1
|
Kulkarni T, Robinson OM, Dutta A, Mukhopadhyay D, Bhattacharya S. Machine learning-based approach for automated classification of cell and extracellular matrix using nanomechanical properties. Mater Today Bio 2024; 25:100970. [PMID: 38312803 PMCID: PMC10835007 DOI: 10.1016/j.mtbio.2024.100970] [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: 10/10/2023] [Revised: 01/16/2024] [Accepted: 01/19/2024] [Indexed: 02/06/2024] Open
Abstract
Fibrosis characterized by excess accumulation of extracellular matrix (ECM) due to complex cell-ECM interactions plays a pivotal role in pathogenesis. Herein, we employ the pancreatic ductal adenocarcinoma (PDAC) model to investigate dynamic alterations in nanomechanical attributes arising from the cell-ECM interactions to study the fibrosis paradigm. Several segregated studies performed on cellular and ECM components fail to recapitulate their complex collaboration. We utilized collagen and fibronectin, the two most abundant PDAC ECM components, and studied their nanomechanical attributes. We demonstrate alteration in morphology and nanomechanical attributes of collagen with varying thicknesses of collagen gel. Furthermore, by mixing collagen and fibronectin in various stoichiometry, their nanomechanical attributes were observed to vary. To demonstrate the dynamicity and complexity of cell-ECM, we utilized Panc-1 and AsPC-1 cells with or without collagen. We observed that Panc-1 and AsPC-1 cells interact differently with collagen and vice versa, evident from their alteration in nanomechanical properties. Further, using nanomechanics data, we demonstrate that ML-based techniques were able to classify between ECM as well as cell, and cell subtypes in the presence/absence of collagen with higher accuracy. This work demonstrates a promising avenue to explore other ECM components facilitating deeper insights into tumor microenvironment and fibrosis paradigm.
Collapse
Affiliation(s)
- Tanmay Kulkarni
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, 4500 San Pablo Road South, Jacksonville, FL, 32224, USA
| | - Olivia-Marie Robinson
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, 4500 San Pablo Road South, Jacksonville, FL, 32224, USA
| | - Ayan Dutta
- School of Computing, University of North Florida, Jacksonville, FL, 32224 USA
| | - Debabrata Mukhopadhyay
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, 4500 San Pablo Road South, Jacksonville, FL, 32224, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Science, 4500 San Pablo Road South, Jacksonville, FL, 32224, USA
| | - Santanu Bhattacharya
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, 4500 San Pablo Road South, Jacksonville, FL, 32224, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Science, 4500 San Pablo Road South, Jacksonville, FL, 32224, USA
| |
Collapse
|
2
|
De Luca F, Di Chio C, Zappalà M, Ettari R. Dihydrochalcones as antitumor agents. Curr Med Chem 2022; 29:5042-5061. [PMID: 35430969 DOI: 10.2174/0929867329666220415113219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 01/16/2022] [Accepted: 01/25/2022] [Indexed: 11/22/2022]
Abstract
Dihydrochalcones are a class of secondary metabolites, possessing several biological properties such as antitumor, antioxidant, antibacterial, antidiabetic, estrogenic, anti-inflammatory, antithrombotic, antiviral, neuroprotective and immunomodulator properties; therefore, they are currently considered promising candidates in the drug discovery process. This review intend to debate their pharmacological actions with a particular attention to their antitumor activity against a panel of cancer cell-lines and to the description of the inhibition mechanisms of cell proliferation such as the regulation of angiogenesis, apoptosis, etc etc.
Collapse
Affiliation(s)
- Fabiola De Luca
- Department of Chemical, Biological, Pharmaceutical and Environmental Chemistry, University of Messina, Italy
| | - Carla Di Chio
- Department of Chemical, Biological, Pharmaceutical and Environmental Chemistry, University of Messina, Italy
| | - Maria Zappalà
- Department of Chemical, Biological, Pharmaceutical and Environmental Chemistry, University of Messina, Italy
| | - Roberta Ettari
- Department of Chemical, Biological, Pharmaceutical and Environmental Chemistry, University of Messina, Italy
| |
Collapse
|
3
|
Feng Y, Jiang Y, Hao F. GSK2126458 has the potential to inhibit the proliferation of pancreatic cancer uncovered by bioinformatics analysis and pharmacological experiments. J Transl Med 2021; 19:373. [PMID: 34461940 PMCID: PMC8406597 DOI: 10.1186/s12967-021-03050-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 08/24/2021] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Pancreatic cancer is one of the most serious digestive malignancies. At present, there is an extreme lack of effective strategies in clinical treatment. The purpose of this study is to identify key genes and pathways in the development of pancreatic cancer and provide targets for the treatment of pancreatic cancer. METHODS GSE15471 and GSE62165 were used to screen differentially expressed genes by GEO2R tool. Hub genes prognostic potential assessed using the GEPIA and Kaplan-Meier plotter databases. The drug susceptibility data of pan-cancer cell lines is provided by The Genomics of Drug Sensitivity in Cancer Project (GDSC). Finally, the effects of PI3K-Akt signaling pathway inhibitors on cell viability of pancreatic cancer cells were detected by cell proliferation and invasion assays. RESULTS A total of 609 differentially expressed genes were screened and enriched in the focal adhesion, phagosome and PI3K-Akt signaling pathway. Of the 15 hub genes we found, four were primarily associated with the PI3K-Akt signaling pathway, including COL3A1, EGF, FN1 and ITGA2. GDSC analysis showed that mTOR inhibitors are very sensitive to pancreatic cancer cells with mutations in EWSR1.FLI1 and RNF43. Cell proliferation and invasion results showed that mTOR inhibitors (GSK2126458) can inhibit the proliferation of pancreatic cancer cells. CONCLUSIONS This study suggested that the PI3K-Akt signaling pathway may be a key pathway for pancreatic cancer, our study uncovered the potential therapeutic potential of GSK2126458, a specific mTOR inhibitor, for pancreatic cancer.
Collapse
Affiliation(s)
- Yueqin Feng
- Department of Ultrasound, The First Affiliated Hospital of China Medical University, Shenyang, 110022, Liaoning, China.
| | - Yuguan Jiang
- School of Pharmacy, China Medical University, Shenyang, 110122, Liaoning, China
| | - Fengjin Hao
- Department of Biochemistry and Molecular Biology, China Medical University, Shenyang, 110122, Liaoning, China
| |
Collapse
|
4
|
Unger K, Mehta KY, Kaur P, Wang Y, Menon SS, Jain SK, Moonjelly RA, Suman S, Datta K, Singh R, Fogel P, Cheema AK. Metabolomics based predictive classifier for early detection of pancreatic ductal adenocarcinoma. Oncotarget 2018; 9:23078-23090. [PMID: 29796173 PMCID: PMC5955422 DOI: 10.18632/oncotarget.25212] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 04/06/2018] [Indexed: 12/13/2022] Open
Abstract
The availability of robust classification algorithms for the identification of high risk individuals with resectable disease is critical to improving early detection strategies and ultimately increasing survival rates in PC. We leveraged high quality biospecimens with extensive clinical annotations from patients that received treatment at the Medstar-Georgetown University hospital. We used a high resolution mass spectrometry based global tissue profiling approach in conjunction with multivariate analysis for developing a classification algorithm that would predict early stage PC with high accuracy. The candidate biomarkers were annotated using tandem mass spectrometry. We delineated a six metabolite panel that could discriminate early stage PDAC from benign pancreatic disease with >95% accuracy of classification (Specificity = 0.85, Sensitivity = 0.9). Subsequently, we used multiple reaction monitoring mass spectrometry for evaluation of this panel in plasma samples obtained from the same patients. The pattern of expression of these metabolites in plasma was found to be discordant as compared to that in tissue. Taken together, our results show the value of using a metabolomics approach for developing highly predictive panels for classification of early stage PDAC. Future investigations will likely lead to the development of validated biomarker panels with potential for clinical translation in conjunction with CA-19-9 and/or other biomarkers.
