represented stronger immune infiltration, inflammatory activity, and better immunotherapeutic response in TME. Functional studies revealed that CMTM6 promoted the ability to proliferate, migrate, and invade. Additionally, CMTM6 and PD-L1 had a positive relationship, and CMTM6 can co-immunocoprecipitate with PD-L1 protein in pancreatic cell lines. CMTM6 overexpression shapes the inflammatory TME with a strong immune response. These findings support that CMTM6 is an immunotherapeutic target with promising effect to treat PAAD.
Collapse
Affiliation(s)
- Hongli Gao
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Jianqiao Yin
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Xin Guan
- Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Shuang Zhang
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Songlin Peng
- Department of General Surgery, the Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
| | - Xun Liu
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Fei Xing
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110004, China.
| |
Collapse
|
31
|
Massagué J, Sheppard D. TGF-β signaling in health and disease. Cell 2023; 186:4007-4037. [PMID: 37714133 PMCID: PMC10772989 DOI: 10.1016/j.cell.2023.07.036] [Citation(s) in RCA: 191] [Impact Index Per Article: 95.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 07/21/2023] [Accepted: 07/28/2023] [Indexed: 09/17/2023]
Abstract
The TGF-β regulatory system plays crucial roles in the preservation of organismal integrity. TGF-β signaling controls metazoan embryo development, tissue homeostasis, and injury repair through coordinated effects on cell proliferation, phenotypic plasticity, migration, metabolic adaptation, and immune surveillance of multiple cell types in shared ecosystems. Defects of TGF-β signaling, particularly in epithelial cells, tissue fibroblasts, and immune cells, disrupt immune tolerance, promote inflammation, underlie the pathogenesis of fibrosis and cancer, and contribute to the resistance of these diseases to treatment. Here, we review how TGF-β coordinates multicellular response programs in health and disease and how this knowledge can be leveraged to develop treatments for diseases of the TGF-β system.
Collapse
Affiliation(s)
- Joan Massagué
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| | - Dean Sheppard
- Department of Medicine and Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94158, USA
| |
Collapse
|
32
|
Jan IS, Ch'ang HJ. Selection of patients with pancreatic adenocarcinoma who may benefit from radiotherapy. Radiat Oncol 2023; 18:137. [PMID: 37596627 PMCID: PMC10439654 DOI: 10.1186/s13014-023-02328-y] [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: 03/28/2023] [Accepted: 08/03/2023] [Indexed: 08/20/2023] Open
Abstract
Despite combination chemotherapy demonstrating a positive effect on survival, the clinical outcomes of pancreatic adenocarcinoma (PDAC) remain poor. Radiotherapy was previously a component of the curative treatment of PDAC. Advances in imaging and computer sciences have enabled the prescription of higher dosage of radiation focused on tumours with minimal toxicity to normal tissue. However, the role of radiotherapy has not been established in the curative treatment of localized PDAC because of the conflicting results from large prospective trials. Most studies have demonstrated improved locoregional control but no survival benefit from additional chemoradiotherapy (CRT) in addition to chemotherapy for resectable, borderline or locally advanced PDAC. The improved locoregional control enabled by CRT does not cause extended survival because of rapid distant progression in a significant proportion of patients with PDAC. Several single-institute studies of prescribing intensive chemotherapy with modern ablative radiotherapy for locally advanced PDAC have demonstrated extended survival with an acceptable safety profile. In an analysis after long-term follow-up, the PREOPANC study demonstrated a survival benefit from neoadjuvant gemcitabine-based CRT in resected PDAC relative to upfront surgery followed by adjuvant gemcitabine only. These observations indicated that the role of radiotherapy in PDAC should be evaluated in a subgroup of patients without rapid distant progression because systemic therapy for PDAC remains underdeveloped. We reviewed critical imaging, tissue, liquid and clinical biomarkers to differentiate the heterogeneous biologic spectra of patients with PDAC to identify those who may benefit the most from local radiotherapy. Exclusion of patients with localised PDAC who develop distant progression in a short time and undergo extended upfront chemotherapy for over 4 months may enable the identification of a survival benefit of local radiotherapy. Though promising, the effectiveness of biomarkers must be validated in a multi-institutional prospective study of patients with PDAC receiving CRT or not receiving CRT.
Collapse
Affiliation(s)
- I-Shiow Jan
- Department of Laboratory Medicine, College of Medicine, National Taiwan University Hospital, National Taiwan University, Taipei, Taiwan
| | - Hui Ju Ch'ang
- National Institute of Cancer Research, National Health Research Institutes, Miaoli, Taiwan.
- Program for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan.
- Department of Radiation Oncology, Taipei Medical University, Taipei, Taiwan.
- Department of Oncology, School of Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
33
|
Yousef A, Yousef M, Chowdhury S, Abdilleh K, Knafl M, Edelkamp P, Alfaro-Munoz K, Chacko R, Peterson J, Smaglo BG, Wolff RA, Pant S, Lee MS, Willis J, Overman M, Doss S, Matrisian L, Hurd MW, Snyder R, Katz MH, Wang H, Maitra A, Shen JP, Zhao D. Impact of KRAS Mutations and Co-mutations on Clinical Outcomes in Pancreatic Ductal Adenocarcinoma. RESEARCH SQUARE 2023:rs.3.rs-3195257. [PMID: 37609177 PMCID: PMC10441514 DOI: 10.21203/rs.3.rs-3195257/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/24/2023]
Abstract
The relevance of KRAS mutation alleles to clinical outcome remains inconclusive in pancreatic adenocarcinoma (PDAC). We conducted a retrospective study of 803 PDAC patients (42% with metastatic disease) at MD Anderson Cancer Center. Overall survival (OS) analysis demonstrated that KRAS mutation status and subtypes were prognostic (p<0.001). Relative to patients with KRAS wildtype tumors (median OS 38 months), patients with KRASG12R had a similar OS (median 34 months), while patients with KRASQ61 and KRASG12D mutated tumors had shorter OS (median 20 months [HR: 1.9, 95% CI 1.2-3.0, p=0.006] and 22 months [HR: 1.7, 95% CI 1.3-2.3, p<0.001], respectively). There was enrichment of KRASG12D mutation in metastatic tumors (34% vs 24%, OR: 1.7, 95% CI 1.2-2.4, p=0.001) and enrichment of KRASG12R in well and moderately differentiated tumors (14% vs 9%, OR: 1.7, 95% CI 1.05-2.99, p=0.04). Similar findings were observed in the external validation cohort (PanCAN's Know Your Tumor® dataset, n=408).
