1
|
Pfaffl MW. Combining Cre-LoxP and single-cell sequencing technologies: insights into the extracellular vesicle cargo transfer. EXTRACELLULAR VESICLES AND CIRCULATING NUCLEIC ACIDS 2024; 5:714-717. [PMID: 39811732 PMCID: PMC11725425 DOI: 10.20517/evcna.2024.58] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 10/22/2024] [Accepted: 10/28/2024] [Indexed: 01/16/2025]
Abstract
The recent study from the Pogge von Strandmann group published in Cellular and Molecular Immunology, by Alashkar Alhamwe et al., combined for the first time the Cre-LoxP recombination system with single-cell sequencing. The group monitored the tumor-derived extracellular vesicle (EV) uptake and the EV functions in the recipient non-malignant cells in a pancreatic ductal adenocarcinoma mouse model. Recombination events and EV uptake, together with resulting gene expression changes in macrophages, neutrophils, and mast cells, were detected by single-cell sequencing technology of the tumor tissue. This new approach is highly specific, as it can identify single EV recipient cells without interfering with the EV biogenesis or the phenotype.
Collapse
Affiliation(s)
- Michael W. Pfaffl
- Animal Physiology and Immunology, School of Life Sciences, Technical University of Munich, Freising 85354, Germany.
| |
Collapse
|
2
|
Sabljo K, Ischyropoulou M, Napp J, Alves F, Feldmann C. High-load nanoparticles with a chemotherapeutic SN-38/FdUMP drug cocktail. NANOSCALE 2024; 16:14853-14860. [PMID: 39034735 DOI: 10.1039/d4nr01403k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/23/2024]
Abstract
[Gd(OH)]2+[(SN-38)0.5(FdUMP)0.5]2- inorganic-organic hybrid nanoparticles (IOH-NPs) with a chemotherapeutic cocktail of ethyl-10-hydroxycamptothecin (SN-38, active form of irinotecan) and 5-fluoro-2'-deoxyuridine-5'-phosphate (FdUMP, active form of 5'-fluoruracil), 40 nm in size, are prepared in water. The IOH-NPs contain a total drug load of 63 wt% with 33 wt% of SN-38 and 30 wt% of FdUMP. Cell-based assays show efficient cellular uptake and promising anti-tumour activity on two pancreatic cancer cell lines of murine origin (KPC, Panc02). Beside the high-load drug cocktail, especially the option to use SN-38, which - although 100- to 1000-times more potent than irinotecan - is usually unsuitable for systemic administration due to poor solubility, low stability, and high toxicity upon non-selective delivery. The [Gd(OH)]2+[(SN-38)0.5(FdUMP)0.5]2- IOH-NPs are a new concept to deliver a drug cocktail with SN-38 and FdUMP directly to the tumour, shielded in a nanoparticle, to reduce side effects.
Collapse
Affiliation(s)
- Kristina Sabljo
- Karlsruhe Institute of Technology (KIT), Institute for Inorganic Chemistry, Engesserstrasse 15, 76131 Karlsruhe, Germany.
| | - Myrto Ischyropoulou
- University Medical Center Goettingen (UMG), Institute for Diagnostic and Interventional Radiology, Robert Koch Str. 40, 37075 Goettingen, Germany
| | - Joanna Napp
- University Medical Center Goettingen (UMG), Institute for Diagnostic and Interventional Radiology, Robert Koch Str. 40, 37075 Goettingen, Germany
| | - Frauke Alves
- University Medical Center Goettingen (UMG), Institute for Diagnostic and Interventional Radiology, Robert Koch Str. 40, 37075 Goettingen, Germany
- Max Planck Institute for Multidisciplinary Sciences, Translational Molecular Imaging, Hermann-Rein-Strasse 3, 37075 Goettingen, Germany
- University Medical Center Goettingen (UMG), Clinic for Haematology and Medical Oncology, Robert Koch Str. 40, 37075 Goettingen, Germany
| | - Claus Feldmann
- Karlsruhe Institute of Technology (KIT), Institute for Inorganic Chemistry, Engesserstrasse 15, 76131 Karlsruhe, Germany.
| |
Collapse
|
3
|
Ji Y, An Q, Wen X, Xu Z, Xia Z, Xia Z, Hu Q, Lei S. Liver cancer from the perspective of single-cell sequencing: a review combined with bibliometric analysis. J Cancer Res Clin Oncol 2024; 150:316. [PMID: 38910204 PMCID: PMC11194221 DOI: 10.1007/s00432-024-05855-7] [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: 03/16/2024] [Accepted: 06/17/2024] [Indexed: 06/25/2024]
Abstract
BACKGROUND Liver cancer (LC) is a prevalent malignancy and a leading cause of cancer-related mortality worldwide. Extensive research has been conducted to enhance patient outcomes and develop effective prevention strategies, ranging from molecular mechanisms to clinical interventions. Single-cell sequencing, as a novel bioanalysis technology, has significantly contributed to the understanding of the global cognition and dynamic changes in liver cancer. However, there is a lack of bibliometric analysis in this specific research area. Therefore, the objective of this study is to provide a comprehensive overview of the knowledge structure and research hotspots in the field of single-cell sequencing in liver cancer research through the use of bibliometrics. METHOD Publications related to the application of single-cell sequencing technology to liver cancer research as of December 31, 2023, were searched on the web of science core collection (WoSCC) database. VOSviewers, CiteSpace, and R package "bibliometrix" were used to conduct this bibliometric analysis. RESULTS A total of 331 publications from 34 countries, primarily led by China and the United States, were included in this study. The research focuses on the application of single cell sequencing technology to liver cancer, and the number of related publications has been increasing year by year. The main research institutions involved in this field are Fudan University, Sun Yat-Sen University, and the Chinese Academy of Sciences. Frontiers in Immunology and Nature Communications is the most popular journal in this field, while Cell is the most frequently co-cited journal. These publications are authored by 2799 individuals, with Fan Jia and Zhou Jian having the most published papers, and Llovet Jm being the most frequently co-cited author. The use of single cell sequencing to explore the immune microenvironment of liver cancer, as well as its implications in immunotherapy and chemotherapy, remains the central focus of this field. The emerging research hotspots are characterized by keywords such as 'Gene-Expression', 'Prognosis', 'Tumor Heterogeneity', 'Immunoregulation', and 'Tumor Immune Microenvironment'. CONCLUSION This is the first bibliometric study that comprehensively summarizes the research trends and developments on the application of single cell sequencing in liver cancer. The study identifies recent research frontiers and hot directions, providing a valuable reference for researchers exploring the landscape of liver cancer, understanding the composition of the immune microenvironment, and utilizing single-cell sequencing technology to guide and enhance the prognosis of liver cancer patients.
Collapse
Affiliation(s)
- Yanwei Ji
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Qi An
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xinyu Wen
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhou Xu
- The Second Clinical Medical College, Jiangxi Medical College, Nanchang University, Jiangxi, Nanchang, China
| | - Zhengyuan Xia
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
- Faculty of Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macao, China
| | - Zhongyuan Xia
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Qinyong Hu
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, China.
| | - Shaoqing Lei
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China.
| |
Collapse
|
4
|
Shinde A, Panchal K, Patra P, Singh S, Enakolla S, Paliwal R, Chaurasiya A. QbD Enabled Development and Evaluation of Pazopanib Loaded Nanoliposomes for PDAC Treatment. AAPS PharmSciTech 2024; 25:97. [PMID: 38710894 DOI: 10.1208/s12249-024-02806-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 04/10/2024] [Indexed: 05/08/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the highly fatal types of cancer with high mortality/incidence. Considering the crucial role of vascular endothelial growth factor (VEGF) in PDAC progression, its inhibition can be a viable strategy for the treatment. Pazopanib, a second-generation VEGF inhibitor, is approved for the treatment of various oncological conditions. However, due to associated limitations like low oral bioavailability (14-39%), high inter/intra-subject variability, stability issues, etc., high doses (800 mg) are required, which further lead to non-specific toxicities and also contribute toward cancer resistance. Thus, to overcome these challenges, pazopanib-loaded PEGylated nanoliposomes were developed and evaluated against pancreatic cancer cell lines. The nanoliposomes were prepared by thin-film hydration method, followed by characterization and stability studies. This QbD-enabled process design successfully led to the development of a suitable pazopanib liposomal formulation with desirable properties. The % entrapment of PZP-loaded non-PEGylated and PEGylated nanoliposomes was found to be 75.2% and 84.9%, respectively, whereas their particle size was found to be 129.7 nm and 182.0 nm, respectively. The developed liposomal formulations exhibited a prolonged release and showed desirable physicochemical properties. Furthermore, these liposomal formulations were also assessed for in vitro cell lines, such as cell cytotoxicity assay and cell uptake. These studies confirm the effectiveness of developed liposomal formulations against pancreatic cancer cell lines. The outcomes of this work provide encouraging results and a way forward to thoroughly investigate its potential for PDAC treatment.
Collapse
Affiliation(s)
- Aishwarya Shinde
- Translational Pharmaceutics Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science, Hyderabad, Pilani, India
| | - Kanan Panchal
- Translational Pharmaceutics Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science, Hyderabad, Pilani, India
| | - Parameswar Patra
- Translational Pharmaceutics Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science, Hyderabad, Pilani, India
| | - Sonali Singh
- Translational Pharmaceutics Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science, Hyderabad, Pilani, India
| | - Sucharitha Enakolla
- Translational Pharmaceutics Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science, Hyderabad, Pilani, India
| | - Rishi Paliwal
- Nanomedicine and Bioengineering Research Laboratory, Department of Pharmacy, Indira Gandhi National Tribal University, Amarkantak, India
| | - Akash Chaurasiya
- Translational Pharmaceutics Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science, Hyderabad, Pilani, India.
| |
Collapse
|
5
|
Lara O, Janssen P, Mambretti M, De Pauw L, Ates G, Mackens L, De Munck J, Walckiers J, Pan Z, Beckers P, Espinet E, Sato H, De Ridder M, Marks DL, Barbé K, Aerts JL, Hermans E, Rooman I, Massie A. Compartmentalized role of xCT in supporting pancreatic tumor growth, inflammation and mood disturbance in mice. Brain Behav Immun 2024; 118:275-286. [PMID: 38447884 DOI: 10.1016/j.bbi.2024.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 02/05/2024] [Accepted: 03/02/2024] [Indexed: 03/08/2024] Open
Abstract
xCT (Slc7a11), the specific subunit of the cystine/glutamate antiporter system xc-, is present in the brain and on immune cells, where it is known to modulate behavior and inflammatory responses. In a variety of cancers -including pancreatic ductal adenocarcinoma (PDAC)-, xCT is upregulated by tumor cells to support their growth and spread. Therefore, we studied the impact of xCT deletion in pancreatic tumor cells (Panc02) and/or the host (xCT-/- mice) on tumor burden, inflammation, cachexia and mood disturbances. Deletion of xCT in the tumor strongly reduced tumor growth. Targeting xCT in the host and not the tumor resulted only in a partial reduction of tumor burden, while it did attenuate tumor-related systemic inflammation and prevented an increase in immunosuppressive regulatory T cells. The latter effect could be replicated by specific xCT deletion in immune cells. xCT deletion in the host or the tumor differentially modulated neuroinflammation. When mice were grafted with xCT-deleted tumor cells, hypothalamic inflammation was reduced and, accordingly, food intake improved. Tumor bearing xCT-/- mice showed a trend of reduced hippocampal neuroinflammation with less anxiety- and depressive-like behavior. Taken together, targeting xCT may have beneficial effects on pancreatic cancer-related comorbidities, beyond reducing tumor burden. The search for novel and specific xCT inhibitors is warranted as they may represent a holistic therapy in pancreatic cancer.
Collapse
Affiliation(s)
- Olaya Lara
- Laboratory of Neuro-Aging & Viro-Immunotherapy, Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB), Brussels 1090, Belgium; Laboratory for Medical and Molecular Oncology, Translational Oncology Research Center (TORC), VUB, Brussels 1090, Belgium
| | - Pauline Janssen
- Laboratory of Neuro-Aging & Viro-Immunotherapy, Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB), Brussels 1090, Belgium; Laboratory for Medical and Molecular Oncology, Translational Oncology Research Center (TORC), VUB, Brussels 1090, Belgium
| | - Marco Mambretti
- Laboratory for Medical and Molecular Oncology, Translational Oncology Research Center (TORC), VUB, Brussels 1090, Belgium
| | - Laura De Pauw
- Laboratory of Neuro-Aging & Viro-Immunotherapy, Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB), Brussels 1090, Belgium
| | - Gamze Ates
- Laboratory of Neuro-Aging & Viro-Immunotherapy, Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB), Brussels 1090, Belgium
| | - Liselotte Mackens
- Laboratory of Neuro-Aging & Viro-Immunotherapy, Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB), Brussels 1090, Belgium
| | - Jolien De Munck
- Laboratory of Neuro-Aging & Viro-Immunotherapy, Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB), Brussels 1090, Belgium
| | - Jarne Walckiers
- Laboratory of Neuro-Aging & Viro-Immunotherapy, Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB), Brussels 1090, Belgium
| | - Zhaolong Pan
- Laboratory for Medical and Molecular Oncology, Translational Oncology Research Center (TORC), VUB, Brussels 1090, Belgium
| | - Pauline Beckers
- Institute of Neuroscience, Université catholique de Louvain, Brussels 1200, Belgium
| | - Elisa Espinet
- Pancreatic Cancer Lab, Department of Pathology and Experimental Therapy, School of Medicine, University of Barcelona, L'Hospitalet de Llobregat, Barcelona 08907, Spain; Molecular Mechanisms and Experimental Therapy in Oncology Program, Institut d'Investigació Biomèdica de Bellvitge, L'Hospitalet de Llobregat, Barcelona 08907, Spain
| | - Hideyo Sato
- Department of Medical Technology, Niigata University, Niigata 950-3198, Japan
| | - Mark De Ridder
- Department of Radiotherapy, UZ Brussels, VUB, Brussels 1090, Belgium
| | - Daniel L Marks
- Papé Family Pediatric Research Institute, Oregon Health and Science University, Portland, OR 97239, USA
| | - Kurt Barbé
- The Biostatistics and Medical Informatics Department, VUB, Brussels 1090, Belgium
| | - Joeri L Aerts
- Laboratory of Neuro-Aging & Viro-Immunotherapy, Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB), Brussels 1090, Belgium
| | - Emmanuel Hermans
- Institute of Neuroscience, Université catholique de Louvain, Brussels 1200, Belgium
| | - Ilse Rooman
- Laboratory for Medical and Molecular Oncology, Translational Oncology Research Center (TORC), VUB, Brussels 1090, Belgium.
| | - Ann Massie
- Laboratory of Neuro-Aging & Viro-Immunotherapy, Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB), Brussels 1090, Belgium.
| |
Collapse
|
6
|
Griffin JI, Chen X, Duan L, Mu Q, Ho RJY. Inoculation of Pan02 cells produces tumor nodules in mouse pancreas: Characterization of a novel orthotopic pancreatic ductal adenocarcinoma tumor model for interventional studies. PLoS One 2024; 19:e0300723. [PMID: 38547077 PMCID: PMC10977750 DOI: 10.1371/journal.pone.0300723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 03/02/2024] [Indexed: 04/02/2024] Open
Abstract
Preclinical models of cancer are vital for assessing and predicting efficacies and toxicities of novel treatments prior to testing in human subjects. Current pancreatic tumor models exhibit variable growth rates, unpredictable tumor size after implantation in non-native tissues, or require surgical implantation. Surgical implantation in the pancreas may produce not only unpredictable tumor uptake but could also elicit additional inflammatory responses. In searching for a pancreatic carcinoma cell that can be introduced into a mouse via simple injection, we found that Pan02, a murine ductal pancreatic adenocarcinoma derived from a pancreatic lesion of a C57BL/6 mouse, inoculated peritoneally can consistently produce pancreatic tumors. This intraperitoneal, but not intravenous, introduction of Pan02 cells leads to the attachment and growth of Pan02 in the pancreas before spreading to other tissues. Time-course tissue analysis indicates that the Pan02 cells first find, infiltrate, and grow within the pancreas, producing a pancreatic tumor model. This model appears to mimic pancreatic cancer development in humans and is the first reported use of Pan02 cells to produce orthotopic pancreatic and metastatic neoplasms in a mouse model without the need for tumor implantation within matrices or survival surgeries. This orthotopic pancreatic tumor model, with consistent tumor uptake, synchronized tumor development and survival, and predictable outcomes may enable and accelerate the preclinical evaluation of treatment candidates for pancreatic cancer.
