1
|
Otowa Y, Kishimoto S, Saida Y, Yamashita K, Yamamoto K, Chandramouli GV, Devasahayam N, Mitchell JB, Krishna MC, Brender JR. Evofosfamide and Gemcitabine Act Synergistically in Pancreatic Cancer Xenografts by Dual Action on Tumor Vasculature and Inhibition of Homologous Recombination DNA Repair. Antioxid Redox Signal 2023; 39:432-444. [PMID: 37051681 PMCID: PMC10623073 DOI: 10.1089/ars.2022.0118] [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: 08/28/2022] [Revised: 03/02/2023] [Accepted: 03/13/2023] [Indexed: 04/14/2023]
Abstract
Aims: Pancreatic ductal adenocarcinomas (PDACs) form hypovascular and hypoxic tumors, which are difficult to treat with current chemotherapy regimens. Gemcitabine (GEM) is often used as a first-line treatment for PDACs but has issues with chemoresistance and penetration in the interior of the tumor. Evofosfamide, a hypoxia-activated prodrug, has been shown to be effective in combination with GEM, although the mechanism of each drug on the other has not been established. We used mouse xenografts from two cell lines (MIA Paca-2 and SU.86.86) with different tumor microenvironmental characteristics to probe the action of each drug on the other. Results: GEM treatment enhanced survival times in mice with SU.86.86 leg xenografts (hazard ratio [HR] = 0.35, p = 0.03) but had no effect on MIA Paca-2 mice (HR = 0.91, 95% confidence interval = 0.37-2.25, p = 0.84). Conversely, evofosfamide did not improve survival times in SU.86.86 mice to a statistically significant degree (HR = 0.57, p = 0.22). Electron paramagnetic resonance imaging showed that oxygenation worsened in MIA Paca-2 tumors when treated with GEM, providing a direct mechanism for the activation of the hypoxia-activated prodrug evofosfamide by GEM. Sublethal amounts of either treatment enhanced the toxicity of other treatment in vitro in SU.86.86 but not in MIA Paca-2. By the biomarker γH2AX, combination treatment increased the number of double-stranded DNA lesions in vitro for SU.86.86 but not MIA Paca-2. Innovation and Conclusion: The synergy between GEM and evofosfamide appears to stem from the dual action of GEMs effect on tumor vasculature and inhibition by GEM of the homologous recombination DNA repair process. Antioxid. Redox Signal. 39, 432-444.
Collapse
Affiliation(s)
- Yasunori Otowa
- Radiation Biology Branch, Center for Cancer Research, National Cancer Institute (NCI), NIH, Bethesda, Maryland, USA
| | - Shun Kishimoto
- Radiation Biology Branch, Center for Cancer Research, National Cancer Institute (NCI), NIH, Bethesda, Maryland, USA
| | - Yu Saida
- Radiation Biology Branch, Center for Cancer Research, National Cancer Institute (NCI), NIH, Bethesda, Maryland, USA
| | - Kota Yamashita
- Radiation Biology Branch, Center for Cancer Research, National Cancer Institute (NCI), NIH, Bethesda, Maryland, USA
| | - Kazutoshi Yamamoto
- Radiation Biology Branch, Center for Cancer Research, National Cancer Institute (NCI), NIH, Bethesda, Maryland, USA
| | - Gadisetti V.R. Chandramouli
- Radiation Biology Branch, Center for Cancer Research, National Cancer Institute (NCI), NIH, Bethesda, Maryland, USA
| | - Nallathamby Devasahayam
- Radiation Biology Branch, Center for Cancer Research, National Cancer Institute (NCI), NIH, Bethesda, Maryland, USA
| | - James B. Mitchell
- Radiation Biology Branch, Center for Cancer Research, National Cancer Institute (NCI), NIH, Bethesda, Maryland, USA
| | - Murali C. Krishna
- Radiation Biology Branch, Center for Cancer Research, National Cancer Institute (NCI), NIH, Bethesda, Maryland, USA
| | - Jeffrey R. Brender
- Radiation Biology Branch, Center for Cancer Research, National Cancer Institute (NCI), NIH, Bethesda, Maryland, USA
| |
Collapse
|
2
|
Zanini S, Renzi S, Limongi AR, Bellavite P, Giovinazzo F, Bermano G. A review of lifestyle and environment risk factors for pancreatic cancer. Eur J Cancer 2021; 145:53-70. [PMID: 33423007 DOI: 10.1016/j.ejca.2020.11.040] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 11/04/2020] [Accepted: 11/24/2020] [Indexed: 12/12/2022]
Abstract
Pancreatic cancer (PaCa) is one of the deadliest cancers known and its incidence is increasing in the developed countries. Because of the lack of biomarkers that allow early detection and the tendency of the disease to be asymptomatic, the diagnosis comes often too late for effective surgical or chemotherapy intervention. Lifestyle factors, that may cause common genetic modifications occurring in the disease, interfere with pancreatic physiology or function, and play a role in PaCa development, have been of concern recently, since a strategy to prevent this severe cancer is needed. This review identifies the latest evidences related to increased risk of developing PaCa due to dietary habits such as high alcohol, fructose and red or processed meat intake, and pathological conditions such as diabetes, obesity and infections in addition to stress and smoking behaviour. It aims to highlight the importance of intervening on modifiable risk factors: the action on these factors could prevent a considerable number of new cases of PaCa.
