1
|
Cutrona MB, Wu J, Yang K, Peng J, Chen T. Pancreatic cancer organoid-screening captures personalized sensitivity and chemoresistance suppression upon cytochrome P450 3A5-targeted inhibition. iScience 2024; 27:110289. [PMID: 39055940 PMCID: PMC11269815 DOI: 10.1016/j.isci.2024.110289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 04/12/2024] [Accepted: 06/13/2024] [Indexed: 07/28/2024] Open
Abstract
Cytochrome P450 3A5 (CYP3A5) has been proposed as a predictor of therapy response in subtypes of pancreatic ductal adenocarcinoma cancer (PDAC). To validate CYP3A5 as a therapeutic target, we developed a high-content image organoid-based screen to quantify the phenotypic responses to the selective inhibition of CYP3A5 enzymatic activity by clobetasol propionate (CBZ), using a cohort of PDAC-derived organoids (PDACOs). The chemoresistance of PDACOs to a panel of standard-of-care drugs, alone or in combination with CBZ, was investigated. PDACO pharmaco-profiling revealed CBZ to have anti-cancer activity that was dependent on the CYP3A5 level. In addition, CBZ restored chemo-vulnerability to cisplatin in a subset of PDACOs. A correlative proteomic analysis established that CBZ caused the suppression of multiple cancer pathways sustained by or associated with a mutant form of p53. Limiting the active pool of CYP3A5 enables targeted and personalized therapy to suppress pro-oncogenic mechanisms that fuel chemoresistance in some PDAC tumors.
Collapse
Affiliation(s)
- Meritxell B. Cutrona
- Department of Chemical Biology and Therapeutics, St. Jude Children’s Research Hospital, Memphis, TN 38105-3678, USA
| | - Jing Wu
- Department of Chemical Biology and Therapeutics, St. Jude Children’s Research Hospital, Memphis, TN 38105-3678, USA
| | - Ka Yang
- Department of Structural Biology, St. Jude Children’s Research Hospital, Memphis, TN 38105-3678, USA
| | - Junmin Peng
- Department of Structural Biology, St. Jude Children’s Research Hospital, Memphis, TN 38105-3678, USA
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, TN 38105-3678, USA
| | - Taosheng Chen
- Department of Chemical Biology and Therapeutics, St. Jude Children’s Research Hospital, Memphis, TN 38105-3678, USA
| |
Collapse
|
2
|
Xiang F, Luo F. Stem cell factor modulates HIF-1α levels and diminishes 5-FU sensitivity in 5-FU resistant pancreatic cells by altering the anabolic glucose metabolism. J Biochem Mol Toxicol 2023; 37:e23487. [PMID: 37718545 DOI: 10.1002/jbt.23487] [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: 03/30/2023] [Revised: 06/19/2023] [Accepted: 07/31/2023] [Indexed: 09/19/2023]
Abstract
Resistance to chemotherapy in cancer leads to poor therapeutic outcomes and also leads to challenges in treatment. The present work evaluated the mechanism involved in the resistance of 5-flurouracil (5-FU) in pancreatic cancer. At least 14 different pancreatic cancer (PC) cell lines were used for the study. For in vivo study female nude mice were selected. Patient-derived tumor xenograft samples were obtained from patients. The study involved, study for glucose uptake, fluorescence-activated cell sorting for glucose transporter, 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide for cell survival, Picto-micrography for clonogenic assay, glutamine uptake assay, extracellular acidification and oxygen consumption rate, carbon dioxide release assay and lactate assay were also done. In addition to this, quantitative real-time polymerase chain reaction analysis for expression of genes, chromatin immunoprecipitation assay, western blot for protein expression, and immunohistochemical analysis in tumor sections, the tumors were studied by imaging for hypoxia and localization of TKT and CTPS-2. Also, patient-derived xenograft tumors were engrafted in nude mice, followed by a glucose uptake assay. We reported that elevated glycolytic flux causes dependence on glucose in cancer cells and, at the same time, increases pyrimidine biosynthesis. It was also found that stem cell factor-mediated stabilization of hypoxia-inducible factor-1a (HIF-1α) modulates the resistance in PC. Targeting HIF-1α in combination with 5-FU, strongly reduced the tumor burden. The study concludes that stem cell factor modulates HIF-1α and decreases the sensitivity in 5-FU resistant pancreatic cancer cells by targeting glucose metabolism. Deceased expression levels of CTPS-2 and TKT, which are regulators of pyrimidine biosynthesis could better the chance of survival in patients of pancreatic cancer receiving treatment of 5-FU.
Collapse
Affiliation(s)
- Fu Xiang
- Department of General Surgery, Dalian Medical University, Dalian, Liaoning, China
| | - Fuwen Luo
- Department of Acute Abdominal Surgery, Dalian Medical University, Dalian, Liaoning, China
| |
Collapse
|
3
|
Nasimi Shad A, Fanoodi A, Maharati A, Akhlaghipour I, Moghbeli M. Molecular mechanisms of microRNA-301a during tumor progression and metastasis. Pathol Res Pract 2023; 247:154538. [PMID: 37209575 DOI: 10.1016/j.prp.2023.154538] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 05/10/2023] [Accepted: 05/16/2023] [Indexed: 05/22/2023]
Abstract
Cancer is known as one of the leading causes of human deaths globally. Late diagnosis is considered as one of the main reasons for the high mortality rate among cancer patients. Therefore, the introduction of early diagnostic tumor markers can improve the efficiency of therapeutic modalities. MicroRNAs (miRNAs) have a key role in regulation of cell proliferation and apoptosis. MiRNAs deregulation has been frequently reported during tumor progressions. Since, miRNAs have a high stability in body fluids; they can be used as the reliable non-invasive tumor markers. Here, we discussed the role of miR-301a during tumor progressions. MiR-301a mainly functions as an oncogene via the modulation of transcription factors, autophagy, epithelial-mesenchymal transition (EMT), and signaling pathways. This review paves the way to suggest miR-301a as a non-invasive marker for the early tumor diagnosis. MiR-301a can also be suggested as an effective target in cancer therapy.
Collapse
Affiliation(s)
- Arya Nasimi Shad
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Fanoodi
- Student Research Committee, Faculty of Medicine, Birjand University of Medical Sciences, Mashhad, Iran
| | - Amirhosein Maharati
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Iman Akhlaghipour
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Meysam Moghbeli
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
4
|
Majeed S, Shamsunazatul NINB, Danish M, Ibrahim MNM, Muthukumarasamy R, Ansari MT. Mitochondrial Membrane Depolarization, Oxidative Stress Induced Cell Death in Human Pancreatic Adenocarcinoma Cells (PANC-1) Treated with Biologically Engineered Gallic Acid Coated Copper Oxide Nanoparticles. J CLUST SCI 2023. [DOI: 10.1007/s10876-023-02420-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/17/2023]
|
5
|
Tumor Treating Fields (TTFields) Therapy Concomitant with Taxanes for Cancer Treatment. Cancers (Basel) 2023; 15:cancers15030636. [PMID: 36765594 PMCID: PMC9913762 DOI: 10.3390/cancers15030636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 01/13/2023] [Accepted: 01/16/2023] [Indexed: 01/22/2023] Open
Abstract
Non-small cell lung cancer, ovarian cancer, and pancreatic cancer all present with high morbidity and mortality. Systemic chemotherapies have historically been the cornerstone of standard of care (SOC) regimens for many cancers, but are associated with systemic toxicity. Multimodal treatment combinations can help improve patient outcomes; however, implementation is limited by additive toxicities and potential drug-drug interactions. As such, there is a high unmet need to develop additional therapies to enhance the efficacy of SOC treatments without increasing toxicity. Tumor Treating Fields (TTFields) are electric fields that exert physical forces to disrupt cellular processes critical for cancer cell viability and tumor progression. The therapy is locoregional and is delivered noninvasively to the tumor site via a portable medical device that consists of field generator and arrays that are placed on the patient's skin. As a noninvasive treatment modality, TTFields therapy-related adverse events mainly consist of localized skin reactions, which are manageable with effective acute and prophylactic treatments. TTFields selectively target cancer cells through a multi-mechanistic approach without affecting healthy cells and tissues. Therefore, the application of TTFields therapy concomitant with other cancer treatments may lead to enhanced efficacy, with low risk of further systemic toxicity. In this review, we explore TTFields therapy concomitant with taxanes in both preclinical and clinical settings. The summarized data suggest that TTFields therapy concomitant with taxanes may be beneficial in the treatment of certain cancers.
Collapse
|
6
|
Roth HE, De Lima Leite A, Palermo NY, Powers R. Leveraging the Structure of DNAJA1 to Discover Novel Potential Pancreatic Cancer Therapies. Biomolecules 2022; 12:1391. [PMID: 36291603 PMCID: PMC9599757 DOI: 10.3390/biom12101391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 09/23/2022] [Accepted: 09/26/2022] [Indexed: 11/16/2022] Open
Abstract
Pancreatic cancer remains one of the deadliest forms of cancer with a 5-year survival rate of only 11%. Difficult diagnosis and limited treatment options are the major causes of the poor outcome for pancreatic cancer. The human protein DNAJA1 has been proposed as a potential therapeutic target for pancreatic cancer, but its cellular and biological functions remain unclear. Previous studies have suggested that DNAJA1's cellular activity may be dependent upon its protein binding partners. To further investigate this assertion, the first 107 amino acid structures of DNAJA1 were solved by NMR, which includes the classical J-domain and its associated linker region that is proposed to be vital to DNAJA1 functionality. The DNAJA1 NMR structure was then used to identify both protein and ligand binding sites and potential binding partners that may suggest the intracellular roles of DNAJA1. Virtual drug screenings followed by NMR and isothermal titration calorimetry identified 5 drug-like compounds that bind to two different sites on DNAJA1. A pull-down assay identified 8 potentially novel protein binding partners of DNAJA1. These proteins in conjunction with our previously published metabolomics study support a vital role for DNAJA1 in cellular oncogenesis and pancreatic cancer.
Collapse
Affiliation(s)
- Heidi E. Roth
- Department of Chemistry, University of Nebraska-Lincoln, Lincoln, NE 68588, USA
| | - Aline De Lima Leite
- Department of Chemistry, University of Nebraska-Lincoln, Lincoln, NE 68588, USA
- Nebraska Center for Integrated Biomolecular Communication, University of Nebraska-Lincoln, Lincoln, NE 68588, USA
| | - Nicolas Y. Palermo
- Computational Chemistry Core Facility, VCR Cores, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Robert Powers
- Department of Chemistry, University of Nebraska-Lincoln, Lincoln, NE 68588, USA
- Nebraska Center for Integrated Biomolecular Communication, University of Nebraska-Lincoln, Lincoln, NE 68588, USA
| |
Collapse
|
7
|
Roth HE, Powers R. Meta-Analysis Reveals Both the Promises and the Challenges of Clinical Metabolomics. Cancers (Basel) 2022; 14:3992. [PMID: 36010984 PMCID: PMC9406125 DOI: 10.3390/cancers14163992] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 08/09/2022] [Accepted: 08/16/2022] [Indexed: 11/17/2022] Open
Abstract
Clinical metabolomics is a rapidly expanding field focused on identifying molecular biomarkers to aid in the efficient diagnosis and treatment of human diseases. Variations in study design, metabolomics methodologies, and investigator protocols raise serious concerns about the accuracy and reproducibility of these potential biomarkers. The explosive growth of the field has led to the recent availability of numerous replicate clinical studies, which permits an evaluation of the consistency of biomarkers identified across multiple metabolomics projects. Pancreatic ductal adenocarcinoma (PDAC) is the third-leading cause of cancer-related death and has the lowest five-year survival rate primarily due to the lack of an early diagnosis and the limited treatment options. Accordingly, PDAC has been a popular target of clinical metabolomics studies. We compiled 24 PDAC metabolomics studies from the scientific literature for a detailed meta-analysis. A consistent identification across these multiple studies allowed for the validation of potential clinical biomarkers of PDAC while also highlighting variations in study protocols that may explain poor reproducibility. Our meta-analysis identified 10 metabolites that may serve as PDAC biomarkers and warrant further investigation. However, 87% of the 655 metabolites identified as potential biomarkers were identified in single studies. Differences in cohort size and demographics, p-value choice, fold-change significance, sample type, handling and storage, data collection, and analysis were all factors that likely contributed to this apparently large false positive rate. Our meta-analysis demonstrated the need for consistent experimental design and normalized practices to accurately leverage clinical metabolomics data for reliable and reproducible biomarker discovery.
