1
|
Uddin MJ, Lo JHJ, Gupta MK, Werfel TA, Asaduzzaman A, Oltman CG, Gbur EF, Mohyuddin MT, Nazmin F, Rahman MS, Jashim A, Crews BC, Kingsley PJ, Klendworth JE, Marnett LJ, Duvall CL, Cook RS. Polymeric Nanoparticles Enable Targeted Visualization of Drug Delivery in Breast Cancer. Mol Pharm 2025; 22:2392-2401. [PMID: 40257460 PMCID: PMC12056698 DOI: 10.1021/acs.molpharmaceut.4c00695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 03/31/2025] [Accepted: 04/03/2025] [Indexed: 04/22/2025]
Abstract
We report the coencapsulation of fluorocoxib Q (FQ) and chemocoxib A (CA) in micellar nanoparticles (FQ-CA-NPs) of a new PPS135-b-POEGA17 diblock polymer, which exhibited a hydrodynamic diameter of 109.2 ± 4.1 nm and a zeta potential (ζ) of -1.59 ± 0.3 mV. The uptake of FQ-CA-NPs by 4T1 mouse mammary cancer cells and intracellular cargo release were assessed by fluorescence microscopy that resulted in increased fluorescence in 4T1 cells compared to cells pretreated with celecoxib. The viability of primary human mammary epithelial cells (HMECs) or 4T1 mouse mammary carcinoma cells treated with FQ-CA-NPs were assessed, which showed decreased growth of 4T1 breast cancer cells but showed no effect on the growth of primary human mammary epithelial cells (HMECs). Intravenous dosing of FQ-CA-NPs in mice enabled ROS-induced cargo (FQ and CA) release and fluorescence activation of FQ and resulted in increased fluorescence in breast tumors compared to the tumors of animals pretreated with tempol or celecoxib, and minimum fluorescence was detected in the tumors of animals treated with nothing or empty-NPs. In addition, tumor tissues from treated animals were analyzed ex vivo by liquid chromatography-mass spectrometry (LC-MS)/MS, and identified increased levels of cargo delivery and retention in the tumor compared to tempol- or celecoxib-pretreated animal tumors. These in vivo and ex vivo results confirmed the targeted delivery of loaded NPs followed by ROS-mediated cargo release and fluorescence activation for targeted visualization of drug delivery in breast tumors and CA-induced therapeutic effect in an in vivo tumor growth inhibition assay and an ex vivo hematoxylin and eosin (H&E) staining of tumor tissues. Thus, coencapsulation of FQ and CA into polymeric micellar nanoparticles (FQ-CA-NPs) enabled their ROS-sensitive release followed by fluorescence activation and COX-2-dependent tumor targeting and retention in the visualization of CA delivery in solid breast tumors.
Collapse
Affiliation(s)
- Md. Jashim Uddin
- Department
of Biochemistry, Vanderbilt University School
of Medicine, Nashville, Tennessee 37232, United States
| | - Justin Han-Je Lo
- Department
of Medicine, Vanderbilt University Medical
Center, Nashville, Tennessee 37232, United States
| | - Mukesh K. Gupta
- Department
of Biomedical Engineering, Vanderbilt University
School of Engineering, Nashville, Tennessee 37232, United States
| | - Thomas A. Werfel
- Department
of Biomedical Engineering, Vanderbilt University
School of Engineering, Nashville, Tennessee 37232, United States
| | - Abu Asaduzzaman
- Departments
of Electrical and Computer Engineering, Wichita State University School of Engineering, Wichita, Kansas 67260, United States
| | - Connor G. Oltman
- Department
of Biochemistry, Vanderbilt University School
of Medicine, Nashville, Tennessee 37232, United States
| | - Eva F. Gbur
- Department
of Biomedical Engineering, Vanderbilt University
School of Engineering, Nashville, Tennessee 37232, United States
| | - Mohammed T. Mohyuddin
- Department
of Biomedical Engineering, Vanderbilt University
School of Engineering, Nashville, Tennessee 37232, United States
| | - Farhana Nazmin
- Department
of Biochemistry, Vanderbilt University School
of Medicine, Nashville, Tennessee 37232, United States
| | - Md. Saidur Rahman
- Department
of Medicine, Vanderbilt University Medical
Center, Nashville, Tennessee 37232, United States
| | - Ahan Jashim
- Department
of Biochemistry, Vanderbilt University School
of Medicine, Nashville, Tennessee 37232, United States
| | - Brenda C. Crews
- Department
of Biochemistry, Vanderbilt University School
of Medicine, Nashville, Tennessee 37232, United States
| | - Philip J. Kingsley
- Department
of Biochemistry, Vanderbilt University School
of Medicine, Nashville, Tennessee 37232, United States
| | - Jamie E. Klendworth
- Department
of Biochemistry, Vanderbilt University School
of Medicine, Nashville, Tennessee 37232, United States
| | - Lawrence J. Marnett
- Departments
of Biochemistry, Chemistry and Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Craig L. Duvall
- Department
of Biomedical Engineering, Vanderbilt University
School of Engineering, Nashville, Tennessee 37232, United States
| | - Rebecca S. Cook
- Department
of Biomedical Engineering, Vanderbilt University
School of Engineering, Nashville, Tennessee 37232, United States
| |
Collapse
|
2
|
Uzuegbunam BC, Rummel C, Librizzi D, Culmsee C, Hooshyar Yousefi B. Radiotracers for Imaging of Inflammatory Biomarkers TSPO and COX-2 in the Brain and in the Periphery. Int J Mol Sci 2023; 24:17419. [PMID: 38139248 PMCID: PMC10743508 DOI: 10.3390/ijms242417419] [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: 10/24/2023] [Revised: 12/01/2023] [Accepted: 12/04/2023] [Indexed: 12/24/2023] Open
Abstract
Inflammation involves the activation of innate immune cells and is believed to play an important role in the development and progression of both infectious and non-infectious diseases such as neurodegeneration, autoimmune diseases, pulmonary and cancer. Inflammation in the brain is marked by the upregulation of translocator protein (TSPO) in microglia. High TSPO levels are also found, for example, in macrophages in cases of rheumatoid arthritis and in malignant tumor cells compared to their relatively low physiological expression. The same applies for cyclooxgenase-2 (COX-2), which is constitutively expressed in the kidney, brain, thymus and gastrointestinal tract, but induced in microglia, macrophages and synoviocytes during inflammation. This puts TSPO and COX-2 in the spotlight as important targets for the diagnosis of inflammation. Imaging modalities, such as positron emission tomography and single-photon emission tomography, can be used to localize inflammatory processes and to track their progression over time. They could also enable the monitoring of the efficacy of therapy and predict its outcome. This review focuses on the current development of PET and SPECT tracers, not only for the detection of neuroinflammation, but also for emerging diagnostic measures in infectious and other non-infectious diseases such as rheumatic arthritis, cancer, cardiac inflammation and in lung diseases.
