1
|
Khan AEMA, Arutla V, Srivenugopal KS. Human NQO1 as a Selective Target for Anticancer Therapeutics and Tumor Imaging. Cells 2024; 13:1272. [PMID: 39120303 PMCID: PMC11311714 DOI: 10.3390/cells13151272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Revised: 07/22/2024] [Accepted: 07/25/2024] [Indexed: 08/10/2024] Open
Abstract
Human NAD(P)H-quinone oxidoreductase1 (HNQO1) is a two-electron reductase antioxidant enzyme whose expression is driven by the NRF2 transcription factor highly active in the prooxidant milieu found in human malignancies. The resulting abundance of NQO1 expression (up to 200-fold) in cancers and a barely detectable expression in body tissues makes it a selective marker of neoplasms. NQO1 can catalyze the repeated futile redox cycling of certain natural and synthetic quinones to their hydroxyquinones, consuming NADPH and generating rapid bursts of cytotoxic reactive oxygen species (ROS) and H2O2. A greater level of this quinone bioactivation due to elevated NQO1 content has been recognized as a tumor-specific therapeutic strategy, which, however, has not been clinically exploited. We review here the natural and new quinones activated by NQO1, the catalytic inhibitors, and the ensuing cell death mechanisms. Further, the cancer-selective expression of NQO1 has opened excellent opportunities for distinguishing cancer cells/tissues from their normal counterparts. Given this diagnostic, prognostic, and therapeutic importance, we and others have engineered a large number of specific NQO1 turn-on small molecule probes that remain latent but release intense fluorescence groups at near-infrared and other wavelengths, following enzymatic cleavage in cancer cells and tumor masses. This sensitive visualization/quantitation and powerful imaging technology based on NQO1 expression offers promise for guided cancer surgery, and the reagents suggest a theranostic potential for NQO1-targeted chemotherapy.
Collapse
Affiliation(s)
| | | | - Kalkunte S. Srivenugopal
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, 1406 Amarillo Research Bldg., Rm. 1102, Amarillo, TX 79106, USA; (A.E.M.A.K.); (V.A.)
| |
Collapse
|
2
|
Sharma A, Verwilst P, Li M, Ma D, Singh N, Yoo J, Kim Y, Yang Y, Zhu JH, Huang H, Hu XL, He XP, Zeng L, James TD, Peng X, Sessler JL, Kim JS. Theranostic Fluorescent Probes. Chem Rev 2024; 124:2699-2804. [PMID: 38422393 PMCID: PMC11132561 DOI: 10.1021/acs.chemrev.3c00778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 01/31/2024] [Accepted: 02/08/2024] [Indexed: 03/02/2024]
Abstract
The ability to gain spatiotemporal information, and in some cases achieve spatiotemporal control, in the context of drug delivery makes theranostic fluorescent probes an attractive and intensely investigated research topic. This interest is reflected in the steep rise in publications on the topic that have appeared over the past decade. Theranostic fluorescent probes, in their various incarnations, generally comprise a fluorophore linked to a masked drug, in which the drug is released as the result of certain stimuli, with both intrinsic and extrinsic stimuli being reported. This release is then signaled by the emergence of a fluorescent signal. Importantly, the use of appropriate fluorophores has enabled not only this emerging fluorescence as a spatiotemporal marker for drug delivery but also has provided modalities useful in photodynamic, photothermal, and sonodynamic therapeutic applications. In this review we highlight recent work on theranostic fluorescent probes with a particular focus on probes that are activated in tumor microenvironments. We also summarize efforts to develop probes for other applications, such as neurodegenerative diseases and antibacterials. This review celebrates the diversity of designs reported to date, from discrete small-molecule systems to nanomaterials. Our aim is to provide insights into the potential clinical impact of this still-emerging research direction.
Collapse
Affiliation(s)
- Amit Sharma
- Amity
School of Chemical Sciences, Amity University
Punjab, Sector 82A, Mohali 140 306, India
| | - Peter Verwilst
- Rega
Institute for Medical Research, Medicinal Chemistry, KU Leuven, Herestraat 49, Box 1041, 3000 Leuven, Belgium
| | - Mingle Li
- College
of Materials Science and Engineering, Shenzhen
University, Shenzhen 518060, China
| | - Dandan Ma
- College
of Materials Science and Engineering, Shenzhen
University, Shenzhen 518060, China
- College
of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
| | - Nem Singh
- Department
of Chemistry, Korea University, Seoul 02841, Korea
| | - Jiyoung Yoo
- Department
of Chemistry, Korea University, Seoul 02841, Korea
| | - Yujin Kim
- Department
of Chemistry, Korea University, Seoul 02841, Korea
| | - Ying Yang
- School of
Light Industry and Food Engineering, Guangxi
University, Nanning, Guangxi 530004, China
| | - Jing-Hui Zhu
- College
of Materials Science and Engineering, Shenzhen
University, Shenzhen 518060, China
- College
of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
| | - Haiqiao Huang
- College
of Materials Science and Engineering, Shenzhen
University, Shenzhen 518060, China
- College
of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
| | - Xi-Le Hu
- Key
Laboratory for Advanced Materials and Joint International Research
Laboratory of Precision Chemistry and Molecular Engineering, Feringa
Nobel Prize Scientist Joint Research Center, School of Chemistry and
Molecular Engineering, East China University
of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Xiao-Peng He
- Key
Laboratory for Advanced Materials and Joint International Research
Laboratory of Precision Chemistry and Molecular Engineering, Feringa
Nobel Prize Scientist Joint Research Center, School of Chemistry and
Molecular Engineering, East China University
of Science and Technology, 130 Meilong Road, Shanghai 200237, China
- National
Center for Liver Cancer, the International Cooperation Laboratory
on Signal Transduction, Eastern Hepatobiliary
Surgery Hospital, Shanghai 200438, China
| | - Lintao Zeng
- School of
Light Industry and Food Engineering, Guangxi
University, Nanning, Guangxi 530004, China
| | - Tony D. James
- Department
of Chemistry, University of Bath, Bath BA2 7AY, United Kingdom
- School
of Chemistry and Chemical Engineering, Henan
Normal University, Xinxiang 453007, China
| | - Xiaojun Peng
- College
of Materials Science and Engineering, Shenzhen
University, Shenzhen 518060, China
- State
Key Laboratory of Fine Chemicals, Dalian
University of Technology, Dalian 116024, China
| | - Jonathan L. Sessler
- Department
of Chemistry, The University of Texas at
Austin, Texas 78712-1224, United
States
| | - Jong Seung Kim
- Department
of Chemistry, Korea University, Seoul 02841, Korea
- TheranoChem Incorporation, Seongbuk-gu, Seoul 02841, Korea
| |
Collapse
|
3
|
Shao Z, Wang H, Ren H, Sun Y, Chen X. The Anticancer Effect of Napabucasin (BBI608), a Natural Naphthoquinone. Molecules 2023; 28:5678. [PMID: 37570646 PMCID: PMC10420168 DOI: 10.3390/molecules28155678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 07/23/2023] [Accepted: 07/24/2023] [Indexed: 08/13/2023] Open
Abstract
Napabucasin (also known as BBI608) is a natural naphthoquinone originally identified as a cancer cell stemness inhibitor. Accumulated in vitro and in vivo evidence demonstrated that napabucasin showed significant anticancer effects in various types of cancers. Napabucasin inhibits cancer cell proliferation, induces apoptosis and cell cycle arrest, and suppresses metastasis and relapse. Such anticancer activities of napabucasin mainly rely on the inhibition of cancer stemness by targeting signal transducer and activator of transcription 3 (STAT3) and its related gene inhibition. However, several novel molecular targets for napabucasin, such as NAD(P)H:quinone oxidoreductase 1 (NQO1) and thioredoxin reductase 1 (TrxR1), have been reported. Napabucasin represents a promising anticancer lead for multiple cancers. In this mini review, the anticancer potential and the molecular mechanism of napabucasin will be briefly highlighted.
Collapse
Affiliation(s)
- Zeyang Shao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical, University of Macau, Macao, China; (Z.S.); (H.W.); (H.R.)
| | - Heng Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical, University of Macau, Macao, China; (Z.S.); (H.W.); (H.R.)
| | - Haiyan Ren
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical, University of Macau, Macao, China; (Z.S.); (H.W.); (H.R.)
| | - Yinxiang Sun
- Zhuhai People’s Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai 519000, China
| | - Xiuping Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical, University of Macau, Macao, China; (Z.S.); (H.W.); (H.R.)
- Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Macao, China
- MoE Frontiers Science Center for Precision Oncology, University of Macau, Macao, China
- GMU-GIBH Joint School of Life Sciences, The Guangdong-Hong Kong-Macau Joint Laboratory for Cell Fate Regulation and Disease, Guangzhou Medical University, Guangzhou 511436, China
| |
Collapse
|
4
|
Otsuka N, Shimizu K, Taniai M, Tokushige K. Risk factors for fatty pancreas and effects of fatty infiltration on pancreatic cancer. Front Physiol 2023; 14:1243983. [PMID: 37664430 PMCID: PMC10470060 DOI: 10.3389/fphys.2023.1243983] [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: 06/21/2023] [Accepted: 07/25/2023] [Indexed: 09/05/2023] Open
Abstract
Objective: This study clarified the risk factors and pathophysiology of pancreatic cancer by examining the factors associated with fatty pancreas. Methods: The degree of fatty pancreas, background factors, and incidence of pancreatic cancer were examined among nonalcoholic fatty liver disease (NAFLD) patients (n = 281) and intraductal papillary mucinous neoplasm (IPMN) patients with a family history of pancreatic cancer (n = 38). The presence of fatty pancreas was confirmed by the pancreatic CT value/splenic CT value ratio (P/S ratio). Immunohistochemical staining was performed on 10 cases with fatty pancreas, confirmed via postoperative pathology. Results: Fatty pancreas occurred in 126 patients (44.8%) in the NAFLD group who were older (p = 0.0002) and more likely to have hypertension (p < 0.0001). The IPMN group had 18 patients (47.4%) with fatty pancreas, included more men than women (p = 0.0056), and was more likely to have patients with hypertension (p = 0.0010). On histological examination, a significant infiltration of adipocytes into the acini from the pancreatic interstitium induced atrophy of the pancreatic parenchyma, and both M1 and M2 macrophages were detected in the area where adipocytes invaded the pancreatic parenchyma. Accumulation of p62 and increased positive staining of NQO1 molecules related to autophagy dysfunction were detected in pancreatic acinar cells in the fatty area, acinar-ductal metaplasia, and pancreatic cancer cells. The rate of p62-positive cell area and that of NQO1-positive cell area were significantly higher in the fatty pancreatic region than those in the control lesion (pancreatic region with few adipocyte infiltration). Furthermore, the rate of p62-positive cell area or that of NQO1-positive cell area showed strong positive correlations with the rate of fatty pancreatic lesion. These results suggest that adipocyte invasion into the pancreatic parenthyme induced macrophage infiltration and autophagy substrate p62 accumulation. High levels of NQO1 expression in the fatty area may be dependent on p62 accumulation. Conclusion: Hypertension was a significant risk factor for fatty pancreas in patients with NAFLD and IPMN. In fatty pancreas, fatty infiltration into the pancreatic parenchyme might induce autophagy dysfunction, resulting in activation of antioxidant proteins NQO1. Thus, patients with fatty pancreas require careful follow-up.
Collapse
Affiliation(s)
- Nao Otsuka
- Department of Internal Medicine, Institute of Gastroenterology, Tokyo Women’s Medical University, Tokyo, Japan
| | - Kyoko Shimizu
- Department of Internal Medicine, Institute of Gastroenterology, Tokyo Women’s Medical University, Tokyo, Japan
- Shinjuku Mitsui Building Clinic, Tokyo, Japan
| | - Makiko Taniai
- Department of Internal Medicine, Institute of Gastroenterology, Tokyo Women’s Medical University, Tokyo, Japan
| | - Katsutoshi Tokushige
- Department of Internal Medicine, Institute of Gastroenterology, Tokyo Women’s Medical University, Tokyo, Japan
| |
Collapse
|
5
|
Gong Q, Wang P, Li T, Yu Z, Yang L, Wu C, Hu J, Yang F, Zhang X, Li X. Novel NQO1 substrates bearing two nitrogen redox centers: Design, synthesis, molecular dynamics simulations, and antitumor evaluation. Bioorg Chem 2023; 134:106480. [PMID: 36958178 DOI: 10.1016/j.bioorg.2023.106480] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 02/20/2023] [Accepted: 03/12/2023] [Indexed: 03/17/2023]
Abstract
By analyzing the crystal structure of NQO1, an additional binding region for the ligand was discovered. In this study, a series of derivatives with a novel skeleton bearing two nitrogen redox centers were designed by introducing amines or hydrazines to fit with the novel binding region of NQO1. Compound 24 with a (4-fluorophenyl)hydrazine substituent was identified as the most efficient substrate for NQO1 with the reduction rate and catalytic efficiency of 1972 ± 82 μmol NADPH/min/μmol NQO1 and 6.4 ± 0.4 × 106 M-1s-1, respectively. Molecular dynamics (MD) simulation revealed that the distances between the nitrogen atom of the redox centers and the key Tyr128 and Tyr126 residues were 3.5 Å (N1-Tyr128) and 3.4 Å (N2-Tyr126), respectively. Compound 24 (IC50/A549 = 0.69 ± 0.09 μM) showed potent antitumor activity against A549 cells both in vitro and in vivo through ROS generation via NQO1-mediated redox cycling, leading to a promising NQO1-targeting antitumor candidate.
