51
|
Siraj AK, Kumar Parvathareddy S, Pratheeshkumar P, Padmaja Divya S, Ahmed SO, Melosantos R, Begum R, Concepcion RMJA, Al-Sanea N, Ashari LH, Abduljabbar A, Al-Dayel F, Al-Kuraya KS. APC truncating mutations in Middle Eastern Population: Tankyrase inhibitor is an effective strategy to sensitize APC mutant CRC To 5-FU chemotherapy. Biomed Pharmacother 2019; 121:109572. [PMID: 31704613 DOI: 10.1016/j.biopha.2019.109572] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 10/16/2019] [Accepted: 10/24/2019] [Indexed: 02/07/2023] Open
Abstract
Colorectal Cancer (CRC) is highly heterogeneous for which prognosis is dependent mainly on clinical staging. There is a need to stratify subpopulations of CRC on molecular basis to better predict outcome and therapy response. Truncating mutations in adenomatous polyposis coli (APC) are well-described events in CRC carcinogenesis. Clinical and genotypic characterization of Middle Eastern CRC based on presence and type of APC was determined in 412 CRC tumors using modern next generation sequencing. APC truncating mutations were identified in 58.2% (240/412) of CRCs. Overall, mutation was significant predictor of superior overall survival. Further, the type of APC mutations (short or long) did not have impact on clinical outcome. However, in vitro analysis showed difference between CRC cell lines carrying short truncating APC vs CRC cells that carry long truncating APC mutation in response to 5-flourouracil (5-FU). Importantly, we were able to overcome the resistance to 5-FU seen in CRC cells carrying short APC by tankyrase inhibitor, XAV939, thereby inhibiting Wnt/β-catenin signaling cascade. Overall, our results showed that APC mutation status plays an important role in predicting overall survival in Middle Eastern population. Furthermore, in vitro data showed that selective targeting of APC mutated CRC by tankyrase inhibitor can be an effective strategy to overcome 5-FU resistance in CRC cells.
Collapse
Affiliation(s)
- Abdul K Siraj
- Human Cancer Genomic Research, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | | | - Poyil Pratheeshkumar
- Human Cancer Genomic Research, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Sasidharan Padmaja Divya
- Human Cancer Genomic Research, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Saeeda Omer Ahmed
- Human Cancer Genomic Research, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Roxanne Melosantos
- Human Cancer Genomic Research, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Rafia Begum
- Human Cancer Genomic Research, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | | | - Nasser Al-Sanea
- Department of Surgery, Colorectal Unit, Riyadh, Saudi Arabia
| | - Luai H Ashari
- Department of Surgery, Colorectal Unit, Riyadh, Saudi Arabia
| | | | - Fouad Al-Dayel
- Department of Pathology, King Faisal Specialist Hospital and Research Centre, P.O. Box 3354, Riyadh, 11211, Saudi Arabia
| | - Khawla S Al-Kuraya
- Human Cancer Genomic Research, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia.
| |
Collapse
|
52
|
Braun JA, Herrmann AL, Blase JI, Frensemeier K, Bulkescher J, Scheffner M, Galy B, Hoppe-Seyler K, Hoppe-Seyler F. Effects of the antifungal agent ciclopirox in HPV-positive cancer cells: Repression of viral E6/E7 oncogene expression and induction of senescence and apoptosis. Int J Cancer 2019; 146:461-474. [PMID: 31603527 DOI: 10.1002/ijc.32709] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 09/05/2019] [Accepted: 09/18/2019] [Indexed: 01/02/2023]
Abstract
The malignant growth of human papillomavirus (HPV)-positive cancer cells is dependent on the continuous expression of the viral E6/E7 oncogenes. Here, we examined the effects of iron deprivation on the phenotype of HPV-positive cervical cancer cells. We found that iron chelators, such as the topical antifungal agent ciclopirox (CPX), strongly repress HPV E6/E7 oncogene expression, both at the transcript and protein level. CPX efficiently blocks the proliferation of HPV-positive cancer cells by inducing cellular senescence. Although active mTOR signaling is considered to be critical for the cellular senescence response towards a variety of prosenescent agents, CPX-induced senescence occurs under conditions of severely impaired mTOR signaling. Prolonged CPX treatment leads to p53-independent Caspase-3/7 activation and induction of apoptosis. CPX also eliminates HPV-positive cancer cells under hypoxic conditions through induction of apoptosis. Taken together, these results show that iron deprivation exerts profound antiviral and antiproliferative effects in HPV-positive cancer cells and suggest that iron chelators, such as CPX, possess therapeutic potential as HPV-inhibitory, prosenescent and proapoptotic agents in both normoxic and hypoxic environments.
Collapse
Affiliation(s)
- Julia A Braun
- Molecular Therapy of Virus-Associated Cancers, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Anja L Herrmann
- Molecular Therapy of Virus-Associated Cancers, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Johanna I Blase
- Molecular Therapy of Virus-Associated Cancers, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Kristin Frensemeier
- Molecular Therapy of Virus-Associated Cancers, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Julia Bulkescher
- Molecular Therapy of Virus-Associated Cancers, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Martin Scheffner
- Department of Biology, University of Konstanz, Konstanz, Germany
| | - Bruno Galy
- Virus-Associated Carcinogenesis, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Karin Hoppe-Seyler
- Molecular Therapy of Virus-Associated Cancers, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Felix Hoppe-Seyler
- Molecular Therapy of Virus-Associated Cancers, German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
53
|
Wang F, Lv H, Zhao B, Zhou L, Wang S, Luo J, Liu J, Shang P. Iron and leukemia: new insights for future treatments. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:406. [PMID: 31519186 PMCID: PMC6743129 DOI: 10.1186/s13046-019-1397-3] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 08/27/2019] [Indexed: 01/19/2023]
Abstract
Iron, an indispensable element for life, is involved in all kinds of important physiological activities. Iron promotes cell growth and proliferation, but it also causes oxidative stress damage. The body has a strict regulation mechanism of iron metabolism due to its potential toxicity. As a cancer of the bone marrow and blood cells, leukemia threatens human health seriously. Current studies suggest that dysregulation of iron metabolism and subsequent accumulation of excess iron are closely associated with the occurrence and progress of leukemia. Specifically, excess iron promotes the development of leukemia due to the pro-oxidative nature of iron and its damaging effects on DNA. On the other hand, leukemia cells acquire large amounts of iron to maintain rapid growth and proliferation. Therefore, targeting iron metabolism may provide new insights for approaches to the treatment of leukemia. This review summarizes physiologic iron metabolism, alternations of iron metabolism in leukemia and therapeutic opportunities of targeting the altered iron metabolism in leukemia, with a focus on acute leukemia.
Collapse
Affiliation(s)
- Fang Wang
- School of Life Science, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Huanhuan Lv
- School of Life Science, Northwestern Polytechnical University, Xi'an, 710072, China.,Research & Development Institute of Northwestern Polytechnical University in Shenzhen, Shenzhen, 518057, China.,Key Laboratory for Space Bioscience and Biotechnology, Institute of Special Environment Biophysics, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Bin Zhao
- School of Life Science, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Liangfu Zhou
- School of Life Science, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Shenghang Wang
- School of Life Science, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Jie Luo
- School of Life Science, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Junyu Liu
- School of Life Science, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Peng Shang
- Research & Development Institute of Northwestern Polytechnical University in Shenzhen, Shenzhen, 518057, China. .,Key Laboratory for Space Bioscience and Biotechnology, Institute of Special Environment Biophysics, Northwestern Polytechnical University, Xi'an, 710072, China.
| |
Collapse
|
54
|
Evaluation of the Cytotoxic Effect of Hydroxypyridinone Derivatives on HCT116 and SW480 Colon Cancer Cell Lines. Pharm Chem J 2019. [DOI: 10.1007/s11094-019-02010-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
55
|
Corton JC, Witt KL, Yauk CL. Identification of p53 Activators in a Human Microarray Compendium. Chem Res Toxicol 2019; 32:1748-1759. [PMID: 31397557 DOI: 10.1021/acs.chemrestox.9b00052] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Biomarkers predictive of molecular and toxicological effects are needed to interpret emerging high-throughput transcriptomic data streams. The previously characterized 63 gene TGx-DDI biomarker that includes 20 genes known to be regulated by p53 was previously shown to accurately predict DNA damage in chemically treated cells. We comprehensively evaluated whether the molecular basis of the DDI predictions was based on a p53-dependent response. The biomarker was compared to microarray data in a compendium derived from human cells using the Running Fisher test, a nonparametric correlation test. Using the biomarker, we identified conditions that led to p53 activation, including exposure to the chemical nutlin-3 which disrupts interactions between p53 and the negative regulator MDM2 or by knockdown of MDM2. The expression of most of the genes in the biomarker (75%) were found to depend on p53 activation status based on gene behavior after TP53 overexpression or knockdown. The biomarker identified DDI chemicals that were strong inducers of p53 in wild-type cells; these p53 responses were decreased or abolished in cells after p53 knockdown by siRNAs. Using the biomarker, we screened ∼1950 chemicals in ∼9800 human cell line chemical vs control comparisons and identified ∼100 chemicals that caused p53 activation. Among the positive chemicals were many that are known to activate p53 through direct and indirect DNA damaging mechanisms. These results contribute to the evidence that the TGx-DDI biomarker is useful for identifying chemicals that cause DDI and activate p53.
Collapse
Affiliation(s)
- J Christopher Corton
- Integrated Systems Toxicology Division, NHEERL , United States Environmental Protection Agency , Research Triangle Park, Durham , North Carolina 27711 , United States
| | - Kristine L Witt
- Division of the National Toxicology Program , National Institute of Environmental Health Sciences , Research Triangle Park, Durham , North Carolina 27709 , United States
| | - Carole L Yauk
- Environmental Health Science and Research Bureau, Health Canada , Ottawa , Ontario K1A 0K9 , Canada
| |
Collapse
|
56
|
Weir SJ, Wood R, Schorno K, Brinker AE, Ramamoorthy P, Heppert K, Rajewski L, Tanol M, Ham T, McKenna MJ, McCulloch W, Dalton M, Reed GA, Jensen RA, Baltezor MJ, Anant S, Taylor JA. Preclinical Pharmacokinetics of Fosciclopirox, a Novel Treatment of Urothelial Cancers, in Rats and Dogs. J Pharmacol Exp Ther 2019; 370:148-159. [PMID: 31113837 PMCID: PMC6614794 DOI: 10.1124/jpet.119.257972] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 05/08/2019] [Indexed: 12/20/2022] Open
Abstract
Pharmacokinetic studies in rats and dogs were performed to characterize the in vivo performance of a novel prodrug, fosciclopirox. Ciclopirox olamine (CPX-O) is a marketed topical antifungal agent with demonstrated in vitro and in vivo preclinical anticancer activity in several solid tumor and hematologic malignancies. The oral route of administration for CPX-O is not feasible due to low bioavailability and dose-limiting gastrointestinal toxicities. To enable parenteral administration, the phosphoryl-oxymethyl ester of ciclopirox (CPX), fosciclopirox (CPX-POM), was synthesized and formulated as an injectable drug product. In rats and dogs, intravenous CPX-POM is rapidly and completely metabolized to its active metabolite, CPX. The bioavailability of the active metabolite is complete following CPX-POM administration. CPX and its inactive metabolite, ciclopirox glucuronide (CPX-G), are excreted in urine, resulting in delivery of drug to the entire urinary tract. The absolute bioavailability of CPX following subcutaneous administration of CPX-POM is excellent in rats and dogs, demonstrating the feasibility of this route of administration. These studies confirmed the oral bioavailability of CPX-O is quite low in rats and dogs compared with intravenous CPX-POM. Given its broad-spectrum anticancer activity in several solid tumor and hematologic cancers and renal elimination, CPX-POM is being developed for the treatment of urothelial cancer. The safety, dose tolerance, pharmacokinetics, and pharmacodynamics of intravenous CPX-POM are currently being characterized in a United States multicenter first-in-human Phase 1 clinical trial in patients with advanced solid tumors (NCT03348514).
Collapse
Affiliation(s)
- Scott J Weir
- University of Kansas Cancer Center, Kansas City, Kansas (S.J.W., R.W., A.E.B., G.A.R., R.A.J., M.J.B., S.A., J.A.T.); Institute for Advancing Medical Innovation (S.J.W., R.W., A.E.B., M.J.B.) and Departments of Cancer Biology (S.J.W., P.R., S.A.), Pharmacology, Toxicology, and Therapeutics (S.J.W., G.A.R.) Pathology (R.A.J.), and Urology (J.A.T.), University of Kansas Medical Center, Kansas City, Kansas; Biotechnology Innovation and Optimization Center, University of Kansas, Lawrence, Kansas (K.S., K.H., L.R., M.T., M.J.B.); School of Pharmacy, Istanbul Kemerburgaz University, Istanbul, Turkey (M.T.); CicloMed LLC, Kansas City, Missouri (T.H.); Navigator LSA, Wilmington, North Carolina (M.J.M.); Alba BioPharm Advisors Inc., Durham, North Carolina (W.M.); and The Gnomon Group, Carrboro, North Carolina (M.D.)
| | - Robyn Wood
- University of Kansas Cancer Center, Kansas City, Kansas (S.J.W., R.W., A.E.B., G.A.R., R.A.J., M.J.B., S.A., J.A.T.); Institute for Advancing Medical Innovation (S.J.W., R.W., A.E.B., M.J.B.) and Departments of Cancer Biology (S.J.W., P.R., S.A.), Pharmacology, Toxicology, and Therapeutics (S.J.W., G.A.R.) Pathology (R.A.J.), and Urology (J.A.T.), University of Kansas Medical Center, Kansas City, Kansas; Biotechnology Innovation and Optimization Center, University of Kansas, Lawrence, Kansas (K.S., K.H., L.R., M.T., M.J.B.); School of Pharmacy, Istanbul Kemerburgaz University, Istanbul, Turkey (M.T.); CicloMed LLC, Kansas City, Missouri (T.H.); Navigator LSA, Wilmington, North Carolina (M.J.M.); Alba BioPharm Advisors Inc., Durham, North Carolina (W.M.); and The Gnomon Group, Carrboro, North Carolina (M.D.)
| | - Karl Schorno
- University of Kansas Cancer Center, Kansas City, Kansas (S.J.W., R.W., A.E.B., G.A.R., R.A.J., M.J.B., S.A., J.A.T.); Institute for Advancing Medical Innovation (S.J.W., R.W., A.E.B., M.J.B.) and Departments of Cancer Biology (S.J.W., P.R., S.A.), Pharmacology, Toxicology, and Therapeutics (S.J.W., G.A.R.) Pathology (R.A.J.), and Urology (J.A.T.), University of Kansas Medical Center, Kansas City, Kansas; Biotechnology Innovation and Optimization Center, University of Kansas, Lawrence, Kansas (K.S., K.H., L.R., M.T., M.J.B.); School of Pharmacy, Istanbul Kemerburgaz University, Istanbul, Turkey (M.T.); CicloMed LLC, Kansas City, Missouri (T.H.); Navigator LSA, Wilmington, North Carolina (M.J.M.); Alba BioPharm Advisors Inc., Durham, North Carolina (W.M.); and The Gnomon Group, Carrboro, North Carolina (M.D.)
| | - Amanda E Brinker
- University of Kansas Cancer Center, Kansas City, Kansas (S.J.W., R.W., A.E.B., G.A.R., R.A.J., M.J.B., S.A., J.A.T.); Institute for Advancing Medical Innovation (S.J.W., R.W., A.E.B., M.J.B.) and Departments of Cancer Biology (S.J.W., P.R., S.A.), Pharmacology, Toxicology, and Therapeutics (S.J.W., G.A.R.) Pathology (R.A.J.), and Urology (J.A.T.), University of Kansas Medical Center, Kansas City, Kansas; Biotechnology Innovation and Optimization Center, University of Kansas, Lawrence, Kansas (K.S., K.H., L.R., M.T., M.J.B.); School of Pharmacy, Istanbul Kemerburgaz University, Istanbul, Turkey (M.T.); CicloMed LLC, Kansas City, Missouri (T.H.); Navigator LSA, Wilmington, North Carolina (M.J.M.); Alba BioPharm Advisors Inc., Durham, North Carolina (W.M.); and The Gnomon Group, Carrboro, North Carolina (M.D.)