Collapse
Affiliation(s)
| | - Christoph Rummel
- Institute of Veterinary Physiology and Biochemistry, Justus Liebig University Giessen, 35392 Gießen, Germany;
- Center for Mind Brain and Behavior, Universities Giessen and Marburg, 35043 Marburg, Germany;
| | - Damiano Librizzi
- Department of Nuclear Medicine, Philipps University of Marburg, 35043 Marburg, Germany;
| | - Carsten Culmsee
- Center for Mind Brain and Behavior, Universities Giessen and Marburg, 35043 Marburg, Germany;
- Institute of Pharmacology and Clinical Pharmacy, Philipps University of Marburg, 35037 Marburg, Germany
| | | |
Collapse
|
3
|
Chen L, Lyu Y, Zhang X, Zheng L, Li Q, Ding D, Chen F, Liu Y, Li W, Zhang Y, Huang Q, Wang Z, Xie T, Zhang Q, Sima Y, Li K, Xu S, Ren T, Xiong M, Wu Y, Song J, Yuan L, Yang H, Zhang XB, Tan W. Molecular imaging: design mechanism and bioapplications. Sci China Chem 2023. [DOI: 10.1007/s11426-022-1461-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/09/2023]
|
4
|
Uddin MJ, Lo JHJ, Oltman CG, Crews BC, Huda T, Liu J, Kingsley PJ, Lin S, Milad M, Aleem AM, Asaduzzaman A, McIntyre JO, Duvall CL, Marnett LJ. Discovery of a Redox-Activatable Chemical Probe for Detection of Cyclooxygenase-2 in Cells and Animals. ACS Chem Biol 2022; 17:1714-1722. [PMID: 35786843 PMCID: PMC10464600 DOI: 10.1021/acschembio.1c00961] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Cyclooxygenase-2 (COX-2) expression is up-regulated in inflammatory tissues and many premalignant and malignant tumors. Assessment of COX-2 protein in vivo, therefore, promises to be a powerful strategy to distinguish pathologic cells from normal cells in a complex disease setting. Herein, we report the first redox-activatable COX-2 probe, fluorocoxib Q (FQ), for in vivo molecular imaging of pathogenesis. FQ inhibits COX-2 selectively in purified enzyme and cell-based assays. FQ exhibits extremely low fluorescence and displays time- and concentration-dependent fluorescence enhancement upon exposure to a redox environment. FQ enters the cells freely and binds to the COX-2 enzyme. FQ exhibits high circulation half-life and metabolic stability sufficient for target site accumulation and demonstrates COX-2-targeted uptake and retention in cancer cells and pathologic tissues. Once taken up, it undergoes redox-mediated transformation into a fluorescent compound fluorocoxib Q-H that results in high signal-to-noise contrast and differentiates pathologic tissues from non-pathologic tissues for real-time in vivo imaging.
Collapse
Affiliation(s)
- Md. Jashim Uddin
- Department of Biochemistry, Vanderbilt University, Nashville, Tennessee 37232 USA
| | - Justin Han-Je Lo
- Department of Biochemistry, Vanderbilt University, Nashville, Tennessee 37232 USA
- Division of Hematology and Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232 USA
| | - Connor G. Oltman
- Department of Biochemistry, Vanderbilt University, Nashville, Tennessee 37232 USA
| | - Brenda C. Crews
- Department of Biochemistry, Vanderbilt University, Nashville, Tennessee 37232 USA
| | - Tamanna Huda
- Department of Biochemistry, Vanderbilt University, Nashville, Tennessee 37232 USA
| | - Justin Liu
- Department of Biochemistry, Vanderbilt University, Nashville, Tennessee 37232 USA
- Department of Neuroscience, Columbia University, New York City, New York, 10027 USA
| | - Philip J. Kingsley
- Department of Biochemistry, Vanderbilt University, Nashville, Tennessee 37232 USA
| | - Shuyang Lin
- Department of Biochemistry, Vanderbilt University, Nashville, Tennessee 37232 USA
| | - Mathew Milad
- Department of Biochemistry, Vanderbilt University, Nashville, Tennessee 37232 USA
| | - Ansari M. Aleem
- Department of Biochemistry, Vanderbilt University, Nashville, Tennessee 37232 USA
| | - Abu Asaduzzaman
- Department of Electrical Engineering and Computer Science, Wichita State University, Wichita, Kansas 67260 USA
| | - J. Oliver McIntyre
- Departments of Radiology and Radiological Sciences, and Pharmacology, Vanderbilt Institute of Imaging Science, Vanderbilt University School of Medicine, Nashville, Tennessee 37232 USA
| | - Craig L. Duvall
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37232 USA
| | - Lawrence J. Marnett
- Departments of Biochemistry, Chemistry, and Pharmacology, Vanderbilt Institute of Chemical Biology, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee 37232 USA
| |
Collapse
|
5
|
Litchfield M, Wuest M, Glubrecht D, Briard E, Auberson YP, McMullen TPW, Brindley DN, Wuest F. Positron Emission Tomography Imaging of Autotaxin in Thyroid and Breast Cancer Models Using [ 18F]PRIMATX. Mol Pharm 2021; 18:3352-3364. [PMID: 34319110 DOI: 10.1021/acs.molpharmaceut.1c00265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Autotaxin (ATX) is a secreted enzyme responsible for producing lysophosphatidic acid (LPA). The ATX/LPA signaling axis is typically activated in wound healing and tissue repair processes. The ATX/LPA axis is highjacked and upregulated in the progression and persistence of several chronic inflammatory diseases, including cancer. As ATX inhibitors are now progressing to clinical testing, innovative diagnostic tools such as positron emission tomography (PET) are needed to measure ATX expression in vivo accurately. The radiotracer, [18F]PRIMATX, was recently developed and tested for PET imaging of ATX in vivo in a murine melanoma model. The goal of the present work was to further validate [18F]PRIMATX as a PET imaging agent by analyzing its in vivo metabolic stability and suitability for PET imaging of ATX in models of human 8305C thyroid tumor and murine 4T1 breast cancer. [18F]PRIMATX displayed favorable metabolic stability in vivo (65% of intact radiotracer after 60 min p.i.) and provided sufficient tumor uptake profiles in both tumor models. Radiotracer uptake could be blocked by 8-12% in 8305C thyroid tumors in the presence of ATX inhibitor AE-32-NZ70 as determined by PET and ex vivo biodistribution analyses. [18F]PRIMATX also showed high brain uptake, which was reduced by 50% through the administration of ATX inhibitor AE-32-NZ70. [18F]PRIMATX is a suitable radiotracer for PET imaging of ATX in the brain and peripheral tumor tissues.