Collapse
Affiliation(s)
- Heidi E. Roth
- Department of Chemistry, University of Nebraska-Lincoln, Lincoln, NE 68588-0304, USA
| | - Robert Powers
- Department of Chemistry, University of Nebraska-Lincoln, Lincoln, NE 68588-0304, USA
- Nebraska Center for Integrated Biomolecular Communication, University of Nebraska-Lincoln, Lincoln, NE 68588-0304, USA
| |
Collapse
|
8
|
Rana M, Kansal R, Chaib M, Teng B, Morrrison M, Hayes DN, Stanfill AG, Shibata D, Carson JA, Makowski L, Glazer ES. The pancreatic cancer immune tumor microenvironment is negatively remodeled by gemcitabine while TGF-β receptor plus dual checkpoint inhibition maintains antitumor immune cells. Mol Carcinog 2022; 61:549-557. [PMID: 35319799 DOI: 10.1002/mc.23401] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 01/25/2022] [Accepted: 02/15/2022] [Indexed: 01/19/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDA) tumors have a highly immunosuppressive desmoplastic tumor microenvironment (TME) where immune checkpoint inhibition (ICI) therapy has been exceptionally ineffective. Transforming growth factor-β (TGF-β) receptor activation leads to cancer and immune cell proliferation and phenotype, and cytokine production leading to tumor progression and worse overall survival in PDA patients. We hypothesized that TGF-β receptor inhibition may alter PDA progression and antitumor immunity in the TME. Here, we used a syngeneic preclinical murine model of PDA to explore the impact of TGF-β pathway inhibitor galunisertib (GAL), dual checkpoint immunotherapy (anti-PD-L1 and CTLA-4), the chemotherapy gemcitabine (GEM), and their combinations on antitumor immune responses. Blockade of TGF-β and ICI in immune-competent mice bearing orthotopically injected murine PDA cells significantly inhibited tumor growth and was accompanied by antitumor M1 macrophage infiltration. In contrast, GEM treatment resulted in increased PDA tumor growth, decreased antitumor M1 macrophages, and decreased cytotoxic CD8+ T cell subpopulation compared to control mice. Together, these findings demonstrate the ability of TGF-β inhibition with GAL to prime antitumor immunity in the TME and the curative potential of combining GAL with dual ICI. These preclinical results indicate that targeted inhibition of TGF-β may enhance the efficacy of dual immunotherapy in PDA. Optimal manipulation of the immune TME with non-ICI therapy may enhance therapeutic efficacy.
Collapse
Affiliation(s)
- Manjul Rana
- Department of Surgery, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Rita Kansal
- Department of Surgery, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Mehdi Chaib
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Bin Teng
- Center for Cancer Research, College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Michelle Morrrison
- Center for Cancer Research, College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - David Neil Hayes
- Center for Cancer Research, College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
- Department of Medicine, Division of Hematology and Oncology, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Ansley G Stanfill
- Department of Nursing Science, College of Nursing, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - David Shibata
- Department of Surgery, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
- Center for Cancer Research, College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - James A Carson
- Department of Physical Therapy, College of Health Professions, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Liza Makowski
- Center for Cancer Research, College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
- Department of Medicine, Division of Hematology and Oncology, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Evan S Glazer
- Department of Surgery, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
- Center for Cancer Research, College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
| |
Collapse
|
9
|
Kumar AA, Buckley BJ, Ranson M. The Urokinase Plasminogen Activation System in Pancreatic Cancer: Prospective Diagnostic and Therapeutic Targets. Biomolecules 2022; 12:152. [PMID: 35204653 PMCID: PMC8961517 DOI: 10.3390/biom12020152] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/13/2022] [Accepted: 01/16/2022] [Indexed: 02/07/2023] Open
Abstract
Pancreatic cancer is a highly aggressive malignancy that features high recurrence rates and the poorest prognosis of all solid cancers. The urokinase plasminogen activation system (uPAS) is strongly implicated in the pathophysiology and clinical outcomes of patients with pancreatic ductal adenocarcinoma (PDAC), which accounts for more than 90% of all pancreatic cancers. Overexpression of the urokinase-type plasminogen activator (uPA) or its cell surface receptor uPAR is a key step in the acquisition of a metastatic phenotype via multiple mechanisms, including the increased activation of cell surface localised plasminogen which generates the serine protease plasmin. This triggers multiple downstream processes that promote tumour cell migration and invasion. Increasing clinical evidence shows that the overexpression of uPA, uPAR, or of both is strongly associated with worse clinicopathological features and poor prognosis in PDAC patients. This review provides an overview of the current understanding of the uPAS in the pathogenesis and progression of pancreatic cancer, with a focus on PDAC, and summarises the substantial body of evidence that supports the role of uPAS components, including plasminogen receptors, in this disease. The review further outlines the clinical utility of uPAS components as prospective diagnostic and prognostic biomarkers for PDAC, as well as a rationale for the development of novel uPAS-targeted therapeutics.
Collapse
Affiliation(s)
- Ashna A. Kumar
- Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia; (A.A.K.); (B.J.B.)
- School of Chemistry and Molecular Biosciences, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, NSW 2522, Australia
| | - Benjamin J. Buckley
- Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia; (A.A.K.); (B.J.B.)
- School of Chemistry and Molecular Biosciences, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, NSW 2522, Australia
| | - Marie Ranson
- Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia; (A.A.K.); (B.J.B.)
- School of Chemistry and Molecular Biosciences, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, NSW 2522, Australia
| |
Collapse
|
10
|
Kim YE, Kim EK, Song MJ, Kim TY, Jang HH, Kang D. SILAC-Based Quantitative Proteomic Analysis of Oxaliplatin-Resistant Pancreatic Cancer Cells. Cancers (Basel) 2021; 13:cancers13040724. [PMID: 33578797 PMCID: PMC7916634 DOI: 10.3390/cancers13040724] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 02/07/2021] [Indexed: 12/13/2022] Open
Abstract
Simple Summary Resistance to oxaliplatin remains a major challenge in pancreatic cancer therapy. However, molecular mechanisms underlying oxaliplatin resistance in pancreatic cancer is still unclear. The aim of this study was to identify global changes of proteins involved in oxaliplatin resistance in pancreatic cancer cells, thereby elucidating the multiple mechanisms of oxaliplatin resistance in pancreatic cancer. We presented the quantitative proteomic profiling of oxaliplatin-resistant pancreatic cancer cells via a stable isotope labelling by amino acids in cell culture (SILAC)-based shotgun proteomic approach. Multiple biological processes including DNA repair, cell cycle process, and type I interferon signaling pathway were enriched in oxaliplatin-resistant pancreatic cancer cells. Furthermore, we demonstrated that both Wntless homolog protein (WLS) and myristoylated alanine-rich C-kinase substrate (MARCKS) could participate in oxaliplatin resistance in pancreatic cancer cells. Abstract Oxaliplatin is a commonly used chemotherapeutic drug for the treatment of pancreatic cancer. Understanding the cellular mechanisms of oxaliplatin resistance is important for developing new strategies to overcome drug resistance in pancreatic cancer. In this study, we performed a stable isotope labelling by amino acids in cell culture (SILAC)-based quantitative proteomics analysis of oxaliplatin-resistant and sensitive pancreatic cancer PANC-1 cells. We identified 107 proteins whose expression levels changed (thresholds of 2-fold changes and p-value ≤ 0.05) between oxaliplatin-resistant and sensitive cells, which were involved in multiple biological processes, including DNA repair, cell cycle process, and type I interferon signaling pathway. Notably, myristoylated alanine-rich C-kinase substrate (MARCKS) and Wntless homolog protein (WLS) were upregulated in oxaliplatin-resistant cells compared to sensitive cells, as confirmed by qRT-PCR and Western blot analysis. We further demonstrated the activation of AKT and β-catenin signaling (downstream targets of MARCKS and WLS, respectively) in oxaliplatin-resistant PANC-1 cells. Additionally, we show that the siRNA-mediated suppression of both MARCKS and WLS enhanced oxaliplatin sensitivity in oxaliplatin-resistant PANC-1 cells. Taken together, our results provide insights into multiple mechanisms of oxaliplatin resistance in pancreatic cancer cells and reveal that MARCKS and WLS might be involved in the oxaliplatin resistance.
Collapse
Affiliation(s)
- Young Eun Kim
- Center for Bioanalysis, Division of Chemical and Medical Metrology, Korea Research Institute of Standards and Science, Daejeon 34113, Korea;
- School of Earth Sciences and Environmental Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Korea;
| | - Eun-Kyung Kim
- Department of Biochemistry, College of Medicine, Gachon University, Incheon 21999, Korea; (E.-K.K.); (M.-J.S.)
| | - Min-Jeong Song
- Department of Biochemistry, College of Medicine, Gachon University, Incheon 21999, Korea; (E.-K.K.); (M.-J.S.)
| | - Tae-Young Kim
- School of Earth Sciences and Environmental Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Korea;
| | - Ho Hee Jang
- Department of Biochemistry, College of Medicine, Gachon University, Incheon 21999, Korea; (E.-K.K.); (M.-J.S.)
- Correspondence: (H.H.J.); (D.K.)
| | - Dukjin Kang
- Center for Bioanalysis, Division of Chemical and Medical Metrology, Korea Research Institute of Standards and Science, Daejeon 34113, Korea;
- Correspondence: (H.H.J.); (D.K.)
| |
Collapse
|
11
|
Sharma N, Bhushan A, He J, Kaushal G, Bhardwaj V. Metabolic plasticity imparts erlotinib-resistance in pancreatic cancer by upregulating glucose-6-phosphate dehydrogenase. Cancer Metab 2020; 8:19. [PMID: 32974013 PMCID: PMC7507640 DOI: 10.1186/s40170-020-00226-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 09/06/2020] [Indexed: 12/12/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most malignant forms of cancer. Lack of effective treatment options and drug resistance contributes to the low survival among PDAC patients. In this study, we investigated the metabolic alterations in pancreatic cancer cells that do not respond to the EGFR inhibitor erlotinib. We selected erlotinib-resistant pancreatic cancer cells from MiaPaCa2 and AsPC1 cell lines. Metabolic profiling of erlotinib-resistant cells revealed a significant downregulation of glycolytic activity and reduced level of glycolytic metabolites compared to the sensitive cells. The resistant cells displayed elevated expression of the pentose phosphate pathway (PPP) enzymes involved in ROS regulation and nucleotide biosynthesis. The enhanced PPP elevated cellular NADPH/NADP+ ratio and protected the cells from reactive oxygen species (ROS)-induced damage. Inhibition of PPP using 6-aminonicotinamide (6AN) elevated ROS levels, induced G1 cell cycle arrest, and sensitized resistant cells to erlotinib. Genetic studies identified elevated PPP enzyme glucose-6-phosphate dehydrogenase (G6PD) as an important contributor to erlotinib resistance. Mechanistically, our data showed that upregulation of inhibitor of differentiation (ID1) regulates G6PD expression in resistant cells thus contributing to altered metabolic phenotype and reduced response to erlotinib. Together, our results highlight an underlying role of tumor metabolism in PDAC drug response and identify G6PD as a target to overcome drug resistance.