Collapse
Affiliation(s)
- Abdelrahman Yousef
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mahmoud Yousef
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Saikat Chowdhury
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kawther Abdilleh
- Pancreatic Cancer Action Network, Manhattan Beach, Los Angeles, CA, USA
| | - Mark Knafl
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Paul Edelkamp
- Department of Data Engineering & Analytics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kristin Alfaro-Munoz
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ray Chacko
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jennifer Peterson
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Brandon G. Smaglo
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Robert A. Wolff
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Shubham Pant
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Michael S. Lee
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jason Willis
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Michael Overman
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sudheer Doss
- Pancreatic Cancer Action Network, Manhattan Beach, Los Angeles, CA, USA
| | - Lynn Matrisian
- Pancreatic Cancer Action Network, Manhattan Beach, Los Angeles, CA, USA
| | - Mark W. Hurd
- Sheikh Ahmed Center for Pancreatic Cancer Research, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Rebecca Snyder
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Matthew H.G. Katz
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Huamin Wang
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Anirban Maitra
- Sheikh Ahmed Center for Pancreatic Cancer Research, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - John Paul Shen
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Dan Zhao
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
34
|
Liu H, Tsai H, Yang M, Li G, Bian Q, Ding G, Wu D, Dai J. Three-dimensional genome structure and function. MedComm (Beijing) 2023; 4:e326. [PMID: 37426677 PMCID: PMC10329473 DOI: 10.1002/mco2.326] [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: 10/19/2022] [Revised: 05/31/2023] [Accepted: 06/09/2023] [Indexed: 07/11/2023] Open
Abstract
Linear DNA undergoes a series of compression and folding events, forming various three-dimensional (3D) structural units in mammalian cells, including chromosomal territory, compartment, topologically associating domain, and chromatin loop. These structures play crucial roles in regulating gene expression, cell differentiation, and disease progression. Deciphering the principles underlying 3D genome folding and the molecular mechanisms governing cell fate determination remains a challenge. With advancements in high-throughput sequencing and imaging techniques, the hierarchical organization and functional roles of higher-order chromatin structures have been gradually illuminated. This review systematically discussed the structural hierarchy of the 3D genome, the effects and mechanisms of cis-regulatory elements interaction in the 3D genome for regulating spatiotemporally specific gene expression, the roles and mechanisms of dynamic changes in 3D chromatin conformation during embryonic development, and the pathological mechanisms of diseases such as congenital developmental abnormalities and cancer, which are attributed to alterations in 3D genome organization and aberrations in key structural proteins. Finally, prospects were made for the research about 3D genome structure, function, and genetic intervention, and the roles in disease development, prevention, and treatment, which may offer some clues for precise diagnosis and treatment of related diseases.
Collapse
Affiliation(s)
- Hao Liu
- Department of Oral and Cranio‐Maxillofacial SurgeryShanghai Ninth People's Hospital, Shanghai Jiao Tong University School of MedicineCollege of Stomatology, Shanghai Jiao Tong UniversityNational Center for StomatologyNational Clinical Research Center for Oral DiseasesShanghai Key Laboratory of StomatologyShanghaiChina
- School of StomatologyWeifang Medical UniversityWeifangChina
| | - Hsiangyu Tsai
- Department of Oral and Cranio‐Maxillofacial SurgeryShanghai Ninth People's Hospital, Shanghai Jiao Tong University School of MedicineCollege of Stomatology, Shanghai Jiao Tong UniversityNational Center for StomatologyNational Clinical Research Center for Oral DiseasesShanghai Key Laboratory of StomatologyShanghaiChina
| | - Maoquan Yang
- School of Clinical MedicineWeifang Medical UniversityWeifangChina
| | - Guozhi Li
- Department of Oral and Cranio‐Maxillofacial SurgeryShanghai Ninth People's Hospital, Shanghai Jiao Tong University School of MedicineCollege of Stomatology, Shanghai Jiao Tong UniversityNational Center for StomatologyNational Clinical Research Center for Oral DiseasesShanghai Key Laboratory of StomatologyShanghaiChina
| | - Qian Bian
- Shanghai Institute of Precision MedicineShanghaiChina
| | - Gang Ding
- School of StomatologyWeifang Medical UniversityWeifangChina
| | - Dandan Wu
- Department of Oral and Cranio‐Maxillofacial SurgeryShanghai Ninth People's Hospital, Shanghai Jiao Tong University School of MedicineCollege of Stomatology, Shanghai Jiao Tong UniversityNational Center for StomatologyNational Clinical Research Center for Oral DiseasesShanghai Key Laboratory of StomatologyShanghaiChina
| | - Jiewen Dai
- Department of Oral and Cranio‐Maxillofacial SurgeryShanghai Ninth People's Hospital, Shanghai Jiao Tong University School of MedicineCollege of Stomatology, Shanghai Jiao Tong UniversityNational Center for StomatologyNational Clinical Research Center for Oral DiseasesShanghai Key Laboratory of StomatologyShanghaiChina
| |
Collapse
|
35
|
Sarkar R, Xu Z, Perera CJ, Apte MV. Emerging role of pancreatic stellate cell-derived extracellular vesicles in pancreatic cancer. Semin Cancer Biol 2023; 93:114-122. [PMID: 37225047 DOI: 10.1016/j.semcancer.2023.05.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 04/17/2023] [Accepted: 05/19/2023] [Indexed: 05/26/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive cancer that is characterised by a prominent collagenous stromal reaction/desmoplasia surrounding tumour cells. Pancreatic stellate cells (PSCs) are responsible for the production of this stroma and have been shown to facilitate PDAC progression. Recently, extracellular vesicles (EVs), in particular, small extracellular vesicles (exosomes) have been a topic of interest in the field of cancer research for their emerging roles in cancer progression and diagnosis. EVs act as a form of intercellular communication by carrying their molecular cargo from one cell to another, regulating functions of the recipient cells. Although the knowledge of the bi-directional interactions between the PSCs and cancer cells that promote disease progression has advanced significantly over the past decade, studies on PSC-derived EVs in PDAC are currently rather limited. This review provides an overview of PDAC, pancreatic stellate cells and their interactions with cancer cells, as well as the currently known role of extracellular vesicles derived from PSCs in PDAC progression.