Collapse
Affiliation(s)
- Keith Unger
- MedStar Georgetown University Hospital, Washington, DC, United States of America
| | - Khyati Y Mehta
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, United States of America
| | - Prabhjit Kaur
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, United States of America
| | - Yiwen Wang
- Department of Biostatistics and Biomathematics, Georgetown University Medical Center, Washington, DC, United States of America
| | - Smrithi S Menon
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, United States of America
| | - Shreyans K Jain
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, United States of America
| | - Rose A Moonjelly
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, United States of America
| | - Shubhankar Suman
- Departments of Biochemistry and Molecular and Cellular Biology, Georgetown University Medical Center, Washington, DC, United States of America
| | - Kamal Datta
- Departments of Biochemistry and Molecular and Cellular Biology, Georgetown University Medical Center, Washington, DC, United States of America
| | - Rajbir Singh
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, United States of America
| | - Paul Fogel
- Unité MéDIAN, UMR CNRS 6237 MEDYC, Université de Reims, Reims, France
| | - Amrita K Cheema
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, United States of America.,Departments of Biochemistry and Molecular and Cellular Biology, Georgetown University Medical Center, Washington, DC, United States of America
| |
Collapse
|
5
|
Dorjee P, Long ZW. A mixed treatment comparison of toxicity of gemcitabine combined with different targeted drugs in the treatment of advanced or metastatic pancreatic cancer. Cancer Biol Ther 2018; 19:497-506. [PMID: 29658816 DOI: 10.1080/15384047.2018.1433503] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The mixed treatment comparison study was performed in order to compare the toxicities of Gemcitabine and different targeted drug combinations in the treatment of advanced/metastatic pancreatic cancer (PC). Searches were performed from the inception of PubMed and Cochrane Library databases to February 2017. This study included randomized controlled trials (RCTs) of Gemcitabine and different targeted drug combinations in the treatment of advanced/metastatic PC. Odds ratio (OR) values were calculated by direct and indirect comparisons, and the surface under the cumulative ranking curves (SUCRA) were drawn. A total of six RCTs were finally incorporated into the study. These studies included six therapy regimens: Gemcitabine + Axitinib, Gemcitabine + Trametinib, Gemcitabine + Sorafenib, Gemcitabine + Bevacizumab, Gemcitabine + Erlotinib and Gemcitabine + Tipifarnib. The results showed that Gemcitabine + Axitinib combinations showed lower incidence rates of rashes (all grades) in comparison to Gemcitabine + Trametinib and Gemcitabine + Erlotinib combinations. Compared with Gemcitabine+ Trametinib combinations, Gemcitabine + Axitinib combinations showed lower incidence rates of diarrhea (grade ≥ 3). Moreover, the cluster analyses results revealed that Gemcitabine + Axitinib combinations and Gemcitabine + Sorafenib combinations showed lower incidence rates of hematotoxicity, while Gemcitabine + Axitinib combinations showed lower incidence rates of non-hematotoxicity. Collectively, the data provided strong evidence of Gemcitabine + Axitinib combinations showing lower incidence rates of non-hematotoxicity, and Gemcitabine + Axitinib and Gemcitabine + Sorafenib combinations may have lower incidence rates of hematotoxicity in the treatment of advanced/metastatic PC.
Collapse
Affiliation(s)
- Penpa Dorjee
- a Department of Medicine , Shigatse People's Hospital , Shigatse , P.R. China.,b Department of Gastric Cancer and Soft-Tissue Sarcoma Surgery , Fudan University Shanghai Cancer Center , Shanghai , P.R. China.,c Department of Oncology , Shanghai Medical College of Fudan University , Shanghai , P.R. China
| | - Zi-Wen Long
- a Department of Medicine , Shigatse People's Hospital , Shigatse , P.R. China.,b Department of Gastric Cancer and Soft-Tissue Sarcoma Surgery , Fudan University Shanghai Cancer Center , Shanghai , P.R. China.,c Department of Oncology , Shanghai Medical College of Fudan University , Shanghai , P.R. China
| |
Collapse
|
6
|
Wang XF, Huang WF, Nie J, Zhou Y, Tan DW, Jiang JH. Toxicity of chemotherapy regimens in advanced and metastatic pancreatic cancer therapy: A network meta-analysis. J Cell Biochem 2018; 119:5082-5103. [PMID: 28681936 DOI: 10.1002/jcb.26266] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 07/05/2017] [Indexed: 02/06/2023]
Abstract
This network meta-analysis is adopted in order to compare the toxicity of different chemotherapy regimens in the treatment of advanced/metastatic pancreatic cancer (PC). Randomized controlled trials (RCTs) about different chemotherapy regimens for advanced/metastatic PC were included in this network meta-analysis using Cochrane Library and PubMed electronic databases. The network meta-analysis was performed to combine direct and indirect evidence in order to calculate the odd ratios (OR) and draw a surface under the cumulative ranking (SUCRA) curve. A total of 19 RCTs were enrolled in this network meta-analysis including 12 chemotherapy regimens (Gemcitabine, Gemcitabine + S-1 [tegafur], Gemcitabine + nab-paclitaxel, Gemcitabine + Capecitabine, Gemcitabine + Cisplatin, FOLFIRINOX [oxaliplatin + irinotecan + fluorouracil + leucovorin], Gemcitabine + oxaliplatin, Gemcitabine + irinotecan, Gemcitabine + Exatecan, Gemcitabine + pemetrexed, Gemcitabine + 5-FU, S-1). The incidence of anemia of Gemcitabine + Capecitabine regimen was higher compared with Gemcitabine regimen, Gemcitabine + pemetrexed regimen exhibited the highest incidence rates of anemia and neutropenia; while Gemcitabine + S-1, Gemcitabine + Cisplatin and FOLFIRINOX regimens exhibited the highest incidence rates of neutropenia. However, S-1 regimen exhibited lower incidence rates of leukopenia and thrombocytopenia. Moreover, the incidence rates of nausea/vomiting and rash of Gemcitabine + S-1 regimen were higher compared with Gemcitabine regimen, while Gemcitabine + Cisplatin regimen had the highest incidence rate of nausea/vomiting. This study demonstrated that the hematologic toxicity of S-1 regimen was the lowest, while Gemcitabine regimen exhibited the lowest incidence rate of non-hematologic toxicity, providing guidance for the treatment of advanced/metastatic PC.