Collapse
Affiliation(s)
- Sara Zanini
- Centre for Obesity Research and Education [CORE], School of Pharmacy & Life Sciences, Robert Gordon University, Aberdeen, UK
| | - Serena Renzi
- Centre for Obesity Research and Education [CORE], School of Pharmacy & Life Sciences, Robert Gordon University, Aberdeen, UK
| | - Antonina R Limongi
- Department of Science, University of Basilicata, Potenza, Italy; BioInnova Srl, Potenza, Italy
| | - Paolo Bellavite
- Department of Medicine, Section of General Pathology, University of Verona, Italy
| | | | - Giovanna Bermano
- Centre for Obesity Research and Education [CORE], School of Pharmacy & Life Sciences, Robert Gordon University, Aberdeen, UK.
| |
Collapse
|
3
|
Malla RR, Kumari S, Amajala KC, Deepak KGK, Gugalavath S, Rokkam P. Methods and Models in Exploring Pancreatic Functions. EXPLORING PANCREATIC METABOLISM AND MALIGNANCY 2019:253-268. [DOI: 10.1007/978-981-32-9393-9_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
4
|
Moutinho-Ribeiro P, Macedo G, Melo SA. Pancreatic Cancer Diagnosis and Management: Has the Time Come to Prick the Bubble? Front Endocrinol (Lausanne) 2018; 9:779. [PMID: 30671023 PMCID: PMC6331408 DOI: 10.3389/fendo.2018.00779] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 12/11/2018] [Indexed: 02/01/2023] Open
Abstract
Pancreatic cancer (PC) is associated with poor prognosis and very dismal survival rates. The most effective possibility of cure is tumor resection, which is only possible in about 15% of patients diagnosed at early stages of disease progression. Recent whole-genome sequencing studies pointed genetic alterations in 12 core signaling pathways in PC. These observations hint at the possibility that the initial mutation in PC might appear nearly 20 years before any symptoms occur, suggesting that a large window of opportunity may exist for early detection. Biomarkers with the potential to identify pre-neoplastic disease or very early stages of cancer are of great promise to improve patient survival. The concept of liquid biopsy refers to a minimally invasive sampling and analysis of liquid biomarkers that can be isolated from body fluids, primarily blood, urine and saliva. A myriad of circulating molecules may be useful as tumor markers, including cell-free DNA (cfDNA), cell-free RNA (cfRNA), circulating tumor cells (CTC), circulating tumor proteins, and extracellular vesicles, more specifically exosomes. In this review, we discuss with more detail the potential role of exosomes in several aspects related to PC, from initiation to tumor progression and its applicability in early detection and treatment. Exosomes are small circulating extracellular vesicles of 50-150 nm in diameter released from the plasma membrane by almost all cells and exhibit some advantages over other biomarkers. Exosomes are central players of intercellular communication and they have been implicated in a series of biological process, including tumorigenesis, migration and metastasis. Several exosomal microRNAs and proteins have been observed to distinguish PC from benign pancreatic diseases and healthy controls. Besides their possible role in diagnosis, understanding exosomes functions in cancer has clarified the importance of microenvironment in PC progression as well as its influence in proliferation, metastasis and resistance to chemotherapy. Increasing knowledge on cancer exosomes provides valuable insights on new therapeutic targets and can potentially open new strategies to treat this disease. Continuous research is needed to ascertain the reliability of using exosomes and their content as potential biomarkers, so that, hopefully, in the near future, they will provide the opportunity for early diagnosis, treatment intervention and increase survival of PC patients.
