1
|
Mishra L, Mishra M. Recent progress towards the development of fluorescent probes for the detection of disease-related enzymes. J Mater Chem B 2024. [PMID: 39639834 DOI: 10.1039/d4tb01960a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2024]
Abstract
Normal physiological functions as well as regulatory mechanisms for various pathological conditions depend on the activity of enzymes. Thus, determining the in vivo activity of enzymes is crucial for monitoring the physiological metabolism and diagnosis of diseases. Traditional enzyme detection methods are inefficient for in vivo detection, which have different limitations, such as high cost, laborious, and inevitable invasive procedures, low spatio-temporal resolution, weak anti-interference ability, and restricted scope of application. Because of its non-destructive nature, ultra-environmental sensitivity, and high spatiotemporal resolution, fluorescence imaging technology has emerged as a potent tool for the real-time visualization of live cells, thereby imaging the motility of proteins and intracellular signalling networks in tissues and cells and evaluating the binding and attraction of molecules. In the last few years, significant advancements have been achieved in detecting and imaging enzymes in biological systems. In this regard, the high sensitivity and unparalleled spatiotemporal resolution of fluorescent probes in association with confocal microscopy have garnered significant interest. In this review, we focus on providing a concise summary of the latest developments in the design of fluorogenic probes used for monitoring disease-associated enzymes and their application in biological imaging. We anticipate that this study will attract considerable attention among researchers in the relevant field, encouraging them to pursue advances in the development and application of fluorescent probes for the real-time monitoring of enzyme activity in live cells and in vivo models while ensuring excellent biocompatibility.
Collapse
Affiliation(s)
- Lopamudra Mishra
- Neural Developmental Biology Lab, Department of Life Sciences, National Institute of Technology, Rourkela, Odisha, 769008, India.
| | - Monalisa Mishra
- Neural Developmental Biology Lab, Department of Life Sciences, National Institute of Technology, Rourkela, Odisha, 769008, India.
| |
Collapse
|
2
|
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
|
3
|
Liu H, Hu F, Cao Z, Qu Y, Wen H, Wang X, Li W. High-contrast NIR fluorescent probes for selective detection of NQO1 in breast cancer. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2024; 311:123898. [PMID: 38340443 DOI: 10.1016/j.saa.2024.123898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 01/10/2024] [Accepted: 01/14/2024] [Indexed: 02/12/2024]
Abstract
NAD(P)H:quinone oxidoreductase 1 (NQO1) is a potential biomarker for breast cancer (BC) diagnosis and prognosis. However, existing fluorescent probes for NQO1 detection have limitations such as short emission wavelength, weak fluorescence response, or large background interference. Here, we developed two novel near-infrared (NIR) fluorescent probes, DCl-Q and DCl2-Q, that selectively detect NQO1 activity in BC cells and tissues. They consist of a trimethyl-locked quinone as the recognition group and a donor-π-acceptor structure with halogen atoms as the reporter group. They exhibit strong fluorescence emission at around 660 nm upon binding to NQO1. We demonstrated that they can distinguish BC cells with different NQO1 expression levels and image endogenous NQO1 in tumor-bearing mice. Our probes provide a convenient and highly sensitive tool for BC diagnosis and prognosis based on NQO1 detection.
Collapse
Affiliation(s)
- Hongjing Liu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Feiyang Hu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Zehong Cao
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yi Qu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Hongmei Wen
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Xinzhi Wang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Wei Li
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| |
Collapse
|
4
|
Wang D, Li M, Zhang H, Feng C, Wu L, Yan L. A Novel Redox-Sensitive Drug Delivery System Based on Trimethyl-Locked Polycarbonate. Biomacromolecules 2023; 24:4303-4315. [PMID: 37585690 DOI: 10.1021/acs.biomac.3c00702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/18/2023]
Abstract
Stimuli-responsive polymer nanocarriers, capable of exploiting subtle changes in the tumor microenvironment for controlled drug release, have gained significant attention in cancer therapy. Notably, NAD(P)H: quinone oxidoreductase 1 (NQO1), found to be upregulated in various solid tumors, represents a promising therapeutic target due to its effective capability to enzymatically reduce trimethyl-locked (TML) benzoquinone structures in a physiological condition. In this study, a novel redox-sensitive carbonate monomer, MTC, was synthesized, and its amphiphilic block copolymers were prepared through ring-opening polymerization. By successfully self-assembling poly(ethylene glycol)-b-PMTC micelles, the model drug doxorubicin (DOX) was encapsulated with high efficiency. The micelles exhibited redox-responsive behavior, leading to rapid drug release. In vitro assessments confirmed their excellent biocompatibility and hemocompatibility. Furthermore, the inhibition of the NQO1 enzyme reduced drug release in NQO1-overexpressed cells but not in control cells, resulting in decreased cytotoxicity in the presence of NQO1 enzyme inhibitors. Overall, this study showcases the potential of MTC-based polycarbonate micelles to achieve targeted and specific drug release in the NQO1 enzyme-mediated tumor microenvironment. Therefore, the self-assembly of MTC-based polymers into nanomicelles holds immense promise as intelligent nanocarriers in drug delivery applications.
Collapse
Affiliation(s)
- Dongdong Wang
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan 430070, China
| | - Mu Li
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan 430070, China
| | - Hanning Zhang
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan 430070, China
| | - Congshu Feng
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan 430070, China
| | - Lili Wu
- School of Materials Science and Engineering, Wuhan University of Technology, Wuhan 430070, China
| | - Lesan Yan
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan 430070, China
| |
Collapse
|
5
|
Xie J, Tian S, Zhang H, Feng C, Han Y, Dai H, Yan L. A Novel NQO1 Enzyme-Responsive Polyurethane Nanocarrier for Redox-Triggered Intracellular Drug Release. Biomacromolecules 2023; 24:2225-2236. [PMID: 37040694 DOI: 10.1021/acs.biomac.3c00134] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/13/2023]
Abstract
The design of nano-drug delivery vehicles responsive to tumor microenvironment stimuli has become a crucial aspect in developing cancer therapy in recent years. Among them, the enzyme-responsive nano-drug delivery system is particularly effective, as it utilizes tumor-specific and highly expressed enzymes as precise targets, leading to increased drug release at the target sites, reduced nonspecific release, and improved efficacy while minimizing toxic side effects on normal tissues. NAD(P)H:quinone oxidoreductase 1 (NQO1) is an important reductase associated with cancer and is overexpressed in some cancer cells, particularly in lung and breast cancer. Thus, the design of nanocarriers with high selectivity and responsiveness to NQO1 is of great significance for tumor diagnosis and treatment. It has been reported that under physiological conditions, NQO1 can specifically reduce the trimethyl-locked benzoquinone structure through a two-electron reduction, resulting in rapid lactonization via an enzymatic reaction. Based on this, a novel reduction-sensitive polyurethane (PEG-PTU-PEG) block copolymer was designed and synthesized by copolymerizing diisocyanate, a reduction-sensitive monomer (TMBQ), and poly(ethylene glycol). The successful synthesis of monomers and polymers was verified by nuclear magnetic resonance (1H NMR) and gel permeation chromatography (GPC). Then, the PEG-PTU-PEG micelles were successfully prepared by self-assembly, and their reductive dissociation behavior in the presence of Na2S2O4 was verified by dynamic light scattering (DLS), 1H NMR, and GPC. Next, the model drug doxorubicin (DOX) was encapsulated into the hydrophobic core of this polyurethane micelles by microemulsion method. It was observed that the drug-loaded micelles could also achieve a redox response and rapidly release the encapsulated substances. In vitro cell experiments demonstrated that PEG-PTU-PEG micelles had good biocompatibility and a low hemolysis rate (<5%). Furthermore, in the presence of an NQO1 enzyme inhibitor (dicoumarol), lower drug release from micelles was observed in A549 and 4T1 cells by both fluorescence microscopy and flow cytometry assays, but not in NIH-3T3 control cells. Predictably, DOX-loaded micelles also showed lower cytotoxicity in 4T1 cells in the presence of NQO1 enzyme inhibitors. These results indicate that drug-loaded polyurethane micelles could accomplish specific drug release in the reducing environment in the presence of NQO1 enzymes. Therefore, this study provides a new option for the construction of polyurethane nanocarriers for precise targeting and reductive release, which could benefit the intracellular drug-specific release and precision therapy of tumors.