Collapse
Affiliation(s)
- James I. Griffin
- Departments of Pharmaceutics, University of Washington, Seattle, Washington, United States of America
| | - Xinyue Chen
- Departments of Pharmaceutics, University of Washington, Seattle, Washington, United States of America
| | - Luqi Duan
- Departments of Pharmaceutics, University of Washington, Seattle, Washington, United States of America
| | - Qingxin Mu
- Departments of Pharmaceutics, University of Washington, Seattle, Washington, United States of America
| | - Rodney J. Y. Ho
- Departments of Pharmaceutics, University of Washington, Seattle, Washington, United States of America
- Departments of Bioengineering, University of Washington, Seattle, Washington, United States of America
| |
Collapse
|
7
|
Goad DW, Nesmelova AY, Yohe LR, Grdzelishvili VZ. Intertumoral heterogeneity impacts oncolytic vesicular stomatitis virus efficacy in mouse pancreatic cancer cells. J Virol 2023; 97:e0100523. [PMID: 37671865 PMCID: PMC10537684 DOI: 10.1128/jvi.01005-23] [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: 07/07/2023] [Accepted: 07/11/2023] [Indexed: 09/07/2023] Open
Abstract
Oncolytic virus (OV) therapy is a promising virus-based approach against various malignancies, including pancreatic ductal adenocarcinoma (PDAC). Our previous studies demonstrated that human PDAC cell lines are highly variable in their permissiveness to OVs. Mouse PDAC cell lines, which are widely used for in vivo examination of the adaptive immune responses during OV and other cancer therapies, have never been examined systematically for the impact of intertumoral heterogeneity (the differences observed between tumors in different patients) on OV virus efficacy. Here, we examined phenotypically and genotypically three commonly used allograftable mouse PDAC cell lines (C57BL6 genetic background): Panc02 (derived from chemically induced PDAC; also known as Pan02), and two cell lines originated from PDACs developed in two different KPC (KrasG12D, Trp53R172H, and PDX-1-Cre) mouse models. Our study (i) characterized the ability of a widely used attenuated oncolytic vesicular stomatitis virus VSV-ΔM51-GFP to infect, replicate in, and kill mouse PDAC cells; (ii) examined their innate antiviral responses; (iii) compared their permissiveness to a non-attenuated VSV-Mwt-GFP and chemotherapeutic drugs; and (iv) analyzed their karyotype and exome. Mouse PDAC cell lines showed high divergence in their permissiveness to VSV-ΔM51-GFP, which negatively correlated with their abilities to mount innate antiviral responses, while all three cell lines were highly permissive to VSV-Mwt-GFP. No correlation was found between resistance to VSV-ΔM51-GFP and chemotherapy. Also, mouse PDAC cell lines showed high divergence in their karyotype and exome. The exome analysis demonstrated that more VSV-ΔM51-GFP-permissive mouse PDAC cell lines harbor mutations in multiple important antiviral genes, such as TYK2, JAK2, and JAK3. IMPORTANCE Oncolytic virus (OV) therapy is a promising virus-based approach against various malignancies, including pancreatic ductal adenocarcinoma (PDAC). Our previous studies using various human PDAC cell lines demonstrated that they are highly variable in their permissiveness to OVs. In this study, we examined phenotypically and genotypically three commonly used allograftable mouse PDAC cell lines, which are widely used for in vivo examination of the adaptive immune responses during cancer therapies. Mouse PDAC cell lines showed high divergence in their permissiveness to oncolytic vesicular stomatitis virus (VSV), which negatively correlated with their abilities to mount innate antiviral responses. Also, we discovered that more VSV-permissive mouse PDAC cell lines harbor mutations in multiple important antiviral genes, such as TYK2, JAK2, and JAK3. Our study provides essential information about three model mouse PDAC cell lines and proposes a novel platform to study OV-based therapies against different PDACs in immunocompetent mice.
Collapse
Affiliation(s)
- Dakota W. Goad
- Department of Biological Sciences, University of North Carolina at Charlotte, Charlotte, North Carolina, USA
| | - Anna Y. Nesmelova
- Department of Biological Sciences, University of North Carolina at Charlotte, Charlotte, North Carolina, USA
| | - Laurel R. Yohe
- Department of Bioinformatics and Genomics, University of North Carolina at Charlotte, Charlotte, North Carolina, USA
| | - Valery Z. Grdzelishvili
- Department of Biological Sciences, University of North Carolina at Charlotte, Charlotte, North Carolina, USA
- School of Data Science, University of North Carolina at Charlotte, Charlotte, North Carolina, USA
| |
Collapse
|
8
|
Fan YC, Fong YC, Kuo CT, Li CW, Chen WY, Lin JD, Bürtin F, Linnebacher M, Bui QT, Lee KD, Tsai YC. Tumor-derived interleukin-1 receptor antagonist exhibits immunosuppressive functions and promotes pancreatic cancer. Cell Biosci 2023; 13:147. [PMID: 37563620 PMCID: PMC10416534 DOI: 10.1186/s13578-023-01090-8] [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: 04/21/2023] [Accepted: 07/19/2023] [Indexed: 08/12/2023] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDA) is a pernicious disease characterized by an immunosuppressive milieu that is unresponsive to current immunotherapies. Interleukin-1 receptor antagonist (IL-1Ra) is a natural anti-inflammatory cytokine; however, its contribution to cancer pathogenesis and immunosuppression remains elusive. In this research, we investigated the role and mechanism of IL-1Ra in malignant progression of PDA. RESULTS Through analyzing clinical dataset and examining the pathological tumor tissues and serum samples, we have demonstrated that IL-1Ra expression is elevated in human PDA and positively associated with malignant progression of PDA. To study the biological function of IL-1Ra in tumors, we generated a set of mouse pancreatic cancer cell lines with a knockout (KO) of the Il1rn gene, encoding IL-1Ra, and compared the tumor growth rates in immune-competent and immune-deficient mice. We found that the Il1rn KO cells exhibited greater tumor inhibition in immune-competent mice, highlighting the crucial role of a functional immune system in Il1rn KO-mediated anti-tumor response. Consistently, we found an increase in CD8+ T cells and a decrease in CD11b+Ly6G- immunosuppressive mononuclear population in the tumor microenvironment of Il1rn KO-derived tumors. To monitor the inhibitory effects of IL-1Ra on immune cells, we utilized a luciferase-based reporter CD4+ T cell line and splenocytes, which were derived from transgenic mice expressing ovalbumin-specific T cell receptors in CD8+ T cells, and mice immunized with ovalbumin. We showed that IL-1Ra suppressed T cell receptor signaling and inhibited antigen-specific interferon-γ (IFN-γ) secretion and cytolytic activity in splenocytes. CONCLUSIONS Our findings illustrate the immunosuppressive properties of the natural anti-inflammatory cytokine IL-1Ra, and provide a rationale for considering IL-1Ra-targeted therapies in the treatment of PDA.
Collapse
Affiliation(s)
- Yu-Ching Fan
- Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei, Taiwan
| | - Yu-Cin Fong
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Chun-Tse Kuo
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Chia-Wei Li
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Wei-Yu Chen
- Department of Pathology, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
- Department of Pathology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Jian-Da Lin
- Department of Biochemical Science and Technology, College of Life Science, National Taiwan University, Taipei City, 10617, Taiwan
- Center for Computational and Systems Biology, National Taiwan University, Taipei City, 10617, Taiwan
| | - Florian Bürtin
- Clinic of General Surgery, University Medical Center Rostock, Schillingallee 35, 18057, Rostock, Germany
| | - Michael Linnebacher
- Clinic of General Surgery, Molecular Oncology and Immunotherapy, University Medical Center Rostock, Schillingallee 69, 18057, Rostock, Germany
| | - Quoc Thang Bui
- International Ph.D. Program for Cell Therapy and Regeneration Medicine (IPCTRM), School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Kuan-Der Lee
- International Ph.D. Program for Cell Therapy and Regeneration Medicine (IPCTRM), School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Post-Baccalaureate Medicine, College of Medicine, Natioanl Chung Hsing University, Taichung, Taiwan
- Cell Therapy and Regenerative Medicine Center and Comprehensive Cancer Center, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Yuan-Chin Tsai
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan.
- Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
9
|
Medler TR, Blair TC, Alice AF, Dowdell AK, Piening BD, Crittenden MR, Gough MJ. Myeloid MyD88 restricts CD8 + T cell response to radiation therapy in pancreatic cancer. Sci Rep 2023; 13:8634. [PMID: 37244938 PMCID: PMC10224952 DOI: 10.1038/s41598-023-35834-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: 09/23/2022] [Accepted: 05/24/2023] [Indexed: 05/29/2023] Open
Abstract
Radiation therapy induces immunogenic cell death in cancer cells, whereby released endogenous adjuvants are sensed by immune cells to direct adaptive immune responses. TLRs expressed on several immune subtypes recognize innate adjuvants to direct downstream inflammatory responses in part via the adapter protein MyD88. We generated Myd88 conditional knockout mice to interrogate its contribution to the immune response to radiation therapy in distinct immune populations in pancreatic cancer. Surprisingly, Myd88 deletion in Itgax (CD11c)-expressing dendritic cells had little discernable effects on response to RT in pancreatic cancer and elicited normal T cell responses using a prime/boost vaccination strategy. Myd88 deletion in Lck-expressing T cells resulted in similar or worsened responses to radiation therapy compared to wild-type mice and lacked antigen-specific CD8+ T cell responses from vaccination, similar to observations in Myd88-/- mice. Lyz2-specific loss of Myd88 in myeloid populations rendered tumors more susceptible to radiation therapy and elicited normal CD8+ T cell responses to vaccination. scRNAseq in Lyz2-Cre/Myd88fl/fl mice revealed gene signatures in macrophages and monocytes indicative of enhanced type I and II interferon responses, and improved responses to RT were dependent on CD8+ T cells and IFNAR1. Together, these data implicate MyD88 signaling in myeloid cells as a critical source of immunosuppression that hinders adaptive immune tumor control following radiation therapy.
Collapse
Affiliation(s)
- Terry R Medler
- Earle A. Chiles Research Institute, Providence Cancer Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, 4805 NE Glisan Street, Suite 2N100, Portland, OR, 97213, USA
| | - Tiffany C Blair
- Earle A. Chiles Research Institute, Providence Cancer Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, 4805 NE Glisan Street, Suite 2N100, Portland, OR, 97213, USA
| | - Alejandro F Alice
- Earle A. Chiles Research Institute, Providence Cancer Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, 4805 NE Glisan Street, Suite 2N100, Portland, OR, 97213, USA
| | - Alexa K Dowdell
- Earle A. Chiles Research Institute, Providence Cancer Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, 4805 NE Glisan Street, Suite 2N100, Portland, OR, 97213, USA
| | - Brian D Piening
- Earle A. Chiles Research Institute, Providence Cancer Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, 4805 NE Glisan Street, Suite 2N100, Portland, OR, 97213, USA
| | - Marka R Crittenden
- Earle A. Chiles Research Institute, Providence Cancer Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, 4805 NE Glisan Street, Suite 2N100, Portland, OR, 97213, USA
- The Oregon Clinic, Portland, OR, USA
| | - Michael J Gough
- Earle A. Chiles Research Institute, Providence Cancer Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, 4805 NE Glisan Street, Suite 2N100, Portland, OR, 97213, USA.
| |
Collapse
|
10
|
Medler TR, Kramer G, Bambina S, Gunderson AJ, Alice A, Blair T, Zebertavage L, Duhen T, Duhen R, Young K, Crittenden MR, Gough MJ. Tumor resident memory CD8 T cells and concomitant tumor immunity develop independently of CD4 help. Sci Rep 2023; 13:6277. [PMID: 37072485 PMCID: PMC10113239 DOI: 10.1038/s41598-023-33508-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 04/13/2023] [Indexed: 05/03/2023] Open
Abstract
Tissue resident memory (Trm) CD8 T cells infiltrating tumors represent an enriched population of tumor antigen-specific T cells, and their presence is associated with improved outcomes in patients. Using genetically engineered mouse pancreatic tumor models we demonstrate that tumor implantation generates a Trm niche that is dependent on direct antigen presentation by cancer cells. However, we observe that initial CCR7-mediated localization of CD8 T cells to tumor draining lymph nodes is required to subsequently generate CD103+ CD8 T cells in tumors. We observe that the formation of CD103+ CD8 T cells in tumors is dependent on CD40L but independent of CD4 T cells, and using mixed chimeras we show that CD8 T cells can provide their own CD40L to permit CD103+ CD8 T cell differentiation. Finally, we show that CD40L is required to provide systemic protection against secondary tumors. These data suggest that CD103+ CD8 T cell formation in tumors can occur independent of the two-factor authentication provided by CD4 T cells and highlight CD103+ CD8 T cells as a distinct differentiation decision from CD4-dependent central memory.
Collapse
Affiliation(s)
- Terry R Medler
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, NE Glisan St., Portland, OR, 480597213, USA
| | - Gwen Kramer
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, NE Glisan St., Portland, OR, 480597213, USA
| | - Shelly Bambina
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, NE Glisan St., Portland, OR, 480597213, USA
| | - Andrew J Gunderson
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, NE Glisan St., Portland, OR, 480597213, USA
- Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center - Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, The OH State University, Columbus, OH, 43210, USA
| | - Alejandro Alice
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, NE Glisan St., Portland, OR, 480597213, USA
| | - Tiffany Blair
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, NE Glisan St., Portland, OR, 480597213, USA
| | - Lauren Zebertavage
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, NE Glisan St., Portland, OR, 480597213, USA
| | - Thomas Duhen
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, NE Glisan St., Portland, OR, 480597213, USA
| | - Rebekka Duhen
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, NE Glisan St., Portland, OR, 480597213, USA
| | - Kristina Young
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, NE Glisan St., Portland, OR, 480597213, USA
- The Oregon Clinic, Portland, OR, 97213, USA
| | - Marka R Crittenden
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, NE Glisan St., Portland, OR, 480597213, USA
- The Oregon Clinic, Portland, OR, 97213, USA
| | - Michael J Gough
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, NE Glisan St., Portland, OR, 480597213, USA.
| |
Collapse
|
11
|
Revskij D, Runst J, Umstätter C, Ehlers L, Rohde S, Zechner D, Bastian M, Müller-Hilke B, Fuellen G, Henze L, Murua Escobar H, Junghanss C, Kowald A, Walter U, Köhling R, Wolkenhauer O, Jaster R. Uncoupling protein 2 deficiency of non-cancerous tissues inhibits the progression of pancreatic cancer in mice. Hepatobiliary Pancreat Dis Int 2023; 22:190-199. [PMID: 36549966 DOI: 10.1016/j.hbpd.2022.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 12/08/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is a disease of the elderly mostly because its development from preneoplastic lesions depends on the accumulation of gene mutations and epigenetic alterations over time. How aging of non-cancerous tissues of the host affects tumor progression, however, remains largely unknown. METHODS We took advantage of a model of accelerated aging, uncoupling protein 2-deficient (Ucp2 knockout, Ucp2 KO) mice, to investigate the growth of orthotopically transplanted Ucp2 wild-type (WT) PDAC cells (cell lines Panc02 and 6606PDA) in vivo and to study strain-dependent differences of the PDAC microenvironment. RESULTS Measurements of tumor weights and quantification of proliferating cells indicated a significant growth advantage of Panc02 and 6606PDA cells in WT mice compared to Ucp2 KO mice. In tumors in the knockout strain, higher levels of interferon-γ mRNA despite similar numbers of tumor-infiltrating T cells were observed. 6606PDA cells triggered a stronger stromal reaction in Ucp2 KO mice than in WT animals. Accordingly, pancreatic stellate cells from Ucp2 KO mice proliferated at a higher rate than cells of the WT strain when they were incubated with conditioned media from PDAC cells. CONCLUSIONS Ucp2 modulates PDAC microenvironment in a way that favors tumor progression and implicates an altered stromal response as one of the underlying mechanisms.