Collapse
Affiliation(s)
- Qijie Gong
- Jiangsu Key Laboratory of Drug Design and Optimization, and Department of Chemistry, China Pharmaceutical University, Nanjing 211198, China
| | - Pengfei Wang
- Jiangsu Key Laboratory of Drug Design and Optimization, and Department of Chemistry, China Pharmaceutical University, Nanjing 211198, China; Department of Pharmaceutical Engineering, China Pharmaceutical University, Nanjing 211198, China
| | - Tian Li
- Jiangsu Key Laboratory of Drug Design and Optimization, and Department of Chemistry, China Pharmaceutical University, Nanjing 211198, China; Department of Pharmaceutical Engineering, China Pharmaceutical University, Nanjing 211198, China
| | - Zhan Yu
- The Affiliated Jiangning Hospital of NJMU, Nanjing Medical University (NJMU), Nanjing 211199, China; Jiangning Clinical Medical College of Jiangsu University, Nanjing 211100, China.
| | - Le Yang
- Jiangsu Key Laboratory of Drug Design and Optimization, and Department of Chemistry, China Pharmaceutical University, Nanjing 211198, China
| | - Chenyang Wu
- Jiangsu Key Laboratory of Drug Design and Optimization, and Department of Chemistry, China Pharmaceutical University, Nanjing 211198, China
| | - Jiabao Hu
- Jiangsu Key Laboratory of Drug Design and Optimization, and Department of Chemistry, China Pharmaceutical University, Nanjing 211198, China
| | - Fulai Yang
- Jiangsu Key Laboratory of Drug Design and Optimization, and Department of Chemistry, China Pharmaceutical University, Nanjing 211198, China.
| | - Xiaojin Zhang
- Jiangsu Key Laboratory of Drug Design and Optimization, and Department of Chemistry, China Pharmaceutical University, Nanjing 211198, China.
| | - Xiang Li
- Department of Pharmaceutical Engineering, China Pharmaceutical University, Nanjing 211198, China.
| |
Collapse
|
6
|
Juvekar V, Lee HW, Lee DJ, Kim HM. Two-photon fluorescent probes for quantitative bio-imaging analysis in live tissues. Trends Analyt Chem 2022. [DOI: 10.1016/j.trac.2022.116787] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
7
|
Preethi S, Arthiga K, Patil AB, Spandana A, Jain V. Review on NAD(P)H dehydrogenase quinone 1 (NQO1) pathway. Mol Biol Rep 2022; 49:8907-8924. [PMID: 35347544 DOI: 10.1007/s11033-022-07369-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 03/11/2022] [Indexed: 12/14/2022]
Abstract
NQO1 is an enzyme present in humans which is encoded by NQO1 gene. It is a protective antioxidant agent, versatile cytoprotective agent and regulates the oxidative stresses of chromatin binding proteins for DNA damage in cancer cells. The oxidization of cellular pyridine nucleotides causes structural alterations to NQO1 and changes in its capacity to binding of proteins. A strategy based on NQO1 to have protective effect against cancer was developed by organic components to enhance NQO1 expression. The quinone derivative compounds like mitomycin C, RH1, E09 (Apaziquone) and β-lapachone causes cell death by NQO1 reduction of two electrons. It was also known to be overexpressed in various tumor cells of breast, lung, cervix, pancreas and colon when it was compared with normal cells in humans. The mechanism of NQO1 by the reduction of FAD by NADPH to form FADH2 is by two ways to inhibit cancer cell development such as suppression of carcinogenic metabolic activation and prevention of carcinogen formation. The NQO1 exhibit suppression of chemical-mediated carcinogenesis by various properties of NQO1 which includes, detoxification of quinone scavenger of superoxide anion radical, antioxidant enzyme, protein stabilizer. This review outlines the NQO1 structure, mechanism of action to inhibit the cancer cell, functions of NQO1 against oxidative stress, drugs acting on NQO1 pathways, clinical significance.
Collapse
Affiliation(s)
- S Preethi
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Sri Shivarathreeshwara Nagar, Mysuru, Karnataka, 570015, India
| | - K Arthiga
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Sri Shivarathreeshwara Nagar, Mysuru, Karnataka, 570015, India
| | - Amit B Patil
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Sri Shivarathreeshwara Nagar, Mysuru, Karnataka, 570015, India
| | - Asha Spandana
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Sri Shivarathreeshwara Nagar, Mysuru, Karnataka, 570015, India
| | - Vikas Jain
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Sri Shivarathreeshwara Nagar, Mysuru, Karnataka, 570015, India.
| |
Collapse
|
8
|
Identification of DNA methylation-driven genes and construction of a nomogram to predict overall survival in pancreatic cancer. BMC Genomics 2021; 22:791. [PMID: 34732125 PMCID: PMC8567715 DOI: 10.1186/s12864-021-08097-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Accepted: 10/12/2021] [Indexed: 12/11/2022] Open
Abstract
Background The incidence and mortality of pancreatic cancer (PC) has gradually increased. The aim of this study was to identify survival-related DNA methylation (DNAm)-driven genes and establish a nomogram to predict outcomes in patients with PC. Methods The gene expression, DNA methylation database, and PC clinical samples were downloaded from TCGA. DNAm-driven genes were identified by integrating analyses of gene expression and DNA methylation data. Survival-related DNAm-driven genes were screened via univariate, least absolute shrinkage and selection operator (LASSO), and multivariate Cox regression analyses to develop a risk score model for prognosis. Based on analyses of clinical parameters and risk score, a nomogram was built and validated. The independent cohort from GEO database were used for external validation. Results A total of 16 differentially expressed methylation-driven genes were identified. Based on LASSO Cox regression and multivariate Cox regression analysis, six genes (FERMT1, LIPH, LAMA3, PPP1R14D, NQO1, VSIG2) were chosen to develop the risk score model. In the Kaplan–Meier analysis, age, T stage, N stage, AJCC stage, radiation therapy history, tumor size, surgery type performed, pathological type, chemotherapy history, and risk score were potential prognostic factors in PC (P < 0.1). In the multivariate analysis, stage, chemotherapy, and risk score were significantly correlated to overall survival (P < 0.05). The nomogram was constructed with the three variables (stage, chemotherapy, and risk score) for predicting the 1-year, 2-year, and 3-year survival rates of PC patients. Nomogram performance was assessed by receiver operating characteristic (ROC) curves and calibration curves. 1-year, 2-year and 3-year AUC of nomogram model was 0.899, 0.765 and 0.776, respectively. Conclusions In our study, we successfully identified the six DNAm-driven genes (FERMT1, LIPH, LAMA3, PPP1R14D, NQO1, VSIG2) with a relationship to the outcomes of PC patients. The nomogram including stage, chemotherapy, and risk score could be used to predict survival in PC patients. Supplementary Information The online version contains supplementary material available at 10.1186/s12864-021-08097-w.
Collapse
|
9
|
Yu J, Li S, Zeng X, Song J, Hu S, Cheng S, Chen C, Luo H, Pan W. Design, synthesis, and evaluation of proliferation inhibitory activity of novel L-shaped ortho-quinone analogs as anticancer agents. Bioorg Chem 2021; 117:105383. [PMID: 34656969 DOI: 10.1016/j.bioorg.2021.105383] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 09/06/2021] [Accepted: 09/20/2021] [Indexed: 12/24/2022]
Abstract
In this study, we present the design and synthesis of novel fully synthetic L-shaped ortho-quinone analogs with tanshinone IIA as the lead compoud, which is a molecule with numerous pharmacological benefits and potential to treat life-threatening diseases, such as cancer and viral infections. 24 L-shaped ortho-quinone analogs were designed and synthesized via click chemistry and introduced 1,2,3-triazole at the C-2 terminal of the furan ring. The cytotoxicity of these analogs toward different cancer cell lines was investigated in vitro. The new TD compounds showed potent inhibitory activities toward prostate cancer (PC3), leukemia (K562), breast cancer (MDA-231), lung cancer (A549), and cervical cancer (Hela) cell lines. Among them, TD1, TD11, and TD17 showed excellent broad-spectrum cytotoxic effects on five cancer cell lines by inducing apoptosis and arresting the cell cycle phase. Besides, TD1, TD11, and TD17 could target-bind with NQO1 protein in the prostate cancer cells PC3 leukemia cells K562. The results showed that removing the methyl group at C-3 and introducing 1,2,3-triazoles at the C-2 terminal of the furan ring were effective strategies for improving the broad-spectrum anticancer activity of L-shaped ortho-quinone analogs.
Collapse
Affiliation(s)
- Jia Yu
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang 550014, China
| | - Shengyou Li
- The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang 550014, China
| | - Xueyi Zeng
- The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang 550014, China
| | - Junrong Song
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang 550014, China
| | - Shengcao Hu
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang 550014, China
| | - Sha Cheng
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang 550014, China
| | - Chao Chen
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang 550014, China.
| | - Heng Luo
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang 550014, China.
| | - Weidong Pan
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang 550014, China.
| |
Collapse
|
10
|
Gong Q, Yang F, Hu J, Li T, Wang P, Li X, Zhang X. Rational designed highly sensitive NQO1-activated near-infrared fluorescent probe combined with NQO1 substrates in vivo: An innovative strategy for NQO1-overexpressing cancer theranostics. Eur J Med Chem 2021; 224:113707. [PMID: 34303080 DOI: 10.1016/j.ejmech.2021.113707] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 07/05/2021] [Accepted: 07/16/2021] [Indexed: 12/16/2022]
Abstract
Since NQO1 is overexpressed in many cancer cells, it can be used as a biomarker for cancer diagnosis and targeted therapy. NQO1 substrates show potent anticancer activity through the redox cycle mediated by NQO1, while the NQO1 probes can monitor NQO1 levels in cancers. High sensitivity of probes is needed for diagnostic imaging in clinic. In this study, based on the analysis of NQO1 catalytic pocket, the naphthoquinone trigger group 13 rationally designed by expanding the aromatic plane of the benzoquinone trigger group 10 shows significantly increased sensitivity to NQO1. The sensitivity of the naphthoquinone trigger group-based probe A was eight times higher than that of benzoquinone trigger group-based probe B in vivo. Probe A was selectively and efficiently sensitive to NQO1 with good safety profile and plasma stability, enabling its combination with NQO1 substrates in vivo for NQO1-overexpressing cancer theranostics for the first time.
Collapse
Affiliation(s)
- Qijie Gong
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China; Laboratory of Drug Design and Discovery, Department of Chemistry, China Pharmaceutical University, Nanjing, 211198, China
| | - Fulai Yang
- Laboratory of Drug Design and Discovery, Department of Chemistry, China Pharmaceutical University, Nanjing, 211198, China
| | - Jiabao Hu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China; Laboratory of Drug Design and Discovery, Department of Chemistry, China Pharmaceutical University, Nanjing, 211198, China
| | - Tian Li
- Department of Pharmaceutical Engineering, China Pharmaceutical University, Nanjing, 211198, China
| | - Pengfei Wang
- Department of Pharmaceutical Engineering, China Pharmaceutical University, Nanjing, 211198, China
| | - Xiang Li
- Department of Pharmaceutical Engineering, China Pharmaceutical University, Nanjing, 211198, China.
| | - Xiaojin Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China; Laboratory of Drug Design and Discovery, Department of Chemistry, China Pharmaceutical University, Nanjing, 211198, China.
| |
Collapse
|
11
|
Wu Z, Wang Q, Yang H, Wang J, Li W, Liu G, Yang Y, Zhao Y, Tang Y. Discovery of Natural Products Targeting NQO1 via an Approach Combining Network-Based Inference and Identification of Privileged Substructures. J Chem Inf Model 2021; 61:2486-2498. [PMID: 33955748 DOI: 10.1021/acs.jcim.1c00260] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
NAD(P)H:quinone oxidoreductase 1 (NQO1) has been shown to be a potential therapeutic target for various human diseases, such as cancer and neurodegenerative disorders. Recent advances in computational methods, especially network-based methods, have made it possible to identify novel compounds for a target with high efficiency and low cost. In this study, we designed a workflow combining network-based methods and identification of privileged substructures to discover new compounds targeting NQO1 from a natural product library. According to the prediction results, we purchased 56 compounds for experimental validation. Without the assistance of privileged substructures, 31 compounds (31/56 = 55.4%) showed IC50 < 100 μM, and 11 compounds (11/56 = 19.6%) showed IC50 < 10 μM. With the assistance of privileged substructures, the two success rates were increased to 61.8 and 26.5%, respectively. Seven natural products further showed inhibitory activity on NQO1 at the cellular level, which may serve as lead compounds for further development. Moreover, network analysis revealed that osthole may exert anticancer effects against multiple cancer types by inhibiting not only carbonic anhydrases IX and XII but also NQO1. Our workflow and computational methods can be easily applied in other targets and become useful tools in drug discovery and development.
Collapse
Affiliation(s)
- Zengrui Wu
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Qiaohui Wang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China.,Optogenetics & Synthetic Biology Interdisciplinary Research Center, State Key Laboratory of Bioreactor Engineering, Shanghai Collaborative Innovation Center for Biomanufacturing Technology, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Hongbin Yang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Jiye Wang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Weihua Li
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Guixia Liu
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Yi Yang
- Optogenetics & Synthetic Biology Interdisciplinary Research Center, State Key Laboratory of Bioreactor Engineering, Shanghai Collaborative Innovation Center for Biomanufacturing Technology, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China.,CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of Neuroscience, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yuzheng Zhao
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China.,Optogenetics & Synthetic Biology Interdisciplinary Research Center, State Key Laboratory of Bioreactor Engineering, Shanghai Collaborative Innovation Center for Biomanufacturing Technology, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Yun Tang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| |
Collapse
|
12
|
Wiechmann S, Saupp E, Schilling D, Heinzlmeir S, Schneider G, Schmid RM, Combs SE, Kuster B, Dobiasch S. Radiosensitization by Kinase Inhibition Revealed by Phosphoproteomic Analysis of Pancreatic Cancer Cells. Mol Cell Proteomics 2020; 19:1649-1663. [PMID: 32651227 PMCID: PMC8014995 DOI: 10.1074/mcp.ra120.002046] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 06/22/2020] [Indexed: 01/12/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive cancers and known for its extensive genetic heterogeneity, high therapeutic resistance, and strong variation in intrinsic radiosensitivity. To understand the molecular mechanisms underlying radioresistance, we screened the phenotypic response of 38 PDAC cell lines to ionizing radiation. Subsequent phosphoproteomic analysis of two representative sensitive and resistant lines led to the reproducible identification of 7,800 proteins and 13,000 phosphorylation sites (p-sites). Approximately 700 p-sites on 400 proteins showed abundance changes after radiation in all cell lines regardless of their phenotypic sensitivity. Apart from recapitulating known radiation response phosphorylation markers such as on proteins involved in DNA damage repair, the analysis uncovered many novel members of a radiation-responsive signaling network that was apparent only at the level of protein phosphorylation. These regulated p-sites were enriched in potential ATM substrates and in vitro kinase assays corroborated 10 of these. Comparing the proteomes and phosphoproteomes of radiosensitive and -resistant cells pointed to additional tractable radioresistance mechanisms involving apoptotic proteins. For instance, elevated NADPH quinine oxidoreductase 1 (NQO1) expression in radioresistant cells may aid in clearing harmful reactive oxygen species. Resistant cells also showed elevated phosphorylation levels of proteins involved in cytoskeleton organization including actin dynamics and focal adhesion kinase (FAK) activity and one resistant cell line showed a strong migration phenotype. Pharmacological inhibition of the kinases FAK by Defactinib and of CHEK1 by Rabusertib showed a statistically significant sensitization to radiation in radioresistant PDAC cells. Together, the presented data map a comprehensive molecular network of radiation-induced signaling, improves the understanding of radioresistance and provides avenues for developing radiotherapeutic strategies.