Collapse
Affiliation(s)
- Jinhai Xie
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan 430070, China
| | - Shuangyu Tian
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan 430070, China
| | - Hanning Zhang
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan 430070, China
| | - Congshu Feng
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan 430070, China
| | - Yingchao Han
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan 430070, China
| | - Honglian Dai
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan 430070, China
| | - Lesan Yan
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan 430070, China
| |
Collapse
|
6
|
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: 1.0] [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
|
7
|
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: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
8
|
Iyanagi T. Roles of Ferredoxin-NADP + Oxidoreductase and Flavodoxin in NAD(P)H-Dependent Electron Transfer Systems. Antioxidants (Basel) 2022; 11:2143. [PMID: 36358515 PMCID: PMC9687028 DOI: 10.3390/antiox11112143] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/19/2022] [Accepted: 10/20/2022] [Indexed: 07/21/2023] Open
Abstract
Distinct isoforms of FAD-containing ferredoxin-NADP+ oxidoreductase (FNR) and ferredoxin (Fd) are involved in photosynthetic and non-photosynthetic electron transfer systems. The FNR (FAD)-Fd [2Fe-2S] redox pair complex switches between one- and two-electron transfer reactions in steps involving FAD semiquinone intermediates. In cyanobacteria and some algae, one-electron carrier Fd serves as a substitute for low-potential FMN-containing flavodoxin (Fld) during growth under low-iron conditions. This complex evolves into the covalent FNR (FAD)-Fld (FMN) pair, which participates in a wide variety of NAD(P)H-dependent metabolic pathways as an electron donor, including bacterial sulfite reductase, cytochrome P450 BM3, plant or mammalian cytochrome P450 reductase and nitric oxide synthase isoforms. These electron transfer systems share the conserved Ser-Glu/Asp pair in the active site of the FAD module. In addition to physiological electron acceptors, the NAD(P)H-dependent diflavin reductase family catalyzes a one-electron reduction of artificial electron acceptors such as quinone-containing anticancer drugs. Conversely, NAD(P)H: quinone oxidoreductase (NQO1), which shares a Fld-like active site, functions as a typical two-electron transfer antioxidant enzyme, and the NQO1 and UDP-glucuronosyltransfease/sulfotransferase pairs function as an antioxidant detoxification system. In this review, the roles of the plant FNR-Fd and FNR-Fld complex pairs were compared to those of the diflavin reductase (FAD-FMN) family. In the final section, evolutionary aspects of NAD(P)H-dependent multi-domain electron transfer systems are discussed.
Collapse
Affiliation(s)
- Takashi Iyanagi
- Graduate School of Life Science, University of Hyogo, 3-2-1 Koto, Akoh 678-1297, Hyogo, Japan
| |
Collapse
|
9
|
Probes and nano-delivery systems targeting NAD(P)H:quinone oxidoreductase 1: a mini-review. Front Chem Sci Eng 2022. [DOI: 10.1007/s11705-022-2194-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/10/2022]
|
10
|
Optical substrates for drug-metabolizing enzymes: Recent advances and future perspectives. Acta Pharm Sin B 2022; 12:1068-1099. [PMID: 35530147 PMCID: PMC9069481 DOI: 10.1016/j.apsb.2022.01.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 10/06/2021] [Accepted: 11/03/2021] [Indexed: 02/08/2023] Open
Abstract
Drug-metabolizing enzymes (DMEs), a diverse group of enzymes responsible for the metabolic elimination of drugs and other xenobiotics, have been recognized as the critical determinants to drug safety and efficacy. Deciphering and understanding the key roles of individual DMEs in drug metabolism and toxicity, as well as characterizing the interactions of central DMEs with xenobiotics require reliable, practical and highly specific tools for sensing the activities of these enzymes in biological systems. In the last few decades, the scientists have developed a variety of optical substrates for sensing human DMEs, parts of them have been successfully used for studying target enzyme(s) in tissue preparations and living systems. Herein, molecular design principals and recent advances in the development and applications of optical substrates for human DMEs have been reviewed systematically. Furthermore, the challenges and future perspectives in this field are also highlighted. The presented information offers a group of practical approaches and imaging tools for sensing DMEs activities in complex biological systems, which strongly facilitates high-throughput screening the modulators of target DMEs and studies on drug/herb‒drug interactions, as well as promotes the fundamental researches for exploring the relevance of DMEs to human diseases and drug treatment outcomes.
Collapse
|
11
|
Yang Y, Zhai H, Yuan J, Wang K, Zhang H. Recent Advances in Fluorescent Probes for Flavinase Activity: Design and Applications. Chem Asian J 2022; 17:e202200043. [PMID: 35174973 DOI: 10.1002/asia.202200043] [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: 01/16/2022] [Revised: 02/14/2022] [Indexed: 11/11/2022]
Abstract
Flavinases, including monoamine oxidase (MAO-A/MAO-B), quinone oxidoreductase (NQO1), thioredoxin reductase (TrxR), nitroreductase (NTR) and so on, are important redox enzymes in organisms. They are considered as biomarkers of cell energy metabolism and cell vitality. Importantly, their aberrant expression is related to various disease processes. Therefore, the accurate measurement of flavinase is useful for the early diagnosis of diseases, which has aroused great concern in the scientific community. Various methods are also available for the detection of flavinases, fluorescence probes are considered to be one of the best detection methods due to their easy and accurate sensing capability. This review aims to introduce the advances in the design and application of flavinase probes in the last five years. This study focuses on analyzing the design strategies and reaction mechanisms of flavinases fluorescent probes and discusses the current challenges, which will further advance the development of diagnostic and therapeutic approaches for flavinase-related diseases.
Collapse
Affiliation(s)
- Yiting Yang
- Henan Normal University School of Chemistry and Chemical Engineering, School of Chemistry and Chemical Engineering, CHINA
| | - Hongchen Zhai
- Henan Normal University School of Chemistry and Chemical Engineering, School of Chenistry and chemical Engineering, CHINA
| | - Jie Yuan
- Henan Normal University School of Chemistry and Chemical Engineering, School of Chemistry and Chemical Engineering, CHINA
| | - Kui Wang
- Henan Normal University School of Chemistry and Chemical Engineering, School of Chemistry and Chemical Engineering, CHINA
| | - Hua Zhang
- Henan Normal University, School of Chemistry and Chemical Engineering, 46 Jianshe Road, Muye Zone,, 453007, Xinxiang, CHINA
| |
Collapse
|
12
|
Research advances in NQO1-responsive prodrugs and nanocarriers for cancer treatment. Future Med Chem 2022; 14:363-383. [PMID: 35102756 DOI: 10.4155/fmc-2021-0289] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
NAD(P)H: quinine oxidoreductase (NQO1) is a class of flavoprotein enzymes commonly expressed in eukaryotic cells. It actively participates in the metabolism of various quinones and their in vivo bioactivation through electron reduction reactions. The expression level of NQO1 is highly upregulated in many solid tumor cells compared with that in normal cells. NQO1 has been considered a candidate molecular target because of its overexpression and bioactivity in different tumors. NQO1-responsive prodrugs and nanocarriers have recently been identified as effective objectives for achieving controlled drug release, reducing adverse reactions and improving clinical efficacy. This review systematically introduces the research advances in applying NQO1-responsive prodrugs and nanocarriers to cancer treatment. It also discusses the existing problems and the developmental prospects of these two antitumor drug delivery systems.
Collapse
|
13
|
Luo Y, Wang W, Zeng Y, Wang S, Guo X, Hu R, Yang G. A bioluminescent probe for NQO1 overexpressing cancer cell imaging in vitro and in vivo. Analyst 2022; 147:5264-5268. [DOI: 10.1039/d2an01435a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
A bioluminescent probe NQO1-Luc toward NQO1 was constructed, which exhibits high selectivity and sensitivity toward NQO1 in vitro and adequate capability of distinguishing NQO1-overexpressing tumors in vivo.
Collapse
Affiliation(s)
- Yuhong Luo
- Beijing National Laboratory for Molecular Sciences (BNLMS), Key Laboratory of Photochemistry, Institute of Chemistry, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100190, China
| | - Wentao Wang
- Beijing National Laboratory for Molecular Sciences (BNLMS), Key Laboratory of Photochemistry, Institute of Chemistry, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100190, China
| | - Yi Zeng
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100190, China
| | - Shuangqing Wang
- Beijing National Laboratory for Molecular Sciences (BNLMS), Key Laboratory of Photochemistry, Institute of Chemistry, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100190, China
| | - Xudong Guo
- Beijing National Laboratory for Molecular Sciences (BNLMS), Key Laboratory of Photochemistry, Institute of Chemistry, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100190, China
| | - Rui Hu
- Beijing National Laboratory for Molecular Sciences (BNLMS), Key Laboratory of Photochemistry, Institute of Chemistry, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100190, China
| | - Guoqiang Yang
- Beijing National Laboratory for Molecular Sciences (BNLMS), Key Laboratory of Photochemistry, Institute of Chemistry, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100190, China
| |
Collapse
|
14
|
Pei Q, Lu S, Zhou J, Jiang B, Li C, Xie Z, Jing X. Intracellular Enzyme-Responsive Profluorophore and Prodrug Nanoparticles for Tumor-Specific Imaging and Precise Chemotherapy. ACS APPLIED MATERIALS & INTERFACES 2021; 13:59708-59719. [PMID: 34879654 DOI: 10.1021/acsami.1c19058] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Responsive drug delivery systems possess great potential in disease diagnosis and treatment. Herein, we develop an activatable prodrug and fluorescence imaging material by engineering the endogenous NAD(P)H:quinone oxidoreductase-1 (NQO1) responsive linker. The as-prepared nanomaterials possess the NQO1-switched drug release and fluorescence enablement, which realizes the tumor-specific chemotherapy and imaging in living mice. The enzyme-sensitive prodrug nanoparticles exhibit selectively potent anticancer performance to NQO1-positive cancer and ignorable off-target toxicity. This work provides an alternative strategy for constructing smart prodrug nanoplatforms with precision, selectivity, and practicability for advanced cancer imaging and therapy.