| | - Prabhu Ramamoorthy
- University of Kansas Cancer Center, Kansas City, Kansas (S.J.W., R.W., A.E.B., G.A.R., R.A.J., M.J.B., S.A., J.A.T.); Institute for Advancing Medical Innovation (S.J.W., R.W., A.E.B., M.J.B.) and Departments of Cancer Biology (S.J.W., P.R., S.A.), Pharmacology, Toxicology, and Therapeutics (S.J.W., G.A.R.) Pathology (R.A.J.), and Urology (J.A.T.), University of Kansas Medical Center, Kansas City, Kansas; Biotechnology Innovation and Optimization Center, University of Kansas, Lawrence, Kansas (K.S., K.H., L.R., M.T., M.J.B.); School of Pharmacy, Istanbul Kemerburgaz University, Istanbul, Turkey (M.T.); CicloMed LLC, Kansas City, Missouri (T.H.); Navigator LSA, Wilmington, North Carolina (M.J.M.); Alba BioPharm Advisors Inc., Durham, North Carolina (W.M.); and The Gnomon Group, Carrboro, North Carolina (M.D.)
| | - Kathy Heppert
- University of Kansas Cancer Center, Kansas City, Kansas (S.J.W., R.W., A.E.B., G.A.R., R.A.J., M.J.B., S.A., J.A.T.); Institute for Advancing Medical Innovation (S.J.W., R.W., A.E.B., M.J.B.) and Departments of Cancer Biology (S.J.W., P.R., S.A.), Pharmacology, Toxicology, and Therapeutics (S.J.W., G.A.R.) Pathology (R.A.J.), and Urology (J.A.T.), University of Kansas Medical Center, Kansas City, Kansas; Biotechnology Innovation and Optimization Center, University of Kansas, Lawrence, Kansas (K.S., K.H., L.R., M.T., M.J.B.); School of Pharmacy, Istanbul Kemerburgaz University, Istanbul, Turkey (M.T.); CicloMed LLC, Kansas City, Missouri (T.H.); Navigator LSA, Wilmington, North Carolina (M.J.M.); Alba BioPharm Advisors Inc., Durham, North Carolina (W.M.); and The Gnomon Group, Carrboro, North Carolina (M.D.)
| | - Lian Rajewski
- University of Kansas Cancer Center, Kansas City, Kansas (S.J.W., R.W., A.E.B., G.A.R., R.A.J., M.J.B., S.A., J.A.T.); Institute for Advancing Medical Innovation (S.J.W., R.W., A.E.B., M.J.B.) and Departments of Cancer Biology (S.J.W., P.R., S.A.), Pharmacology, Toxicology, and Therapeutics (S.J.W., G.A.R.) Pathology (R.A.J.), and Urology (J.A.T.), University of Kansas Medical Center, Kansas City, Kansas; Biotechnology Innovation and Optimization Center, University of Kansas, Lawrence, Kansas (K.S., K.H., L.R., M.T., M.J.B.); School of Pharmacy, Istanbul Kemerburgaz University, Istanbul, Turkey (M.T.); CicloMed LLC, Kansas City, Missouri (T.H.); Navigator LSA, Wilmington, North Carolina (M.J.M.); Alba BioPharm Advisors Inc., Durham, North Carolina (W.M.); and The Gnomon Group, Carrboro, North Carolina (M.D.)
| | - Mehmet Tanol
- University of Kansas Cancer Center, Kansas City, Kansas (S.J.W., R.W., A.E.B., G.A.R., R.A.J., M.J.B., S.A., J.A.T.); Institute for Advancing Medical Innovation (S.J.W., R.W., A.E.B., M.J.B.) and Departments of Cancer Biology (S.J.W., P.R., S.A.), Pharmacology, Toxicology, and Therapeutics (S.J.W., G.A.R.) Pathology (R.A.J.), and Urology (J.A.T.), University of Kansas Medical Center, Kansas City, Kansas; Biotechnology Innovation and Optimization Center, University of Kansas, Lawrence, Kansas (K.S., K.H., L.R., M.T., M.J.B.); School of Pharmacy, Istanbul Kemerburgaz University, Istanbul, Turkey (M.T.); CicloMed LLC, Kansas City, Missouri (T.H.); Navigator LSA, Wilmington, North Carolina (M.J.M.); Alba BioPharm Advisors Inc., Durham, North Carolina (W.M.); and The Gnomon Group, Carrboro, North Carolina (M.D.)
| | - Tammy Ham
- University of Kansas Cancer Center, Kansas City, Kansas (S.J.W., R.W., A.E.B., G.A.R., R.A.J., M.J.B., S.A., J.A.T.); Institute for Advancing Medical Innovation (S.J.W., R.W., A.E.B., M.J.B.) and Departments of Cancer Biology (S.J.W., P.R., S.A.), Pharmacology, Toxicology, and Therapeutics (S.J.W., G.A.R.) Pathology (R.A.J.), and Urology (J.A.T.), University of Kansas Medical Center, Kansas City, Kansas; Biotechnology Innovation and Optimization Center, University of Kansas, Lawrence, Kansas (K.S., K.H., L.R., M.T., M.J.B.); School of Pharmacy, Istanbul Kemerburgaz University, Istanbul, Turkey (M.T.); CicloMed LLC, Kansas City, Missouri (T.H.); Navigator LSA, Wilmington, North Carolina (M.J.M.); Alba BioPharm Advisors Inc., Durham, North Carolina (W.M.); and The Gnomon Group, Carrboro, North Carolina (M.D.)
| | - Michael J McKenna
- University of Kansas Cancer Center, Kansas City, Kansas (S.J.W., R.W., A.E.B., G.A.R., R.A.J., M.J.B., S.A., J.A.T.); Institute for Advancing Medical Innovation (S.J.W., R.W., A.E.B., M.J.B.) and Departments of Cancer Biology (S.J.W., P.R., S.A.), Pharmacology, Toxicology, and Therapeutics (S.J.W., G.A.R.) Pathology (R.A.J.), and Urology (J.A.T.), University of Kansas Medical Center, Kansas City, Kansas; Biotechnology Innovation and Optimization Center, University of Kansas, Lawrence, Kansas (K.S., K.H., L.R., M.T., M.J.B.); School of Pharmacy, Istanbul Kemerburgaz University, Istanbul, Turkey (M.T.); CicloMed LLC, Kansas City, Missouri (T.H.); Navigator LSA, Wilmington, North Carolina (M.J.M.); Alba BioPharm Advisors Inc., Durham, North Carolina (W.M.); and The Gnomon Group, Carrboro, North Carolina (M.D.)
| | - William McCulloch
- University of Kansas Cancer Center, Kansas City, Kansas (S.J.W., R.W., A.E.B., G.A.R., R.A.J., M.J.B., S.A., J.A.T.); Institute for Advancing Medical Innovation (S.J.W., R.W., A.E.B., M.J.B.) and Departments of Cancer Biology (S.J.W., P.R., S.A.), Pharmacology, Toxicology, and Therapeutics (S.J.W., G.A.R.) Pathology (R.A.J.), and Urology (J.A.T.), University of Kansas Medical Center, Kansas City, Kansas; Biotechnology Innovation and Optimization Center, University of Kansas, Lawrence, Kansas (K.S., K.H., L.R., M.T., M.J.B.); School of Pharmacy, Istanbul Kemerburgaz University, Istanbul, Turkey (M.T.); CicloMed LLC, Kansas City, Missouri (T.H.); Navigator LSA, Wilmington, North Carolina (M.J.M.); Alba BioPharm Advisors Inc., Durham, North Carolina (W.M.); and The Gnomon Group, Carrboro, North Carolina (M.D.)
| | - Michael Dalton
- University of Kansas Cancer Center, Kansas City, Kansas (S.J.W., R.W., A.E.B., G.A.R., R.A.J., M.J.B., S.A., J.A.T.); Institute for Advancing Medical Innovation (S.J.W., R.W., A.E.B., M.J.B.) and Departments of Cancer Biology (S.J.W., P.R., S.A.), Pharmacology, Toxicology, and Therapeutics (S.J.W., G.A.R.) Pathology (R.A.J.), and Urology (J.A.T.), University of Kansas Medical Center, Kansas City, Kansas; Biotechnology Innovation and Optimization Center, University of Kansas, Lawrence, Kansas (K.S., K.H., L.R., M.T., M.J.B.); School of Pharmacy, Istanbul Kemerburgaz University, Istanbul, Turkey (M.T.); CicloMed LLC, Kansas City, Missouri (T.H.); Navigator LSA, Wilmington, North Carolina (M.J.M.); Alba BioPharm Advisors Inc., Durham, North Carolina (W.M.); and The Gnomon Group, Carrboro, North Carolina (M.D.)
| | - Gregory A Reed
- University of Kansas Cancer Center, Kansas City, Kansas (S.J.W., R.W., A.E.B., G.A.R., R.A.J., M.J.B., S.A., J.A.T.); Institute for Advancing Medical Innovation (S.J.W., R.W., A.E.B., M.J.B.) and Departments of Cancer Biology (S.J.W., P.R., S.A.), Pharmacology, Toxicology, and Therapeutics (S.J.W., G.A.R.) Pathology (R.A.J.), and Urology (J.A.T.), University of Kansas Medical Center, Kansas City, Kansas; Biotechnology Innovation and Optimization Center, University of Kansas, Lawrence, Kansas (K.S., K.H., L.R., M.T., M.J.B.); School of Pharmacy, Istanbul Kemerburgaz University, Istanbul, Turkey (M.T.); CicloMed LLC, Kansas City, Missouri (T.H.); Navigator LSA, Wilmington, North Carolina (M.J.M.); Alba BioPharm Advisors Inc., Durham, North Carolina (W.M.); and The Gnomon Group, Carrboro, North Carolina (M.D.)
| | - Roy A Jensen
- University of Kansas Cancer Center, Kansas City, Kansas (S.J.W., R.W., A.E.B., G.A.R., R.A.J., M.J.B., S.A., J.A.T.); Institute for Advancing Medical Innovation (S.J.W., R.W., A.E.B., M.J.B.) and Departments of Cancer Biology (S.J.W., P.R., S.A.), Pharmacology, Toxicology, and Therapeutics (S.J.W., G.A.R.) Pathology (R.A.J.), and Urology (J.A.T.), University of Kansas Medical Center, Kansas City, Kansas; Biotechnology Innovation and Optimization Center, University of Kansas, Lawrence, Kansas (K.S., K.H., L.R., M.T., M.J.B.); School of Pharmacy, Istanbul Kemerburgaz University, Istanbul, Turkey (M.T.); CicloMed LLC, Kansas City, Missouri (T.H.); Navigator LSA, Wilmington, North Carolina (M.J.M.); Alba BioPharm Advisors Inc., Durham, North Carolina (W.M.); and The Gnomon Group, Carrboro, North Carolina (M.D.)
| | - Michael J Baltezor
- University of Kansas Cancer Center, Kansas City, Kansas (S.J.W., R.W., A.E.B., G.A.R., R.A.J., M.J.B., S.A., J.A.T.); Institute for Advancing Medical Innovation (S.J.W., R.W., A.E.B., M.J.B.) and Departments of Cancer Biology (S.J.W., P.R., S.A.), Pharmacology, Toxicology, and Therapeutics (S.J.W., G.A.R.) Pathology (R.A.J.), and Urology (J.A.T.), University of Kansas Medical Center, Kansas City, Kansas; Biotechnology Innovation and Optimization Center, University of Kansas, Lawrence, Kansas (K.S., K.H., L.R., M.T., M.J.B.); School of Pharmacy, Istanbul Kemerburgaz University, Istanbul, Turkey (M.T.); CicloMed LLC, Kansas City, Missouri (T.H.); Navigator LSA, Wilmington, North Carolina (M.J.M.); Alba BioPharm Advisors Inc., Durham, North Carolina (W.M.); and The Gnomon Group, Carrboro, North Carolina (M.D.)
| | - Shrikant Anant
- University of Kansas Cancer Center, Kansas City, Kansas (S.J.W., R.W., A.E.B., G.A.R., R.A.J., M.J.B., S.A., J.A.T.); Institute for Advancing Medical Innovation (S.J.W., R.W., A.E.B., M.J.B.) and Departments of Cancer Biology (S.J.W., P.R., S.A.), Pharmacology, Toxicology, and Therapeutics (S.J.W., G.A.R.) Pathology (R.A.J.), and Urology (J.A.T.), University of Kansas Medical Center, Kansas City, Kansas; Biotechnology Innovation and Optimization Center, University of Kansas, Lawrence, Kansas (K.S., K.H., L.R., M.T., M.J.B.); School of Pharmacy, Istanbul Kemerburgaz University, Istanbul, Turkey (M.T.); CicloMed LLC, Kansas City, Missouri (T.H.); Navigator LSA, Wilmington, North Carolina (M.J.M.); Alba BioPharm Advisors Inc., Durham, North Carolina (W.M.); and The Gnomon Group, Carrboro, North Carolina (M.D.)
| | - John A Taylor
- University of Kansas Cancer Center, Kansas City, Kansas (S.J.W., R.W., A.E.B., G.A.R., R.A.J., M.J.B., S.A., J.A.T.); Institute for Advancing Medical Innovation (S.J.W., R.W., A.E.B., M.J.B.) and Departments of Cancer Biology (S.J.W., P.R., S.A.), Pharmacology, Toxicology, and Therapeutics (S.J.W., G.A.R.) Pathology (R.A.J.), and Urology (J.A.T.), University of Kansas Medical Center, Kansas City, Kansas; Biotechnology Innovation and Optimization Center, University of Kansas, Lawrence, Kansas (K.S., K.H., L.R., M.T., M.J.B.); School of Pharmacy, Istanbul Kemerburgaz University, Istanbul, Turkey (M.T.); CicloMed LLC, Kansas City, Missouri (T.H.); Navigator LSA, Wilmington, North Carolina (M.J.M.); Alba BioPharm Advisors Inc., Durham, North Carolina (W.M.); and The Gnomon Group, Carrboro, North Carolina (M.D.)
| |
Collapse
|
57
|
Basu S, Ghosh A, Barui A, Basu B. Epithelial cell functionality on electroconductive Fe/Sr co-doped biphasic calcium phosphate. J Biomater Appl 2019; 33:1035-1052. [PMID: 30630385 DOI: 10.1177/0885328218821549] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
In the perspective of dental restorative applications, co-doped bioceramics have not been explored much. From the clinical perspective, a successful dental implant is expected to interact with peri-prosthetic bones, gingival tissue, and surrounding connective tissues. The interaction of implant and implant coating materials with bone tissue is well studied. However, their interaction with surrounding epithelial components needs scientific validation. In this context, the present study aims at quantitative evaluation of the electrical properties of Fe/Sr co-doped biphasic calcium phosphate (BCP) samples and assessment of their cytocompatibility with epithelial (vero) cells. Sr/Fe co-doped BCPs were prepared by sol-gel synthesis technique, with different dopant concentration. Impact of co-doping on conductivity was assessed and interestingly an increase in conductivity with dopant amount was recorded in different co-doped BCPs. Cellular study showed the significant ( p = 0.01) increase in both cellular viability and functionality with increasing conductivity of samples. Higher epithelial cell adhesion indicates that (Sr/Fe) co-doped BCP would be favorable for faster epithelial sealing and also would reduce the chances of infection. Real-time PCR and immunofluorescence studies indicated that the expression of the epithelial marker (E-cadherin) significantly ( p = 0.01) increased in 10, 30 and 40 mol% co-doped samples in comparison to undoped BCP. In contrast to E-cadherin, fold change of β-catenin remains unchanged amongst the co-doped ceramics, implying the absence of tumorigenic potential of (Sr/Fe) co-doped BCP. In addition, immune-fluorescence signatures for cellular polarity are established from enhanced expression PARD3 protein, which has major relevance for cellular morphogenesis and cell division. Summarizing, the present study establishes the efficacy of Sr/Fe co-doped BCPs as a dental implant coating material and its ability to modulate vero cell functionality.