Collapse
Affiliation(s)
- Svenja Wiechmann
- Chair of Proteomics and Bioanalytics, Technical University of Munich, Freising, Germany; German Cancer Consortium, Munich, Germany; German Cancer Center, Heidelberg, Germany
| | - Elena Saupp
- Department of Radiation Oncology, Technical University of Munich, Klinikum rechts der Isar, Munich, Germany
| | - Daniela Schilling
- Department of Radiation Oncology, Technical University of Munich, Klinikum rechts der Isar, Munich, Germany; Institute of Radiation Medicine, Department of Radiation Sciences, Helmholtz Zentrum München, Neuherberg, Germany
| | - Stephanie Heinzlmeir
- Chair of Proteomics and Bioanalytics, Technical University of Munich, Freising, Germany
| | - Günter Schneider
- Medical Clinic and Polyclinic II, Klinikum rechts der Isar, Technical University Munich, München, Germany
| | - Roland M Schmid
- Medical Clinic and Polyclinic II, Klinikum rechts der Isar, Technical University Munich, München, Germany
| | - Stephanie E Combs
- German Cancer Consortium, Munich, Germany; German Cancer Center, Heidelberg, Germany; Department of Radiation Oncology, Technical University of Munich, Klinikum rechts der Isar, Munich, Germany; Institute of Radiation Medicine, Department of Radiation Sciences, Helmholtz Zentrum München, Neuherberg, Germany
| | - Bernhard Kuster
- Chair of Proteomics and Bioanalytics, Technical University of Munich, Freising, Germany; German Cancer Consortium, Munich, Germany; German Cancer Center, Heidelberg, Germany; Bavarian Center for Biomolecular Mass Spectrometry, Technical University of Munich, Freising, Germany
| | - Sophie Dobiasch
- German Cancer Consortium, Munich, Germany; German Cancer Center, Heidelberg, Germany; Department of Radiation Oncology, Technical University of Munich, Klinikum rechts der Isar, Munich, Germany; Institute of Radiation Medicine, Department of Radiation Sciences, Helmholtz Zentrum München, Neuherberg, Germany.
| |
Collapse
|
13
|
Tsao YC, Chang YJ, Wang CH, Chen L. Discovery of Isoplumbagin as a Novel NQO1 Substrate and Anti-Cancer Quinone. Int J Mol Sci 2020; 21:E4378. [PMID: 32575541 PMCID: PMC7352187 DOI: 10.3390/ijms21124378] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 06/16/2020] [Accepted: 06/18/2020] [Indexed: 12/27/2022] Open
Abstract
Isoplumbagin (5-hydroxy-3-methyl-1,4-naphthoquinone), a naturally occurring quinone from Lawsonia inermis and Plumbago europaea, has been reported to have anti-inflammatory and antimicrobial activity. Inflammation has long been implicated in cancer progression. In this study, we examined the anticancer effect of chemically synthesized isoplumbagin. Our results revealed that isoplumbagin treatment suppressed cell viability and invasion of highly invasive oral squamous cell carcinoma (OSCC) OC3-IV2 cells, glioblastoma U87 cells, non-small cell lung carcinoma H1299 cells, prostate cancer PC3 cells, and cervical cancer HeLa cells by using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) and Boyden chamber assays. In vivo studies demonstrate the inhibitory effect of 2 mg/kg isoplumbagin on the growth of orthotopic xenograft tumors derived from OSCC cells. Mechanistically, isoplumbagin exerts its cytotoxic effect through acting as a substrate of reduced nicotinamide adenine dinucleotide phosphate [NAD(P)H] dehydrogenase quinone 1 (NQO1) to generate hydroquinone, which reverses mitochondrial fission phenotype, reduces mitochondrial complex IV activity, and thus compromises mitochondrial function. Collectively, this work reveals an anticancer activity of isoplumbagin mainly through modulating mitochondrial dynamics and function.
Collapse
Affiliation(s)
- Yen-Chi Tsao
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu 30013, Taiwan; (Y.-C.T.); (C.-H.W.)
| | - Yu-Jung Chang
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu 30013, Taiwan; (Y.-C.T.); (C.-H.W.)
| | - Chun-Hsien Wang
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu 30013, Taiwan; (Y.-C.T.); (C.-H.W.)
| | - Linyi Chen
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu 30013, Taiwan; (Y.-C.T.); (C.-H.W.)
- Department of Medical Science, National Tsing Hua University, Hsinchu 30013, Taiwan
| |
Collapse
|
14
|
Xue T, Jia X, Wang J, Xiang J, Wang W, Du J, He Y. “Turn‐On” Activatable AIE Dots for Tumor Hypoxia Imaging. Chemistry 2019; 25:9634-9638. [DOI: 10.1002/chem.201902296] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Indexed: 12/27/2022]
Affiliation(s)
- Tianhao Xue
- Department of Chemical EngineeringKey Laboratory of Advanced Materials (MOE)Tsinghua University Beijing 100084 China
| | - Xiangqian Jia
- School of Pharmaceutical SciencesTsinghua University Beijing 100084 China
| | - Jilei Wang
- Department of Chemical EngineeringKey Laboratory of Advanced Materials (MOE)Tsinghua University Beijing 100084 China
| | - Jingyuan Xiang
- Department of Chemical EngineeringKey Laboratory of Advanced Materials (MOE)Tsinghua University Beijing 100084 China
| | - Wei Wang
- Department of Chemical EngineeringKey Laboratory of Advanced Materials (MOE)Tsinghua University Beijing 100084 China
| | - Juanjuan Du
- School of Pharmaceutical SciencesTsinghua University Beijing 100084 China
| | - Yaning He
- Department of Chemical EngineeringKey Laboratory of Advanced Materials (MOE)Tsinghua University Beijing 100084 China
| |
Collapse
|
15
|
Selvakumar R, Anantha Krishnan D, Ramakrishnan C, Velmurugan D, Gunasekaran K. Identification of novel NAD(P)H dehydrogenase [quinone] 1 antagonist using computational approaches. J Biomol Struct Dyn 2019; 38:682-696. [DOI: 10.1080/07391102.2019.1585291] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Rajendran Selvakumar
- CAS in Crystallography and Biophysics, University of Madras, Chennai, Tamil Nadu, India
| | | | - Chandrasekaran Ramakrishnan
- Department of Biotechnology, Bhupat Jyoti Mehta School of Biosciences, Indian Institute of Technology (IIT) Madras, Chennai, Tamil Nadu, India
| | - Devadasan Velmurugan
- CAS in Crystallography and Biophysics, University of Madras, Chennai, Tamil Nadu, India
| | - Krishnasamy Gunasekaran
- CAS in Crystallography and Biophysics, University of Madras, Chennai, Tamil Nadu, India
- Bioinformatics Infrastructure Facility, University of Madras, Chennai, Tamil Nadu, India
| |
Collapse
|
16
|
Cho MK, Juvekar V, Lim CS, Noh C, Shin SJ, Kim HM. A Highly Sensitive Two‐Photon Ratiometric Probe for Rapid Detection of the hNQO1 Enzyme in Colon Cancer Tissue. ASIAN J ORG CHEM 2019. [DOI: 10.1002/ajoc.201800694] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Myoung Ki Cho
- Department of Energy System Research and Department of ChemistryAjou University Suwon, Gyeonggi-do 443-749 Republic of Korea
| | - Vinayak Juvekar
- Department of Energy System Research and Department of ChemistryAjou University Suwon, Gyeonggi-do 443-749 Republic of Korea
| | - Chang Su Lim
- Department of Energy System Research and Department of ChemistryAjou University Suwon, Gyeonggi-do 443-749 Republic of Korea
| | - Choong‐Kyun Noh
- Department of GastroenterologyAjou University School of Medicine Suwon 443-721 Republic of Korea
| | - Sung Jae Shin
- Department of GastroenterologyAjou University School of Medicine Suwon 443-721 Republic of Korea
| | - Hwan Myung Kim
- Department of Energy System Research and Department of ChemistryAjou University Suwon, Gyeonggi-do 443-749 Republic of Korea
| |
Collapse
|
17
|
Medina-Carmona E, Rizzuti B, Martín-Escolano R, Pacheco-García JL, Mesa-Torres N, Neira JL, Guzzi R, Pey AL. Phosphorylation compromises FAD binding and intracellular stability of wild-type and cancer-associated NQO1: Insights into flavo-proteome stability. Int J Biol Macromol 2018; 125:1275-1288. [PMID: 30243998 DOI: 10.1016/j.ijbiomac.2018.09.108] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 08/30/2018] [Accepted: 09/18/2018] [Indexed: 02/07/2023]
Abstract
Over a quarter million of protein phosphorylation sites have been identified so far, although the effects of site-specific phosphorylation on protein function and stability, as well as their possible impact in the phenotypic manifestation in genetic diseases are vastly unknown. We investigated here the effects of phosphorylating S82 in human NADP(H):quinone oxidoreductase 1, a representative example of disease-associated flavoprotein in which protein stability is coupled to the intracellular flavin levels. Additionally, the cancer-associated P187S polymorphism causes inactivation and destabilization of the enzyme. By using extensive in vitro and in silico characterization of phosphomimetic S82D mutations, we showed that S82D locally affected the flavin binding site of the wild-type (WT) and P187S proteins thus altering flavin binding affinity, conformational stability and aggregation propensity. Consequently, the phosphomimetic S82D may destabilize the WT protein intracellularly by promoting the formation of the degradation-prone apo-protein. Noteworthy, WT and P187S proteins respond differently to the phosphomimetic mutation in terms of intracellular stability, further supporting differences in molecular recognition of these two variants by the proteasomal degradation pathway. We propose that phosphorylation could have critical consequences on stability and function of human flavoproteins, important for our understanding of genotype-phenotype relationships in their related genetic diseases.
Collapse
Affiliation(s)
| | - Bruno Rizzuti
- CNR-NANOTEC, Licryl-UOS Cosenza and CEMIF.Cal, Department of Physics, University of Calabria, 87036 Rende, Italy
| | - Rubén Martín-Escolano
- Department of Parasitology, Instituto de Investigación Biosanitaria (ibs.Granada), Hospitales Universitarios De Granada/University of Granada, 18071 Granada, Spain
| | | | - Noel Mesa-Torres
- Department of Physical Chemistry, University of Granada, 18071 Granada, Spain
| | - José L Neira
- Instituto de Biología Molecular y Celular, Universidad Miguel Hernández, Avda. del Ferrocarril s/n, 03202 Elche, Alicante, Spain; Instituto de Biocomputación y Física de los Sistemas Complejos (BIFI), 50009 Zaragoza, Spain
| | - Rita Guzzi
- CNR-NANOTEC, Licryl-UOS Cosenza and CEMIF.Cal, Department of Physics, University of Calabria, 87036 Rende, Italy; Molecular Biophysics Laboratory, Department of Physics, University of Calabria, 87036 Rende, Italy
| | - Angel L Pey
- Department of Physical Chemistry, University of Granada, 18071 Granada, Spain.
| |
Collapse
|
18
|
Guo P, Zhang H, Zhou J, Gallou F, Parmentier M, Wang H. Micelle-Enabled Suzuki–Miyaura Cross-Coupling of Heteroaryl Boronate Esters. J Org Chem 2018; 83:7523-7527. [DOI: 10.1021/acs.joc.8b00257] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Pengfei Guo
- Chemical & Analytical Development, Suzhou Novartis Pharma Technology Company Limited, Changshu, Jiangsu 215537, China
| | - Hao Zhang
- Chemical & Analytical Development, Suzhou Novartis Pharma Technology Company Limited, Changshu, Jiangsu 215537, China
| | - Jianguang Zhou
- Chemical & Analytical Development, Suzhou Novartis Pharma Technology Company Limited, Changshu, Jiangsu 215537, China
| | - Fabrice Gallou
- Chemical & Analytical Development, Novartis Pharma AG, 4056 Basel, Switzerland
| | - Michael Parmentier
- Chemical & Analytical Development, Novartis Pharma AG, 4056 Basel, Switzerland
| | - Hui Wang
- Chemical & Analytical Development, Suzhou Novartis Pharma Technology Company Limited, Changshu, Jiangsu 215537, China
| |
Collapse
|
19
|
Fatima R, Akhtar K, Hossain MM, Ahmad R. Chromium oxide nanoparticle-induced biochemical and histopathological alterations in the kidneys and brain of Wistar rats. Toxicol Ind Health 2017; 33:911-921. [PMID: 29069986 DOI: 10.1177/0748233717735266] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Chromium oxide nanoparticles (Cr2O3 NPs) have a wide range of applications in industry. They are used as pigments, catalysts, wear-resistant or high-temperature-resistant coating material and are used in liquid crystal displays. In view of ever escalating use of NPs, risk assessment becomes obligatory to ensure the safety of both human health and the ecosystem. The present study was designed and conducted to evaluate biochemical changes and histopathological alterations in kidneys and brain of rats, following exposure to Cr2O3 NPs. Male Wistar rats were divided into low-dose (50 µg/100 g body weight (bwt) groups and high-dose (200 µg/100 g bwt) groups. Each group type received oral administration of Cr2O3 NPs for multiple durations (single dosing, once daily for 7 days and once daily for 14 days, respectively). According to our data, this allotment presented a meaningful picture of NPs behaviour in different scenarios. In the kidneys and brain of Cr2O3 NPs-exposed animals, reactive oxygen species (ROS) production caused a significant increase in malondialdehyde (MDA) concentration along with a significant decrease in superoxide dismutase and glutathione levels, as compared to controls. Histopathological changes in these organs confirmed cellular injury and functional damage due to exposure to Cr2O3 NPs. In this study, we have distinguished pathological alterations consequent to deleterious oxidative stress due to enhanced ROS generation after Cr2O3 NPs exposure.
Collapse
Affiliation(s)
- Ravish Fatima
- 1 Section of Genetics, Department of Zoology, Aligarh Muslim University, Aligarh, Uttar Pradesh, India
| | - Kafil Akhtar
- 2 Department of Pathology, Jawaharlal Nehru Medical College, Aligarh Muslim University, Aligarh, Uttar Pradesh, India
| | - M Mobarak Hossain
- 3 Department of Physiology, Jawaharlal Nehru Medical College, Aligarh Muslim University, Aligarh, Uttar Pradesh, India
| | - Riaz Ahmad
- 1 Section of Genetics, Department of Zoology, Aligarh Muslim University, Aligarh, Uttar Pradesh, India
| |
Collapse
|
20
|
Beg MS, Huang X, Silvers MA, Gerber DE, Bolluyt J, Sarode V, Fattah F, Deberardinis RJ, Merritt ME, Xie XJ, Leff R, Laheru D, Boothman DA. Using a novel NQO1 bioactivatable drug, beta-lapachone (ARQ761), to enhance chemotherapeutic effects by metabolic modulation in pancreatic cancer. J Surg Oncol 2017; 116:83-88. [PMID: 28346693 PMCID: PMC5509448 DOI: 10.1002/jso.24624] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2017] [Accepted: 03/05/2017] [Indexed: 12/26/2022]
Abstract
Novel, tumor-selective therapies are needed to increase the survival rate of pancreatic cancer patients. K-Ras-mutant-driven NAD(P)H:quinone oxidoreductase 1 (NQO1) is over-expressed in pancreatic tumor versus associated normal tissue, while catalase expression is lowered compared to levels in associated normal pancreas tissue. ARQ761 undergoes a robust, futile redox cycle in NQO1+ cancer cells, producing massive hydrogen peroxide (H2 O2 ) levels; normal tissues are spared by low NQO1 and high catalase expression. DNA damage created by ARQ761 in pancreatic cancer cells "hyperactivates" PARP1, causing metabolic catastrophe and NAD ± keresis cell death. NQO1: catalase levels (high in tumor, low in normal tissue) are an attractive therapeutic window to treat pancreatic cancer. Based on a growing body of literature, we are leading a clinical trial to evaluate the combination of ARQ761 and chemotherapy in patients with pancreatic cancer.