Collapse
Affiliation(s)
- Xiao-Fang Wang
- Department of Hepatobiliary Surgery, Jiangxi Pingxiang People's Hospital, Pingxiang, P. R. China
| | - Wen-Feng Huang
- Department of Hepatobiliary Surgery, Jiangxi Pingxiang People's Hospital, Pingxiang, P. R. China
| | - Jian Nie
- Department of Hepatobiliary Surgery, Jiangxi Pingxiang People's Hospital, Pingxiang, P. R. China
| | - Yong Zhou
- Department of Hepatobiliary Surgery, Jiangxi Pingxiang People's Hospital, Pingxiang, P. R. China
| | - Ding-Wu Tan
- Department of Hepatobiliary Surgery, Jiangxi Pingxiang People's Hospital, Pingxiang, P. R. China
| | - Ji-Hao Jiang
- Department of Hepatobiliary Surgery, Jiangxi Pingxiang People's Hospital, Pingxiang, P. R. China
| |
Collapse
|
7
|
Liu SY, Xu N, Liu LJ, Wang YX, Ma DY. An Efficient Synthesis of Angelmarin and its Analogs. Nat Prod Commun 2017. [DOI: 10.1177/1934578x1701201215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Angelmarin, a naturally occurring coumarin, exhibited highly anti-austerity potency towards human pancreatic carcinoma cell line PANC-1. In this paper, an efficient and eco-friendly synthesis of angelmarin and its analogs from columbianetin has been developed via a ZnO mediated esterification and a Wittig reaction.
Collapse
Affiliation(s)
- Su-You Liu
- School of Pharmaceutical Sciences, Central South University, Changsha 410013, China
| | - Na Xu
- School of Pharmaceutical Sciences, Central South University, Changsha 410013, China
| | - Li-Jun Liu
- School of Life Sciences, Central South University, Changsha 410013, China
| | - Ying-Xiong Wang
- Shanxi Engineering Research Center of Biorefinery, Institute of Coal Chemistry, Chinese Academy of Sciences, Taiyuan 030001, China
| | - Da-You Ma
- School of Pharmaceutical Sciences, Central South University, Changsha 410013, China
| |
Collapse
|
8
|
Mehta KY, Wu HJ, Menon SS, Fallah Y, Zhong X, Rizk N, Unger K, Mapstone M, Fiandaca MS, Federoff HJ, Cheema AK. Metabolomic biomarkers of pancreatic cancer: a meta-analysis study. Oncotarget 2017; 8:68899-68915. [PMID: 28978166 PMCID: PMC5620306 DOI: 10.18632/oncotarget.20324] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 08/04/2017] [Indexed: 02/07/2023] Open
Abstract
Pancreatic cancer (PC) is an aggressive disease with high mortality rates, however, there is no blood test for early detection and diagnosis of this disease. Several research groups have reported on metabolomics based clinical investigations to identify biomarkers of PC, however there is a lack of a centralized metabolite biomarker repository that can be used for meta-analysis and biomarker validation. Furthermore, since the incidence of PC is associated with metabolic syndrome and Type 2 diabetes mellitus (T2DM), there is a need to uncouple these common metabolic dysregulations that may otherwise diminish the clinical utility of metabolomic biosignatures. Here, we attempted to externally replicate proposed metabolite biomarkers of PC reported by several other groups in an independent group of PC subjects. Our study design included a T2DM cohort that was used as a non-cancer control and a separate cohort diagnosed with colorectal cancer (CRC), as a cancer disease control to eliminate possible generic biomarkers of cancer. We used targeted mass spectrometry for quantitation of literature-curated metabolite markers and identified a biomarker panel that discriminates between normal controls (NC) and PC patients with high accuracy. Further evaluation of our model with CRC, however, showed a drop in specificity for the PC biomarker panel. Taken together, our study underscores the need for a more robust study design for cancer biomarker studies so as to maximize the translational value and clinical implementation.
Collapse
Affiliation(s)
- Khyati Y Mehta
- Department of Oncology, Georgetown University Medical Center, Washington, DC, United States of America
| | - Hung-Jen Wu
- Department of Biochemistry and Molecular and Cellular Biology, Georgetown University Medical Center, Washington, DC, United States of America
| | - Smrithi S Menon
- Department of Oncology, Georgetown University Medical Center, Washington, DC, United States of America
| | - Yassi Fallah
- Department of Oncology, Georgetown University Medical Center, Washington, DC, United States of America
| | - Xiaogang Zhong
- Department of Biostatistics Bioinformatics and Biomathematics, Georgetown University, Washington, DC, United States of America
| | - Nasser Rizk
- Department of Health Sciences, Qatar University, Doha, Qatar
| | - Keith Unger
- Lombardi Comprehensive Cancer Center, Med-Star Georgetown University Hospital, Washington, DC, United States of America
| | - Mark Mapstone
- Department of Neurology, University of California, Irvine, CA, United States of America
| | - Massimo S Fiandaca
- Department of Neurology, University of California, Irvine, CA, United States of America.,Department of Neurological Surgery, University of California, Irvine, CA, United States of America
| | - Howard J Federoff
- Department of Neurology, University of California, Irvine, CA, United States of America
| | - Amrita K Cheema
- Department of Oncology, Georgetown University Medical Center, Washington, DC, United States of America.,Department of Biochemistry and Molecular and Cellular Biology, Georgetown University Medical Center, Washington, DC, United States of America
| |
Collapse
|
9
|
Liu GF, Li GJ, Zhao H. Efficacy and Toxicity of Different Chemotherapy Regimens in the Treatment of Advanced or Metastatic Pancreatic Cancer: A Network Meta-Analysis. J Cell Biochem 2017; 119:511-523. [PMID: 28608558 DOI: 10.1002/jcb.26210] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Accepted: 06/12/2017] [Indexed: 12/29/2022]
Abstract
Objective A network meta-analysis was conducted to compare the efficacy and toxicity of different chemotherapy regimens in treating advanced or metastatic pancreatic cancer (PC). PubMed, Cochrane Library and EMBASE databases from inception to June 2016 were searched. A combination of direct and indirect evidences was referred to for calculating the weighted mean difference (WMD) or the odds ratio (OR) and to establish surface under the cumulative ranking (SUCRA) curves, so as to evaluate the efficacy and toxicity of different chemotherapy regimens in treating advanced or metastatic PC. Twenty randomized controlled trials were enrolled. Twelve chemotherapy regimens included Gemcitabine, S-1 (Tegafur), Gemcitabine + Cisplatin, Gemcitabine + Capecitabine, Gemcitabine + S-1, Gemcitabine + 5-FU (5-fluorouracil), Gemcitabine + Exatecan, Gemcitabine + Irinotecan, Gemcitabine + Nab-paclitaxel, FOLFIRINOX (Oxaliplatin + Irinotecan + Fluorouracil + Leucovorin), Gemcitabine + Oxaliplatin, and Gemcitabine + Pemetrexed. Higher overall response rate (ORR) was observed in patients treated with the gemcitabine + S-1 and FOLFIRINO regimens. Thrombocytopenia reduced in patients treated with the S-1 regimen. The Gemcitabine + S-1 and FOLFIRINO regimens had better short- and long-term efficacies than the other regimens; S-1 regimen had the lowest hematologic toxicity, while Gemcitabine + Nab-paclitaxel, FOLFIRINOX, and Gemcitabine + Pemetrexed regimens had higher incidence of non-hematologic toxicity among twelve chemotherapy regimens. The efficacy of Gemcitabine + S-1 and FOLFIRINOX regimens may be better in treating patients with advanced or metastatic pancreatic cancer, while FOLFIRINOX and Gemcitabine + Pemetrexed regimens may have relatively higher incidence of toxicity than other regimens. J. Cell. Biochem. 119: 511-523, 2018. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Gui-Feng Liu
- Department of Radiology, China-Japan Union Hospital of Jilin University, Changchun 130033, P.R. China
| | - Gui-Jie Li
- Department of ENT, China-Japan Union Hospital of Jilin University, Changchun 130033, P.R. China
| | - Hang Zhao
- Department of Neurosurgery, China-Japan Union Hospital of Jilin University, Changchun 130033, P.R. China
| |
Collapse
|
10
|
Nguyen HX, Do TNV, Le TH, Nguyen MTT, Nguyen NT, Esumi H, Awale S. Chemical Constituents of Mangifera indica and Their Antiausterity Activity against the PANC-1 Human Pancreatic Cancer Cell Line. JOURNAL OF NATURAL PRODUCTS 2016; 79:2053-2059. [PMID: 27466882 DOI: 10.1021/acs.jnatprod.6b00381] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Human pancreatic cancer cell lines such as PANC-1 have an altered metabolism, enabiling them to tolerate and survive under extreme conditions of nutrient starvation. The search for candidates that inhibit their viability during nutrition starvation represents a novel antiausterity strategy in anticancer drug discovery. A methanol extract of the bark of Mangifera indica was found to inhibit the survival of PANC-1 human pancreatic cancer cells preferentially under nutrient-deprived conditions with a PC50 value of 15.5 μg/mL, without apparent toxicity, in normal nutrient-rich conditions. Chemical investigation on this bioactive extract led to the isolation of 19 compounds (1-19), including two new cycloartane-type triterpenes, mangiferolate A (1) and mangiferolate B (2). The structures of 1 and 2 were determined by NMR spectroscopic analysis. Among the isolated compounds, mangiferolate B (2) and isoambolic acid (12) exhibited potent preferential cytotoxicity against PANC-1 human pancreatic cancer cells under the nutrition-deprived condition with PC50 values of 11.0 and 4.8 μM, respectively.