Collapse
Affiliation(s)
- Pedro Moutinho-Ribeiro
- Department of Gastroenterology, Centro Hospitalar São João, Porto, Portugal
- Faculty of Medicine of the University of Porto, Porto, Portugal
| | - Guilherme Macedo
- Department of Gastroenterology, Centro Hospitalar São João, Porto, Portugal
- Faculty of Medicine of the University of Porto, Porto, Portugal
- *Correspondence: Guilherme Macedo
| | - Sónia A. Melo
- Faculty of Medicine of the University of Porto, Porto, Portugal
- Institute for Research Innovation in Health (i3S), Porto, Portugal
- Institute of Pathology and Molecular Immunology of the University of Porto, Porto, Portugal
- Sónia A. Melo
| |
Collapse
|
5
|
Xu MD, Liu SL, Feng YZ, Liu Q, Shen M, Zhi Q, Liu Z, Gu DM, Yu J, Shou LM, Gong FR, Zhu Q, Duan W, Chen K, Zhang J, Wu MY, Tao M, Li W. Genomic characteristics of pancreatic squamous cell carcinoma, an investigation by using high throughput sequencing after in-solution hybrid capture. Oncotarget 2017; 8:14620-14635. [PMID: 28099906 PMCID: PMC5362430 DOI: 10.18632/oncotarget.14678] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 01/09/2017] [Indexed: 12/23/2022] Open
Abstract
Squamous cell carcinoma (SCC) of pancreas is a rare histotype of pancreatic ductal carcinoma which is distinct from pancreatic adenocarcinoma (AC). Although there are standard treatments for pancreatic AC, no precise therapies exist for pancreatic SCC. Here, we screened 1033 cases of pancreatic cancer and identified 2 cases of pure SCC, which were pathologically diagnosed on the basis of finding definite intercellular bridges and/or focal keratin peal formation in the tumor cells. Immunohistochemistry assay confirmed the positive expression of CK5/6 and p63 in pancreatic SCC. To verify the genomic characteristics of pancreatic SCC, we employed in-solution hybrid capture targeting 137 cancer-related genes accompanied by high throughput sequencing (HTS) to compare the different genetic variants in SCC and AC of pancreas. We compared the genetic alterations of known biomarkers of pancreatic adenocarcinoma in different pancreatic cancer tissues, and identified nine mutated genes in SCC of pancreas: C7orf70, DNHD1, KPRP, MDM4, MUC6, OR51Q1, PTPRD, TCF4, TET2, and nine genes (ABCB1, CSF1R, CYP2C18, FBXW7, ITPA, KIAA0748, SOD2, SULT1A2, ZNF142) that are mutated in pancreatic AC. This study may have taken one step forward on the discovery of potential biomarkers for the targeted treatment of SCC of the pancreas.
Collapse
Affiliation(s)
- Meng-Dan Xu
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Shu-Ling Liu
- Department of Radiation Oncology, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Yi-Zhong Feng
- Department of Pathology, The Second Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Qiang Liu
- Department of Pathology, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200000, China
| | - Meng Shen
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Qiaoming Zhi
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Zeyi Liu
- Department of Respiratory Medicine, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Dong-Mei Gu
- Department of Pathology, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Jie Yu
- Department of Pathology, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Liu-Mei Shou
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
- Department of Oncology, The First Affiliated Hospital of Zhejiang Chinese Medicine University, Hangzhou 310006, China
| | - Fei-Ran Gong
- Department of Hematology, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Qi Zhu
- Xi'an Tianlong Science and Technology Co., Ltd., Xi'an 710018, China
| | - Weiming Duan
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Kai Chen
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Junning Zhang
- Department of Radiation Oncology, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Meng-Yao Wu
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Min Tao
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
- PREMED Key Laboratory for Precision Medicine, Soochow University, Suzhou 215021, China
- Jiangsu Institute of Clinical Immunology, Suzhou 215006, China
- Institute of Medical Biotechnology, Soochow University, Suzhou 215021, China
| | - Wei Li
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
- PREMED Key Laboratory for Precision Medicine, Soochow University, Suzhou 215021, China
- Jiangsu Institute of Clinical Immunology, Suzhou 215006, China
| |
Collapse
|
6
|
Alhasan SF, Haugk B, Ogle LF, Beale GS, Long A, Burt AD, Tiniakos D, Televantou D, Coxon F, Newell DR, Charnley R, Reeves HL. Sulfatase-2: a prognostic biomarker and candidate therapeutic target in patients with pancreatic ductal adenocarcinoma. Br J Cancer 2016; 115:797-804. [PMID: 27560551 PMCID: PMC5046211 DOI: 10.1038/bjc.2016.264] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 07/13/2016] [Accepted: 07/28/2016] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is the fifth most common cause of cancer death in the UK. Its poor prognosis is attributed to late detection and limited therapeutic options. Expression of SULF2, an endosulfatase that modulates heparan sulfate proteoglycan 6-O-sulfation and is reportedly tumourigenic in different types of cancer, was investigated. METHODS SULF2 expression was determined immunohistochemically in archival surgical resection tissue sections from 93 patients with a confirmed histological diagnosis of PDAC between 2002 and 2008 followed for a median of 9 years. Relationships with clinico-pathological parameters and patient survival were explored. RESULTS The majority of PDACs showed positive