Collapse
Affiliation(s)
- Rohit Sarkar
- Pancreatic Research Group, South West Sydney Clinical Campuses, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Sydney 2052, Australia; Ingham Institute of Applied Medical Research, Sydney 2170, Australia
| | - Zhihong Xu
- Pancreatic Research Group, South West Sydney Clinical Campuses, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Sydney 2052, Australia; Ingham Institute of Applied Medical Research, Sydney 2170, Australia
| | - Chamini J Perera
- Pancreatic Research Group, South West Sydney Clinical Campuses, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Sydney 2052, Australia; Ingham Institute of Applied Medical Research, Sydney 2170, Australia.
| | - Minoti V Apte
- Pancreatic Research Group, South West Sydney Clinical Campuses, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Sydney 2052, Australia; Ingham Institute of Applied Medical Research, Sydney 2170, Australia
| |
Collapse
|
36
|
Pidkovka N, Belkhiri A. Altered expression of AXL receptor tyrosine kinase in gastrointestinal cancers: a promising therapeutic target. Front Oncol 2023; 13:1079041. [PMID: 37469409 PMCID: PMC10353021 DOI: 10.3389/fonc.2023.1079041] [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: 10/24/2022] [Accepted: 05/31/2023] [Indexed: 07/21/2023] Open
Abstract
Gastrointestinal (GI) cancers that include all cancers of the digestive tract organs are generally associated with obesity, lack of exercising, smoking, poor diet, and heavy alcohol consumption. Treatment of GI cancers typically involves surgery followed by chemotherapy and/or radiation. Unfortunately, intrinsic or acquired resistance to these therapies underscore the need for more effective targeted therapies that have been proven in other malignancies. The aggressive features of GI cancers share distinct signaling pathways that are connected to each other by the overexpression and activation of AXL receptor tyrosine kinase. Several preclinical and clinical studies involving anti-AXL antibodies and small molecule AXL kinase inhibitors to test their efficacy in solid tumors, including GI cancers, have been recently carried out. Therefore, AXL may be a promising therapeutic target for overcoming the shortcomings of standard therapies in GI cancers.
Collapse
Affiliation(s)
- Nataliya Pidkovka
- Department of Health Science, South College, Nashville, TN, United States
| | - Abbes Belkhiri
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, United States
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, United States
| |
Collapse
|
37
|
Haraoka Y, Miyake M, Ishitani T. Zebrafish imaging reveals hidden oncogenic-normal cell communication during primary tumorigenesis. Cell Struct Funct 2023; 48:113-121. [PMID: 37164759 PMCID: PMC10721949 DOI: 10.1247/csf.23026] [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: 03/28/2023] [Accepted: 05/08/2023] [Indexed: 05/12/2023] Open
Abstract
Oncogenic mutations drive tumorigenesis, and single cells with oncogenic mutations act as the tumor seeds that gradually evolve into fully transformed tumors. However, oncogenic cell behavior and communication with neighboring cells during primary tumorigenesis remain poorly understood. We used the zebrafish, a small vertebrate model suitable for in vivo cell biology, to address these issues. We describe the cooperative and competitive communication between oncogenic cells and neighboring cells, as revealed by our recent zebrafish imaging studies. Newly generated oncogenic cells are actively eliminated by neighboring cells in healthy epithelia, whereas oncogenic cells cooperate with their neighbors to prime tumorigenesis in unhealthy epithelia via additional mutations or inflammation. In addition, we discuss the potential of zebrafish in vivo imaging to determine the initial steps of human tumorigenesis.Key words: zebrafish, imaging, cell-cell communication, cell competition, EDAC, senescence, primary tumorigenesis.
Collapse
Affiliation(s)
- Yukinari Haraoka
- Department of Homeostatic Regulation, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan
| | - Mai Miyake
- Department of Homeostatic Regulation, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan
| | - Tohru Ishitani
- Department of Homeostatic Regulation, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan
- Center for Infectious Disease Education and Research (CiDER), Osaka University, Suita, Osaka 565-0871, Japan
| |
Collapse
|
38
|
Wang Q, Šabanović B, Awada A, Reina C, Aicher A, Tang J, Heeschen C. Single-cell omics: a new perspective for early detection of pancreatic cancer? Eur J Cancer 2023; 190:112940. [PMID: 37413845 DOI: 10.1016/j.ejca.2023.112940] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 06/04/2023] [Indexed: 07/08/2023]
Abstract
Pancreatic cancer is one of the most lethal cancers, mostly due to late diagnosis and limited treatment options. Early detection of pancreatic cancer in high-risk populations bears the potential to greatly improve outcomes, but current screening approaches remain of limited value despite recent technological advances. This review explores the possible advantages of liquid biopsies for this application, particularly focusing on circulating tumour cells (CTCs) and their subsequent single-cell omics analysis. Originating from both primary and metastatic tumour sites, CTCs provide important information for diagnosis, prognosis and tailoring of treatment strategies. Notably, CTCs have even been detected in the blood of subjects with pancreatic precursor lesions, suggesting their suitability as a non-invasive tool for the early detection of malignant transformation in the pancreas. As intact cells, CTCs offer comprehensive genomic, transcriptomic, epigenetic and proteomic information that can be explored using rapidly developing techniques for analysing individual cells at the molecular level. Studying CTCs during serial sampling and at single-cell resolution will help to dissect tumour heterogeneity for individual patients and among different patients, providing new insights into cancer evolution during disease progression and in response to treatment. Using CTCs for non-invasive tracking of cancer features, including stemness, metastatic potential and expression of immune targets, provides important and readily accessible molecular insights. Finally, the emerging technology of ex vivo culturing of CTCs could create new opportunities to study the functionality of individual cancers at any stage and develop personalised and more effective treatment approaches for this lethal disease.
Collapse
Affiliation(s)
- Qi Wang
- Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Berina Šabanović
- Pancreatic Cancer Heterogeneity, Candiolo Cancer Institute FPO-IRCCS, Candiolo, Turin, Italy
| | - Azhar Awada
- Pancreatic Cancer Heterogeneity, Candiolo Cancer Institute FPO-IRCCS, Candiolo, Turin, Italy; Molecular Biotechnology Center, University of Turin (UniTO), Turin, Italy
| | - Chiara Reina
- Pancreatic Cancer Heterogeneity, Candiolo Cancer Institute FPO-IRCCS, Candiolo, Turin, Italy
| | - Alexandra Aicher
- Precision Immunotherapy, Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
| | - Jiajia Tang
- Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai, China; South Chongqing Road 227, Shanghai, China.
| | - Christopher Heeschen
- Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Pancreatic Cancer Heterogeneity, Candiolo Cancer Institute FPO-IRCCS, Candiolo, Turin, Italy; South Chongqing Road 227, Shanghai, China.