Collapse
Affiliation(s)
- Subhadip Basu
- 1 Laboratory for Biomaterials, Materials Research Center, Indian Institute of Science, Bangalore, India
| | - Aritri Ghosh
- 2 Centre for Healthcare Science and Technology, Indian Institute of Engineering Science and Technology, Howrah, India
| | - Ananya Barui
- 2 Centre for Healthcare Science and Technology, Indian Institute of Engineering Science and Technology, Howrah, India
| | - Bikramjit Basu
- 1 Laboratory for Biomaterials, Materials Research Center, Indian Institute of Science, Bangalore, India.,3 Center for Biosystems Science and Engineering, Indian Institute of Science, Bangalore, India
| |
Collapse
|
58
|
Zhang MW, Zhao P, Yung WH, Sheng Y, Ke Y, Qian ZM. Tissue iron is negatively correlated with TERC or TERT mRNA expression: A heterochronic parabiosis study in mice. Aging (Albany NY) 2018; 10:3834-3850. [PMID: 30555055 PMCID: PMC6326661 DOI: 10.18632/aging.101676] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 11/18/2018] [Indexed: 12/21/2022]
Abstract
To test the hypothesis that iron accumulation in tissues with age is a key harmful factor for the development of aging, we established heterochronic parabiosis-pairings and investigated changes in serum iron, the expression of major iron transport proteins and iron contents, as well as telomerase reverse transcriptase (TERT), telomerase RNA component (TERC), and telomere length in the liver, kidney and heart of Y-O(O) (old pairing with young), Y-O(Y) (young pairing with old), O-O (pairings between two old) and Y-Y (pairings between two young) mice. We demonstrated that the reduced serum iron, increased iron and reduced expression of TERT and TERC in the tissues of aged mice are reversible by exposure to a younger mouse’s circulation. All of these measurements in young mice are reversible by exposure to an older mouse’s circulation. Correlation analysis showed that tissue iron is negatively correlated with TERT and TERC expression in the liver, kidney and heart of parabiotic mice. These findings provide new evidence for the key role of iron in aging and also imply the existence of rejuvenating factors in young serum with an anti-ageing role that act by reversing the impaired activity of iron metabolism in old mice.
Collapse
Affiliation(s)
- Meng-Wan Zhang
- National Clinical Research Center for Aging and Medicine, Huashan Hostital, Laboratory of Neuropharmacology, School of Pharmacy, Fudan University, Shanghai 201203, PRC
| | - Peng Zhao
- National Clinical Research Center for Aging and Medicine, Huashan Hostital, Laboratory of Neuropharmacology, School of Pharmacy, Fudan University, Shanghai 201203, PRC
| | - Wing-Ho Yung
- School of Biomedical Sciences and Gerald Choa Neuroscience Centre, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, NT, Hong Kong
| | - Yuan Sheng
- National Clinical Research Center for Aging and Medicine, Huashan Hostital, Laboratory of Neuropharmacology, School of Pharmacy, Fudan University, Shanghai 201203, PRC
| | - Ya Ke
- School of Biomedical Sciences and Gerald Choa Neuroscience Centre, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, NT, Hong Kong
| | - Zhong-Ming Qian
- National Clinical Research Center for Aging and Medicine, Huashan Hostital, Laboratory of Neuropharmacology, School of Pharmacy, Fudan University, Shanghai 201203, PRC.,Laboratory of Neuropharmacology, Institute of Translational & Precision Medicine, Nantong University, Nantong 226019, PRC
| |
Collapse
|
59
|
Al-Dali AM, Weiher H, Schmidt-Wolf IGH. Utilizing ethacrynic acid and ciclopirox olamine in liver cancer. Oncol Lett 2018; 16:6854-6860. [PMID: 30405829 DOI: 10.3892/ol.2018.9472] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 08/30/2018] [Indexed: 01/20/2023] Open
Abstract
Once aberrantly activated, the Wnt/β-catenin pathway may result in uncontrolled proliferation and eventually cancer. Efforts to counter and inhibit this pathway are mainly directed against β-catenin, as it serves a role on the cytoplasm and the nucleus. In addition, specially-generated lymphocytes are recruited for the purpose of treating liver cancer. Peripheral blood mononuclear lymphocytes are expanded by the timely addition of interferon γ, interleukin (IL)-1β, IL-2 and anti-cluster of differentiation 3 antibody. The resulting cells are called cytokine-induced killer (CIK) cells. The present study utilised these cells and combine them with drugs inhibiting the Wnt pathway in order to examine whether this resulted in an improvement in the killing ability of CIK cells against liver cancer cells. Drugs including ethacrynic acid (EA) and ciclopirox olamine (CPX) were determined to be suitable candidates, as determined by previous studies. Drugs were administered on their own and combined with CIK cells and then a cell viability assay was performed. These results suggest that EA-treated cells demonstrated apoptosis and were significantly affected compared with untreated cells. Unlike EA, CPX killed normal and cancerous cells even at low concentrations. Subsequent to combining EA with CIK cells, the potency of killing was increased and a greater number of cells died, which proves a synergistic action. In summary, EA may be used as an anti-hepatocellular carcinoma drug, while CPX possesses a high toxicity to cancerous as well as to normal cells. It was proposed that EA should be integrated into present therapeutic methods for cancer.
Collapse
Affiliation(s)
- Ahmad M Al-Dali
- Center for Integrated Oncology, University Hospital Bonn, Bonn D-53105, Germany.,Department of Immunology and Cell Biology, Bonn-Rhein-Sieg University of Applied Sciences, Rheinbach D-53359, Germany
| | - Hans Weiher
- Department of Immunology and Cell Biology, Bonn-Rhein-Sieg University of Applied Sciences, Rheinbach D-53359, Germany
| | | |
Collapse
|
60
|
Han M, Xu R, Wang S, Yang N, Ni S, Zhang Q, Xu Y, Zhang X, Zhang C, Wei Y, Ji J, Huang B, Zhang D, Chen A, Li W, Bjerkvig R, Li X, Wang J. Six-Transmembrane Epithelial Antigen of Prostate 3 Predicts Poor Prognosis and Promotes Glioblastoma Growth and Invasion. Neoplasia 2018; 20:543-554. [PMID: 29730475 PMCID: PMC5994776 DOI: 10.1016/j.neo.2018.04.002] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2017] [Revised: 12/20/2017] [Accepted: 04/02/2018] [Indexed: 01/20/2023] Open
Abstract
Recent evidence suggests that dysregulation of iron regulatory factors may play essential roles in cancer pathophysiology. Six-transmembrane epithelial antigen of prostate 3 (STEAP3) is a metalloreductase, which is vital for cellular iron uptake and homeostasis. However, the clinical significance and function of STEAP3 in the development of human gliomas remain unclear. Through analysis of publicly available databases, we found that STEAP3 was highly expressed in malignant gliomas, especially in the mesenchymal glioma molecular subtype and isocitrate dehydrogenase 1/2 (IDH1/2) wild-type gliomas. Expression levels of STEAP3 in gliomas correlated inversely with patient overall survival (OS) and served as an independent prognostic marker by multivariate Cox regression analysis. In functional assays performed with RNA knockdown, loss of STEAP3 attenuated aggressive phenotypes in glioma cells, including cell proliferation, invasion, and sphere formation in vitro and tumor growth in vivo. Finally, STEAP3 drives these activities by inducing mesenchymal transition, promoting transferrin receptor (TfR) expression, and activating STAT3-FoxM1 axis signaling. Taken together, these results indicate that STEAP3 functions as an oncogenic mediator in glioma progression and is thus a potential therapeutic target for the treatment of the disease.
Collapse
Affiliation(s)
- Mingzhi Han
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, Key Laboratory of Brain Functional Remodeling, Shandong, 107# Wenhua Xi Road, Jinan, 250012, China; K G Jebsen Brain Tumor Research Center, Department of Biomedicine, University of Bergen, 5009, Bergen, Norway
| | - Ran Xu
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, Key Laboratory of Brain Functional Remodeling, Shandong, 107# Wenhua Xi Road, Jinan, 250012, China; Brain and Mind Center, Sydney Medical School and Faculty of Health Sciences, University of Sydney, Sydney, NSW, 2050, Australia
| | - Shuai Wang
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, Key Laboratory of Brain Functional Remodeling, Shandong, 107# Wenhua Xi Road, Jinan, 250012, China
| | - Ning Yang
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, Key Laboratory of Brain Functional Remodeling, Shandong, 107# Wenhua Xi Road, Jinan, 250012, China
| | - Shilei Ni
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, Key Laboratory of Brain Functional Remodeling, Shandong, 107# Wenhua Xi Road, Jinan, 250012, China
| | - Qing Zhang
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, Key Laboratory of Brain Functional Remodeling, Shandong, 107# Wenhua Xi Road, Jinan, 250012, China
| | - Yangyang Xu
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, Key Laboratory of Brain Functional Remodeling, Shandong, 107# Wenhua Xi Road, Jinan, 250012, China
| | - Xin Zhang
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, Key Laboratory of Brain Functional Remodeling, Shandong, 107# Wenhua Xi Road, Jinan, 250012, China
| | - Chao Zhang
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, Key Laboratory of Brain Functional Remodeling, Shandong, 107# Wenhua Xi Road, Jinan, 250012, China
| | - Yuzhen Wei
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, Key Laboratory of Brain Functional Remodeling, Shandong, 107# Wenhua Xi Road, Jinan, 250012, China; Department of Neurosurgery, Jining No.1 People's Hospital, Jiankang Road, Jining, 272011, China
| | - Jianxiong Ji
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, Key Laboratory of Brain Functional Remodeling, Shandong, 107# Wenhua Xi Road, Jinan, 250012, China
| | - Bin Huang
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, Key Laboratory of Brain Functional Remodeling, Shandong, 107# Wenhua Xi Road, Jinan, 250012, China
| | - Di Zhang
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, Key Laboratory of Brain Functional Remodeling, Shandong, 107# Wenhua Xi Road, Jinan, 250012, China
| | - Anjing Chen
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, Key Laboratory of Brain Functional Remodeling, Shandong, 107# Wenhua Xi Road, Jinan, 250012, China
| | - Wenjie Li
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, Key Laboratory of Brain Functional Remodeling, Shandong, 107# Wenhua Xi Road, Jinan, 250012, China
| | - Rolf Bjerkvig
- K G Jebsen Brain Tumor Research Center, Department of Biomedicine, University of Bergen, 5009, Bergen, Norway
| | - Xingang Li
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, Key Laboratory of Brain Functional Remodeling, Shandong, 107# Wenhua Xi Road, Jinan, 250012, China.
| | - Jian Wang
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, Key Laboratory of Brain Functional Remodeling, Shandong, 107# Wenhua Xi Road, Jinan, 250012, China; K G Jebsen Brain Tumor Research Center, Department of Biomedicine, University of Bergen, 5009, Bergen, Norway.
| |
Collapse
|
61
|
Shen T, Huang S. Repositioning the Old Fungicide Ciclopirox for New Medical Uses. Curr Pharm Des 2018; 22:4443-50. [PMID: 27238364 DOI: 10.2174/1381612822666160530151209] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2016] [Accepted: 05/27/2016] [Indexed: 02/06/2023]
Abstract
BACKGROUND Ciclopirox (CPX) has been used as an antifungal agent in various formulations to treat superficial fungal infection for decades. Its effectiveness and safety in treatments have been demonstrated by multiple studies. METHODS Here we briefly summarize the pharmacological and toxicological properties of CPX as an antifungal agent, the new medical uses of CPX, as well as the correspondent molecular mechanisms. RESULTS Increasing evidence has demonstrated that CPX is able to inhibit tumor growth, ameliorate diabetes and its complications, prevent human immunodeficiency virus (HIV) infection, and improve age-associated cardiovascular defects. Interestingly, its antifungal activity and all those newly observed effects are more or less related to its capability of chelating iron and interfering with the related signaling pathways. Mechanistically, CPX is capable of modulating the activities of certain enzymes or signaling pathways, such as ribonucleotide reductase (RR), deoxyhypusine hydroxylase (DOHH)/eukaryotic translation initiation factor 5A (eIF5A), Wnt/β-catenin, hypoxia-inducible factor-1α (HIF-1 α)/vascular endothelial growth factor (VEGF), vascular endothelial growth factor receptor 3 (VEGFR-3)/extracellular signal-regulated protein kinases 1/2, mammalian target of rapamycin, and cyclin dependent kinases (CDKs). Most of these activities are related to its chelation of iron. CONCLUSION CPX, as an antifungal agent, may be repositioned for treatment of cancer and other human diseases.
Collapse
Affiliation(s)
| | - Shile Huang
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA 71130-3932, USA
| |
Collapse
|
62
|
Ibrahim H, Temerk Y, Farhan N. A novel sensor based on nanobiocomposite Au-In2O3-chitosan modified acetylene black paste electrode for sensitive detection of antimycotic ciclopirox olamine. Talanta 2018; 179:75-85. [DOI: 10.1016/j.talanta.2017.10.036] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 10/18/2017] [Accepted: 10/20/2017] [Indexed: 10/18/2022]
|
63
|
Mihailidou C, Papakotoulas P, Papavassiliou AG, Karamouzis MV. Superior efficacy of the antifungal agent ciclopirox olamine over gemcitabine in pancreatic cancer models. Oncotarget 2018; 9:10360-10374. [PMID: 29535812 PMCID: PMC5828195 DOI: 10.18632/oncotarget.23164] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 11/17/2017] [Indexed: 12/15/2022] Open
Abstract
Ciclopirox olamine (CPX) is an antifungal agent that has recently demonstrated promising anti-neoplastic activity against hematologic and solid tumors. Here, we evaluated CPX compared with gemcitabine alone as well as their combination in human pancreatic cancer cell lines; BxPC-3, Panc-1, and MIA PaCa-2 and in humanized xenograft mouse models. We also examined the preclinical pharmacodynamic activity of CPX. CPX caused a pronounced decrease in cell proliferation and clonogenic growth potential. These inhibitory effects were accompanied by induction of reactive oxygen species (ROS), which were strongly associated with reduced Bcl-xL and survivin levels and activation of a panel of caspases, especially caspase-3, and finally resulted in apoptotic death. CPX-induced apoptosis was associated with reduced pEGFR (Y1068) and pAkt (Ser473) protein levels. Additionally, decreased proliferation was observed in CPX-treated xenografts tumors, demonstrating unique tumor regression and a profound survival benefit. Finally, we showed that CPX significantly abrogated gemcitabine-induced ROS levels in pancreatic tissues. These pre-clinical results have verified the superior antitumor efficacy of CPX over gemcitabine alone, while their combination is even more effective, providing the rationale for further clinical testing of CPX plus gemcitabine in pancreatic cancer patients.
Collapse
Affiliation(s)
- Chrysovalantou Mihailidou
- Molecular Oncology Unit, Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Pavlos Papakotoulas
- Molecular Oncology Unit, Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
- 2 Department of Medical Oncology, Theagenion Hospital, 54007 Thessaloniki, Greece
| | - Athanasios G. Papavassiliou
- Molecular Oncology Unit, Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Michalis V. Karamouzis
- Molecular Oncology Unit, Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
- First Department of Internal Medicine, Laiko Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| |
Collapse
|
64
|
Kamihara Y, Takada K, Sato T, Kawano Y, Murase K, Arihara Y, Kikuchi S, Hayasaka N, Usami M, Iyama S, Miyanishi K, Sato Y, Kobune M, Kato J. The iron chelator deferasirox induces apoptosis by targeting oncogenic Pyk2/β-catenin signaling in human multiple myeloma. Oncotarget 2018; 7:64330-64341. [PMID: 27602957 PMCID: PMC5325446 DOI: 10.18632/oncotarget.11830] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 08/21/2016] [Indexed: 01/19/2023] Open
Abstract
Deregulated iron metabolism underlies the pathogenesis of many human cancers. Recently, low expression of ferroportin, which is the only identified non-heme iron exporter, has been associated with significantly reduced overall survival in multiple myeloma (MM); however, the altered iron metabolism in MM biology remains unclear. In this study we demonstrated, by live cell imaging, that MM cells have increased intracellular iron levels as compared with normal cells. In experiments to test the effect of iron chelation on the growth of MM cells, we found that deferasirox (DFX), an oral iron chelator used to treat iron overload in clinical practice, inhibits MM cell growth both in vivo and in vitro. Mechanistically, DFX was found to induce apoptosis of MM cells via the inhibition of proline-rich tyrosine kinase 2 (Pyk2), which is known to promote tumor growth in MM. Inhibition of Pyk2 is caused by the suppression of reactive oxygen species, and leads to downregulation of the Wnt/β-catenin signaling pathway. Taken together, our findings indicate that high levels of intracellular iron, which might be due to low ferroportin expression, play a role in MM pathophysiology. Therefore, DFX may provide a therapeutic option for MM that is driven by deregulated iron homeostasis and/or Pyk2/Wnt signaling.