SULF2 staining in tumour cells and intratumoural or tumour-adjacent stroma. Greater than 25% SULF2-positive tumour cells was present in 60% of cancers and correlated with tumour stage (P=0.002) and perineural invasion (P=0.024). SULF2 intensity was scored moderate or strong in 81% of cancers and positively correlated with vascular invasion (P=0.015). High SULF2 expression, defined as >50% SULF2-positive tumour cells and strong SULF2 staining, was associated with shorter time to radiological progression (P=0.018, HR 1.98, CI 1.13-3.47). Similarly, by multivariate analysis, high SULF2 expression was independently associated with poorer survival (P=0.004, HR 2.10, CI 1.26-3.54), with a median survival of 11 months vs 21 months for lower PDAC SULF2. CONCLUSIONS Elevated SULF2 in PDAC was associated with advanced tumour stage, vascular invasion, shorter interval to radiological progression and shorter overall survival. SULF2 may have roles as a prognostic biomarker and as a therapeutic target for patients with PDAC.
Collapse
Affiliation(s)
- Sari F Alhasan
- Northern Institute for Cancer Research, Paul O'Gorman Building, The Medical School, Framlington Place, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Beate Haugk
- Department of Cellular Pathology, Royal Victoria Infirmary, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne NE1 4LP, UK
| | - Laura F Ogle
- Northern Institute for Cancer Research, Paul O'Gorman Building, The Medical School, Framlington Place, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Gary S Beale
- Northern Institute for Cancer Research, Paul O'Gorman Building, The Medical School, Framlington Place, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Anna Long
- Department of Cellular Pathology, Royal Victoria Infirmary, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne NE1 4LP, UK
| | - Alastair D Burt
- Department of Cellular Pathology, Royal Victoria Infirmary, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne NE1 4LP, UK
- School of Medicine, Eleanor Harrald Building, Frome Road, The University of Adelaide, Adelaide, 5000 South Australia, Australia
| | - Dina Tiniakos
- Department of Cellular Pathology, Royal Victoria Infirmary, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne NE1 4LP, UK
- Institute of Cellular Medicine, The Medical School, Framlington Place, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Despina Televantou
- Northern Institute for Cancer Research, Paul O'Gorman Building, The Medical School, Framlington Place, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
- Department of Cellular Pathology, Royal Victoria Infirmary, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne NE1 4LP, UK
| | - Fareeda Coxon
- Hepatopancreatobiliary multidisciplinary team, Freeman Hospital, Newcastle-upon-Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne NE7 7DN, UK
| | - David R Newell
- Northern Institute for Cancer Research, Paul O'Gorman Building, The Medical School, Framlington Place, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Richard Charnley
- Hepatopancreatobiliary multidisciplinary team, Freeman Hospital, Newcastle-upon-Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne NE7 7DN, UK
| | - Helen L Reeves
- Northern Institute for Cancer Research, Paul O'Gorman Building, The Medical School, Framlington Place, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
- Hepatopancreatobiliary multidisciplinary team, Freeman Hospital, Newcastle-upon-Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne NE7 7DN, UK
| |
Collapse
|
7
|
Tayao M, Andrici J, Farzin M, Clarkson A, Sioson L, Watson N, Chua TC, Sztynda T, Samra JS, Gill AJ. Loss of BAP1 Expression Is Very Rare in Pancreatic Ductal Adenocarcinoma. PLoS One 2016; 11:e0150338. [PMID: 26982343 PMCID: PMC4794169 DOI: 10.1371/journal.pone.0150338] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 02/12/2016] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Pancreatic cancer is both common and highly lethal and therefore new biomarkers or potential targets for treatment are needed. Loss of BRCA associated protein-1 (BAP1) expression has been found in up to a quarter of intrahepatic cholangiocarcinomas. Given the close anatomical relationship between intrahepatic cholangiocarcinoma and pancreatic ductal adenocarcinoma, we therefore sought to investigate the frequency of loss of BAP1 expression in pancreatic ductal adenocarcinoma. METHODS The records of the department of Anatomical Pathology Royal North Shore Hospital, Sydney, Australia, were searched for cases of pancreatic ductal adenocarcinoma diagnosed between 1992 and 2014 with material available in archived formalin fixed paraffin embedded tissue blocks. Immunohistochemistry for BAP1 was performed on tissue microarray sections and if staining was equivocal or negative it was confirmed on whole sections. Negative staining for BAP1 was defined as loss of expression in all neoplastic nuclei, with preserved expression in non-neoplastic cells which acted as an internal positive control. RESULTS Loss of BAP1 expression was found in only 1 of 306 (0.33%) pancreatic ductal adenocarcinomas. This case was confirmed to demonstrate diffuse loss of expression throughout all neoplastic cells in multiple blocks, consistent with BAP1 loss being an early clonal event. All other cases demonstrated positive expression of BAP1. CONCLUSION We conclude that, in contrast to intrahepatic cholangiocarcinoma, loss of expression of BAP1 occurs very rarely in pancreatic ductal adenocarcinoma. Therefore BAP1 inactivation is unlikely to be a frequent driver abnormality in pancreatic adenocarcinoma.