Collapse
Affiliation(s)
- Qing Pei
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, P. R. China
| | - Shaojin Lu
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, P. R. China
- University of Science and Technology of China, Hefei, Anhui 230026, P. R. China
| | - Junli Zhou
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, P. R. China
- University of Science and Technology of China, Hefei, Anhui 230026, P. R. China
| | - Bowen Jiang
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, P. R. China
- University of Science and Technology of China, Hefei, Anhui 230026, P. R. China
| | - Chaonan Li
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, P. R. China
- University of Science and Technology of China, Hefei, Anhui 230026, P. R. China
| | - Zhigang Xie
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, P. R. China
- University of Science and Technology of China, Hefei, Anhui 230026, P. R. China
| | - Xiabin Jing
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, P. R. China
| |
Collapse
|
15
|
Tantak MP, Sekhar V, Tao X, Zhai RG, Phanstiel O. Development of a Redox-Sensitive Spermine Prodrug for the Potential Treatment of Snyder Robinson Syndrome. J Med Chem 2021; 64:15593-15607. [PMID: 34695351 DOI: 10.1021/acs.jmedchem.1c00419] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Snyder Robinson Syndrome (SRS) is a rare disease associated with a defective spermine synthase gene and low intracellular spermine levels. In this study, a spermine replacement therapy was developed using a spermine prodrug that enters cells via the polyamine transport system. The prodrug was comprised of three components: a redox-sensitive quinone "trigger", a "trimethyl lock (TML)" aryl "release mechanism", and spermine. The presence of spermine in the design facilitated uptake by the polyamine transport system. The quinone-TML motifs provided a redox-sensitive agent, which upon intracellular reduction generated a hydroquinone, which underwent intramolecular cyclization to release free spermine and a lactone byproduct. Rewardingly, most SRS fibroblasts treated with the prodrug revealed a significant increase in intracellular spermine. Administering the spermine prodrug through feeding in a Drosophila model of SRS showed significant beneficial effects. In summary, a spermine prodrug is developed and provides a lead compound for future spermine replacement therapy experiments.
Collapse
Affiliation(s)
- Mukund P Tantak
- Department of Medical Education, College of Medicine, University of Central Florida, 12722 Research Parkway, Orlando, Florida 32826-3227, United States
| | - Vandana Sekhar
- Department of Medical Education, College of Medicine, University of Central Florida, 12722 Research Parkway, Orlando, Florida 32826-3227, United States
| | - Xianzun Tao
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, 1600 NW 10th Ave, Miami, Florida 33136, United States
| | - R Grace Zhai
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, 1600 NW 10th Ave, Miami, Florida 33136, United States
| | - Otto Phanstiel
- Department of Medical Education, College of Medicine, University of Central Florida, 12722 Research Parkway, Orlando, Florida 32826-3227, United States
| |
Collapse
|
16
|
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: 3.3] [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
|
17
|
In silico design of novel diamino-quinoline-5,8‑dione derivatives as putative inhibitors of NAD(P)H:Quinone oxidoreductase 1 based on docking studies and molecular dynamics simulations. J Mol Struct 2021. [DOI: 10.1016/j.molstruc.2021.129906] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
18
|
Sun Y, Davis E. Nanoplatforms for Targeted Stimuli-Responsive Drug Delivery: A Review of Platform Materials and Stimuli-Responsive Release and Targeting Mechanisms. NANOMATERIALS (BASEL, SWITZERLAND) 2021; 11:746. [PMID: 33809633 PMCID: PMC8000772 DOI: 10.3390/nano11030746] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 03/05/2021] [Accepted: 03/08/2021] [Indexed: 12/12/2022]
Abstract
To achieve the promise of stimuli-responsive drug delivery systems for the treatment of cancer, they should (1) avoid premature clearance; (2) accumulate in tumors and undergo endocytosis by cancer cells; and (3) exhibit appropriate stimuli-responsive release of the payload. It is challenging to address all of these requirements simultaneously. However, the numerous proof-of-concept studies addressing one or more of these requirements reported every year have dramatically expanded the toolbox available for the design of drug delivery systems. This review highlights recent advances in the targeting and stimuli-responsiveness of drug delivery systems. It begins with a discussion of nanocarrier types and an overview of the factors influencing nanocarrier biodistribution. On-demand release strategies and their application to each type of nanocarrier are reviewed, including both endogenous and exogenous stimuli. Recent developments in stimuli-responsive targeting strategies are also discussed. The remaining challenges and prospective solutions in the field are discussed throughout the review, which is intended to assist researchers in overcoming interdisciplinary knowledge barriers and increase the speed of development. This review presents a nanocarrier-based drug delivery systems toolbox that enables the application of techniques across platforms and inspires researchers with interdisciplinary information to boost the development of multifunctional therapeutic nanoplatforms for cancer therapy.
Collapse
Affiliation(s)
| | - Edward Davis
- Materials Engineering Program, Mechanical Engineering Department, Auburn University, 101 Wilmore Drive, Auburn, AL 36830, USA;
| |
Collapse
|
19
|
Huang J, Wang C, Lin M, Zeng F, Wu S. Synthesis of NQO1-activatable Optoacoustic Probe and Its Imaging of Breast Cancer. ACTA CHIMICA SINICA 2021. [DOI: 10.6023/a20100459] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
20
|
Su D, Chen X, Zhang Y, Gao X. Activatable imaging probes for cancer-linked NAD(P)H:quinone oxidoreductase-1 (NQO1): Advances and future prospects. Trends Analyt Chem 2020. [DOI: 10.1016/j.trac.2020.116112] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
21
|
Nemeikaitė-Čėnienė A, Šarlauskas J, Misevičienė L, Marozienė A, Jonušienė V, Lesanavičius M, Čėnas N. Aerobic Cytotoxicity of Aromatic N-Oxides: The Role of NAD(P)H:Quinone Oxidoreductase (NQO1). Int J Mol Sci 2020; 21:ijms21228754. [PMID: 33228195 PMCID: PMC7699506 DOI: 10.3390/ijms21228754] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 11/13/2020] [Accepted: 11/18/2020] [Indexed: 12/24/2022] Open
Abstract
Derivatives of tirapazamine and other heteroaromatic N-oxides (ArN→O) exhibit tumoricidal, antibacterial, and antiprotozoal activities, which are typically attributed to bioreductive activation and free radical generation. In this work, we aimed to clarify the role of NAD(P)H:quinone oxidoreductase (NQO1) in ArN→O aerobic cytotoxicity. We synthesized 9 representatives of ArN→O with uncharacterized redox properties and examined their single-electron reduction by rat NADPH:cytochrome P-450 reductase (P-450R) and Plasmodium falciparum ferredoxin:NADP+ oxidoreductase (PfFNR), and by rat NQO1. NQO1 catalyzed both redox cycling and the formation of stable reduction products of ArN→O. The reactivity of ArN→O in NQO1-catalyzed reactions did not correlate with the geometric average of their activity towards P-450R- and PfFNR, which was taken for the parameter of their redox cycling efficacy. The cytotoxicity of compounds in murine hepatoma MH22a cells was decreased by antioxidants and the inhibitor of NQO1, dicoumarol. The multiparameter regression analysis of the data of this and a previous study (DOI: 10.3390/ijms20184602) shows that the cytotoxicity of ArN→O (n = 18) in MH22a and human colon carcinoma HCT-116 cells increases with the geometric average of their reactivity towards P-450R and PfFNR, and with their reactivity towards NQO1. These data demonstrate that NQO1 is a potentially important target of action of heteroaromatic N-oxides.
Collapse
Affiliation(s)
- Aušra Nemeikaitė-Čėnienė
- State Research Institute Center for Innovative Medicine, Santariškių St. 5, LT-08406 Vilnius, Lithuania;
| | - Jonas Šarlauskas
- Institute of Biochemistry of Vilnius University, Saulėtekio 7, LT-10257 Vilnius, Lithuania; (J.Š.); (L.M.); (A.M.); (M.L.)
| | - Lina Misevičienė
- Institute of Biochemistry of Vilnius University, Saulėtekio 7, LT-10257 Vilnius, Lithuania; (J.Š.); (L.M.); (A.M.); (M.L.)
| | - Audronė Marozienė
- Institute of Biochemistry of Vilnius University, Saulėtekio 7, LT-10257 Vilnius, Lithuania; (J.Š.); (L.M.); (A.M.); (M.L.)
| | - Violeta Jonušienė
- Institute of Biosciences of Vilnius University, Saulėtekio 7, LT-10257 Vilnius, Lithuania;
| | - Mindaugas Lesanavičius
- Institute of Biochemistry of Vilnius University, Saulėtekio 7, LT-10257 Vilnius, Lithuania; (J.Š.); (L.M.); (A.M.); (M.L.)
| | - Narimantas Čėnas
- Institute of Biochemistry of Vilnius University, Saulėtekio 7, LT-10257 Vilnius, Lithuania; (J.Š.); (L.M.); (A.M.); (M.L.)
- Correspondence: ; Tel.: +370-5-223-4392
| |
Collapse
|
22
|
Synthesis and biological evaluation of NQO1-activated prodrugs of podophyllotoxin as antitumor agents. Bioorg Med Chem 2020; 28:115821. [PMID: 33091789 DOI: 10.1016/j.bmc.2020.115821] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 10/05/2020] [Indexed: 12/27/2022]
Abstract
Podophyllotoxin (PPT), a toxic polyphenol derived from the roots of genus Podophyllum, had been reported with strong inhibition on both normal human cells and tumor cells, which hindered the development of PPT as the candidate antitumor agent. In the present work, multiple NQO1-activatable PPT prodrugs were synthesized for reducing normal cell toxicity and keeping tumor cell toxicity. The antiproliferative activities in vitro showed prodrug 3 was greatly selectively toxic to tumor cells over-expressing NQO1, taxol-resistant A549, hypoxia A549 and HepG2, and lower damage to normal cells in comparison with podophyllotoxin, prodrug 1 and 2. As elucidated by further mechanistic research, prodrug 3 was activated via NQO1 to efficiently while gently produce cytotoxic PPT units and kill tumor cells. In additions, in vivo study revealed that 3 significantly suppressed cancer growth in HepG2 xenograft models without obvious toxicity. Therefore, this NQO1-activatable prodrug delivery system exhibits good biosafety and provides a novel strategy for the development of drug delivery systems.