Collapse
Affiliation(s)
- Muhammad Shaalan Beg
- Division of Hematology and Oncology, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX 75390
- Harold Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX 75390
| | - Xiumei Huang
- Harold Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX 75390
- Departments of Pharmacology and Radiation Oncology, UT Southwestern Medical Center, Dallas, TX 75390
| | - Molly A. Silvers
- Harold Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX 75390
| | - David E. Gerber
- Division of Hematology and Oncology, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX 75390
- Harold Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX 75390
| | - Joyce Bolluyt
- Harold Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX 75390
| | - Venetia Sarode
- Harold Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX 75390
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX 75390
| | - Farjana Fattah
- Harold Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX 75390
| | - Ralph J. Deberardinis
- Departments of Pharmacology and Radiation Oncology, UT Southwestern Medical Center, Dallas, TX 75390
- Children’s Medical Center Research Institute, UT Southwestern Medical Center, Dallas, TX 75390
| | - Matthew E. Merritt
- Department of Biochemistry and Molecular Biology, University of Florida. University of Florida, Gainesville, FL
| | - Xian-Jin Xie
- Harold Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX 75390
| | - Richard Leff
- Clinical Pharmacology & Experimental Therapeutics Center, Texas Tech University Health Sciences Center, School of Pharmacy Dallas, Texas 75235
| | - Daniel Laheru
- Sidney Kimmel Cancer Center at The Johns Hopkins University, Baltimore, MD
| | - David A. Boothman
- Harold Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX 75390
- Departments of Pharmacology and Radiation Oncology, UT Southwestern Medical Center, Dallas, TX 75390
| |
Collapse
|
21
|
Yang Y, Zhou X, Xu M, Piao J, Zhang Y, Lin Z, Chen L. β-lapachone suppresses tumour progression by inhibiting epithelial-to-mesenchymal transition in NQO1-positive breast cancers. Sci Rep 2017; 7:2681. [PMID: 28578385 PMCID: PMC5457413 DOI: 10.1038/s41598-017-02937-0] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 04/20/2017] [Indexed: 01/28/2023] Open
Abstract
NQO1 is a FAD-binding protein that can form homodimers and reduce quinones to hydroquinones, and a growing body of evidence currently suggests that NQO1 is dramatically elevated in solid cancers. Here, we demonstrated that NQO1 was elevated in breast cancer and that its expression level was positively correlated with invasion and reduced disease free survival (DFS) and overall survival (OS) rates. Next, we found that β-lapachone exerted significant anti-proliferation and anti-metastasis effects in breast cancer cell lines due to its effects on NQO1 expression. Moreover, we revealed that the anti-cancer effects of β-lapachone were mediated by the inactivation of the Akt/mTOR pathway. In conclusion, these results demonstrated that NQO1 could be a useful prognostic biomarker for patients with breast cancer, and its bioactivatable drug, β-lapachone represented a promising new development and an effective strategy for indicating the progression of NQO1-positive breast cancers.
Collapse
Affiliation(s)
- Yang Yang
- Department of Pathology & Cancer Research Center, Yanbian University Medical College, Yanji, 133002, China
| | - Xianchun Zhou
- Department of Internal Medicine, Yanbian University Hospital, Yanji, 133000, China
| | - Ming Xu
- Department of Pathology & Cancer Research Center, Yanbian University Medical College, Yanji, 133002, China
| | - Junjie Piao
- Department of Pathology & Cancer Research Center, Yanbian University Medical College, Yanji, 133002, China.,Department of Internal Medicine, Yanbian University Hospital, Yanji, 133000, China
| | - Yuan Zhang
- Department of Pathology & Cancer Research Center, Yanbian University Medical College, Yanji, 133002, China
| | - Zhenhua Lin
- Department of Pathology & Cancer Research Center, Yanbian University Medical College, Yanji, 133002, China.
| | - Liyan Chen
- Department of Pathology & Cancer Research Center, Yanbian University Medical College, Yanji, 133002, China.
| |
Collapse
|
22
|
Kwon N, Cho MK, Park SJ, Kim D, Nam SJ, Cui L, Kim HM, Yoon J. An efficient two-photon fluorescent probe for human NAD(P)H:quinone oxidoreductase (hNQO1) detection and imaging in tumor cells. Chem Commun (Camb) 2017; 53:525-528. [DOI: 10.1039/c6cc08971b] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The probe TPQ, which displays high selectivity and anti-interference ability, was successfully applied to endogenous hNQO1 imaging and for the identification of different cancer cells.
Collapse
Affiliation(s)
- Nahyun Kwon
- Department of Chemistry and Nano Science
- Ewha Womans University
- Seoul 120-750
- Korea
| | - Myoung Ki Cho
- Department of Energy Systems Research
- Ajou University
- Suwon 443-749
- Korea
| | - Sang Jun Park
- Department of Energy Systems Research
- Ajou University
- Suwon 443-749
- Korea
| | - Dayoung Kim
- Department of Chemistry and Nano Science
- Ewha Womans University
- Seoul 120-750
- Korea
| | - Sang-Jip Nam
- Department of Chemistry and Nano Science
- Ewha Womans University
- Seoul 120-750
- Korea
| | - Lei Cui
- Department of Chemistry and Nano Science
- Ewha Womans University
- Seoul 120-750
- Korea
- College of Science
| | - Hwan Myung Kim
- Department of Energy Systems Research
- Ajou University
- Suwon 443-749
- Korea
| | - Juyoung Yoon
- Department of Chemistry and Nano Science
- Ewha Womans University
- Seoul 120-750
- Korea
| |
Collapse
|
23
|
Lundberg AP, Francis JM, Pajak M, Parkinson EI, Wycislo KL, Rosol TJ, Brown ME, London CA, Dirikolu L, Hergenrother PJ, Fan TM. Pharmacokinetics and derivation of an anticancer dosing regimen for the novel anti-cancer agent isobutyl-deoxynyboquinone (IB-DNQ), a NQO1 bioactivatable molecule, in the domestic felid species. Invest New Drugs 2016; 35:134-144. [PMID: 27975234 DOI: 10.1007/s10637-016-0414-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Accepted: 11/25/2016] [Indexed: 12/17/2022]
Abstract
Isobutyl-deoxynyboquinone (IB-DNQ) is a selective substrate for NAD(P)H:quinone oxidoreductase (NQO1), an enzyme overexpressed in many solid tumors. Following activation by NQO1, IB-DNQ participates in a catalytic futile reduction/reoxidation cycle with consequent toxic reactive oxygen species generation within the tumor microenvironment. To elucidate the potential of IB-DNQ to serve as a novel anticancer agent, in vitro studies coupled with in vivo pharmacokinetic and toxicologic investigations in the domestic felid species were conducted to investigate the tractability of IB-DNQ as a translationally applicable anticancer agent. First, using feline oral squamous cell carcinoma (OSCC) as a comparative cancer model, expressions of NQO1 were characterized in not only human, but also feline OSCC tissue microarrays. Second, IB-DNQ mediated cytotoxicity in three immortalized feline OSCC cell lines were studied under dose-dependent and sequential exposure conditions. Third, the feasibility of administering IB-DNQ at doses predicted to achieve cytotoxic plasma concentrations and biologically relevant durations of exposure were investigated through pharmacokinetic and tolerability studies in healthy research felines. Intravenous administration of IB-DNQ at 1.0-2.0 mg/kg achieved peak plasma concentrations and durations of exposure reaching or exceeding predicted in vitro cytotoxic concentrations. Clinical adverse side effects including ptyalism and tachypnea exhibited during and post-IV infusion of IB-DNQ were transient and tolerable. Additionally, IB-DNQ administration did not produce acute or delayed-onset unacceptable hematologic, non-hematologic, or off-target oxidative toxicities. Collectively, the findings reported here within provide important safety and pharmacokinetic data to support the continued development of IB-DNQ as a novel anticancer strategy for NQO1 expressing cancers.
Collapse
Affiliation(s)
- Alycen P Lundberg
- Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, 61802, USA.,Carle R. Woese Institute for Genomic Biology, Anticancer Discovery from Pets to People, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Joshua M Francis
- Carle R. Woese Institute for Genomic Biology, Anticancer Discovery from Pets to People, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA.,Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Malgorzata Pajak
- Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, 61802, USA
| | - Elizabeth I Parkinson
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Kathryn L Wycislo
- Department of Pathobiology, University of Illinois at Urbana-Champaign, Urbana, IL, 61802, USA
| | - Thomas J Rosol
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, 43210, USA
| | - Megan E Brown
- Department of Veterinary Clinical Sciences, The Ohio State University, Columbus, OH, 43210, USA
| | - Cheryl A London
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, 43210, USA
| | - Levent Dirikolu
- Department of Comparative Biomedical Sciences, Louisiana State University, Baton Rouge, LA, 70803, USA
| | - Paul J Hergenrother
- Carle R. Woese Institute for Genomic Biology, Anticancer Discovery from Pets to People, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA.,Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Timothy M Fan
- Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, 61802, USA. .,Carle R. Woese Institute for Genomic Biology, Anticancer Discovery from Pets to People, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA.
| |
Collapse
|
24
|
Ryan A. Azoreductases in drug metabolism. Br J Pharmacol 2016; 174:2161-2173. [PMID: 27487252 DOI: 10.1111/bph.13571] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 07/27/2016] [Accepted: 07/29/2016] [Indexed: 02/06/2023] Open
Abstract
Azoreductases are flavoenzymes that have been characterized in a range of prokaryotes and eukaryotes. Bacterial azoreductases are associated with the activation of two classes of drug, azo drugs for the treatment of inflammatory bowel disease and nitrofuran antibiotics. The mechanism of reduction of azo compounds is presented; it requires tautomerisation of the azo compound to a quinoneimine and provides a unifying mechanism for the reduction of azo and quinone substrates by azoreductases. The importance of further work in the characterization of azoreductases from enteric bacteria is highlighted to aid in the development of novel drugs for the treatment of colon related disorders. Human azoreductases are known to play a crucial role in the metabolism of a number of quinone-containing cancer chemotherapeutic drugs. The mechanism of hydride transfer to quinones, which is shared not only between eukaryotic and prokaryotic azoreductases but also the wider family of NAD(P)H quinone oxidoreductases, is outlined. The importance of common single nucleotide polymorphisms (SNPs) in human azoreductases is described not only in cancer prognosis but also with regard to their effects on the efficacy of quinone drug-based cancer chemotherapeutic regimens. This highlights the need to screen patients for azoreductase SNPs ahead of treatment with these regimens. LINKED ARTICLES This article is part of a themed section on Drug Metabolism and Antibiotic Resistance in Micro-organisms. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.14/issuetoc.
Collapse
Affiliation(s)
- Ali Ryan
- Faculty of Science, Engineering and Computing, Kingston University, Kingston upon Thames, UK
| |
Collapse
|
25
|
Shin WS, Han J, Verwilst P, Kumar R, Kim JH, Kim JS. Cancer Targeted Enzymatic Theranostic Prodrug: Precise Diagnosis and Chemotherapy. Bioconjug Chem 2016; 27:1419-26. [DOI: 10.1021/acs.bioconjchem.6b00184] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Weon Sup Shin
- Department
of Chemistry, Korea University, Seoul 136-701, Korea
| | - Jiyou Han
- Department of Biotechnology, Laboratory of Stem Cells and Tissue Regeneration, College of Life Sciences & Biotechnology, Korea University, Seoul 136-713, Republic of Korea
| | - Peter Verwilst
- Department
of Chemistry, Korea University, Seoul 136-701, Korea
| | - Rajesh Kumar
- Department
of Chemistry, Korea University, Seoul 136-701, Korea
| | - Jong-Hoon Kim
- Department of Biotechnology, Laboratory of Stem Cells and Tissue Regeneration, College of Life Sciences & Biotechnology, Korea University, Seoul 136-713, Republic of Korea
| | - Jong Seung Kim
- Department
of Chemistry, Korea University, Seoul 136-701, Korea
| |
Collapse
|
26
|
Aras D, Cinar O, Cakar Z, Ozkavukcu S, Can A. Can dicoumarol be used as a gonad-safe anticancer agent: an in vitro and in vivo experimental study. Mol Hum Reprod 2015; 22:57-67. [PMID: 26612783 DOI: 10.1093/molehr/gav065] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Accepted: 11/20/2015] [Indexed: 12/16/2022] Open
Abstract
STUDY HYPOTHESIS Dicoumarol (DC) has potential for use as a gonad-safe anticancer agent. STUDY FINDING DC altered cell proliferation, decreased viability and increased apoptosis in Vero and MCF-7 cell lines but did not show any toxic effect on mouse ovarian tissues and developing oocytes in vitro and in vivo. WHAT IS KNOWN ALREADY DC suppresses cell proliferation and increases apoptosis in various cancer cells such as breast, urogenital and melanoma. DC has also been reported to alter the anticancer effects of several chemotherapeutics, including cisplatin, gemcitabine and doxorubicin in prostate, liver and uroepithelial cancer cells, respectively. STUDY DESIGN, SAMPLES/MATERIALS, METHODS Vero (African green monkey kidney epithelial cells) and MCF-7 (human cancerous breast epithelial cells) cell lines and mouse granulosa cells isolated from 21-day-old female BALB/c mice (n = 21) were used to assess the effects of DC (10, 50, 100 and 200 µm) for 24 and 48 h on cell proliferation, viability and apoptotic cell death. In vivo experiments were performed with a single i.p. injection of 32 mg/kg DC in 21-day-old female BALB/c mice (n = 12). Following 48 h, animals were sacrificed by cervical dislocation and histological sections of isolated ovaries were evaluated for apoptosis. Viability assays were based on the trypan blue dye exclusion method and an automated cell counter device was used. Terminal deoxynucleotidyltransferase-mediated dUTP nick-end labelling (TUNEL) and Annexin-V immunofluorescence were assessed by 3D confocal microscopy to address apoptotic cell death. We also assessed whether DC inhibits cell proliferation and viability through NQO1 [NAD(P)H Quinone Oxidoreductase 1], an intracellular inhibitor of reactive oxygen species (ROS). The meiotic spindle and chromosomes were studied in mouse oocytes by α-β-tubulin and 7-aminoactinomycine D (7-AAD) immunostaining in vitro and in vivo. MAIN RESULTS AND THE ROLE OF CHANCE DC does not block oocyte maturation and no significant alteration was noted in meiotic spindle or chromosome morphology in metaphase-II (M-II) stage oocytes following DC treatment in vitro or in vivo. In contrast, exposure to DC for 24 h suppressed cell proliferation (P = 0.026 at 200 µm), decreased viability (P = 0.002 at 200 µm) and increased apoptosis (P = 0.048 at 100 µm) in Vero and MCF-7 cell lines, compared with controls. These changes were not related to intracellular NQO1 levels. Mouse granulosa cells were unaffected by 50 or 100 µm DC treatment for 24 and 48 h in vitro. DC treatment in vivo did not alter the number of primordial follicles or the ratio of apoptosis in primordial, primary and secondary follicles, as well as in antral follicles, compared with the controls. LIMITATIONS, REASONS FOR CAUTION DC was tested for ovarian toxicity only in isolated mouse oocytes/ovaries and healthy BALB/c mice. No cancer formation was used as an in vivo test model. The possibility that DC may potentiate ovarian toxicity when combined with traditional chemotherapeutic agents, such as mitomycin-C, cisplatin, gemcitabine and doxorubicin, must be taken into account, as DC is known to alter their effects in some cancer cells. WIDER IMPLICATIONS OF THE FINDINGS The present study evaluated, for the first time, the effect of DC on ovarian tissue. The results suggested that DC is not toxic to ovarian tissues and developing oocytes; therefore, DC should be assessed further as a potential anticancer agent when female fertility preservation is a concern. LARGE SCALE DATA N/A. STUDY FUNDING AND COMPETING INTERESTS This work includes data from dissertation thesis entitled 'Effects of dicoumarol on mitotic and meiotic cells as an anticancer agent' by DA, 2014 and was partly supported by The National Scientific and Technological Research Council of Turkey (SBAG-109S415) to AC, OC and SO. The authors confirm that this article content presents no conflicts of interest.