| |
Collapse
|
39
|
Mirzayeh Fashami F, Levine M, Xie F, Blackhouse G, Tarride JE. Olaparib versus Placebo in Maintenance Treatment of Germline BRCA-Mutated Metastatic Pancreatic Cancer: A Cost-Utility Analysis from the Canadian Public Payer's Perspective. Curr Oncol 2023; 30:4688-4699. [PMID: 37232812 DOI: 10.3390/curroncol30050354] [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: 03/03/2023] [Revised: 04/11/2023] [Accepted: 04/28/2023] [Indexed: 05/27/2023] Open
Abstract
Pancreatic cancer has an annual incidence of 2/10,000 in Canada, with a one-year mortality rate greater than 80%. In the absence of a cost-effectiveness analysis in Canada, this study's objective was to assess the cost-effectiveness of olaparib versus a placebo in adult patients with deleterious or suspected deleterious BRCA metastatic pancreatic adenocarcinoma, who did not show any progression for at least 16 weeks with first-line platinum-based chemotherapy. A partitioned survival model with a 5-year time horizon was adopted to estimate the costs and effectiveness. All of the costs were extracted from the public payer's available resources, effectiveness data were obtained from the POLO trial, and Canadian studies were used for utility inputs. Probabilistic sensitivity analyses and scenario analyses were performed. The total costs of olaparib and the placebo over five years were CAD 179,477 and CAD 68,569, with overall quality-adjusted life-years (QALYs) of 1.70 and 1.36, respectively. The incremental cost-effectiveness ratio (ICER) of the olaparib group compared with the placebo was CAD 329,517 per QALY. With a commonly cited willingness to pay (WTP) threshold of CAD 50,000 per QALY, the drug does not achieve acceptable cost-effectiveness mainly due to the high price of the medication and insufficient impact on the overall survival of patients with metastatic pancreatic cancer.
Collapse
Affiliation(s)
- Fatemeh Mirzayeh Fashami
- Health Research Methodology Graduate Program, McMaster University, 1280 Main St. West, Hamilton, ON L8S 4K1, Canada
| | - Mitchell Levine
- Department of Health Research Methods, Evidence, and Impact, McMaster University, 1280 Main St. West, Hamilton, ON L8S 4K1, Canada
- Center for Health Economics and Policy Analysis (CHEPA), McMaster University, 1280 Main St. West, Hamilton, ON L8S 4K1, Canada
- Programs for Assessment of Technology in Health (PATH), The Research Institute of St. Joe's Hamilton, St. Joseph's Healthcare Hamilton, 50 Charlton Ave E, Hamilton, ON L8N 4A6, Canada
| | - Feng Xie
- Department of Health Research Methods, Evidence, and Impact, McMaster University, 1280 Main St. West, Hamilton, ON L8S 4K1, Canada
- Center for Health Economics and Policy Analysis (CHEPA), McMaster University, 1280 Main St. West, Hamilton, ON L8S 4K1, Canada
| | - Gordon Blackhouse
- Department of Health Research Methods, Evidence, and Impact, McMaster University, 1280 Main St. West, Hamilton, ON L8S 4K1, Canada
- Programs for Assessment of Technology in Health (PATH), The Research Institute of St. Joe's Hamilton, St. Joseph's Healthcare Hamilton, 50 Charlton Ave E, Hamilton, ON L8N 4A6, Canada
| | - Jean-Eric Tarride
- Center for Health Economics and Policy Analysis (CHEPA), McMaster University, 1280 Main St. West, Hamilton, ON L8S 4K1, Canada
- Programs for Assessment of Technology in Health (PATH), The Research Institute of St. Joe's Hamilton, St. Joseph's Healthcare Hamilton, 50 Charlton Ave E, Hamilton, ON L8N 4A6, Canada
- McMaster Chair in Health Technology Management, McMaster University, 1280 Main St. West, Hamilton, ON L8S 4K1, Canada
| |
Collapse
|
40
|
Mortoglou M, Miralles F, Mould RR, Sengupta D, Uysal-Onganer P. Inhibiting CDK4/6 in pancreatic ductal adenocarcinoma via microRNA-21. Eur J Cell Biol 2023; 102:151318. [PMID: 37105116 DOI: 10.1016/j.ejcb.2023.151318] [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: 02/20/2023] [Revised: 04/17/2023] [Accepted: 04/22/2023] [Indexed: 04/29/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive malignancies, with a 5-year survival rate of 5-10 %. The high mortality rate is due to the asymptomatic progression of clinical features in metastatic stages of the disease, which renders standard therapeutic options futile. PDAC is characterised by alterations in several genes that drive carcinogenesis and limit therapeutic response. The two most common genetic aberrations in PDAC are the mutational activation of KRAS and loss of the tumour suppressor CDK inhibitor 2A (CDKN2A), which culminate the activation of the cyclin-dependent kinase 4 and 6 (CDK4/6), that promote G1 cell cycle progression. Therapeutic strategies focusing on the CDK4/6 inhibitors such as palbociclib (PD-0332991) may potentially improve outcomes in this malignancy. MicroRNAs (miRs/miRNAs) are small endogenous non-coding RNA molecules associated with cellular proliferation, invasion, apoptosis, and cell cycle. Primarily, miR-21 promotes cell proliferation and a higher proportion of PDAC cells in the S phase, while knockdown of miR-21 has been linked to cell cycle arrest at the G2/M phase and inhibition of cell proliferation. In this study, using a CRISPR/Cas9 loss-of-function screen, we individually silenced the expression of miR-21 in two PDAC cell lines and in combination with PD-0332991 treatment, we examined the synergetic mechanisms of CDK4/6 inhibitors and miR-21 knockouts (KOs) on cell survival and death. This combination reduced cell proliferation, cell viability, increased apoptosis and G1 arrest in vitro. We further analysed the mitochondrial respiration and glycolysis of PDAC cells; then assessed the protein content of these cells and revealed numerous Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways associated with PD-0332991 treatment and miR-21 knocking out. Our results demonstrate that combined targeting of CDK4/6 and silencing of miR-21 represents a novel therapeutic strategy in PDAC.
Collapse
Affiliation(s)
- Maria Mortoglou
- Cancer Mechanisms and Biomarkers Research Group, School of Life Sciences, University of Westminster, W1W 6UW London, UK
| | - Francesc Miralles
- Centre of Biomedical Education/Molecular and Clinical Sciences, Cell Biology Research Centre, St. George's, University of London, Cranmer Terrace, London SW17 0RE, UK
| | - Rhys Richard Mould
- Research Centre for Optimal Health, School of Life Sciences, University of Westminster, W1W 6UW London, UK
| | - Dipankar Sengupta
- Health Data Sciences Research Group, Research Centre for Optimal Health, School of Life Sciences, University of Westminster, W1W 6UW London, UK
| | - Pinar Uysal-Onganer
- Cancer Mechanisms and Biomarkers Research Group, School of Life Sciences, University of Westminster, W1W 6UW London, UK.