Collapse
|
23
|
Zhang C, Qu Y, Ma X, Li M, Li S, Li Y, Wu L. NQO1-selective activated prodrugs of combretastatin A-4: Synthesis and biological evaluation. Bioorg Chem 2020; 103:104200. [DOI: 10.1016/j.bioorg.2020.104200] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 07/28/2020] [Accepted: 07/31/2020] [Indexed: 10/23/2022]
|
24
|
Nemeikaitė-Čėnienė A, Šarlauskas J, Jonušienė V, Marozienė A, Misevičienė L, Yantsevich AV, Čėnas N. Kinetics of Flavoenzyme-Catalyzed Reduction of Tirapazamine Derivatives: Implications for Their Prooxidant Cytotoxicity. Int J Mol Sci 2019; 20:ijms20184602. [PMID: 31533349 PMCID: PMC6769651 DOI: 10.3390/ijms20184602] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 09/06/2019] [Accepted: 09/11/2019] [Indexed: 12/23/2022] Open
Abstract
Derivatives of tirapazamine and other heteroaromatic N-oxides (ArN→O) exhibit promising antibacterial, antiprotozoal, and tumoricidal activities. Their action is typically attributed to bioreductive activation and free radical generation. In this work, we aimed to clarify the mechanism(s) of aerobic mammalian cell cytotoxicity of ArN→O performing the parallel studies of their reactions with NADPH:cytochrome P-450 reductase (P-450R), adrenodoxin reductase/adrenodoxin (ADR/ADX), and NAD(P)H:quinone oxidoreductase (NQO1); we found that in P-450R and ADR/ADX-catalyzed single-electron reduction, the reactivity of ArN→O (n = 9) increased with their single-electron reduction midpoint potential (E17), and correlated with the reactivity of quinones. NQO1 reduced ArN→O at low rates with concomitant superoxide production. The cytotoxicity of ArN→O in murine hepatoma MH22a and human colon adenocarcinoma HCT-116 cells increased with their E17, being systematically higher than that of quinones. The cytotoxicity of both groups of compounds was prooxidant. Inhibitor of NQO1, dicoumarol, and inhibitors of cytochromes P-450 α-naphthoflavone, isoniazid and miconazole statistically significantly (p < 0.02) decreased the toxicity of ArN→O, and potentiated the cytotoxicity of quinones. One may conclude that in spite of similar enzymatic redox cycling rates, the cytotoxicity of ArN→O is higher than that of quinones. This is partly attributed to ArN→O activation by NQO1 and cytochromes P-450. A possible additional factor in the aerobic cytotoxicity of ArN→O is their reductive activation in oxygen-poor cell compartments, leading to the formation of DNA-damaging species similar to those forming under hypoxia.
Collapse
Affiliation(s)
- Aušra Nemeikaitė-Čėnienė
- State Research Institute Center for Innovative Medicine, Santariškių St. 5, LT-08406 Vilnius, Lithuania.
| | - Jonas Šarlauskas
- Department of Xenobiotics Biochemistry, Institute of Biochemistry of Vilnius University, Saulėtekio 7, LT-10257 Vilnius, Lithuania.
| | - Violeta Jonušienė
- Department of Biochemistry and Molecular Biology, Institute of Biosciences of Vilnius University, Saulėtekio 7, LT-10257 Vilnius, Lithuania.
| | - Audronė Marozienė
- Department of Xenobiotics Biochemistry, Institute of Biochemistry of Vilnius University, Saulėtekio 7, LT-10257 Vilnius, Lithuania.
| | - Lina Misevičienė
- Department of Xenobiotics Biochemistry, Institute of Biochemistry of Vilnius University, Saulėtekio 7, LT-10257 Vilnius, Lithuania.
| | - Aliaksei V Yantsevich
- Institute of Bioorganic Chemistry, NAS of Belarus, Kuprevicha 5/2, BY-220072 Minsk, Belarus.
| | - Narimantas Čėnas
- Department of Xenobiotics Biochemistry, Institute of Biochemistry of Vilnius University, Saulėtekio 7, LT-10257 Vilnius, Lithuania.
| |
Collapse
|
25
|
Punganuru SR, Madala HR, Arutla V, Zhang R, Srivenugopal KS. Characterization of a highly specific NQO1-activated near-infrared fluorescent probe and its application for in vivo tumor imaging. Sci Rep 2019; 9:8577. [PMID: 31189950 PMCID: PMC6562040 DOI: 10.1038/s41598-019-44111-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 05/09/2019] [Indexed: 12/30/2022] Open
Abstract
The Near-infrared Fluorescence (NIRF) molecular imaging of cancer is known to be superior in sensitivity, deeper penetration, and low phototoxicity compared to other imaging modalities. In view of an increased need for efficient and targeted imaging agents, we synthesized a NAD(P)H quinone oxidoreductase 1 (NQO1)-activatable NIR fluorescent probe (NIR-ASM) by conjugating dicyanoisophorone (ASM) fluorophore with the NQO1 substrate quinone propionic acid (QPA). The probe remained non-fluorescent until activation by NQO1, whose expression is largely limited to malignant tissues. With a large Stokes shift (186 nm) and a prominent near-infrared emission (646 nm) in response to NQO1, NIR-ASM was capable of monitoring NQO1 activity in vitro and in vivo with high specificity and selectivity. We successfully employed the NIR-ASM to differentiate cancer cells from normal cells based on NQO1 activity using fluorescence microscopy and flow cytometry. Chemical and genetic approaches involving the use of ES936, a specific inhibitor of NQO1 and siRNA and gene transfection procedures unambiguously demonstrated NQO1 to be the sole target activating the NIR-ASM in cell cultures. NIR-ASM was successfully used to detect and image the endogenous NQO1 in three live tumor-bearing mouse models (A549 lung cancer, Lewis lung carcinoma, and MDMAMB 231 xenografts) with a high signal-to-low noise ratiometric NIR fluorescence response. When the NQO1-proficient A549 tumors and NQO1-deficient MDA-MB-231 tumors were developed in the same animal, only the A549 malignancies activated the NIR-ASM probe with a strong signal. Because of its high sensitivity, rapid activation, tumor selectivity, and nontoxic properties, the NIR-ASM appears to be a promising agent with clinical applications.
Collapse
Affiliation(s)
- Surendra Reddy Punganuru
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, 79106, USA.
| | - Hanumantha Rao Madala
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, 79106, USA
| | - Viswanath Arutla
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, 79106, USA
| | - Ruiwen Zhang
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, 77204, USA
| | - Kalkunte S Srivenugopal
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, 79106, USA.
| |
Collapse
|
26
|
Wu X, Li X, Li Z, Yu Y, You Q, Zhang X. Discovery of Nonquinone Substrates for NAD(P)H: Quinone Oxidoreductase 1 (NQO1) as Effective Intracellular ROS Generators for the Treatment of Drug-Resistant Non-Small-Cell Lung Cancer. J Med Chem 2018; 61:11280-11297. [PMID: 30508483 DOI: 10.1021/acs.jmedchem.8b01424] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The elevation of oxidative stress preferentially in cancer cells by efficient NQO1 substrates, which promote ROS generation through redox cycling, has emerged as an effective strategy for cancer therapy, even for treating drug-resistant cancers. Here, we described the identification and structural optimization studies of the hit compound 1, a new chemotype of nonquinone substrate for NQO1 as an efficient ROS generator. Further structure-activity relationship studies resulted in the most active compound 20k, a tricyclic 2,3-dicyano indenopyrazinone, which selectively inhibited the proliferation of NQO1-overexpressing A549 and A549/Taxol cancer cells. Furthermore, 20k dramatically elevated the intracellular ROS levels through NQO1-catalyzed redox cycling and induced PARP-1-mediated cell apoptosis in A549/Taxol cells. In addition, 20k significantly suppressed the growth of A549/Taxol xenograft tumors in mice with no apparent toxicity observed in vivo. Together, 20k acts as an efficient NQO1 substrate and may be a new option for the treatment of NQO1-overexpresssing drug-resistant NSCLC.