Collapse
Affiliation(s)
- Marcus Litchfield
- Department of Oncology, University of Alberta, 11560 University Avenue, Edmonton T6G 1Z2, Alberta, Canada.,Cancer Research Institute of Northern Alberta, University of Alberta, Edmonton T6G 2S2, Alberta, Canada
| | - Melinda Wuest
- Department of Oncology, University of Alberta, 11560 University Avenue, Edmonton T6G 1Z2, Alberta, Canada
| | - Daryl Glubrecht
- Department of Oncology, University of Alberta, 11560 University Avenue, Edmonton T6G 1Z2, Alberta, Canada
| | - Emmanuelle Briard
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research, Fabristrasse 2, Novartis Campus, Basel CH-4056, Switzerland
| | - Yves P Auberson
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research, Fabristrasse 2, Novartis Campus, Basel CH-4056, Switzerland
| | - Todd P W McMullen
- Department of Oncology, University of Alberta, 11560 University Avenue, Edmonton T6G 1Z2, Alberta, Canada.,Cancer Research Institute of Northern Alberta, University of Alberta, Edmonton T6G 2S2, Alberta, Canada
| | - David N Brindley
- Department of Biochemistry, University of Alberta, Edmonton T6G 1Z2, Alberta, Canada.,Cancer Research Institute of Northern Alberta, University of Alberta, Edmonton T6G 2S2, Alberta, Canada
| | - Frank Wuest
- Department of Oncology, University of Alberta, 11560 University Avenue, Edmonton T6G 1Z2, Alberta, Canada.,Cancer Research Institute of Northern Alberta, University of Alberta, Edmonton T6G 2S2, Alberta, Canada
| |
Collapse
|
6
|
Brandão SCS, Ramos JDOX, de Arruda GFA, Godoi ETAM, Carreira LCTF, Lopes RW, Grossman GB, de Souza Leão Lima R. Mapping COVID-19 functional sequelae: the perspective of nuclear medicine. AMERICAN JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING 2020; 10:319-333. [PMID: 33329934 PMCID: PMC7724276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 10/03/2020] [Indexed: 06/12/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 infection is capable of affecting several organs. Direct viral toxicity, pro-inflammatory and pro-thrombotic induction, endothelial damage, immune imbalance, and dysregulation of the renin-angiotensin-aldosterone system are the mechanisms underlying the viral potential of multiple organ damage. The impairment of four organs stands out among severe patients: lung, heart, kidney, and endothelium. The nuclear medicine field holds accurate and safe exam techniques, such as positron emission tomography-computed tomography and scintigraphy, that allow the anatomophysiological study of the majority of human organ systems. By choosing the most appropriate method and radiopharmaceutical, analyzing the presence of inflammation, fibrosis, changes in perfusion, and function of desired organs is possible. Therefore, its use in the monitoring of patients with coronavirus disease 2019 becomes relevant, especially for monitoring sequelae. In this review, we discuss the use of Nuclear Medicine in the detection, monitoring, and therapeutic evaluation of pulmonary and extrapulmonary sequelae by coronavirus disease 2019.
Collapse
Affiliation(s)
- Simone Cristina Soares Brandão
- Department of Internal Medicine, Divisions of Cardiology and Nuclear Imaging, Hospital das Clínicas, Federal University of Pernambuco (UFPE)Recife, PE, Brazil
| | | | | | | | | | | | - Gabriel Blacher Grossman
- Department of Nuclear Medicine, Hospital Moinhos de VentoPorto Alegre, RS, Brazil
- Cardionuclear ClinicPorto Alegre, RS, Brazil
| | - Ronaldo de Souza Leão Lima
- Department of Cardiology and Division of Nuclear Imaging, Federal University of Rio de Janeiro (UFRJ)Rio de Janeiro, RJ, Brazil
| |
Collapse
|
7
|
Kaur J, Bhardwaj A, Wuest F. In Cellulo Generation of Fluorescent Probes for Live-Cell Imaging of Cylooxygenase-2. Chemistry 2020; 27:3326-3337. [PMID: 32786126 DOI: 10.1002/chem.202003315] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 08/09/2020] [Indexed: 02/01/2023]
Abstract
Live-cell imaging with fluorescent probes is an essential tool in chemical biology to visualize the dynamics of biological processes in real-time. Intracellular disease biomarker imaging remains a formidable challenge due to the intrinsic limitations of conventional fluorescent probes and the complex nature of cells. This work reports the in cellulo assembly of a fluorescent probe to image cyclooxygenase-2 (COX-2). We developed celecoxib-azide derivative 14, possessing favorable biophysical properties and excellent COX-2 selectivity profile. In cellulo strain-promoted fluorogenic click chemistry of COX-2-engaged compound 14 with non/weakly-fluorescent compounds 11 and 17 formed fluorescent probes 15 and 18 for the detection of COX-2 in living cells. Competitive binding studies, biophysical, and comprehensive computational analyses were used to describe protein-ligand interactions. The reported new chemical toolbox enables precise visualization and tracking of COX-2 in live cells with superior sensitivity in the visible range.