Collapse
Affiliation(s)
- Denis Revskij
- Department of Medicine II, Division of Gastroenterology, Rostock University Medical Center, Rostock, Germany
| | - Jakob Runst
- Department of Medicine II, Division of Gastroenterology, Rostock University Medical Center, Rostock, Germany
| | - Camilla Umstätter
- Department of Medicine II, Division of Gastroenterology, Rostock University Medical Center, Rostock, Germany
| | - Luise Ehlers
- Department of Medicine II, Division of Gastroenterology, Rostock University Medical Center, Rostock, Germany
| | - Sarah Rohde
- Department of Medicine II, Division of Gastroenterology, Rostock University Medical Center, Rostock, Germany
| | - Dietmar Zechner
- Rudolf-Zenker-Institute of Experimental Surgery, Rostock University Medical Center, Rostock, Germany
| | - Manuela Bastian
- Institute of Clinical Chemistry and Laboratory Medicine, Rostock University Medical Center, Rostock, Germany
| | - Brigitte Müller-Hilke
- Facility for Cell Sorting and Cell Analysis and Institute of Immunology, Rostock University Medical Center, Rostock, Germany
| | - Georg Fuellen
- Institute for Biostatistics and Informatics in Medicine and Ageing Research, Rostock University Medical Center, Rostock, Germany
| | - Larissa Henze
- Department of Medicine, Clinic III, Hematology, Oncology, Palliative Medicine, Rostock University Medical Center, Rostock, Germany
| | - Hugo Murua Escobar
- Department of Medicine, Clinic III, Hematology, Oncology, Palliative Medicine, Rostock University Medical Center, Rostock, Germany
| | - Christian Junghanss
- Department of Medicine, Clinic III, Hematology, Oncology, Palliative Medicine, Rostock University Medical Center, Rostock, Germany
| | - Axel Kowald
- Institute for Biostatistics and Informatics in Medicine and Ageing Research, Rostock University Medical Center, Rostock, Germany
| | - Uwe Walter
- Department of Neurology, Rostock University Medical Center, Rostock, Germany
| | - Rüdiger Köhling
- Oscar Langendorff Institute of Physiology, Rostock University Medical Center, Rostock, Germany
| | - Olaf Wolkenhauer
- Department of Systems Biology and Bioinformatics, University of Rostock, Rostock, Germany
| | - Robert Jaster
- Department of Medicine II, Division of Gastroenterology, Rostock University Medical Center, Rostock, Germany.
| |
Collapse
|
12
|
Czaplinska D, Ialchina R, Andersen HB, Yao J, Stigliani A, Dannesboe J, Flinck M, Chen X, Mitrega J, Gnosa SP, Dmytriyeva O, Alves F, Napp J, Sandelin A, Pedersen SF. Crosstalk between tumor acidosis, p53 and extracellular matrix regulates pancreatic cancer aggressiveness. Int J Cancer 2023; 152:1210-1225. [PMID: 36408933 PMCID: PMC10108304 DOI: 10.1002/ijc.34367] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 10/14/2022] [Accepted: 11/07/2022] [Indexed: 11/22/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an extremely aggressive malignancy with minimal treatment options and a global rise in prevalence. PDAC is characterized by frequent driver mutations including KRAS and TP53 (p53), and a dense, acidic tumor microenvironment (TME). The relation between genotype and TME in PDAC development is unknown. Strikingly, when wild type (WT) Panc02 PDAC cells were adapted to growth in an acidic TME and returned to normal pH to mimic invasive cells escaping acidic regions, they displayed a strong increase of aggressive traits such as increased growth in 3-dimensional (3D) culture, adhesion-independent colony formation and invasive outgrowth. This pattern of acidosis-induced aggressiveness was observed in 3D spheroid culture as well as upon organotypic growth in matrigel, collagen-I and combination thereof, mimicking early and later stages of PDAC development. Acid-adaptation-induced gain of cancerous traits was further increased by p53 knockout (KO), but only in specific extracellular matrix (ECM) compositions. Akt- and Transforming growth factor-β (TGFβ) signaling, as well as expression of the Na+ /H+ exchanger NHE1, were increased by acid adaptation. Whereas Akt inhibition decreased spheroid growth regardless of treatment and genotype, stimulation with TGFβI increased growth of WT control spheroids, and inhibition of TGFβ signaling tended to limit growth under acidic conditions only. Our results indicate that a complex crosstalk between tumor acidosis, ECM composition and genotype contributes to PDAC development. The findings may guide future strategies for acidosis-targeted therapies.
Collapse
Affiliation(s)
- Dominika Czaplinska
- Section for Cell Biology and Physiology, Department of Biology, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Renata Ialchina
- Section for Cell Biology and Physiology, Department of Biology, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Henriette Berg Andersen
- Section for Cell Biology and Physiology, Department of Biology, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Jiayi Yao
- Section for Computational and RNA Biology, Department of Biology, Faculty of Science, University of Copenhagen, Copenhagen, Denmark.,Biotech Research and Innovation Centre (BRIC), Faculty of Health, University of Copenhagen, Copenhagen, Denmark
| | - Arnaud Stigliani
- Section for Computational and RNA Biology, Department of Biology, Faculty of Science, University of Copenhagen, Copenhagen, Denmark.,Biotech Research and Innovation Centre (BRIC), Faculty of Health, University of Copenhagen, Copenhagen, Denmark
| | - Johs Dannesboe
- Section for Cell Biology and Physiology, Department of Biology, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Mette Flinck
- Section for Cell Biology and Physiology, Department of Biology, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Xiaoming Chen
- Section for Cell Biology and Physiology, Department of Biology, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Jakub Mitrega
- Max-Planck-Institute for Multidisciplinary Sciences, Goettingen, Germany.,Institute for Diagnostic and Interventional Radiology, University Medical Center Goettingen, Goettingen, Germany
| | - Sebastian Peter Gnosa
- Biotech Research and Innovation Centre (BRIC), Faculty of Health, University of Copenhagen, Copenhagen, Denmark
| | - Oksana Dmytriyeva
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health, University of Copenhagen, Copenhagen, Denmark
| | - Frauke Alves
- Max-Planck-Institute for Multidisciplinary Sciences, Goettingen, Germany.,Institute for Diagnostic and Interventional Radiology, University Medical Center Goettingen, Goettingen, Germany.,Clinic of Haematology and Medical Oncology, University Medical Center Goettingen, Goettingen, Germany
| | - Joanna Napp
- Max-Planck-Institute for Multidisciplinary Sciences, Goettingen, Germany.,Institute for Diagnostic and Interventional Radiology, University Medical Center Goettingen, Goettingen, Germany.,Clinic of Haematology and Medical Oncology, University Medical Center Goettingen, Goettingen, Germany
| | - Albin Sandelin
- Section for Computational and RNA Biology, Department of Biology, Faculty of Science, University of Copenhagen, Copenhagen, Denmark.,Biotech Research and Innovation Centre (BRIC), Faculty of Health, University of Copenhagen, Copenhagen, Denmark
| | - Stine Falsig Pedersen
- Section for Cell Biology and Physiology, Department of Biology, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
13
|
Araki H, Tazawa H, Kanaya N, Kajiwara Y, Yamada M, Hashimoto M, Kikuchi S, Kuroda S, Yoshida R, Umeda Y, Urata Y, Kagawa S, Fujiwara T. Oncolytic virus-mediated p53 overexpression promotes immunogenic cell death and efficacy of PD-1 blockade in pancreatic cancer. Mol Ther Oncolytics 2022; 27:3-13. [PMID: 36212775 PMCID: PMC9513735 DOI: 10.1016/j.omto.2022.09.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Accepted: 09/09/2022] [Indexed: 10/26/2022] Open
|
14
|
PD-L1-directed PlGF/VEGF blockade synergizes with chemotherapy by targeting CD141 + cancer-associated fibroblasts in pancreatic cancer. Nat Commun 2022; 13:6292. [PMID: 36272973 PMCID: PMC9588060 DOI: 10.1038/s41467-022-33991-6] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 10/11/2022] [Indexed: 12/25/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has a poor 5-year overall survival rate. Patients with PDAC display limited benefits after undergoing chemotherapy or immunotherapy modalities. Herein, we reveal that chemotherapy upregulates placental growth factor (PlGF), which directly activates cancer-associated fibroblasts (CAFs) to induce fibrosis-associated collagen deposition in PDAC. Patients with poor prognosis have high PIGF/VEGF expression and an increased number of PIGF/VEGF receptor-expressing CAFs, associated with enhanced collagen deposition. We also develop a multi-paratopic VEGF decoy receptor (Ate-Grab) by fusing the single-chain Fv of atezolizumab (anti-PD-L1) to VEGF-Grab to target PD-L1-expressing CAFs. Ate-Grab exerts anti-tumor and anti-fibrotic effects in PDAC models via the PD-L1-directed PlGF/VEGF blockade. Furthermore, Ate-Grab synergizes with gemcitabine by relieving desmoplasia. Single-cell RNA sequencing identifies that a CD141+ CAF population is reduced upon Ate-Grab and gemcitabine combination treatment. Overall, our results elucidate the mechanism underlying chemotherapy-induced fibrosis in PDAC and highlight a combinatorial therapeutic strategy for desmoplastic cancers.
Collapse
|
15
|
Deborde S, Gusain L, Powers A, Marcadis A, Yu Y, Chen CH, Frants A, Kao E, Tang LH, Vakiani E, Amisaki M, Balachandran VP, Calo A, Omelchenko T, Jessen KR, Reva B, Wong RJ. Reprogrammed Schwann Cells Organize into Dynamic Tracks that Promote Pancreatic Cancer Invasion. Cancer Discov 2022; 12:2454-2473. [PMID: 35881881 PMCID: PMC9533012 DOI: 10.1158/2159-8290.cd-21-1690] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 06/16/2022] [Accepted: 07/22/2022] [Indexed: 01/07/2023]
Abstract
Nerves are a component of the tumor microenvironment contributing to cancer progression, but the role of cells from nerves in facilitating cancer invasion remains poorly understood. Here we show that Schwann cells (SC) activated by cancer cells collectively function as tumor-activated Schwann cell tracks (TAST) that promote cancer cell migration and invasion. Nonmyelinating SCs form TASTs and have cell gene expression signatures that correlate with diminished survival in patients with pancreatic ductal adenocarcinoma. In TASTs, dynamic SCs form tracks that serve as cancer pathways and apply forces on cancer cells to enhance cancer motility. These SCs are activated by c-Jun, analogous to their reprogramming during nerve repair. This study reveals a mechanism of cancer cell invasion that co-opts a wound repair process and exploits the ability of SCs to collectively organize into tracks. These findings establish a novel paradigm of how cancer cells spread and reveal therapeutic opportunities. SIGNIFICANCE How the tumor microenvironment participates in pancreatic cancer progression is not fully understood. Here, we show that SCs are activated by cancer cells and collectively organize into tracks that dynamically enable cancer invasion in a c-Jun-dependent manner. See related commentary by Amit and Maitra, p. 2240. This article is highlighted in the In This Issue feature, p. 2221.
Collapse
Affiliation(s)
- Sylvie Deborde
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Laxmi Gusain
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ann Powers
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Andrea Marcadis
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Yasong Yu
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Chun-Hao Chen
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Anna Frants
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Elizabeth Kao
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Laura H. Tang
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Efsevia Vakiani
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Masataka Amisaki
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Vinod P. Balachandran
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, New York
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
- Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Annalisa Calo
- Institute for Bioengineering of Catalonia, Barcelona, Spain
| | - Tatiana Omelchenko
- Laboratory of Mammalian Cell Biology and Development, The Rockefeller University, New York, New York
| | - Kristjan R. Jessen
- Cell and Developmental Biology, University College London, London, United Kingdom
| | - Boris Reva
- Department of Genetics and Genomics Sciences, Mount Sinai Medical Center, New York, New York
| | - Richard J. Wong
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
16
|
DNA Polymerase Theta Plays a Critical Role in Pancreatic Cancer Development and Metastasis. Cancers (Basel) 2022; 14:cancers14174077. [PMID: 36077614 PMCID: PMC9454495 DOI: 10.3390/cancers14174077] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 08/12/2022] [Accepted: 08/16/2022] [Indexed: 11/16/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC), due to its genomic heterogeneity and lack of effective treatment, despite decades of intensive research, will become the second leading cause of cancer-related deaths by 2030. Step-wise acquisition of mutations, due to genomic instability, is considered to drive the development of PDAC; the KRAS mutation occurs in 95 to 100% of human PDAC, and is already detectable in early premalignant lesions designated as pancreatic intraepithelial neoplasia (PanIN). This mutation is possibly the key event leading to genomic instability and PDAC development. Our study aimed to investigate the role of the error-prone DNA double-strand breaks (DSBs) repair pathway, alt-EJ, in the presence of the KRAS G12D mutation in pancreatic cancer development. Our findings show that oncogenic KRAS contributes to increasing the expression of Polθ, Lig3, and Mre11, key components of alt-EJ in both mouse and human PDAC models. We further confirm increased catalytic activity of alt-EJ in a mouse and human model of PDAC bearing the KRAS G12D mutation. Subsequently, we focused on estimating the impact of alt-EJ inactivation by polymerase theta (Polθ) deletion on pancreatic cancer development, and survival in genetically engineered mouse models (GEMMs) and cancer patients. Here, we show that even though Polθ deficiency does not fully prevent the development of pancreatic cancer, it significantly delays the onset of PanIN formation, prolongs the overall survival of experimental mice, and correlates with the overall survival of pancreatic cancer patients in the TCGA database. Our study clearly demonstrates the role of alt-EJ in the development of PDAC, and alt-EJ may be an attractive therapeutic target for pancreatic cancer patients.
Collapse
|
17
|
Lu C, Liu Z, Klement JD, Yang D, Merting AD, Poschel D, Albers T, Waller JL, Shi H, Liu K. WDR5-H3K4me3 epigenetic axis regulates OPN expression to compensate PD-L1 function to promote pancreatic cancer immune escape. J Immunother Cancer 2021; 9:e002624. [PMID: 34326167 PMCID: PMC8323468 DOI: 10.1136/jitc-2021-002624] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/29/2021] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Despite PD-L1 (Programmed death receptor ligand-1) expression on tumor cells and cytotoxic T lymphocytes tumor infiltration in the tumor microenvironment, human pancreatic cancer stands out as one of the human cancers that does not respond to immune checkpoint inhibitor (ICI) immunotherapy. Epigenome dysregulation has emerged as a major mechanism in T cell exhaustion and non-response to ICI immunotherapy, we, therefore, aimed at testing the hypothesis that an epigenetic mechanism compensates PD-L1 function to render pancreatic cancer non-response to ICI immunotherapy. METHODS Two orthotopic pancreatic tumor mouse models were used for chromatin immunoprecipitation-Seq and RNA-Seq to identify genome-wide dysregulation of H3K4me3 and gene expression. Human pancreatic tumor and serum were analyzed for osteopontin (OPN) protein level and for correlation with patient prognosis. OPN and PD-L1 cellular location were determined in the tumors using flow cytometry. The function of WDR5-H3K4me3 axis in OPN expression were determined by Western blotting. The function of H3K4me3-OPN axis in pancreatic cancer immune escape and response to ICI immunotherapy was determined in an orthotopic pancreatic tumor mouse model. RESULTS Mouse pancreatic tumors have a genome-wide increase in H3K4me3 deposition as compared with normal pancreas. OPN and its receptor CD44 were identified being upregulated in pancreatic tumors by their promoter H3K4me3 deposition. OPN protein is increased in both tumor cells and tumor-infiltrating immune cells in human pancreatic carcinoma and is inversely correlated with pancreatic cancer patient survival. OPN is primarily expressed in tumor cells and monocytic myeloid-derived suppressor cells (M-MDSCs), whereas PD-L1 is expressed in tumor cells, M-MDSCs, polymorphonuclear MDSCs and tumor-associated macrophages. WDR5 is essential for H3K4me3-specific histone methyltransferase activity that regulates OPN expression in tumor cells and MDSCs. Inhibition of WDR5 significantly decreased OPN protein level. Inhibition of WDR5 or knocking out of OPN suppressed orthotopic mouse pancreatic tumor growth. Inhibition of WDR5 also significantly increased efficacy of anti-PD-1 immunotherapy in suppression of mouse pancreatic tumor growth in vivo. CONCLUSIONS OPN compensates PD-L1 function to promote pancreatic cancer immune escape. Pharmacological inhibition of the WDR5-H3K4me3 epigenetic axis is effective in suppressing pancreatic tumor immune escape and in improving efficacy of anti-PD-1 immunotherapy in pancreatic cancer.
Collapse
Affiliation(s)
- Chunwan Lu
- School of Life Sciences, Tianjin University, Tianjin, China
| | - Zhuoqi Liu
- Department of Biochemistry and Molecular Biology, Nanchang University, Nanchang, China
| | - John D Klement
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta, Georgia, USA
- Georgia Cancer Center, Medical College of Georgia, Augusta, Georgia, USA
- Charlie Norwood VA Medical Center, Augusta, Georgia, USA
| | - Dafeng Yang
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta, Georgia, USA
- Georgia Cancer Center, Medical College of Georgia, Augusta, Georgia, USA
- Charlie Norwood VA Medical Center, Augusta, Georgia, USA
| | - Alyssa D Merting
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta, Georgia, USA
- Georgia Cancer Center, Medical College of Georgia, Augusta, Georgia, USA
- Charlie Norwood VA Medical Center, Augusta, Georgia, USA
| | - Dakota Poschel
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta, Georgia, USA
- Georgia Cancer Center, Medical College of Georgia, Augusta, Georgia, USA
- Charlie Norwood VA Medical Center, Augusta, Georgia, USA
| | - Thomas Albers
- Georgia Cancer Center, Medical College of Georgia, Augusta, Georgia, USA
| | - Jennifer L Waller
- Department of Population Health Science, Medical College of Georgia, Augusta, Georgia, USA
| | - Huidong Shi
- Georgia Cancer Center, Medical College of Georgia, Augusta, Georgia, USA
| | - Kebin Liu
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta, Georgia, USA
- Georgia Cancer Center, Medical College of Georgia, Augusta, Georgia, USA
- Charlie Norwood VA Medical Center, Augusta, Georgia, USA
| |
Collapse
|
18
|
Heinrich MA, Mostafa AMRH, Morton JP, Hawinkels LJAC, Prakash J. Translating complexity and heterogeneity of pancreatic tumor: 3D in vitro to in vivo models. Adv Drug Deliv Rev 2021; 174:265-293. [PMID: 33895214 DOI: 10.1016/j.addr.2021.04.018] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 04/16/2021] [Accepted: 04/19/2021] [Indexed: 02/08/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an extremely aggressive type of cancer with an overall survival rate of less than 7-8%, emphasizing the need for novel effective therapeutics against PDAC. However only a fraction of therapeutics which seemed promising in the laboratory environment will eventually reach the clinic. One of the main reasons behind this low success rate is the complex tumor microenvironment (TME) of PDAC, a highly fibrotic and dense stroma surrounding tumor cells, which supports tumor progression as well as increases the resistance against the treatment. In particular, the growing understanding of the PDAC TME points out a different challenge in the development of efficient therapeutics - a lack of biologically relevant in vitro and in vivo models that resemble the complexity and heterogeneity of PDAC observed in patients. The purpose and scope of this review is to provide an overview of the recent developments in different in vitro and in vivo models, which aim to recapitulate the complexity of PDAC in a laboratory environment, as well to describe how 3D in vitro models can be integrated into drug development pipelines that are already including sophisticated in vivo models. Hereby a special focus will be given on the complexity of in vivo models and the challenges in vitro models face to reach the same levels of complexity in a controllable manner. First, a brief introduction of novel developments in two dimensional (2D) models and ex vivo models is provided. Next, recent developments in three dimensional (3D) in vitro models are described ranging from spheroids, organoids, scaffold models, bioprinted models to organ-on-chip models including a discussion on advantages and limitations for each model. Furthermore, we will provide a detailed overview on the current PDAC in vivo models including chemically-induced models, syngeneic and xenogeneic models, highlighting hetero- and orthotopic, patient-derived tissues (PDX) models, and genetically engineered mouse models. Finally, we will provide a discussion on overall limitations of both, in vitro and in vivo models, and discuss necessary steps to overcome these limitations to reach an efficient drug development pipeline, as well as discuss possibilities to include novel in silico models in the process.