Collapse
Affiliation(s)
- Yusuke Kamihara
- Department of Medical Oncology and Hematology, Sapporo Medical University School of Medicine, Japan
| | - Kohichi Takada
- Department of Medical Oncology and Hematology, Sapporo Medical University School of Medicine, Japan
| | - Tsutomu Sato
- Department of Medical Oncology and Hematology, Sapporo Medical University School of Medicine, Japan
| | - Yutaka Kawano
- Department of Medical Oncology and Hematology, Sapporo Medical University School of Medicine, Japan
| | - Kazuyuki Murase
- Department of Medical Oncology and Hematology, Sapporo Medical University School of Medicine, Japan
| | - Yohei Arihara
- Department of Medical Oncology and Hematology, Sapporo Medical University School of Medicine, Japan
| | - Shohei Kikuchi
- Department of Medical Oncology and Hematology, Sapporo Medical University School of Medicine, Japan
| | - Naotaka Hayasaka
- Department of Medical Oncology and Hematology, Sapporo Medical University School of Medicine, Japan
| | - Makoto Usami
- Department of Medical Oncology and Hematology, Sapporo Medical University School of Medicine, Japan
| | - Satoshi Iyama
- Department of Medical Oncology and Hematology, Sapporo Medical University School of Medicine, Japan
| | - Koji Miyanishi
- Department of Medical Oncology and Hematology, Sapporo Medical University School of Medicine, Japan
| | - Yasushi Sato
- Department of Medical Oncology and Hematology, Sapporo Medical University School of Medicine, Japan
| | - Masayoshi Kobune
- Department of Medical Oncology and Hematology, Sapporo Medical University School of Medicine, Japan
| | - Junji Kato
- Department of Medical Oncology and Hematology, Sapporo Medical University School of Medicine, Japan
| |
Collapse
|
65
|
Shen T, Zhou H, Shang C, Luo Y, Wu Y, Huang S. Ciclopirox activates ATR-Chk1 signaling pathway leading to Cdc25A protein degradation. Genes Cancer 2018; 9:39-52. [PMID: 29725502 PMCID: PMC5931253 DOI: 10.18632/genesandcancer.166] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 02/11/2018] [Indexed: 02/05/2023] Open
Abstract
Ciclopirox olamine (CPX), an off-patent anti-fungal drug, has been found to inhibit the G1-cyclin dependent kinases partly by increasing the phosphorylation and degradation of Cdc25A. However, little is known about the molecular target(s) of CPX responsible for Cdc25A degradation. Here, we show that CPX induced the degradation of Cdc25A neither by increasing CK1α or decreasing DUB3 expression, nor via activating GSK3β, but through activating Chk1 in rhabdomyosarcoma (Rh30) and breast carcinoma (MDA-MB-231) cells. This is strongly supported by the findings that inhibition of Chk1 with TCS2312 or knockdown of Chk1 profoundly attenuated CPX-induced Cdc25A degradation in the cells. Furthermore, we observed that CPX caused DNA damage, which was independent of reactive oxygen species (ROS) induction, but related to iron chelation. CPX treatment resulted in the activation of ataxia telangiectasia mutated (ATM) and ATM-and RAD3-related (ATR) kinases. Treatment with Ku55933 (a selective ATM inhibitor) failed to prevent CPX-induced Chk1 phosphorylation and Cdc25A degradation. In contrast, knockdown of ATR conferred high resistance to CPX-induced Chk1 phosphorylation and Cdc25A degradation. Therefore, the results suggest that CPX-induced degradation of Cdc25A is attributed to the activation of ATR-Chk1 signaling pathway, a consequence of iron chelation-induced DNA damage.
Collapse
Affiliation(s)
- Tao Shen
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
- Feist-Weiller Cancer Center, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Hongyu Zhou
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Chaowei Shang
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
- Feist-Weiller Cancer Center, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Yan Luo
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
- State Key Laboratory of Biotherapy / Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, P.R. China
| | - Yang Wu
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
- State Key Laboratory of Biotherapy / Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, P.R. China
| | - Shile Huang
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
- Feist-Weiller Cancer Center, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| |
Collapse
|
66
|
Raggi C, Gammella E, Correnti M, Buratti P, Forti E, Andersen JB, Alpini G, Glaser S, Alvaro D, Invernizzi P, Cairo G, Recalcati S. Dysregulation of Iron Metabolism in Cholangiocarcinoma Stem-like Cells. Sci Rep 2017; 7:17667. [PMID: 29247214 PMCID: PMC5732280 DOI: 10.1038/s41598-017-17804-1] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 11/26/2017] [Indexed: 12/13/2022] Open
Abstract
Cholangiocarcinoma (CCA) is a devastating liver tumour arising from malignant transformation of bile duct epithelial cells. Cancer stem cells (CSC) are a subset of tumour cells endowed with stem-like properties, which play a role in tumour initiation, recurrence and metastasis. In appropriate conditions, CSC form 3D spheres (SPH), which retain stem-like tumour-initiating features. Here, we found different expression of iron proteins indicating increased iron content, oxidative stress and higher expression of CSC markers in CCA-SPH compared to tumour cells growing as monolayers. Exposure to the iron chelator desferrioxamine decreased SPH forming efficiency and the expression of CSC markers and stem-like genes, whereas iron had an opposite effect. Microarray profiles in CCA samples (n = 104) showed decreased H ferritin, hepcidin and ferroportin expression in tumours respect to surrounding liver, whereas transferrin receptor was up-regulated. Moreover, we found a trend toward poorer outcome in CCA patients with elevated expression of ferritin and hepcidin, two major proteins of iron metabolism. These findings, which represent the first evidence of a role for iron in the stem cell compartment as a novel metabolic factor involved in CCA growth, may have implications for a better therapeutic approach.
Collapse
Affiliation(s)
- Chiara Raggi
- Center for Autoimmune Liver Diseases, Humanitas Clinical and Research Center, Rozzano, Italy
- Dipartimento Medicina Sperimentale e Clinica, University of Firence, Firenze, Italy
| | - Elena Gammella
- Department of Biomedical Sciences for Health, University of Milan, Milano, Italy
| | - Margherita Correnti
- Center for Autoimmune Liver Diseases, Humanitas Clinical and Research Center, Rozzano, Italy
| | - Paolo Buratti
- Department of Biomedical Sciences for Health, University of Milan, Milano, Italy
| | - Elisa Forti
- Center for Autoimmune Liver Diseases, Humanitas Clinical and Research Center, Rozzano, Italy
| | - Jesper B Andersen
- Biotech Research and Innovation Centre, Department of Health and Medical Sciences University of Copenhagen, Copenhagen, Denmark
| | - Gianfranco Alpini
- Research, Central Texas Veterans Health Care System, Baylor Scott & White Digestive Disease Research Center, Scott & White Health, Department of Medicine, Texas A&M Health Science Center, Temple, TX, USA
| | - Shannon Glaser
- Research, Central Texas Veterans Health Care System, Baylor Scott & White Digestive Disease Research Center, Scott & White Health, Department of Medicine, Texas A&M Health Science Center, Temple, TX, USA
| | - Domenico Alvaro
- Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Rome, Italy
| | - Pietro Invernizzi
- Division of Gastroenterology and Program for Autoimmune Liver Diseases, International Center for Digestive Health, Department of Medicine and Surgery, University of Milan-Bicocca, Monza, Italy.
| | - Gaetano Cairo
- Department of Biomedical Sciences for Health, University of Milan, Milano, Italy.
| | - Stefania Recalcati
- Department of Biomedical Sciences for Health, University of Milan, Milano, Italy
| |
Collapse
|
67
|
Jiang W, Li Y, Ou J, Wang X, Zhang C, Yi L, Xue L, Zhang M. Expression analysis of E-cad and vascular endothelial growth factor in triple-negative breast cancer patients of different ethnic groups in western China. Medicine (Baltimore) 2017; 96:e8155. [PMID: 29049198 PMCID: PMC5662364 DOI: 10.1097/md.0000000000008155] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The aim of this article is to investigate the expression of E-cadherin (E-cad) and vascular endothelial growth factor (VEGF) in triple-negative breast cancer (TNBC) of Han and Uygur women patients in western China, and their relationship with clinical features of TNBC.Totally, 172 cases of Han TNBC patients and 79 cases of Uighur TNBC patients were enrolled. The expressions of E-cad and VEGF were detected with immunohistochemistry. The correlation of E-cad and VEGF expression with lymph node metastasis, TNM stage, and histological grade were analyzed. The 5-year disease-free survival rate of the 2 groups was also evaluated.There was no significant difference in the 5-year disease-free survival rate (P > .05) and the expression of E-cad between the 2 groups. The positive rate of VEGF in Han was significantly lower than that in Uygur (P < .05). The expression of E-cad was negatively correlated with lymph node metastasis, TNM stage, and histological grade (-1≤r < 1, P < .05). However, the expression of VEGF was positively correlated with lymph node metastasis and TNM staging (0 < r < 1, P < 0.05), but not with histological grading.The expression of E-cad and VEGF and their relationship with clinical features of TNBC suggest that Uygur TNBC patients might have different prognostic factors as compared with Han patients.
Collapse
|
68
|
Cheng Q, Liu L, Fu Y, Zhang Y, Yang Y, Liu J. RhPDCD5 combined with dexamethasone increases antitumor activity in multiple myeloma partially via inhibiting the Wnt signalling pathway. Clin Exp Pharmacol Physiol 2017; 45:140-145. [PMID: 28945941 DOI: 10.1111/1440-1681.12859] [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: 06/10/2017] [Revised: 09/12/2017] [Accepted: 09/18/2017] [Indexed: 11/29/2022]
Abstract
Multiple myeloma (MM) is one of the most common hematological malignancies and characterized by the clonal accumulation of malignant plasma cells. Significant progress has been made in MM treatment recently, while MM still remains incurable. Our previous studies showed that the recombined human programmed cell death 5 (rhPDCD5) can promote MM apoptosis induced by dexamethasone (Dex). Here, we expanded the findings by showing that the rhPDCD5 alone could not induce an obvious growth inhibition of U266 cells (a MM cell line). Of note, with the combination of dexamethasone (Dex), the growth of MM cells was significantly inhibited and accompanied with the cell cycle arrest in G0/G1. For mechanism study, we found that the combination treatment of rhPDCD5 plus Dex downregulated the mRNA and protein expressions of Wnt effectors including β-catenin, β-catenin (Ser675), TCF4, survivin and c-Myc when compared to Dex only. Moreover, the activation of WNT pathway induced by LiCl can also be inhibited by this combination treatment. Taken together, our study demonstrated that the combination of rhPDCD5 and Dex can suppress the proliferation of multiple myeloma cells partially via inhibiting the WNT signalling pathway.
Collapse
Affiliation(s)
- Qian Cheng
- Department of Hematology, the Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Liping Liu
- Department of General Surgery, the Affiliated Zhuzhou Hospital, Xiangya Medical College, Central South University, Zhuzhou, Hunan, China
| | - Yunfeng Fu
- Department of Hematology, the Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yanan Zhang
- Department of Hematology, the Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ye Yang
- Basic Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Jing Liu
- Department of Hematology, the Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
69
|
Preziosi ME, Singh S, Valore EV, Jung CL, Popovic B, Poddar M, Nagarajan S, Ganz T, Monga SP. Mice lacking liver-specific β-catenin develop steatohepatitis and fibrosis after iron overload. J Hepatol 2017; 67:360-369. [PMID: 28341391 PMCID: PMC5515705 DOI: 10.1016/j.jhep.2017.03.012] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Revised: 03/06/2017] [Accepted: 03/13/2017] [Indexed: 12/21/2022]
Abstract
BACKGROUND & AIMS Iron overload disorders such as hereditary hemochromatosis and iron loading anemias are a common cause of morbidity from liver diseases and increase risk of hepatic fibrosis and hepatocellular carcinoma (HCC). Treatment options for iron-induced damage are limited, partly because there is lack of animal models of human disease. Therefore, we investigated the effect of iron overload in liver-specific β-catenin knockout mice (KO), which are susceptible to injury, fibrosis and tumorigenesis following chemical carcinogen exposure. METHODS Iron overload diet was administered to KO and littermate control (CON) mice for various times. To ameliorate an oxidant-mediated component of tissue injury, N-Acetyl-L-(+)-cysteine (NAC) was added to drinking water of mice on iron overload diet. RESULTS KO on iron diet (KO +Fe) exhibited remarkable inflammation, followed by steatosis, oxidative stress, fibrosis, regenerating nodules and occurrence of occasional HCC. Increased injury in KO +Fe was associated with activated protein kinase B (AKT), ERK, and NF-κB, along with reappearance of β-catenin and target gene Cyp2e1, which promoted lipid peroxidation and hepatic damage. Addition of NAC to drinking water protected KO +Fe from hepatic steatosis, injury and fibrosis, and prevented activation of AKT, ERK, NF-κB and reappearance of β-catenin. CONCLUSIONS The absence of hepatic β-catenin predisposes mice to hepatic injury and fibrosis following iron overload, which was reminiscent of hemochromatosis and associated with enhanced steatohepatitis and fibrosis. Disease progression was notably alleviated by antioxidant therapy, which supports its chemopreventive role in the management of chronic iron overload disorders. LAY SUMMARY Lack of animal models for iron overload disorders makes it hard to study the disease process for improving therapies. Feeding high iron diet to mice that lack the β-catenin gene in liver cells led to increased inflammation followed by fat accumulation, cell death and wound healing that mimicked human disease. Administration of an antioxidant prevented hepatic injury in this model.
Collapse
Affiliation(s)
- Morgan E. Preziosi
- Department of Pathology (Division of Experimental Pathology),Pittsburgh Liver Research Center
| | - Sucha Singh
- Department of Pathology (Division of Experimental Pathology),Pittsburgh Liver Research Center
| | - Erika V. Valore
- Department of Medicine and Laboratory Medicine, University of California at Los Angeles, Los Angeles, CA,Department of Pathology and Laboratory Medicine, University of California at Los Angeles, Los Angeles, CA
| | - Chun-Ling Jung
- Department of Medicine and Laboratory Medicine, University of California at Los Angeles, Los Angeles, CA,Department of Pathology and Laboratory Medicine, University of California at Los Angeles, Los Angeles, CA
| | | | - Minakshi Poddar
- Department of Pathology (Division of Experimental Pathology),Pittsburgh Liver Research Center
| | - Shanmugam Nagarajan
- Department of Pathology (Division of Experimental Pathology),Pittsburgh Liver Research Center
| | - Tomas Ganz
- Department of Medicine and Laboratory Medicine, University of California at Los Angeles, Los Angeles, CA,Department of Pathology and Laboratory Medicine, University of California at Los Angeles, Los Angeles, CA
| | - Satdarshan P Monga
- Department of Pathology (Division of Experimental Pathology), University of Pittsburgh, Pennsylvania, United States; Pittsburgh Liver Research Center, University of Pittsburgh, Pennsylvania, United States; Department of Medicine (Division of Gastroenterology, Hepatology and Nutrition), University of Pittsburgh, Pennsylvania, United States.
| |
Collapse
|
70
|
Yang J, Milasta S, Hu D, AlTahan AM, Interiano RB, Zhou J, Davidson J, Low J, Lin W, Bao J, Goh P, Nathwani AC, Wang R, Wang Y, Ong SS, Boyd VA, Young B, Das S, Shelat A, Wu Y, Li Z, Zheng JJ, Mishra A, Cheng Y, Qu C, Peng J, Green DR, White S, Guy RK, Chen T, Davidoff AM. Targeting Histone Demethylases in MYC-Driven Neuroblastomas with Ciclopirox. Cancer Res 2017; 77:4626-4638. [PMID: 28684529 DOI: 10.1158/0008-5472.can-16-0826] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Revised: 11/28/2016] [Accepted: 06/29/2017] [Indexed: 12/21/2022]
Abstract
Histone lysine demethylases facilitate the activity of oncogenic transcription factors, including possibly MYC. Here we show that multiple histone demethylases influence the viability and poor prognosis of neuroblastoma cells, where MYC is often overexpressed. We also identified the approved small-molecule antifungal agent ciclopirox as a novel pan-histone demethylase inhibitor. Ciclopirox targeted several histone demethylases, including KDM4B implicated in MYC function. Accordingly, ciclopirox inhibited Myc signaling in parallel with mitochondrial oxidative phosphorylation, resulting in suppression of neuroblastoma cell viability and inhibition of tumor growth associated with an induction of differentiation. Our findings provide new insights into epigenetic regulation of MYC function and suggest a novel pharmacologic basis to target histone demethylases as an indirect MYC-targeting approach for cancer therapy. Cancer Res; 77(17); 4626-38. ©2017 AACR.