Collapse
Affiliation(s)
- Duru Aras
- Ankara University Biotechnology Institute, Tandogan, Ankara 06500, Turkey
| | - Ozgur Cinar
- Department of Histology and Embryology, Laboratory for Stem Cells and Reproductive Biology, Ankara University School of Medicine, Sihhiye, Ankara 06100, Turkey
| | - Zeynep Cakar
- Department of Histology and Embryology, Laboratory for Stem Cells and Reproductive Biology, Ankara University School of Medicine, Sihhiye, Ankara 06100, Turkey
| | - Sinan Ozkavukcu
- Department of Obstetric and Gynaecology, Centre for Assisted Reproduction, Ankara University School of Medicine, Cebeci, Ankara 06590, Turkey
| | - Alp Can
- Department of Histology and Embryology, Laboratory for Stem Cells and Reproductive Biology, Ankara University School of Medicine, Sihhiye, Ankara 06100, Turkey
| |
Collapse
|
27
|
Abstract
One of the major goals of cancer therapy is the selective targeting of cancer cells over normal cells. Unfortunately, even with recent advances, the majority of chemotherapeutics still indiscriminately kill all rapidly dividing cells. Although these drugs are effective in certain settings, their inability to specifically target cancer results in significant dose-limiting toxicities. One way to avoid such toxicities is to target an aspect of the cancer cell that is not shared by normal cells. A potential cancer-specific target is the enzyme NAD(P)H quinone oxidoreductase 1 (NQO1). NQO1 is a 2-electron reductase responsible for the detoxification of quinones. Its expression is typically quite low in normal tissue, but it has been found to be greatly overexpressed in many types of solid tumors, including lung, breast, pancreatic, and colon cancers. This overexpression is thought to be in response to the higher oxidative stress of the cancer cell, and it is possible that NQO1 contributes to tumor progression. The overexpression of NQO1 and its correlation with poor patient outcome make it an intriguing target. Although some have explored inhibiting NQO1 as an anticancer strategy, this has generally been unsuccessful. A more promising strategy is to utilize NQO1 substrates that are activated upon reduction by NQO1. For example, in principle, reduction of a quinone can result in a hydroquinone that is a DNA alkylator, protein inhibitor, or reduction-oxidation cycler. Although there are many proposed NQO1 substrates, head-to-head assays reveal only two classes of compounds that convincingly induce cancer cell death through NQO1-mediated activation. In this Account, we describe the discovery and development of one of these compounds, the natural product deoxynyboquinone (DNQ), an excellent NQO1 substrate and anticancer agent. A modular synthesis of DNQ was developed that enabled access to the large compound quantities needed to conduct extensive mechanistic evaluations and animal experiments. During these evaluations, we found that DNQ is an outstanding NQO1 substrate that is processed much more efficiently than other putative NQO1 substrates. Importantly, its anticancer activity is strictly dependent on the overexpression of active NQO1. Using previous crystal structures of NQO1, novel DNQ derivatives were designed that are also excellent NQO1 substrates and possess properties that make them more attractive than the parent natural product for translational development. Given their selectivity, potency, outstanding pharmacokinetic properties, and the ready availability of diagnostics to assess NQO1 in patients, DNQ and its derivatives have considerable potential as personalized medicines for the treatment of cancer.
Collapse
Affiliation(s)
- Elizabeth I. Parkinson
- Department of Chemistry,
Roger Adams Laboratory, University of Illinois at Urbana−Champaign, Urbana, Illinois 61801, United States
| | - Paul J. Hergenrother
- Department of Chemistry,
Roger Adams Laboratory, University of Illinois at Urbana−Champaign, Urbana, Illinois 61801, United States
| |
Collapse
|
28
|
Chakrabarti G, Moore ZR, Luo X, Ilcheva M, Ali A, Padanad M, Zhou Y, Xie Y, Burma S, Scaglioni PP, Cantley LC, DeBerardinis RJ, Kimmelman AC, Lyssiotis CA, Boothman DA. Targeting glutamine metabolism sensitizes pancreatic cancer to PARP-driven metabolic catastrophe induced by ß-lapachone. Cancer Metab 2015; 3:12. [PMID: 26462257 PMCID: PMC4601138 DOI: 10.1186/s40170-015-0137-1] [Citation(s) in RCA: 93] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2015] [Accepted: 09/17/2015] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinomas (PDA) activate a glutamine-dependent pathway of cytosolic nicotinamide adenine dinucleotide phosphate (NADPH) production to maintain redox homeostasis and support proliferation. Enzymes involved in this pathway (GLS1 (mitochondrial glutaminase 1), GOT1 (cytoplasmic glutamate oxaloacetate transaminase 1), and GOT2 (mitochondrial glutamate oxaloacetate transaminase 2)) are highly upregulated in PDA, and among these, inhibitors of GLS1 were recently deployed in clinical trials to target anabolic glutamine metabolism. However, single-agent inhibition of this pathway is cytostatic and unlikely to provide durable benefit in controlling advanced disease. RESULTS Here, we report that reducing NADPH pools by genetically or pharmacologically (bis-2-(5-phenylacetamido-1,2,4-thiadiazol-2-yl)ethyl sulfide (BPTES) or CB-839) inhibiting glutamine metabolism in mutant Kirsten rat sarcoma viral oncogene homolog (KRAS) PDA sensitizes cell lines and tumors to ß-lapachone (ß-lap, clinical form ARQ761). ß-Lap is an NADPH:quinone oxidoreductase (NQO1)-bioactivatable drug that leads to NADPH depletion through high levels of reactive oxygen species (ROS) from the futile redox cycling of the drug and subsequently nicotinamide adenine dinucleotide (NAD)+ depletion through poly(ADP ribose) polymerase (PARP) hyperactivation. NQO1 expression is highly activated by mutant KRAS signaling. As such, ß-lap treatment concurrent with inhibition of glutamine metabolism in mutant KRAS, NQO1 overexpressing PDA leads to massive redox imbalance, extensive DNA damage, rapid PARP-mediated NAD+ consumption, and PDA cell death-features not observed in NQO1-low, wild-type KRAS expressing cells. CONCLUSIONS This treatment strategy illustrates proof of principle that simultaneously decreasing glutamine metabolism-dependent tumor anti-oxidant defenses and inducing supra-physiological ROS formation are tumoricidal and that this rationally designed combination strategy lowers the required doses of both agents in vitro and in vivo. The non-overlapping specificities of GLS1 inhibitors and ß-lap for PDA tumors afford high tumor selectivity, while sparing normal tissue.
Collapse
Affiliation(s)
- Gaurab Chakrabarti
- Department of Pharmacology, University of Texas Southwestern Medical Center, 6001 Forest Park Drive, Dallas, 75390-8807 TX USA ; Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX USA
| | - Zachary R Moore
- Department of Pharmacology, University of Texas Southwestern Medical Center, 6001 Forest Park Drive, Dallas, 75390-8807 TX USA ; Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX USA
| | - Xiuquan Luo
- Department of Pharmacology, University of Texas Southwestern Medical Center, 6001 Forest Park Drive, Dallas, 75390-8807 TX USA ; Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX USA
| | - Mariya Ilcheva
- Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX USA
| | - Aktar Ali
- Touchstone Diabetes Center, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX USA
| | - Mahesh Padanad
- Department of Internal Medicine, Weill Cornell Medical College, 413 East 69th Street, BB-1362, New York, NY 10021 USA
| | - Yunyun Zhou
- Department of Bioinformatics and Biostatistics, Clinical Sciences, UT Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390 USA
| | - Yang Xie
- Department of Bioinformatics and Biostatistics, Clinical Sciences, UT Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390 USA
| | - Sandeep Burma
- Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX USA
| | - Pier P Scaglioni
- Department of Internal Medicine, Weill Cornell Medical College, 413 East 69th Street, BB-1362, New York, NY 10021 USA
| | - Lewis C Cantley
- Department of Medicine, Weill Cornell Medical College, 413 East 69th Street, BB-1362, New York, NY 10021 USA
| | - Ralph J DeBerardinis
- Children's Medical Center Research Institute, UT Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390 USA
| | - Alec C Kimmelman
- Department of Radiation Oncology, Division of Genomic Stability and DNA Repair, Dana-Farber Cancer Institute, Boston, MA 02215 USA
| | - Costas A Lyssiotis
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109 USA ; Internal Medicine, Division of Gastroenterology, University of Michigan, Ann Arbor, MI 48109 USA
| | - David A Boothman
- Department of Pharmacology, University of Texas Southwestern Medical Center, 6001 Forest Park Drive, Dallas, 75390-8807 TX USA ; Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX USA
| |
Collapse
|
29
|
Ma X, Moore ZR, Huang G, Huang X, Boothman DA, Gao J. Nanotechnology-enabled delivery of NQO1 bioactivatable drugs. J Drug Target 2015; 23:672-80. [DOI: 10.3109/1061186x.2015.1073296] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
|
30
|
Zhang L, Chen Z, Yang K, Liu C, Gao J, Qian F. β-Lapachone and Paclitaxel Combination Micelles with Improved Drug Encapsulation and Therapeutic Synergy as Novel Nanotherapeutics for NQO1-Targeted Cancer Therapy. Mol Pharm 2015; 12:3999-4010. [PMID: 26415823 DOI: 10.1021/acs.molpharmaceut.5b00448] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
β-Lapachone (LPC) is a novel cytotoxic agent that is bioactivated by NADP(H): quinone oxidoreductase 1 (NQO1), an enzyme elevated in a variety of tumors, such as non-small cell lung cancer (NSCLC), pancreatic cancer, liver cancer, and breast cancer. Despite its unique mechanism of action, its clinical evaluation has been largely hindered by low water solubility, short blood half-life, and narrow therapeutic window. Although encapsulation into poly(ethylene glycol)-b-poly(D,L-lactic acid) (PEG-PLA) micelles could modestly improve its solubility and prolong its half-life, the extremely fast intrinsic crystallization tendency of LPC prevents drug loading higher than ∼2 wt %. The physical stability of the LPC-loaded micelles is also far from satisfactory for further development. In this study, we demonstrate that paclitaxel (PTX), a front-line drug for many cancers, can provide two functions when coencapsulated together with LPC in the PEG-PLA micelles; first, as a strong crystallization inhibitor for LPC, thus to significantly increase the LPC encapsulation efficiency in the micelle from 11.7 ± 2.4% to 100.7 ± 2.2%. The total drug loading efficiency of both PTX and LPC in the combination polymeric micelle reached 100.3 ± 3.0%, and the drug loading density reached 33.2 ± 1.0%. Second, the combination of LPC/PTX demonstrates strong synergistic cytotoxicity effect against the NQO1 overexpressing cancer cells, including A549 NSCLC cells, and several pancreatic cancer cells (combination index <1). In vitro drug release study showed that LPC was released faster than PTX either in phosphate-buffered saline (PH = 7.4) or in 1 M sodium salicylate, which agrees with the desired dosing sequence of the two drugs to exert synergistic pharmacologic effect at different cell checkpoints. The PEG-PLA micelles coloaded with LPC and PTX offer a novel nanotherapeutic, with high drug loading, sufficient physical stability, and biological synergy to increase drug delivery efficiency and optimize the therapeutic window for NOQ1-targeted therapy of cancer.
Collapse
Affiliation(s)
- Ling Zhang
- Department of Pharmacology and Pharmaceutical Sciences and Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Medicine, Tsinghua University , Beijing 100084, P.R. China
| | - Zhen Chen
- Department of Pharmacology and Pharmaceutical Sciences and Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Medicine, Tsinghua University , Beijing 100084, P.R. China
| | - Kuan Yang
- Department of Pharmacology and Pharmaceutical Sciences and Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Medicine, Tsinghua University , Beijing 100084, P.R. China
| | - Chun Liu
- Department of Pharmacology and Pharmaceutical Sciences and Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Medicine, Tsinghua University , Beijing 100084, P.R. China
| | - Jinming Gao
- Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center at Dallas , Dallas, Texas 75390, United States
| | - Feng Qian
- Department of Pharmacology and Pharmaceutical Sciences and Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Medicine, Tsinghua University , Beijing 100084, P.R. China
| |
Collapse
|
31
|
Hayes AJ, Skouras C, Haugk B, Charnley RM. Keap1-Nrf2 signalling in pancreatic cancer. Int J Biochem Cell Biol 2015; 65:288-99. [PMID: 26117456 DOI: 10.1016/j.biocel.2015.06.017] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Revised: 06/21/2015] [Accepted: 06/22/2015] [Indexed: 12/16/2022]
Abstract
Transcription factor NF-E2 p45-related factor 2 (Nrf2, also called Nfe2l2), a master regulator of redox homeostasis, and its dominant negative regulator, Kelch-like ECH-associated protein 1 (Keap1), together tightly control the expression of numerous detoxifying and antioxidant genes. Nrf2 and the 'antioxidant response element' (ARE)-driven genes it controls are frequently upregulated in pancreatic cancer and correlate with poor survival. Upregulation of Nrf2 is, at least in part, K-Ras oncogene-driven and contributes to pancreatic cancer proliferation and chemoresistance. In this review, we aim to provide an overview of Keap1-Nrf2 signalling as it relates to pancreatic cancer, discussing the effects of inhibiting Nrf2 or Nrf2/ARE effector proteins to increase chemosensitivity.