Collapse
Affiliation(s)
- Neha Sharma
- Department of Pharmaceutical Sciences, Jefferson College of Pharmacy, Thomas Jefferson University, Philadelphia, PA USA
| | - Alok Bhushan
- Department of Pharmaceutical Sciences, Jefferson College of Pharmacy, Thomas Jefferson University, Philadelphia, PA USA
| | - Jun He
- Department of Pathology, Anatomy & Cell Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, USA
| | - Gagan Kaushal
- Department of Pharmaceutical Sciences, Jefferson College of Pharmacy, Thomas Jefferson University, Philadelphia, PA USA
| | - Vikas Bhardwaj
- Department of Pharmaceutical Sciences, Jefferson College of Pharmacy, Thomas Jefferson University, Philadelphia, PA USA
| |
Collapse
|
12
|
Gillson J, Ramaswamy Y, Singh G, Gorfe AA, Pavlakis N, Samra J, Mittal A, Sahni S. Small Molecule KRAS Inhibitors: The Future for Targeted Pancreatic Cancer Therapy? Cancers (Basel) 2020; 12:cancers12051341. [PMID: 32456277 PMCID: PMC7281596 DOI: 10.3390/cancers12051341] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 05/21/2020] [Accepted: 05/21/2020] [Indexed: 02/06/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest solid tumors in the world. Currently, there are no approved targeted therapies for PDAC. Mutations in Kirsten rat sarcoma viral oncogene homologue (KRAS) are known to be a major driver of PDAC progression, but it was considered an undruggable target until recently. Moreover, PDAC also suffers from drug delivery issues due to the highly fibrotic tumor microenvironment. In this perspective, we provide an overview of recent developments in targeting mutant KRAS and strategies to overcome drug delivery issues (e.g., nanoparticle delivery). Overall, we propose that the antitumor effects from novel KRAS inhibitors along with strategies to overcome drug delivery issues could be a new therapeutic way forward in PDAC.
Collapse
Affiliation(s)
- Josef Gillson
- Northern Clinical School, Faculty of Medicine and Health, University of Sydney, St Leonards 2065, NSW, Australia; (J.G.); (N.P.); (J.S.); (A.M.)
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute of Medical Research, University of Sydney, St Leonards 2065, NSW, Australia
- Australian Pancreatic Centre, St Leonards 2065, NSW, Australia
| | - Yogambha Ramaswamy
- School of Biomedical Engineering, Faculty of Engineering, The University of Sydney 2006, Sydney, Australia; (Y.R.); (G.S.)
| | - Gurvinder Singh
- School of Biomedical Engineering, Faculty of Engineering, The University of Sydney 2006, Sydney, Australia; (Y.R.); (G.S.)
| | - Alemayehu A. Gorfe
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center Houston, Houston, TX 77030, USA;
| | - Nick Pavlakis
- Northern Clinical School, Faculty of Medicine and Health, University of Sydney, St Leonards 2065, NSW, Australia; (J.G.); (N.P.); (J.S.); (A.M.)
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute of Medical Research, University of Sydney, St Leonards 2065, NSW, Australia
- Northern Sydney Cancer Center, Royal North Shore Hospital, St Leonards 2065, NSW, Australia
- Genesis Care, St Leonards and Frenchs Forest 2065, NSW, Australia
| | - Jaswinder Samra
- Northern Clinical School, Faculty of Medicine and Health, University of Sydney, St Leonards 2065, NSW, Australia; (J.G.); (N.P.); (J.S.); (A.M.)
- Australian Pancreatic Centre, St Leonards 2065, NSW, Australia
- Upper GI Surgical Unit, Royal North Shore Hospital and North Shore Private Hospital, St Leonards 2065, NSW, Australia
| | - Anubhav Mittal
- Northern Clinical School, Faculty of Medicine and Health, University of Sydney, St Leonards 2065, NSW, Australia; (J.G.); (N.P.); (J.S.); (A.M.)
- Australian Pancreatic Centre, St Leonards 2065, NSW, Australia
- Upper GI Surgical Unit, Royal North Shore Hospital and North Shore Private Hospital, St Leonards 2065, NSW, Australia
| | - Sumit Sahni
- Northern Clinical School, Faculty of Medicine and Health, University of Sydney, St Leonards 2065, NSW, Australia; (J.G.); (N.P.); (J.S.); (A.M.)
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute of Medical Research, University of Sydney, St Leonards 2065, NSW, Australia
- Australian Pancreatic Centre, St Leonards 2065, NSW, Australia
- Correspondence: ; Tel.: +61-2-9926-7829
| |
Collapse
|
13
|
Azmi AS, Khan HY, Muqbil I, Aboukameel A, Neggers JE, Daelemans D, Mahipal A, Dyson G, Kamgar M, Al-Hallak MN, Tesfaye A, Kim S, Shidham V, M Mohammad R, Philip PA. Preclinical Assessment with Clinical Validation of Selinexor with Gemcitabine and Nab-Paclitaxel for the Treatment of Pancreatic Ductal Adenocarcinoma. Clin Cancer Res 2020; 26:1338-1348. [PMID: 31831564 PMCID: PMC7073299 DOI: 10.1158/1078-0432.ccr-19-1728] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 10/11/2019] [Accepted: 12/06/2019] [Indexed: 01/23/2023]
Abstract
PURPOSE Pancreatic ductal adenocarcinoma (PDAC) remains a deadly disease urgently requiring new treatments. Overexpression of the protein transporter exportin-1 (XPO1) leads to mislocalization of tumor-suppressor proteins (TSP) and their inactivation. Earlier, we showed that blocking XPO1 by CRISPR/Cas9 validated Selective Inhibitor of Nuclear Export (SINE) compounds (selinexor and analogs) restores the antitumor activity of multiple TSPs leading to suppression of PDAC in vitro and in orthotopic models. EXPERIMENTAL DESIGN We evaluate the synergy between SINE compounds and standard-of-care treatments in preclinical models and in a PDAC Phase Ib trial. RESULTS SINE compounds synergize with gemcitabine (GEM) and nanoparticle albumin-bound (nab)-paclitaxel leading to suppression of PDAC cellular growth and cancer stem cell (CSC) spheroids disintegration. Label-free quantitative proteome profiling with nuclear and cytoplasmic enrichment showed superior enhancement in nuclear protein fraction in combination treatment. Selinexor inhibited the growth of PDAC CSC and two patient-derived (PDX) subcutaneous xenografts. Selinexor-GEM-nab-paclitaxel blocked PDX and orthotopic tumor growth. In a phase 1b study (NCT02178436), 9 patients were exposed to selinexor (60 mg oral) with GEM (1,000 mg/m2 i.v.) and nab-paclitaxel (125 mg/m2 i.v.) on days 1, 8, and 15 of 28-day cycle. Two patients showed partial response, and 2 had stable disease. An outstanding, durable objective response was observed in one of the responders with progression-free survival of 16 months and overall survival of 22 months. CONCLUSIONS Our preclinical and ongoing clinical study lends support to the use of selinexor-GEM-nab-paclitaxel as an effective therapy for metastatic PDAC.
Collapse
Affiliation(s)
- Asfar S Azmi
- Department of Oncology, Wayne State University School of Medicine, Detroit, Michigan.
| | - Husain Yar Khan
- Department of Oncology, Wayne State University School of Medicine, Detroit, Michigan
| | - Irfana Muqbil
- Department of Chemistry, University of Detroit Mercy, Detroit, Michigan
| | - Amro Aboukameel
- Department of Oncology, Wayne State University School of Medicine, Detroit, Michigan
| | - Jasper E Neggers
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
- KU Leuven Department of Microbiology, Immunology and Transplantation, Laboratory of Virology and Chemotherapy, Rega Institute, KU Leuven, Leuven, Belgium
| | - Dirk Daelemans
- KU Leuven Department of Microbiology, Immunology and Transplantation, Laboratory of Virology and Chemotherapy, Rega Institute, KU Leuven, Leuven, Belgium
| | | | - Gregory Dyson
- Department of Oncology, Wayne State University School of Medicine, Detroit, Michigan
| | | | | | - Anteneh Tesfaye
- Department of Oncology, Wayne State University School of Medicine, Detroit, Michigan
| | - Steve Kim
- Department of Oncology, Wayne State University School of Medicine, Detroit, Michigan
| | - Vinod Shidham
- Department of Oncology, Wayne State University School of Medicine, Detroit, Michigan
| | - Ramzi M Mohammad
- Department of Oncology, Wayne State University School of Medicine, Detroit, Michigan
| | - Philip A Philip
- Department of Oncology, Wayne State University School of Medicine, Detroit, Michigan.
| |
Collapse
|
14
|
Yang D, Zhang Q, Ma Y, Che Z, Zhang W, Wu M, Wu L, Liu F, Chu Y, Xu W, McGrath M, Song C, Liu J. Augmenting the therapeutic efficacy of adenosine against pancreatic cancer by switching the Akt/p21-dependent senescence to apoptosis. EBioMedicine 2019; 47:114-127. [PMID: 31495718 PMCID: PMC6796568 DOI: 10.1016/j.ebiom.2019.08.068] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 08/20/2019] [Accepted: 08/28/2019] [Indexed: 01/02/2023] Open
Abstract
Background There are many reports of the anti-tumour effects of exogenous adenosine in gastrointestinal tumours. Gemcitabine, a first line agent for patients with poor performance status, and adenosine have structural similarities. For these reasons, it is worth exploring the therapeutic efficacy of adenosine and its underlying mechanism in pancreatic cancer. Methods Tumour volumes and survival periods were measured in a patient-derived xenograft (PDX) model of pancreatic cancer. The Akt-p21 signalling axis was blocked by p21 silencing or by the Akt inhibitor GSK690693. The combined effect of GSK690693 and adenosine was calculated by the Chou-Talalay equation and verified by measuring fluorescent areas in orthotopic models. Findings Among the PDX mice, the tumour volume in the adenosine treatment group was only 61% of that in the saline treatment group. Adenosine treatment in combination with the Akt inhibitor, GSK690693, or the silencing of p21 to interfere with the Akt-p21 axis can switch the senescence-to-apoptosis signal and alleviate drug resistance. A GSK690693-adenosine combination caused 37.4% further reduction of tumour fluorescent areas in orthotopic models compared with that observed in adenosine monotherapy. Interpretation: Our data confirmed the therapeutic effect of adenosine on pancreatic cancer, and revealed the potential of Akt inhibitors as sensitization agents in this treatment. Fund The work is supported by grants from the National Natural Science Foundation of China to Dongqin Yang (81572336, 81770579) and Jie Liu (81630016, 81830080), and jointly by the Development Fund for Shanghai Talents (201660).
Collapse
Affiliation(s)
- Dongqin Yang
- Department of Digestive Diseases of Huashan Hospital, Fudan University, Shanghai, China.
| | - Qi Zhang
- Department of Digestive Diseases of Huashan Hospital, Fudan University, Shanghai, China
| | - Yunfang Ma
- Department of Digestive Diseases of Huashan Hospital, Fudan University, Shanghai, China
| | - Zhihui Che
- Department of Digestive Diseases of Huashan Hospital, Fudan University, Shanghai, China
| | - Wenli Zhang
- Department of Digestive Diseases of Huashan Hospital, Fudan University, Shanghai, China
| | - Mengmeng Wu
- Department of Digestive Diseases of Huashan Hospital, Fudan University, Shanghai, China
| | - Lijun Wu
- Department of Library, Fudan University, Shanghai, China
| | - Fuchen Liu
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Yiwei Chu
- Department of Immunology of School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Wei Xu
- Department of Immunology of School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Mary McGrath
- Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Chunhua Song
- Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Jie Liu
- Department of Digestive Diseases of Huashan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
15
|
Yu Y, Ding F, Gao M, Jia YF, Ren L. Establishment and characterization of the gemcitabine-resistant human pancreatic cancer cell line SW1990/gemcitabine. Oncol Lett 2019; 18:3065-3071. [PMID: 31452783 PMCID: PMC6676397 DOI: 10.3892/ol.2019.10627] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2018] [Accepted: 04/08/2019] [Indexed: 02/06/2023] Open
Abstract
Due to its rapid progression, metastasis and resistance to chemotherapy, pancreatic cancer is one of the most malignant tumor types to affect the digestive system. Gemcitabine chemotherapy is typically the first choice of treatment for advanced pancreatic cancer; however, chemoresistance is a major obstacle to successful treatment. In order to elucidate the underlying mechanisms of gemcitabine resistance in pancreatic cancer, the drug-resistant cell line SW1990-gemcitabine (SW1990-GZ) was established using the human pancreatic cancer cell line SW1990. The IC50, resistance index and growth of SW1990 and SW1990-GZ cells were also assessed using 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide (MTT) assays. The cellular uptake of gemcitabine in SW1990 and SW1990-GZ was measured using high performance liquid chromatography (HPLC). The protein expression of p53 was also assessed by western blot analysis. The results demonstrated that the IC50 of SW1990 and SW1990-Gz was 0.07±0.0021 and 87.5±3.24 µg/ml, respectively, and that the resistance index ratio of SW1990-Gz was 1,250. The growth rate of SW1990-GZ cells was low compared with that of SW1990 cells. The HPLC results indicated that gemcitabine uptake was markedly reduced in SW1990-GZ cells compared with in SW1990 cells at different time points. The protein expression of p53 was significantly higher in GEM-resistant SW1990-GZ cells compared with that in SW1990 cells (P<0.01). These results suggest that a human gemcitabine-resistant pancreatic cancer cell line was successfully established, with stable and significant drug resistance. The results of the present study suggest that the decreased cellular uptake of gemcitabine may serve an important role in gemcitabine chemoresistance in SW1990-GZ cells; thus, this cell line may be used as an effective in vitro model to improve our understanding of gemcitabine-resistance in pancreatic cancer.