Collapse
Affiliation(s)
- Jun Yang
- Department of Surgery, St. Jude Children's Research Hospital, Memphis, Tennessee.
| | - Sandra Milasta
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Dongli Hu
- Department of Surgery, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Alaa M AlTahan
- Department of Surgery, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Rodrigo B Interiano
- Department of Surgery, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Junfang Zhou
- Department of Surgery, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Jesse Davidson
- Department of Surgery, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Jonathan Low
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Wenwei Lin
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Ju Bao
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Pollyanna Goh
- Department of Oncology, University College London Cancer Institute, London, United Kingdom
| | - Amit C Nathwani
- Department of Oncology, University College London Cancer Institute, London, United Kingdom
| | - Ruoning Wang
- Department of Pediatrics, The Ohio State University School of Medicine, The Research Institute at Nationwide Children's Hospital, Center for Childhood Cancer and Blood Disease, Columbus, Ohio
| | - Yingdi Wang
- Yale Cardiovascular Research Center, Yale School of Medicine, New Haven, Connecticut
| | - Su Sien Ong
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Vincent A Boyd
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Brandon Young
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Sourav Das
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Anang Shelat
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Yinan Wu
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Zhenmei Li
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Jie J Zheng
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Ashutosh Mishra
- Department of Structural Biology, Department of Developmental Neurobiology and St. Jude Proteomics Facility, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Yong Cheng
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Chunxu Qu
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Junmin Peng
- Department of Structural Biology, Department of Developmental Neurobiology and St. Jude Proteomics Facility, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Douglas R Green
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Stephen White
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - R Kiplin Guy
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Taosheng Chen
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Andrew M Davidoff
- Department of Surgery, St. Jude Children's Research Hospital, Memphis, Tennessee
| |
Collapse
|
71
|
Horniblow RD, Bedford M, Hollingworth R, Evans S, Sutton E, Lal N, Beggs A, Iqbal TH, Tselepis C. BRAF mutations are associated with increased iron regulatory protein-2 expression in colorectal tumorigenesis. Cancer Sci 2017; 108:1135-1143. [PMID: 28281325 PMCID: PMC5480081 DOI: 10.1111/cas.13234] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Revised: 03/02/2017] [Accepted: 03/03/2017] [Indexed: 01/01/2023] Open
Abstract
A role for iron in carcinogenesis is supported by evidence that iron metabolism proteins are modulated in cancer progression. To date, however, the expression of iron regulatory protein‐2 (IRP2), which is known to regulate several iron metabolism proteins, has not been assessed in colorectal cancer. Expression of IRP2 was assessed by quantitative RT‐PCR and immunohistochemistry in human colorectal cancer tissue. By interrogating The Cancer Genome Atlas (TCGA) database, expression of IRP2 and transferrin receptor‐1 (TfR1) was assessed relative to common mutations that are known to occur in cancer. The impact of suppressing IRP2 on cellular iron metabolism was also determined by using siRNA and by using the MEK inhibitor trametinib. IRP2 was overexpressed in colorectal cancer compared to normal colonic mucosa and its expression was positively correlated with TfR1 expression. In addition, IRP2 expression was associated with mutations in BRAF. The MEK inhibitor trametinib suppressed IRP2 and this was associated with a suppression in TfR1 and the labile iron pool (LIP). Moreover, epidermal growth factor stimulation resulted in decreased ferritin expression and an increase in the LIP which were independent of IRP2. Results presented here suggest that ablating IRP2 provides a therapeutic platform for intervening in colorectal tumorigenesis.
Collapse
Affiliation(s)
- Richard D Horniblow
- Institutes of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - Matthew Bedford
- Institutes of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - Robert Hollingworth
- Institutes of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - Sarah Evans
- Institutes of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - Emily Sutton
- Institutes of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - Neeraj Lal
- Immunity and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Andrew Beggs
- Institutes of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - Tariq H Iqbal
- Institutes of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - Chris Tselepis
- Institutes of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| |
Collapse
|
72
|
Abstract
Ciclopirox olamine (CPX), an off-patent fungicide, has recently been identified as a novel anticancer agent. However, the molecular mechanism underlying its anticancer action remains to be elucidated. Here we show that CPX inhibits cell proliferation in part by downregulating the protein level of Cdc25A in tumor cells. Our studies revealed that CPX did not significantly reduce Cdc25A mRNA level or Cdc25A protein synthesis, but remarkably promoted Cdc25A protein degradation. This resulted in inhibition of G1-cyclin dependent kinases (CDKs), as evidenced by increased inhibitory phosphorylation of G1-CDKs. Since Cdc25A degradation is tightly related to its phosphorylation status, we further examined whether CPX alters Cdc25A phosphorylation. The results showed that CPX treatment increased the phosphorylation of Cdc25A (S76 and S82), but only Cdc25A-S82A mutant was resistant to CPX-induced degradation. Furthermore, ectopic expression of Cdc25A-S82A partially conferred resistance to CPX inhibition of cell proliferation. Therefore, our findings indicate that CPX inhibits cell proliferation at least in part by promoting Cdc25A degradation.
Collapse
|
73
|
González N, Prieto I, del Puerto-Nevado L, Portal-Nuñez S, Ardura JA, Corton M, Fernández-Fernández B, Aguilera O, Gomez-Guerrero C, Mas S, Moreno JA, Ruiz-Ortega M, Sanz AB, Sanchez-Niño MD, Rojo F, Vivanco F, Esbrit P, Ayuso C, Alvarez-Llamas G, Egido J, García-Foncillas J, Ortiz A. 2017 update on the relationship between diabetes and colorectal cancer: epidemiology, potential molecular mechanisms and therapeutic implications. Oncotarget 2017; 8:18456-18485. [PMID: 28060743 PMCID: PMC5392343 DOI: 10.18632/oncotarget.14472] [Citation(s) in RCA: 125] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 12/26/2016] [Indexed: 02/06/2023] Open
Abstract
Worldwide deaths from diabetes mellitus (DM) and colorectal cancer increased by 90% and 57%, respectively, over the past 20 years. The risk of colorectal cancer was estimated to be 27% higher in patients with type 2 DM than in non-diabetic controls. However, there are potential confounders, information from lower income countries is scarce, across the globe there is no correlation between DM prevalence and colorectal cancer incidence and the association has evolved over time, suggesting the impact of additional environmental factors. The clinical relevance of these associations depends on understanding the mechanism involved. Although evidence is limited, insulin use has been associated with increased and metformin with decreased incidence of colorectal cancer. In addition, colorectal cancer shares some cellular and molecular pathways with diabetes target organ damage, exemplified by diabetic kidney disease. These include epithelial cell injury, activation of inflammation and Wnt/β-catenin pathways and iron homeostasis defects, among others. Indeed, some drugs have undergone clinical trials for both cancer and diabetic kidney disease. Genome-wide association studies have identified diabetes-associated genes (e.g. TCF7L2) that may also contribute to colorectal cancer. We review the epidemiological evidence, potential pathophysiological mechanisms and therapeutic implications of the association between DM and colorectal cancer. Further studies should clarify the worldwide association between DM and colorectal cancer, strengthen the biological plausibility of a cause-and-effect relationship through characterization of the molecular pathways involved, search for specific molecular signatures of colorectal cancer under diabetic conditions, and eventually explore DM-specific strategies to prevent or treat colorectal cancer.
Collapse
Affiliation(s)
- Nieves González
- Renal, Vascular and Diabetes Research Laboratory, IIS-Fundacion Jimenez Diaz-UAM, Spanish Biomedical Research Network in Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid, Spain
| | - Isabel Prieto
- Radiation Oncology, Oncohealth Institute, IIS-Fundacion Jimenez Diaz-UAM, Madrid, Spain
| | - Laura del Puerto-Nevado
- Translational Oncology Division, Oncohealth Institute, IIS-Fundacion Jimenez Diaz-UAM, Madrid, Spain
| | - Sergio Portal-Nuñez
- Bone and Mineral Metabolism laboratory, IIS-Fundacion Jimenez Diaz-UAM, Madrid, Spain
| | - Juan Antonio Ardura
- Bone and Mineral Metabolism laboratory, IIS-Fundacion Jimenez Diaz-UAM, Madrid, Spain
| | - Marta Corton
- Genetics, IIS-Fundacion Jimenez Diaz-UAM, Madrid, Spain
| | | | - Oscar Aguilera
- Translational Oncology Division, Oncohealth Institute, IIS-Fundacion Jimenez Diaz-UAM, Madrid, Spain
| | | | - Sebastián Mas
- Nephrology, IIS-Fundacion Jimenez Diaz-UAM, Madrid, Spain
| | | | | | - Ana Belen Sanz
- Nephrology, IIS-Fundacion Jimenez Diaz-UAM, Madrid, Spain
- REDINREN, Madrid, Spain
| | | | - Federico Rojo
- Pathology, IIS-Fundacion Jimenez Diaz-UAM, Madrid, Spain
| | | | - Pedro Esbrit
- Bone and Mineral Metabolism laboratory, IIS-Fundacion Jimenez Diaz-UAM, Madrid, Spain
| | - Carmen Ayuso
- Genetics, IIS-Fundacion Jimenez Diaz-UAM, Madrid, Spain
| | | | - Jesús Egido
- Renal, Vascular and Diabetes Research Laboratory, IIS-Fundacion Jimenez Diaz-UAM, Spanish Biomedical Research Network in Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid, Spain
- Nephrology, IIS-Fundacion Jimenez Diaz-UAM, Madrid, Spain
| | - Jesús García-Foncillas
- Translational Oncology Division, Oncohealth Institute, IIS-Fundacion Jimenez Diaz-UAM, Madrid, Spain
| | - Alberto Ortiz
- Nephrology, IIS-Fundacion Jimenez Diaz-UAM, Madrid, Spain
- REDINREN, Madrid, Spain
| | | |
Collapse
|
74
|
Brenner AK, Tvedt THA, Nepstad I, Rye KP, Hagen KM, Reikvam H, Bruserud Ø. Patients with acute myeloid leukemia can be subclassified based on the constitutive cytokine release of the leukemic cells; the possible clinical relevance and the importance of cellular iron metabolism. Expert Opin Ther Targets 2017; 21:357-369. [PMID: 28281897 DOI: 10.1080/14728222.2017.1300255] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
OBJECTIVE Acute myeloid leukaemia (AML) is a heterogeneous malignancy; we studied how the constitutive cytokine release by the AML cells varies among patients. METHODS We investigated the constitutive release of 28 mediators during in vitro culture for 79 consecutive patients. RESULTS Constitutive cytokine release profiles differed among patients, and hierarchical clustering identified three subsets with high, intermediate and low release, respectively. The high-release subset showed high levels of most mediators, usually monocytic differentiation as well as altered mRNA expression of proteins involved in intracellular iron homeostasis and molecular trafficking; this subset also included 4 out of 6 patients with inv(16). Spontaneous in vitro apoptosis did not differ among the subsets. For the high-release patients, cytokines were released both by CD34+ and CD34- cells. The mRNA and released protein levels showed statistically significant correlations only for eleven of the cytokines. The overall survival after intensive anti-leukemic therapy was significantly higher for high-release compared with low-release patients. Pharmacological targeting of iron metabolism (iron chelation, transferrin receptor blocking) altered the cytokine release profile. CONCLUSIONS Subclassification of AML patients based on the constitutive cytokine release may be clinically relevant and a part of a low-risk (i.e. chemosensitive) AML cell phenotype.
Collapse
Affiliation(s)
- Annette K Brenner
- a Section for Haematology, Department of Clinical Science , University of Bergen , Bergen , Norway
| | | | - Ina Nepstad
- a Section for Haematology, Department of Clinical Science , University of Bergen , Bergen , Norway
| | - Kristin P Rye
- a Section for Haematology, Department of Clinical Science , University of Bergen , Bergen , Norway
| | - Karen M Hagen
- a Section for Haematology, Department of Clinical Science , University of Bergen , Bergen , Norway
| | - Håkon Reikvam
- a Section for Haematology, Department of Clinical Science , University of Bergen , Bergen , Norway.,b Department of Medicine , Haukeland University Hospital , Bergen , Norway
| | - Øystein Bruserud
- a Section for Haematology, Department of Clinical Science , University of Bergen , Bergen , Norway.,b Department of Medicine , Haukeland University Hospital , Bergen , Norway
| |
Collapse
|
75
|
Rychtarcikova Z, Lettlova S, Tomkova V, Korenkova V, Langerova L, Simonova E, Zjablovskaja P, Alberich-Jorda M, Neuzil J, Truksa J. Tumor-initiating cells of breast and prostate origin show alterations in the expression of genes related to iron metabolism. Oncotarget 2017; 8:6376-6398. [PMID: 28031527 PMCID: PMC5351639 DOI: 10.18632/oncotarget.14093] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 11/30/2016] [Indexed: 12/22/2022] Open
Abstract
The importance of iron in the growth and progression of tumors has been widely documented. In this report, we show that tumor-initiating cells (TICs), represented by spheres derived from the MCF7 cell line, exhibit higher intracellular labile iron pool, mitochondrial iron accumulation and are more susceptible to iron chelation. TICs also show activation of the IRP/IRE system, leading to higher iron uptake and decrease in iron storage, suggesting that level of properly assembled cytosolic iron-sulfur clusters (FeS) is reduced. This finding is confirmed by lower enzymatic activity of aconitase and FeS cluster biogenesis enzymes, as well as lower levels of reduced glutathione, implying reduced FeS clusters synthesis/utilization in TICs. Importantly, we have identified specific gene signature related to iron metabolism consisting of genes regulating iron uptake, mitochondrial FeS cluster biogenesis and hypoxic response (ABCB10, ACO1, CYBRD1, EPAS1, GLRX5, HEPH, HFE, IREB2, QSOX1 and TFRC). Principal component analysis based on this signature is able to distinguish TICs from cancer cells in vitro and also Leukemia-initiating cells (LICs) from non-LICs in the mouse model of acute promyelocytic leukemia (APL). Majority of the described changes were also recapitulated in an alternative model represented by MCF7 cells resistant to tamoxifen (TAMR) that exhibit features of TICs. Our findings point to the critical importance of redox balance and iron metabolism-related genes and proteins in the context of cancer and TICs that could be potentially used for cancer diagnostics or therapy.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents/pharmacology
- Biological Transport
- Breast Neoplasms/drug therapy
- Breast Neoplasms/enzymology
- Breast Neoplasms/genetics
- Breast Neoplasms/pathology
- Dose-Response Relationship, Drug
- Drug Resistance, Neoplasm
- Female
- Gene Expression Profiling
- Gene Expression Regulation, Enzymologic
- Gene Expression Regulation, Neoplastic
- Humans
- Iron/metabolism
- Iron Chelating Agents/pharmacology
- Leukemia, Promyelocytic, Acute/enzymology
- Leukemia, Promyelocytic, Acute/genetics
- MCF-7 Cells
- Male
- Mice, Transgenic
- Mitochondria/enzymology
- Neoplastic Stem Cells/drug effects
- Neoplastic Stem Cells/enzymology
- Neoplastic Stem Cells/pathology
- Phenotype
- Principal Component Analysis
- Prostatic Neoplasms/drug therapy
- Prostatic Neoplasms/enzymology
- Prostatic Neoplasms/genetics
- Prostatic Neoplasms/pathology
- Spheroids, Cellular
- Tamoxifen/pharmacology
- Transcriptome
Collapse
Affiliation(s)
- Zuzana Rychtarcikova
- Institute of Biotechnology, Czech Academy of Sciences, Prague, Czech Republic
- Charles University in Prague, Faculty of Pharmacy in Hradec Kralove, Hradec Kralove, Czech Republic
| | - Sandra Lettlova
- Institute of Biotechnology, Czech Academy of Sciences, Prague, Czech Republic
- Charles University in Prague, Faculty of Sciences, Prague, Czech Republic
| | - Veronika Tomkova
- Institute of Biotechnology, Czech Academy of Sciences, Prague, Czech Republic
- Charles University in Prague, Faculty of Sciences, Prague, Czech Republic
| | - Vlasta Korenkova
- Institute of Biotechnology, Czech Academy of Sciences, Prague, Czech Republic
| | - Lucie Langerova
- Institute of Biotechnology, Czech Academy of Sciences, Prague, Czech Republic
| | - Ekaterina Simonova
- Institute of Biotechnology, Czech Academy of Sciences, Prague, Czech Republic
| | - Polina Zjablovskaja
- Institute of Molecular Genetics, Czech Academy of Sciences, Prague, Czech Republic
| | | | - Jiri Neuzil
- Institute of Biotechnology, Czech Academy of Sciences, Prague, Czech Republic
- School of Medical Science, Menzies Health Institute Queensland, Southport, Queensland, Australia
| | - Jaroslav Truksa
- Institute of Biotechnology, Czech Academy of Sciences, Prague, Czech Republic
| |
Collapse
|
76
|
Xue X, Shah YM. Iron, Cancer, and Hypoxia-Inducible Factor Signaling. MOLECULAR, GENETIC, AND NUTRITIONAL ASPECTS OF MAJOR AND TRACE MINERALS 2017:203-213. [DOI: 10.1016/b978-0-12-802168-2.00017-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
77
|
Horniblow RD, Henesy D, Iqbal TH, Tselepis C. Modulation of iron transport, metabolism and reactive oxygen status by quercetin-iron complexes in vitro. Mol Nutr Food Res 2016; 61. [PMID: 27794191 DOI: 10.1002/mnfr.201600692] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Revised: 10/21/2016] [Accepted: 10/25/2016] [Indexed: 02/06/2023]
Abstract
SCOPE Excess free-iron is detrimental to health through its ability to participate in free radical generation and amplification of oncogenic pathways. The study aims were to identify polyphenols with iron-chelating potential. METHODS AND RESULTS Of four polyphenols tested quercetin demonstrated potent iron binding with the physiological outcome dictated by the location of interaction. In the presence of extracellular iron and quercetin, ferritin expression and cellular iron concentrations decreased suggesting the resulting quercetin-iron complex is not internalised. However, in the relative absence of extracellular iron, quercetin becomes internalised and complexes with both intracellular iron, and iron which subsequently becomes absorbed as indicated by increased cellular 59 Fe post pre-culture with quercetin. This increased intracellular iron complexed to quercetin does not associate with the labile iron pool and cells behave as though they are iron deficient (increased transferrin receptor-1 and iron regulatory protein-2 expression and low ferritin expression). Additionally, a suppression in reactive oxygen species was observed. CONCLUSION Quercetin, an exogenous iron chelator, is able to render the cell functionally iron-deficient which not only provides a therapeutic platform for chelating excess free luminal iron but also may be of use in limiting processes such as cancer-cell growth, inflammation and bacterial infections, which all require iron.