| |
Collapse
|
41
|
Murthy D, Attri KS. PTGES Expression Is Associated with Metabolic and Immune Reprogramming in Pancreatic Ductal Adenocarcinoma. Int J Mol Sci 2023; 24:ijms24087304. [PMID: 37108468 PMCID: PMC10138618 DOI: 10.3390/ijms24087304] [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: 02/28/2023] [Revised: 03/18/2023] [Accepted: 04/13/2023] [Indexed: 04/29/2023] Open
Abstract
Metabolic reprogramming is an established hallmark of multiple cancers, including pancreatic cancer. Dysregulated metabolism is utilized by cancer cells for tumor progression, metastasis, immune microenvironment remodeling, and therapeutic resistance. Prostaglandin metabolites have been shown to be critical for inflammation and tumorigenesis. While the functional role of prostaglandin E2 metabolite has been extensively studied, there is a limited understanding of the PTGES enzyme in pancreatic cancer. Here, we investigated the relationship between expression of prostaglandin E synthase (PTGES) isoforms and the pathogenesis and regulation of pancreatic cancer. Our analysis identified higher expression of PTGES in pancreatic tumors compared to normal pancreatic tissues, suggesting an oncogenic function. Only PTGES1 expression was significantly correlated with worse prognosis of pancreatic cancer patients. Further, utilizing cancer genome atlas data, PTGES was found to be positively correlated with epithelial-mesenchymal transition, metabolic pathways, mucin oncogenic proteins, and immune pathways in cancer cells. PTGES expression was also correlated with higher mutational burden in key driver genes, such as TP53 and KRAS. Furthermore, our analysis indicated that the oncogenic pathway controlled by PTGES1 could be regulated via DNA methylation-dependent epigenetic mechanisms. Notably, the glycolysis pathway was positively correlated with PTGES and may fuel cancer cell growth. PTGES expression was also associated with downregulation of the MHC pathway and negatively correlated with CD8+ T cell activation markers. In summary, our study established an association of PTGES expression with pancreatic cancer metabolism and the immune microenvironment.
Collapse
Affiliation(s)
- Divya Murthy
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Kuldeep S Attri
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
42
|
LaRue KC, Fernandez-Zapico ME. EGFR Reloaded: Finding New Ways to Shape Pancreatic Cancer Epigenome. Cell Mol Gastroenterol Hepatol 2023; 15:1253-1254. [PMID: 36882150 PMCID: PMC10140288 DOI: 10.1016/j.jcmgh.2023.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 02/13/2023] [Indexed: 03/09/2023]
Affiliation(s)
- Kayla C LaRue
- Schulze Center for Novel Therapeutics, Division of Oncology Research; Mayo Clinic Graduate School of Biomedical Sciences, Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota
| | - Martin E Fernandez-Zapico
- Schulze Center for Novel Therapeutics, Division of Oncology Research, Mayo Clinic, Rochester, Minnesota.
| |
Collapse
|
43
|
Hoshi D, Kita E, Maru Y, Kogashi H, Nakamura Y, Tatsumi Y, Shimozato O, Nakamura K, Sudo K, Tsujimoto A, Yokoyama R, Kato A, Ushiku T, Fukayama M, Itami M, Yamaguchi T, Hippo Y. Derivation of pancreatic acinar cell carcinoma cell line HS-1 as a patient-derived tumor organoid. Cancer Sci 2023; 114:1165-1179. [PMID: 36382538 PMCID: PMC9986095 DOI: 10.1111/cas.15656] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 10/31/2022] [Accepted: 11/09/2022] [Indexed: 11/17/2022] Open
Abstract
Acinar cell carcinoma (ACC) of the pancreas is a malignant tumor of the exocrine cell lineage with a poor prognosis. Due to its rare incidence and technical difficulties, few authentic human cell lines are currently available, hampering detailed investigations of ACC. Therefore, we applied the organoid culture technique to various types of specimens, such as bile, biopsy, and resected tumor, obtained from a single ACC patient. Despite the initial propagation, none of these organoids achieved long-term proliferation or tolerated cryopreservation, confirming the challenging nature of establishing ACC cell lines. Nevertheless, the biopsy-derived early passage organoid developed subcutaneous tumors in immunodeficient mice. The xenograft tumor histologically resembled the original tumor and gave rise to infinitely propagating organoids with solid features and high levels of trypsin secretion. Moreover, the organoid stained positive for carboxylic ester hydrolase, a specific ACC marker, but negative for the duct cell marker CD133 and the endocrine lineage marker synaptophysin. Hence, we concluded the derivation of a novel ACC cell line of the pure exocrine lineage, designated HS-1. Genomic analysis revealed extensive copy number alterations and mutations in EP400 in the original tumor, which were enriched in primary organoids. HS-1 displayed homozygous deletion of CDKN2A, which might underlie xenograft formation from organoids. Although resistant to standard cytotoxic agents, the cell line was highly sensitive to the proteasome inhibitor bortezomib, as revealed by an in vitro drug screen and in vivo validation. In summary, we document a novel ACC cell line, which could be useful for ACC studies in the future.
Collapse
Affiliation(s)
- Daisuke Hoshi
- Department of Molecular Carcinogenesis, Chiba Cancer Center Research Institute, Chiba, Japan.,Department of Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Emiri Kita
- Department of Molecular Carcinogenesis, Chiba Cancer Center Research Institute, Chiba, Japan.,Department of Gastroenterology, Chiba Cancer Center, Chiba, Japan
| | - Yoshiaki Maru
- Department of Molecular Carcinogenesis, Chiba Cancer Center Research Institute, Chiba, Japan
| | - Hiroyuki Kogashi
- Department of Molecular Carcinogenesis, Chiba Cancer Center Research Institute, Chiba, Japan
| | - Yuki Nakamura
- Division of Oncogenomics, Chiba Cancer Center Research Institute, Chiba, Japan
| | - Yasutoshi Tatsumi
- Division of Oncogenomics, Chiba Cancer Center Research Institute, Chiba, Japan
| | - Osamu Shimozato
- Division of Oncogenomics, Chiba Cancer Center Research Institute, Chiba, Japan
| | | | - Kentaro Sudo
- Department of Gastroenterology, Chiba Cancer Center, Chiba, Japan
| | - Akiko Tsujimoto
- Department of Gastroenterology, Chiba Cancer Center, Chiba, Japan
| | - Ryo Yokoyama
- Division of Pathological Diagnosis, Matsudo City General Hospital, Chiba, Japan
| | - Atsushi Kato
- Department of Hepatobiliary-Pancreatic Surgery, Chiba Cancer Center, Chiba, Japan
| | - Tetsuo Ushiku
- Department of Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Masashi Fukayama
- Department of Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Department of Pathology, Asahi General Hospital, Chiba, Japan
| | - Makiko Itami
- Division of Surgical Pathology, Chiba Cancer Center, Chiba, Japan
| | - Taketo Yamaguchi
- Department of Gastroenterology, Chiba Cancer Center, Chiba, Japan.,Department of Internal Medicine, Funabashi Central Hospital, Chiba, Japan
| | - Yoshitaka Hippo
- Department of Molecular Carcinogenesis, Chiba Cancer Center Research Institute, Chiba, Japan
| |
Collapse
|
44
|