Collapse
Affiliation(s)
- Marcel A Heinrich
- Department of Biomaterials Science and Technology, Section Targeted Therapeutics, Technical Medical Centre, University of Twente, 7500AE Enschede, the Netherlands
| | - Ahmed M R H Mostafa
- Department of Biomaterials Science and Technology, Section Targeted Therapeutics, Technical Medical Centre, University of Twente, 7500AE Enschede, the Netherlands
| | - Jennifer P Morton
- Cancer Research UK, Beatson Institute, Garscube Estate, Switchback Rd, Glasgow G61 1BD, UK; Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Rd, Glasgow G61 1QH, UK
| | - Lukas J A C Hawinkels
- Department of Gastroenterology-Hepatology, Leiden University Medical Centre, PO-box 9600, 2300 RC Leiden, the Netherlands
| | - Jai Prakash
- Department of Biomaterials Science and Technology, Section Targeted Therapeutics, Technical Medical Centre, University of Twente, 7500AE Enschede, the Netherlands.
| |
Collapse
|
19
|
Doello K, Mesas C, Quiñonero F, Perazzoli G, Cabeza L, Prados J, Melguizo C, Ortiz R. The Antitumor Activity of Sodium Selenite Alone and in Combination with Gemcitabine in Pancreatic Cancer: An In Vitro and In Vivo Study. Cancers (Basel) 2021; 13:cancers13133169. [PMID: 34201986 PMCID: PMC8268835 DOI: 10.3390/cancers13133169] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 06/17/2021] [Accepted: 06/22/2021] [Indexed: 01/19/2023] Open
Abstract
Sodium selenite acts by depleting enzymes that protect against cellular oxidative stress. To determine its effect alone or in combination with gemcitabine (GMZ) in pancreatic cancer, we used PANC-1 and Pan02 cell lines and C57BL mice bearing a Pan02-generated tumor. Our results demonstrated a significant inhibition of pancreatic cancer cell viability with the use of sodium selenite alone and a synergistic effect when associated with GMZ. The molecular mechanisms of the antitumor effect of sodium selenite alone involved apoptosis-inducing factor (AIF) and the expression of phospho-p38 in the combined therapy. In addition, sodium selenite alone and in association with GMZ significantly decreased the migration capacity and colony-forming ability, reduced tumor activity in multicellular tumor spheroids (MTS) and decreased sphere formation of cancer stem cells. In vivo studies demonstrated that combined therapy not only inhibited tumor growth (65%) compared to the untreated group but also relative to sodium selenite or GMZ used as monotherapy (up to 40%), increasing mice survival. These results were supported by the analysis of C57BL/6 albino mice bearing a Pan02-generated tumor, using the IVIS system. In conclusion, our results showed that sodium selenite is a potential agent for the improvement in the treatment of pancreatic cancer and should be considered for future human clinical trials.
Collapse
Affiliation(s)
- Kevin Doello
- Medical Oncology Service, Virgen de las Nieves Hospital, 18014 Granada, Spain;
- Instituto Biosanitario de Granada (ibs. GRANADA), 18014 Granada, Spain; (C.M.); (F.Q.); (G.P.); (L.C.); (C.M.); (R.O.)
| | - Cristina Mesas
- Instituto Biosanitario de Granada (ibs. GRANADA), 18014 Granada, Spain; (C.M.); (F.Q.); (G.P.); (L.C.); (C.M.); (R.O.)
- Center of Biomedical Research (CIBM), Institute of Biopathology and Regenerative Medicine (IBIMER), University of Granada, 18100 Granada, Spain
| | - Francisco Quiñonero
- Instituto Biosanitario de Granada (ibs. GRANADA), 18014 Granada, Spain; (C.M.); (F.Q.); (G.P.); (L.C.); (C.M.); (R.O.)
- Center of Biomedical Research (CIBM), Institute of Biopathology and Regenerative Medicine (IBIMER), University of Granada, 18100 Granada, Spain
- Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, 18071 Granada, Spain
| | - Gloria Perazzoli
- Instituto Biosanitario de Granada (ibs. GRANADA), 18014 Granada, Spain; (C.M.); (F.Q.); (G.P.); (L.C.); (C.M.); (R.O.)
- Center of Biomedical Research (CIBM), Institute of Biopathology and Regenerative Medicine (IBIMER), University of Granada, 18100 Granada, Spain
- Department of Medicine, Faculty of Health Sciences, University of Almería, 04120 Granada, Spain
| | - Laura Cabeza
- Instituto Biosanitario de Granada (ibs. GRANADA), 18014 Granada, Spain; (C.M.); (F.Q.); (G.P.); (L.C.); (C.M.); (R.O.)
- Center of Biomedical Research (CIBM), Institute of Biopathology and Regenerative Medicine (IBIMER), University of Granada, 18100 Granada, Spain
- Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, 18071 Granada, Spain
| | - Jose Prados
- Instituto Biosanitario de Granada (ibs. GRANADA), 18014 Granada, Spain; (C.M.); (F.Q.); (G.P.); (L.C.); (C.M.); (R.O.)
- Center of Biomedical Research (CIBM), Institute of Biopathology and Regenerative Medicine (IBIMER), University of Granada, 18100 Granada, Spain
- Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, 18071 Granada, Spain
- Correspondence:
| | - Consolacion Melguizo
- Instituto Biosanitario de Granada (ibs. GRANADA), 18014 Granada, Spain; (C.M.); (F.Q.); (G.P.); (L.C.); (C.M.); (R.O.)
- Center of Biomedical Research (CIBM), Institute of Biopathology and Regenerative Medicine (IBIMER), University of Granada, 18100 Granada, Spain
- Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, 18071 Granada, Spain
| | - Raul Ortiz
- Instituto Biosanitario de Granada (ibs. GRANADA), 18014 Granada, Spain; (C.M.); (F.Q.); (G.P.); (L.C.); (C.M.); (R.O.)
- Center of Biomedical Research (CIBM), Institute of Biopathology and Regenerative Medicine (IBIMER), University of Granada, 18100 Granada, Spain
- Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, 18071 Granada, Spain
| |
Collapse
|
20
|
Holbrook MC, Goad DW, Grdzelishvili VZ. Expanding the Spectrum of Pancreatic Cancers Responsive to Vesicular Stomatitis Virus-Based Oncolytic Virotherapy: Challenges and Solutions. Cancers (Basel) 2021; 13:1171. [PMID: 33803211 PMCID: PMC7963195 DOI: 10.3390/cancers13051171] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 03/05/2021] [Accepted: 03/05/2021] [Indexed: 02/07/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a devastating malignancy with poor prognosis and a dismal survival rate, expected to become the second leading cause of cancer-related deaths in the United States. Oncolytic virus (OV) is an anticancer approach that utilizes replication-competent viruses to preferentially infect and kill tumor cells. Vesicular stomatitis virus (VSV), one such OV, is already in several phase I clinical trials against different malignancies. VSV-based recombinant viruses are effective OVs against a majority of tested PDAC cell lines. However, some PDAC cell lines are resistant to VSV. Upregulated type I IFN signaling and constitutive expression of a subset of interferon-simulated genes (ISGs) play a major role in such resistance, while other mechanisms, such as inefficient viral attachment and resistance to VSV-mediated apoptosis, also play a role in some PDACs. Several alternative approaches have been shown to break the resistance of PDACs to VSV without compromising VSV oncoselectivity, including (i) combinations of VSV with JAK1/2 inhibitors (such as ruxolitinib); (ii) triple combinations of VSV with ruxolitinib and polycations improving both VSV replication and attachment; (iii) combinations of VSV with chemotherapeutic drugs (such as paclitaxel) arresting cells in the G2/M phase; (iv) arming VSV with p53 transgenes; (v) directed evolution approach producing more effective OVs. The latter study demonstrated impressive long-term genomic stability of complex VSV recombinants encoding large transgenes, supporting further clinical development of VSV as safe therapeutics for PDAC.
Collapse
Affiliation(s)
| | | | - Valery Z. Grdzelishvili
- Department of Biological Sciences, University of North Carolina at Charlotte, Charlotte, NC 28223, USA; (M.C.H.); (D.W.G.)
| |
Collapse
|
21
|
Pham TND, Shields MA, Spaulding C, Principe DR, Li B, Underwood PW, Trevino JG, Bentrem DJ, Munshi HG. Preclinical Models of Pancreatic Ductal Adenocarcinoma and Their Utility in Immunotherapy Studies. Cancers (Basel) 2021; 13:cancers13030440. [PMID: 33503832 PMCID: PMC7865443 DOI: 10.3390/cancers13030440] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 01/13/2021] [Accepted: 01/20/2021] [Indexed: 12/18/2022] Open
Abstract
Simple Summary Immune checkpoint blockade has provided durable clinical responses in a number of human malignancies, but not in patients with pancreatic cancer. Efforts to understand mechanisms of resistance and increase efficacy of immune checkpoint blockade in pancreatic cancer require the use of appropriate preclinical models in the laboratory. Here, we discuss the benefits, caveats, and potentials for improvement of the most commonly used models, including murine-based and patient-derived models. Abstract The advent of immunotherapy has transformed the treatment landscape for several human malignancies. Antibodies against immune checkpoints, such as anti-PD-1/PD-L1 and anti-CTLA-4, demonstrate durable clinical benefits in several cancer types. However, checkpoint blockade has failed to elicit effective anti-tumor responses in pancreatic ductal adenocarcinoma (PDAC), which remains one of the most lethal malignancies with a dismal prognosis. As a result, there are significant efforts to identify novel immune-based combination regimens for PDAC, which are typically first tested in preclinical models. Here, we discuss the utility and limitations of syngeneic and genetically-engineered mouse models that are currently available for testing immunotherapy regimens. We also discuss patient-derived xenograft mouse models, human PDAC organoids, and ex vivo slice cultures of human PDAC tumors that can complement murine models for a more comprehensive approach to predict response and resistance to immunotherapy regimens.
Collapse
Affiliation(s)
- Thao N. D. Pham
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; (M.A.S.); (C.S.)
- Jesse Brown VA Medical Center, Chicago, IL 60612, USA;
- Correspondence: (T.N.D.P.); (H.G.M.); Tel.: +1-312-503-0312 (T.N.D.P.); +1-312-503-2301 (H.G.M.)
| | - Mario A. Shields
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; (M.A.S.); (C.S.)
- Robert H. Lurie Comprehensive Cancer Center, Chicago, IL 60611, USA
| | - Christina Spaulding
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; (M.A.S.); (C.S.)
- Jesse Brown VA Medical Center, Chicago, IL 60612, USA;
| | - Daniel R. Principe
- Medical Scientist Training Program, University of Illinois, Chicago, IL 60612, USA;
| | - Bo Li
- Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA;
| | - Patrick W. Underwood
- Department of Surgery, University of Florida, Gainesville, FL 32611, USA; (P.W.U.); (J.G.T.)
| | - Jose G. Trevino
- Department of Surgery, University of Florida, Gainesville, FL 32611, USA; (P.W.U.); (J.G.T.)
| | - David J. Bentrem
- Jesse Brown VA Medical Center, Chicago, IL 60612, USA;
- Robert H. Lurie Comprehensive Cancer Center, Chicago, IL 60611, USA
- Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA;
| | - Hidayatullah G. Munshi
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; (M.A.S.); (C.S.)
- Jesse Brown VA Medical Center, Chicago, IL 60612, USA;
- Robert H. Lurie Comprehensive Cancer Center, Chicago, IL 60611, USA
- Correspondence: (T.N.D.P.); (H.G.M.); Tel.: +1-312-503-0312 (T.N.D.P.); +1-312-503-2301 (H.G.M.)
| |
Collapse
|
22
|
Jiang W, Li H, Liu X, Zhang J, zhang W, Li T, Liu L, Yu X. Precise and efficient silencing of mutant Kras G12D by CRISPR-CasRx controls pancreatic cancer progression. Theranostics 2020; 10:11507-11519. [PMID: 33052229 PMCID: PMC7545986 DOI: 10.7150/thno.46642] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 08/31/2020] [Indexed: 12/12/2022] Open
Abstract
Rationale: Pancreatic ductal adenocarcinoma (PDAC) is a highly lethal disease with few therapeutic targets and rare effective treatments. Over 90% of PDAC tumors bear a Kras mutation, and the single-site mutation G12D (KrasG12D) is most prevalent. Methods: Here, we applied the CRISPR-CasRx system to silence the mutant KrasG12D transcript in PDAC cells. We also used a capsid-optimized adenovirus-associated virus 8 vector (AAV8) to deliver the CRISPR-CasRx system into PDAC orthotopic tumors and patient-derived tumor xenografts (PDX). Results: Our data showed that guided by a KrasG12D-specific gRNA, CasRx is able to precisely and efficiently silence the mutant KrasG12D expression in PDAC cells. The knockdown of mutant KrasG12D by CasRx abolishes the aberrant activation of downstream signaling induced by mutant KrasG12D and subsequently suppresses the tumor growth and improves the sensitivity of gemcitabine in PDAC. Additionally, delivering CasRx-gRNA via AAV8 into the orthotopic KrasG12D PDAC tumors substantially improves the survival of mice without obvious toxicity. Furthermore, targeting KrasG12D through CasRx suppresses the growth of PDAC PDXs. In conclusion, our study provides a proof-of-concept that CRISPR-CasRx can be utilized to target and silence mutant KrasG12D transcripts and therefore inhibit PDAC malignancy.
Collapse
Affiliation(s)
- Wang Jiang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, PR China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, PR China
- Shanghai Pancreatic Cancer Institute, Shanghai 200032, PR China
- Pancreatic Cancer Institute, Fudan University, Shanghai 200032, PR China
| | - Hao Li
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, PR China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, PR China
- Shanghai Pancreatic Cancer Institute, Shanghai 200032, PR China
- Pancreatic Cancer Institute, Fudan University, Shanghai 200032, PR China
| | - Xiyu Liu
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, PR China
| | - Jianping Zhang
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Centre, Shanghai 200032, PR China
| | - Wuhu zhang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, PR China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, PR China
- Shanghai Pancreatic Cancer Institute, Shanghai 200032, PR China
- Pancreatic Cancer Institute, Fudan University, Shanghai 200032, PR China
| | - Tianjiao Li
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, PR China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, PR China
- Shanghai Pancreatic Cancer Institute, Shanghai 200032, PR China
- Pancreatic Cancer Institute, Fudan University, Shanghai 200032, PR China
| | - Liang Liu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, PR China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, PR China
- Shanghai Pancreatic Cancer Institute, Shanghai 200032, PR China
- Pancreatic Cancer Institute, Fudan University, Shanghai 200032, PR China
| | - Xianjun Yu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, PR China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, PR China
- Shanghai Pancreatic Cancer Institute, Shanghai 200032, PR China
- Pancreatic Cancer Institute, Fudan University, Shanghai 200032, PR China
| |
Collapse
|
23
|
He M, Henderson M, Muth S, Murphy A, Zheng L. Preclinical mouse models for immunotherapeutic and non-immunotherapeutic drug development for pancreatic ductal adenocarcinoma. ACTA ACUST UNITED AC 2020; 3. [PMID: 32832900 PMCID: PMC7440242 DOI: 10.21037/apc.2020.03.03] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is in urgent need of better diagnostic and therapeutic methods due to its late diagnosis, limited treatment options and poor prognosis. Finding the right animal models to recapitulate the tumor molecular pathogenesis and tumor microenvironment (TME) complexity is critical for preclinical immunotherapeutic and non-immunotherapeutic treatment developments. In this review, we summarize and evaluate popular preclinical animal models including patient-derived xenograft models, humanized mouse models, genetically engineered mouse models, and syngeneic mouse models. Through comparisons between these models in different research settings, we hope to provide guidance in finding the most relevant preclinical models to suit various research purposes.