Collapse
Affiliation(s)
- Yue Yu
- Department of Gastroenterology, Affiliated Provincial Hospital, Anhui Medical University, Hefei, Anhui 230001, P.R. China
| | - Fei Ding
- Department of Gastroenterology, Affiliated Provincial Hospital, Anhui Medical University, Hefei, Anhui 230001, P.R. China.,Department of Gastroenterology, Anqing First People's Hospital, Anqing, Anhui 246000, P.R. China
| | - Meng Gao
- Department of Gastroenterology, Affiliated Provincial Hospital, Anhui Medical University, Hefei, Anhui 230001, P.R. China
| | - Yi Fu Jia
- Department of Gastroenterology, Affiliated Provincial Hospital, Anhui Medical University, Hefei, Anhui 230001, P.R. China
| | - Le Ren
- Department of Gastroenterology, Affiliated Provincial Hospital, Anhui Medical University, Hefei, Anhui 230001, P.R. China
| |
Collapse
|
16
|
Fang Y, Zhou W, Rong Y, Kuang T, Xu X, Wu W, Wang D, Lou W. Exosomal miRNA-106b from cancer-associated fibroblast promotes gemcitabine resistance in pancreatic cancer. Exp Cell Res 2019; 383:111543. [PMID: 31374207 DOI: 10.1016/j.yexcr.2019.111543] [Citation(s) in RCA: 159] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 07/22/2019] [Accepted: 07/29/2019] [Indexed: 12/25/2022]
Abstract
Gemcitabine (GEM)-based chemotherapy is commonly used to treat pancreatic cancer. However, acquired resistance to GEM remains a challenge in pancreatic cancer patients. Here we tested whether cancer-associated fibroblasts (CAFs) play vital roles in regulating drug resistance by transferring exosomal miRNA to cancer cells. CAFs were isolated from primary fibroblast of pancreatic cancer patients, and exosomes were collected and identified through transmission electron microscopy and western blotting analysis. The functions of CAFs-derived exosomal miRNA in regulating drug resistance were further investigated. We found that CAFs were innately resistant to GEM. The conditioned medium (CM) and the exosomes derived from CAFs contributed to GEM resistance, and GEM treatment further enhanced the effect of CAFs or CAFs-exosomes on pancreatic cancer cells proliferation. MiR-106b level was upregulated in CAFs and CAFs-exosomes following GEM treatment. MiR-106b was directly transferred from CAFs to pancreatic cancer cells through exosomes. Pretreatment of CAFs with miR-106b inhibitor suppressed miR-106b expression in CAFs-exosomes and resulted in a decreased resistance of cancer cells to GEM. MiR-106b promoted GEM resistance of cancer cells by directly targeting TP53INP1. Summarily, our data demonstrated that CAFs-derived exosomal miR-106b plays a vital role in causing GEM resistance of pancreatic cancer, thus offering a new target for sensitizing pancreatic cancer cells to GEM.
Collapse
Affiliation(s)
- Yuan Fang
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Wentao Zhou
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Yefei Rong
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Tiantao Kuang
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Xuefeng Xu
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Wenchuan Wu
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Dansong Wang
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| | - Wenhui Lou
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
17
|
Zhang S, Yang Y, Weng W, Guo B, Cai G, Ma Y, Cai S. Fusobacterium nucleatum promotes chemoresistance to 5-fluorouracil by upregulation of BIRC3 expression in colorectal cancer. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:14. [PMID: 30630498 PMCID: PMC6327560 DOI: 10.1186/s13046-018-0985-y] [Citation(s) in RCA: 195] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 11/29/2018] [Indexed: 12/16/2022]
Abstract
Background Emerging evidence suggests a potential relationship between gut microbiota and the host response to chemotherapeutic drugs including 5-fluorouracil (5-Fu). Fusobacterium nucleatum (Fn) has been linked to the initiation and progression of colorectal cancer (CRC). Unfortunately, little was known about the relationship between Fn infection and chemotherapeutic efficacy. Here, we investigate the potential relationship between Fn infection and chemotherapeutic efficacy of 5-Fu in CRC. Methods Differentially expressed genes of CRC cell lines induced by Fn infection were analyzed based on a whole genome microarray analysis Then, we explored the relationship between upregulation of BIRC3 induced by Fn infection and chemoresistance to 5-Fu in vitro and in vivo. Furthermore, we dissected the mechanisms involved in Fn-induced BIRC3 expression. Finally, we investigated the clinical relevance of Fn infection, BIRC3 protein expression and chemoresistance to 5-Fu treatment in CRC patients. Results BIRC3 was the most upregulated gene induced by Fn infection via the TLR4/NF-κB pathway in CRC cells; Fn infection reduced the chemosensitivity of CRC cells to 5-Fu through upregulation of BIRC3 in vitro and in vivo. High Fn abundance correlated with chemoresistance in advanced CRC patients who received standard 5-Fu-based adjuvant chemotherapy after radical surgery. Conclusions Our evidence suggests that Fn and BIRC3 may serve as promising therapeutic targets for reducing chemoresistance to 5-Fu treatment in advanced CRC. Electronic supplementary material The online version of this article (10.1186/s13046-018-0985-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sheng Zhang
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, 270 Dong'an Road, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yongzhi Yang
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, 270 Dong'an Road, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Wenhao Weng
- Department of Clinical Laboratory, Yangpu Hospital, Tongji University School of Medicine, Shanghai, China.,Center for Translational Medicine, Yangpu Hospital, Tongji University School of Medicine, Shanghai, China
| | - Bomin Guo
- Department of Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Guoxiang Cai
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, 270 Dong'an Road, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yanlei Ma
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, 270 Dong'an Road, Shanghai, 200032, China. .,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| | - Sanjun Cai
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, 270 Dong'an Road, Shanghai, 200032, China. .,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
18
|
Harbuzariu A, Gonzalez-Perez RR. Leptin-Notch axis impairs 5-fluorouracil effects on pancreatic cancer. Oncotarget 2018; 9:18239-18253. [PMID: 29719602 PMCID: PMC5915069 DOI: 10.18632/oncotarget.24435] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 01/30/2018] [Indexed: 12/15/2022] Open
Abstract
5-FU chemotherapy is a current strategy to treat pancreatic cancer (PC), but unfortunately chemoresistance is eventually developed in most patients. Obesity is a risk factor for PC that could affect 5-FU effectiveness through the adipokine leptin, which is a known proliferation, survival factor and Notch inducer. We investigated whether leptin signaling affects 5-FU cytotoxicity on PC. To this end, tumorspheres developed from BxPC-3 and MiaPaCa-2 PC cells were treated with 5-FU, leptin, inhibitors for Notch (DAPT) and leptin signaling (IONP-LPrA2) and ATP-binding cassette of proteins (Probenecid). Leptin treatment decreased 5-FU cytotoxicity, and increased cell proliferation, colony forming ability, stem cell, pluripotency, EMT markers, drug efflux proteins (ABCC5, ABCC11) and Notch. In addition, leptin reduced the 5-FU effects on apoptosis by decreasing pro-apoptotic (Bax, Caspase-3 activation and PARP degradation) and increasing anti-apoptotic factors (RIP and Bcl-XL). Leptin's effects on PC tumorspheres treated with 5-FU were reduced by IONP-LPrA2 and were mainly Notch signaling- dependent and more evident in MiaPaCa-2-derived tumorspheres. Present results suggest that leptin could impair 5-FU cytotoxicity and promote chemoresistance. Therefore, targeting the leptin-Notch axis could be a novel way to improve 5-FU therapy for PC patients, especially in obesity context.
Collapse
Affiliation(s)
- Adriana Harbuzariu
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Ruben Rene Gonzalez-Perez
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA 30310, USA
| |
Collapse
|
19
|
Sun L, Hu L, Cogdell D, Lu L, Gao C, Tian W, Zhang Z, Kang Y, Fleming JB, Zhang W. MIR506 induces autophagy-related cell death in pancreatic cancer cells by targeting the STAT3 pathway. Autophagy 2017; 13:703-714. [PMID: 28121485 DOI: 10.1080/15548627.2017.1280217] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the most aggressive and lethal cancer. The role of autophagy in the pathobiology of PDAC is intricate, with opposing functions manifested in different cellular contexts. MIR506 functions as a tumor suppressor in many cancer types through the regulation of multiple pathways. In this study, we hypothesized that MIR506 exerted a tumor suppression function in PDAC by inducing autophagy-related cell death. Our results provided evidence that downregulation of MIR506 expression was associated with disease progression in human PDAC. MIR506 triggered autophagic flux in PDAC cells, which led to autophagy-related cell death through direct targeting of the STAT3 (signal transducer and activator of transcription 3)-BCL2-BECN1 axis. Silencing and inhibiting STAT3 recapitulated the effects of MIR506, whereas forced expression of STAT3 abrogated the effects of MIR506. We propose that the apoptosis-inhibitory protein BCL2, which also inhibits induction of autophagy by blocking BECN1, was inhibited by MIR506 through targeting STAT3, thus augmenting BECN1 and promoting autophagy-related cell death. Silencing BECN1 and overexpression of BCL2 abrogated the effects of MIR506. These findings expand the known mechanisms of MIR506-mediated tumor suppression to activation of autophagy-related cell death and suggest a strategy for using MIR506 as an anti-STAT3 approach to PDAC treatment.
Collapse
Affiliation(s)
- Longhao Sun
- a Department of Cancer Biology , Comprehensive Cancer Center of Wake Forest Baptist Medical Center , Winston-Salem , NC , USA.,b Department of Pathology , The University of Texas MD Anderson Cancer Center , Houston , TX , USA.,c Department of General Surgery , Tianjin Medical University General Hospital , Tianjin , China
| | - Limei Hu
- b Department of Pathology , The University of Texas MD Anderson Cancer Center , Houston , TX , USA
| | - David Cogdell
- b Department of Pathology , The University of Texas MD Anderson Cancer Center , Houston , TX , USA
| | - Li Lu
- c Department of General Surgery , Tianjin Medical University General Hospital , Tianjin , China
| | - Chao Gao
- a Department of Cancer Biology , Comprehensive Cancer Center of Wake Forest Baptist Medical Center , Winston-Salem , NC , USA.,b Department of Pathology , The University of Texas MD Anderson Cancer Center , Houston , TX , USA
| | - Weijun Tian
- c Department of General Surgery , Tianjin Medical University General Hospital , Tianjin , China
| | - Zhixiang Zhang
- c Department of General Surgery , Tianjin Medical University General Hospital , Tianjin , China
| | - Ya'an Kang
- d Department of Surgical Oncology , The University of Texas MD Anderson Cancer Center , Houston , TX , USA
| | - Jason B Fleming
- d Department of Surgical Oncology , The University of Texas MD Anderson Cancer Center , Houston , TX , USA
| | - Wei Zhang
- a Department of Cancer Biology , Comprehensive Cancer Center of Wake Forest Baptist Medical Center , Winston-Salem , NC , USA.,b Department of Pathology , The University of Texas MD Anderson Cancer Center , Houston , TX , USA
| |
Collapse
|
20
|
Hoang NTH, Kadonosono T, Kuchimaru T, Kizaka-Kondoh S. Hypoxia-inducible factor-targeting prodrug TOP3 combined with gemcitabine or TS-1 improves pancreatic cancer survival in an orthotopic model. Cancer Sci 2016; 107:1151-8. [PMID: 27270607 PMCID: PMC4982586 DOI: 10.1111/cas.12982] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 05/31/2016] [Accepted: 06/02/2016] [Indexed: 01/25/2023] Open
Abstract
Pancreatic cancer is one of the most lethal digestive system cancers with a 5‐year survival rate of 4–7%. Despite extensive efforts, recent chemotherapeutic regimens have provided only limited benefits to pancreatic cancer patients. Gemcitabine and TS‐1, the current standard‐of‐care chemotherapeutic drugs for treatment of this severe cancer, have a low response rate. Hypoxia is one of the factors contributing to treatment resistance. Specifically, overexpression of hypoxia‐inducible factor, a master transcriptional regulator of cell adaption to hypoxia, is strongly correlated with poor prognosis in many human cancers. TAT‐ODD‐procaspase‐3 (TOP3) is a protein prodrug that is specifically processed and activated in hypoxia‐inducible factor‐active cells in cancers, leading to cell death. Here, we report combination therapies in which TOP3 was combined with gemcitabine or TS‐1. As monotherapy, gemcitabine and TS‐1 showed a limited effect on hypoxic and starved pancreatic cancer cells, whereas co‐treatment with TOP3 successfully overcame this limitation in vitro. Furthermore, combination therapies of TOP3 with these drugs resulted in a significant improvement in survival of orthotopic pancreatic cancer models involving the human pancreatic cancer cell line SUIT‐2. Overall, our study indicates that the combination of TOP3 with current chemotherapeutic drugs can significantly improve treatment outcome, offering a promising new therapeutic option for patients with pancreatic cancer.