Collapse
Affiliation(s)
- Xingsen Wu
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization , China Pharmaceutical University , Nanjing , 210009 , China.,Department of Chemistry, School of Science , China Pharmaceutical University , Nanjing , 211198 , China
| | - Xiang Li
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization , China Pharmaceutical University , Nanjing , 210009 , China.,Department of Pharmaceutical Engineering , China Pharmaceutical University , Nanjing , 211198 , China
| | - Zhihong Li
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization , China Pharmaceutical University , Nanjing , 210009 , China.,Department of Chemistry, School of Science , China Pharmaceutical University , Nanjing , 211198 , China
| | - Yancheng Yu
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization , China Pharmaceutical University , Nanjing , 210009 , China.,Department of Chemistry, School of Science , China Pharmaceutical University , Nanjing , 211198 , China
| | - Qidong You
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization , China Pharmaceutical University , Nanjing , 210009 , China
| | - Xiaojin Zhang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization , China Pharmaceutical University , Nanjing , 210009 , China.,Department of Chemistry, School of Science , China Pharmaceutical University , Nanjing , 211198 , China
| |
Collapse
|
27
|
Cancer-Specific Biomarker hNQO1-Activatable Fluorescent Probe for Imaging Cancer Cells In Vitro and In Vivo. Cancers (Basel) 2018; 10:cancers10120470. [PMID: 30487423 PMCID: PMC6316840 DOI: 10.3390/cancers10120470] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 11/21/2018] [Accepted: 11/27/2018] [Indexed: 01/19/2023] Open
Abstract
Human NAD(P)H quinone oxidoreductase-1 (hNQO1) is an important cancer-related biomarker, which shows significant overexpression in malignant cells. Developing an effective method for detecting NQO1 activity with high sensitivity and selectivity in tumors holds a great potential for cancer diagnosis, treatment, and management. In the present study, we report a new dicyanoisophorone (DCP) based fluorescent probe (NQ-DCP) capable of monitoring hNQO1 activity in vitro and in vivo in both ratiometric and turn-on model. NQ-DCP was prepared by conjugating dicyanoisophorone fluoroprobe with hNQO1 activatable quinone propionic acid (QPA), which remain non-fluorescent until activation by tumor-specific hNQO1. NQ-DCP featured a large Stokes shift (145 nm), excellent biocompatibility, cell permeability, and selectivity towards hNQO1 allowed to differentiate cancer cells from healthy cells. We have successfully employed NQ-DCP to monitor non-invasive endogenous hNQO1 activity in brain tumor cells in vitro and in xenografted tumors developed in nude mice.
Collapse
|
28
|
Zeng Y, Ma J, Zhan Y, Xu X, Zeng Q, Liang J, Chen X. Hypoxia-activated prodrugs and redox-responsive nanocarriers. Int J Nanomedicine 2018; 13:6551-6574. [PMID: 30425475 PMCID: PMC6202002 DOI: 10.2147/ijn.s173431] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Hypoxia is one of the marked features of malignant tumors, which is associated with several adaptation changes in the microenvironment of tumor cells. Therefore, targeting tumor hypoxia is a research hotspot for cancer therapy. In this review, we summarize the developing chemotherapeutic drugs for targeting hypoxia, including quinones, nitroaromatic/nitroimidazole, N-oxides, and transition metal complexes. In addition, redox-responsive bonds, such as nitroimidazole groups, azogroups, and disulfide bonds, are frequently used in drug delivery systems for targeting the redox environment of tumors. Both hypoxia-activated prodrugs and redox-responsive drug delivery nanocarriers have significant effects on targeting tumor hypoxia for cancer therapy. Hypoxia-activated prodrugs are commonly used in clinical trials with favorable prospects, while redox-responsive nanocarriers are currently at the experimental stage.
Collapse
Affiliation(s)
- Yun Zeng
- Engineering Research Center of Molecular and Neuro Imaging of the Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an 710071, Shaanxi Province, People's Republic of China, ,
| | - Jingwen Ma
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing 210009, Jiangsu Province, People's Republic of China
| | - Yonghua Zhan
- Engineering Research Center of Molecular and Neuro Imaging of the Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an 710071, Shaanxi Province, People's Republic of China, ,
| | - Xinyi Xu
- Engineering Research Center of Molecular and Neuro Imaging of the Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an 710071, Shaanxi Province, People's Republic of China, ,
| | - Qi Zeng
- Engineering Research Center of Molecular and Neuro Imaging of the Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an 710071, Shaanxi Province, People's Republic of China, ,
| | - Jimin Liang
- Engineering Research Center of Molecular and Neuro Imaging of the Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an 710071, Shaanxi Province, People's Republic of China, ,
| | - Xueli Chen
- Engineering Research Center of Molecular and Neuro Imaging of the Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an 710071, Shaanxi Province, People's Republic of China, ,
| |
Collapse
|
29
|
Okoh OA, Klahn P. Trimethyl Lock: A Multifunctional Molecular Tool for Drug Delivery, Cellular Imaging, and Stimuli-Responsive Materials. Chembiochem 2018; 19:1668-1694. [PMID: 29888433 DOI: 10.1002/cbic.201800269] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Indexed: 12/13/2022]
Abstract
Trimethyl lock (TML) systems are based on ortho-hydroxydihydrocinnamic acid derivatives displaying increased lactonization reactivity owing to unfavorable steric interactions of three pendant methyl groups, and this leads to the formation of hydrocoumarins. Protection of the phenolic hydroxy function or masking of the reactivity as benzoquinone derivatives prevents lactonization and provides a trigger for controlled release of molecules attached to the carboxylic acid function through amides, esters, or thioesters. Their easy synthesis and possible chemical adaption to several different triggers make TML a highly versatile module for the development of drug-delivery systems, prodrug approaches, cell-imaging tools, molecular tools for supramolecular chemistry, as well as smart stimuliresponsive materials.
Collapse
Affiliation(s)
- Okoh Adeyi Okoh
- Institute for Organic Chemistry, Technische Universität Braunschweig, Hagenring 30, 38106, Braunschweig, Germany
| | - Philipp Klahn
- Institute for Organic Chemistry, Technische Universität Braunschweig, Hagenring 30, 38106, Braunschweig, Germany
| |
Collapse
|
30
|
Zhang X, Li X, Li Z, Wu X, Wu Y, You Q, Zhang X. An NAD(P)H:Quinone Oxidoreductase 1 Responsive and Self-Immolative Prodrug of 5-Fluorouracil for Safe and Effective Cancer Therapy. Org Lett 2018; 20:3635-3638. [PMID: 29847952 DOI: 10.1021/acs.orglett.8b01409] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Tripartite prodrug 1, composed of an NAD(P)H:quinone oxidoreductase 1 (NQO1)-responsive trigger group, a self-immolative linker, and the active drug 5-fluorouracil (5-FU), was designed and synthesized for site-specific cancer therapy. Upon bioreductive activation by NQO1, 1 can release the parent drug 5-FU specifically in NQO1-overexpressing cancer cells. This prodrug exerts comparable antitumor activity and a more favorable safety profile compared with 5-FU both in vitro and in vivo.
Collapse
Affiliation(s)
- Xian Zhang
- Sate Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization , China Pharmaceutical University , Nanjing 210009 , China
| | - Xiang Li
- Sate Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization , China Pharmaceutical University , Nanjing 210009 , China
| | - Zhihong Li
- Sate Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization , China Pharmaceutical University , Nanjing 210009 , China.,Department of Organic Chemistry, School of Science , China Pharmaceutical University , Nanjing 211198 , China
| | - Xingsen Wu
- Sate Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization , China Pharmaceutical University , Nanjing 210009 , China.,Department of Organic Chemistry, School of Science , China Pharmaceutical University , Nanjing 211198 , China
| | - Yue Wu
- Sate Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization , China Pharmaceutical University , Nanjing 210009 , China
| | - Qidong You
- Sate Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization , China Pharmaceutical University , Nanjing 210009 , China
| | - Xiaojin Zhang
- Sate Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization , China Pharmaceutical University , Nanjing 210009 , China.,Department of Organic Chemistry, School of Science , China Pharmaceutical University , Nanjing 211198 , China
| |
Collapse
|
31
|
Zhang K, Chen D, Ma K, Wu X, Hao H, Jiang S. NAD(P)H:Quinone Oxidoreductase 1 (NQO1) as a Therapeutic and Diagnostic Target in Cancer. J Med Chem 2018; 61:6983-7003. [DOI: 10.1021/acs.jmedchem.8b00124] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Kuojun Zhang
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Dong Chen
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Kun Ma
- Center for Drug Evaluation, China Food and Drug Administration, Beijing 100038, China
| | - Xiaoxing Wu
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Haiping Hao
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Sheng Jiang
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| |
Collapse
|
32
|
Lee Y, Thompson DH. Stimuli-responsive liposomes for drug delivery. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2017; 9:10.1002/wnan.1450. [PMID: 28198148 PMCID: PMC5557698 DOI: 10.1002/wnan.1450] [Citation(s) in RCA: 240] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Revised: 11/23/2016] [Accepted: 11/27/2016] [Indexed: 12/25/2022]
Abstract
The ultimate goal of drug delivery is to increase the bioavailability and reduce the toxic side effects of the active pharmaceutical ingredient (API) by releasing them at a specific site of action. In the case of antitumor therapy, association of the therapeutic agent with a carrier system can minimize damage to healthy, nontarget tissues, while limit systemic release and promoting long circulation to enhance uptake at the cancerous site due to the enhanced permeation and retention effect (EPR). Stimuli-responsive systems have become a promising way to deliver and release payloads in a site-selective manner. Potential carrier systems have been derived from a wide variety of materials, including inorganic nanoparticles, lipids, and polymers that have been imbued with stimuli-sensitive properties to accomplish triggered release based on an environmental cue. The unique features in the tumor microenvironment can serve as an endogenous stimulus (pH, redox potential, or unique enzymatic activity) or the locus of an applied external stimulus (heat or light) to trigger the controlled release of API. In liposomal carrier systems triggered release is generally based on the principle of membrane destabilization from local defects within bilayer membranes to effect release of liposome-entrapped drugs. This review focuses on the literature appearing between November 2008-February 2016 that reports new developments in stimuli-sensitive liposomal drug delivery strategies using pH change, enzyme transformation, redox reactions, and photochemical mechanisms of activation. WIREs Nanomed Nanobiotechnol 2017, 9:e1450. doi: 10.1002/wnan.1450 For further resources related to this article, please visit the WIREs website.