Collapse
Affiliation(s)
- Hai Xuan Nguyen
- Faculty of Chemistry, University of Science, Vietnam National University-Hochiminh City , 227 Nguyen Van Cu Street, District 5, Vietnam
| | - Truong Nhat Van Do
- Faculty of Chemistry, University of Science, Vietnam National University-Hochiminh City , 227 Nguyen Van Cu Street, District 5, Vietnam
| | - Tho Huu Le
- Faculty of Chemistry, University of Science, Vietnam National University-Hochiminh City , 227 Nguyen Van Cu Street, District 5, Vietnam
| | - Mai Thanh Thi Nguyen
- Faculty of Chemistry, University of Science, Vietnam National University-Hochiminh City , 227 Nguyen Van Cu Street, District 5, Vietnam
| | - Nhan Trung Nguyen
- Faculty of Chemistry, University of Science, Vietnam National University-Hochiminh City , 227 Nguyen Van Cu Street, District 5, Vietnam
| | - Hiroyasu Esumi
- Research Institute for Biomedical Sciences, Tokyo University of Science , Chiba 278-8510, Japan
| | - Suresh Awale
- Division of Natural Drug Discovery, Department of Translational Research, Institute of Natural Medicine, University of Toyama , 2630 Sugitani, Toyama 930-0194, Japan
| |
Collapse
|
11
|
Eskander MF, Bliss LA, Tseng JF. Pancreatic adenocarcinoma. Curr Probl Surg 2016; 53:107-54. [DOI: 10.1067/j.cpsurg.2016.01.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Accepted: 01/04/2016] [Indexed: 12/17/2022]
|
12
|
Zhang H, Zhou R, Jun M, Bacay AF, Eyring K, Webb A, Carrico-Moniz D. Identification of the Factors Responsible for the Selective in vitro Cytotoxic Activity of Isoprenylated Coumarin Derivatives under Nutrient-deprived Conditions. J Cancer 2016; 7:160-6. [PMID: 26819639 PMCID: PMC4716848 DOI: 10.7150/jca.13243] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 10/30/2015] [Indexed: 01/01/2023] Open
Abstract
Pancreatic cancer is one of the most devastating forms of human cancer. The lack of effective clinical treatments for pancreatic cancer has led to one of the lowest five-year survival rates among all cancers. Recently, our laboratory has developed a novel series of isoprenylated coumarin derivatives that have exhibited anti-pancreatic cancer activity exclusively under nutrient-deprived conditions. In this study, we report the effect of the various cell culture medium components on the preferential cytotoxicity of our lead isoprenylated coumarin compound against the pancreatic adenocarcinoma cell line PANC-1. In particular, our findings show a clear link between observed cytotoxicity and glucose deprivation, suggesting that our compound targets a salvage pathway when glycolysis is no longer an option for cancer cell survival. The cytotoxicity of our lead compound was also examined in vitro against two other pancreatic cancer cell lines, BxPC-3 and Capan-2 under both nutrient-rich and nutrient-deprived conditions.
Collapse
Affiliation(s)
- Hong Zhang
- 1. Department of Chemistry, Wellesley College, Wellesley, Massachusetts 02481, USA
| | - Ronghao Zhou
- 1. Department of Chemistry, Wellesley College, Wellesley, Massachusetts 02481, USA
| | - Maria Jun
- 1. Department of Chemistry, Wellesley College, Wellesley, Massachusetts 02481, USA
| | - Alyssa F Bacay
- 1. Department of Chemistry, Wellesley College, Wellesley, Massachusetts 02481, USA
| | - Katherine Eyring
- 1. Department of Chemistry, Wellesley College, Wellesley, Massachusetts 02481, USA
| | - Andrew Webb
- 2. Department of Biological Sciences, Wellesley College, Wellesley, Massachusetts 02481, USA
| | - Dora Carrico-Moniz
- 1. Department of Chemistry, Wellesley College, Wellesley, Massachusetts 02481, USA
| |
Collapse
|
13
|
Haab BB, Huang Y, Balasenthil S, Partyka K, Tang H, Anderson M, Allen P, Sasson A, Zeh H, Kaul K, Kletter D, Ge S, Bern M, Kwon R, Blasutig I, Srivastava S, Frazier ML, Sen S, Hollingsworth MA, Rinaudo JA, Killary AM, Brand RE. Definitive Characterization of CA 19-9 in Resectable Pancreatic Cancer Using a Reference Set of Serum and Plasma Specimens. PLoS One 2015; 10:e0139049. [PMID: 26431551 PMCID: PMC4592020 DOI: 10.1371/journal.pone.0139049] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 09/07/2015] [Indexed: 01/05/2023] Open
Abstract
The validation of candidate biomarkers often is hampered by the lack of a reliable means of assessing and comparing performance. We present here a reference set of serum and plasma samples to facilitate the validation of biomarkers for resectable pancreatic cancer. The reference set includes a large cohort of stage I-II pancreatic cancer patients, recruited from 5 different institutions, and relevant control groups. We characterized the performance of the current best serological biomarker for pancreatic cancer, CA 19–9, using plasma samples from the reference set to provide a benchmark for future biomarker studies and to further our knowledge of CA 19–9 in early-stage pancreatic cancer and the control groups. CA 19–9 distinguished pancreatic cancers from the healthy and chronic pancreatitis groups with an average sensitivity and specificity of 70–74%, similar to previous studies using all stages of pancreatic cancer. Chronic pancreatitis patients did not show CA 19–9 elevations, but patients with benign biliary obstruction had elevations nearly as high as the cancer patients. We gained additional information about the biomarker by comparing two distinct assays. The two CA 9–9 assays agreed well in overall performance but diverged in measurements of individual samples, potentially due to subtle differences in antibody specificity as revealed by glycan array analysis. Thus, the reference set promises be a valuable resource for biomarker validation and comparison, and the CA 19–9 data presented here will be useful for benchmarking and for exploring relationships to CA 19–9.