Collapse
Affiliation(s)
- Jatinder Kaur
- Department of Oncology, University of Alberta, Edmonton, AB, Canada.,Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
| | - Atul Bhardwaj
- Department of Oncology, University of Alberta, Edmonton, AB, Canada.,Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
| | - Frank Wuest
- Department of Oncology, University of Alberta, Edmonton, AB, Canada.,Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada.,Department of Chemistry, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
8
|
Laube M, Gassner C, Neuber C, Wodtke R, Ullrich M, Haase-Kohn C, Löser R, Köckerling M, Kopka K, Kniess T, Hey-Hawkins E, Pietzsch J. Deuteration versus ethylation - strategies to improve the metabolic fate of an 18F-labeled celecoxib derivative. RSC Adv 2020; 10:38601-38611. [PMID: 35517533 PMCID: PMC9057277 DOI: 10.1039/d0ra04494f] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 10/11/2020] [Indexed: 12/14/2022] Open
Abstract
The inducible isoenzyme cyclooxygenase-2 (COX-2) is closely associated with chemo-/radioresistance and poor prognosis of solid tumors. Therefore, COX-2 represents an attractive target for functional characterization of tumors by positron emission tomography (PET). In this study, the celecoxib derivative 3-([18F]fluoromethyl)-1-[4-(methylsulfonyl)phenyl]-5-(p-tolyl)-1H-pyrazole ([18F]5a) was chosen as a lead compound having a reported high COX-2 inhibitory potency and a potentially low carbonic anhydrase binding tendency. The respective deuterated analog [D2,18F]5a and the fluoroethyl-substituted derivative [18F]5b were selected to study the influence of these modifications with respect to COX inhibition potency in vitro and metabolic stability of the radiolabeled tracers in vivo. COX-2 inhibitory potency was found to be influenced by elongation of the side chain but, as expected, not by deuteration. An automated radiosynthesis comprising 18F-fluorination and purification under comparable conditions provided the radiotracers [18F]5a,b and [D2,18F]5a in good radiochemical yields (RCY) and high radiochemical purity (RCP). Biodistribution and PET studies comparing all three compounds revealed bone accumulation of 18F-activity to be lowest for the ethyl derivative [18F]5b. However, the deuterated analog [D2,18F]5a turned out to be the most stable compound of the three derivatives studied here. Time-dependent degradation of [18F]5a,b and [D2,18F]5a after incubation in murine liver microsomes was in accordance with the data on metabolism in vivo. Furthermore, metabolites were identified based on UPLC-MS/MS. The aim of this study is to investigate the influence of deuteration and elongation on an 18F-labeled COX-2 inhibitor with focus on metabolic stability to develop suitable COX-2 targeting radiotracers.![]()
Collapse
Affiliation(s)
- Markus Laube
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research Bautzner Landstrasse 400 01328 Dresden Germany
| | - Cemena Gassner
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research Bautzner Landstrasse 400 01328 Dresden Germany .,Faculty of Chemistry and Food Chemistry, School of Science, Technische Universität Dresden Mommsenstrasse 4 D-01062 Dresden Germany
| | - Christin Neuber
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research Bautzner Landstrasse 400 01328 Dresden Germany
| | - Robert Wodtke
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research Bautzner Landstrasse 400 01328 Dresden Germany
| | - Martin Ullrich
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research Bautzner Landstrasse 400 01328 Dresden Germany
| | - Cathleen Haase-Kohn
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research Bautzner Landstrasse 400 01328 Dresden Germany
| | - Reik Löser
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research Bautzner Landstrasse 400 01328 Dresden Germany
| | - Martin Köckerling
- University of Rostock, Institute of Chemistry, Department of Inorganic Solid State Chemistry Albert-Einstein-Str. 3a D-18059 Rostock Germany
| | - Klaus Kopka
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research Bautzner Landstrasse 400 01328 Dresden Germany
| | - Torsten Kniess
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research Bautzner Landstrasse 400 01328 Dresden Germany
| | - Evamarie Hey-Hawkins
- Leipzig University, Faculty of Chemistry and Mineralogy, Institute of Inorganic Chemistry Johannisallee 29 D-04103 Leipzig Germany
| | - Jens Pietzsch
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research Bautzner Landstrasse 400 01328 Dresden Germany .,Faculty of Chemistry and Food Chemistry, School of Science, Technische Universität Dresden Mommsenstrasse 4 D-01062 Dresden Germany
| |
Collapse
|
9
|
Malerba P, Crews BC, Ghebreselasie K, Daniel CK, Jashim E, Aleem AM, Salam RA, Marnett LJ, Uddin MJ. Targeted Detection of Cyclooxygenase-1 in Ovarian Cancer. ACS Med Chem Lett 2020; 11:1837-1842. [PMID: 33062161 DOI: 10.1021/acsmedchemlett.9b00280] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 07/24/2019] [Indexed: 12/20/2022] Open
Abstract
Overexpression of cyclooxygenase-1 (COX-1) is associated with the initiation and progression of ovarian cancer, and targeted imaging of COX-1 is a promising strategy for early detection of this disease. We report the discovery of N-[(5-carboxy-X-rhodaminyl)but-4-yl]-3-(1-(4-methoxyphenyl)-5-(p-tolyl)-1H-pyrazol-3-yl)propenamide (CMP) as the first COX-1-targeted optical agent for imaging of ovarian cancer. CMP exhibits light emission at 604 nm (λmax), thereby minimizing tissue autofluorescence interference. In both purified enzyme and COX-1-expressing human ovarian adenocarcinoma (OVCAR-3) cells, CMP inhibits COX-1 at low nanomolar potencies (IC50 = 94 and 44 nM, respectively). CMP's selective binding to COX-1 in OVCAR-3 cells was visualized microscopically as intense intracellular fluorescence. In vivo optical imaging of xenografts in athymic nude mice revealed COX-1-dependent accumulation of CMP in COX-1-expressing mouse ovarian surface epithelial carcinoma (ID8-NGL) and OVCAR-3 cells. These results establish proof-of-principle for the feasibility of targeting COX-1 in the development of new imaging and therapeutic strategies for ovarian cancer.