Collapse
Affiliation(s)
- Richard D Horniblow
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, England, UK
| | - Daisy Henesy
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, England, UK
| | - Tariq H Iqbal
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, England, UK
| | - Chris Tselepis
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, England, UK
| |
Collapse
|
78
|
Nakanishi S, Cleveland JL. Targeting the polyamine-hypusine circuit for the prevention and treatment of cancer. Amino Acids 2016; 48:2353-62. [PMID: 27357307 PMCID: PMC5573165 DOI: 10.1007/s00726-016-2275-3] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Accepted: 06/08/2016] [Indexed: 01/19/2023]
Abstract
The unique amino acid hypusine is present in only two proteins in eukaryotic cells, eukaryotic translation initiation factor 5A-1 (eIF5A1), and eIF5A2, where it is covalently linked to the lysine-50 residue of these proteins via a post-translational modification coined hypusination. This unique modification is directed by two highly conserved and essential enzymes, deoxyhypusine synthase (DHPS), and deoxyhypusine hydroxylase (DOHH), which selectively use the polyamine spermidine as a substrate to generate hypusinated eIF5A. Notably, elevated levels of polyamines are a hallmark of most tumor types, and increased levels of polyamines can also be detected in the urine and blood of cancer patients. Further, in-clinic agents that block the function of key biosynthetic enzymes in the polyamine pathway markedly impair tumor progression and maintenance of the malignant state. Thus, the polyamine pathway is attractive as a prognostic, prevention and therapeutic target. As we review, recent advances in our understanding of the specific functions of hypusinated eIF5A and its role in tumorigenesis suggest that the polyamine-hypusine circuit is a high priority target for cancer therapeutics.
Collapse
Affiliation(s)
- Shima Nakanishi
- Department of Tumor Biology, The Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA
| | - John L Cleveland
- Department of Tumor Biology, The Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA.
| |
Collapse
|
79
|
Chavez-Gonzalez A, Bakhshinejad B, Pakravan K, Guzman ML, Babashah S. Novel strategies for targeting leukemia stem cells: sounding the death knell for blood cancer. Cell Oncol (Dordr) 2016; 40:1-20. [PMID: 27678246 DOI: 10.1007/s13402-016-0297-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/31/2016] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Cancer stem cells (CSCs), also known as tumor-initiating cells (TICs), are characterized by high self-renewal and multi-lineage differentiation capacities. CSCs are thought to play indispensable roles in the initiation, progression and metastasis of many types of cancer. Leukemias are thought to be initiated and maintained by a specific sub-type of CSC, the leukemia stem cell (LSC). An important feature of LSCs is their resistance to standard therapy, which may lead to relapse. Increasing efforts are aimed at developing novel therapeutic strategies that selectively target LSCs, while sparing their normal counterparts and, thus, minimizing adverse treatment-associated side-effects. These LSC targeting therapies aim to eradicate LSCs through affecting mechanisms that control their survival, self-renewal, differentiation, proliferation and cell cycle progression. Some LSC targeting therapies have already been proven successful in pre-clinical studies and they are now being tested in clinical studies, mainly in combination with conventional treatment regimens. CONCLUSIONS A growing body of evidence indicates that the selective targeting of LSCs represents a promising approach to improve disease outcome. Beyond doubt, the CSC hypothesis has added a new dimension to the area of anticancer research, thereby paving the way for shaping a new trend in cancer therapy.
Collapse
Affiliation(s)
| | - Babak Bakhshinejad
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, P.O. Box: 14115-154, Tehran, Iran
| | - Katayoon Pakravan
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, P.O. Box: 14115-154, Tehran, Iran
| | - Monica L Guzman
- Department of Medicine, Weill Medical College of Cornell University, 1300 York Ave, Box 113, New York, NY, 10065, USA.
| | - Sadegh Babashah
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, P.O. Box: 14115-154, Tehran, Iran.
| |
Collapse
|
80
|
Abstract
Increased dietary iron intake and elevated systemic iron levels are associated with increased cancer risk. In this issue, Xue et al. (2016) have identified an unexpected link between intracellular iron accumulation and pro-inflammatory signaling that provides, at least in part, a molecular explanation for the tumor-promoting effects of iron.
Collapse
Affiliation(s)
- Florian R Greten
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, Paul-Ehrlich-Str. 42-44, 60596 Frankfurt am Main, Germany.
| |
Collapse
|
81
|
Xue X, Ramakrishnan SK, Weisz K, Triner D, Xie L, Attili D, Pant A, Győrffy B, Zhan M, Carter-Su C, Hardiman KM, Wang TD, Dame MK, Varani J, Brenner D, Fearon ER, Shah YM. Iron Uptake via DMT1 Integrates Cell Cycle with JAK-STAT3 Signaling to Promote Colorectal Tumorigenesis. Cell Metab 2016; 24:447-461. [PMID: 27546461 PMCID: PMC5023486 DOI: 10.1016/j.cmet.2016.07.015] [Citation(s) in RCA: 182] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Revised: 06/07/2016] [Accepted: 07/20/2016] [Indexed: 12/31/2022]
Abstract
Dietary iron intake and systemic iron balance are implicated in colorectal cancer (CRC) development, but the means by which iron contributes to CRC are unclear. Gene expression and functional studies demonstrated that the cellular iron importer, divalent metal transporter 1 (DMT1), is highly expressed in CRC through hypoxia-inducible factor 2α-dependent transcription. Colon-specific Dmt1 disruption resulted in a tumor-selective inhibitory effect of proliferation in mouse colon tumor models. Proteomic and genomic analyses identified an iron-regulated signaling axis mediated by cyclin-dependent kinase 1 (CDK1), JAK1, and STAT3 in CRC progression. A pharmacological inhibitor of DMT1 antagonized the ability of iron to promote tumor growth in a CRC mouse model and a patient-derived CRC enteroid orthotopic model. Our studies implicate a growth-promoting signaling network instigated by elevated intracellular iron levels in tumorigenesis, offering molecular insights into how a key dietary component may contribute to CRC.
Collapse
Affiliation(s)
- Xiang Xue
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Sadeesh K Ramakrishnan
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Kevin Weisz
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Daniel Triner
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Liwei Xie
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Durga Attili
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Asha Pant
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Balázs Győrffy
- MTA TTK Lendület Cancer Biomarker Research Group, Budapest 1117, Hungary; 2nd Department of Pediatrics, Semmelweis University, Budapest 1085, Hungary
| | - Mingkun Zhan
- Department of Plastic Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, China
| | - Christin Carter-Su
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA; Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Karin M Hardiman
- Department of Surgery, University of Michigan, Ann Arbor, MI 48109, USA
| | - Thomas D Wang
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA; Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA; Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Michael K Dame
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109, USA
| | - James Varani
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Dean Brenner
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109, USA; Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Eric R Fearon
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109, USA; Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA; Department of Human Genetics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Yatrik M Shah
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA; Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
82
|
McCubrey JA, Rakus D, Gizak A, Steelman LS, Abrams SL, Lertpiriyapong K, Fitzgerald TL, Yang LV, Montalto G, Cervello M, Libra M, Nicoletti F, Scalisi A, Torino F, Fenga C, Neri LM, Marmiroli S, Cocco L, Martelli AM. Effects of mutations in Wnt/β-catenin, hedgehog, Notch and PI3K pathways on GSK-3 activity-Diverse effects on cell growth, metabolism and cancer. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1863:2942-2976. [PMID: 27612668 DOI: 10.1016/j.bbamcr.2016.09.004] [Citation(s) in RCA: 116] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 08/14/2016] [Accepted: 09/02/2016] [Indexed: 02/07/2023]
Abstract
Glycogen synthase kinase-3 (GSK-3) is a serine/threonine kinase that participates in an array of critical cellular processes. GSK-3 was first characterized as an enzyme that phosphorylated and inactivated glycogen synthase. However, subsequent studies have revealed that this moon-lighting protein is involved in numerous signaling pathways that regulate not only metabolism but also have roles in: apoptosis, cell cycle progression, cell renewal, differentiation, embryogenesis, migration, regulation of gene transcription, stem cell biology and survival. In this review, we will discuss the roles that GSK-3 plays in various diseases as well as how this pivotal kinase interacts with multiple signaling pathways such as: PI3K/PTEN/Akt/mTOR, Ras/Raf/MEK/ERK, Wnt/beta-catenin, hedgehog, Notch and TP53. Mutations that occur in these and other pathways can alter the effects that natural GSK-3 activity has on regulating these signaling circuits that can lead to cancer as well as other diseases. The novel roles that microRNAs play in regulation of the effects of GSK-3 will also be evaluated. Targeting GSK-3 and these other pathways may improve therapy and overcome therapeutic resistance.
Collapse
Affiliation(s)
- James A McCubrey
- Department of Microbiology and Immunology, Brody School of Medicine at East Carolina University Greenville, NC 27858, USA.
| | - Dariusz Rakus
- Department of Animal Molecular Physiology, Institute of Experimental Biology, Wroclaw University, Wroclaw, Poland
| | - Agnieszka Gizak
- Department of Animal Molecular Physiology, Institute of Experimental Biology, Wroclaw University, Wroclaw, Poland
| | - Linda S Steelman
- Department of Microbiology and Immunology, Brody School of Medicine at East Carolina University Greenville, NC 27858, USA
| | - Steve L Abrams
- Department of Microbiology and Immunology, Brody School of Medicine at East Carolina University Greenville, NC 27858, USA
| | - Kvin Lertpiriyapong
- Department of Comparative Medicine, Brody School of Medicine at East Carolina University, USA
| | - Timothy L Fitzgerald
- Department of Surgery, Brody School of Medicine at East Carolina University, USA
| | - Li V Yang
- Department of Internal Medicine, Hematology/Oncology Section, Brody School of Medicine at East Carolina University, USA
| | - Giuseppe Montalto
- Biomedical Department of Internal Medicine and Specialties, University of Palermo, Palermo, Italy; Consiglio Nazionale delle Ricerche, Istituto di Biomedicina e Immunologia Molecolare "Alberto Monroy", Palermo, Italy
| | - Melchiorre Cervello
- Consiglio Nazionale delle Ricerche, Istituto di Biomedicina e Immunologia Molecolare "Alberto Monroy", Palermo, Italy
| | - Massimo Libra
- Department of Bio-medical Sciences, University of Catania, Catania, Italy
| | | | - Aurora Scalisi
- Unit of Oncologic Diseases, ASP-Catania, Catania 95100, Italy
| | - Francesco Torino
- Department of Systems Medicine, Chair of Medical Oncology, Tor Vergata University of Rome, Rome, Italy
| | - Concettina Fenga
- Department of Biomedical, Odontoiatric, Morphological and Functional Images, Occupational Medicine Section - Policlinico "G. Martino" - University of Messina, Messina 98125, Italy
| | - Luca M Neri
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Sandra Marmiroli
- Department of Surgery, Medicine, Dentistry and Morphology, University of Modena and Reggio Emilia, Modena, Italy
| | - Lucio Cocco
- Dipartimento di Scienze Biomediche e Neuromotorie, Università di Bologna, Bologna, Italy
| | - Alberto M Martelli
- Dipartimento di Scienze Biomediche e Neuromotorie, Università di Bologna, Bologna, Italy
| |
Collapse
|
83
|
Lu B, Green BA, Farr JM, Lopes FCM, Van Raay TJ. Wnt Drug Discovery: Weaving Through the Screens, Patents and Clinical Trials. Cancers (Basel) 2016; 8:cancers8090082. [PMID: 27598201 PMCID: PMC5040984 DOI: 10.3390/cancers8090082] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Revised: 08/09/2016] [Accepted: 08/15/2016] [Indexed: 12/17/2022] Open
Abstract
The Wnt signaling pathway is intricately involved in many aspects of development and is the root cause of an increasing number of diseases. For example, colorectal cancer is the second leading cause of death in the industrialized world and aberration of Wnt signaling within the colonic stem cell is the cause of more than 90% of these cancers. Despite our advances in successfully targeting other pathways, such as Human Epidermal Growth Factor Receptor 2 (HER2), there are no clinically relevant therapies available for Wnt-related diseases. Here, we investigated where research activities are focused with respect to Wnt signaling modulators by searching the United States Patent and Trade Office (USPTO) for patents and patent applications related to Wnt modulators and compared this to clinical trials focusing on Wnt modulation. We found that while the transition of intellectual property surrounding the Wnt ligand-receptor interface to clinical trials is robust, this is not true for specific inhibitors of β-catenin, which is constitutively active in many cancers. Considering the ubiquitous use of the synthetic T-cell Factor/Lymphoid Enhancer Factor (TCF/Lef) reporter system and its success in identifying novel modulators in vitro, we speculate that this model of drug discovery does not capture the complexity of in vivo Wnt signaling that may be required if we are to successfully target the Wnt pathway in the clinic. Notwithstanding, increasingly more complex models are being developed, which may not be high throughput, but more pragmatic in our pursuit to control Wnt signaling.