Collapse
Affiliation(s)
- Mengni He
- Department of Cell Biology, Baltimore, MD, USA
| | - MacKenzie Henderson
- Department of Oncology, Baltimore, MD, USA.,The Sidney Kimmel Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Stephen Muth
- Department of Oncology, Baltimore, MD, USA.,The Sidney Kimmel Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Adrian Murphy
- Department of Oncology, Baltimore, MD, USA.,The Sidney Kimmel Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,The Precision Medicine Center of Excellence (PMCoE) Program for Pancreatic Cancer, Baltimore, MD, USA
| | - Lei Zheng
- Department of Oncology, Baltimore, MD, USA.,The Sidney Kimmel Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,The Precision Medicine Center of Excellence (PMCoE) Program for Pancreatic Cancer, Baltimore, MD, USA
| |
Collapse
|
24
|
Awaji M, Saxena S, Wu L, Prajapati DR, Purohit A, Varney ML, Kumar S, Rachagani S, Ly QP, Jain M, Batra SK, Singh RK. CXCR2 signaling promotes secretory cancer-associated fibroblasts in pancreatic ductal adenocarcinoma. FASEB J 2020; 34:9405-9418. [PMID: 32453916 PMCID: PMC7501205 DOI: 10.1096/fj.201902990r] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 04/21/2020] [Accepted: 05/04/2020] [Indexed: 12/21/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains one of the most challenging malignancies. Desmoplasia and tumor-supporting inflammation are hallmarks of PDAC. The tumor microenvironment contributes significantly to tumor progression and spread. Cancer-associated fibroblasts (CAFs) facilitate therapy resistance and metastasis. Recent reports emphasized the concurrence of multiple subtypes of CAFs with diverse roles, fibrogenic, and secretory. C-X-C motif chemokine receptor 2 (CXCR2) is a chemokine receptor known for its role during inflammation and its adverse role in PDAC. Oncogenic Kras upregulates CXCR2 and its ligands and, thus, contribute to tumor proliferation and immunosuppression. CXCR2 deletion in a PDAC syngeneic mouse model produced increased fibrosis revealing a potential undescribed role of CXCR2 in CAFs. In this study, we demonstrate that the oncogenic Kras-CXCR2 axis regulates the CAFs function in PDAC and contributes to CAFs heterogeneity. We observed that oncogenic Kras and CXCR2 signaling alter CAFs, producing a secretory CAF phenotype with low fibrogenic features; and increased secretion of pro-tumor cytokines and CXCR2 ligands, utilizing the NF-κB activity. Finally, using syngeneic mouse models, we demonstrate that oncogenic Kras is associated with secretory CAFs and that CXCR2 inhibition promotes activation of fibrotic cells (myofibroblasts) and impact tumors in a mutation-dependent manner.
Collapse
MESH Headings
- Animals
- Apoptosis
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Cancer-Associated Fibroblasts/metabolism
- Cancer-Associated Fibroblasts/pathology
- Carcinoma, Pancreatic Ductal/genetics
- Carcinoma, Pancreatic Ductal/metabolism
- Carcinoma, Pancreatic Ductal/pathology
- Cell Proliferation
- Gene Expression Regulation, Neoplastic
- Mice
- Mice, Knockout
- Mutation
- Pancreatic Neoplasms/genetics
- Pancreatic Neoplasms/metabolism
- Pancreatic Neoplasms/pathology
- Receptors, Interleukin-8B/genetics
- Receptors, Interleukin-8B/metabolism
- Signal Transduction
- Tumor Cells, Cultured
- Tumor Microenvironment
- Pancreatic Neoplasms
Collapse
Affiliation(s)
- Mohammad Awaji
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha NE 68198-5900
- Department of Pathology and Laboratory Medicine, King Fahad Specialist Hospital, Dammam, Saudi Arabia 31444
| | - Sugandha Saxena
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha NE 68198-5900
| | - Lingyun Wu
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha NE 68198-5900
| | - Dipakkumar R. Prajapati
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha NE 68198-5900
| | - Abhilasha Purohit
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha NE 68198-5900
| | - Michelle L. Varney
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha NE 68198-5900
| | - Sushil Kumar
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha NE 68198-5870
| | - Satyanarayana Rachagani
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha NE 68198-5870
| | - Quan P. Ly
- Department of Surgical Oncology, University of Nebraska Medical Center, Omaha NE 68198-6880
| | - Maneesh Jain
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha NE 68198-5870
| | - Surinder K. Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha NE 68198-5870
| | - Rakesh K. Singh
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha NE 68198-5900
| |
Collapse
|
25
|
Huang Y, Chen Y, Zhou S, Chen L, Wang J, Pei Y, Xu M, Feng J, Jiang T, Liang K, Liu S, Song Q, Jiang G, Gu X, Zhang Q, Gao X, Chen J. Dual-mechanism based CTLs infiltration enhancement initiated by Nano-sapper potentiates immunotherapy against immune-excluded tumors. Nat Commun 2020; 11:622. [PMID: 32001695 PMCID: PMC6992734 DOI: 10.1038/s41467-020-14425-7] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 01/06/2020] [Indexed: 12/29/2022] Open
Abstract
The failure of immunotherapies in immune-excluded tumor (IET) is largely ascribed to the void of intratumoral cytotoxic T cells (CTLs). The major obstacles are the excessive stroma, defective vasculatures and the deficiency of signals recruiting CTLs. Here we report a dual-mechanism based CTLs infiltration enhancer, Nano-sapper, which can simultaneously reduce the physical obstacles in tumor microenvironment and recruiting CTLs to potentiate immunotherapy in IET. Nano-sapper consists a core that co-loaded with antifibrotic phosphates-modified α-mangostin and plasmid encoding immune-enhanced cytokine LIGHT. Through reversing the abnormal activated fibroblasts, decreasing collagen deposition, normalizing the intratumoral vasculatures, and in situ stimulating the lymphocyte-recruiting chemoattractants expression, Nano-sapper paves the road for the CTLs infiltration, induces the intratumoral tertiary lymphoid structures, thus reshapes tumor microenvironment and potentiates checkpoint inhibitor against IET. This study demonstrates that the combination of antifibrotic agent and immune-enhanced cytokine might represent a modality in promoting immunotherapy against IET. The exclusion of cytotoxic T cells remains an important barrier to the efficacy of immunotherapies. Here the authors demonstrate that the combination anti-fibrosis agents and immune-enhanced cytokines can enhance T cell infiltration in a mouse model of pancreatic cancer.
Collapse
Affiliation(s)
- Yukun Huang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Lane 826, Zhangheng Road, Shanghai, 201203, P.R. China
| | - Yu Chen
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Lane 826, Zhangheng Road, Shanghai, 201203, P.R. China
| | - Songlei Zhou
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Lane 826, Zhangheng Road, Shanghai, 201203, P.R. China
| | - Liang Chen
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Lane 826, Zhangheng Road, Shanghai, 201203, P.R. China
| | - Jiahao Wang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Lane 826, Zhangheng Road, Shanghai, 201203, P.R. China
| | - Yuanyuan Pei
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Lane 826, Zhangheng Road, Shanghai, 201203, P.R. China
| | - Minjun Xu
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Lane 826, Zhangheng Road, Shanghai, 201203, P.R. China
| | - Jingxian Feng
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Lane 826, Zhangheng Road, Shanghai, 201203, P.R. China
| | - Tianze Jiang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Lane 826, Zhangheng Road, Shanghai, 201203, P.R. China
| | - Kaifan Liang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Lane 826, Zhangheng Road, Shanghai, 201203, P.R. China
| | - Shanshan Liu
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Lane 826, Zhangheng Road, Shanghai, 201203, P.R. China
| | - Qingxiang Song
- Department of Pharmacology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, P.R. China
| | - Gan Jiang
- Department of Pharmacology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, P.R. China
| | - Xiao Gu
- Department of Pharmacology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, P.R. China
| | - Qian Zhang
- Department of Pharmacology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, P.R. China
| | - Xiaoling Gao
- Department of Pharmacology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, P.R. China.
| | - Jun Chen
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Lane 826, Zhangheng Road, Shanghai, 201203, P.R. China. .,Department of Pharmacy, Shanghai Pudong Hospital, Fudan University, 2800 Gongwei Road, Shanghai, 201399, P.R. China.
| |
Collapse
|
26
|
Kong K, Guo M, Liu Y, Zheng J. Progress in Animal Models of Pancreatic Ductal Adenocarcinoma. J Cancer 2020; 11:1555-1567. [PMID: 32047562 PMCID: PMC6995380 DOI: 10.7150/jca.37529] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 11/10/2019] [Indexed: 12/22/2022] Open
Abstract
As a common gastrointestinal tumor, the incidence of pancreatic cancer has been increasing in recent years. The disease shows multi-gene, multi-step complex evolution from occurrence to dissemination. Furthermore, pancreatic cancer has an insidious onset and an extremely poor prognosis, so it is difficult to obtain cinical specimens at different stages of the disease, and it is, therefore, difficult to observe tumorigenesis and tumor development in patients with pancreatic cancer. At present, no standard protocols stipulate clinical treatment of pancreatic cancer, and the benefit rate of new targeted therapies is low. For this reason, a well-established preclinical model of pancreatic cancer must be established to allow further exploration of the occurrence, development, invasion, and metastasis mechanism of pancreatic cancer, as well as to facilitate research into new therapeutic targets. A large number of animal models of pancreatic cancer are currently available, including a cancer cell line-based xenograft, a patient-derived xenograft, several mouse models (including transgenic mice), and organoid models. These models have their own characteristics, but they still cannot perfectly predict the clinical outcome of the new treatment. In this paper, we present the distinctive features of the currently popular pancreatic cancer models, and discuss their preparation methods, clinical relations, scientific purposes and limitations.
Collapse
Affiliation(s)
- Kaiwen Kong
- Pathology Department of Changhai Hospital, Second Military Medical University
| | - Meng Guo
- Institute of Organ Transplantation, Changzheng Hospital, Second Military Medical University, Shanghai, China; National Key Laboratory of Medical Immunology &Institute of Immunology, Second Military Medical University
| | - Yanfang Liu
- Pathology Department of Changhai Hospital, Second Military Medical University; National Key Laboratory of Medical Immunology &Institute of Immunology, Second Military Medical University
| | - Jianming Zheng
- Pathology Department of Changhai Hospital, Second Military Medical University
| |
Collapse
|
27
|
Yang Y, Stang A, Schweickert PG, Lanman NA, Paul EN, Monia BP, Revenko AS, Palumbo JS, Mullins ES, Elzey BD, Janssen EM, Konieczny SF, Flick MJ. Thrombin Signaling Promotes Pancreatic Adenocarcinoma through PAR-1-Dependent Immune Evasion. Cancer Res 2019; 79:3417-3430. [PMID: 31048498 DOI: 10.1158/0008-5472.can-18-3206] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 03/11/2019] [Accepted: 04/29/2019] [Indexed: 12/22/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is associated with robust activity of the coagulation system. To determine mechanisms by which clotting factors influence PDAC tumor progression, we generated and characterized C57Bl/6-derived KPC (KRasG12D, TRP53R172H ) cell lines. Tissue factor (TF) and protease-activated receptor-1 (PAR-1) were highly expressed in primary KPC pancreatic lesions and KPC cell lines similar to expression profiles observed in biopsies of patients with PDAC. In allograft studies, tumor growth and metastatic potential were significantly diminished by depletion of TF or Par-1 in cancer cells or by genetic or pharmacologic reduction of the coagulation zymogen prothrombin in mice. Notably, PAR-1-deleted KPC cells (KPC-Par-1KO) failed to generate sizable tumors, a phenotype completely rescued by restoration of Par-1 expression. Expression profiling of KPC and KPC-Par-1KO cells indicated that thrombin-PAR-1 signaling significantly altered immune regulation pathways. Accordingly, KPC-Par-1KO cells failed to form tumors in immune-competent mice but displayed robust tumor growth comparable to that observed with control KPC cells in immune-compromised NSG mice. Immune cell depletion studies indicated that CD8 T cells, but not CD4 cells or natural killer cells, mediated elimination of KPC-Par-1KO tumor cells in C57Bl/6 mice. These results demonstrate that PDAC is driven by activation of the coagulation system through tumor cell-derived TF, circulating prothrombin, and tumor cell-derived PAR-1 and further indicate that one key mechanism of thrombin/PAR-1-mediated tumor growth is suppression of antitumor immunity in the tumor microenvironment. SIGNIFICANCE: The tissue factor-thrombin-PAR-1 signaling axis in tumor cells promotes PDAC growth and disease progression with one key mechanism being suppression of antitumor immunity in the microenvironment.
Collapse
Affiliation(s)
- Yi Yang
- Department of Biological Science and the Purdue Center for Cancer Research, Purdue University, West Lafayette, Indiana
| | - Amanda Stang
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital, Cincinnati, Ohio
| | - Patrick G Schweickert
- Department of Biological Science and the Purdue Center for Cancer Research, Purdue University, West Lafayette, Indiana
| | - Nadia A Lanman
- Department of Biological Science and the Purdue Center for Cancer Research, Purdue University, West Lafayette, Indiana
| | - Erin N Paul
- Department of Biological Science and the Purdue Center for Cancer Research, Purdue University, West Lafayette, Indiana
| | - Brett P Monia
- Ionis Pharmaceuticals, Inc., Antisense Drug Discovery, Carlsbad, California
| | - Alexey S Revenko
- Ionis Pharmaceuticals, Inc., Antisense Drug Discovery, Carlsbad, California
| | - Joseph S Palumbo
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital, Cincinnati, Ohio
| | - Eric S Mullins
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital, Cincinnati, Ohio
| | - Bennett D Elzey
- Department of Comparative Pathobiology, Purdue University, West Lafayette, Indiana
| | - Edith M Janssen
- Division of Immunobiology, Cincinnati Children's Hospital, Cincinnati, Ohio
| | - Stephen F Konieczny
- Department of Biological Science and the Purdue Center for Cancer Research, Purdue University, West Lafayette, Indiana.
| | - Matthew J Flick
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital, Cincinnati, Ohio.
| |
Collapse
|
28
|
Nanoparticle Delivery of RIG-I Agonist Enables Effective and Safe Adjuvant Therapy in Pancreatic Cancer. Mol Ther 2018; 27:507-517. [PMID: 30545600 DOI: 10.1016/j.ymthe.2018.11.012] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 11/09/2018] [Accepted: 11/11/2018] [Indexed: 01/14/2023] Open
Abstract
Local immunomodulation can be a promising strategy to augment the efficacy and decrease off-target toxicities associated with cancer treatment. Pancreatic cancer is resistant to immunotherapies due to the immunosuppressive tumor microenvironment. Herein, we investigated a therapeutic approach involving delivery of a short interfering double-stranded RNA (dsRNA), specific to Bcl2, with 5' triphosphate ends, by lipid calcium phosphate nanoparticles, in an orthotopic allograft KPC model of pancreatic cancer. Retinoic acid-inducible gene I (RIG-I)-like receptors can bind to 5' triphosphate dsRNA (ppp dsRNA), a pathogen-associated molecular pattern, producing type I interferon, while Bcl2 silencing can drive apoptosis of cancer cells. Our approach demonstrated a robust enrichment of tumor tissue with therapeutic nanoparticles and enabled a significant tumor growth inhibition, prolonging median overall survival. Nanoparticles encapsulating dual-therapeutic ppp dsRNA allowed strong induction in levels of pro-inflammatory Th1 cytokines, further increasing proportions of CD8+ T cells over regulatory T cells, M1 over M2 macrophages, and decreased levels of immunosuppressive B regulatory and plasma cells in the tumor microenvironment. Thus, these results provide a new immunotherapy approach for pancreatic cancer.
Collapse
|
29
|
Tekin C, Shi K, Daalhuisen JB, ten Brink MS, Bijlsma MF, Spek CA. PAR1 signaling on tumor cells limits tumor growth by maintaining a mesenchymal phenotype in pancreatic cancer. Oncotarget 2018; 9:32010-32023. [PMID: 30174793 PMCID: PMC6112838 DOI: 10.18632/oncotarget.25880] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 07/21/2018] [Indexed: 12/19/2022] Open
Abstract
Protease activated receptor-1 (PAR1) expression is associated with disease progression and overall survival in a variety of cancers. However, the importance of tumor cell PAR1 in pancreatic ductal adenocarcinomas (PDAC) remains unexplored. Utilizing orthotopic models with wild type and PAR1-targeted PDAC cells, we show that tumor cell PAR1 negatively affects PDAC growth, yet promotes metastasis. Mechanistically, we show that tumor cell-specific PAR1 expression correlates with mesenchymal signatures in PDAC and that PAR1 is linked to the maintenance of a partial mesenchymal cell state. Indeed, loss of PAR1 expression results in well-differentiated pancreatic tumors in vivo, with enhanced epithelial characteristics both in vitro and in vivo. Taken together, we have identified a novel growth inhibitory role of PAR1 in PDAC, which is linked to the induction, and maintenance of a mesenchymal-like phenotype. The recognition that PAR1 actively limits pancreatic cancer cell growth suggest that the contributions of PAR1 to tumor growth differ between cancers of epithelial origin and that its targeting should be applied with care.