Collapse
Affiliation(s)
- Alastair J Hayes
- MRC Centre for Inflammation Research, University of Edinburgh, Queen's Medical Research Centre, Room C2.18, 47 Little France Crescent, Edinburgh, Scotland EH16 4TJ, United Kingdom.
| | - Christos Skouras
- School of Clinical Surgery, College of Medicine and Veterinary Medicine, University of Edinburgh, Room SU 305, Chancellor's Building, 49 Little France Crescent, Edinburgh, Scotland EH16 4SB, United Kingdom.
| | - Beate Haugk
- Department of Cellular Pathology, Newcastle upon Tyne Hospitals NHS Foundation Trust, Royal Victoria Infirmary, Queen Victoria Road, Newcastle upon Tyne, England NE1 4LP, United Kingdom.
| | - Richard M Charnley
- Department of Hepato-Pancreatico-Biliary Surgery, Newcastle upon Tyne Hospitals NHS Foundation Trust, Freeman Hospital, High Heaton, Newcastle upon Tyne, England NE7 7DN, United Kingdom.
| |
Collapse
|
32
|
Moore Z, Chakrabarti G, Luo X, Ali A, Hu Z, Fattah FJ, Vemireddy R, DeBerardinis RJ, Brekken RA, Boothman DA. NAMPT inhibition sensitizes pancreatic adenocarcinoma cells to tumor-selective, PAR-independent metabolic catastrophe and cell death induced by β-lapachone. Cell Death Dis 2015; 6:e1599. [PMID: 25590809 PMCID: PMC4669762 DOI: 10.1038/cddis.2014.564] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Revised: 11/26/2014] [Accepted: 11/28/2014] [Indexed: 01/01/2023]
Abstract
Nicotinamide phosphoribosyltransferase (NAMPT) inhibitors (e.g., FK866) target the most active pathway of NAD(+) synthesis in tumor cells, but lack tumor-selectivity for use as a single agent. Reducing NAD(+) pools by inhibiting NAMPT primed pancreatic ductal adenocarcinoma (PDA) cells for poly(ADP ribose) polymerase (PARP1)-dependent cell death induced by the targeted cancer therapeutic, β-lapachone (β-lap, ARQ761), independent of poly(ADP ribose) (PAR) accumulation. β-Lap is bioactivated by NADPH:quinone oxidoreductase 1 (NQO1) in a futile redox cycle that consumes oxygen and generates high levels of reactive oxygen species (ROS) that cause extensive DNA damage and rapid PARP1-mediated NAD(+) consumption. Synergy with FK866+β-lap was tumor-selective, only occurring in NQO1-overexpressing cancer cells, which is noted in a majority (∼85%) of PDA cases. This treatment strategy simultaneously decreases NAD(+) synthesis while increasing NAD(+) consumption, reducing required doses and treatment times for both drugs and increasing potency. These complementary mechanisms caused profound NAD(P)(+) depletion and inhibited glycolysis, driving down adenosine triphosphate levels and preventing recovery normally observed with either agent alone. Cancer cells died through an ROS-induced, μ-calpain-mediated programmed cell death process that kills independent of caspase activation and is not driven by PAR accumulation, which we call NAD(+)-Keresis. Non-overlapping specificities of FK866 for PDA tumors that rely heavily on NAMPT-catalyzed NAD(+) synthesis and β-lap for cancer cells with elevated NQO1 levels affords high tumor-selectivity. The concept of reducing NAD(+) pools in cancer cells to sensitize them to ROS-mediated cell death by β-lap is a novel strategy with potential application for pancreatic and other types of NQO1+ solid tumors.
Collapse
Affiliation(s)
- Z Moore
- Pharmacology and Radiation Oncology, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - G Chakrabarti
- Pharmacology and Radiation Oncology, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - X Luo
- Pharmacology and Radiation Oncology, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - A Ali
- Internal Medicine and Touchstone Diabetes Center, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Z Hu
- Children's Medical Center Research Institute, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - F J Fattah
- Pharmacology and Radiation Oncology, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - R Vemireddy
- Pharmacology and Radiation Oncology, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - R J DeBerardinis
- Children's Medical Center Research Institute, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - R A Brekken
- Pharmacology and Radiation Oncology, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Surgical Oncology, Department of Surgery and Hamon Center for Therapeutic Oncology Research, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - D A Boothman
- Pharmacology and Radiation Oncology, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
33
|
Cao L, Li LS, Spruell C, Xiao L, Chakrabarti G, Bey EA, Reinicke KE, Srougi MC, Moore Z, Dong Y, Vo P, Kabbani W, Yang CR, Wang X, Fattah F, Morales JC, Motea EA, Bornmann WG, Yordy JS, Boothman DA. Tumor-selective, futile redox cycle-induced bystander effects elicited by NQO1 bioactivatable radiosensitizing drugs in triple-negative breast cancers. Antioxid Redox Signal 2014; 21:237-50. [PMID: 24512128 PMCID: PMC4060774 DOI: 10.1089/ars.2013.5462] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
AIMS β-Lapachone (β-lap), a novel radiosensitizer with potent antitumor efficacy alone, selectively kills solid cancers that over-express NAD(P)H quinone oxidoreductase 1 (NQO1). Since breast or other solid cancers have heterogeneous NQO1 expression, therapies that reduce the resistance (e.g., NQO1(low)) of tumor cells will have significant clinical advantages. We tested whether NQO1-proficient (NQO1(+)) cells generated sufficient hydrogen peroxide (H2O2) after β-lap treatment to elicit bystander effects, DNA damage, and cell death in neighboring NQO1(low) cells. RESULTS β-Lap showed NQO1-dependent efficacy against two triple-negative breast cancer (TNBC) xenografts. NQO1 expression variations in human breast cancer patient samples were noted, where ~60% cancers over-expressed NQO1, with little or no expression in associated normal tissue. Differential DNA damage and lethality were noted in NQO1(+) versus NQO1-deficient (NQO1(-)) TNBC cells and xenografts after β-lap treatment. β-Lap-treated NQO1(+) cells died by programmed necrosis, whereas co-cultured NQO1(-) TNBC cells exhibited DNA damage and caspase-dependent apoptosis. NQO1 inhibition (dicoumarol) or H2O2 scavenging (catalase [CAT]) blocked all responses. Only NQO1(-) cells neighboring NQO1(+) TNBC cells responded to β-lap in vitro, and bystander effects correlated well with H2O2 diffusion. Bystander effects in NQO1(-) cells in vivo within mixed 50:50 co-cultured xenografts were dramatic and depended on NQO1(+) cells. However, normal human cells in vitro or in vivo did not show bystander effects, due to elevated endogenous CAT levels. Innovation and Conclusions: NQO1-dependent bystander effects elicited by NQO1 bioactivatable drugs (β-lap or deoxynyboquinone [DNQ]) likely contribute to their efficacies, killing NQO1(+) solid cancer cells and eliminating surrounding heterogeneous NQO1(low) cancer cells. Normal cells/tissue are protected by low NQO1:CAT ratios.
Collapse
Affiliation(s)
- Lifen Cao
- 1 Department of General Surgery, The Second Xiangya Hospital of Central South University , Changsha, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Sztiller-Sikorska M, Koprowska K, Majchrzak K, Hartman M, Czyz M. Natural compounds' activity against cancer stem-like or fast-cycling melanoma cells. PLoS One 2014; 9:e90783. [PMID: 24595456 PMCID: PMC3940936 DOI: 10.1371/journal.pone.0090783] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Accepted: 02/04/2014] [Indexed: 12/21/2022] Open
Abstract
Background Accumulating evidence supports the concept that melanoma is highly heterogeneous and sustained by a small subpopulation of melanoma stem-like cells. Those cells are considered as responsible for tumor resistance to therapies. Moreover, melanoma cells are characterized by their high phenotypic plasticity. Consequently, both melanoma stem-like cells and their more differentiated progeny must be eradicated to achieve durable cure. By reevaluating compounds in heterogeneous melanoma populations, it might be possible to select compounds with activity not only against fast-cycling cells but also against cancer stem-like cells. Natural compounds were the focus of the present study. Methods We analyzed 120 compounds from The Natural Products Set II to identify compounds active against melanoma populations grown in an anchorage-independent manner and enriched with cells exerting self-renewing capacity. Cell viability, cell cycle arrest, apoptosis, gene expression, clonogenic survival and label-retention were analyzed. Findings Several compounds efficiently eradicated cells with clonogenic capacity and nanaomycin A, streptonigrin and toyocamycin were effective at 0.1 µM. Other anti-clonogenic but not highly cytotoxic compounds such as bryostatin 1, siomycin A, illudin M, michellamine B and pentoxifylline markedly reduced the frequency of ABCB5 (ATP-binding cassette, sub-family B, member 5)-positive cells. On the contrary, treatment with maytansine and colchicine selected for cells expressing this transporter. Maytansine, streptonigrin, toyocamycin and colchicine, even if highly cytotoxic, left a small subpopulation of slow-dividing cells unaffected. Compounds selected in the present study differentially altered the expression of melanocyte/melanoma specific microphthalmia-associated transcription factor (MITF) and proto-oncogene c-MYC. Conclusion Selected anti-clonogenic compounds might be further investigated as potential adjuvants targeting melanoma stem-like cells in the combined anti-melanoma therapy, whereas selected cytotoxic but not anti-clonogenic compounds, which increased the frequency of ABCB5-positive cells and remained slow-cycling cells unaffected, might be considered as a tool to enrich cultures with cells exhibiting melanoma stem cell characteristics.
Collapse
Affiliation(s)
| | - Kamila Koprowska
- Department of Molecular Biology of Cancer, Medical University of Lodz, Lodz, Poland
| | - Kinga Majchrzak
- Department of Molecular Biology of Cancer, Medical University of Lodz, Lodz, Poland
| | - Mariusz Hartman
- Department of Molecular Biology of Cancer, Medical University of Lodz, Lodz, Poland
| | - Malgorzata Czyz
- Department of Molecular Biology of Cancer, Medical University of Lodz, Lodz, Poland
- * E-mail:
| |
Collapse
|
35
|
Lin L, Qin Y, Jin T, Liu S, Zhang S, Shen X, Lin Z. Significance of NQO1 overexpression for prognostic evaluation of gastric adenocarcinoma. Exp Mol Pathol 2013; 96:200-5. [PMID: 24384455 DOI: 10.1016/j.yexmp.2013.12.008] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Accepted: 12/03/2013] [Indexed: 12/15/2022]
Abstract
NQO1 (NAD(P)H: quinone oxidoreductase, also known as DT-diaphorase) plays a prominent role in maintaining cellular homeostasis. NQO1 is abnormally elevated in many solid cancer types, including those of the adrenal gland, breast, colon, lung, ovary, and thyroid. However, little is known about the status of NQO1 in gastric adenocarcinoma (GAC). To investigate the clinicopathological significance of NQO1 expression in GAC, and thus evaluate its role as a potential prognostic marker, 203 cases of primary GAC, 31 of gastric dysplasia, and 53 of adjacent non-tumor tissues were selected for immunohistochemical staining of NQO1 protein. Correlations between NQO1 overexpression and clinicopathological characteristics were evaluated by χ(2) test and Fisher's exact test, while survival rates were calculated by Kaplan-Meier method. The relationship between prognostic factors and patient survival was analyzed by Cox proportional hazards model. Through these analyses it was found that the strongly positive rate of NQO1 protein in GAC was significantly higher than that in gastric dysplasia and adjacent non-tumor tissues. Analysis by qRT-PCR also confirmed that NQO1 mRNA levels were increased in GAC compared with those detected in either adjacent non-tumor tissues or normal gastric mucosa. Additionally, the NQO1 expression rate was positively correlated with tumor size, serosal invasion, tumor stage, and both disease-free survival and 5-year survival rates. Further analysis showed that although NQO1 was not an independent predictor of GAC, elevated expression of NQO1 could predict lower disease-free survival and 5-year survival times in late-stage patients. In conclusion, NQO1 plays an important role in the progression of GAC, and might be a potential, but not an independent, poor prognostic biomarker and therapeutic target of GAC.
Collapse
Affiliation(s)
- Lijuan Lin
- Department of Pathology, Yanbian University Medical College, Yanji 133002, China; Department of Medical Imaging, College of Medicine, Eastern Liaoning University, Dandong 118003, China.
| | - Yunzhi Qin
- Department of Anesthesiology, Yanbian University Hospital, Yanji 133000, China.
| | - Tiefeng Jin
- Department of Pathology, Yanbian University Medical College, Yanji 133002, China.
| | - Shuangping Liu
- Department of Pathology, Yanbian University Medical College, Yanji 133002, China.
| | - Songnan Zhang
- Department of Oncology, Yanbian University Hospital, Yanji 133000, China.
| | - Xionghu Shen
- Department of Oncology, Yanbian University Hospital, Yanji 133000, China.
| | - Zhenhua Lin
- Department of Pathology, Yanbian University Medical College, Yanji 133002, China.
| |
Collapse
|
36
|
Liu M, Wang Q, Liu F, Cheng X, Wu X, Wang H, Wu M, Ma Y, Wang G, Hao H. UDP-glucuronosyltransferase 1A compromises intracellular accumulation and anti-cancer effect of tanshinone IIA in human colon cancer cells. PLoS One 2013; 8:e79172. [PMID: 24244442 PMCID: PMC3828323 DOI: 10.1371/journal.pone.0079172] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2013] [Accepted: 09/20/2013] [Indexed: 01/11/2023] Open
Abstract
Background and Purpose NAD(P)H: quinone oxidoreductase 1 (NQO1) mediated quinone reduction and subsequent UDP-glucuronosyltransferases (UGTs) catalyzed glucuronidation is the dominant metabolic pathway of tanshinone IIA (TSA), a promising anti-cancer agent. UGTs are positively expressed in various tumor tissues and play an important role in the metabolic elimination of TSA. This study aims to explore the role of UGT1A in determining the intracellular accumulation and the resultant apoptotic effect of TSA. Experimental Approach We examined TSA intracellular accumulation and glucuronidation in HT29 (UGT1A positive) and HCT116 (UGT1A negative) human colon cancer cell lines. We also examined TSA-mediated reactive oxygen species (ROS) production, cytotoxicity and apoptotic effect in HT29 and HCT116 cells to investigate whether UGT1A levels are directly associated with TSA anti-cancer effect. UGT1A siRNA or propofol, a UGT1A9 competitive inhibitor, was used to inhibit UGT1A expression or UGT1A9 activity. Key Results Multiple UGT1A isoforms are positively expressed in HT29 but not in HCT116 cells. Cellular S9 fractions prepared from HT29 cells exhibit strong glucuronidation activity towards TSA, which can be inhibited by propofol or UGT1A siRNA interference. TSA intracellular accumulation in HT29 cells is much lower than that in HCT116 cells, which correlates with high expression levels of UGT1A in HT29 cells. Consistently, TSA induces less intracellular ROS, cytotoxicity, and apoptotic effect in HT29 cells than those in HCT116 cells. Pretreatment of HT29 cells with UGT1A siRNA or propofol can decrease TSA glucuronidation and simultaneously improve its intracellular accumulation, as well as enhance TSA anti-cancer effect. Conclusions and Implications UGT1A can compromise TSA cytotoxicity via reducing its intracellular exposure and switching the NQO1-triggered redox cycle to metabolic elimination. Our study may shed a light in understanding the cellular pharmacokinetic and molecular mechanism by which UGTs determine the chemotherapy effects of drugs that are UGTs’ substrates.