Collapse
Affiliation(s)
- Ngoc Thi Hong Hoang
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Japan
| | - Tetsuya Kadonosono
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Japan
| | - Takahiro Kuchimaru
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Japan
| | - Shinae Kizaka-Kondoh
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Japan
| |
Collapse
|
21
|
Mohammad RM, Muqbil I, Lowe L, Yedjou C, Hsu HY, Lin LT, Siegelin MD, Fimognari C, Kumar NB, Dou QP, Yang H, Samadi AK, Russo GL, Spagnuolo C, Ray SK, Chakrabarti M, Morre JD, Coley HM, Honoki K, Fujii H, Georgakilas AG, Amedei A, Niccolai E, Amin A, Ashraf SS, Helferich WG, Yang X, Boosani CS, Guha G, Bhakta D, Ciriolo MR, Aquilano K, Chen S, Mohammed SI, Keith WN, Bilsland A, Halicka D, Nowsheen S, Azmi AS. Broad targeting of resistance to apoptosis in cancer. Semin Cancer Biol 2015; 35 Suppl:S78-S103. [PMID: 25936818 PMCID: PMC4720504 DOI: 10.1016/j.semcancer.2015.03.001] [Citation(s) in RCA: 596] [Impact Index Per Article: 59.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Revised: 03/04/2015] [Accepted: 03/04/2015] [Indexed: 12/15/2022]
Abstract
Apoptosis or programmed cell death is natural way of removing aged cells from the body. Most of the anti-cancer therapies trigger apoptosis induction and related cell death networks to eliminate malignant cells. However, in cancer, de-regulated apoptotic signaling, particularly the activation of an anti-apoptotic systems, allows cancer cells to escape this program leading to uncontrolled proliferation resulting in tumor survival, therapeutic resistance and recurrence of cancer. This resistance is a complicated phenomenon that emanates from the interactions of various molecules and signaling pathways. In this comprehensive review we discuss the various factors contributing to apoptosis resistance in cancers. The key resistance targets that are discussed include (1) Bcl-2 and Mcl-1 proteins; (2) autophagy processes; (3) necrosis and necroptosis; (4) heat shock protein signaling; (5) the proteasome pathway; (6) epigenetic mechanisms; and (7) aberrant nuclear export signaling. The shortcomings of current therapeutic modalities are highlighted and a broad spectrum strategy using approaches including (a) gossypol; (b) epigallocatechin-3-gallate; (c) UMI-77 (d) triptolide and (e) selinexor that can be used to overcome cell death resistance is presented. This review provides a roadmap for the design of successful anti-cancer strategies that overcome resistance to apoptosis for better therapeutic outcome in patients with cancer.
Collapse
Affiliation(s)
- Ramzi M Mohammad
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, United States; Interim translational Research Institute, Hamad Medical Corporation, Doha, Qatar.
| | - Irfana Muqbil
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, United States
| | - Leroy Lowe
- Getting to Know Cancer, Truro, Nova Scotia, Canada
| | - Clement Yedjou
- C-SET, [Jackson, #229] State University, Jackson, MS, United States
| | - Hsue-Yin Hsu
- Department of Life Sciences, Tzu-Chi University, Hualien, Taiwan
| | - Liang-Tzung Lin
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Markus David Siegelin
- Department of Pathology and Cell Biology, Columbia University, New York City, NY, United States
| | - Carmela Fimognari
- Dipartimento di Scienze per la Qualità della Vita Alma Mater Studiorum-Università di Bologna, Italy
| | - Nagi B Kumar
- Moffit Cancer Center, University of South Florida College of Medicine, Tampa, FL, United States
| | - Q Ping Dou
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, United States; Departments of Pharmacology and Pathology, Karmanos Cancer Institute, Detroit MI, United States
| | - Huanjie Yang
- The School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang, China
| | | | - Gian Luigi Russo
- Institute of Food Sciences National Research Council, Avellino, Italy
| | - Carmela Spagnuolo
- Institute of Food Sciences National Research Council, Avellino, Italy
| | - Swapan K Ray
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine, Columbia, SC, United States
| | - Mrinmay Chakrabarti
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine, Columbia, SC, United States
| | - James D Morre
- Mor-NuCo, Inc, Purdue Research Park, West Lafayette, IN, United States
| | - Helen M Coley
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, Surrey, United Kingdom
| | - Kanya Honoki
- Department of Orthopedic Surgery, Nara Medical University, Kashihara, Japan
| | - Hiromasa Fujii
- Department of Orthopedic Surgery, Nara Medical University, Kashihara, Japan
| | - Alexandros G Georgakilas
- Department of Physics, School of Applied Mathematical and Physical Sciences, National Technical University of Athens, Zografou 15780, Athens, Greece
| | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, university of florence, Italy
| | - Elena Niccolai
- Department of Experimental and Clinical Medicine, university of florence, Italy
| | - Amr Amin
- Department of Biology, College of Science, UAE University, United Arab Emirates; Faculty of Science, Cairo University, Egypt
| | - S Salman Ashraf
- Department of Chemistry, College of Science, UAE University, United Arab Emirates
| | - William G Helferich
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Xujuan Yang
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Chandra S Boosani
- Department of BioMedical Sciences, School of Medicine Creighton University, Omaha NE, United States
| | - Gunjan Guha
- School of Chemical and Bio Technology, SASTRA University, Thanjavur, India
| | - Dipita Bhakta
- School of Chemical and Bio Technology, SASTRA University, Thanjavur, India
| | | | - Katia Aquilano
- Department of Biology, University of Rome "Tor Vergata", Italy
| | - Sophie Chen
- Ovarian and Prostate Cancer Research Trust Laboratory, Guildford, Surrey, United Kingdom
| | - Sulma I Mohammed
- Department of Comparative Pathobiology and Purdue University Center for Cancer Research, Purdue, West Lafayette, IN, United States
| | - W Nicol Keith
- Institute of Cancer Sciences, University of Glasgow, Glasgow, Ireland
| | - Alan Bilsland
- Institute of Cancer Sciences, University of Glasgow, Glasgow, Ireland
| | - Dorota Halicka
- Department of Pathology, New York Medical College, Valhalla, NY, United States
| | - Somaira Nowsheen
- Mayo Graduate School, Mayo Medical School, Mayo Clinic Medical Scientist Training Program, Rochester, MN, United States
| | - Asfar S Azmi
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, United States
| |
Collapse
|
22
|
Dhayat SA, Mardin WA, Seggewiß J, Ströse AJ, Matuszcak C, Hummel R, Senninger N, Mees ST, Haier J. MicroRNA Profiling Implies New Markers of Gemcitabine Chemoresistance in Mutant p53 Pancreatic Ductal Adenocarcinoma. PLoS One 2015; 10:e0143755. [PMID: 26606261 PMCID: PMC4659591 DOI: 10.1371/journal.pone.0143755] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Accepted: 11/09/2015] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND No reliable predictors of susceptibility to gemcitabine chemotherapy exist in pancreatic ductal adenocarcinoma (PDAC). MicroRNAs (miR) are epigenetic gene regulators with tumorsuppressive or oncogenic roles in various carcinomas. This study assesses chemoresistant PDAC for its specific miR expression pattern. METHODS Gemcitabine-resistant variants of two mutant p53 human PDAC cell lines were established. Survival rates were analyzed by cytotoxicity and apoptosis assays. Expression of 1733 human miRs was investigated by microarray and validated by qRT-PCR. After in-silico analysis of specific target genes and proteins of dysregulated miRs, expression of MRP-1, Bcl-2, mutant p53, and CDK1 was quantified by Western blot. RESULTS Both established PDAC clones showed a significant resistance to gemcitabine (p<0.02) with low apoptosis rate (p<0.001) vs. parental cells. MiR-screening revealed significantly upregulated (miR-21, miR-99a, miR-100, miR-125b, miR-138, miR-210) and downregulated miRs (miR-31*, miR-330, miR-378) in chemoresistant PDAC (p<0.05). Bioinformatic analysis suggested involvement of these miRs in pathways controlling cell death and cycle. MRP-1 (p<0.02) and Bcl-2 (p<0.003) were significantly overexpressed in both resistant cell clones and mutant p53 (p = 0.023) in one clone. CONCLUSION Consistent miR expression profiles, in part regulated by mutant TP53 gene, were identified in gemcitabine-resistant PDAC with significant MRP-1 and Bcl-2 overexpression. These results provide a basis for further elucidation of chemoresistance mechanisms and therapeutic approaches to overcome chemoresistance in PDAC.
Collapse
Affiliation(s)
- Sameer A. Dhayat
- Department of General and Visceral Surgery, University Hospital Muenster, Muenster, Germany
| | - Wolf Arif Mardin
- Department of General and Visceral Surgery, University Hospital Muenster, Muenster, Germany
| | - Jochen Seggewiß
- Integrated Functional Genomics, Interdisciplinary Center for Clinical Research, Medical Faculty of the University of Muenster, Muenster, Germany
| | - Anda Jana Ströse
- Comprehensive Cancer Center Muenster, University Hospital Muenster, Muenster, Germany
| | - Christiane Matuszcak
- Department of General and Visceral Surgery, University Hospital Muenster, Muenster, Germany
| | - Richard Hummel
- Department of General and Visceral Surgery, University Hospital Muenster, Muenster, Germany
| | - Norbert Senninger
- Department of General and Visceral Surgery, University Hospital Muenster, Muenster, Germany
| | - Sören Torge Mees
- Department of General and Visceral Surgery, University Hospital Muenster, Muenster, Germany
| | - Jörg Haier
- Comprehensive Cancer Center Muenster, University Hospital Muenster, Muenster, Germany
| |
Collapse
|
23
|
Wang WB, Yang Y, Zhao YP, Zhang TP, Liao Q, Shu H. Recent studies of 5-fluorouracil resistance in pancreatic cancer. World J Gastroenterol 2014; 20:15682-15690. [PMID: 25400452 PMCID: PMC4229533 DOI: 10.3748/wjg.v20.i42.15682] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Revised: 05/04/2014] [Accepted: 07/25/2014] [Indexed: 02/06/2023] Open
Abstract
Resistance to 5-fluorouracil (5-FU), an important anticancer drug, is a serious challenge in the treatment of pancreatic cancer. Equilibrative nucleoside transporter 1 and multidrug-resistance protein (MRP) 5 and MRP8, rather than P-glycoprotein, play important roles in 5-FU transport. Thymidylate synthase, dihydropyrimidine dehydrogenase, methylenetetrahydrofolate reductase and thymidine phosphorylase are four key enzymes involved in 5-FU metabolism. Other metabolic enzymes, including uridine monophosphate synthetase, also contribute to chemoresistance. Intracellular signaling pathways are an integrated network, and nuclear factor kappa-light-chain-enhancer of activated B cells, AKT and extracellular signal-regulated kinases are signaling pathways that are particularly relevant to 5-FU resistance. In addition, recent reports indicate that STAT-3 is a crucial survival protein. Proteomic assays provide a powerful tool for identifying target proteins and understanding the role of microRNAs and stromal factors to facilitate the development of strategies to combat 5-FU resistance.