Collapse
Affiliation(s)
- Y Lee
- Department of Chemistry, Purdue University, West Lafayette, IN, USA
| | - D H Thompson
- Department of Chemistry, Purdue University, West Lafayette, IN, USA
| |
Collapse
|
33
|
Zhang X, Bian J, Li X, Wu X, Dong Y, You Q. 2-Substituted 3,7,8-trimethylnaphtho[1,2- b ]furan-4,5-diones as specific L-shaped NQO1-mediated redox modulators for the treatment of non-small cell lung cancer. Eur J Med Chem 2017; 138:616-629. [DOI: 10.1016/j.ejmech.2017.06.028] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2017] [Revised: 06/10/2017] [Accepted: 06/14/2017] [Indexed: 02/03/2023]
|
34
|
López-Lira C, Alzate-Morales JH, Paulino M, Mella-Raipán J, Salas CO, Tapia RA, Soto-Delgado J. Combined molecular modelling and 3D-QSAR study for understanding the inhibition of NQO1 by heterocyclic quinone derivatives. Chem Biol Drug Des 2017. [PMID: 28643389 DOI: 10.1111/cbdd.13051] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
A combination of three-dimensional quantitative structure-activity relationship (3D-QSAR), and molecular modelling methods were used to understand the potent inhibitory NAD(P)H:quinone oxidoreductase 1 (NQO1) activity of a set of 52 heterocyclic quinones. Molecular docking results indicated that some favourable interactions of key amino acid residues at the binding site of NQO1 with these quinones would be responsible for an improvement of the NQO1 activity of these compounds. The main interactions involved are hydrogen bond of the amino group of residue Tyr128, π-stacking interactions with Phe106 and Phe178, and electrostatic interactions with flavin adenine dinucleotide (FADH) cofactor. Three models were prepared by 3D-QSAR analysis. The models derived from Model I and Model III, shown leave-one-out cross-validation correlation coefficients (q2LOO ) of .75 and .73 as well as conventional correlation coefficients (R2 ) of .93 and .95, respectively. In addition, the external predictive abilities of these models were evaluated using a test set, producing the predicted correlation coefficients (r2pred ) of .76 and .74, respectively. The good concordance between the docking results and 3D-QSAR contour maps provides helpful information about a rational modification of new molecules based in quinone scaffold, in order to design more potent NQO1 inhibitors, which would exhibit highly potent antitumor activity.
Collapse
Affiliation(s)
- Claudia López-Lira
- Departamento de Química Orgánica, Facultad de Química, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Jans H Alzate-Morales
- Facultad de Ingeniería, Centro de Bioinformática y Simulación Molecular, Universidad de Talca, Talca, Chile
| | - Margot Paulino
- Facultad de Química, Centro de Bioinformática Estructural-DETEMA, Universidad de la República, Montevideo, Uruguay
| | - Jaime Mella-Raipán
- Facultad de Ciencias, Instituto de Química y Bioquímica, Universidad de Valparaíso, Valparaíso, Casilla, Chile
| | - Cristian O Salas
- Departamento de Química Orgánica, Facultad de Química, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Ricardo A Tapia
- Departamento de Química Orgánica, Facultad de Química, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Jorge Soto-Delgado
- Departamento de Ciencias Químicas, Facultad de Ciencias Exactas, Universidad Andrés Bello, Viña del Mar, Chile
| |
Collapse
|
35
|
Shen Z, Prasai B, Nakamura Y, Kobayashi H, Jackson MS, McCarley RL. A Near-Infrared, Wavelength-Shiftable, Turn-on Fluorescent Probe for the Detection and Imaging of Cancer Tumor Cells. ACS Chem Biol 2017; 12:1121-1132. [PMID: 28240865 DOI: 10.1021/acschembio.6b01094] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Fast, selective, and noninvasive reporting of intracellular cancer-associated events and species will lead to a better understanding of tumorigenesis at the molecular level and development of precision medicine approaches in oncology. Overexpressed reductase presence in solid tumor cells is key to cancer progression and protection of those diseased cells from the oxidative effects of therapeutics meant to kill them. Human NAD(P)H:quinone oxidoreductase isozyme I (hNQO1), a cytoprotective 2-electron-specific reductase found at unusually high activity levels in cancer cells of multiple origins, has attracted significant attention due to its major role in metastatic pathways and its link to low survival rates in patients, as well as its ability to effectively activate quinone-based, anticancer drugs. Accurate assessment of hNQO1 activities in living tumor models and ready differentiation of metastases from healthy tissue by fluorescent light-based protocols requires creation of hNQO1-responsive, near-infrared probes that offer deep tissue penetration and low background fluorescence. Herein, we disclose a quinone-trigger-based, near-infrared probe whose fluorescence is effectively turned on several hundred-fold through highly selective reduction of the quinone trigger group by hNQO1, with unprecedented, catalytically efficient formation of a fluorescent reporter. hNQO1 activity-specific production of a fluorescence signal in two-dimensional cultures of respiring human cancer cells that harbor the reductase enzyme allows for their quick (30 min) high-integrity recognition. The characteristics of the near-infrared probe make possible the imaging of clinically relevant three-dimensional colorectal tumor models possessing spatially heterogeneous hNQO1 activities and provide for fluorescence-assisted identification of submillimeter dimension metastases in a preclinical mouse model of human ovarian serous adenocarcinoma.
Collapse
Affiliation(s)
- Zhenhua Shen
- Department
of Chemistry, Louisiana State University, Baton Rouge, Louisiana 70803-1804, United States
| | - Bijeta Prasai
- Department
of Chemistry, Louisiana State University, Baton Rouge, Louisiana 70803-1804, United States
| | - Yuko Nakamura
- Molecular
Imaging Program, Center for Cancer Research, National Cancer Institute, United States National Institutes of Health, Bethesda, Maryland 20892-1088, United States
| | - Hisataka Kobayashi
- Molecular
Imaging Program, Center for Cancer Research, National Cancer Institute, United States National Institutes of Health, Bethesda, Maryland 20892-1088, United States
| | - Milcah S. Jackson
- Department
of Chemistry, Louisiana State University, Baton Rouge, Louisiana 70803-1804, United States
| | - Robin L. McCarley
- Department
of Chemistry, Louisiana State University, Baton Rouge, Louisiana 70803-1804, United States
| |
Collapse
|
36
|
Best QA, Prasai B, Rouillere A, Johnson AE, McCarley RL. Efficacious fluorescence turn-on probe for high-contrast imaging of human cells overexpressing quinone reductase activity. Chem Commun (Camb) 2017; 53:783-786. [DOI: 10.1039/c6cc08306d] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A turn-on substrate probe is activated by an oxidoreductase, offering fluorescence images of cancer cells with unprecedented positive signal-to-negative background ratios.
Collapse
Affiliation(s)
- Quinn A. Best
- Department of Chemistry
- Louisiana State University
- Baton Rouge
- USA
| | - Bijeta Prasai
- Department of Chemistry
- Louisiana State University
- Baton Rouge
- USA
| | | | | | | |
Collapse
|
37
|
Gayam SR, Venkatesan P, Sung YM, Sung SY, Hu SH, Hsu HY, Wu SP. An NAD(P)H:quinone oxidoreductase 1 (NQO1) enzyme responsive nanocarrier based on mesoporous silica nanoparticles for tumor targeted drug delivery in vitro and in vivo. NANOSCALE 2016; 8:12307-17. [PMID: 27271875 DOI: 10.1039/c6nr03525f] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
UNLABELLED The synthesis and characterization of an NAD(P)H quinone oxidoreductase 1 (NQO1) enzyme responsive nanocarrier based on mesoporous silica nanoparticles (MSNPs) for on-command delivery applications has been described in this paper. Gatekeeping of MSNPs is achieved by the integration of mechanically interlocked rotaxane nanovalves on the surface of MSNPs. The rotaxane nanovalve system is composed of a linear stalk anchoring on the surface of MSNPs, an α-cyclodextrin ring that encircles it and locks the payload "cargo" molecules in the mesopores, and a benzoquinone stopper incorporated at the end of the stalk. The gate opening and controlled release of the cargo are triggered by cleavage of the benzoquinone stopper using an endogenous NQO1 enzyme. In addition to having efficient drug loading and controlled release mechanisms, this smart biocompatible carrier system showed obvious uptake and consequent release of the drug in tumor cells, could selectively induce the tumor cell death and enhance the capability of inhibition of tumor growth in vivo. The controlled drug delivery system demonstrated its use as a potential theranostic material.