Collapse
Affiliation(s)
- Michael Tayao
- Cancer Diagnosis and Pathology Research Group, Kolling Institute of Medical Research, St Leonards, NSW, Australia, 2065
- School of Life Sciences, University of Technology Sydney, Ultimo, NSW, Australia, 2007
| | - Juliana Andrici
- Cancer Diagnosis and Pathology Research Group, Kolling Institute of Medical Research, St Leonards, NSW, Australia, 2065
- Sydney Medical School, University of Sydney, Sydney, NSW, Australia, 2006
- Department of Anatomical Pathology, Royal North Shore Hospital, St Leonards, NSW, Australia, 2065
| | - Mahtab Farzin
- Cancer Diagnosis and Pathology Research Group, Kolling Institute of Medical Research, St Leonards, NSW, Australia, 2065
- Department of Anatomical Pathology, Royal North Shore Hospital, St Leonards, NSW, Australia, 2065
| | - Adele Clarkson
- Cancer Diagnosis and Pathology Research Group, Kolling Institute of Medical Research, St Leonards, NSW, Australia, 2065
- Department of Anatomical Pathology, Royal North Shore Hospital, St Leonards, NSW, Australia, 2065
| | - Loretta Sioson
- Cancer Diagnosis and Pathology Research Group, Kolling Institute of Medical Research, St Leonards, NSW, Australia, 2065
- Department of Anatomical Pathology, Royal North Shore Hospital, St Leonards, NSW, Australia, 2065
| | - Nicole Watson
- Cancer Diagnosis and Pathology Research Group, Kolling Institute of Medical Research, St Leonards, NSW, Australia, 2065
| | - Terence C Chua
- Department of Gastrointestinal Surgery, Royal North Shore Hospital, St Leonards, NSW, Australia, and Discipline of Surgery, University of Sydney, Sydney, NSW, Australia
- Macquarie University Hospital, Macquarie University, North Ryde, NSW, Australia
| | - Tamara Sztynda
- School of Life Sciences, University of Technology Sydney, Ultimo, NSW, Australia, 2007
| | - Jaswinder S Samra
- Sydney Medical School, University of Sydney, Sydney, NSW, Australia, 2006
- Department of Gastrointestinal Surgery, Royal North Shore Hospital, St Leonards, NSW, Australia, and Discipline of Surgery, University of Sydney, Sydney, NSW, Australia
- Macquarie University Hospital, Macquarie University, North Ryde, NSW, Australia
| | - Anthony J Gill
- Cancer Diagnosis and Pathology Research Group, Kolling Institute of Medical Research, St Leonards, NSW, Australia, 2065
- Sydney Medical School, University of Sydney, Sydney, NSW, Australia, 2006
- Department of Anatomical Pathology, Royal North Shore Hospital, St Leonards, NSW, Australia, 2065
- Sydney Vital Translational Research Centre, Royal North Shore Hospital, Pacific Highway, St Leonards, NSW, Australia, 2065
| |
Collapse
|
8
|
Lu L, Risch HA. Exosomes: potential for early detection in pancreatic cancer. Future Oncol 2016; 12:1081-90. [PMID: 26860951 DOI: 10.2217/fon-2015-0005] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Progress in the treatment of patients with pancreatic cancer at earlier stages has motivated research in identifying novel noninvasive or minimally invasive biomarkers for early detection. Exosomes, which contain bioactive molecules (such as proteins, RNAs and lipids), are membrane-structured nanovesicles that are secreted from living cells and are found in human body fluids. As functional mediators, exosomes play key roles in cell-cell communications, regulating diverse biological processes. Here we aim to examine recent findings in the potential diagnostic value of serum exosomes in pancreatic cancer.