Collapse
Affiliation(s)
- Paola Malerba
- A. B. Hancock, Jr. Memorial Laboratory for Cancer Research, Departments of Biochemistry, Chemistry, and Pharmacology, Vanderbilt Institute of Chemical Biology, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee 37232 United States
- Department of Pharmacy and Pharmaceutical Sciences, University of Bari “A. Moro”, Via Orabona 4, 70125 Bari, Italy
| | - Brenda C. Crews
- A. B. Hancock, Jr. Memorial Laboratory for Cancer Research, Departments of Biochemistry, Chemistry, and Pharmacology, Vanderbilt Institute of Chemical Biology, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee 37232 United States
| | - Kebreab Ghebreselasie
- A. B. Hancock, Jr. Memorial Laboratory for Cancer Research, Departments of Biochemistry, Chemistry, and Pharmacology, Vanderbilt Institute of Chemical Biology, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee 37232 United States
| | - Cristina K. Daniel
- A. B. Hancock, Jr. Memorial Laboratory for Cancer Research, Departments of Biochemistry, Chemistry, and Pharmacology, Vanderbilt Institute of Chemical Biology, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee 37232 United States
| | - Elma Jashim
- A. B. Hancock, Jr. Memorial Laboratory for Cancer Research, Departments of Biochemistry, Chemistry, and Pharmacology, Vanderbilt Institute of Chemical Biology, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee 37232 United States
| | - Ansari M. Aleem
- A. B. Hancock, Jr. Memorial Laboratory for Cancer Research, Departments of Biochemistry, Chemistry, and Pharmacology, Vanderbilt Institute of Chemical Biology, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee 37232 United States
| | - Redoan A. Salam
- A. B. Hancock, Jr. Memorial Laboratory for Cancer Research, Departments of Biochemistry, Chemistry, and Pharmacology, Vanderbilt Institute of Chemical Biology, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee 37232 United States
- Department of Biology, Washington University in St. Louis, St. Louis, Missouri 63130 United States
| | - Lawrence J. Marnett
- A. B. Hancock, Jr. Memorial Laboratory for Cancer Research, Departments of Biochemistry, Chemistry, and Pharmacology, Vanderbilt Institute of Chemical Biology, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee 37232 United States
| | - Md. Jashim Uddin
- A. B. Hancock, Jr. Memorial Laboratory for Cancer Research, Departments of Biochemistry, Chemistry, and Pharmacology, Vanderbilt Institute of Chemical Biology, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee 37232 United States
| |
Collapse
|
10
|
Affiliation(s)
- Xinping Zhang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering Southeast University Nanjing China
| | - Xiaoyang Liu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering Southeast University Nanjing China
| | - Yuxin Guo
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering Southeast University Nanjing China
| | - Fu‐Gen Wu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering Southeast University Nanjing China
| |
Collapse
|
11
|
Cekanova M, Pandey S, Olin S, Ryan P, Stokes JE, Hecht S, Martin-Jimenez T, Uddin MJ, Marnett LJ. Pharmacokinetic characterization of fluorocoxib D, a cyclooxygenase-2-targeted optical imaging agent for detection of cancer. JOURNAL OF BIOMEDICAL OPTICS 2020; 25:JBO-200044R. [PMID: 32860356 PMCID: PMC7456637 DOI: 10.1117/1.jbo.25.8.086005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 08/11/2020] [Indexed: 06/11/2023]
Abstract
SIGNIFICANCE Fluorocoxib D, N-[(rhodamin-X-yl)but-4-yl]-2-[1-(4-chlorobenzoyl)-5-methoxy-2-methyl-1H-indol-3-yl]acetamide, is a water-soluble optical imaging agent to detect cyclooxygenase-2 (COX-2)-expressing cancer cells. AIM We evaluated the pharmacokinetic and safety properties of fluorocoxib D and its ability to detect cancer cells in vitro and in vivo. APPROACH Pharmacokinetic parameters of fluorocoxib D were assessed from plasma collected at designated time points after intravenous administration of 1 mg / kg fluorocoxib D in six research dogs using a high-performance liquid chromatography analysis. Safety of fluorocoxib D was assessed for 3 days after its administration using physical assessment, complete blood count, serum chemistry profile, and complete urinalysis in six research dogs. The ability of fluorocoxib D to detect COX-2-expressing cancer cells was performed using human 5637 cells in vitro and during rhinoscopy evaluation of specific fluorocoxib D uptake by canine cancer cells in vivo. RESULTS No evidence of toxicity and no clinically relevant adverse events were noted in dogs. Peak concentration of fluorocoxib D (114.8 ± 50.5 ng / ml) was detected in plasma collected at 0.5 h after its administration. Pretreatment of celecoxib blocked specific uptake of fluorocoxib D in COX-2-expressing human 5637 cancer cells. Fluorocoxib D uptake was detected in histology-confirmed COX-2-expressing head and neck cancer during rhinoscopy in a client-owned dog in vivo. Specific tumor-to-normal tissue ratio of detected fluorocoxib D signal was in an average of 3.7 ± 0.9 using Image J analysis. CONCLUSIONS Our results suggest that fluorocoxib D is a safe optical imaging agent used for detection of COX-2-expressing cancers and their margins during image-guided minimally invasive biopsy and surgical procedures.