Menjivar RE, Nwosu ZC, Du W, Donahue KL, Hong HS, Espinoza C, Brown K, Velez-Delgado A, Yan W, Lima F, Bischoff A, Kadiyala P, Salas-Escabillas D, Crawford HC, Bednar F, Carpenter E, Zhang Y, Halbrook CJ, Lyssiotis CA, Pasca di Magliano M. Arginase 1 is a key driver of immune suppression in pancreatic cancer. eLife 2023; 12:e80721. [PMID: 36727849 PMCID: PMC10260021 DOI: 10.7554/elife.80721] [Citation(s) in RCA: 57] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 01/30/2023] [Indexed: 02/03/2023] Open
Abstract
An extensive fibroinflammatory stroma rich in macrophages is a hallmark of pancreatic cancer. In this disease, it is well appreciated that macrophages are immunosuppressive and contribute to the poor response to immunotherapy; however, the mechanisms of immune suppression are complex and not fully understood. Immunosuppressive macrophages are classically defined by the expression of the enzyme Arginase 1 (ARG1), which we demonstrated is potently expressed in pancreatic tumor-associated macrophages from both human patients and mouse models. While routinely used as a polarization marker, ARG1 also catabolizes arginine, an amino acid required for T cell activation and proliferation. To investigate this metabolic function, we used a genetic and a pharmacologic approach to target Arg1 in pancreatic cancer. Genetic inactivation of Arg1 in macrophages, using a dual recombinase genetically engineered mouse model of pancreatic cancer, delayed formation of invasive disease, while increasing CD8+ T cell infiltration. Additionally, Arg1 deletion induced compensatory mechanisms, including Arg1 overexpression in epithelial cells, namely Tuft cells, and Arg2 overexpression in a subset of macrophages. To overcome these compensatory mechanisms, we used a pharmacological approach to inhibit arginase. Treatment of established tumors with the arginase inhibitor CB-1158 exhibited further increased CD8+ T cell infiltration, beyond that seen with the macrophage-specific knockout, and sensitized the tumors to anti-PD1 immune checkpoint blockade. Our data demonstrate that Arg1 drives immune suppression in pancreatic cancer by depleting arginine and inhibiting T cell activation.
Collapse
Affiliation(s)
- Rosa E Menjivar
- Cellular and Molecular Biology Program, University of Michigan-Ann ArborAnn ArborUnited States
| | - Zeribe C Nwosu
- Department of Molecular and Integrative Physiology, University of Michigan-Ann ArborAnn ArborUnited States
| | - Wenting Du
- Department of Surgery, University of Michigan-Ann ArborAnn ArborUnited States
| | - Katelyn L Donahue
- Cancer Biology Program, University of Michigan-Ann ArborAnn ArborUnited States
| | - Hanna S Hong
- Department of Immunology, University of Michigan-Ann ArborAnn ArborUnited States
| | - Carlos Espinoza
- Department of Surgery, University of Michigan-Ann ArborAnn ArborUnited States
| | - Kristee Brown
- Department of Surgery, University of Michigan-Ann ArborAnn ArborUnited States
| | - Ashley Velez-Delgado
- Department of Cell and Developmental Biology, University of Michigan-Ann ArborAnn ArborUnited States
| | - Wei Yan
- Department of Surgery, University of Michigan-Ann ArborAnn ArborUnited States
| | - Fatima Lima
- Department of Surgery, University of Michigan-Ann ArborAnn ArborUnited States
| | - Allison Bischoff
- Cancer Biology Program, University of Michigan-Ann ArborAnn ArborUnited States
| | - Padma Kadiyala
- Department of Immunology, University of Michigan-Ann ArborAnn ArborUnited States
| | | | | | - Filip Bednar
- Department of Surgery, University of Michigan-Ann ArborAnn ArborUnited States
- Rogel Cancer CenterAnn ArborUnited States
| | - Eileen Carpenter
- Rogel Cancer CenterAnn ArborUnited States
- Department of Internal Medicine, Division of Gastroenterolog, University of Michigan-Ann ArborAnn ArborUnited States
| | - Yaqing Zhang
- Department of Surgery, University of Michigan-Ann ArborAnn ArborUnited States
- Rogel Cancer CenterAnn ArborUnited States
| | - Christopher J Halbrook
- Department of Molecular Biology and Biochemistry, University of California, IrvineIrvineUnited States
- Chao Family Comprehensive Cancer Center, University of California, IrvineIrvineUnited States
| | - Costas A Lyssiotis
- Department of Molecular and Integrative Physiology, University of Michigan-Ann ArborAnn ArborUnited States
- Cancer Biology Program, University of Michigan-Ann ArborAnn ArborUnited States
- Rogel Cancer CenterAnn ArborUnited States
- Department of Internal Medicine, Division of Gastroenterolog, University of Michigan-Ann ArborAnn ArborUnited States
| | - Marina Pasca di Magliano
- Cellular and Molecular Biology Program, University of Michigan-Ann ArborAnn ArborUnited States
- Cancer Biology Program, University of Michigan-Ann ArborAnn ArborUnited States
- Department of Cell and Developmental Biology, University of Michigan-Ann ArborAnn ArborUnited States
- Henry Ford Pancreatic Cancer CenterDetroitUnited States
- Rogel Cancer CenterAnn ArborUnited States
| |
Collapse
|
45
|
Cao P, Wu Y, Sun D, Zhang W, Qiu J, Tang Z, Xue X, Qin L. IGF2BP2 promotes pancreatic carcinoma progression by enhancing the stability of B3GNT6 mRNA via m6A methylation. Cancer Med 2023; 12:4405-4420. [PMID: 35908253 PMCID: PMC9972174 DOI: 10.1002/cam4.5096] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 05/08/2022] [Accepted: 07/14/2022] [Indexed: 11/07/2022] Open
Abstract
BACKGROUND Pancreatic carcinoma (PC) is a highly lethal cancer with an increasing mortality rate, its five-year survival rate is only approximately 4%. N6-methyladenosine (m6A) modification is the most common posttranscriptional modification of RNA, it could affect tumor formation by regulating m6A modifications in the mRNA of key oncogenes or tumor suppressor genes. However, its role in PC remains unclear. METHODS We combined bioinformatic analysis with in vitro and in vivo experiments to investigate the expression profile of methylation modulators and identify key m6A regulators in the progression of PC. Further study focused on exploring the target genes binding to the regulators through RIP and immunofluorescence staining experiment. RESULTS TCGA and Gene Expression Omnibus (GEO) analyses revealed an overall increasing trend in the expression of m6A regulators in PC, and consensus clustering analysis of m6A modification showed that the expression of regulators was negatively correlated with the survival rate. LASSO-Cox regression analysis revealed that IGF2BP2, METTL3, ALKBH5 and KIAA1429 were associated with hazard ratios (HR), but only IGF2BP2 was sufficiently appropriate for the m6A survival prognosis model. The IHC and WB results verified high protein expression of IGF2BP2 in PC, and IGF2BP2 knockdown inhibited the proliferation and migration of PC cells. We predicted and verified B3GNT6 was observably regulated by IGF2BP2 via RIP assays. In addition, IF staining confirmed the co-expression of IGF2BP2 and B3GNT6. The tumor-promoting effect of IGF2BP2 and its co-expression with B3GNT6 were verified in an animal model. CONCLUSIONS Elevated m6A levels promote PC progression. IGF2BP2 is a credible marker and modulates B3GNT6 mRNA stability, indicating that IGF2BP2 is a potential prognostic marker and therapeutic target in PC progression.