Collapse
Affiliation(s)
- Benjamin Lu
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON N1G 2W1, Canada.
| | - Brooke A Green
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON N1G 2W1, Canada.
| | - Jacqueline M Farr
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON N1G 2W1, Canada.
| | - Flávia C M Lopes
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON N1G 2W1, Canada.
| | - Terence J Van Raay
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON N1G 2W1, Canada.
| |
Collapse
|
84
|
Li Y, Lauriola M, Kim D, Francesconi M, D'Uva G, Shibata D, Malafa MP, Yeatman TJ, Coppola D, Solmi R, Cheng JQ. Adenomatous polyposis coli (APC) regulates miR17-92 cluster through β-catenin pathway in colorectal cancer. Oncogene 2016; 35:4558-4568. [PMID: 26804172 PMCID: PMC4960006 DOI: 10.1038/onc.2015.522] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Revised: 11/09/2015] [Accepted: 11/30/2015] [Indexed: 02/06/2023]
Abstract
Adenomatous polyposis coli (APC) mutation is the most common genetic change in sporadic colorectal cancer (CRC). Although deregulations of miRNAs have been frequently reported in this malignancy, APC-regulated miRNAs have not been extensively documented. Here, by using an APC-inducible cell line and array analysis, we identified a total of 26 deregulated miRNAs. Among them, members of miR-17-92 cluster were dramatically inhibited by APC and induced by enforced expression of β-catenin. Furthermore, we demonstrate that activated β-catenin resulted from APC loss binds to and activates the miR-17-92 promoter. Notably, enforced expression of miR-19a overrides APC tumor suppressor activity, and knockdown of miR-19a in cancer cells with compromised APC function reduced their aggressive features in vitro. Finally, we observed that expression of miR-19a significantly correlates with β-catenin levels in colorectal cancer specimens, and it is associated to the aggressive stage of tumor progression. Thus, our study reveals that miR-17-92 cluster is directly regulated by APC/β-catenin pathway and could be a potential therapeutic target in colon cancers with aberrant APC/β-catenin signaling.
Collapse
Affiliation(s)
- Yajuan Li
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Mattia Lauriola
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
- Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale, Università di Bologna, via Massarenti, 9, 40138 Bologna, Italy
| | - Donghwa Kim
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Mirko Francesconi
- EMBL-CRG Systems Biology Unit, Centre for Genomic Regulation (CRG), Dr. Aiguader 88, 08003 Barcelona, Spain
| | - Gabriele D'Uva
- Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale, Università di Bologna, via Massarenti, 9, 40138 Bologna, Italy
| | - Dave Shibata
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Mokenge P Malafa
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Timothy J Yeatman
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Domenico Coppola
- Department of Pathology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Rossella Solmi
- Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale, Università di Bologna, via Massarenti, 9, 40138 Bologna, Italy
| | - Jin Q Cheng
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| |
Collapse
|
85
|
Gallium and its competing roles with iron in biological systems. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1863:2044-53. [DOI: 10.1016/j.bbamcr.2016.04.027] [Citation(s) in RCA: 128] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 04/27/2016] [Accepted: 04/30/2016] [Indexed: 12/12/2022]
|
86
|
Lui GYL, Kovacevic Z, Richardson V, Merlot AM, Kalinowski DS, Richardson DR. Targeting cancer by binding iron: Dissecting cellular signaling pathways. Oncotarget 2016; 6:18748-79. [PMID: 26125440 PMCID: PMC4662454 DOI: 10.18632/oncotarget.4349] [Citation(s) in RCA: 130] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 06/12/2015] [Indexed: 12/30/2022] Open
Abstract
Newer and more potent therapies are urgently needed to effectively treat advanced cancers that have developed resistance and metastasized. One such strategy is to target cancer cell iron metabolism, which is altered compared to normal cells and may facilitate their rapid proliferation. This is supported by studies reporting the anti-neoplastic activities of the clinically available iron chelators, desferrioxamine and deferasirox. More recently, ligands of the di-2-pyridylketone thiosemicarbazone (DpT) class have demonstrated potent and selective anti-proliferative activity across multiple cancer-types in vivo, fueling studies aimed at dissecting their molecular mechanisms of action. In the past five years alone, significant advances have been made in understanding how chelators not only modulate cellular iron metabolism, but also multiple signaling pathways implicated in tumor progression and metastasis. Herein, we discuss recent research on the targeting of iron in cancer cells, with a focus on the novel and potent DpT ligands. Several key studies have revealed that iron chelation can target the AKT, ERK, JNK, p38, STAT3, TGF-β, Wnt and autophagic pathways to subsequently inhibit cellular proliferation, the epithelial-mesenchymal transition (EMT) and metastasis. These developments emphasize that these novel therapies could be utilized clinically to effectively target cancer.
Collapse
Affiliation(s)
- Goldie Y L Lui
- Department of Pathology and Bosch Institute, Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Zaklina Kovacevic
- Department of Pathology and Bosch Institute, Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Vera Richardson
- Department of Pathology and Bosch Institute, Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Angelica M Merlot
- Department of Pathology and Bosch Institute, Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Danuta S Kalinowski
- Department of Pathology and Bosch Institute, Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Des R Richardson
- Department of Pathology and Bosch Institute, Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
87
|
Salehi S, Saljooghi AS, Shiri A. Synthesis, characterization and in vitro anticancer evaluations of two novel derivatives of deferasirox iron chelator. Eur J Pharmacol 2016; 781:209-17. [DOI: 10.1016/j.ejphar.2016.04.026] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Revised: 04/11/2016] [Accepted: 04/13/2016] [Indexed: 01/08/2023]
|
88
|
Kim DS, Na YJ, Kang MH, Yoon SY, Choi CW. Use of deferasirox, an iron chelator, to overcome imatinib resistance of chronic myeloid leukemia cells. Korean J Intern Med 2016; 31:357-66. [PMID: 26874514 PMCID: PMC4773721 DOI: 10.3904/kjim.2015.024] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Revised: 03/18/2015] [Accepted: 03/30/2015] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND/AIMS The treatment of chronic myeloid leukemia (CML) has achieved impressive success since the development of the Bcr-Abl tyrosine kinase inhibitor, imatinib mesylate. Nevertheless, resistance to imatinib has been observed, and a substantial number of patients need alternative treatment strategies. METHODS We have evaluated the effects of deferasirox, an orally active iron chelator, and imatinib on K562 and KU812 human CML cell lines. Imatinib-resistant CML cell lines were created by exposing cells to gradually increasing concentrations of imatinib. RESULTS Co-treatment of cells with deferasirox and imatinib induced a synergistic dose-dependent inhibition of proliferation of both CML cell lines. Cell cycle analysis showed an accumulation of cells in the subG1 phase. Western blot analysis of apoptotic proteins showed that co-treatment with deferasirox and imatinib induced an increased expression of apoptotic proteins. These tendencies were clearly identified in imatinib-resistant CML cell lines. The results also showed that co-treatment with deferasirox and imatinib reduced the expression of BcrAbl, phosphorylated Bcr-Abl, nuclear factor-κB (NF-κB) and β-catenin. CONCLUSIONS We observed synergistic effects of deferasirox and imatinib on both imatinib-resistant and imatinib-sensitive cell lines. These effects were due to induction of apoptosis and cell cycle arrest by down-regulated expression of NF-κB and β-catenin levels. Based on these results, we suggest that a combination treatment of deferasirox and imatinib could be considered as an alternative treatment option for imatinib-resistant CML.
Collapse
Affiliation(s)
- Dae Sik Kim
- Division of Hematology and Oncology, Department of Internal Medicine, Korea University School of Medicine, Seoul, Korea
| | - Yoo Jin Na
- Graduate School of Medicine, Korea University School of Medicine, Seoul, Korea
| | - Myoung Hee Kang
- Graduate School of Medicine, Korea University School of Medicine, Seoul, Korea
| | - Soo-Young Yoon
- Department of Laboratory Medicine, Korea University School of Medicine, Seoul, Korea
| | - Chul Won Choi
- Division of Hematology and Oncology, Department of Internal Medicine, Korea University School of Medicine, Seoul, Korea
- Correspondence to Chul Won Choi, M.D. Division of Hematology and Oncology, Department of Internal Medicine, Korea University Guro Hospital, 148 Gurodong-ro, Guro-gu, Seoul 08308, Korea Tel: +82-2-2626-3058 Fax: +82-2-862-6453 E-mail:
| |
Collapse
|
89
|
Legendre C, Avril S, Guillet C, Garcion E. Low oxygen tension reverses antineoplastic effect of iron chelator deferasirox in human glioblastoma cells. BMC Cancer 2016; 16:51. [PMID: 26832741 PMCID: PMC4736662 DOI: 10.1186/s12885-016-2074-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Accepted: 01/19/2016] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Overcoming resistance to treatment is an essential issue in many cancers including glioblastoma (GBM), the deadliest primary tumor of the central nervous system. As dependence on iron is a key feature of tumor cells, using chelators to reduce iron represents an opportunity to improve conventional GBM therapies. The aim of the present study was, therefore, to investigate the cytostatic and cytotoxic impact of the new iron chelator deferasirox (DFX) on human GBM cells in well-defined clinical situations represented by radiation therapy and mild-hypoxia. RESULTS Under experimental normoxic condition (21% O2), deferasirox (DFX) used at 10 μM for 3 days reduced proliferation, led cell cycle arrest in S and G2-M phases and induced cytotoxicity and apoptosis in U251 and U87 GBM cells. The abolition of the antineoplastic DFX effects when cells were co-treated with ferric ammonium sulfate supports the hypothesis that its effects result from its ability to chelate iron. As radiotherapy is the main treatment for GBM, the combination of DFX and X-ray beam irradiation was also investigated. Irradiation at a dose of 16 Gy repressed proliferation, cytotoxicity and apoptosis, but only in U251 cells, while no synergy with DFX was observed in either cell line. Importantly, when the same experiment was conducted in mild-hypoxic conditions (3% O2), the antiproliferative and cytotoxic effects of DFX were abolished, and its ability to deplete iron was also impaired. CONCLUSIONS Taken together, these in vitro results could raise the question of the benefit of using iron chelators in their native forms under the hypoxic conditions often encountered in solid tumors such as GBM. Developing new chemistry or a new drug delivery system that would keep DFX active in hypoxic cells may be the next step toward their application.
Collapse
Affiliation(s)
- Claire Legendre
- INSERM U1066, Micro et Nanomédecines Biomimétiques, IBS - CHU, 4 Rue Larrey, F-49933, Angers, France
| | - Sylvie Avril
- INSERM U1066, Micro et Nanomédecines Biomimétiques, IBS - CHU, 4 Rue Larrey, F-49933, Angers, France
| | - Catherine Guillet
- PACeM : Plate-forme d'Analyses Cellulaire et Moléculaire, IBS - CHU, 4 Rue Larrey, F-49933, Angers, France
| | - Emmanuel Garcion
- INSERM U1066, Micro et Nanomédecines Biomimétiques, IBS - CHU, 4 Rue Larrey, F-49933, Angers, France.
| |
Collapse
|
90
|
Xue D, Zhou CX, Shi YB, Lu H, He XZ. Decreased expression of ferroportin in prostate cancer. Oncol Lett 2015; 10:913-916. [PMID: 26622594 DOI: 10.3892/ol.2015.3363] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Accepted: 05/07/2015] [Indexed: 12/21/2022] Open
Abstract
The present study examined the expression levels of ferroportin, a transmembrane protein that transports iron from the inside of a cell to the outside, in the prostate cancer PC3, DU145 and LNCAP cell lines, in the normal prostate RWPE2 cell line, and in tissue samples from different differentiation stages of prostatic carcinoma and prostatic hyperplasia. The study also investigated the role of ferroportin protein expression in the diagnosis and prognosis of prostate cancer. Reverse transcription-quantitative polymerase chain reaction and western blot analysis were employed to measure the mRNA and protein expression levels of ferroportin in the PC3, DU145, LNCAP and RWPE2 cells. Immunohistochemistry was used to determine ferroportin protein expression in the prostate cancer and prostatic hyperplasia tissues. Compared with the normal prostate RWPE2 cells, ferroportin protein expression was significantly lower in the prostate cancer PC3, DU145 and LNCAP cells (P<0.05). Compared with the prostatic hyperplasia tissues, ferroportin protein expression was significantly reduced in the prostate cancer tissues (P<0.05). Overall, the expression levels of ferroportin in the prostate cancer tissues were lower than those in the normal prostate tissues, which may provide valuable clinical information for the diagnosis and prediction of disease progression in prostate cancer, and may indicate a potential therapeutic target for treating prostate cancer by regulating iron metabolism.
Collapse
Affiliation(s)
- Dong Xue
- Department of Urology, Third Affiliated Hospital, Suzhou University, Changzhou, Jiangsu 213003, P.R. China
| | - Cui-Xing Zhou
- Department of Urology, Third Affiliated Hospital, Suzhou University, Changzhou, Jiangsu 213003, P.R. China
| | - Yun-Bo Shi
- Foreign Languages School, Changzhou Institute of Technology, Changzhou, Jiangsu 213002, P.R. China
| | - Hao Lu
- Department of Urology, Third Affiliated Hospital, Suzhou University, Changzhou, Jiangsu 213003, P.R. China
| | - Xiao-Zhou He
- Department of Urology, Third Affiliated Hospital, Suzhou University, Changzhou, Jiangsu 213003, P.R. China
| |
Collapse
|
91
|
Bloomer SA, Brown KE. Tumour promotion versus tumour suppression in chronic hepatic iron overload. Cell Biochem Funct 2015; 33:241-8. [DOI: 10.1002/cbf.3110] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Revised: 02/25/2015] [Accepted: 03/26/2015] [Indexed: 12/14/2022]
Affiliation(s)
- Steven A. Bloomer
- Division of Science and Engineering; Penn State Abington College; Abington PA USA
| | - Kyle E. Brown
- Iowa City Veterans Administration Medical Center; Iowa City IA USA
- Division of Gastroenterology-Hepatology; University of Iowa Roy J. and Lucille A. Carver College of Medicine; Iowa City IA USA
- Program in Free Radical and Radiation Biology; University of Iowa Roy J. and Lucille A. Carver College of Medicine; Iowa City IA USA
| |
Collapse
|
92
|
Padmanabhan H, Brookes MJ, Iqbal T. Iron and colorectal cancer: evidence from in vitro and animal studies. Nutr Rev 2015; 73:308-17. [DOI: 10.1093/nutrit/nuu015] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
|
93
|
Ziaei A, Ardakani MRP, Hashemi MS, Peymani M, Ghaedi K, Baharvand H, Nasr-Esfahani MH. Acute course of deferoxamine promoted neuronal differentiation of neural progenitor cells through suppression of Wnt/β-catenin pathway: a novel efficient protocol for neuronal differentiation. Neurosci Lett 2015; 590:138-44. [PMID: 25660235 DOI: 10.1016/j.neulet.2015.01.083] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2014] [Revised: 01/27/2015] [Accepted: 01/30/2015] [Indexed: 12/27/2022]
Abstract
Neural progenitor cells (NPCs) are feasible therapeutically model cells in regenerative medicine. However, a number of obstacles oppose their applications including insufficiency in differentiation protocols. These complications should be overwhelmed to obtain a significant clinical application. Deferoxamine (DFO), as a small molecule with a clinically high-affinity to chelate intracellular Iron, has been granted orphan drug status for treatment of traumatic spinal cord injury, while its neuroprotective function is not well understood. The aim of the present study is evaluating whether DFO could modulate neuronal differentiation process of NPCs. A varies concentrations of DFO were used to promote neuronal differentiation of mouse and human NPCs with different serum condition as an extracellular source of Iron. Several neural markers were assessed by RT-qPCR and Western analysis. Meanwhile β-catenin content was evaluated as key member of Wnt pathway. The maximal neuronal differentiation rate was observed when treating cells were treated with acute dosage of DFO (100 μM) for 6h in serum free condition. This treatment produced a significant increase in expression of neuronal markers and resulted in dramatically decrease in expression of glial markers. The protein content of β-catenin was also decreased by this treatment. Despite of chronic concentration of DFO, which reduced the size of EBs apparently due to G1/S arrest of cell cycle as known features of DFO. Application of acute courses of DFO increased neuronal differentiation rate of NPCs in serum free conditions. We concluded that suppression of Wnt/β-catenin pathway was induced through chelating of intracellular Iron due to DFO treatment. These findings help to understand therapeutic benefit of DFO as a neuroprotective agent.