Collapse
Affiliation(s)
- Lingeng Lu
- Department of Chronic Disease Epidemiology, Yale School of Public Health, Yale School of Medicine, Yale Cancer Center, New Haven, CT 06520-8034, USA
| | - Harvey A Risch
- Department of Chronic Disease Epidemiology, Yale School of Public Health, Yale School of Medicine, Yale Cancer Center, New Haven, CT 06520-8034, USA
| |
Collapse
|
9
|
Wylie N, Koea JB. Prediction in pancreatic cancer. ANZ J Surg 2015; 85:796. [PMID: 26498563 DOI: 10.1111/ans.13288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Neil Wylie
- Department of Surgery, North Shore Hospital, Auckland, New Zealand
| | - Jonathan B Koea
- Department of Surgery, North Shore Hospital, Auckland, New Zealand
| |
Collapse
|
10
|
Miller BW, Morton JP, Pinese M, Saturno G, Jamieson NB, McGhee E, Timpson P, Leach J, McGarry L, Shanks E, Bailey P, Chang D, Oien K, Karim S, Au A, Steele C, Carter CR, McKay C, Anderson K, Evans TRJ, Marais R, Springer C, Biankin A, Erler JT, Sansom OJ. Targeting the LOX/hypoxia axis reverses many of the features that make pancreatic cancer deadly: inhibition of LOX abrogates metastasis and enhances drug efficacy. EMBO Mol Med 2015; 7:1063-76. [PMID: 26077591 PMCID: PMC4551344 DOI: 10.15252/emmm.201404827] [Citation(s) in RCA: 202] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2014] [Revised: 05/12/2015] [Accepted: 05/21/2015] [Indexed: 12/20/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the leading causes of cancer-related mortality. Despite significant advances made in the treatment of other cancers, current chemotherapies offer little survival benefit in this disease. Pancreaticoduodenectomy offers patients the possibility of a cure, but most will die of recurrent or metastatic disease. Hence, preventing metastatic disease in these patients would be of significant benefit. Using principal component analysis (PCA), we identified a LOX/hypoxia signature associated with poor patient survival in resectable patients. We found that LOX expression is upregulated in metastatic tumors from Pdx1-Cre Kras(G12D/+) Trp53(R172H/+) (KPC) mice and that inhibition of LOX in these mice suppressed metastasis. Mechanistically, LOX inhibition suppressed both migration and invasion of KPC cells. LOX inhibition also synergized with gemcitabine to kill tumors and significantly prolonged tumor-free survival in KPC mice with early-stage tumors. This was associated with stromal alterations, including increased vasculature and decreased fibrillar collagen, and increased infiltration of macrophages and neutrophils into tumors. Therefore, LOX inhibition is able to reverse many of the features that make PDAC inherently refractory to conventional therapies and targeting LOX could improve outcome in surgically resectable disease.
Collapse
Affiliation(s)
- Bryan W Miller
- Cancer Research UK Beatson Institute Garscube Estate, Glasgow, UK
| | | | - Mark Pinese
- The Garvan Institute of Medical Research, Sydney, NSW, Australia
| | - Grazia Saturno
- Cancer Research UK Manchester Institute, Withington Manchester, UK
| | - Nigel B Jamieson
- West of Scotland Pancreatic Unit, Glasgow Royal Infirmary, Glasgow, UK
| | - Ewan McGhee
- Cancer Research UK Beatson Institute Garscube Estate, Glasgow, UK
| | - Paul Timpson
- The Garvan Institute of Medical Research, Sydney, NSW, Australia
| | - Joshua Leach
- Cancer Research UK Beatson Institute Garscube Estate, Glasgow, UK
| | - Lynn McGarry
- Cancer Research UK Beatson Institute Garscube Estate, Glasgow, UK
| | - Emma Shanks
- Cancer Research UK Beatson Institute Garscube Estate, Glasgow, UK
| | - Peter Bailey
- Institute of Cancer Sciences University of Glasgow Garscube Estate, Glasgow, UK
| | - David Chang
- Institute of Cancer Sciences University of Glasgow Garscube Estate, Glasgow, UK
| | - Karin Oien
- Institute of Cancer Sciences University of Glasgow Garscube Estate, Glasgow, UK
| | - Saadia Karim
- Cancer Research UK Beatson Institute Garscube Estate, Glasgow, UK
| | - Amy Au
- Cancer Research UK Beatson Institute Garscube Estate, Glasgow, UK