Collapse
Affiliation(s)
- Miao Liu
- Key Lab of Drug Metabolism and Pharmacokinetics, State Key Lab of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Qiong Wang
- Key Lab of Drug Metabolism and Pharmacokinetics, State Key Lab of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Fang Liu
- Key Lab of Drug Metabolism and Pharmacokinetics, State Key Lab of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Xuefang Cheng
- Key Lab of Drug Metabolism and Pharmacokinetics, State Key Lab of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Xiaolan Wu
- Key Lab of Drug Metabolism and Pharmacokinetics, State Key Lab of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Hong Wang
- Key Lab of Drug Metabolism and Pharmacokinetics, State Key Lab of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Mengqiu Wu
- Key Lab of Drug Metabolism and Pharmacokinetics, State Key Lab of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Ying Ma
- Key Lab of Drug Metabolism and Pharmacokinetics, State Key Lab of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Guangji Wang
- Key Lab of Drug Metabolism and Pharmacokinetics, State Key Lab of Natural Medicines, China Pharmaceutical University, Nanjing, China
- * E-mail: (GW); (HH)
| | - Haiping Hao
- Key Lab of Drug Metabolism and Pharmacokinetics, State Key Lab of Natural Medicines, China Pharmaceutical University, Nanjing, China
- * E-mail: (GW); (HH)
| |
Collapse
|
37
|
Parkinson EI, Bair JS, Cismesia M, Hergenrother PJ. Efficient NQO1 substrates are potent and selective anticancer agents. ACS Chem Biol 2013; 8:2173-83. [PMID: 23937670 DOI: 10.1021/cb4005832] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
A major goal of personalized medicine in oncology is the identification of drugs with predictable efficacy based on a specific trait of the cancer cell, as has been demonstrated with gleevec (presence of Bcr-Abl protein), herceptin (Her2 overexpression), and iressa (presence of a specific EGFR mutation). This is a challenging task, as it requires identifying a cellular component that is altered in cancer, but not normal cells, and discovering a compound that specifically interacts with it. The enzyme NQO1 is a potential target for personalized medicine, as it is overexpressed in many solid tumors. In normal cells NQO1 is inducibly expressed, and its major role is to detoxify quinones via bioreduction; however, certain quinones become more toxic after reduction by NQO1, and these compounds have potential as selective anticancer agents. Several quinones of this type have been reported, including mitomycin C, RH1, EO9, streptonigrin, β-lapachone, and deoxynyboquinone (DNQ). However, no unified picture has emerged from these studies, and the key question regarding the relationship between NQO1 processing and anticancer activity remains unanswered. Here, we directly compare these quinones as substrates for NQO1 in vitro, and for their ability to kill cancer cells in culture in an NQO1-dependent manner. We show that DNQ is a superior NQO1 substrate, and we use computationally guided design to create DNQ analogues that have a spectrum of activities with NQO1. Assessment of these compounds definitively establishes a strong relationship between in vitro NQO1 processing and induction of cancer cell death and suggests these compounds are outstanding candidates for selective anticancer therapy.
Collapse
Affiliation(s)
- Elizabeth I. Parkinson
- Department of Chemistry, Roger Adams Laboratory, University of Illinois at Urbana−Champaign, Urbana, Illinois 61801, United States
| | - Joseph S. Bair
- Department of Chemistry, Roger Adams Laboratory, University of Illinois at Urbana−Champaign, Urbana, Illinois 61801, United States
| | - Megan Cismesia
- Department of Chemistry, Roger Adams Laboratory, University of Illinois at Urbana−Champaign, Urbana, Illinois 61801, United States
| | - Paul J. Hergenrother
- Department of Chemistry, Roger Adams Laboratory, University of Illinois at Urbana−Champaign, Urbana, Illinois 61801, United States
| |
Collapse
|
38
|
Pathania D, Sechi M, Palomba M, Sanna V, Berrettini F, Sias A, Taheri L, Neamati N. Design and discovery of novel quinazolinedione-based redox modulators as therapies for pancreatic cancer. Biochim Biophys Acta Gen Subj 2013; 1840:332-43. [PMID: 23954204 DOI: 10.1016/j.bbagen.2013.08.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2013] [Revised: 07/23/2013] [Accepted: 08/08/2013] [Indexed: 01/09/2023]
Abstract
BACKGROUND Altered cellular bioenergetics and oxidative stress are emerging hallmarks of most cancers including pancreatic cancer. Elevated levels of intrinsic reactive oxygen species (ROS) in tumors make them more susceptible to exogenously induced oxidative stress. Excessive oxidative insults overwhelm their adaptive antioxidant capacity and trigger ROS-mediated cell death. Recently, we have discovered a novel class of quinazolinediones that exert their cytotoxic effects by modulating ROS-mediated signaling. METHODS Cytotoxic potential was determined by colorimetric and colony formation assays. An XF24 Extracellular Flux Analyzer, and colorimetric and fluorescent techniques were used to assess the bioenergetics and oxidative stress effects, respectively. Mechanism was determined by Western blots. RESULTS Compound 3a (6-[(2-acetylphenyl)amino]quinazoline-5,8-dione) was identified through a medium throughput screen of ~1000 highly diverse in-house compounds and chemotherapeutic agents for their ability to alter cellular bioenergetics. Further structural optimizations led to the discovery of a more potent analog, 3b (6-[(3-acetylphenyl)amino]quinazoline-5,8-dione) that displayed anti-proliferative activities in low micromolar range in both drug-sensitive and drug-resistant cancer cells. Treatment with 3b causes Akt activation resulting in increased cellular oxygen consumption and oxidative stress in pancreatic cancer cells. Moreover, oxidative stress induced by 3b promoted activation of stress kinases (p38/JNK) resulting in cancer cell death. Treatment with antioxidants was able to reduce cell death confirming ROS-mediated cytotoxicity. CONCLUSION In conclusion, our novel quinazolinediones are promising lead compounds that selectively induce ROS-mediated cell death in cancer cells and warrant further preclinical studies. GENERAL SIGNIFICANCE Since 3b (6-[(3-acetylphenyl)amino]quinazoline-5,8-dione) exerts Akt-dependent ROS-mediated cell death, it might provide potential therapeutic options for chemoresistant and Akt-overexpressing cancers.
Collapse
Affiliation(s)
- Divya Pathania
- Department of Pharmacology and Pharmaceutical Sciences, University of Southern California, School of Pharmacy, 1985 Zonal Avenue, Los Angeles, CA, USA
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Bey EA, Reinicke KE, Srougi MC, Varnes M, Anderson VE, Pink JJ, Li LS, Patel M, Cao L, Moore Z, Rommel A, Boatman M, Lewis C, Euhus DM, Bornmann WG, Buchsbaum DJ, Spitz DR, Gao J, Boothman DA. Catalase abrogates β-lapachone-induced PARP1 hyperactivation-directed programmed necrosis in NQO1-positive breast cancers. Mol Cancer Ther 2013; 12:2110-20. [PMID: 23883585 DOI: 10.1158/1535-7163.mct-12-0962] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Improving patient outcome by personalized therapy involves a thorough understanding of an agent's mechanism of action. β-Lapachone (clinical forms, Arq501/Arq761) has been developed to exploit dramatic cancer-specific elevations in the phase II detoxifying enzyme NAD(P)H:quinone oxidoreductase (NQO1). NQO1 is dramatically elevated in solid cancers, including primary and metastatic [e.g., triple-negative (ER-, PR-, Her2/Neu-)] breast cancers. To define cellular factors that influence the efficacy of β-lapachone using knowledge of its mechanism of action, we confirmed that NQO1 was required for lethality and mediated a futile redox cycle where ∼120 moles of superoxide were formed per mole of β-lapachone in 2 minutes. β-Lapachone induced reactive oxygen species (ROS), stimulated DNA single-strand break-dependent poly(ADP-ribose) polymerase-1 (PARP1) hyperactivation, caused dramatic loss of essential nucleotides (NAD(+)/ATP), and elicited programmed necrosis in breast cancer cells. Although PARP1 hyperactivation and NQO1 expression were major determinants of β-lapachone-induced lethality, alterations in catalase expression, including treatment with exogenous enzyme, caused marked cytoprotection. Thus, catalase is an important resistance factor and highlights H2O2 as an obligate ROS for cell death from this agent. Exogenous superoxide dismutase enhanced catalase-induced cytoprotection. β-Lapachone-induced cell death included apoptosis-inducing factor (AIF) translocation from mitochondria to nuclei, TUNEL+ staining, atypical PARP1 cleavage, and glyceraldehyde 3-phosphate dehydrogenase S-nitrosylation, which were abrogated by catalase. We predict that the ratio of NQO1:catalase activities in breast cancer versus associated normal tissue are likely to be the major determinants affecting the therapeutic window of β-lapachone and other NQO1 bioactivatable drugs.
Collapse
Affiliation(s)
- Erik A Bey
- Corresponding Authors: Erik A. Bey, West Virginia University, 1 Medical Center Drive, Box 9300, Room 1835, Morgantown, WV 26506.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Du J, Nelson ES, Simons AL, Olney KE, Moser JC, Schrock HE, Wagner BA, Buettner GR, Smith BJ, Teoh ML, Tsao MS, Cullen JJ. Regulation of pancreatic cancer growth by superoxide. Mol Carcinog 2013; 52:555-67. [PMID: 22392697 PMCID: PMC3375391 DOI: 10.1002/mc.21891] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2011] [Revised: 01/31/2012] [Accepted: 02/06/2012] [Indexed: 12/29/2022]
Abstract
K-ras mutations have been identified in up to 95% of pancreatic cancers, implying their critical role in the molecular pathogenesis. Expression of K-ras oncogene in an immortalized human pancreatic ductal epithelial cell line, originally derived from normal pancreas (H6c7), induced the formation of carcinoma in mice. We hypothesized that K-ras oncogene correlates with increased non-mitochondrial-generated superoxide (O 2.-), which could be involved in regulating cell growth contributing to tumor progression. In the H6c7 cell line and its derivatives, H6c7er-Kras+ (H6c7 cells expressing K-ras oncogene), and H6c7eR-KrasT (tumorigenic H6c7 cells expressing K-ras oncogene), there was an increase in hydroethidine fluorescence in cell lines that express K-ras. Western blots and activity assays for the antioxidant enzymes that detoxify O 2.- were similar in these cell lines suggesting that the increase in hydroethidine fluorescence was not due to decreased antioxidant capacity. To determine a possible non-mitochondrial source of the increased levels of O 2.-, Western analysis demonstrated the absence of NADPH oxidase-2 (NOX2) in H6c7 cells but present in the H6c7 cell lines expressing K-ras and other pancreatic cancer cell lines. Inhibition of NOX2 decreased hydroethidine fluorescence and clonogenic survival. Furthermore, in the cell lines with the K-ras oncogene, overexpression of superoxide dismutases that detoxify non-mitochondrial sources of O 2.-, and treatment with the small molecule O 2.- scavenger Tempol, also decreased hydroethidine fluorescence, inhibited clonogenic survival and inhibited growth of tumor xenografts. Thus, O 2.- produced by NOX2 in pancreatic cancer cells with K-ras, may regulate pancreatic cancer cell growth.
Collapse
Affiliation(s)
- Juan Du
- Department of Radiation Oncology, University of Iowa College of Medicine, Iowa City, IA
| | - Elke S. Nelson
- Department of Radiation Oncology, University of Iowa College of Medicine, Iowa City, IA
| | - Andrean L. Simons
- Department of Pathology, University of Iowa College of Medicine, Iowa City, IA
- Holden Comprehensive Cancer Center, Iowa City, IA
| | - Kristen E. Olney
- Department of Radiation Oncology, University of Iowa College of Medicine, Iowa City, IA
| | - Justin C. Moser
- Department of Radiation Oncology, University of Iowa College of Medicine, Iowa City, IA
| | - Hannah E. Schrock
- Department of Radiation Oncology, University of Iowa College of Medicine, Iowa City, IA
| | - Brett A. Wagner
- Department of Radiation Oncology, University of Iowa College of Medicine, Iowa City, IA
| | - Garry R. Buettner
- Department of Radiation Oncology, University of Iowa College of Medicine, Iowa City, IA
- Holden Comprehensive Cancer Center, Iowa City, IA
| | | | - Melissa L.T. Teoh
- Department of Radiation Oncology, University of Iowa College of Medicine, Iowa City, IA
| | - Ming-Sound Tsao
- Department of Pathology and Division of Cellular Molecular Biology and the Ontario Cancer Institute/Princess Margaret Hospital Toronto, and University of Toronto, Ontario, Canada
| | - Joseph J. Cullen
- Department of Radiation Oncology, University of Iowa College of Medicine, Iowa City, IA
- Department of Surgery, University of Iowa College of Medicine, Iowa City, IA
- Holden Comprehensive Cancer Center, Iowa City, IA
- Veterans Affairs Medical Center, Iowa City, IA
| |
Collapse
|
41
|
Thurow S, Webber R, Perin G, Lenardão EJ, Alves D. Glycerol/hypophosphorous acid: an efficient system solvent-reducing agent for the synthesis of 2-organylselanyl pyridines. Tetrahedron Lett 2013. [DOI: 10.1016/j.tetlet.2013.04.057] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
42
|
Tang Z, Wu M, Li Y, Zheng X, Liu H, Cheng X, Xu L, Wang G, Hao H. Absolute quantification of NAD(P)H:quinone oxidoreductase 1 in human tumor cell lines and tissues by liquid chromatography-mass spectrometry/mass spectrometry using both isotopic and non-isotopic internal standards. Anal Chim Acta 2013; 772:59-67. [PMID: 23540248 DOI: 10.1016/j.aca.2013.02.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2012] [Revised: 02/01/2013] [Accepted: 02/08/2013] [Indexed: 12/19/2022]
Abstract
NAD(P)H:quinone oxidoreductase 1 (NQO1, DT-diaphorase) is a prognostic biomarker and a potential therapeutic target for various tumors. Therefore, it is of significance to develop a robust method for the absolute quantification of NQO1. This study aimed to develop and validate a LC-MS/MS based method and to test the appropriateness of using non-isotopic analog peptide as the internal standard (IS) by comparing with a stable isotope labeled (SIL) peptide. The chromatographic performance and mass spectra between the selected signature peptide of NQO1 and the non-isotopic peptide were observed to be very similar. The use of the two internal standards was validated appropriate for the absolute quantification of NQO1, as evidenced by satisfactory validation results over a concentration range of 1.62-162 fmol μL(-1). This method has been successfully applied to the absolute quantification of NQO1 expression in various tumor cell lines and tissues. NQO1 expression in human tumor tissues is much higher than that in the neighboring normal tissues in both the cases of lung and colon cancer. The quantitative results obtained from the isotopic and non-isotopic methods are quite similar, further supporting that the use of non-isotopic analog peptide as internal standard is appropriate and feasible for the quantification of NQO1. By comparing with a classical isotopic IS, the present study indicates that the use of a non-isotopic peptide analog to the proteotypic peptide as the internal standard can get equal accuracy and preciseness in measuring NQO1. The universal applicability of the non-isotopic IS approach for the quantification of proteins warrants further research.