Collapse
Affiliation(s)
- Cansu Tekin
- Amsterdam UMC, University of Amsterdam, Center of Experimental and Molecular Medicine, Amsterdam, The Netherlands
- Amsterdam UMC, University of Amsterdam, Laboratory for Experimental Oncology and Radiobiology, Cancer Center Amsterdam, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - Kun Shi
- Amsterdam UMC, University of Amsterdam, Center of Experimental and Molecular Medicine, Amsterdam, The Netherlands
| | - Joost B. Daalhuisen
- Amsterdam UMC, University of Amsterdam, Center of Experimental and Molecular Medicine, Amsterdam, The Netherlands
| | - Marieke S. ten Brink
- Amsterdam UMC, University of Amsterdam, Center of Experimental and Molecular Medicine, Amsterdam, The Netherlands
| | - Maarten F. Bijlsma
- Amsterdam UMC, University of Amsterdam, Laboratory for Experimental Oncology and Radiobiology, Cancer Center Amsterdam, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - C. Arnold Spek
- Amsterdam UMC, University of Amsterdam, Center of Experimental and Molecular Medicine, Amsterdam, The Netherlands
| |
Collapse
|
30
|
Bakst RL, Xiong H, Chen CH, Deborde S, Lyubchik A, Zhou Y, He S, McNamara W, Lee SY, Olson OC, Leiner IM, Marcadis AR, Keith JW, Al-Ahmadie HA, Katabi N, Gil Z, Vakiani E, Joyce JA, Pamer E, Wong RJ. Inflammatory Monocytes Promote Perineural Invasion via CCL2-Mediated Recruitment and Cathepsin B Expression. Cancer Res 2017; 77:6400-6414. [PMID: 28951461 PMCID: PMC5831809 DOI: 10.1158/0008-5472.can-17-1612] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 08/21/2017] [Accepted: 09/18/2017] [Indexed: 12/14/2022]
Abstract
Perineural invasion (PNI) is an ominous event strongly linked to poor clinical outcome. Cells residing within peripheral nerves collaborate with cancer cells to enable PNI, but the contributing conditions within the tumor microenvironment are not well understood. Here, we show that CCR2-expressing inflammatory monocytes (IM) are preferentially recruited to sites of PNI, where they differentiate into macrophages and potentiate nerve invasion through a cathepsin B-mediated process. A series of adoptive transfer experiments with genetically engineered donors and recipients demonstrated that IM recruitment to nerves was driven by CCL2 released from Schwann cells at the site of PNI, but not CCL7, an alternate ligand for CCR2. Interruption of either CCL2-CCR2 signaling or cathepsin B function significantly impaired PNI in vivo Correlative studies in human specimens demonstrated that cathepsin B-producing macrophages were enriched in invaded nerves, which was associated with increased local tumor recurrence. These findings deepen our understanding of PNI pathogenesis and illuminate how PNI is driven in part by corruption of a nerve repair program. Further, they support the exploration of inhibiting IM recruitment and function as a targeted therapy for PNI. Cancer Res; 77(22); 6400-14. ©2017 AACR.
Collapse
MESH Headings
- Animals
- Cathepsin B/metabolism
- Cell Line
- Cell Line, Tumor
- Chemokine CCL2/genetics
- Chemokine CCL2/metabolism
- Humans
- Macrophages/metabolism
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Nude
- Monocytes/metabolism
- Monocytes/pathology
- Neoplasm Invasiveness
- Neoplasms, Experimental/genetics
- Neoplasms, Experimental/metabolism
- Neoplasms, Experimental/pathology
- Pancreatic Neoplasms/genetics
- Pancreatic Neoplasms/metabolism
- Pancreatic Neoplasms/pathology
- Peripheral Nerves/metabolism
- Peripheral Nerves/pathology
- Receptors, CCR2/genetics
- Receptors, CCR2/metabolism
- Schwann Cells/metabolism
- Transplantation, Heterologous
Collapse
Affiliation(s)
- Richard L Bakst
- Department of Radiation Oncology, Mount Sinai School of Medicine, New York, New York
| | - Huizhong Xiong
- Immunology Program, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Chun-Hao Chen
- Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Sylvie Deborde
- Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Anna Lyubchik
- Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Yi Zhou
- Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Shizhi He
- Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - William McNamara
- Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Sei-Young Lee
- Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Oakley C Olson
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ingrid M Leiner
- Immunology Program, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Andrea R Marcadis
- Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - James W Keith
- Immunology Program, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Hikmat A Al-Ahmadie
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Nora Katabi
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ziv Gil
- Department of Otolaryngology, Rambam Healthcare Campus, The Technion-Israel Institute of Technology, Haifa, Israel
| | - Efsevia Vakiani
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Johanna A Joyce
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, New York
- Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland
| | - Eric Pamer
- Immunology Program, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Richard J Wong
- Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, New York.
| |
Collapse
|
31
|
Luheshi NM, Coates-Ulrichsen J, Harper J, Mullins S, Sulikowski MG, Martin P, Brown L, Lewis A, Davies G, Morrow M, Wilkinson RW. Transformation of the tumour microenvironment by a CD40 agonist antibody correlates with improved responses to PD-L1 blockade in a mouse orthotopic pancreatic tumour model. Oncotarget 2017; 7:18508-20. [PMID: 26918344 PMCID: PMC4951305 DOI: 10.18632/oncotarget.7610] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 02/14/2016] [Indexed: 01/05/2023] Open
Abstract
Despite the availability of recently developed chemotherapy regimens, survival times for pancreatic cancer patients remain poor. These patients also respond poorly to immune checkpoint blockade therapies (anti-CTLA-4, anti-PD-L1, anti-PD-1), which suggests the presence of additional immunosuppressive mechanisms in the pancreatic tumour microenvironment (TME). CD40 agonist antibodies (αCD40) promote antigen presenting cell (APC) maturation and enhance macrophage tumouricidal activity, and may therefore alter the pancreatic TME to increase sensitivity to immune checkpoint blockade. Here, we test whether αCD40 transforms the TME in a mouse syngeneic orthotopic model of pancreatic cancer, to increase sensitivity to PD-L1 blockade. We found that whilst mice bearing orthotopic Pan02 tumours responded poorly to PD-L1 blockade, αCD40 improved overall survival. αCD40 transformed the TME, upregulating Th1 chemokines, increasing cytotoxic T cell infiltration and promoting formation of an immune cell-rich capsule separating the tumour from the normal pancreas. Furthermore, αCD40 drove systemic APC maturation, memory T cell expansion, and upregulated tumour and systemic PD-L1 expression. Combining αCD40 with PD-L1 blockade enhanced anti-tumour immunity and improved overall survival versus either monotherapy. These data provide further support for the potential of combining αCD40 with immune checkpoint blockade to promote anti-tumour immunity in pancreatic cancer.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Lee Brown
- MedImmune Ltd., Cambridge CB21 6GH, UK
| | | | | | | | | |
Collapse
|
32
|
Lu C, Talukder A, Savage NM, Singh N, Liu K. JAK-STAT-mediated chronic inflammation impairs cytotoxic T lymphocyte activation to decrease anti-PD-1 immunotherapy efficacy in pancreatic cancer. Oncoimmunology 2017; 6:e1291106. [PMID: 28405527 DOI: 10.1080/2162402x.2017.1291106] [Citation(s) in RCA: 115] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 01/27/2017] [Accepted: 01/31/2017] [Indexed: 12/14/2022] Open
Abstract
Human pancreatic cancer does not respond to immune check point blockade immunotherapy. One key feature of pancreatic cancer is the association between its progression and chronic inflammation. Emerging evidence supports a key role for the JAK-STAT pathway in pancreatic cancer inflammation. We aimed at testing the hypothesis that sustained JAK-STAT signaling suppresses cytotoxic T lymphocyte (CTL) activation to counteract anti-PD-1 immunotherapy-induced CTL activity in pancreatic cancer. We show that human pancreatic carcinomas express high level of PD-L1 and exhibit low level of CTL infiltration. JAK-STAT inhibitor Ruxolitinib selectively inhibits STAT1 and STAT3 activation and increases CTL infiltration to induce a Tc1/Th1 immune response in the tumor microenvironment in an orthotopic pancreatic cancer mouse model. Ruxilitinib-mediated tumor suppressive efficacy diminishes in T-cell-deficient mice. Pancreatic tumor grows significantly faster in IFNγ-deficient mice. However, neutralizing IFNγ does not alter tumor growth but diminishes Ruxolitinib-induced tumor suppression in vivo, indicating that lymphocytes and IFNγ are essential for Ruxolitinib-induced host antitumor immune response. Both type I and type II interferons upregulate PD-L1 expression through the JAK-STAT signaling pathway in mouse pancreatic tumor cells. Tumor cells respond to activated T cells by activating STAT3. The inhibition of STAT3 downregulates immune suppressive cytokines production by tumor cells, resulting in increased T cell activation and effector function. Consequently, Ruxolitinib significantly improves the efficacy of anti-PD-1 immunotherapy. Our data demonstrate that Ruxolitinib is effective in the inhibition of systemic inflammation in the tumor microenvironment and therefore upregulates CTL infiltration and activation to overcome pancreatic cancer resistance to anti-PD-1 immunotherapy.
Collapse
Affiliation(s)
- Chunwan Lu
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta, GA, USA; Georgia Cancer Center, Augusta, GA, USA; Charlie Norwood VA Medical Center, Augusta, GA, USA
| | - Asif Talukder
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta, GA, USA; Department of Surgery, Pathology, Medical College of Georgia, Augusta, GA, USA
| | | | - Nagendra Singh
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta, GA, USA; Georgia Cancer Center, Augusta, GA, USA
| | - Kebin Liu
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta, GA, USA; Georgia Cancer Center, Augusta, GA, USA; Charlie Norwood VA Medical Center, Augusta, GA, USA
| |
Collapse
|
33
|
Lu C, Paschall AV, Shi H, Savage N, Waller JL, Sabbatini ME, Oberlies NH, Pearce C, Liu K. The MLL1-H3K4me3 Axis-Mediated PD-L1 Expression and Pancreatic Cancer Immune Evasion. J Natl Cancer Inst 2017; 109:2962333. [PMID: 28131992 DOI: 10.1093/jnci/djw283] [Citation(s) in RCA: 178] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Revised: 09/21/2016] [Accepted: 10/26/2016] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Pancreatic cancer is one of the cancers where anti-PD-L1/PD-1 immunotherapy has been unsuccessful. What confers pancreatic cancer resistance to checkpoint immunotherapy is unknown. The aim of this study is to elucidate the underlying mechanism of PD-L1 expression regulation in the context of pancreatic cancer immune evasion. METHODS Pancreatic cancer mouse models and human specimens were used to determine PD-L1 and PD-1 expression and cancer immune evasion. Histone methyltransferase inhibitors, RNAi, and overexpression were used to elucidate the underlying molecular mechanism of PD-L1 expression regulation. All statistical tests were two-sided. RESULTS PD-L1 is expressed in 60% to 90% of tumor cells in human pancreatic carcinomas and in nine of 10 human pancreatic cancer cell lines. PD-1 is expressed in 51.2% to 52.1% of pancreatic tumor-infiltrating cytotoxic T lymphocytes (CTLs). Tumors grow statistically significantly faster in FasL-deficient mice than in wild-type mice (P = .03-.001) and when CTLs are neutralized (P = .03-<.001). H3K4 trimethylation (H3K4me3) is enriched in the cd274 promoter in pancreatic tumor cells. MLL1 directly binds to the cd274 promoter to catalyze H3K4me3 to activate PD-L1 transcription in tumor cells. Inhibition or silencing of MLL1 decreases the H3K4me3 level in the cd274 promoter and PD-L1 expression in tumor cells. Accordingly, inhibition of MLL1 in combination with anti-PD-L1 or anti-PD-1 antibody immunotherapy effectively suppresses pancreatic tumor growth in a FasL- and CTL-dependent manner. CONCLUSIONS The Fas-FasL/CTLs and the MLL1-H3K4me3-PD-L1 axis play contrasting roles in pancreatic cancer immune surveillance and evasion. Targeting the MLL1-H3K4me3 axis is an effective approach to enhance the efficacy of checkpoint immunotherapy against pancreatic cancer.
Collapse
Affiliation(s)
- Chunwan Lu
- Affiliations of authors: Department of Biochemistry and Molecular Biology (CL, AVP, KL), Department of Pathology (NS), and Department of Biostatistics and Epidemiology (JLW), Medical College of Georgia, Augusta, GA; Georgia Cancer Center (CL, AVP, HS, KL) and Department of Biological Sciences (MES), Augusta University, Augusta, GA; Charlie Norwood VA Medical Center, Augusta, GA (CL, AVP, KL); Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, Greensboro, NC (NHO); Mycosynthetix, Inc., Hillsborough, NC (CP)
| | - Amy V Paschall
- Affiliations of authors: Department of Biochemistry and Molecular Biology (CL, AVP, KL), Department of Pathology (NS), and Department of Biostatistics and Epidemiology (JLW), Medical College of Georgia, Augusta, GA; Georgia Cancer Center (CL, AVP, HS, KL) and Department of Biological Sciences (MES), Augusta University, Augusta, GA; Charlie Norwood VA Medical Center, Augusta, GA (CL, AVP, KL); Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, Greensboro, NC (NHO); Mycosynthetix, Inc., Hillsborough, NC (CP)
| | - Huidong Shi
- Affiliations of authors: Department of Biochemistry and Molecular Biology (CL, AVP, KL), Department of Pathology (NS), and Department of Biostatistics and Epidemiology (JLW), Medical College of Georgia, Augusta, GA; Georgia Cancer Center (CL, AVP, HS, KL) and Department of Biological Sciences (MES), Augusta University, Augusta, GA; Charlie Norwood VA Medical Center, Augusta, GA (CL, AVP, KL); Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, Greensboro, NC (NHO); Mycosynthetix, Inc., Hillsborough, NC (CP)
| | - Natasha Savage
- Affiliations of authors: Department of Biochemistry and Molecular Biology (CL, AVP, KL), Department of Pathology (NS), and Department of Biostatistics and Epidemiology (JLW), Medical College of Georgia, Augusta, GA; Georgia Cancer Center (CL, AVP, HS, KL) and Department of Biological Sciences (MES), Augusta University, Augusta, GA; Charlie Norwood VA Medical Center, Augusta, GA (CL, AVP, KL); Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, Greensboro, NC (NHO); Mycosynthetix, Inc., Hillsborough, NC (CP)
| | - Jennifer L Waller
- Affiliations of authors: Department of Biochemistry and Molecular Biology (CL, AVP, KL), Department of Pathology (NS), and Department of Biostatistics and Epidemiology (JLW), Medical College of Georgia, Augusta, GA; Georgia Cancer Center (CL, AVP, HS, KL) and Department of Biological Sciences (MES), Augusta University, Augusta, GA; Charlie Norwood VA Medical Center, Augusta, GA (CL, AVP, KL); Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, Greensboro, NC (NHO); Mycosynthetix, Inc., Hillsborough, NC (CP)
| | - Maria E Sabbatini
- Affiliations of authors: Department of Biochemistry and Molecular Biology (CL, AVP, KL), Department of Pathology (NS), and Department of Biostatistics and Epidemiology (JLW), Medical College of Georgia, Augusta, GA; Georgia Cancer Center (CL, AVP, HS, KL) and Department of Biological Sciences (MES), Augusta University, Augusta, GA; Charlie Norwood VA Medical Center, Augusta, GA (CL, AVP, KL); Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, Greensboro, NC (NHO); Mycosynthetix, Inc., Hillsborough, NC (CP)
| | - Nicholas H Oberlies
- Affiliations of authors: Department of Biochemistry and Molecular Biology (CL, AVP, KL), Department of Pathology (NS), and Department of Biostatistics and Epidemiology (JLW), Medical College of Georgia, Augusta, GA; Georgia Cancer Center (CL, AVP, HS, KL) and Department of Biological Sciences (MES), Augusta University, Augusta, GA; Charlie Norwood VA Medical Center, Augusta, GA (CL, AVP, KL); Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, Greensboro, NC (NHO); Mycosynthetix, Inc., Hillsborough, NC (CP)
| | - Cedric Pearce
- Affiliations of authors: Department of Biochemistry and Molecular Biology (CL, AVP, KL), Department of Pathology (NS), and Department of Biostatistics and Epidemiology (JLW), Medical College of Georgia, Augusta, GA; Georgia Cancer Center (CL, AVP, HS, KL) and Department of Biological Sciences (MES), Augusta University, Augusta, GA; Charlie Norwood VA Medical Center, Augusta, GA (CL, AVP, KL); Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, Greensboro, NC (NHO); Mycosynthetix, Inc., Hillsborough, NC (CP)
| | - Kebin Liu
- Affiliations of authors: Department of Biochemistry and Molecular Biology (CL, AVP, KL), Department of Pathology (NS), and Department of Biostatistics and Epidemiology (JLW), Medical College of Georgia, Augusta, GA; Georgia Cancer Center (CL, AVP, HS, KL) and Department of Biological Sciences (MES), Augusta University, Augusta, GA; Charlie Norwood VA Medical Center, Augusta, GA (CL, AVP, KL); Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, Greensboro, NC (NHO); Mycosynthetix, Inc., Hillsborough, NC (CP)
| |
Collapse
|
34
|
Lu C, Redd PS, Lee JR, Savage N, Liu K. The expression profiles and regulation of PD-L1 in tumor-induced myeloid-derived suppressor cells. Oncoimmunology 2016; 5:e1247135. [PMID: 28123883 DOI: 10.1080/2162402x.2016.1247135] [Citation(s) in RCA: 173] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 09/27/2016] [Accepted: 10/05/2016] [Indexed: 12/22/2022] Open
Abstract
Programmed death-ligand 1 (PD-L1) is an inhibitory ligand that binds to PD-1 to suppress T cell activation. PD-L1 is constitutively expressed and inducible in tumor cells, but the expression profiles and regulatory mechanism of PD-L1 in myeloid-derived suppressor cells (MDSCs) are largely unknown. We report that PD-L1 is abundantly expressed in tumor-infiltrating leukocytes in human patients with both microsatellite instable and microsatellite stable colon cancer. About 60% CD11b+CD33+HLA-DR- MDSCs from peripheral blood of human colon cancer patients are PD-L1+. PD-L1+ MDSCs are also significantly higher in tumor-bearing mice than in tumor-free mice. Interestingly, the highest PD-L1+ MDSCs were observed in the tumor microenvironment in which 56-71% tumor-infiltrating MDSCs are PD-L1+in vivo. In contrast, PD-L1+ MDSCs are significantly less in secondary lymphoid organs and peripheral blood as compared to the tumor tissues, whereas bone marrow MDSCs are essentially PD-L1- in tumor-bearing mice. IFNγ is highly expressed in cells of the tumor tissues and IFNγ neutralization significantly decreased PD-L1+ MDSCs in the tumor microenvironment in vivo. However, IFNγ-activated pSTAT1 does not bind to the cd274 promoter in MDSCs. Instead, pSTAT1 activates expression of IRF1, IRF5, IRF7 and IRF8 in MDSCs, and only pSTAT1-activated IRF1 binds to a unique IRF-binding sequence element in vitro and chromatin in vivo in the cd274 promoter to activate PD-L1 transcription. Our data determine that PD-L1 is highly expressed in tumor-infiltrating MDSCs and in a lesser degree in lymphoid organs, and the pSTAT1-IRF1 axis regulates PD-L1 expression in MDSCs.