| | - Colin Steele
- Cancer Research UK Beatson Institute Garscube Estate, Glasgow, UK
| | | | - Colin McKay
- West of Scotland Pancreatic Unit, Glasgow Royal Infirmary, Glasgow, UK
| | - Kurt Anderson
- Cancer Research UK Beatson Institute Garscube Estate, Glasgow, UK
| | - Thomas R Jeffry Evans
- Cancer Research UK Beatson Institute Garscube Estate, Glasgow, UK Institute of Cancer Sciences University of Glasgow Garscube Estate, Glasgow, UK
| | - Richard Marais
- Cancer Research UK Manchester Institute, Withington Manchester, UK
| | | | - Andrew Biankin
- Institute of Cancer Sciences University of Glasgow Garscube Estate, Glasgow, UK
| | - Janine T Erler
- Biotech Research & Innovation Centre (BRIC), University of Copenhagen, Copenhagen (UCPH), Denmark
| | - Owen J Sansom
- Cancer Research UK Beatson Institute Garscube Estate, Glasgow, UK
| |
Collapse
|
11
|
Neureiter D, Jäger T, Ocker M, Kiesslich T. Epigenetics and pancreatic cancer: pathophysiology and novel treatment aspects. World J Gastroenterol 2014; 20:7830-7848. [PMID: 24976721 PMCID: PMC4069312 DOI: 10.3748/wjg.v20.i24.7830] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2013] [Revised: 02/07/2014] [Accepted: 03/12/2014] [Indexed: 02/06/2023] Open
Abstract
An improvement in pancreatic cancer treatment represents an urgent medical goal. Late diagnosis and high intrinsic resistance to conventional chemotherapy has led to a dismal overall prognosis that has remained unchanged during the past decades. Increasing knowledge about the molecular pathogenesis of the disease has shown that genetic alterations, such as mutations of K-ras, and especially epigenetic dysregulation of tumor-associated genes, such as silencing of the tumor suppressor p16(ink4a), are hallmarks of pancreatic cancer. Here, we describe genes that are commonly affected by epigenetic dysregulation in pancreatic cancer via DNA methylation, histone acetylation or miRNA (microRNA) expression, and review the implications on pancreatic cancer biology such as epithelial-mesenchymal transition, morphological pattern formation, or cancer stem cell regulation during carcinogenesis from PanIN (pancreatic intraepithelial lesions) to invasive cancer and resistance development. Epigenetic drugs, such as DNA methyltransferases or histone deactylase inhibitors, have shown promising preclinical results in pancreatic cancer and are currently in early phases of clinical development. Combinations of epigenetic drugs with established cytotoxic drugs or targeted therapies are promising approaches to improve the poor response and survival rate of pancreatic cancer patients.
Collapse
|
12
|
The use of multidimensional data to identify the molecular biomarker for pancreatic ductal adenocarcinoma. BIOMED RESEARCH INTERNATIONAL 2014; 2013:798054. [PMID: 24171174 PMCID: PMC3793503 DOI: 10.1155/2013/798054] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2013] [Accepted: 08/23/2013] [Indexed: 12/14/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a lethal disease, and the patient has an extremely poor overall survival with a less than 5% 5-year survival rate. Development of potential biomarkers provides a critical foundation for the diagnosis of PDAC. In this project, we have adopted an integrative approach to simultaneously identify biomarker and generate testable hypothesis from multidimensional omics data. We first examine genes for which expression levels are correlated with survival data. The gene list was screened with TF regulation, predicted miRNA targets information, and KEGG pathways. We identified that 273 candidate genes are correlated with patient survival data. 12 TF regulation gene sets, 11 miRNAs targets gene sets, and 15 KEGG pathways are enriched with these survival genes. Notably, CEBPA/miRNA32/PER2 signaling to the clock rhythm qualifies this pathway as a suitable target for therapeutic intervention in PDAC. PER2 expression was highly associated with survival data, thus representing a novel biomarker for earlier detection of PDAC.