Collapse
Affiliation(s)
- Brian B. Haab
- Van Andel Research Institute, Grand Rapids, MI, United States of America
| | - Ying Huang
- Fred Hutchinson Cancer Research Center, Seattle, WA, United States of America
| | | | - Katie Partyka
- Van Andel Research Institute, Grand Rapids, MI, United States of America
| | - Huiyuan Tang
- Van Andel Research Institute, Grand Rapids, MI, United States of America
| | | | - Peter Allen
- Memorial Sloan Kettering Cancer Center, New York, NY, United States of America
| | - Aaron Sasson
- University of Nebraska Medical Center, Omaha, NE, United States of America
| | - Herbert Zeh
- University of Pittsburgh Medical Center, Pittsburgh, PA, United States of America
| | - Karen Kaul
- Northshore University Healthsystems, Evanston, IL, United States of America
| | - Doron Kletter
- Palo Alto Research Center, Palo Alto, CA, United States of America
| | - Shaokui Ge
- Fred Hutchinson Cancer Research Center, Seattle, WA, United States of America
| | - Marshall Bern
- Palo Alto Research Center, Palo Alto, CA, United States of America
| | - Richard Kwon
- University of Michigan, Ann Arbor, MI, United States of America
| | | | | | - Marsha L. Frazier
- The University of Texas MD Anderson Cancer Center, Houston, TX, United States of America
| | - Subrata Sen
- The University of Texas MD Anderson Cancer Center, Houston, TX, United States of America
| | | | - Jo Ann Rinaudo
- National Cancer Institute, Rockville, MD, United States of America
| | - Ann M. Killary
- The University of Texas MD Anderson Cancer Center, Houston, TX, United States of America
| | - Randall E. Brand
- University of Pittsburgh Medical Center, Pittsburgh, PA, United States of America
- * E-mail:
| |
Collapse
|
14
|
Inman M, Visconti A, Yan C, Siegel D, Ross D, Moody CJ. Antitumour indolequinones: synthesis and activity against human pancreatic cancer cells. Org Biomol Chem 2015; 12:4848-61. [PMID: 24848343 DOI: 10.1039/c4ob00711e] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
An important determinant of the growth inhibitory activity of indolequinones against pancreatic cancer cells is substitution on the 2-position with 2-unsubstituted derivatives being markedly more potent. A series of indolequinones bearing a range of substituents on nitrogen and at the indolylcarbinyl position was prepared by copper(II)-mediated reaction of bromoquinones and enamines, followed by functional group interconversions. The compounds were then assayed for their ability to inhibit the growth of pancreatic cancer cells. The pKa of the leaving group at the 3-position was shown to influence growth inhibitory activity that is consistent with the proposed mechanism of action of reduction, loss of leaving group and formation of a reactive iminium species. Substitutions on the indole nitrogen were well tolerated with little influence on growth inhibitory activity while substitutions at the 5- and 6-positions larger than methoxy led to decreased activity. The studies presented define the range of substitutions of 2-unsubstituted indolequinones required for optimal growth inhibitory activity.
Collapse
Affiliation(s)
- Martyn Inman
- School of Chemistry, University of Nottingham, University Park, Nottingham NG7 2RD, UK.
| | | | | | | | | | | |
Collapse
|
15
|
Nguyen MTT, Nguyen NT, Awale S. Prenylated Dihydrochalcones from Artocarpus altilis as Antiausterity Agents. ACTA ACUST UNITED AC 2015; 37:95-110. [DOI: 10.1016/bs.enz.2015.05.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2023]
|
16
|
Zhu C, Zhou GX. Inhibition of arachidonic acid metabolism to decrease pancreatic cancer cell proliferation. Shijie Huaren Xiaohua Zazhi 2014; 22:1106-1111. [DOI: 10.11569/wcjd.v22.i8.1106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the effect of inhibition of arachidonic acid metabolism on proliferation of pancreatic cancer cells.
METHODS: Cultured pancreatic cancer SW1990 cells were treated with different concentrations of MK886, celecoxib, or MK886 + celecoxib. After treatment, the expression of leukotriene B4 receptor 1 (BLT1) and vascular endothelial growth factor (VEGF) mRNAs was detected by semi-quantitative RT-PCR and the expression of phosphorylated extracellular regulated protein (p-Erk) was measured by Western blot.
RESULTS: Treatment with MK886 significantly decreased the expression of BLT1 and VEGF mRNAs (P < 0.01 for both) and p-Erk (P < 0.05). Treatment with celecoxib did not alter the expression of BLT1 mRNA and decreased the expression of VEGF mRNA compared with untreated cells (P < 0.01), but increased the expression of p-Erk compared with the MK886 group (P < 0.01). Treatment with MK886 and celecoxib significantly decreased the expression of BLT1 and VEGF mRNAs (P < 0.01 for both), but did not alter the expression of p-Erk.
CONCLUSION: Both two pathways of arachidonic acid metabolism are associated with pancreatic cancer cell proliferation, with the pathway involving 5-lipoxygenase being more important.
Collapse
|
17
|
Powerful inhibition of experimental human pancreatic cancers by receptor targeted cytotoxic LH-RH analog AEZS-108. Oncotarget 2014; 4:751-60. [PMID: 23744510 PMCID: PMC3742835 DOI: 10.18632/oncotarget.1044] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Pancreatic carcinoma is one of the cancers with the worse prognosis, thus any therapeutic improvement is imperative. Cytotoxic LH-RH analog, AN-152 (proprietary designation, AEZS-108), consisting of doxorubicin (DOX) conjugated to D-Lys⁶LH-RH, is now in clinical trials for targeted therapy of several sex hormone-dependent tumors that express LH-RH receptors. We investigated LH-RH receptors in human pancreatic carcinoma and the effects of AN-152 (AEZS-108) on experimental pancreatic cancers. We determined LH-RH receptor presence in human pancreatic cancer samples by immunohistochemistry and, in three human pancreatic cancer lines (SW-1990, Panc-1 and CFPAC-1), by binding assays and Western blotting. The effects of the cytotoxic LH-RH analog were investigated on growth of these same cancer lines xenografted into nude mice. We also analyzed differences between the antitumor effects of the cytotoxic analog and its cytotoxic radical alone, doxorubicin (DOX), on the expression of cancer-related genes by PCR arrays. LH-RH receptors were expressed in two randomly selected surgically removed human pancreatic cancer samples and in all three cancer lines. Cytotoxic LH-RH analogs powerfully inhibited growth of all three tumor lines in nude mice; AN-152 was significantly stronger than DOX on Panc-1 and CFPAC-1 cancers. PCR array showed that cytotoxic LH-RH analog AN-152 affected the expression of genes associated with cellular migration, invasion, metastasis and angiogenesis more favorably than DOX, however the changes in gene expression varied considerably among the three cancer lines. Cytotoxic LH-RH analog, AEZS-108, may be a useful agent for the treatment of LH-RH receptor positive advanced pancreatic carcinoma.