Collapse
Affiliation(s)
- Maria Cekanova
- The University of Tennessee, College of Veterinary Medicine, Department of Small Animal Clinical Sciences, Knoxville, Tennessee, United States
- The University of Tennessee, UT-ORNL Graduate School of Genome, Science and Technology, Knoxville, Tennessee, United States
| | - Sony Pandey
- The University of Tennessee, College of Veterinary Medicine, Department of Small Animal Clinical Sciences, Knoxville, Tennessee, United States
| | - Shelly Olin
- The University of Tennessee, College of Veterinary Medicine, Department of Small Animal Clinical Sciences, Knoxville, Tennessee, United States
| | - Phillip Ryan
- The University of Tennessee, College of Veterinary Medicine, Department of Small Animal Clinical Sciences, Knoxville, Tennessee, United States
| | - Jennifer E. Stokes
- The University of Tennessee, College of Veterinary Medicine, Department of Small Animal Clinical Sciences, Knoxville, Tennessee, United States
| | - Silke Hecht
- The University of Tennessee, College of Veterinary Medicine, Department of Small Animal Clinical Sciences, Knoxville, Tennessee, United States
| | - Tomas Martin-Jimenez
- The University of Tennessee, College of Veterinary Medicine, Department of Biomedical and Diagnostic Sciences, Knoxville, Tennessee, United States
| | - Md. Jashim Uddin
- Vanderbilt University School of Medicine, Vanderbilt Institute of Chemical Biology, Center for Molecular Toxicology and Vanderbilt-Ingram Cancer Center, A. B. Hancock, Jr., Memorial Laboratory for Cancer Research, Departments of Biochemistry, Chemistry and Pharmacology, Nashville, Tennessee, United States
| | - Lawrence J. Marnett
- Vanderbilt University School of Medicine, Vanderbilt Institute of Chemical Biology, Center for Molecular Toxicology and Vanderbilt-Ingram Cancer Center, A. B. Hancock, Jr., Memorial Laboratory for Cancer Research, Departments of Biochemistry, Chemistry and Pharmacology, Nashville, Tennessee, United States
| |
Collapse
|
12
|
Litchfield M, Wuest M, Glubrecht D, Wuest F. Radiosynthesis and Biological Evaluation of [ 18F]Triacoxib: A New Radiotracer for PET Imaging of COX-2. Mol Pharm 2019; 17:251-261. [PMID: 31816246 DOI: 10.1021/acs.molpharmaceut.9b00986] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Inducible isozyme cyclooxygenase-2 (COX-2) is upregulated under acute and chronic inflammatory conditions, including cancer, wherein it promotes angiogenesis, tissue invasion, and resistance to apoptosis. Due to its high expression in various cancers, COX-2 has become an important biomarker for molecular imaging and therapy of cancer. Recently, our group applied in situ click chemistry for the identification of the highly potent and selective COX-2 inhibitor triacoxib. In this study, we present the radiosynthesis in vitro and in vivo radiopharmacological validation of [18F]triacoxib, a novel radiotracer for PET imaging of COX-2. Radiosynthesis of [18F]triacoxib was accomplished using copper-mediated late-stage radiofluorination chemistry. The radiosynthesis, including radio-HPLC purification, of [18F]triacoxib was accomplished within 90 min in decay-corrected radiochemical yields of 72% (n = 7) at molar activities exceeding 90 GBq/μmol. Cellular uptake and inhibition studies with [18F]triacoxib were carried out in COX-2 expressing HCA-7 cells. Cellular uptake of [18F]triacoxib in HCA-7 cells reached 25% radioactivity/mg protein after 60 min. Cellular uptake was reduced by 63% upon pretreatment with 0.1 mM celecoxib, and 90% of the radiotracer remained intact in vivo after 60 min p.i. in mice. [18F]Triacoxib was further evaluated in HCA-7 tumor-bearing mice using dynamic PET imaging, radiometabolite analysis, autoradiography, and immunohistochemistry. PET imaging revealed a favorable baseline radiotracer uptake in HCA-7 tumors (SUV60min = 0.76 ± 0.02 (n = 4)), which could be blocked by 20% through i.p. pretreatment with 2 mg of celecoxib. Autoradiography and immunohistochemistry experiments further the confirmed blocking of COX-2 in vivo. [18F]Triacoxib, whose nonradioactive analogue was identified through in situ click chemistry, is a novel radiotracer for PET imaging of COX-2 in cancer. Despite a substantial amount of nonspecific uptake in vivo, [18F]triacoxib displayed specific binding to COX-2 in vivo and reinforced the feasibility of optimal structure selection by in situ click chemistry. It remains to be elucidated how this novel radiotracer would perform in first-in-human studies to detect COX-2 with PET.
Collapse
Affiliation(s)
- Marcus Litchfield
- Department of Oncology , University of Alberta , 11560 University Avenue , Edmonton , Alberta T6G 1Z2 , Canada
| | - Melinda Wuest
- Department of Oncology , University of Alberta , 11560 University Avenue , Edmonton , Alberta T6G 1Z2 , Canada.,Cancer Research Institute of Northern Alberta , University of Alberta , Edmonton , Alberta T6G 2S2 , Canada
| | - Darryl Glubrecht
- Department of Oncology , University of Alberta , 11560 University Avenue , Edmonton , Alberta T6G 1Z2 , Canada
| | - Frank Wuest
- Department of Oncology , University of Alberta , 11560 University Avenue , Edmonton , Alberta T6G 1Z2 , Canada.,Cancer Research Institute of Northern Alberta , University of Alberta , Edmonton , Alberta T6G 2S2 , Canada
| |
Collapse
|
13
|
Yamali C, Gul HI, Ece A, Bua S, Angeli A, Sakagami H, Sahin E, Supuran CT. Synthesis, biological evaluation and in silico modelling studies of 1,3,5-trisubstituted pyrazoles carrying benzenesulfonamide as potential anticancer agents and selective cancer-associated hCA IX isoenzyme inhibitors. Bioorg Chem 2019; 92:103222. [PMID: 31499260 DOI: 10.1016/j.bioorg.2019.103222] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 08/16/2019] [Accepted: 08/26/2019] [Indexed: 01/10/2023]
Abstract
Inhibition of carbonic anhydrases (CAs, EC 4.2.1.1) has clinical importance for the treatment of several diseases. They participate in crucial regulatory mechanisms for balancing intracellular and extracellular pH of the cells. Among CA isoforms, selective inhibition of hCA IX has been linked to decreasing of cell growth for both primary tumors and metastases. The discovery of novel CA inhibitors as anticancer drug candidates is a current topic in medicinal chemistry. 1,3,5-Trisubstituted pyrazoles carrying benzenesulfonamide were evaluated against physiologically abundant cytosolic hCA I and hCA II and trans-membrane, tumor-associated hCA IX isoforms by a stopped-flow CO2 hydrase method. Their in vitro cytotoxicities were screened against human oral squamous cell carcinoma (OSCC) cell lines (HSC-2) and human mesenchymal normal oral cells (HGF) via 3-(4,5-Dimethylthiazol-2-yl)-2,5-Diphenyltetrazolium Bromide (MTT) test. Compounds 6, 8, 9, 11, and 12 showed low nanomolar hCA II inhibitory potency with Ki < 10 nM, whereas compounds 9 and 12 displayed Ki < 10 nM against hCA IX isoenzyme when compared with reference Acetazolamide (AZA). Compound 9, 4-(3-(hydrazinecarbonyl)-5-(4-nitrophenyl)-1H-pyrazol-1-yl)benzenesulfonamide, can be considered as the most selective hCA IX inhibitor over off-target cytosolic isoenzymes hCA I and hCA II with the lowest Ki value of 2.3 nM and selectivity ratios of 3217 (hCA I/hCA IX) and 3.9 (hCA II/hCA IX). Isoform selectivity profiles were also discussed using in silico modelling. Cytotoxicity results pointed out that compounds 5 (CC50 = 37.7 μM) and 11 (CC50 = 58.1 μM) can be considered as lead cytotoxic compounds since they were more cytotoxic than 5-Fluorouracil (5-FU) and Methotrexate (MTX).