Collapse
Affiliation(s)
- Srivardhan Reddy Gayam
- National Chiao Tung University, Department of Applied Chemistry, Science Building 2, 1001 Ta Hsueh Road, Hsinchu, 300 Taiwan.
| | - Parthiban Venkatesan
- National Chiao Tung University, Department of Applied Chemistry, Science Building 2, 1001 Ta Hsueh Road, Hsinchu, 300 Taiwan.
| | - Yi-Ming Sung
- National Chiao Tung University, Department of Applied Chemistry, Science Building 2, 1001 Ta Hsueh Road, Hsinchu, 300 Taiwan.
| | - Shuo-Yuan Sung
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Shang-Hsiu Hu
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Hsin-Yun Hsu
- National Chiao Tung University, Department of Applied Chemistry, Science Building 2, 1001 Ta Hsueh Road, Hsinchu, 300 Taiwan.
| | - Shu-Pao Wu
- National Chiao Tung University, Department of Applied Chemistry, Science Building 2, 1001 Ta Hsueh Road, Hsinchu, 300 Taiwan.
| |
Collapse
|
38
|
Shin WS, Lee MG, Verwilst P, Lee JH, Chi SG, Kim JS. Mitochondria-targeted aggregation induced emission theranostics: crucial importance of in situ activation. Chem Sci 2016; 7:6050-6059. [PMID: 30034745 PMCID: PMC6022148 DOI: 10.1039/c6sc02236g] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 06/07/2016] [Indexed: 01/05/2023] Open
Abstract
A mitochondria targeted AIE fluorophore was further decorated with an NQO1 cleavable masking unit and showed selective targeting to and activation in cancer cells resulting in bright AIE fluorescence and apoptosis triggered by mitochondrial dysfunction.
Tissue selective targeting and specific suborganellular localization combined with an efficient pathology associated enzymatic activation of drugs in drug delivery systems may exhibit a clear advantage over conventional cancer treatment. Here, a mitochondria targeted aggregation induced emission (AIE) fluorophore further conjugated with an NAD(P)H:quinone oxidoreductase-1 (NQO1) cleavable masking unit showed preferential uptake in cancer cells and was selectively activated, resulting in bright AIE fluorescence and apoptosis via the caspase pathway, triggered by mitochondrial dysfunction. In vivo experimental data further support the conclusions from in vitro experiments, clearly showing the dependence of the therapy's success on both the suborganelle localization and specific in situ activation. And the site specific and enzyme dependent activation and aggregation was further supported by in vivo and ex vivo imaging. As a whole, the data comprised in this work represent a strong argument for the further development of this type of novel anticancer drugs.
Collapse
Affiliation(s)
- Weon Sup Shin
- Department of Chemistry , Korea University , Seoul , 136-701 , Korea .
| | - Min-Goo Lee
- School of Life Sciences and Biotechnology , Korea University , Seoul , 136-701 , Korea .
| | - Peter Verwilst
- Department of Chemistry , Korea University , Seoul , 136-701 , Korea .
| | - Joung Hae Lee
- Korea Research Institute of Standards and Science , Daejeon 305-600 , Korea
| | - Sung-Gil Chi
- School of Life Sciences and Biotechnology , Korea University , Seoul , 136-701 , Korea .
| | - Jong Seung Kim
- Department of Chemistry , Korea University , Seoul , 136-701 , Korea .
| |
Collapse
|
39
|
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: 7.1] [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
|
40
|
Li X, Bian J, Wang N, Qian X, Gu J, Mu T, Fan J, Yang X, Li S, Yang T, Sun H, You Q, Zhang X. Novel naphtho[2,1-d]oxazole-4,5-diones as NQO1 substrates with improved aqueous solubility: Design, synthesis, and in vivo antitumor evaluation. Bioorg Med Chem 2016; 24:1006-13. [DOI: 10.1016/j.bmc.2016.01.024] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Revised: 01/13/2016] [Accepted: 01/14/2016] [Indexed: 11/15/2022]
|
41
|
Best QA, Johnson AE, Prasai B, Rouillere A, McCarley RL. Environmentally Robust Rhodamine Reporters for Probe-based Cellular Detection of the Cancer-linked Oxidoreductase hNQO1. ACS Chem Biol 2016; 11:231-40. [PMID: 26555574 DOI: 10.1021/acschembio.5b00792] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
We successfully synthesized a fluorescent probe capable of detecting the cancer-associated NAD(P)H quinoneoxidoreductase isozyme-1 within human cells, based on results from an investigation of the stability of various rhodamines and seminaphthorhodamines toward the biological reductant NADH, present at ∼100-200 μM within cells. While rhodamines are generally known for their chemical stability, we observe that NADH causes significant and sometimes rapid modification of numerous rhodamine analogues, including those oftentimes used in imaging applications. Results from mechanistic studies lead us to rule out a radical-based reduction pathway, suggesting rhodamine reduction by NADH proceeds by a hydride transfer process to yield the reduced leuco form of the rhodamine and oxidized NAD(+). A relationship between the structural features of the rhodamines and their reactivity with NADH is observed. Rhodamines with increased alkylation on the N3- and N6-nitrogens, as well as the xanthene core, react the least with NADH; whereas, nonalkylated variants or analogues with electron-withdrawing substituents have the fastest rates of reaction. These outcomes allowed us to judiciously construct a seminaphthorhodamine-based, turn-on fluorescent probe that is capable of selectively detecting the cancer-associated, NADH-dependent enzyme NAD(P)H quinoneoxidoreductase isozyme-1 in human cancer cells, without the issue of NADH-induced deactivation of the seminaphthorhodamine reporter.
Collapse
Affiliation(s)
- Quinn A. Best
- Department of Chemistry, Louisiana State University, Baton Rouge, Louisiana 70803-1804, United States
| | - Amanda E. Johnson
- Department of Chemistry, Louisiana State University, Baton Rouge, Louisiana 70803-1804, United States
| | - Bijeta Prasai
- Department of Chemistry, Louisiana State University, Baton Rouge, Louisiana 70803-1804, United States
| | - Alexandra Rouillere
- Department of Chemistry, Louisiana State University, Baton Rouge, Louisiana 70803-1804, United States
| | - Robin L. McCarley
- Department of Chemistry, Louisiana State University, Baton Rouge, Louisiana 70803-1804, United States
| |
Collapse
|
42
|
Sung YM, Gayam SR, Hsieh PY, Hsu HY, Diau EWG, Wu SP. Quinone-Modified Mn-Doped ZnS Quantum Dots for Room-Temperature Phosphorescence Sensing of Human Cancer Cells That Overexpress NQO1. ACS APPLIED MATERIALS & INTERFACES 2015; 7:25961-25969. [PMID: 26540617 DOI: 10.1021/acsami.5b09244] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
UNLABELLED Early detection of cancer cells in a rapid and sensitive approach is one of the great challenges in modern clinical cancer care. This study has demonstrated the first example of a rapid, selective, and sensitive phosphorescence probe based on phosphorescence energy transfer (PET) for cancer-associated human NAD(P)H quinone oxidoreductase isozyme 1 (NQO1). An efficient room-temperature phosphorescence NQO1 probe was constructed by using Mn-doped ZnS quantum dots (Mn:ZnS QDs) as donors and trimethylquinone propionic acids as acceptors. Phosphorescence quenching of Mn:ZnS QDs from the Mn:ZnS QDs to a covalently bonded quinone was achieved through PET. Phosphorescence of Mn:ZnS QDs was turned on by the rapid reduction-initiated removal of the quinone quencher by NQO1. This probe shows low cellular toxicity and can rapidly distinguish between NQO1-expressing and -nonexpressing cancer cell lines through phosphorescence imaging.
Collapse
Affiliation(s)
- Yi-Ming Sung
- Department of Applied Chemistry and ‡Center for Interdisciplinary Science (CIS), National Chiao Tung University , Hsinchu 300, Taiwan
| | - Srivardhan Reddy Gayam
- Department of Applied Chemistry and ‡Center for Interdisciplinary Science (CIS), National Chiao Tung University , Hsinchu 300, Taiwan
| | - Pei-Ying Hsieh
- Department of Applied Chemistry and ‡Center for Interdisciplinary Science (CIS), National Chiao Tung University , Hsinchu 300, Taiwan
| | - Hsin-Yun Hsu
- Department of Applied Chemistry and ‡Center for Interdisciplinary Science (CIS), National Chiao Tung University , Hsinchu 300, Taiwan
| | - Eric Wei-Guang Diau
- Department of Applied Chemistry and ‡Center for Interdisciplinary Science (CIS), National Chiao Tung University , Hsinchu 300, Taiwan
| | - Shu-Pao Wu
- Department of Applied Chemistry and ‡Center for Interdisciplinary Science (CIS), National Chiao Tung University , Hsinchu 300, Taiwan
| |
Collapse
|
43
|
Bae J, Maurya A, Shariat-Madar Z, Murthy SN, Jo S. Novel Redox-Responsive Amphiphilic Copolymer Micelles for Drug Delivery: Synthesis and Characterization. AAPS JOURNAL 2015; 17:1357-68. [PMID: 26122497 DOI: 10.1208/s12248-015-9800-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Accepted: 06/13/2015] [Indexed: 11/30/2022]
Abstract
A novel redox-responsive amphiphilic polymer was synthesized with bioreductive trimethyl-locked quinone propionic acid for a potential triggered drug delivery application. The aim of this study was to synthesize and characterize the redox-responsive amphiphilic block copolymer micelles containing pendant bioreductive quinone propionic acid (QPA) switches. The redox-responsive hydrophobic block (polyQPA), synthesized from QPA-serinol and adipoyl chloride, was end-capped with methoxy poly(ethylene glycol) of molecular weight 750 (mPEG750) to achieve a redox-responsive amphiphilic block copolymer, polyQPA-mPEG750. PolyQPA-mPEG750 was able to self-assemble as micelles to show a critical micelle concentration (CMC) of 0.039% w/v (0.39 mg/ml, 0.107 mM) determined by a dye solubilization method using 1,6-diphenyl-1,3,5-hexatriene (DPH) in phosphate-buffered saline (PBS). The mean diameter of polymeric micelles was found to be 27.50 nm (PI = 0.064) by dynamic light scattering. Furthermore, redox-triggered destabilization of the polymeric micelles was confirmed by (1)H-NMR spectroscopy and particle size measurements in a simulated redox state. PolyQPA-mPEG750 underwent triggered reduction to shed pendant redox-responsive QPA groups and its polymeric micelles were swollen to be dissembled in the presence of a reducing agent, thereby enabling the release of loaded model drug, paclitaxel. The redox-responsive polyQPA-mPEG750 polymer micelles would be useful as a drug delivery system allowing triggered drug release in an altered redox state such as tumor microenvironments with an altered redox potential and/or redox enzyme upregulation.