Collapse
Affiliation(s)
- Chunwan Lu
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta, GA, USA; Charlie Norwood VA Medical Center, Augusta, GA, USA
| | - Priscilla S Redd
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta, GA, USA; Charlie Norwood VA Medical Center, Augusta, GA, USA; Georgia Cancer Center, Augusta University, Augusta, GA, USA
| | - Jeffrey R Lee
- Charlie Norwood VA Medical Center, Augusta, GA, USA; Pathology, Medical College of Georgia, Augusta, GA, USA
| | - Natasha Savage
- Pathology, Medical College of Georgia , Augusta, GA, USA
| | - Kebin Liu
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta, GA, USA; Charlie Norwood VA Medical Center, Augusta, GA, USA; Georgia Cancer Center, Augusta University, Augusta, GA, USA
| |
Collapse
|
35
|
Incio J, Liu H, Suboj P, Chin SM, Chen IX, Pinter M, Ng MR, Nia HT, Grahovac J, Kao S, Babykutty S, Huang Y, Jung K, Rahbari NN, Han X, Chauhan VP, Martin JD, Kahn J, Huang P, Desphande V, Michaelson J, Michelakos TP, Ferrone CR, Soares R, Boucher Y, Fukumura D, Jain RK. Obesity-Induced Inflammation and Desmoplasia Promote Pancreatic Cancer Progression and Resistance to Chemotherapy. Cancer Discov 2016; 6:852-69. [PMID: 27246539 PMCID: PMC4972679 DOI: 10.1158/2159-8290.cd-15-1177] [Citation(s) in RCA: 301] [Impact Index Per Article: 33.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 05/23/2016] [Indexed: 12/16/2022]
Abstract
UNLABELLED It remains unclear how obesity worsens treatment outcomes in patients with pancreatic ductal adenocarcinoma (PDAC). In normal pancreas, obesity promotes inflammation and fibrosis. We found in mouse models of PDAC that obesity also promotes desmoplasia associated with accelerated tumor growth and impaired delivery/efficacy of chemotherapeutics through reduced perfusion. Genetic and pharmacologic inhibition of angiotensin-II type-1 receptor reverses obesity-augmented desmoplasia and tumor growth and improves response to chemotherapy. Augmented activation of pancreatic stellate cells (PSC) in obesity is induced by tumor-associated neutrophils (TAN) recruited by adipocyte-secreted IL1β. PSCs further secrete IL1β, and inactivation of PSCs reduces IL1β expression and TAN recruitment. Furthermore, depletion of TANs, IL1β inhibition, or inactivation of PSCs prevents obesity-accelerated tumor growth. In patients with pancreatic cancer, we confirmed that obesity is associated with increased desmoplasia and reduced response to chemotherapy. We conclude that cross-talk between adipocytes, TANs, and PSCs exacerbates desmoplasia and promotes tumor progression in obesity. SIGNIFICANCE Considering the current obesity pandemic, unraveling the mechanisms underlying obesity-induced cancer progression is an urgent need. We found that the aggravation of desmoplasia is a key mechanism of obesity-promoted PDAC progression. Importantly, we discovered that clinically available antifibrotic/inflammatory agents can improve the treatment response of PDAC in obese hosts. Cancer Discov; 6(8); 852-69. ©2016 AACR.See related commentary by Bronte and Tortora, p. 821This article is highlighted in the In This Issue feature, p. 803.
Collapse
Affiliation(s)
- Joao Incio
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts. Department of Internal Medicine, Hospital S. Joao, Porto, Portugal. I3S, Institute for Innovation and Research in Heath, Metabolism, Nutrition and Endocrinology Group, Biochemistry Department, Faculty of Medicine, Porto University, Porto, Portugal
| | - Hao Liu
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts. Biology and Biomedical Sciences, Harvard Medical School, Boston, Massachusetts
| | - Priya Suboj
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts. Department of Botany and Biotechnology, St. Xaviers College, Thumba, Trivandrum, Kerala, India
| | - Shan M Chin
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Ivy X Chen
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts. Harvard School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts
| | - Matthias Pinter
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Mei R Ng
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Hadi T Nia
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Jelena Grahovac
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Shannon Kao
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Suboj Babykutty
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts. Department of Zoology, Mar Ivanios College, Nalanchira, Trivandrum, Kerala, India
| | - Yuhui Huang
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Keehoon Jung
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Nuh N Rahbari
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Xiaoxing Han
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Vikash P Chauhan
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - John D Martin
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Julia Kahn
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Peigen Huang
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Vikram Desphande
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - James Michaelson
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts. Laboratory for Quantitative Medicine, and Division of Surgical Oncology, Gillette Center for Women's Cancers, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Theodoros P Michelakos
- Departments of Gastroenterology and Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Cristina R Ferrone
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts. Departments of Gastroenterology and Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Raquel Soares
- I3S, Institute for Innovation and Research in Heath, Metabolism, Nutrition and Endocrinology Group, Biochemistry Department, Faculty of Medicine, Porto University, Porto, Portugal
| | - Yves Boucher
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Dai Fukumura
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts.
| | - Rakesh K Jain
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
36
|
Yeo D, Huynh N, Beutler JA, Baldwin GS, He H, Nikfarjam M. Glaucarubinone Combined with Gemcitabine Improves Pancreatic Cancer Survival in an Immunocompetent Orthotopic Murine Model. J INVEST SURG 2016; 29:366-372. [PMID: 27027695 DOI: 10.3109/08941939.2016.1160167] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND Pancreatic cancer continues to have a poor survival rate with an urgent need for improved treatments. Glaucarubinone, a natural product first isolated from the seeds of the tree Simarouba glauca, has recently been recognized as having anti-cancer properties that may be particularly applicable to pancreatic cancer. METHODS The effect of glaucarubinone on the growth and migration of murine pancreatic cancer cells was assessed by 3H-thymidine incorporation assay. The survival impact of glaucarubinone alone and in combination with gemcitabine chemotherapy was assessed using an immunocompetent orthotopic murine model of pancreatic cancer. RESULTS Glaucarubinone inhibited the growth of the murine pancreatic cancer cell lines LM-P and PAN02. Treatment with either glaucarubinone or gemcitabine reduced proliferation in vitro and the combination was synergistic. The combination treatment improved survival two-fold compared to gemcitabine treatment alone (p = 0.046) in PAN02 cells. CONCLUSIONS The synergistic inhibition by glaucarubinone and gemcitabine observed in vitro and the improved survival in vivo suggest that glaucarubinone may be a useful adjunct to current chemotherapy regimens.
Collapse
Affiliation(s)
- Dannel Yeo
- a Department of Surgery , University of Melbourne, Austin Health , Melbourne , Victoria , Australia
| | - Nhi Huynh
- a Department of Surgery , University of Melbourne, Austin Health , Melbourne , Victoria , Australia
| | - John A Beutler
- b Molecular Targets Laboratory, National Cancer Institute , Frederick , MD
| | - Graham S Baldwin
- a Department of Surgery , University of Melbourne, Austin Health , Melbourne , Victoria , Australia
| | - Hong He
- a Department of Surgery , University of Melbourne, Austin Health , Melbourne , Victoria , Australia
| | - Mehrdad Nikfarjam
- a Department of Surgery , University of Melbourne, Austin Health , Melbourne , Victoria , Australia
| |
Collapse
|
37
|
Deborde S, Omelchenko T, Lyubchik A, Zhou Y, He S, McNamara WF, Chernichenko N, Lee SY, Barajas F, Chen CH, Bakst RL, Vakiani E, He S, Hall A, Wong RJ. Schwann cells induce cancer cell dispersion and invasion. J Clin Invest 2016; 126:1538-54. [PMID: 26999607 DOI: 10.1172/jci82658] [Citation(s) in RCA: 167] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Accepted: 01/26/2016] [Indexed: 12/23/2022] Open
Abstract
Nerves enable cancer progression, as cancers have been shown to extend along nerves through the process of perineural invasion, which carries a poor prognosis. Furthermore, the innervation of some cancers promotes growth and metastases. It remains unclear, however, how nerves mechanistically contribute to cancer progression. Here, we demonstrated that Schwann cells promote cancer invasion through direct cancer cell contact. Histological evaluation of murine and human cancer specimens with perineural invasion uncovered a subpopulation of Schwann cells that associates with cancer cells. Coculture of cancer cells with dorsal root ganglion extracts revealed that Schwann cells direct cancer cells to migrate toward nerves and promote invasion in a contact-dependent manner. Upon contact, Schwann cells induced the formation of cancer cell protrusions in their direction and intercalated between the cancer cells, leading to cancer cell dispersion. The formation of these processes was dependent on Schwann cell expression of neural cell adhesion molecule 1 (NCAM1) and ultimately promoted perineural invasion. Moreover, NCAM1-deficient mice showed decreased neural invasion and less paralysis. Such Schwann cell behavior reflects normal Schwann cell programs that are typically activated in nerve repair but are instead exploited by cancer cells to promote perineural invasion and cancer progression.
Collapse
|
38
|
Yeo D, He H, Patel O, Lowy AM, Baldwin GS, Nikfarjam M. FRAX597, a PAK1 inhibitor, synergistically reduces pancreatic cancer growth when combined with gemcitabine. BMC Cancer 2016; 16:24. [PMID: 26774265 PMCID: PMC4715347 DOI: 10.1186/s12885-016-2057-z] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Accepted: 01/08/2016] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma remains one of the most lethal of all solid tumours. Treatment options are limited and gemcitabine-based chemotherapy remains the standard of care. Although growing evidence shows that p21-activated kinase 1 (PAK1) plays a crucial role in pancreatic cancer, its role has not been fully elucidated. This study aimed to characterise the expression and functional relevance of PAK1 in pancreatic cancer. METHODS PAK1 expression was measured in pancreatic cancer specimens by immunohistochemistry and in pancreatic cancer cell lines by western blotting. The effect of inhibition of PAK1 by either shRNA knock-down (KD), or by a selective inhibitor, FRAX597, alone or in combination with gemcitabine, on cell proliferation and migration/invasion was measured by thymidine uptake and Boyden chamber assays, respectively. The effect on tumour growth and survival was assessed in orthotopic murine models. RESULTS PAK1 was expressed in all human pancreatic cancer samples tested, an7d was upregulated in all pancreatic cancer cell lines tested. PAK1 KD inhibited pancreatic cancer cell growth and survival, and increased sensitivity to gemcitabine treatment. AKT activity and HIF1α expression were also inhibited. FRAX597 inhibited pancreatic cancer cell proliferation, survival, and migration/invasion. When combined with gemcitabine, FRAX597 synergistically inhibited pancreatic cancer proliferation in vitro and inhibited tumour growth in vivo. CONCLUSIONS These results implicate PAK1 as a regulator of pancreatic cancer cell growth and survival. Combination of a PAK1 inhibitor such as FRAX597 with cytotoxic chemotherapy deserves further study as a novel therapeutic approach to pancreatic cancer treatment.
Collapse
Affiliation(s)
- Dannel Yeo
- Department of Surgery, Austin Health, University of Melbourne, Heidelberg, VIC, Australia.
| | - Hong He
- Department of Surgery, Austin Health, University of Melbourne, Heidelberg, VIC, Australia.
| | - Oneel Patel
- Department of Surgery, Austin Health, University of Melbourne, Heidelberg, VIC, Australia.
| | - Andrew M Lowy
- Department of Surgery, Division of Surgical Oncology, University of California at San Diego, Moores Cancer, La Jolla, CA, USA.
| | - Graham S Baldwin
- Department of Surgery, Austin Health, University of Melbourne, Heidelberg, VIC, Australia.
| | - Mehrdad Nikfarjam
- Department of Surgery, Austin Health, University of Melbourne, Heidelberg, VIC, Australia.
| |
Collapse
|
39
|
Incio J, Suboj P, Chin SM, Vardam-Kaur T, Liu H, Hato T, Babykutty S, Chen I, Deshpande V, Jain RK, Fukumura D. Metformin Reduces Desmoplasia in Pancreatic Cancer by Reprogramming Stellate Cells and Tumor-Associated Macrophages. PLoS One 2015; 10:e0141392. [PMID: 26641266 PMCID: PMC4671732 DOI: 10.1371/journal.pone.0141392] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2015] [Accepted: 10/06/2015] [Indexed: 02/06/2023] Open
Abstract
Background Pancreatic ductal adenocarcinoma (PDAC) is a highly desmoplastic tumor with a dismal prognosis for most patients. Fibrosis and inflammation are hallmarks of tumor desmoplasia. We have previously demonstrated that preventing the activation of pancreatic stellate cells (PSCs) and alleviating desmoplasia are beneficial strategies in treating PDAC. Metformin is a widely used glucose-lowering drug. It is also frequently prescribed to diabetic pancreatic cancer patients and has been shown to associate with a better outcome. However, the underlying mechanisms of this benefit remain unclear. Metformin has been found to modulate the activity of stellate cells in other disease settings. In this study, we examine the effect of metformin on PSC activity, fibrosis and inflammation in PDACs. Methods/Results In overweight, diabetic PDAC patients and pre-clinical mouse models, treatment with metformin reduced levels of tumor extracellular matrix (ECM) components, in particular hyaluronan (HA). In vitro, we found that metformin reduced TGF-ß signaling and the production of HA and collagen-I in cultured PSCs. Furthermore, we found that metformin alleviates tumor inflammation by reducing the expression of inflammatory cytokines including IL-1β as well as infiltration and M2 polarization of tumor-associated macrophages (TAMs) in vitro and in vivo. These effects on macrophages in vitro appear to be associated with a modulation of the AMPK/STAT3 pathway by metformin. Finally, we found in our preclinical models that the alleviation of desmoplasia by metformin was associated with a reduction in ECM remodeling, epithelial-to-mesenchymal transition (EMT) and ultimately systemic metastasis. Conclusion Metformin alleviates the fibro-inflammatory microenvironment in obese/diabetic individuals with pancreatic cancer by reprogramming PSCs and TAMs, which correlates with reduced disease progression. Metformin should be tested/explored as part of the treatment strategy in overweight diabetic PDAC patients.