Collapse
|
24
|
Gonzalez CD, Alvarez S, Ropolo A, Rosenzvit C, Gonzalez Bagnes MF, Vaccaro MI. Autophagy, Warburg, and Warburg reverse effects in human cancer. BIOMED RESEARCH INTERNATIONAL 2014; 2014:926729. [PMID: 25197670 PMCID: PMC4145381 DOI: 10.1155/2014/926729] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Accepted: 07/24/2014] [Indexed: 12/15/2022]
Abstract
Autophagy is a highly regulated-cell pathway for degrading long-lived proteins as well as for clearing cytoplasmic organelles. Autophagy is a key contributor to cellular homeostasis and metabolism. Warburg hypothesized that cancer growth is frequently associated with a deviation of a set of energy generation mechanisms to a nonoxidative breakdown of glucose. This cellular phenomenon seems to rely on a respiratory impairment, linked to mitochondrial dysfunction. This mitochondrial dysfunction results in a switch to anaerobic glycolysis. It has been recently suggested that epithelial cancer cells may induce the Warburg effect in neighboring stromal fibroblasts in which autophagy was activated. These series of observations drove to the proposal of a putative reverse Warburg effect of pathophysiological relevance for, at least, some tumor phenotypes. In this review we introduce the autophagy process and its regulation and its selective pathways and role in cancer cell metabolism. We define and describe the Warburg effect and the newly suggested "reverse" hypothesis. We also discuss the potential value of modulating autophagy with several pharmacological agents able to modify the Warburg effect. The association of the Warburg effect in cancer and stromal cells to tumor-related autophagy may be of relevance for further development of experimental therapeutics as well as for cancer prevention.
Collapse
Affiliation(s)
- Claudio D. Gonzalez
- Institute of Biochemistry and Molecular Medicine, National Council for Scientific and Technological Research, School of Pharmacy and Biochemistry, University of Buenos Aires, Junin 956 p5, 1113 Buenos Aires, Argentina
- Department of Pharmacology, CEMIC University Institute, 1113 Buenos Aires, Argentina
| | - Silvia Alvarez
- Institute of Biochemistry and Molecular Medicine, National Council for Scientific and Technological Research, School of Pharmacy and Biochemistry, University of Buenos Aires, Junin 956 p5, 1113 Buenos Aires, Argentina
| | - Alejandro Ropolo
- Institute of Biochemistry and Molecular Medicine, National Council for Scientific and Technological Research, School of Pharmacy and Biochemistry, University of Buenos Aires, Junin 956 p5, 1113 Buenos Aires, Argentina
| | - Carla Rosenzvit
- Department of Pharmacology, CEMIC University Institute, 1113 Buenos Aires, Argentina
| | | | - Maria I. Vaccaro
- Institute of Biochemistry and Molecular Medicine, National Council for Scientific and Technological Research, School of Pharmacy and Biochemistry, University of Buenos Aires, Junin 956 p5, 1113 Buenos Aires, Argentina
| |
Collapse
|
25
|
Hao WH, Yang LC, Wang JJ, Hsu CS, Chang LC, Hsu KY. Facile method for determination of deoxycytidine kinase activity in biological milieus. J Food Drug Anal 2014. [PMCID: PMC9359329 DOI: 10.1016/j.jfda.2013.09.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
A new analytical method for determining deoxycytidine kinase (dCK) activity in biological milieus using luminescence is reported here. This method, based on utilizing adenosine triphosphate (ATP) as the sole phosphate donor in the kinase reaction and monitoring ATP consumption via a luciferase-based chemiluminescence reaction, is capable of detecting dCK activity without the use of specific substrates or radioisotope techniques. Comparing with the reverse-phase high-performance liquid chromatography method, this new method is suggested to be efficient and sensitive. Further, application of the proposed method for profiling dCK activity in cultured cancer cells revealed that a cervix cell line exhibited the highest dCK activity to gemcitabine metabolism.
Collapse
Affiliation(s)
- Wei-Hua Hao
- InnoPharmax Inc., Taipei,
Taiwan, ROC
- School of Pharmacy, Taipei Medical University, Taipei,
Taiwan, ROC
| | | | | | | | - Li-Chien Chang
- School of Pharmacy, National Defense Medical Center, Taipei,
Taiwan, ROC
| | - Kuang-Yang Hsu
- School of Pharmacy, Taipei Medical University, Taipei,
Taiwan, ROC
- Corresponding author. 250 Wu-Hsing Street, Taipei City, Taiwan, ROC. E-mail address: (K.-Y. Hsu)
| |
Collapse
|
26
|
Designing of promiscuous inhibitors against pancreatic cancer cell lines. Sci Rep 2014; 4:4668. [PMID: 24728108 PMCID: PMC3985076 DOI: 10.1038/srep04668] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Accepted: 03/17/2014] [Indexed: 01/02/2023] Open
Abstract
Pancreatic cancer remains the most devastating disease with worst prognosis. There is a pressing need to accelerate the drug discovery process to identify new effective drug candidates against pancreatic cancer. We have developed QSAR models for predicting promiscuous inhibitors using the pharmacological data. Our models achieved maximum Pearson correlation coefficient of 0.86, when evaluated on 10-fold cross-validation. Our models have also successfully validated the drug-to-oncogene relationship and further we used these models to screen FDA approved drugs and tested them in vitro. We have integrated these models in a webserver named as DiPCell, which will be useful for screening and designing novel promiscuous drug molecules. We have also identified the most and least effective drugs for pancreatic cancer cell lines. On the other side, we have identified resistant pancreatic cancer cell lines, which need investigative scanner on them to put light on resistant mechanism in pancreatic cancer.
Collapse
|
27
|
Stark JL, Mehla K, Chaika N, Acton TB, Xiao R, Singh PK, Montelione GT, Powers R. Structure and function of human DnaJ homologue subfamily a member 1 (DNAJA1) and its relationship to pancreatic cancer. Biochemistry 2014; 53:1360-72. [PMID: 24512202 PMCID: PMC3985919 DOI: 10.1021/bi401329a] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Pancreatic cancer has a dismal 5 year survival rate of 5.5% that has not been improved over the past 25 years despite an enormous amount of effort. Thus, there is an urgent need to identify truly novel yet druggable protein targets for drug discovery. The human protein DnaJ homologue subfamily A member 1 (DNAJA1) was previously shown to be downregulated 5-fold in pancreatic cancer cells and has been targeted as a biomarker for pancreatic cancer, but little is known about the specific biological function for DNAJA1 or the other members of the DnaJ family encoded in the human genome. Our results suggest the overexpression of DNAJA1 suppresses the stress response capabilities of the oncogenic transcription factor, c-Jun, and results in the diminution of cell survival. DNAJA1 likely activates a DnaK protein by forming a complex that suppresses the JNK pathway, the hyperphosphorylation of c-Jun, and the anti-apoptosis state found in pancreatic cancer cells. A high-quality nuclear magnetic resonance solution structure of the J-domain of DNAJA1 combined with a bioinformatics analysis and a ligand affinity screen identifies a potential DnaK binding site, which is also predicted to overlap with an inhibitory binding site, suggesting DNAJA1 activity is highly regulated.
Collapse
Affiliation(s)
- Jaime L Stark
- Department of Chemistry, University of Nebraska-Lincoln , Lincoln, Nebraska 68588, United States
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Du X, Zhao YP, Zhang TP, Zhou L, Chen G, Wang TX, You L, Shu H. Alteration of the intrinsic apoptosis pathway is involved in Notch-induced chemoresistance to gemcitabine in pancreatic cancer. Arch Med Res 2013; 45:15-20. [PMID: 24316112 DOI: 10.1016/j.arcmed.2013.10.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2013] [Accepted: 09/27/2013] [Indexed: 01/26/2023]
Abstract
BACKGROUND AND AIMS Chemoresistance is a major challenge in pancreatic cancer (PC) treatment. Limited data have shown that members of the Notch signaling pathway are involved in resistance to gemcitabine (GEM) in PC. However, further evidence is needed and the underlying mechanisms remain to be elucidated. The current study aims to investigate the role of alterations of the intrinsic apoptosis pathway in Notch-induced GEM resistance of PC. METHODS The Notch signaling pathway was inhibited or activated in three PC cell lines (AsPC-1, BxPC-3, and MIA PaCa-2) by γ-secretase inhibition and Notch intracellular domain (NICD) overexpression, respectively. Subsequent analyses included inhibition rates of cell proliferation by GEM, cell apoptosis, and expression of proteins involved in the intrinsic apoptosis pathway. RESULTS Hes-1 expression was significantly elevated after GEM treatment, indicating Notch activation. Inhibition of the Notch signaling pathway by DAPT, a γ-secretase inhibitor, resulted in a significant increase of the inhibition rates by GEM in all PC cell lines. In addition, there was more frequent apoptosis, higher caspase-3 activity, up-regulation of Bax, and down-regulation of Bcl-2 and Bcl-xL. Conversely, transient transfection of NICD, which enhances the activity of the Notch signaling, caused a remarkable decrease of the chemosensitivity to GEM. CONCLUSIONS An alteration of the intrinsic apoptosis pathway is involved in Notch-induced chemoresistance to GEM in PC cells.
Collapse
Affiliation(s)
- Xiao Du
- Department of General Surgery, Peking Union Medical College Hospital, Beijing, China
| | - Yu-Pei Zhao
- Department of General Surgery, Peking Union Medical College Hospital, Beijing, China; National Laboratory of Medical Molecular Biology, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, China.
| | - Tai-Ping Zhang
- Department of General Surgery, Peking Union Medical College Hospital, Beijing, China
| | - Li Zhou
- Department of General Surgery, Peking Union Medical College Hospital, Beijing, China
| | - Ge Chen
- Department of General Surgery, Peking Union Medical College Hospital, Beijing, China
| | - Tian-Xiao Wang
- Department of General Surgery, Peking Union Medical College Hospital, Beijing, China
| | - Lei You
- Department of General Surgery, Peking Union Medical College Hospital, Beijing, China
| | - Hong Shu
- Department of General Surgery, Peking Union Medical College Hospital, Beijing, China
| |
Collapse
|
29
|
Induction of pancreatic cancer cell apoptosis, invasion, migration, and enhancement of chemotherapy sensitivity of gemcitabine, 5-FU, and oxaliplatin by hnRNP A2/B1 siRNA. Anticancer Drugs 2013; 24:566-76. [PMID: 23525071 DOI: 10.1097/cad.0b013e3283608bc5] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
We investigated the effects of inhibiting heterogeneous nuclear ribonucleoprotein A2/B1 (hnRNP A2/B1) expression on apoptosis, invasion, migration, and the chemotherapy sensitivity of pancreatic cancer cells to gemcitabine, 5-FU, and oxaliplatin chemotherapy using small interfering RNA (siRNA). Chemically synthesized siRNA hnRNP A2/B1 was transfected into the human pancreatic cancer cell lines SW1990 and BxPC-3. The IC(50) of gemcitabine, 5-FU, and oxaliplatin was determined using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) assay. Cell apoptosis and cycle were detected using flow cytometry. The expressions of apoptosis-related genes, p53, Bax, Bcl-2, TRAIL, Survivin, multidrug resistance 1 (MDR1), E-cadherin, and matrix metalloproteinases-2 (MMP-2) were detected using real-time PCR and western blot. Plate colony formation assay, wound scratch assay, invasion, and migration were also examined. Gemcitabine, 5-FU, and oxaliplatin inhibit the proliferation of SW1990 and BxPC-3 cells in a concentration-dependent manner. Inhibition of hnRNP A2/B1 expression significantly reduced the IC(50) of gemcitabine, 5-FU, and oxaliplatin (P<0.01). hnRNP A2/B1 siRNA combined with gemcitabine, 5-FU and oxaliplatin significantly increased (P<0.01) apoptosis of pancreatic cancer cell lines SW1990 and BxPC-3, increased the expression level of Bax mRNA, decreased Bcl-2 mRNA and MDR1 mRNA expression (P<0.01), and induced no change in p53, TRAIL, and Survivin mRNA expression in SW1990. In the western blot analysis, the expression level of Bax protein increased (P<0.01); the expression of both P-glycoprotein (Pg-p) protein and Bcl-2 protein decreased (P<0.01). Silencing hnRNP A2/B1 decreased invasion and migration in the cell line SW1990. Silencing hnRNP A2/B1 in SW1990 also correlated with an increase in E-cadherin expression and a decrease in MMP-2 expression at the same time. Inhibition of hnRNP A2/B1 expression can induce apoptosis in pancreatic cancer cells and improve chemosensitivity to gemcitabine, 5-FU, and oxaliplatin. hnRNP A2/B1 may play a role in invasion and migration in the pancreatic cancer cell line SW1990 through the regulation of E-cadherin and expression of MMP-2.