Collapse
Affiliation(s)
- Amin Ziaei
- Department of Cellular Biotechnology at Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Mohammad Reza Piri Ardakani
- Department of Cellular Biotechnology at Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Motahare-Sadat Hashemi
- Department of Cellular Biotechnology at Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Maryam Peymani
- Department of Cellular Biotechnology at Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Kamran Ghaedi
- Department of Cellular Biotechnology at Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran; Department of Biology, School of Sciences, University of Isfahan, Isfahan, Iran.
| | - Hossein Baharvand
- Department of Stem Cells and Developmental Biology at Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Department of Developmental Biology, University of Science and Culture, ACECR, Tehran, Iran
| | - Mohammad Hossein Nasr-Esfahani
- Department of Cellular Biotechnology at Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran.
| |
Collapse
|
94
|
Arslan M, Ila HB. Deferasirox-induced cytogenetic responses. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2015; 39:787-793. [PMID: 25733130 DOI: 10.1016/j.etap.2015.02.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Revised: 02/04/2015] [Accepted: 02/05/2015] [Indexed: 06/04/2023]
Abstract
Deferasirox (commercially formulated as Exjade(®)) is one of the effective iron chelators used in treatment of iron overload diseases. In this study the effect of this substance for chromosome aberration, sister chromatid exchange and mitotic index was studied by in vitro (by using human peripheral lymphocytes) and in vivo (by using rat) analysis. Deferasirox increased the sister chromatid exchange frequency in all tested concentrations and periods in vitro. Also, in the presence of metabolic activator, the substance led to a statistically significant increase in the sister chromatid exchange frequencies only at high concentration. While in in vitro analysis the substance significantly increased abnormal cell percentages in all concentrations, in in vivo study the substance increased chromosome aberrations only in two concentrations at 12h treatment. In the cultured lymphocytes, deferasirox showed cytotoxicity by significantly reducing proliferation index and mitotic index values. While in the presence of metabolic activation it did not affect the proliferation index frequency, it had a stimulant effect on the mitotic index frequency. Deferasirox reduced significantly the mitotic index value in the bone marrow cells especially in high concentration and short treatment period (12h).
Collapse
Affiliation(s)
- Mehmet Arslan
- Ardahan University, School of Health Sciences, Department of Nursing, 75000 Ardahan, Turkey.
| | - Hasan Basri Ila
- Cukurova University, Faculty of Science and Letters, Department of Biology, 01330 Adana, Turkey
| |
Collapse
|
95
|
Sidarovich V, Adami V, Gatto P, Greco V, Tebaldi T, Tonini GP, Quattrone A. Translational downregulation of HSP90 expression by iron chelators in neuroblastoma cells. Mol Pharmacol 2015; 87:513-24. [PMID: 25564462 DOI: 10.1124/mol.114.095729] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Iron is an essential cellular nutrient, being a critical cofactor of several proteins involved in cell growth and replication. Compared with normal cells, neoplastic cells have been shown to require a greater amount of iron, thus laying the basis for the promising anticancer activity of iron chelators. In this work, we evaluated the effects of molecules with iron chelation activity on neuroblastoma (NB) cell lines. Of the 17 iron chelators tested, six reduced cell viability of two NB cell lines with an inhibition of growth of 50% below 10 µM; four of the six molecules-ciclopirox olamine (CPX), piroctone, 8-hydroxyquinoline, and deferasirox-were also shown to efficiently chelate intracellular iron within minutes after addition. Effects on cell viability of one of the compounds, CPX, were indeed dependent on chelation of intracellular iron and mediated by both G0/G1 cell cycle block and induction of apoptosis. By combined transcriptome and translatome profiling we identified early translational downregulation of several members of the heat shock protein group as a specific effect of CPX treatment. We functionally confirmed iron-dependent depletion of HSP90 and its client proteins at pharmacologically achievable concentrations of CPX, and we extended this effect to piroctone, 8-hydroxyquinoline, and deferasirox. Given the documented sensitivity of NB cells to HSP90 inhibition, we propose CPX and other iron chelators as investigational antitumor agents in NB therapy.
Collapse
Affiliation(s)
- Viktoryia Sidarovich
- Laboratory of Translational Genomics, Centre for Integrative Biology, University of Trento, Trento, Italy (V.S., P.G., V.G., T.T., A.Q.); High-Throughput Screening Core Facility, Centre for Integrative Biology, University of Trento, Trento, Italy (V.A.); and Neuroblastoma Laboratory, Onco/Hematology Laboratory, SDB Department, University of Padua, Pediatric Research Institute, Padua, Italy (G.P.T.)
| | - Valentina Adami
- Laboratory of Translational Genomics, Centre for Integrative Biology, University of Trento, Trento, Italy (V.S., P.G., V.G., T.T., A.Q.); High-Throughput Screening Core Facility, Centre for Integrative Biology, University of Trento, Trento, Italy (V.A.); and Neuroblastoma Laboratory, Onco/Hematology Laboratory, SDB Department, University of Padua, Pediatric Research Institute, Padua, Italy (G.P.T.)
| | - Pamela Gatto
- Laboratory of Translational Genomics, Centre for Integrative Biology, University of Trento, Trento, Italy (V.S., P.G., V.G., T.T., A.Q.); High-Throughput Screening Core Facility, Centre for Integrative Biology, University of Trento, Trento, Italy (V.A.); and Neuroblastoma Laboratory, Onco/Hematology Laboratory, SDB Department, University of Padua, Pediatric Research Institute, Padua, Italy (G.P.T.)
| | - Valentina Greco
- Laboratory of Translational Genomics, Centre for Integrative Biology, University of Trento, Trento, Italy (V.S., P.G., V.G., T.T., A.Q.); High-Throughput Screening Core Facility, Centre for Integrative Biology, University of Trento, Trento, Italy (V.A.); and Neuroblastoma Laboratory, Onco/Hematology Laboratory, SDB Department, University of Padua, Pediatric Research Institute, Padua, Italy (G.P.T.)
| | - Toma Tebaldi
- Laboratory of Translational Genomics, Centre for Integrative Biology, University of Trento, Trento, Italy (V.S., P.G., V.G., T.T., A.Q.); High-Throughput Screening Core Facility, Centre for Integrative Biology, University of Trento, Trento, Italy (V.A.); and Neuroblastoma Laboratory, Onco/Hematology Laboratory, SDB Department, University of Padua, Pediatric Research Institute, Padua, Italy (G.P.T.)
| | - Gian Paolo Tonini
- Laboratory of Translational Genomics, Centre for Integrative Biology, University of Trento, Trento, Italy (V.S., P.G., V.G., T.T., A.Q.); High-Throughput Screening Core Facility, Centre for Integrative Biology, University of Trento, Trento, Italy (V.A.); and Neuroblastoma Laboratory, Onco/Hematology Laboratory, SDB Department, University of Padua, Pediatric Research Institute, Padua, Italy (G.P.T.)
| | - Alessandro Quattrone
- Laboratory of Translational Genomics, Centre for Integrative Biology, University of Trento, Trento, Italy (V.S., P.G., V.G., T.T., A.Q.); High-Throughput Screening Core Facility, Centre for Integrative Biology, University of Trento, Trento, Italy (V.A.); and Neuroblastoma Laboratory, Onco/Hematology Laboratory, SDB Department, University of Padua, Pediatric Research Institute, Padua, Italy (G.P.T.)
| |
Collapse
|
96
|
Anti-Cancer Iron(II) Complexes of Pentadentate N-Donor Ligands: Cytotoxicity, Transcriptomics Analyses, and Mechanisms of Action. Chemistry 2014; 21:3062-72. [DOI: 10.1002/chem.201404749] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Revised: 10/10/2014] [Indexed: 01/10/2023]
|
97
|
Zhang C, Zhang F. Iron homeostasis and tumorigenesis: molecular mechanisms and therapeutic opportunities. Protein Cell 2014; 6:88-100. [PMID: 25476483 PMCID: PMC4312762 DOI: 10.1007/s13238-014-0119-z] [Citation(s) in RCA: 107] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Accepted: 11/04/2014] [Indexed: 12/21/2022] Open
Abstract
Excess iron is tightly associated with tumorigenesis in multiple human cancer types through a variety of mechanisms including catalyzing the formation of mutagenic hydroxyl radicals, regulating DNA replication, repair and cell cycle progression, affecting signal transduction in cancer cells, and acting as an essential nutrient for proliferating tumor cells. Thus, multiple therapeutic strategies based on iron deprivation have been developed in cancer therapy. During the past few years, our understanding of genetic association and molecular mechanisms between iron and tumorigenesis has expanded enormously. In this review, we briefly summarize iron homeostasis in mammals, and discuss recent progresses in understanding the aberrant iron metabolism in numerous cancer types, with a focus on studies revealing altered signal transduction in cancer cells.
Collapse
Affiliation(s)
- Caiguo Zhang
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, CO, 80045, USA,
| | | |
Collapse
|
98
|
Diamantidis MD, Papaioannou M. Toward adjunctive therapy of acute myeloid leukemia: is it feasible? Leuk Res 2014; 38:1016-1017. [PMID: 25063523 DOI: 10.1016/j.leukres.2014.06.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2014] [Revised: 06/12/2014] [Accepted: 06/28/2014] [Indexed: 11/28/2022]
Affiliation(s)
- Michael D Diamantidis
- Department of Hematology, First Department of Internal Medicine, AHEPA University General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece.
| | - Maria Papaioannou
- Department of Hematology, First Department of Internal Medicine, AHEPA University General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
99
|
Minden MD, Hogge DE, Weir SJ, Kasper J, Webster DA, Patton L, Jitkova Y, Hurren R, Gronda M, Goard CA, Rajewski LG, Haslam JL, Heppert KE, Schorno K, Chang H, Brandwein JM, Gupta V, Schuh AC, Trudel S, Yee KWL, Reed GA, Schimmer AD. Oral ciclopirox olamine displays biological activity in a phase I study in patients with advanced hematologic malignancies. Am J Hematol 2014; 89:363-8. [PMID: 24273151 DOI: 10.1002/ajh.23640] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Revised: 11/19/2013] [Accepted: 11/20/2013] [Indexed: 12/12/2022]
Abstract
The antimycotic ciclopirox olamine is an intracellular iron chelator that has anticancer activity in vitro and in vivo. We developed an oral formulation of ciclopirox olamine and conducted the first-in-human phase I study of this drug in patients with relapsed or refractory hematologic malignancies (Trial registration ID: NCT00990587). Patients were treated with 5-80 mg/m² oral ciclopirox olamine once daily for five days in 21-day treatment cycles. Pharmacokinetic and pharmacodynamic companion studies were performed in a subset of patients. Following definition of the half-life of ciclopirox olamine, an additional cohort was enrolled and treated with 80 mg/m² ciclopirox olamine four times daily. Adverse events and clinical response were monitored throughout the trial. Twenty-three patients received study treatment. Ciclopirox was rapidly absorbed and cleared with a short half-life. Plasma concentrations of an inactive ciclopirox glucuronide metabolite were greater than those of ciclopirox. Repression of survivin expression was observed in peripheral blood cells isolated from patients treated once daily with ciclopirox olamine at doses greater than 10 mg/m², demonstrating biological activity of the drug. Dose-limiting gastrointestinal toxicities were observed in patients receiving 80 mg/m² four times daily, and no dose limiting toxicity was observed at 40 mg/m² once daily. Hematologic improvement was observed in two patients. Once-daily dosing of oral ciclopirox olamine was well tolerated in patients with relapsed or refractory hematologic malignancies, and further optimization of dosing regimens is warranted in this patient population.
Collapse
Affiliation(s)
- Mark D. Minden
- Princess Margaret Cancer Centre; University Health Network; Toronto Ontario Canada
| | - Donna E. Hogge
- Division of Hematology and Leukemia/BMT; University of British Columbia; Vancouver British Columbia Canada
| | - Scott J. Weir
- University of Kansas Cancer Center; Kansas City Kansas
| | - Jim Kasper
- The Leukemia & Lymphoma Society; White Plains New York
| | | | | | - Yulia Jitkova
- Princess Margaret Cancer Centre; University Health Network; Toronto Ontario Canada
| | - Rose Hurren
- Princess Margaret Cancer Centre; University Health Network; Toronto Ontario Canada
| | - Marcela Gronda
- Princess Margaret Cancer Centre; University Health Network; Toronto Ontario Canada
| | - Carolyn A. Goard
- Princess Margaret Cancer Centre; University Health Network; Toronto Ontario Canada
| | - Lian G. Rajewski
- Biotechnology Innovation and Optimization (BIO) Center; University of Kansas; Lawrence Kansas
| | - John L. Haslam
- Biotechnology Innovation and Optimization (BIO) Center; University of Kansas; Lawrence Kansas
| | - Kathleen E. Heppert
- Biotechnology Innovation and Optimization (BIO) Center; University of Kansas; Lawrence Kansas
| | - Kevin Schorno
- University of Kansas Cancer Center; Kansas City Kansas
| | - Hong Chang
- Department of Laboratory Hematology; University Health Network; Toronto Ontario Canada
| | - Joseph M. Brandwein
- Princess Margaret Cancer Centre; University Health Network; Toronto Ontario Canada
| | - Vikas Gupta
- Princess Margaret Cancer Centre; University Health Network; Toronto Ontario Canada
| | - Andre C. Schuh
- Princess Margaret Cancer Centre; University Health Network; Toronto Ontario Canada
| | - Suzanne Trudel
- Princess Margaret Cancer Centre; University Health Network; Toronto Ontario Canada
| | - Karen W. L. Yee
- Princess Margaret Cancer Centre; University Health Network; Toronto Ontario Canada
| | | | - Aaron D. Schimmer
- Princess Margaret Cancer Centre; University Health Network; Toronto Ontario Canada
| |
Collapse
|
100
|
Chan KT, Choi MY, Lai KKY, Tan W, Tung LN, Lam HY, Tong DKH, Lee NP, Law S. Overexpression of transferrin receptor CD71 and its tumorigenic properties in esophageal squamous cell carcinoma. Oncol Rep 2014; 31:1296-304. [PMID: 24435655 DOI: 10.3892/or.2014.2981] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Accepted: 11/18/2013] [Indexed: 11/05/2022] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is the predominant type of esophageal cancer in endemic Asian regions. In the present study, we investigated the clinical implication and role of transferrin receptor CD71 in ESCC. CD71 has a physiological role in cellular iron intake and is implicated in the carcinogenesis of various types of tumors. In our cohort, more than a 2-fold upregulation of the CD71 transcript was detected in 61.5% of patients using quantitative polymerase chain reaction. Immunohistochemical analysis also showed strong membranous and cytoplasmic localization of CD71 in paraffin-embedded tumors. Staining parallel tumor sections with the proliferative marker Ki-67 revealed that the pattern of Ki-67 staining was associated with CD71 expression. Analysis of clinicopathological data indicated that CD71 overexpression can be used as an indicator for advanced T4 stage (p=0.0307). These data suggested a strong link between CD71 and ESCC. Subsequent in vitro assays using short interfering RNA (siRNA) to suppress CD71 expression confirmed the tumorigenic properties of CD71 in ESCC; cell growth inhibition and cell cycle arrest at S phase were observed in CD71-suppressed cells. The underlying mechanism involved activation of the MEK/ERK pathway. In summary, the present study provides evidence showing the tumorigenic properties of CD71 in ESCC with clinical correlations and suggests targeting CD71 as a strategy for the treatment of ESCC.
Collapse
Affiliation(s)
- Kin Tak Chan
- Department of Surgery, The University of Hong Kong, Hong Kong, SAR, P.R. China
| | - Mei Yuk Choi
- Department of Surgery, The University of Hong Kong, Hong Kong, SAR, P.R. China
| | - Kenneth K Y Lai
- Department of Surgery, The University of Hong Kong, Hong Kong, SAR, P.R. China
| | - Winnie Tan
- Department of Surgery, The University of Hong Kong, Hong Kong, SAR, P.R. China
| | - Lai Nar Tung
- Department of Surgery, The University of Hong Kong, Hong Kong, SAR, P.R. China
| | - Ho Yu Lam
- Department of Surgery, The University of Hong Kong, Hong Kong, SAR, P.R. China
| | - Daniel K H Tong
- Department of Surgery, The University of Hong Kong, Hong Kong, SAR, P.R. China
| | - Nikki P Lee
- Department of Surgery, The University of Hong Kong, Hong Kong, SAR, P.R. China
| | - Simon Law
- Department of Surgery, The University of Hong Kong, Hong Kong, SAR, P.R. China
| |
Collapse
|