Collapse
Affiliation(s)
- Joao Incio
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital Research Institute, Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Internal Medicine, Hospital S. Joao, Porto, Portugal
- I3S, Institute for Innovation and Research in Heath, Metabolism, Nutrition and Endocrinology group, Biochemistry Department, Faculty of Medicine, Porto University, Porto, Portugal
| | - Priya Suboj
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital Research Institute, Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Botany and Biotechnology, St. Xaviers College, Thumba, Trivandrum, Kerala, India
| | - Shan M. Chin
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital Research Institute, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Trupti Vardam-Kaur
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital Research Institute, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Hao Liu
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital Research Institute, Harvard Medical School, Boston, Massachusetts, United States of America
- Program of Biology and Biomedical Sciences, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Tai Hato
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital Research Institute, Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Suboj Babykutty
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital Research Institute, Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Zoology, Mar Ivanios College, Nalanchira, Trivandrum, Kerala, India
| | - Ivy Chen
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital Research Institute, Harvard Medical School, Boston, Massachusetts, United States of America
- School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts, United States of America
| | - Vikram Deshpande
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Rakesh K. Jain
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital Research Institute, Harvard Medical School, Boston, Massachusetts, United States of America
- * E-mail: (RKJ); (DF)
| | - Dai Fukumura
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital Research Institute, Harvard Medical School, Boston, Massachusetts, United States of America
- * E-mail: (RKJ); (DF)
| |
Collapse
|
40
|
Logsdon CD, Arumugam T, Ramachandran V. Animal Models of Gastrointestinal and Liver Diseases. The difficulty of animal modeling of pancreatic cancer for preclinical evaluation of therapeutics. Am J Physiol Gastrointest Liver Physiol 2015; 309:G283-91. [PMID: 26159697 PMCID: PMC4556944 DOI: 10.1152/ajpgi.00169.2015] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 06/25/2015] [Indexed: 01/31/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is relatively rare but extremely lethal. Standard cytotoxic therapeutics provide little benefit. To date, newer targeted therapeutics have also not been highly successful. Often novel therapeutics that have appeared to perform well in preclinical models have failed in the clinic. Many factors contribute to these failures, but the one most often attributed is the shortcomings of the preclinical models. A plethora of animal models now exist for PDAC, including cell line xenografts, patient-derived xenografts, a wide variety of genetic mouse models, and syngeneic xenografts. These models have generated a tremendous amount of information useful for the understanding of PDAC. Yet none seems to well predict clinical outcomes of new treatments. This review will discuss how genetic instability and cellular heterogeneity make this disease so difficult to model accurately. We will also discuss the strengths and weaknesses of many of the popular models. Ultimately we will argue that there is no perfect model and that the best approach to understanding clinical performance is the use of multiple preclinical models with an understanding of their salient features.
Collapse
Affiliation(s)
- Craig D. Logsdon
- 1Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas and ,2Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Thiruvengadam Arumugam
- 1Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas and
| | - Vijaya Ramachandran
- 1Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas and
| |
Collapse
|
41
|
Adiponectin promotes pancreatic cancer progression by inhibiting apoptosis via the activation of AMPK/Sirt1/PGC-1α signaling. Oncotarget 2015; 5:4732-45. [PMID: 25051362 PMCID: PMC4148095 DOI: 10.18632/oncotarget.1963] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Adiponectin is an adipocyte-secreted adipokine with pleiotropic actions. Clinical evidence has shown that serum adiponectin levels are increased and that adiponectin can protect pancreatic beta cells against apoptosis, which suggests that adiponectin may play an anti-apoptotic role in pancreatic cancer (PC). Here, we investigated the effects of adiponectin on PC development and elucidated the underlying molecular mechanisms. Adiponectin deficiency markedly attenuated pancreatic tumorigenesis in vivo. We found that adiponectin significantly inhibited the apoptosis of both human and mouse pancreatic cancer cells via adipoR1, but not adipoR2. Furthermore, adiponectin can increase AMP-activated protein kinase (AMPK) phosphorylation and NAD-dependent deacetylase sirtuin-1 (Sirt1) of PC cells. Knockdown of AMPK or Sirt1 can increase the apoptosis in PC cells. AMPK up-regulated Sirt1, and Sirt1 can inversely phosphorylate AMPK. Further studies have shown that Sirt1 can deacetylate peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC1α), which can increase the expression levels of mitochondrial genes. Thus, adiponectin exerts potent anti-apoptotic effects on PC cells via the activation of AMPK/Sirt1/PGC1α signaling. Finally, adiponectin can elevate β-catenin levels. Taken together, these novel findings reveal an unconventional role of adiponectin in promoting pancreatic cancers, and suggest that the effects of adiponectin on tumorigenesis are highly tissue-dependent.
Collapse
|
42
|
Cifarelli V, Lashinger LM, Devlin KL, Dunlap SM, Huang J, Kaaks R, Pollak MN, Hursting SD. Metformin and Rapamycin Reduce Pancreatic Cancer Growth in Obese Prediabetic Mice by Distinct MicroRNA-Regulated Mechanisms. Diabetes 2015; 64:1632-42. [PMID: 25576058 PMCID: PMC4407853 DOI: 10.2337/db14-1132] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2014] [Accepted: 11/25/2014] [Indexed: 12/12/2022]
Abstract
Metformin treatment is associated with a decreased risk and better prognosis of pancreatic cancer (PC) in patients with type 2 diabetes, but the mechanism of metformin's PC growth inhibition in the context of a prediabetic state is unknown. We used a Panc02 pancreatic tumor cell transplant model in diet-induced obese (DIO) C57BL/6 mice to compare the effects of metformin and the direct mammalian target of rapamycin (mTOR) inhibitor rapamycin on PC growth, glucose regulation, mTOR pathway signaling, and candidate microRNA (miR) expression. In DIO/prediabetic mice, metformin and rapamycin significantly reduced pancreatic tumor growth and mTOR-related signaling. The rapamycin effects centered on decreased mTOR-regulated growth and survival signaling, including increased expression of let-7b and cell cycle-regulating miRs. Metformin (but not rapamycin) reduced glucose and insulin levels and expression of miR-34a and its direct targets Notch, Slug, and Snail. Metformin also reduced the number and size of Panc02 tumor spheres in vitro and inhibited the expression of Notch in spheroids. Our results suggest that metformin and rapamycin can both inhibit pancreatic tumor growth in obese, prediabetic mice through shared and distinct mechanisms. Metformin and direct mTOR inhibitors, alone or possibly in combination, represent promising intervention strategies for breaking the diabetes-PC link.
Collapse
Affiliation(s)
- Vincenza Cifarelli
- Department of Medicine, Center for Human Nutrition, Washington University School of Medicine, St. Louis, MO
| | - Laura M Lashinger
- Department of Nutritional Sciences, The University of Texas at Austin, Austin, TX
| | - Kaylyn L Devlin
- Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, TX
| | - Sarah M Dunlap
- Department of Nutritional Sciences, The University of Texas at Austin, Austin, TX
| | - Jennifer Huang
- Department of Nutritional Sciences, The University of Texas at Austin, Austin, TX
| | - Rudolf Kaaks
- Division of Epidemiology, German Cancer Research Center, Heidelberg, Germany
| | - Michael N Pollak
- Departments of Medicine and Oncology, McGill University, Montreal, Canada
| | - Stephen D Hursting
- Department of Nutritional Sciences, The University of Texas at Austin, Austin, TX Department of Nutrition, University of North Carolina, Chapel Hill, NC Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC
| |
Collapse
|
43
|
Metabolic tumor burden is associated with major oncogenomic alterations and serum tumor markers in patients with resected pancreatic cancer. Cancer Lett 2015; 360:227-33. [PMID: 25687883 DOI: 10.1016/j.canlet.2015.02.014] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Revised: 02/08/2015] [Accepted: 02/09/2015] [Indexed: 12/27/2022]
Abstract
Pancreatic cancer is an aggressive and lethal disease with an overall 5-year survival rate of only 5%. Studies have demonstrated the ability of (18)F-fludrodeoxyglucose ((18)F-FDG) positron emission tomography/computed tomography (PET/CT) to measure the metabolic tumor burden in patients with various tumors, including pancreatic cancer. In a previous study, we investigated the predictive significance of the metabolic tumor burden in terms of the metabolic tumor volume (MTV) and total lesion glycolysis (TLG). In this study, we analyzed the correlation between metabolic tumor burden and the status of the KRAS, TP53, CDKN2A/p16, and SMAD4/DPC4 genes. Our results showed that the metabolic tumor burden was associated with oncogenomic alterations that reflected the abnormal expression of carbohydrate metabolic enzymes (GLUT1, ALDOA and FBP1). We also identified a linear correlation between serum tumor markers and the metabolic tumor burden. To estimate the metabolic tumor burden when (18)F-FDG PET/CT is not available, we used the linear regression models to establish equations for MTV and TLG using CA19-9 and CA125 as independent variables. Our results suggest that the metabolic tumor burden, as evaluated by (18)F-FDG PET/CT or estimated by serum tumor markers, may be suitable for monitoring treatment response and disease progression of pancreatic cancer. Further research is needed to better understand why pancreatic cancer patients with abnormal expressions of TP53, CDKN2A/p16, and SMAD4/DPC4 get high metabolic tumor burden.
Collapse
|
44
|
Ren H, Jia L, Zhao T, Zhang H, Chen J, Yang S, Liu J, Yu M, Hao J. Hypoxia inducible factor (HIF)-1α directly activates leptin receptor (Ob-R) in pancreatic cancer cells. Cancer Lett 2014; 354:172-80. [PMID: 25130171 DOI: 10.1016/j.canlet.2014.08.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Revised: 07/23/2014] [Accepted: 08/04/2014] [Indexed: 02/07/2023]
Abstract
The aim of this study is to investigate the regulatory mechanism of leptin receptors (Ob-R) in pancreatic cancer. We found that the over-expression of hypoxia inducible factor (HIF-1)α and hypoxia up-regulated the expression of Ob-R in pancreatic cancer cells. When HIF-1α gene was silenced in vitro, the expression of Ob-R was significantly decreased. Xenograft mouse models showed that the inhibition of HIF-1α resulted in the concomitant decrease of Ob-R in vivo. In addition, HIF-1α expression was correlated with Ob-R in pancreatic cancer tissues by immunohistochemical staining. Clinical data showed that over-expression of HIF-1 was associated with pathological tumor node metastasis stage, lymph node metastasis and overall survival. HIF-1α directly bound to the hypoxia-responsive element (HRE) located in Ob-R gene promoter (-828/-832) and activated the transcription. Finally, we demonstrated that the silence of HIF-1α gene reversed the inhibitory effect of leptin/Ob-R in pancreatic cancer cells. Taken together, our results indicate that HIF-1α directly regulated Ob-R expression in pancreatic cancer, which might be a valuable therapeutic target for pancreatic cancer.
Collapse
Affiliation(s)
- He Ren
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Lingling Jia
- School of Public Health, Tianjin Medical University, Tianjin, China
| | - Tiansuo Zhao
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Huan Zhang
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Jing Chen
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Shaoguang Yang
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Jingcheng Liu
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Ming Yu
- School of Public Health, Tianjin Medical University, Tianjin, China
| | - Jihui Hao
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.
| |
Collapse
|
45
|
Shi K, Queiroz KCS, Roelofs JJTH, van Noesel CJM, Richel DJ, Spek CA. Protease-activated receptor 2 suppresses lymphangiogenesis and subsequent lymph node metastasis in a murine pancreatic cancer model. J Pathol 2014; 234:398-409. [DOI: 10.1002/path.4411] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Revised: 07/14/2014] [Accepted: 07/18/2014] [Indexed: 12/18/2022]
Affiliation(s)
- Kun Shi
- Centre for Experimental and Molecular Medicine; Academic Medical Centre; Amsterdam The Netherlands
| | - Karla CS Queiroz
- Centre for Experimental and Molecular Medicine; Academic Medical Centre; Amsterdam The Netherlands
| | - Joris JTH Roelofs
- Department of Pathology; Academic Medical Centre; Amsterdam The Netherlands
| | | | - Dirk J Richel
- Department of Medical Oncology; Academic Medical Centre; Amsterdam The Netherlands
| | - C Arnold Spek
- Centre for Experimental and Molecular Medicine; Academic Medical Centre; Amsterdam The Netherlands
| |
Collapse
|
46
|
Torres MP, Rachagani S, Souchek JJ, Mallya K, Johansson SL, Batra SK. Novel pancreatic cancer cell lines derived from genetically engineered mouse models of spontaneous pancreatic adenocarcinoma: applications in diagnosis and therapy. PLoS One 2013; 8:e80580. [PMID: 24278292 PMCID: PMC3835415 DOI: 10.1371/journal.pone.0080580] [Citation(s) in RCA: 104] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2013] [Accepted: 10/14/2013] [Indexed: 01/05/2023] Open
Abstract
Pancreatic cancer (PC) remains one of the most lethal human malignancies with poor prognosis. Despite all advances in preclinical research, there have not been significant translation of novel therapies into the clinics. The development of genetically engineered mouse (GEM) models that produce spontaneous pancreatic adenocarcinoma (PDAC) have increased our understanding of the pathogenesis of the disease. Although these PDAC mouse models are ideal for studying potential therapies and specific genetic mutations, there is a need for developing syngeneic cell lines from these models. In this study, we describe the successful establishment and characterization of three cell lines derived from two (PDAC) mouse models. The cell line UN-KC-6141 was derived from a pancreatic tumor of a KrasG12D;Pdx1-Cre (KC) mouse at 50 weeks of age, whereas UN-KPC-960 and UN-KPC-961 cell lines were derived from pancreatic tumors of KrasG12D;Trp53R172H;Pdx1-Cre (KPC) mice at 17 weeks of age. The cancer mutations of these parent mice carried over to the daughter cell lines (i.e. KrasG12D mutation was observed in all three cell lines while Trp53 mutation was observed only in KPC cell lines). The cell lines showed typical cobblestone epithelial morphology in culture, and unlike the previously established mouse PDAC cell line Panc02, expressed the ductal marker CK19. Furthermore, these cell lines expressed the epithelial-mesenchymal markers E-cadherin and N-cadherin, and also, Muc1 and Muc4 mucins. In addition, these cell lines were resistant to the chemotherapeutic drug Gemcitabine. Their implantation in vivo produced subcutaneous as well as tumors in the pancreas (orthotopic). The genetic mutations in these cell lines mimic the genetic compendium of human PDAC, which make them valuable models with a high potential of translational relevance for examining diagnostic markers and therapeutic drugs.
Collapse
Affiliation(s)
- María P. Torres
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Satyanarayana Rachagani
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Joshua J. Souchek
- Buffett Cancer Center, Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Kavita Mallya
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Sonny L. Johansson
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Surinder K. Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- Buffett Cancer Center, Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- * E-mail:
| |
Collapse
|
47
|
Costello LC, Franklin RB. A Review of the Current Status and Concept of the Emerging Implications of Zinc and Zinc Transporters in the Development of Pancreatic Cancer. ACTA ACUST UNITED AC 2013; Suppl 4. [PMID: 24195024 DOI: 10.4172/2165-7092.s4-002] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Pancreatic cancer (adenocarcinoma) remains a deadly untreatable cancer with no effective early detection procedure. Little is known concerning the factors involved in the development of pancreatic malignancy, which impedes advancements in its treatment and detection. Altered cellular zinc has been implicated in several cancers. Recent studies provide evidence that zinc and zinc transporters are important factors in pancreatic cancer. This review discusses the current information relating to the status of zinc and zinc transporters in human pancreatic adenocarcinoma. Relationships of the physiology and biochemistry of zinc in mammalian cells are presented, which should be applied to the conduct, interpretation, and translational application of human studies and experimental models. Evidence from human pancreatic tissue studies supports a new concept of the role of zinc in the development of pancreatic adenocarcinoma. The zinc level of the normal ductal and acinar epithelium is markedly decreased in the development of the malignant cells and the premalignant PanIN cells. ZIP3 is identified as the likely zinc uptake transporter, which is down regulated concurrently with the loss of zinc. Ras responsive binding protein (RREB1) is identified as the possible transcription factor involved in the silencing of ZIP3 expression. The evidence supports the current views of transdifferentiation of PanIN epithelium to ductal adenocarcinoma, and the possibility that acinar epithelial dedifferentiation might be a source of premalignant cells. These zinc-associated events occur early in oncogenesis to protect the malignant cells from the cytotoxic effects of zinc levels that exist in the normal cells. Hopefully, this presentation will stimulate interest in and support for much needed research into the implications of zinc and zinc transporters as important events in pancreatic carcinogenesis. The potential exists for the RREB1-ZIP3-zinc concept and/or other implications of zinc as new approaches for the development of effective treatment and for diagnostic biomarkers for pancreatic cancer.
Collapse
Affiliation(s)
- Leslie C Costello
- Department of Oncology and Diagnostic Sciences, University of Maryland Dental School and The University of Maryland Greenebaum Cancer Center, Baltimore, Maryland, USA
| | | |
Collapse
|