Collapse
Affiliation(s)
- Cem Yamali
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Ataturk University, Erzurum, Turkey
| | - Halise Inci Gul
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Ataturk University, Erzurum, Turkey.
| | - Abdulilah Ece
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Biruni University, Istanbul, Turkey
| | - Silvia Bua
- Neurofarba Department, Sezione di Scienza Farmaceutiche e Nutraceutiche, Universita degli Studi di Firenze, Via U. Schiff 6, 50019 Sesto Fiorentino (Florence), Italy
| | - Andrea Angeli
- Neurofarba Department, Sezione di Scienza Farmaceutiche e Nutraceutiche, Universita degli Studi di Firenze, Via U. Schiff 6, 50019 Sesto Fiorentino (Florence), Italy
| | - Hiroshi Sakagami
- Meikai University Research Institute of Odontology (M-RIO), Sakado, Saitama 350-0283, Japan
| | - Ertan Sahin
- Department of Chemistry, Faculty of Science, Ataturk University, Erzurum, Turkey
| | - Claudiu T Supuran
- Neurofarba Department, Sezione di Scienza Farmaceutiche e Nutraceutiche, Universita degli Studi di Firenze, Via U. Schiff 6, 50019 Sesto Fiorentino (Florence), Italy
| |
Collapse
|
14
|
Bourn J, Pandey S, Uddin J, Marnett L, Cekanova M. Detection of tyrosine kinase inhibitors-induced COX-2 expression in bladder cancer by fluorocoxib A. Oncotarget 2019; 10:5168-5180. [PMID: 31497247 PMCID: PMC6718263 DOI: 10.18632/oncotarget.27125] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 07/17/2019] [Indexed: 01/02/2023] Open
Abstract
Among challenges of targeted therapies is the activation of alternative pro-survival signaling pathways in cancer cells, resulting in an acquired drug resistance. Cyclooxygenase-2 (COX-2) is overexpressed in bladder cancer cells, making it an attractive molecular target for the detection and treatment of cancer. Fluorocoxib A is an optical imaging agent that selectively targets COX-2. In this study, we evaluated the ability of fluorocoxib A to monitor the responses of bladder cancer to targeted therapies in vivo. The effects of several tyrosine kinase inhibitors (TKIs: axitinib, AB1010, toceranib, imatinib, erlotinib, gefitinib, imatinib, sorafenib, vandetanib, SP600125, UO126, and AZD 5438) on COX-2 expression were validated in ten human and canine bladder cancer cell lines (J82, RT4, T24, UM-UC-3, 5637, SW780, TCCSUP, K9TCC#1Lillie, K9TCC#2Dakota, K9TCC#5Lilly) in vitro. The effects of TKIs on bladder cancer in vivo were evaluated using the COX-2-expressing K9TCC#5Lilly xenograft mouse model and detected by fluorocoxib A. The increased COX-2 expression was detected by all tested TKIs in at least one of the tested COX-2-expressing bladder cancer cell lines (5637, SW780, TCCSUP, K9TCC#1Lillie, K9TCC#2Dakota, and K9TCC#5Lilly) in vitro. In addition, fluorocoxib A uptake correlated with the AB1010- and imatinib-induced COX-2 expression in the K9TCC#5Lilly xenografts in vivo. In conclusion, these results indicate that fluorocoxib A could be used for the monitoring the early responses to targeted therapies in COX-2-expressing bladder cancer.
Collapse
Affiliation(s)
- Jennifer Bourn
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, The University of Tennessee, Knoxville, TN 37996, USA.,University of Tennessee and Oak Ridge National Laboratory, Graduate School of Genome Science and Technology, The University of Tennessee, Knoxville, TN 37996, USA.,Current address: Department of Cancer Biology, College of Medicine, University of Cincinnati, Cincinnati, OH 45221, USA
| | - Sony Pandey
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, The University of Tennessee, Knoxville, TN 37996, USA
| | - Jashim Uddin
- A. B. Hancock, Jr., Memorial Laboratory for Cancer Research, Departments of Biochemistry, Chemistry and Pharmacology, Vanderbilt Institute of Chemical Biology, Center for Molecular Toxicology and Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Lawrence Marnett
- A. B. Hancock, Jr., Memorial Laboratory for Cancer Research, Departments of Biochemistry, Chemistry and Pharmacology, Vanderbilt Institute of Chemical Biology, Center for Molecular Toxicology and Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Maria Cekanova
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, The University of Tennessee, Knoxville, TN 37996, USA.,University of Tennessee and Oak Ridge National Laboratory, Graduate School of Genome Science and Technology, The University of Tennessee, Knoxville, TN 37996, USA
| |
Collapse
|
15
|
Zhang X, Rakesh KP, Qin HL. Transition-metal-free regioselective construction of 1,5-diaryl-1,2,3-triazoles through dehydrative cycloaddition of alcohols with aryl azides mediated by SO 2F 2. Chem Commun (Camb) 2019; 55:2845-2848. [PMID: 30768105 DOI: 10.1039/c8cc09693g] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
A novel, simple and practical method for mild, efficient, cost-effective and regioselective synthesis of highly valuable 1,5-diaryl-1,2,3-triazoles was achieved through dehydrative annulation of readily available alcohols with aryl azides. The reaction proceeded at room temperature, without any metal catalysts, exhibiting excellent compatibility to a large variety of functional groups (>50 examples), resulting in up to quantitative yields.