Collapse
|
18
|
Shakya B, Yadav PN, Ueda JY, Awale S. Discovery of 2-pyridineformamide thiosemicarbazones as potent antiausterity agents. Bioorg Med Chem Lett 2014; 24:458-61. [DOI: 10.1016/j.bmcl.2013.12.044] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Revised: 12/09/2013] [Accepted: 12/11/2013] [Indexed: 01/22/2023]
|
19
|
Ueda JY, Athikomkulchai S, Miyatake R, Saiki I, Esumi H, Awale S. (+)-Grandifloracin, an antiausterity agent, induces autophagic PANC-1 pancreatic cancer cell death. DRUG DESIGN DEVELOPMENT AND THERAPY 2013; 8:39-47. [PMID: 24379655 PMCID: PMC3872082 DOI: 10.2147/dddt.s52168] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Human pancreatic tumors are known to be highly resistant to nutrient starvation, and this prolongs their survival in the hypovascular (austere) tumor microenvironment. Agents that retard this tolerance to nutrient starvation represent a novel antiausterity strategy in anticancer drug discovery. (+)-Grandifloracin (GF), isolated from Uvaria dac, has shown preferential toxicity to PANC-1 human pancreatic cancer cells under nutrient starvation, with a PC50 value of 14.5 μM. However, the underlying mechanism is not clear. In this study, GF was found to preferentially induce PANC-1 cell death in a nutrient-deprived medium via hyperactivation of autophagy, as evidenced by a dramatic upregulation of microtubule-associated protein 1 light chain 3. No change was observed in expression of the caspase-3 and Bcl-2 apoptosis marker proteins. GF was also found to strongly inhibit the activation of Akt, a key regulator of cancer cell survival and proliferation. Because pancreatic tumors are highly resistant to current therapies that induce apoptosis, the alternative cell death mechanism exhibited by GF provides a novel therapeutic insight into antiausterity drug candidates.
Collapse
Affiliation(s)
- Jun-ya Ueda
- Frontier Research Core for Life Sciences, Toyama, Japan ; Institute of Natural Medicine, University of Toyama, Toyama, Japan
| | | | - Ryuta Miyatake
- Graduate School of Science and Engineering, University of Toyama, Toyama, Japan
| | - Ikuo Saiki
- Institute of Natural Medicine, University of Toyama, Toyama, Japan
| | - Hiroyasu Esumi
- Research Institute for Biomedical Sciences, Tokyo University of Science, Chiba, Japan ; National Cancer Center Hospital East, Chiba, Japan
| | - Suresh Awale
- Frontier Research Core for Life Sciences, Toyama, Japan ; Institute of Natural Medicine, University of Toyama, Toyama, Japan
| |
Collapse
|
20
|
Cleistanthane diterpenes from the seed of Caesalpinia sappan and their antiausterity activity against PANC-1 human pancreatic cancer cell line. Fitoterapia 2013; 91:148-153. [DOI: 10.1016/j.fitote.2013.08.018] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Revised: 08/24/2013] [Accepted: 08/26/2013] [Indexed: 11/22/2022]
|
21
|
Slee RB, Grimes BR, Bansal R, Gore J, Blackburn C, Brown L, Gasaway R, Jeong J, Victorino J, March KL, Colombo R, Herbert BS, Korc M. Selective inhibition of pancreatic ductal adenocarcinoma cell growth by the mitotic MPS1 kinase inhibitor NMS-P715. Mol Cancer Ther 2013; 13:307-315. [PMID: 24282275 DOI: 10.1158/1535-7163.mct-13-0324] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Most solid tumors, including pancreatic ductal adenocarcinoma (PDAC), exhibit structural and numerical chromosome instability (CIN). Although often implicated as a driver of tumor progression and drug resistance, CIN also reduces cell fitness and poses a vulnerability that can be exploited therapeutically. The spindle assembly checkpoint (SAC) ensures correct chromosome-microtubule attachment, thereby minimizing chromosome segregation errors. Many tumors exhibit upregulation of SAC components such as MPS1, which may help contain CIN within survivable limits. Prior studies showed that MPS1 inhibition with the small molecule NMS-P715 limits tumor growth in xenograft models. In cancer cell lines, NMS-P715 causes cell death associated with impaired SAC function and increased chromosome missegregation. Although normal cells appeared more resistant, effects on stem cells, which are the dose-limiting toxicity of most chemotherapeutics, were not examined. Elevated expression of 70 genes (CIN70), including MPS1, provides a surrogate measure of CIN and predicts poor patient survival in multiple tumor types. Our new findings show that the degree of CIN70 upregulation varies considerably among PDAC tumors, with higher CIN70 gene expression predictive of poor outcome. We identified a 25 gene subset (PDAC CIN25) whose overexpression was most strongly correlated with poor survival and included MPS1. In vitro, growth of human and murine PDAC cells is inhibited by NMS-P715 treatment, whereas adipose-derived human mesenchymal stem cells are relatively resistant and maintain chromosome stability upon exposure to NMS-P715. These studies suggest that NMS-P715 could have a favorable therapeutic index and warrant further investigation of MPS1 inhibition as a new PDAC treatment strategy.
Collapse
Affiliation(s)
- Roger B Slee
- Indiana University School of Medicine (IUSM) Department of Medical and Molecular Genetics,Indiana. Nerviano Medical Sciences, Nerviano, Italy.,IU Melvin and Bren Simon Cancer Center (IUSCC), Nerviano Medical Sciences, Nerviano, Italy
| | - Brenda R Grimes
- Indiana University School of Medicine (IUSM) Department of Medical and Molecular Genetics,Indiana. Nerviano Medical Sciences, Nerviano, Italy.,IU Melvin and Bren Simon Cancer Center (IUSCC), Nerviano Medical Sciences, Nerviano, Italy.,IUSCC Center for Pancreatic Cancer Research, Nerviano Medical Sciences, Nerviano, Italy
| | - Ruchi Bansal
- Indiana University School of Medicine (IUSM) Department of Medical and Molecular Genetics,Indiana. Nerviano Medical Sciences, Nerviano, Italy
| | - Jesse Gore
- IUSM Department of Medicine, Nerviano Medical Sciences, Nerviano, Italy
| | - Corinne Blackburn
- Indiana University School of Medicine (IUSM) Department of Medical and Molecular Genetics,Indiana. Nerviano Medical Sciences, Nerviano, Italy
| | - Lyndsey Brown
- Indiana University School of Medicine (IUSM) Department of Medical and Molecular Genetics,Indiana. Nerviano Medical Sciences, Nerviano, Italy
| | - Rachel Gasaway
- Indiana University School of Medicine (IUSM) Department of Medical and Molecular Genetics,Indiana. Nerviano Medical Sciences, Nerviano, Italy
| | - Jaesik Jeong
- IUSM Department of Biostatistics, Nerviano Medical Sciences, Nerviano, Italy
| | - Jose Victorino
- Indiana University School of Medicine (IUSM) Department of Medical and Molecular Genetics,Indiana. Nerviano Medical Sciences, Nerviano, Italy.,California State University Dominguez Hills, Nerviano Medical Sciences, Nerviano, Italy
| | - Keith L March
- IUSM Department of Medicine, Nerviano Medical Sciences, Nerviano, Italy.,IUSM Department of Biochemistry and Molecular Biology, Nerviano Medical Sciences, Nerviano, Italy.,Krannert Institute of Cardiology, Nerviano Medical Sciences, Nerviano, Italy.,Indiana Center for Vascular Biology, Nerviano Medical Sciences, Nerviano, Italy.,R.L. Roudebush Veterans Affairs Medical Center, Nerviano Medical Sciences, Nerviano, Italy
| | - Riccardo Colombo
- Indianapolis, Indiana. Nerviano Medical Sciences, Nerviano, Italy
| | - Brittney-Shea Herbert
- Indiana University School of Medicine (IUSM) Department of Medical and Molecular Genetics,Indiana. Nerviano Medical Sciences, Nerviano, Italy.,IU Melvin and Bren Simon Cancer Center (IUSCC), Nerviano Medical Sciences, Nerviano, Italy
| | - Murray Korc
- IU Melvin and Bren Simon Cancer Center (IUSCC), Nerviano Medical Sciences, Nerviano, Italy.,IUSCC Center for Pancreatic Cancer Research, Nerviano Medical Sciences, Nerviano, Italy.,IUSM Department of Medicine, Nerviano Medical Sciences, Nerviano, Italy.,IUSM Department of Biochemistry and Molecular Biology, Nerviano Medical Sciences, Nerviano, Italy
| |
Collapse
|
22
|
Tomizawa M, Shinozaki F, Sugiyama T, Yamamoto S, Sueishi M, Yoshida T. Frizzled-2: A potential novel target for molecular pancreatic cancer therapy. Oncol Lett 2013; 7:74-78. [PMID: 24348824 PMCID: PMC3861566 DOI: 10.3892/ol.2013.1681] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2013] [Accepted: 11/07/2013] [Indexed: 02/01/2023] Open
Abstract
In the present study, pancreatic cancer cell proliferation was analyzed following the suppression of frizzled (Fz)2 expression. Reverse transcription polymerase chain reaction (PCR) was performed using RNA isolated from pancreatic cancer cell lines, PANC-1, NOR-P1, PK-45H, PK-1, PK-59, MIA-Paca2 and KP4. A surgical specimen of pancreatic cancer was immunostained with antibodies specific to Fz2. Cell proliferation assays were performed with MIA-Paca2 cells transfected with small interfering RNA (siRNA) or short hairpin RNA (shRNA) of Fz2. Fz2 was found to be expressed in all pancreatic cancer cell lines, with the exception of NOR-P1. Immunostaining revealed that Fz2 was not expressed in normal pancreatic tissues, while it was expressed in pancreatic cancer cells. The expression levels of cyclin D1 were analyzed by quantitative PCR. The proliferation and expression of cyclin D1 were suppressed with the siRNA and shRNA of Fz2 in the MIA-Paca2 cells. Therefore, Fz2 is a potential target for the molecular therapy of pancreatic cancer.