Collapse
Affiliation(s)
- Pei Cao
- Department of General SurgeryThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
| | - Yufan Wu
- Department of General SurgeryThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
| | - Ding Sun
- Department of General SurgeryThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
| | - Weigang Zhang
- Department of General SurgeryThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
| | - Junyi Qiu
- Department of General SurgeryThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
| | - Zuxiong Tang
- Department of General SurgeryThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
| | - Xiaofeng Xue
- Department of General SurgeryThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
| | - Lei Qin
- Department of General SurgeryThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
| |
Collapse
|
46
|
Tanaka HY, Nakazawa T, Enomoto A, Masamune A, Kano MR. Therapeutic Strategies to Overcome Fibrotic Barriers to Nanomedicine in the Pancreatic Tumor Microenvironment. Cancers (Basel) 2023; 15:cancers15030724. [PMID: 36765684 PMCID: PMC9913712 DOI: 10.3390/cancers15030724] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/19/2023] [Accepted: 01/21/2023] [Indexed: 01/26/2023] Open
Abstract
Pancreatic cancer is notorious for its dismal prognosis. The enhanced permeability and retention (EPR) effect theory posits that nanomedicines (therapeutics in the size range of approximately 10-200 nm) selectively accumulate in tumors. Nanomedicine has thus been suggested to be the "magic bullet"-both effective and safe-to treat pancreatic cancer. However, the densely fibrotic tumor microenvironment of pancreatic cancer impedes nanomedicine delivery. The EPR effect is thus insufficient to achieve a significant therapeutic effect. Intratumoral fibrosis is chiefly driven by aberrantly activated fibroblasts and the extracellular matrix (ECM) components secreted. Fibroblast and ECM abnormalities offer various potential targets for therapeutic intervention. In this review, we detail the diverse strategies being tested to overcome the fibrotic barriers to nanomedicine in pancreatic cancer. Strategies that target the fibrotic tissue/process are discussed first, which are followed by strategies to optimize nanomedicine design. We provide an overview of how a deeper understanding, increasingly at single-cell resolution, of fibroblast biology is revealing the complex role of the fibrotic stroma in pancreatic cancer pathogenesis and consider the therapeutic implications. Finally, we discuss critical gaps in our understanding and how we might better formulate strategies to successfully overcome the fibrotic barriers in pancreatic cancer.
Collapse
Affiliation(s)
- Hiroyoshi Y. Tanaka
- Department of Pharmaceutical Biomedicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 1-1-1 Tsushima-naka, Kita-ku, Okayama-shi 700-8530, Okayama, Japan
| | - Takuya Nakazawa
- Department of Pharmaceutical Biomedicine, Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, 1-1-1 Tsushima-naka, Kita-ku, Okayama-shi 700-8530, Okayama, Japan
| | - Atsushi Enomoto
- Department of Pathology, Graduate School of Medicine, Nagoya University, 65 Tsurumai-cho, Showa-ku, Nagoya-shi 466-8550, Aichi, Japan
| | - Atsushi Masamune
- Division of Gastroenterology, Graduate School of Medicine, Tohoku University, 1-1 Seiryo-machi, Aoba-ku, Sendai-shi 980-8574, Miyagi, Japan
| | - Mitsunobu R. Kano
- Department of Pharmaceutical Biomedicine, Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, 1-1-1 Tsushima-naka, Kita-ku, Okayama-shi 700-8530, Okayama, Japan
- Correspondence:
| |
Collapse
|
47
|
In Vitro Measurement and Mathematical Modeling of Thermally-Induced Injury in Pancreatic Cancer Cells. Cancers (Basel) 2023; 15:cancers15030655. [PMID: 36765619 PMCID: PMC9913239 DOI: 10.3390/cancers15030655] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/16/2023] [Accepted: 01/18/2023] [Indexed: 01/24/2023] Open
Abstract
Thermal therapies are under investigation as part of multi-modality strategies for the treatment of pancreatic cancer. In the present study, we determined the kinetics of thermal injury to pancreatic cancer cells in vitro and evaluated predictive models for thermal injury. Cell viability was measured in two murine pancreatic cancer cell lines (KPC, Pan02) and a normal fibroblast (STO) cell line following in vitro heating in the range 42.5-50 °C for 3-60 min. Based on measured viability data, the kinetic parameters of thermal injury were used to predict the extent of heat-induced damage. Of the three thermal injury models considered in this study, the Arrhenius model with time delay provided the most accurate prediction (root mean square error = 8.48%) for all cell lines. Pan02 and STO cells were the most resistant and susceptible to hyperthermia treatments, respectively. The presented data may contribute to studies investigating the use of thermal therapies as part of pancreatic cancer treatment strategies and inform the design of treatment planning strategies.
Collapse
|
48
|
Lian Y, Zeng S, Wen S, Zhao X, Fang C, Zeng N. Review and Application of Integrin Alpha v Beta 6 in the Diagnosis and Treatment of Cholangiocarcinoma and Pancreatic Ductal Adenocarcinoma. Technol Cancer Res Treat 2023; 22:15330338231189399. [PMID: 37525872 PMCID: PMC10395192 DOI: 10.1177/15330338231189399] [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: 02/14/2023] [Revised: 06/13/2023] [Accepted: 06/28/2023] [Indexed: 08/02/2023] Open
Abstract
Integrin Alpha v Beta 6 is expressed primarily in solid epithelial tumors, such as cholangiocarcinoma, pancreatic cancer, and colorectal cancer. It has been considered a potential and promising molecular marker for the early diagnosis and treatment of cancer. Cholangiocarcinoma and pancreatic ductal adenocarcinoma share genetic, histological, and pathophysiological similarities due to the shared embryonic origin of the bile duct and pancreas. These cancers share numerous clinicopathological characteristics, including growth pattern, poor response to conventional radiotherapy and chemotherapy, and poor prognosis. This review focuses on the role of integrin Alpha v Beta 6 in cancer progression. It addition, it reviews how the marker can be used in molecular imaging and therapeutic targets. We propose further research explorations and questions that need to be addressed. We conclude that integrin Alpha v Beta 6 may serve as a potential biomarker for cancer disease progression and prognosis.