Collapse
Affiliation(s)
- Zhiyuan Tang
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
A simple metal-free synthesis of 2-substituted pyridine-4,5-dicarboxylates and their N-oxides. Tetrahedron 2012. [DOI: 10.1016/j.tet.2012.04.013] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
44
|
Huang X, Dong Y, Bey EA, Kilgore JA, Bair JS, Li LS, Patel M, Parkinson EI, Wang Y, Williams NS, Gao J, Hergenrother PJ, Boothman DA. An NQO1 substrate with potent antitumor activity that selectively kills by PARP1-induced programmed necrosis. Cancer Res 2012; 72:3038-47. [PMID: 22532167 DOI: 10.1158/0008-5472.can-11-3135] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Agents, such as β-lapachone, that target the redox enzyme, NAD(P)H:quinone oxidoreductase 1 (NQO1), to induce programmed necrosis in solid tumors have shown great promise, but more potent tumor-selective compounds are needed. Here, we report that deoxynyboquinone kills a wide spectrum of cancer cells in an NQO1-dependent manner with greater potency than β-lapachone. Deoxynyboquinone lethality relies on NQO1-dependent futile redox cycling that consumes oxygen and generates extensive reactive oxygen species (ROS). Elevated ROS levels cause extensive DNA lesions, PARP1 hyperactivation, and severe NAD+ /ATP depletion that stimulate Ca2+ -dependent programmed necrosis, unique to this new class of NQO1 "bioactivated" drugs. Short-term exposure of NQO1+ cells to deoxynyboquinone was sufficient to trigger cell death, although genetically matched NQO1- cells were unaffected. Moreover, siRNA-mediated NQO1 or PARP1 knockdown spared NQO1+ cells from short-term lethality. Pretreatment of cells with BAPTA-AM (a cytosolic Ca2+ chelator) or catalase (enzymatic H2O2 scavenger) was sufficient to rescue deoxynyboquinone-induced lethality, as noted with β-lapachone. Investigations in vivo showed equivalent antitumor efficacy of deoxynyboquinone to β-lapachone, but at a 6-fold greater potency. PARP1 hyperactivation and dramatic ATP loss were noted in the tumor, but not in the associated normal lung tissue. Our findings offer preclinical proof-of-concept for deoxynyboquinone as a potent chemotherapeutic agent for treatment of a wide spectrum of therapeutically challenging solid tumors, such as pancreatic and lung cancers.
Collapse
Affiliation(s)
- Xiumei Huang
- Department of Pharmacology, UT Southwestern Medical Center, Dallas, Texas 75390, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Isoxazolo(aza)naphthoquinones: a new class of cytotoxic Hsp90 inhibitors. Eur J Med Chem 2012; 53:64-75. [PMID: 22538015 DOI: 10.1016/j.ejmech.2012.03.036] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2011] [Revised: 01/28/2012] [Accepted: 03/20/2012] [Indexed: 12/20/2022]
Abstract
A series of 3-aryl-naphtho[2,3-d]isoxazole-4,9-diones and some of their 6-aza analogues were synthesized and found to inhibit the heat shock protein 90 (Hsp90). The compounds were tested for their binding to Hsp90 and for their effects on Hsp90 client proteins expression in a series of human tumour cell lines. Representative compounds (7f, 10c) downregulated the Hsp90 client proteins EGFR, Akt, Cdk4, Raf-1, and survivin, and upregulated Hsp70. Most of the compounds, in particular the alkylated 3-pyridyl derivatives, exhibited potent antiproliferative activity, down to two-digit nanomolar range. Preliminary results indicated in vivo activity of 7f against human epithelial carcinoma A431 model growing as tumour xenograft in nude mice, thus supporting the therapeutic potential of this novel series of Hsp90 inhibitors.
Collapse
|
46
|
Bongard RD, Krenz GS, Gastonguay AJ, Williams CL, Lindemer BJ, Merker MP. Characterization of the threshold for NAD(P)H:quinone oxidoreductase activity in intact sulforaphane-treated pulmonary arterial endothelial cells. Free Radic Biol Med 2011; 50:953-62. [PMID: 21238579 PMCID: PMC3851029 DOI: 10.1016/j.freeradbiomed.2011.01.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2009] [Revised: 01/03/2011] [Accepted: 01/06/2011] [Indexed: 12/22/2022]
Abstract
Treatment of bovine pulmonary arterial endothelial cells in culture with the phase II enzyme inducer sulforaphane (5μM, 24h; sulf-treated) increased cell-lysate NAD(P)H:quinone oxidoreductase (NQO1) activity by 5.7 ± 0.6 (mean ± SEM)-fold, but intact-cell NQO1 activity by only 2.8 ± 0.1-fold compared to control cells. To evaluate the hypothesis that the threshold for sulforaphane-induced intact-cell NQO1 activity reflects a limitation in the capacity to supply NADPH at a sufficient rate to drive all the induced NQO1 to its maximum activity, total KOH-extractable pyridine nucleotides were measured in cells treated with duroquinone to stimulate maximal NQO1 activity. NQO1 activation increased NADP(+) in control and sulf-treated cells, with the effect more pronounced in the sulf-treated cells, in which the NADPH was also decreased. Glucose-6-phosphate dehydrogenase (G-6-PDH) inhibition partially blocked NQO1 activity in control and sulf-treated cells, but G-6-PDH overexpression via transient transfection with the human cDNA alleviated neither the restriction on intact sulf-treated cell NQO1 activity nor the impact on the NADPH/NADP(+) ratios. Intracellular ATP levels were not affected by NQO1 activation in control or sulf-treated cells. An increased dependence on extracellular glucose and a rightward shift in the K(m) for extracellular glucose were observed in NQO1-stimulated sulf-treated vs control cells. The data suggest that glucose transport in the sulf-treated cells may be insufficient to support the increased metabolic demand for pentose phosphate pathway-generated NADPH as an explanation for the NQO1 threshold.
Collapse
Affiliation(s)
- Robert D Bongard
- Department of Pulmonary Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | | | | | | | | | | |
Collapse
|
47
|
Siegel D, Shieh B, Yan C, Kepa JK, Ross D. Role for NAD(P)H:quinone oxidoreductase 1 and manganese-dependent superoxide dismutase in 17-(allylamino)-17-demethoxygeldanamycin-induced heat shock protein 90 inhibition in pancreatic cancer cells. J Pharmacol Exp Ther 2011; 336:874-80. [PMID: 21156818 PMCID: PMC3061536 DOI: 10.1124/jpet.110.176438] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2010] [Accepted: 12/13/2010] [Indexed: 01/10/2023] Open
Abstract
Previous work demonstrated that NAD(P)H:quinone oxidoreductase 1 (NQO1) metabolized the heat shock protein 90 (Hsp90) inhibitor 17-(allylamino)-17-demethoxygeldanamycin (17AAG) to the corresponding hydroquinone (17AAGH₂). The formation of 17AAGH₂ by NQO1 results in a molecule that binds with greater affinity to Hsp90 compared with the parent quinone. 17AAG induced substantial growth inhibition in human pancreatic cancer cell lines expressing NQO1. Growth inhibition induced by 17AAG could be reduced by pretreatment with 5-methoxy-1,2-dimethyl-3-[(4-nitrophenoxy)methyl]-indole-4,7-dione (ES936), a mechanism-based inhibitor of NQO1. After treatment with 17AAG, biomarkers of Hsp90 inhibition, including markers of cell-cycle arrest, were more pronounced in NQO1-expressing cells compared with NQO1-null cells. The intracellular concentrations of 17AAG and 17AAGH₂ were measured in human pancreatic cancer cells, and it was observed that larger amounts of 17AAG and 17AAGH₂ could be detected in cells with catalytically active NQO1 compared with cells lacking NQO1 activity or cells pretreated with ES936. These data demonstrate that, in addition to generating an inhibitor with greater affinity for Hsp90 (17AAGH₂), reduction of 17AAG to 17AAGH₂ by NQO1 leads to substantially greater intracellular concentrations of 17AAG and 17AAGH₂. In addition, oxidation of 17AAGH₂ could be prevented by superoxide dismutase (SOD), demonstrating that 17AAGH₂ was sensitive to oxidation by superoxide. Stable transfection of manganese-dependent SOD into MiaPaCa-2 cells resulted in a significantly greater intracellular concentration of 17AAGH₂ with a corresponding increase in growth inhibitory activity. These data confirm the role of NQO1 in sensitivity to 17AAG and demonstrate that SOD functions in conjunction with NQO1 to maintain intracellular levels of 17AAGH₂, the active Hsp90 inhibitor derived from 17AAG.
Collapse
Affiliation(s)
- David Siegel
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Colorado-Denver, Aurora, CO 80045, USA.
| | | | | | | | | |
Collapse
|
48
|
Lindemer BJ, Bongard RD, Hoffmann R, Baumgardt S, Gonzalez FJ, Merker MP. Genetic evidence for NAD(P)H:quinone oxidoreductase 1-catalyzed quinone reduction on passage through the mouse pulmonary circulation. Am J Physiol Lung Cell Mol Physiol 2011; 300:L773-80. [PMID: 21296895 DOI: 10.1152/ajplung.00394.2010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The quinones duroquinone (DQ) and coenzyme Q(1) (CoQ(1)) and quinone reductase inhibitors have been used to identify reductases involved in quinone reduction on passage through the pulmonary circulation. In perfused rat lung, NAD(P)H:quinone oxidoreductase 1 (NQO1) was identified as the predominant DQ reductase and NQO1 and mitochondrial complex I as the CoQ(1) reductases. Since inhibitors have nonspecific effects, the goal was to use Nqo1-null (NQO1(-)/(-)) mice to evaluate DQ as an NQO1 probe in the lung. Lung homogenate cytosol NQO1 activities were 97 ± 11, 54 ± 6, and 5 ± 1 (SE) nmol dichlorophenolindophenol reduced·min(-1)·mg protein(-1) for NQO1(+/+), NQO1(+/-), and NQO1(-/-) lungs, respectively. Intact lung quinone reduction was evaluated by infusion of DQ (50 μM) or CoQ(1) (60 μM) into the pulmonary arterial inflow of the isolated perfused lung and measurement of pulmonary venous effluent hydroquinone (DQH(2) or CoQ(1)H(2)). DQH(2) efflux rates for NQO1(+/+), NQO1(+/-), and NQO1(-/-) lungs were 0.65 ± 0.08, 0.45 ± 0.04, and 0.13 ± 0.05 (SE) μmol·min(-1)·g dry lung(-1), respectively. DQ reduction in NQO1(+/+) lungs was inhibited by 90 ± 4% with dicumarol; there was no inhibition in NQO1(-/-) lungs. There was no significant difference in CoQ(1)H(2) efflux rates for NQO1(+/+) and NQO1(-/-) lungs. Differences in DQ reduction were not due to differences in lung dry weights, wet-to-dry weight ratios, perfusion pressures, perfused surface areas, or total DQ recoveries. The data provide genetic evidence implicating DQ as a specific NQO1 probe in the perfused rodent lung.
Collapse
Affiliation(s)
- Brian J Lindemer
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | | | | | | | | | | |
Collapse
|
49
|
Li LS, Bey EA, Dong Y, Meng J, Patra B, Yan J, Xie XJ, Brekken RA, Barnett CC, Bornmann WG, Gao J, Boothman DA. Modulating endogenous NQO1 levels identifies key regulatory mechanisms of action of β-lapachone for pancreatic cancer therapy. Clin Cancer Res 2011; 17:275-85. [PMID: 21224367 DOI: 10.1158/1078-0432.ccr-10-1983] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
PURPOSE Pancreatic cancer is the fourth leading cause of cancer-related deaths, in which the 5-year survival rate is less than 5%. Current standard of care therapies offer little selectivity and high toxicity. Novel, tumor-selective approaches are desperately needed. Although prior work suggested that β-lapachone (β-lap) could be used for the treatment of pancreatic cancers, the lack of knowledge of the compound's mechanism of action prevented optimal use of this agent. EXPERIMENTAL DESIGN We examined the role of NAD(P)H:quinone oxidoreductase-1 (NQO1) in β-lap-mediated antitumor activity, using a series of MIA PaCa-2 pancreatic cancer clones varying in NQO1 levels by stable shRNA knockdown. The antitumor efficacy of β-lap was determined using an optimal hydroxypropyl-β-cyclodextran (HPβ-CD) vehicle formulation in metastatic pancreatic cancer models. RESULTS β-Lap-mediated cell death required ∼90 enzymatic units of NQO1. Essential downstream mediators of lethality were as follows: (i) reactive oxygen species (ROS); (ii) single-strand DNA breaks induced by ROS; (iii) poly(ADP-ribose)polymerase-1 (PARP1) hyperactivation; (iv) dramatic NAD(+)/ATP depletion; and (v) programmed necrosis. We showed that 1 regimen of β-lap therapy (5 treatments every other day) efficaciously regressed and reduced human pancreatic tumor burden and dramatically extended the survival of athymic mice, using metastatic pancreatic cancer models. CONCLUSIONS Because NQO1 enzyme activities are easily measured and commonly overexpressed (i.e., >70%) in pancreatic cancers 5- to 10-fold above normal tissue, strategies using β-lap to efficaciously treat pancreatic cancers are indicated. On the basis of optimal drug formulation and efficacious antitumor efficacy, such a therapy should be extremely safe and not accompanied with normal tissue toxicity or hemolytic anemia.
Collapse
Affiliation(s)
- Long Shan Li
- Department of Pharmacology, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center at Dallas, Texas 75390, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Du J, Liu J, Smith BJ, Tsao MS, Cullen JJ. Role of Rac1-dependent NADPH oxidase in the growth of pancreatic cancer. Cancer Gene Ther 2010; 18:135-43. [PMID: 21037555 PMCID: PMC3058504 DOI: 10.1038/cgt.2010.64] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
K-ras mutations occur in as high as 95% of patients with pancreatic cancer. K-ras activates Rac1-dependent NADPH oxidase, a key source of superoxide. Superoxide plays an important role in pancreatic cancer cell proliferation and scavenging or decreasing the levels of superoxide inhibits pancreatic cancer cell growth both in vitro and in vivo. DNA microarray analysis and RT-PCR has demonstrated that Rac1 is also upregulated in pancreatic cancer. The aim of this study was to determine if inhibiting Rac1 would alter pancreatic tumor cell behavior. Human pancreatic cancer cells with mutant K-ras (MIA PaCa-2), wild-type K-ras (BxPC-3), and the immortal H6c7 cell line (pancreatic ductal epithelium) expressing K-ras oncogene (H6c7eR-KrasT) that is tumorigenic, were infected with a dominant/negative Rac1 construct (AdN17Rac1). In cells with mutant K-ras, AdN17Rac1 decreased rac activity, decreased superoxide levels, and inhibited in vitro growth. However in the BxPC-3 cell line, AdN17Rac1 did not change rac activity, superoxide levels, or in vitro cell growth. Additionally, AdN17Rac1 decreased superoxide levels and inhibited in vitro growth in the KrasT tumorigenic cell line, but had no effect in the immortalized H6c7 cell line. In human pancreatic tumor xenografts, intratumoral injections of AdN17Rac1 inhibited tumor growth. These results suggest that activation of Rac1-dependent superoxide generation leads to pancreatic cancer cell proliferation. In pancreatic cancer inhibition of Rac1 may be a potential therapeutic target.
Collapse
Affiliation(s)
- J Du
- Department of Radiation Oncology, University of Iowa College of Medicine, Iowa City, IA, USA
| | | | | | | | | |
Collapse
|