Collapse
Affiliation(s)
- Jungeun Bae
- Department of Pharmaceutics and Drug Delivery, School of Pharmacy, The University of Mississippi, University, Mississippi, 38677, USA
| | - Abhijeet Maurya
- Department of Pharmaceutics and Drug Delivery, School of Pharmacy, The University of Mississippi, University, Mississippi, 38677, USA
| | - Zia Shariat-Madar
- Department of BioMolecular Sciences, School of Pharmacy, The University of Mississippi, University, Mississippi, 38677, USA
| | - S Narasimha Murthy
- Department of Pharmaceutics and Drug Delivery, School of Pharmacy, The University of Mississippi, University, Mississippi, 38677, USA.,Institute for Drug Delivery and Biomedical Research (IDBR), Bangalore, India
| | - Seongbong Jo
- Department of Pharmaceutics and Drug Delivery, School of Pharmacy, The University of Mississippi, University, Mississippi, 38677, USA. .,Research Institute of Pharmaceutical Sciences, School of Pharmacy, The University of Mississippi, University, Mississippi, 38677, USA.
| |
Collapse
|
44
|
Prasai B, Silvers WC, McCarley RL. Oxidoreductase-Facilitated Visualization and Detection of Human Cancer Cells. Anal Chem 2015; 87:6411-8. [PMID: 26005900 DOI: 10.1021/acs.analchem.5b01615] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
UNLABELLED Achieving highly selective and sensitive detection/visualization of intracellular biological events through the use of cell-penetrable, bioanalyte-activatable, turn-on probes is dependent on the presence of specific event-linked cellular biomarkers, if and only if there exist activatable probes that appropriately respond to the biomarker analyte. Here is described the evaluation of, and use in cellular imaging studies, a previously undisclosed naphthalimide probe QMeNN, whose fluorescence is deactivated by photoinduced electron transfer (PeT) quenching that results from the presence of a covalently linked biomarker-specific quinone trigger group. Highly selective and rapid activation of the quinone group by the human cancer tumor-linked NAD(P)H quinone oxido-reductase isozyme 1 (hNQO1) results in fast trigger group removal to yield a highly fluorescent green-energy-range reporter that possesses a high molar absorptivity; there is a 136-fold increase in brightness for the enzymatically produced reporter versus probe precursor, a value 4 times greater than previously reported for the hNQO1 analyte. The novel probe is taken up and activated rapidly within only hNQO1-positive human cancer cells; addition of an hNQO1 inhibitor prevents the selective activation of the probe. Comparison of cytosolic fluorescence intensity in positive cells versus background in negative cells yields a quantitative metric (positive-to-negative ratio, PNR) for judging hNQO1 activity. We show it is possible to determine hNQO1 presence in previously studied colorectal cancer cells and the unexplored ovarian cancer cell line NIH:OVCAR-3, with respective PNR values of 926 and 34 being obtained. Even with 10 min probe incubation, ready discrimination of positive cells from negative cells is achieved. Cell viability is unaffected by probe presence, thereby highlighting the practicality of probe use in live-cell imaging applications.
Collapse
Affiliation(s)
- Bijeta Prasai
- Department of Chemistry, Louisiana State University, Baton Rouge, Louisiana 70803-1804, United States
| | - William C Silvers
- Department of Chemistry, Louisiana State University, Baton Rouge, Louisiana 70803-1804, United States
| | - Robin L McCarley
- Department of Chemistry, Louisiana State University, Baton Rouge, Louisiana 70803-1804, United States
| |
Collapse
|
45
|
Ji C, Miller MJ. Siderophore-fluoroquinolone conjugates containing potential reduction-triggered linkers for drug release: synthesis and antibacterial activity. Biometals 2015; 28:541-51. [PMID: 25663417 PMCID: PMC5808879 DOI: 10.1007/s10534-015-9830-3] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2014] [Accepted: 02/01/2015] [Indexed: 11/25/2022]
Abstract
Syntheses of two Siderophore-fluoroquinolone conjugates with a potential reduction triggered linker for drug release are described. The "trimethyl lock" based linker incorporated in the conjugates was designed to be activated by taking advantage of the reductive pathway of bacterial iron metabolism. Electrochemical and LC-MS studies indicated that the linker is thermodynamically reducible by common biological reductants and the expected lactonization proceeds rapidly with concomitant release of the drug. Antibacterial activity assays revealed that conjugates with the reduction-triggered linker were more potent than their counterparts with a stable linker, which suggests that drug release occurs inside bacterial cells.
Collapse
Affiliation(s)
- Cheng Ji
- Department of Chemistry and Biochemistry, 251 Nieuwland Science Hall, University of Notre Dame, Notre Dame, Indiana, 46556, United States
| | - Marvin J. Miller
- Department of Chemistry and Biochemistry, 251 Nieuwland Science Hall, University of Notre Dame, Notre Dame, Indiana, 46556, United States
| |
Collapse
|
46
|
Synthesis and evaluation of (±)-dunnione and its ortho-quinone analogues as substrates for NAD(P)H:quinone oxidoreductase 1 (NQO1). Bioorg Med Chem Lett 2015; 25:1244-8. [DOI: 10.1016/j.bmcl.2015.01.057] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Revised: 01/17/2015] [Accepted: 01/23/2015] [Indexed: 01/10/2023]
|
47
|
2-Substituted 3-methylnaphtho[1,2-b]furan-4,5-diones as novel L-shaped ortho-quinone substrates for NAD(P)H:quinone oxidoreductase (NQO1). Eur J Med Chem 2014; 82:56-67. [DOI: 10.1016/j.ejmech.2014.05.041] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Revised: 05/12/2014] [Accepted: 05/13/2014] [Indexed: 11/18/2022]
|
48
|
Hettiarachchi SU, Prasai B, McCarley RL. Detection and cellular imaging of human cancer enzyme using a turn-on, wavelength-shiftable, self-immolative profluorophore. J Am Chem Soc 2014; 136:7575-8. [PMID: 24813575 PMCID: PMC4046754 DOI: 10.1021/ja5030707] [Citation(s) in RCA: 133] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
![]()
A frontier
area in the development of activatable (turn-on) fluorescence-based
probes is that concerned with rapid and selective stimulus triggering
of probe activation so as to allow for biomarker identification and
cellular imaging. The work here is concerned with a cloaked fluorophore
composed of a reporter whose fluorescence is efficiently quenched
by it being bound to an activatable trigger group through a novel
self-immolative linker. Highly selective and rapid activation of the
trigger group is achieved by chemical and enzymatic means that result
in activated trigger group detachment from the self-immolative linker,
with the latter subsequently cleaved from the reporter autonomously,
thereby unmasking intense, red-shifted fluorescence emission. To achieve
this success, we used a trimethyl-locked quinone propionic acid trigger
group and an N-methyl-p-aminobenzyl
alcohol self-immolative linker attached to the reporter. Delineated
here are the synthesis and characterization of this cloaked fluorophore
and the evaluation of its triggered turning on in the presence of
an up-regulated enzyme in human cancer cells, NAD(P)H:quinone oxidoreductase-1
(NQO1, DT-diaphorase, EC 1.6.99.2).
Collapse
Affiliation(s)
- Suraj U Hettiarachchi
- Department of Chemistry, Louisiana State University , Baton Rouge, Louisiana 70803-1804, United States
| | | | | |
Collapse
|
49
|
Zhou W, Leippe D, Duellman S, Sobol M, Vidugiriene J, O'Brien M, Shultz JW, Kimball JJ, DiBernardo C, Moothart L, Bernad L, Cali J, Klaubert DH, Meisenheimer P. Self-immolative bioluminogenic quinone luciferins for NAD(P)H assays and reducing capacity-based cell viability assays. Chembiochem 2014; 15:670-5. [PMID: 24591148 DOI: 10.1002/cbic.201300744] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Indexed: 12/13/2022]
Abstract
Highly sensitive self-cleavable trimethyl lock quinone-luciferin substrates for diaphorase were designed and synthesized to measure NAD(P)H in biological samples and monitor viable cells via NAD(P)H-dependent cellular oxidoreductase enzymes and their NAD(P)H cofactors.
Collapse
Affiliation(s)
- Wenhui Zhou
- Research and Development, Promega Biosciences, Inc. 277 Granada Drive, San Luis Obispo, CA 93401 (USA).
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
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: 4.5] [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
|