Collapse
Affiliation(s)
- Yunyu Lian
- Zhujiang Hospital, The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
- First Department of Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Silue Zeng
- First Department of Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Guangdong Provincial Clinical and Engineering Technology Center of Digital Medicine, Guangzhou, China
| | - Sai Wen
- First Department of Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Guangdong Provincial Clinical and Engineering Technology Center of Digital Medicine, Guangzhou, China
| | - Xingyang Zhao
- First Department of Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Guangdong Provincial Clinical and Engineering Technology Center of Digital Medicine, Guangzhou, China
| | - Chihua Fang
- Zhujiang Hospital, The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
- First Department of Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Guangdong Provincial Clinical and Engineering Technology Center of Digital Medicine, Guangzhou, China
| | - Ning Zeng
- Zhujiang Hospital, The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
- First Department of Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Guangdong Provincial Clinical and Engineering Technology Center of Digital Medicine, Guangzhou, China
| |
Collapse
|
49
|
Peng J, Madduri S, Clontz AD, Stewart DA. Clinical trial-identified inflammatory biomarkers in breast and pancreatic cancers. Front Endocrinol (Lausanne) 2023; 14:1106520. [PMID: 37181043 PMCID: PMC10173309 DOI: 10.3389/fendo.2023.1106520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 04/04/2023] [Indexed: 05/16/2023] Open
Abstract
Breast cancer and pancreatic cancer are two common cancer types characterized by high prevalence and high mortality rates, respectively. However, breast cancer has been more well-studied than pancreatic cancer. This narrative review curated inflammation-associated biomarkers from clinical studies that were systematically selected for both breast and pancreatic cancers and discusses some of the common and unique elements between the two endocrine-regulated malignant diseases. Finding common ground between the two cancer types and specifically analyzing breast cancer study results, we hoped to explore potential feasible methods and biomarkers that may be useful also in diagnosing and treating pancreatic cancer. A PubMed MEDLINE search was used to identify articles that were published between 2015-2022 of different kinds of clinical trials that measured immune-modulatory biomarkers and biomarker changes of inflammation defined in diagnosis and treatment of breast cancer and pancreatic cancer patients. A total of 105 papers (pancreatic cancer 23, breast cancer 82) were input into Covidence for the title and abstract screening. The final number of articles included in this review was 73 (pancreatic cancer 19, breast cancer 54). The results showed some of the frequently cited inflammatory biomarkers for breast and pancreatic cancers included IL-6, IL-8, CCL2, CD8+ T cells and VEGF. Regarding unique markers, CA15-3 and TNF-alpha were two of several breast cancer-specific, and CA19 and IL-18 were pancreatic cancer-specific. Moreover, we discussed leptin and MMPs as emerging biomarker targets with potential use for managing pancreatic cancer based on breast cancer studies in the future, based on inflammatory mechanisms. Overall, the similarity in how both types of cancers respond to or result in further disruptive inflammatory signaling, and that point to a list of markers that have been shown useful in diagnosis and/or treatment method response or efficacy in managing breast cancer could potentially provide insights into developing the same or more useful diagnostic and treatment measurement inflammatory biomarkers for pancreatic cancer. More research is needed to investigate the relationship and associated inflammatory markers between the similar immune-associated biological mechanisms that contribute to breast and pancreatic cancer etiology, drive disease progression or that impact treatment response and reflect survival outcomes.
Collapse
Affiliation(s)
- Jing Peng
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Supradeep Madduri
- Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, NC, United States
| | - Angela D. Clontz
- Department of Nutrition, Meredith College, Raleigh, NC, United States
| | - Delisha A. Stewart
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, NC, United States
- *Correspondence: Delisha A. Stewart,
| |
Collapse
|
50
|
Mugiyanto E, Adikusuma W, Irham LM, Huang WC, Chang WC, Kuo CN. Integrated genomic analysis to identify druggable targets for pancreatic cancer. Front Oncol 2022; 12:989077. [PMID: 36531045 PMCID: PMC9752886 DOI: 10.3389/fonc.2022.989077] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 10/19/2022] [Indexed: 03/31/2024] Open
Abstract
According to the National Comprehensive Cancer Network and the American Society of Clinical Oncology, the standard treatment for pancreatic cancer (PC) is gemcitabine and fluorouracil. Other chemotherapeutic agents have been widely combined. However, drug resistance remains a huge challenge, leading to the ineffectiveness of cancer therapy. Therefore, we are trying to discover new treatments for PC by utilizing genomic information to identify PC-associated genes as well as drug target genes for drug repurposing. Genomic information from a public database, the cBio Cancer Genomics Portal, was employed to retrieve the somatic mutation genes of PC. Five functional annotations were applied to prioritize the PC risk genes: Kyoto Encyclopedia of Genes and Genomes; biological process; knockout mouse; Gene List Automatically Derived For You; and Gene Expression Omnibus Dataset. DrugBank database was utilized to extract PC drug targets. To narrow down the most promising drugs for PC, CMap Touchstone analysis was applied. Finally, ClinicalTrials.gov and a literature review were used to screen the potential drugs under clinical and preclinical investigation. Here, we extracted 895 PC-associated genes according to the cBioPortal database and prioritized them by using five functional annotations; 318 genes were assigned as biological PC risk genes. Further, 216 genes were druggable according to the DrugBank database. CMap Touchstone analysis indicated 13 candidate drugs for PC. Among those 13 drugs, 8 drugs are in the clinical trials, 2 drugs were supported by the preclinical studies, and 3 drugs are with no evidence status for PC. Importantly, we found that midostaurin (targeted PRKA) and fulvestrant (targeted ESR1) are promising candidate drugs for PC treatment based on the genomic-driven drug repurposing pipelines. In short, integrated analysis using a genomic information database demonstrated the viability for drug repurposing. We proposed two drugs (midostaurin and fulvestrant) as promising drugs for PC.
Collapse
Affiliation(s)
- Eko Mugiyanto
- PhD Program in Clinical Drug Development of Herbal Medicine, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
- Department of Pharmacy, Faculty of Health Science, University of Muhammadiyah Pekajangan Pekalongan, Pekalongan, Indonesia
| | - Wirawan Adikusuma
- Department of Clinical Pharmacy, School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
- Department of Pharmacy, Faculty of Health Science, University of Muhammadiyah Mataram, Mataram, Indonesia
| | | | - Wan-Chen Huang
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| | - Wei-Chiao Chang
- PhD Program in Clinical Drug Development of Herbal Medicine, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
- Department of Clinical Pharmacy, School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
- Department of Pharmacy, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
- Integrative Research Center for Critical Care, Department of Pharmacy, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Chun-Nan Kuo
- Department of Clinical Pharmacy, School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
- Department of Pharmacy, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|