Collapse
Affiliation(s)
- Minoru Tomizawa
- Department of Gastroenterology, National Hospital Organization, Shimoshizu Hospital, Yotsukaido, Chiba 284-0003, Japan
| | - Fuminobu Shinozaki
- Department of Radiology, National Hospital Organization, Shimoshizu Hospital, Yotsukaido, Chiba 284-0003, Japan
| | - Takao Sugiyama
- Department of Rheumatology, National Hospital Organization, Shimoshizu Hospital, Yotsukaido, Chiba 284-0003, Japan
| | - Shigenori Yamamoto
- Department of Pediatrics, National Hospital Organization, Shimoshizu Hospital, Yotsukaido, Chiba 284-0003, Japan
| | - Makoto Sueishi
- Department of Rheumatology, National Hospital Organization, Shimoshizu Hospital, Yotsukaido, Chiba 284-0003, Japan
| | - Takanobu Yoshida
- Department of Internal Medicine, National Hospital Organization, Shimoshizu Hospital, Yotsukaido, Chiba 284-0003, Japan
| |
Collapse
|
23
|
Hermann PC, Trabulo SM, Sainz B, Balic A, Garcia E, Hahn SA, Vandana M, Sahoo SK, Tunici P, Bakker A, Hidalgo M, Heeschen C. Multimodal Treatment Eliminates Cancer Stem Cells and Leads to Long-Term Survival in Primary Human Pancreatic Cancer Tissue Xenografts. PLoS One 2013; 8:e66371. [PMID: 23825539 PMCID: PMC3688976 DOI: 10.1371/journal.pone.0066371] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2012] [Accepted: 05/07/2013] [Indexed: 12/17/2022] Open
Abstract
Purpose In spite of intense research efforts, pancreatic ductal adenocarcinoma remains one of the most deadly malignancies in the world. We and others have previously identified a subpopulation of pancreatic cancer stem cells within the tumor as a critical therapeutic target and additionally shown that the tumor stroma represents not only a restrictive barrier for successful drug delivery, but also serves as a paracrine niche for cancer stem cells. Therefore, we embarked on a large-scale investigation on the effects of combining chemotherapy, hedgehog pathway inhibition, and mTOR inhibition in a preclinical mouse model of pancreatic cancer. Experimental Design Prospective and randomized testing in a set of almost 200 subcutaneous and orthotopic implanted whole-tissue primary human tumor xenografts. Results The combined targeting of highly chemoresistant cancer stem cells as well as their more differentiated progenies, together with abrogation of the tumor microenvironment by targeting the stroma and enhancing tissue penetration of the chemotherapeutic agent translated into significantly prolonged survival in preclinical models of human pancreatic cancer. Most pronounced therapeutic effects were observed in gemcitabine-resistant patient-derived tumors. Intriguingly, the proposed triple therapy approach could be further enhanced by using a PEGylated formulation of gemcitabine, which significantly increased its bioavailability and tissue penetration, resulting in a further improved overall outcome. Conclusions This multimodal therapeutic strategy should be further explored in the clinical setting as its success may eventually improve the poor prognosis of patients with pancreatic ductal adenocarcinoma.
Collapse
Affiliation(s)
- Patrick C. Hermann
- Stem Cells and Cancer Group, Clinical Research Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Sara M. Trabulo
- Stem Cells and Cancer Group, Clinical Research Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Bruno Sainz
- Stem Cells and Cancer Group, Clinical Research Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Anamaria Balic
- Stem Cells and Cancer Group, Clinical Research Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Elena Garcia
- Gastrointestinal Cancer Clinical Research Unit, Clinical Research Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Stephan A. Hahn
- Department of Molecular GI-Oncology, Ruhr-University Bochum, Bochum, Germany
| | - Mallaredy Vandana
- Nanomedicine Laboratory, Institute of Life Sciences, Bhubaneswar, India
| | - Sanjeeb K. Sahoo
- Nanomedicine Laboratory, Institute of Life Sciences, Bhubaneswar, India
| | | | - Annette Bakker
- Children’s Tumor Foundation, New York, New York, United States of America
| | - Manuel Hidalgo
- Gastrointestinal Cancer Clinical Research Unit, Clinical Research Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Christopher Heeschen
- Stem Cells and Cancer Group, Clinical Research Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
- * E-mail:
| |
Collapse
|
24
|
Biological Studies of Turmeric Oil, Part 2: Isolation and Characterization of Turmeric Oil Components; Cytotoxic Activity against Pancreatic Cancer Cells. Nat Prod Commun 2013. [DOI: 10.1177/1934578x1300800633] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
The current study is directed at identifying compounds that have biological activity in the less studied oil fraction of Curcuma longa (turmeric) to treat pancreatic cancer, which has no good treatment options. Fractional distillation and chromatographic separation of turmeric oil (TO) gave column fractions (CF) having biological activity against the PANC-1 pancreatic cancer cell line, with an EC50 in the range of 23 to 33 μg/mL. These fractions were analyzed by NMR and GCMS and found to contain the sesquiterpenes, ar-curcumene, 7-epi-zingiberene, β-sesquiphellandrene, curlone, α-turmerone, β-turmerone and ar-turmerone. The ability of TO components to induce cell death was independent of caspase activity. Potency was higher at lower cell density and was reduced by increasing serum concentration, the latter indicating serum binding of active components.