Collapse
|
13
|
Abstract
Over the last decade, it has been discovered that the transcription factor Sox9 plays several critical roles in governing the development of the embryonic pancreas and the homeostasis of the mature organ. While analysis of pancreata from patients affected by the Sox9 haploinsufficiency syndrome campomelic dysplasia initially alluded to a functional role of Sox9 in pancreatic morphogenesis, transgenic mouse models have been instrumental in mechanistically dissecting such roles. Although initially defined as a marker and maintenance factor for pancreatic progenitors, Sox9 is now considered to fulfill additional indispensable functions during pancreogenesis and in the postnatal organ through its interactions with other transcription factors and signaling pathways such as Fgf and Notch. In addition to maintaining both multipotent and bipotent pancreatic progenitors, Sox9 is also required for initiating endocrine differentiation and maintaining pancreatic ductal identity, and it has recently been unveiled as a key player in the initiation of pancreatic cancer. These functions of Sox9 are discussed in this article, with special emphasis on the knowledge gained from various loss-of-function and lineage tracing mouse models. Also, current controversies regarding Sox9 function in healthy and injured adult pancreas and unanswered questions and avenues of future study are discussed.
Collapse
Affiliation(s)
- Philip A Seymour
- The Danish Stem Cell Center (DanStem), University of Copenhagen, Panum Institute, Blegdamsvej 3B, DK-2200, Copenhagen N, Denmark
| |
Collapse
|
14
|
Zhu ZM, Xu YF, Su QJ, Du JD, Tan XL, Tu YL, Tan JW, Jiao HB. Prognostic significance of microRNA-141 expression and its tumor suppressor function in human pancreatic ductal adenocarcinoma. Mol Cell Biochem 2013; 388:39-49. [PMID: 24242138 DOI: 10.1007/s11010-013-1897-y] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Accepted: 11/05/2013] [Indexed: 01/03/2023]
Abstract
Increasing evidence shows that dysregulation of microRNAs is correlated with tumor development. This study was performed to determine the expression of miR-141 and investigate its clinical significance in pancreatic ductal adenocarcinoma (PDAC). Taqman quantitative RT-PCR was used to detect miR-141 expressions in 94 PDAC tissues and 16 nontumorous pancreatic tissues. Correlations between miR-141 expression and clinicopathologic features and prognosis of patients were statistically analyzed. The effects of miR-141 expression on growth and apoptosis of PDAC cell line (PANC-1) were determined by MTT, colony formation, and flow cytometry assays. Potential target genes were identified by luciferase reporter and Western blot assays. The expression level of miR-141 in PDAC tissues was significantly lower than that in corresponding nontumorous tissues. Downregulation of miR-141 correlated with poorer pT and pN status, advanced clinical stage, and lymphatic invasion. Also, low miR-141 expression in PDAC tissues was significantly correlated with shorter overall survival, and multivariate analysis showed that miR-141 was an independent prognostic factor for PDAC patients. Further, functional researches suggested that miR-141 inhibits growth and colony formation, and enhances caspase-3-dependent apoptosis in PANC-1 cells by targeting Yes-associated protein-1 (YAP1). Therefore, miR-141 is an independent prognostic factor for PDAC patients, and functions as a tumor suppressor gene by targeting YAP1.
Collapse
Affiliation(s)
- Zi-Man Zhu
- Department of Hepatobiliary Surgery, First Hospital Affiliated to the Chinese PLA General Hospital, Fucheng Road 51, Haidian District, Beijing, 100048, China
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Zöller M. Pancreatic cancer diagnosis by free and exosomal miRNA. World J Gastrointest Pathophysiol 2013; 4:74-90. [PMID: 24340225 PMCID: PMC3858795 DOI: 10.4291/wjgp.v4.i4.74] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2013] [Revised: 08/01/2013] [Accepted: 09/18/2013] [Indexed: 02/06/2023] Open
Abstract
Patients with pancreatic adenocarcinoma (PaCa) have a dismal prognosis. This is in part due to late diagnosis prohibiting surgical intervention, which provides the only curative option as PaCa are mostly chemo- and radiation resistance. Hope is raised on a reliable non-invasive/minimally invasive diagnosis that is still missing. Recently two diagnostic options are discussed, serum MicroRNA (miRNA) and serum exosomes. Serum miRNA can be free or vesicle-, particularly, exosomes-enclosed. This review will provide an overview on the current state of the diagnostic trials on free serum miRNA and proceed with an introduction of exosomes that use as a diagnostic tool in serum and other body fluids has not received sufficient attention, although serum exosome miRNA in combination with protein marker expression likely will increase the diagnostic and prognostic power. By their crosstalk with host cells, which includes binding-initiated signal transduction, as well as reprogramming target cells via the transfer of proteins, mRNA and miRNA exosomes are suggested to become a most powerful therapeutics. I will discuss which hurdles have still to be taken as well as the different modalities, which can be envisaged to make therapeutic use of exosomes. PaCa are known to most intensely crosstalk with the host as apparent by desmoplasia and frequent paraneoplastic syndromes. Thus, there is hope that the therapeutic application of exosomes brings about a major breakthrough.
Collapse
|