Collapse
|
30
|
Yeon SE, No DY, Lee SH, Nam SW, Oh IH, Lee J, Kuh HJ. Application of concave microwells to pancreatic tumor spheroids enabling anticancer drug evaluation in a clinically relevant drug resistance model. PLoS One 2013; 8:e73345. [PMID: 24039920 PMCID: PMC3769301 DOI: 10.1371/journal.pone.0073345] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2013] [Accepted: 07/19/2013] [Indexed: 12/11/2022] Open
Abstract
Intrinsic drug resistance of pancreatic ductal adenocarcinoma (PDAC) warrants studies using models that are more clinically relevant for identifying novel resistance mechanisms as well as for drug development. Tumor spheroids (TS) mimic in vivo tumor conditions associated with multicellular resistance and represent a promising model for efficient drug screening, however, pancreatic cancer cells often fail to form spheroids using conventional methods such as liquid overlay. This study describes the induction of TS of human pancreatic cancer cells (Panc-1, Aspc-1, Capan-2) in concave polydimethylsiloxane (PDMS) microwell plates and evaluation of their usefulness as an anticancer efficacy test model. All three cell lines showed TS formation with varying degree of necrosis inside TS. Among these, Panc-1 spheroid with spherical morphology, a rather rough surface, and unique adhesion structures were successfully produced with no notable necrosis in concave microwell plates. Panc-1 TS contained growth factors or enzymes such as TGF-β1, CTGF, and MT1-MMP, and extracellular matrix proteins such as collagen type I, fibronectin, and laminin. Panc-1 cells grown as TS showed changes in stem cell populations and in expression levels of miRNAs that may play roles in chemoresistance. Visualization of drug penetration and detection of viability indicators, such as Ki-67 and MitoSOX, were optimized for TS for quantitative analysis. Water-soluble tetrazolium (MTS) and acid phosphatase (APH) assays were also successfully optimized. Overall, we demonstrated that concave PDMS microwell plates are a novel platform for preparation of TS of weakly aggregating cells and that Panc-1 spheroids may represent a novel three-dimensional model for anti-pancreatic cancer drug screening.
Collapse
Affiliation(s)
- Sang-Eun Yeon
- Lab of Onco-Pharmacology and Experimental Therapeutics, Department of Biomedical Sciences, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Da Yoon No
- Department of Biomedical Engineering, College of Health Science, Korea University, Seoul, Republic of Korea
| | - Sang-Hoon Lee
- Department of Biomedical Engineering, College of Health Science, Korea University, Seoul, Republic of Korea
| | - Suk Woo Nam
- Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Cancer Evolution Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Il-Hoan Oh
- Catholic High Performance Cell Therapy Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Cancer Evolution Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jaehwi Lee
- College of Pharmacy, Chung-Ang University, Seoul, Republic of Korea
| | - Hyo-Jeong Kuh
- Lab of Onco-Pharmacology and Experimental Therapeutics, Department of Biomedical Sciences, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Cancer Evolution Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| |
Collapse
|
31
|
Ultrasound-guided direct delivery of 3-bromopyruvate blocks tumor progression in an orthotopic mouse model of human pancreatic cancer. Target Oncol 2013; 8:145-51. [PMID: 23529644 DOI: 10.1007/s11523-013-0273-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2013] [Accepted: 03/10/2013] [Indexed: 02/07/2023]
Abstract
Studies in animal models of cancer have demonstrated that targeting tumor metabolism can be an effective anticancer strategy. Previously, we showed that inhibition of glucose metabolism by the pyruvate analog, 3-bromopyruvate (3-BrPA), induces anticancer effects both in vitro and in vivo. We have also documented that intratumoral delivery of 3-BrPA affects tumor growth in a subcutaneous tumor model of human liver cancer. However, the efficacy of such an approach in a clinically relevant orthotopic tumor model has not been reported. Here, we investigated the feasibility of ultrasound (US) image-guided delivery of 3-BrPA in an orthotopic mouse model of human pancreatic cancer and evaluated its therapeutic efficacy. In vitro, treatment of Panc-1 cells with 3-BrPA resulted in a dose-dependent decrease in cell viability. The loss of viability correlated with a dose-dependent decrease in the intracellular ATP level and lactate production confirming that disruption of energy metabolism underlies these 3-BrPA-mediated effects. In vivo, US-guided delivery of 3-BrPA was feasible and effective as demonstrated by a marked decrease in tumor size on imaging. Further, the antitumor effect was confirmed by (1) a decrease in the proliferative potential by Ki-67 immunohistochemical staining and (2) the induction of apoptosis by terminal deoxynucleotidyl transferase-mediated deoxyuridine 5-triphospate nick end labeling staining. We therefore demonstrate the technical feasibility of US-guided intratumoral injection of 3-BrPA in a mouse model of human pancreatic cancer as well as its therapeutic efficacy. Our data suggest that this new therapeutic approach consisting of a direct intratumoral injection of antiglycolytic agents may represent an exciting opportunity to treat patients with pancreas cancer.
Collapse
|
32
|
Mechanistic evaluation of a novel small molecule targeting mitochondria in pancreatic cancer cells. PLoS One 2013; 8:e54346. [PMID: 23349858 PMCID: PMC3549929 DOI: 10.1371/journal.pone.0054346] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2012] [Accepted: 12/12/2012] [Indexed: 12/25/2022] Open
Abstract
Background Pancreatic cancer is one of the deadliest cancers with a 5-year survival rate of 6%. Therapeutic options are very limited and there is an unmet medical need for safe and efficacious treatments. Cancer cell metabolism and mitochondria provide unexplored targets for this disease. We recently identified a novel class of triphenylphosphonium salts, TP compounds, with broad- spectrum anticancer properties. We examined the ability of our prototypical compound TP421– chosen for its fluorescent properties – to inhibit the growth of pancreatic cancer cells and further investigated the molecular mechanisms by which it exerts its anticancer effects. Methodology/Principal Findings TP421 exhibited sub-micromolar IC50 values in all the pancreatic cancer cell lines tested using MTT and colony formation assays. TP421 localized predominantly to mitochondria and induced G0/G1 arrest, ROS accumulation, and activation of several stress-regulated kinases. Caspase and PARP-1 cleavage were observed indicating an apoptotic response while LC3B-II and p62 were accumulated indicating inhibition of autophagy. Furthermore, TP421 induced de-phosphorylation of key signaling molecules involved in FAK mediated adhesion that correlated with inhibition of cell migration. Conclusions/Significance TP421 is a representative compound of a new promising class of mitochondrial-targeted agents useful for pancreatic cancer treatment. Because of their unique mechanism of action and efficacy further development is warranted.
Collapse
|
33
|
Liu Y, Chen M, Luo Z, Lin J, Song L. Investigation on the site-selective binding of bovine serum albumin by erlotinib hydrochloride. J Biomol Struct Dyn 2012; 31:1160-74. [PMID: 23072300 DOI: 10.1080/07391102.2012.726532] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
The purpose of this study was to investigate the site-selective binding of erlotinib hydrochloride (ET), a targeted anticancer drug, to bovine serum albumin (BSA) through 1H NMR, spectroscopic, thermodynamic, and molecular modeling methods. The fluorescence quenching of BSA by ET was a result of the formation of BSA-ET complex with high binding affinity. The site marker competition study combined with isothermal titration calorimetry experiment revealed that ET binds to site II of BSA mainly through hydrogen bond and van der Waals force. Molecular docking was further applied to define the specific binding site of ET to BSA. The conformation of BSA was changed in the presence of ET, revealed by synchronous fluorescence, circular dichroism, and three-dimensional fluorescence spectroscopy results. Further, NMR analysis of the complex revealed that the binding capacity contributed by the aromatic protons in the binding site of BSA might be greater than the aliphatic protons. An animated interactive 3D complement (I3DC) is available in Proteopedia at http://proteopedia.org/w/Journal:JBSD:26.
Collapse
Affiliation(s)
- Yan Liu
- a The State Key Lab of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences , Fuzhou , Fujian , 350002 , P.R. China
| | | | | | | | | |
Collapse
|
34
|
Hofmann I, Weiss A, Elain G, Schwaederle M, Sterker D, Romanet V, Schmelzle T, Lai A, Brachmann SM, Bentires-Alj M, Roberts TM, Sellers WR, Hofmann F, Maira SM. K-RAS mutant pancreatic tumors show higher sensitivity to MEK than to PI3K inhibition in vivo. PLoS One 2012; 7:e44146. [PMID: 22952903 PMCID: PMC3432074 DOI: 10.1371/journal.pone.0044146] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2012] [Accepted: 07/30/2012] [Indexed: 12/30/2022] Open
Abstract
Activating K-RAS mutations occur at a frequency of 90% in pancreatic cancer, and to date no therapies exist targeting this oncogene. K-RAS signals via downstream effector pathways such as the MAPK and the PI3K signaling pathways, and much effort has been focused on developing drugs targeting components of these pathways. To better understand the requirements for K-RAS and its downstream signaling pathways MAPK and PI3K in pancreatic tumor maintenance, we established an inducible K-RAS knock down system that allowed us to ablate K-RAS in established tumors. Knock down of K-RAS resulted in impaired tumor growth in all pancreatic xenograft models tested, demonstrating that K-RAS expression is indeed required for tumor maintenance of K-RAS mutant pancreatic tumors. We further examined signaling downstream of K-RAS, and detected a robust reduction of pERK levels upon K-RAS knock down. In contrast, no effect on pAKT levels could be observed due to almost undetectable basal expression levels. To investigate the requirement of the MAPK and the PI3K pathways on tumor maintenance, three selected pancreatic xenograft models were tested for their response to MEK or PI3K inhibition. Tumors of all three models regressed upon MEK inhibition, but showed less pronounced response to PI3K inhibition. The effect of MEK inhibition on pancreatic xenografts could be enhanced further by combined application of a PI3K inhibitor. These data provide further rationale for testing combinations of MEK and PI3K inhibitors in clinical trials comprising a patient population with pancreatic cancer harboring mutations in K-RAS.
Collapse
Affiliation(s)
- Irmgard Hofmann
- Novartis Institutes for Biomedical Research, Oncology Disease Area, Basel, Switzerland.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Li Y, Gu WJ, Liu HL. Induction of pancreatic cancer cell apoptosis and enhancement of gemcitabine sensitivity by RAP80 siRNA. Dig Dis Sci 2012; 57:2072-8. [PMID: 22573342 DOI: 10.1007/s10620-012-2132-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2011] [Accepted: 03/02/2012] [Indexed: 12/20/2022]
Abstract
BACKGROUND Receptor-associated protein 80 (RAP80) increases substantially in pancreatic cancer. The involvement of RAP80 in the chemoresistance of pancreatic cancer should be elucidated. AIMS We investigated the effects of inhibiting RAP80 expression on the sensitivity of pancreatic cancer cells to gemcitabine chemotherapy by using small interfering RNA (siRNA). METHODS Chemically synthesized siRNA RAP80 was transfected into human pancreatic cancer cell lines SW1990 and Capan-2. The IC(50) of gemcitabine was determined by using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) assay. Cell apoptosis was detected by using flow cytometry. Expression of apoptosis-related genes, Bax, Bcl-2, TRAIL, survivin, and caspase-8 was detected by using reverse transcription-polymerase chain reaction (RT-PCR) and western blot. RESULTS Gemcitabine inhibits proliferation of SW1990 and Capan-2 cells in a concentration-dependent manner. Inhibition of RAP80 expression significantly reduced the IC(50) of gemcitabine (P < 0.01). RAP80 siRNA combined with gemcitabine significantly increased (P < 0.01) apoptosis of pancreatic cancer cell lines SW1990 and Capan-2, increased expression of Bax mRNA, reduced Bcl-2 mRNA expression (P < 0.01), and slightly increased TRAIL mRNA expression (P < 0.01). Correspondingly, in the RAP80 siRNA combined with gemcitabine group, both Bax and cleaved caspase-8 protein levels were increased (P < 0.01), whereas Bcl-2 protein decreased significantly (P < 0.01). No change in survivin mRNA expression was observed (P < 0.01). CONCLUSION Inhibition of RAP80 expression can induce apoptosis in pancreatic cancer cells and improve chemosensitivity to gemcitabine.