Collapse
|
25
|
Siegelin MD. Inhibition of the mitochondrial Hsp90 chaperone network: a novel, efficient treatment strategy for cancer? Cancer Lett 2013; 333:133-46. [PMID: 23376257 DOI: 10.1016/j.canlet.2013.01.045] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2013] [Revised: 01/23/2013] [Accepted: 01/24/2013] [Indexed: 12/17/2022]
Abstract
Research has shown that cancer cells exhibit multiple deregulated pathways, involving proliferation, migration and cell death. Heat-shock-proteins have evolved as "central regulators" and are implicated in the modulation of these pathways and in organelle-specific signaling. In this instance, heat-shock-proteins (Hsps) assist cancer cells in the maturation of proteins. Hsp90 is of particular interest because its enzymatic ATPase activity is elevated in malignant cells as compared to non-neoplastic counterparts. Consistent with its high-activity in cancer cells, Hsp90 stabilizes a considerable number of proteins being instrumental in carcinogenesis and the maintenance and growth of highly malignant cancers. Among its distribution Hsp90 is also localized within mitochondria of neoplastic cells of various origin, interacting with another chaperone, TRAP1 (Tumor necrosis factor type 1 receptor-associated protein or Heat-shock-protein 75) to antagonize the cell death promoting properties of the matrix protein, Cyclophilin-D. Several preclinical studies, including in vivo studies in both orthotopic and genetic animal models, have confirmed that targeting mitochondrial Hsp90 may be a novel efficient treatment method for highly recalcitrant tumors. This review summarizes the most recent findings of mitochondrial Hsp90 signaling and its potential implications for cancer therapy.
Collapse
Affiliation(s)
- Markus D Siegelin
- Department of Pathology & Cell Biology, Columbia University College of Physicians & Surgeons, 630 W. 168th Street, VC14-239, New York, NY 10032, USA.
| |
Collapse
|
26
|
Metronomic ceramide analogs inhibit angiogenesis in pancreatic cancer through up-regulation of caveolin-1 and thrombospondin-1 and down-regulation of cyclin D1. Neoplasia 2013; 14:833-45. [PMID: 23019415 DOI: 10.1593/neo.12772] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2012] [Revised: 07/30/2012] [Accepted: 07/30/2012] [Indexed: 02/07/2023] Open
Abstract
AIMS To evaluate the antitumor and antiangiogenic activity of metronomic ceramide analogs and their relevant molecular mechanisms. METHODS Human endothelial cells [human dermal microvascular endothelial cells and human umbilical vascular endothelial cell (HUVEC)] and pancreatic cancer cells (Capan-1 and MIA PaCa-2) were treated with the ceramide analogs (C2, AL6, C6, and C8), at low concentrations for 144 hours to evaluate any antiproliferative and proapoptotic effects and inhibition of migration and to measure the expression of caveolin-1 (CAV-1) and thrombospondin-1 (TSP-1) mRNAs by real-time reverse transcription-polymerase chain reaction. Assessment of extracellular signal-regulated kinases 1 and 2 (ERK1/2) and Akt phosphorylation and of CAV-1 and cyclin D1 protein expression was performed by ELISA. Maximum tolerated dose (MTD) gemcitabine was compared against metronomic doses of the ceramide analogs by evaluating the inhibition of MIA PaCa-2 subcutaneous tumor growth in nude mice. RESULTS Metronomic ceramide analogs preferentially inhibited cell proliferation and enhanced apoptosis in endothelial cells. Low concentrations of AL6 and C2 caused a significant inhibition of HUVEC migration. ERK1/2 and Akt phosphorylation were significantly decreased after metronomic ceramide analog treatment. Such treatment caused the overexpression of CAV-1 and TSP-1 mRNAs and proteins in endothelial cells, whereas cyclin D1 protein levels were reduced. The antiangiogenic and antitumor impact in vivo of metronomic C2 and AL6 regimens was similar to that caused by MTD gemcitabine. CONCLUSIONS Metronomic C2 and AL6 analogs have antitumor and antiangiogenic activity, determining the up-regulation of CAV-1 and TSP-1 and the suppression of cyclin D1.
Collapse
|
27
|
Preclinical evaluation of anticancer efficacy and pharmacological properties of FBA-TPQ, a novel synthetic makaluvamine analog. Mar Drugs 2012; 10:1138-1155. [PMID: 22822362 PMCID: PMC3397457 DOI: 10.3390/md10051138] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2012] [Revised: 05/02/2012] [Accepted: 05/16/2012] [Indexed: 12/02/2022] Open
Abstract
We have recently designed and synthesized a novel iminoquinone anticancer agent, 7-(4-fluorobenzylamino)-1,3,4,8-tetrahydropyrrolo[4,3,2-de]quinolin-8(1H)-one (FBA-TPQ) and initiated its preclinical development. Herein we investigated its efficacy, safety, and pharmacokinetics in in vitro and in vivo models of human pancreatic cancer. Our results demonstrated that FBA-TPQ inhibited pancreatic cancer cell growth, induced apoptosis, and caused cell cycle arrest in vitro. It inhibited the growth of xenograft tumors with minimal host toxicity. To facilitate future preclinical and clinical development of the agent, we also developed and validated a Rapid Resolution Liquid Chromatography (RRLC) method for quantitative analysis of FBA-TPQ in plasma and tissue samples. The method was found to be precise, accurate, and specific. Using this method, we carried out in vitro and in vivo evaluations of the pharmacological properties of FBA-TPQ, including stability in plasma, plasma protein binding, metabolism by S9 enzymes, plasma pharmacokinetics, and tissue distribution. Our results indicate that FBA-TPQ is a potential therapeutic agent for pancreatic cancer, providing a basis for future preclinical and clinical development.
Collapse
|
28
|
Lonardo E, Frias-Aldeguer J, Hermann PC, Heeschen C. Pancreatic stellate cells form a niche for cancer stem cells and promote their self-renewal and invasiveness. Cell Cycle 2012; 11:1282-90. [PMID: 22421149 DOI: 10.4161/cc.19679] [Citation(s) in RCA: 140] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Chronic pancreatitis and pancreatic ductal adenocarcinoma (PDAC) are characterized by extensive fibrosis. Importantly, in PDAC, this results in poor vascularization and impaired drug delivery to the cancer cells. Therefore, the combined targeting of pancreatic tumor stroma and chemotherapy should enhance response rates, but the negative outcome of a recent phase III clinical trial for the combination of chemotherapy and hedgehog pathway inhibition suggests that other means also need to be considered. Emerging data indicate that elimination of cancer stem cells as the root of the cancer is of pivotal importance for efficient treatment of pancreatic cancer. Recently, we demonstrated in a highly relevant preclinical mouse model for primary pancreatic cancers that the combination of cancer stem cell-targeting strategies in combination with a stroma-targeting agent, such as a hedgehog pathway inhibitor and chemotherapy, results in significantly enhanced long-term and progression-free survival. In the present study, we demonstrate mechanistically that Nodal-expressing pancreatic stellate cells are an important component of the tumor stroma for creating a paracrine niche for pancreatic cancer stem cells. Secretion of the embryonic morphogens Nodal/Activin by pancreatic stellate cells promoted in vitro sphere formation and invasiveness of pancreatic cancer stem cells in an Alk4-dependent manner. These data imply that the pancreatic cancer stem cell phenotype is promoted by paracrine Nodal/Activin signaling at the tumor-stroma interface. Therefore, targeting the tumor microenvironment is not only able to improve drug delivery but, even more importantly, destroys the cancer stem cell niche and, therefore, should be an integral part of cancer stem cell-based treatment strategies.
Collapse
Affiliation(s)
- Enza Lonardo
- Stem Cells & Cancer Group, Clinical Research Programme, Spanish National Cancer Research Centre, Madrid, Spain
| | | | | | | |
Collapse
|