Collapse
Affiliation(s)
- Ya Li
- Department of Gastroenterology, The Ninth People's Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai 200011, China
| | | | | |
Collapse
|
36
|
Hornick JR, Spitzer D, Goedegebuure P, Mach RH, Hawkins WG. Therapeutic targeting of pancreatic cancer utilizing sigma-2 ligands. Surgery 2012; 152:S152-6. [PMID: 22763259 DOI: 10.1016/j.surg.2012.05.014] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2012] [Accepted: 05/11/2012] [Indexed: 01/05/2023]
Abstract
One major barrier in the development of pancreas cancer therapeutics is the selective delivery of the drugs to their cellular targets. We have previously developed several sigma-2 ligands and reported the discovery of a component of the receptor for these ligands. Several sigma-2 ligands have been shown to trigger apoptosis in pancreas cancer cells. More importantly, sigma-2 ligands are internalized rapidly by the cancer cells and are capable of delivering other small-molecule therapeutics. Here we review sigma-2 ligands and conjugates as a potential novel therapy suitable for investigation in patients with pancreatic cancer.
Collapse
Affiliation(s)
- John R Hornick
- Department of Surgery, Harvard Medical School, Boston, MA, USA
| | | | | | | | | |
Collapse
|
37
|
Leake K, Singhal J, Nagaprashantha LD, Awasthi S, Singhal SS. RLIP76 regulates PI3K/Akt signaling and chemo-radiotherapy resistance in pancreatic cancer. PLoS One 2012; 7:e34582. [PMID: 22509328 PMCID: PMC3317991 DOI: 10.1371/journal.pone.0034582] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2012] [Accepted: 03/07/2012] [Indexed: 11/18/2022] Open
Abstract
Purpose Pancreatic cancer is an aggressive malignancy with characteristic metastatic course of disease and resistance to conventional chemo-radiotherapy. RLIP76 is a multi-functional cell membrane protein that functions as a major mercapturic acid pathway transporter as well as key regulator of receptor-ligand complexes. In this regard, we investigated the significance of targeting RLIP76 on PI3K/Akt pathway and mechanisms regulating response to chemo-radiotherapy. Research Design and Methods Cell survival was assessed by MTT and colony forming assays. Cellular levels of proteins and phosphorylation was determined by Western blot analyses. The impact on apoptosis was determined by TUNEL assay. The anti-cancer effects of RLIP76 targeted interventions in vivo were determined using mice xenograft model of the pancreatic cancer. The regulation of doxorubicin transport and radiation sensitivity were determined by transport studies and colony forming assays, respectively. Results Our current studies reveal an encompassing model for the role of RLIP76 in regulating the levels of fundamental proteins like PI3K, Akt, E-cadherin, CDK4, Bcl2 and PCNA which are of specific importance in the signal transduction from critical upstream signaling cascades that determine the proliferation, apoptosis and differentiation of pancreatic cancer cells. RLIP76 depletion also caused marked and sustained regression of established human BxPC-3 pancreatic cancer tumors in nude mouse xenograft model. RLIP76 turned out to be a major regulator of drug transport along with contributing to the radiation resistance in pancreatic cancer. Conclusions/Significance RLIP76 represents a mechanistically significant target for developing effective interventions in aggressive and refractory pancreatic cancers.
Collapse
Affiliation(s)
- Kathryn Leake
- Department of Diabetes and Metabolic Diseases Research, Beckman Research Institute, City of Hope, Comprehensive Cancer Center, Duarte, California, United States of America
| | - Jyotsana Singhal
- Department of Diabetes and Metabolic Diseases Research, Beckman Research Institute, City of Hope, Comprehensive Cancer Center, Duarte, California, United States of America
| | - Lokesh Dalasanur Nagaprashantha
- Department of Diabetes and Metabolic Diseases Research, Beckman Research Institute, City of Hope, Comprehensive Cancer Center, Duarte, California, United States of America
| | - Sanjay Awasthi
- Department of Diabetes and Metabolic Diseases Research, Beckman Research Institute, City of Hope, Comprehensive Cancer Center, Duarte, California, United States of America
| | - Sharad S. Singhal
- Department of Diabetes and Metabolic Diseases Research, Beckman Research Institute, City of Hope, Comprehensive Cancer Center, Duarte, California, United States of America
- * E-mail:
| |
Collapse
|
38
|
Li F, Hu G, Jiang Z, Guo J, Wang K, Ouyang K, Wen D, Zhu M, Liang J, Qin X, Zhang L. Identification of NME5 as a contributor to innate resistance to gemcitabine in pancreatic cancer cells. FEBS J 2012; 279:1261-73. [PMID: 22325559 DOI: 10.1111/j.1742-4658.2012.08521.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The limited therapeutic effect of gemcitabine on pancreatic cancer is largely attributed to pre-existing or acquired resistance of the tumor cells. This study was aimed at screening for candidate resistance-related gene(s) and elucidating the underlying mechanisms. NME5 was found to be highly expressed in an innate gemcitabine-resistant human pancreatic cancer sample and the cell line PAXC002 derived from the sample. Downregulation of NME5 significantly reversed gemcitabine resistance in PAXC002 cells, whereas NME5 overexpression induced gemcitabine resistance in the pancreatic cancer cell line BxPC-3. NME5 attenuated the induction of apoptosis and cell cycle arrest induced by gemcitabine, probably accounting for the blunted sensitivity to gemcitabine. Furthermore, NME5 was demonstrated to play its role in a nuclear factor kappaB (NF-κB)-dependent manner. NME5 was capable of directly binding NF-κB, and possibly regulated its expression level in PAXC002 cells. Our results also suggest that NF-κB is a key executor of NME5 in regulating apoptosis and cell cycle. All of these data suggest that NME5 is a promising target for relieving innate gemcitabine resistance in pancreatic cancer cells.
Collapse
Affiliation(s)
- Fu Li
- Jiangsu Center of Drug Screening, China Pharmaceutical University, Nanjing, Jiangsu Province, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Chen D, Zheng X, Kang D, Yan B, Liu X, Gao Y, Zhang K. Apoptosis and expression of the Bcl-2 family of proteins and P53 in human pancreatic ductal adenocarcinoma. Med Princ Pract 2012; 21:68-73. [PMID: 22024503 DOI: 10.1159/000332423] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2011] [Accepted: 06/12/2011] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE The purpose of this study was to clarify the association between P53 and the Bcl-2 family (Bcl-2, Bax, Bcl-xL, Bcl-xS) expression and apoptosis in pancreatic ductal adenocarcinoma (PDAC). SUBJECTS AND METHODS A total of 70 patients with PDAC were studied. The expression of P53 protein in PDAC was assessed using the immunohistochemical method, which categorized the PDAC patients into two groups: group 1: 36 cases with immunonegative P53(-), and group 2: 34 cases with immunopositive P53(+). The expression of Bcl-2, Bax, Bcl-xL, and Bcl-xS in the 70 PDAC cases was detected by immunohistochemical and Western blotting methods. The apoptotic index (AI) was also measured in these samples by the terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling (TUNEL) method. The relation between P53 and the Bcl-2 protein family and apoptosis was then evaluated. RESULTS Bcl-2 and Bcl-xS expression was significantly associated with P53 (p < 0.05). No clear associations were found among P53, Bax and Bcl-xL expression (p > 0.05). The AI of groups 1 and 2 was 12.1 ± 2.47 and 8.1 ± 1.48, respectively (p = 0.023). There was no relationship between AI and Bcl-2, Bax, Bcl-xL and Bcl-xS expression (p > 0.05, respectively). Bcl-2/Bax ratio was significantly associated with AI (p < 0.01). CONCLUSION Bcl-2 and Bcl-xS represent significant anti- and proapoptotic proteins, respectively, modulated through a P53-dependent pathway in PDAC, and P53 modulated apoptosis mainly through Bcl-2/Bax ratio.
Collapse
Affiliation(s)
- Dong Chen
- Department of General Surgery, Affiliated Hospital of Medical College, QingDao University, QingDao, ShanDong Province, People’s Republic of China
| | | | | | | | | | | | | |
Collapse
|
40
|
Ropolo A, Bagnes CI, Molejon MI, Lo Re A, Boggio V, Gonzalez CD, Vaccaro MI. Chemotherapy and autophagy-mediated cell death in pancreatic cancer cells. Pancreatology 2012; 12:1-7. [PMID: 22487466 DOI: 10.1016/j.pan.2011.11.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Autophagy is an evolutionarily preserved degradation process of cytoplasmic cellular constituents and plays important physiological roles in human health and disease. It has been proposed that autophagy plays an important role both in tumor progression and in promotion of cancer cell death, although the molecular mechanisms responsible for this dual action of autophagy in cancer have not been elucidated. Pancreatic ductal adenocarcinoma is one of the most aggressive human malignancies with 2-3% five-year survival rate. Its poor prognosis has been attributed to the lack of specific symptoms and early detection tools, and its relatively refractory to traditional cytotoxic agents and radiotherapy. Experimental evidence pointed at autophagy as a pancreatic cancer cell mechanism to survive under adverse environmental conditions, or as a defective programmed cell death mechanism that favors pancreatic cancer cell resistance to treatment. Here, we consider several phenotypical alterations that have been related to increase or decrease the autophagic process in pancreatic tumor cells. We specially review autophagy as a cell death mechanism in response to chemotherapeutic drugs.
Collapse
Affiliation(s)
- Alejandro Ropolo
- Department of Pathophysiology, School of Pharmacy and Biochemistry, University of Buenos Aires, 956 Junin p5, C1113AAD Buenos Aires, Argentina
| | | | | | | | | | | | | |
Collapse
|
41
|
Chen Q, Wang Z, Zhang K, Liu X, Cao W, Zhang L, Zhang S, Yan B, Wang Y, Xia C. Clusterin confers gemcitabine resistance in pancreatic cancer. World J Surg Oncol 2011; 9:59. [PMID: 21609464 PMCID: PMC3120680 DOI: 10.1186/1477-7819-9-59] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2011] [Accepted: 05/24/2011] [Indexed: 12/19/2022] Open
Abstract
Objective To measure clusterin expression in pancreatic cancer tissues and cell lines and to evaluate whether clusterin confers resistance to gmcitabine in pancreatic cancer cells. Methods Immunohistochemistry for clusterin was performed on 50 primary pancreatic cancer tissues and 25 matched backgrounds, and clusterin expression in 5 pancreatic cancer cell lines was quantified by Western blot and PT-PCR. The correlation between clusterin expression level and gmcitabine IC50 in pancreatic cancer cell lines was evaluated. The effect of an antisense oligonucleotide (ASO) against clusterin(OGX-011) on gmcitabine resistance was evaluated by MTT assays. Xenograft model was used to demonstrate tumor growth. Results Pancreatic cancer tissues expressed significantly higher levels of clusterin than did normal pancreatic tissues (P < 0.01). Clusterin expression levels were correlated with gmcitabine resistance in pancreatic cancer cell lines, and OGX-011 significantly decreased BxPc-3 cells resistance to gmcitabine (P < 0.01). In vivo systemic administration of AS clusterin and gmcitabine significantly decreased the s.c. BxPC-3 tumor volume compared with mismatch control ODN plus gmcitabine. Conclusion Our finding that clusterin expression was significantly higher in pancreatic cancer than in normal pancreatic tissues suggests that clusterin may confer gmcitabine resistance in pancreatic cancer cells.
Collapse
Affiliation(s)
- Qingfeng Chen
- Affiliated Hospital of Medical College, QingDao University, RP China
| | | | | | | | | | | | | | | | | | | |
Collapse
|