Collapse
Affiliation(s)
- Xu Zhang
- State Key Laboratory of Silicate Materials for Architectures, School of Chemistry, Chemical Engineering and Life Science Wuhan University of Technology, Wuhan 430070, China.
| | | | | |
Collapse
|
16
|
Medicinal chemistry of vicinal diaryl scaffold: A mini review. Eur J Med Chem 2018; 162:1-17. [PMID: 30396033 DOI: 10.1016/j.ejmech.2018.10.054] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 10/19/2018] [Accepted: 10/22/2018] [Indexed: 12/22/2022]
Abstract
The privileged structures have been widely used as a valuable template in new drug discovery. 1,2-Diaryl or vicinal diaryl is a simple scaffold found in many drugs and naturally occurring compounds. From synthetic point of view, the vicinal diaryl derivatives are easily accessible due to their facile and expedient syntheses. These scaffolds have shown numerous interesting pharmacological activities against various diseases with lot of clinical potentials. This review aims to highlight the evidence of vicinal diaryl motif as a privileged scaffold in COX-2 inhibitors and CA-4 analogs.
Collapse
|
17
|
Zhang Q, Han Z, Tao J, Zhang W, Li P, Tang L, Gu Y. A novel near-infrared fluorescent probe for monitoring cyclooxygenase-2 in inflammation and tumor. JOURNAL OF BIOPHOTONICS 2018; 11:e201700339. [PMID: 29341436 DOI: 10.1002/jbio.201700339] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 01/14/2018] [Indexed: 06/07/2023]
Abstract
Targeting cyclooxygenase-2 (COX-2) for molecular imaging is an attractive approach applicable for its overexpression in inflammation and many malignancies. Herein, for monitoring COX-2, we synthesize a specific COX-2 probe celecoxib-MPA probe (CMP), based on celecoxib and a water-soluble near-infrared dye dye ICG-Der-02 (MPA). Its high affinity for binding to COX-2 is verified by molecular docking, dynamics simulation and inhibition assay. At cellular level, CMP selectively accumulates in cytoplasm of COX-2-positive cells. in vivo assays, probe guided-imaging in inflamed or cancerous tissues confirms that CMP can bind to the locally endogenic COX-2 and exhibit intense fluorescence. Importantly, we further prove the targeting specificity of CMP as the fluorescence is significantly reduced by blocking COX-2 active site through preinjection with celecoxib. The results suggest that the probe CMP, with favorable hydrophilic property, good biocompatibility, long-term observation, excellent targeting ability and optical imaging capability, could serve as a promising probe for real-time monitoring COX-2 in inflammation and tumor.
Collapse
Affiliation(s)
- Qi Zhang
- State Key Laboratory of Natural Medicines, Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing, China
| | - Zhihao Han
- State Key Laboratory of Natural Medicines, Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing, China
| | - Ji Tao
- State Key Laboratory of Natural Medicines, Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing, China
| | - Wancun Zhang
- State Key Laboratory of Natural Medicines, Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing, China
| | - Ping Li
- State Key Laboratory of Natural Medicines, Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing, China
| | - Liping Tang
- Department of Bioengineering, University of Texas at Arlington, Arlington, Texas
| | - Yueqing Gu
- State Key Laboratory of Natural Medicines, Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
18
|
Kumar N, Ansari MY, Kant R, Kumar A. Copper-catalyzed decarboxylative regioselective synthesis of 1,5-disubstituted 1,2,3-triazoles. Chem Commun (Camb) 2018; 54:2627-2630. [DOI: 10.1039/c7cc09934g] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A copper-catalyzed decarboxylative regioselective protocol for the synthesis of 1,5-disubstituted 1,2,3-triazoles via direct annulation of cinnamic acids with aryl azides has been developed. This is the first example of 1,5-disubstituted 1,2,3-triazoles using Cu(ii) as the catalyst, which were generally synthesized using a ruthenium(ii) catalyst.
Collapse
Affiliation(s)
- Navaneet Kumar
- Medicinal & Process Chemistry Division
- CSIR-Central Drug Research Institute
- Lucknow 226031
- India
| | - Mohd Yeshab Ansari
- Medicinal & Process Chemistry Division
- CSIR-Central Drug Research Institute
- Lucknow 226031
- India
| | - Ruchir Kant
- Molecular and Structural Biology
- CSIR-Central Drug Research Institute
- Lucknow 226031
- India
| | - Atul Kumar
- Medicinal & Process Chemistry Division
- CSIR-Central Drug Research Institute
- Lucknow 226031
- India
- Academy of Scientific and Innovative Research
| |
Collapse
|
19
|
Morgenroth A, Vogg ATJ, Neumaier B, Mottaghy FM, Zlatopolskiy BD. Radioiodinated indomethacin amide for molecular imaging of cyclooxygenase-2 expressing tumors. Oncotarget 2017; 8:18059-18069. [PMID: 28407689 PMCID: PMC5392307 DOI: 10.18632/oncotarget.15437] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Accepted: 01/27/2017] [Indexed: 12/18/2022] Open
Abstract
Cyclooxygenase-2 (COX-2) is an important biomarker in several tumors. Available imaging probes display relatively low tumor to background ratios (smaller than 2:1). We evaluated newly developed indomethacin (Ind) derivatives for in vivo molecular imaging of COX-2 expressing carcinoma. Radioiodinated Ind derivatives Ind-NH-(CH2)4-NH-3-[I-125]I-Bz ([I-125]5), Ind-NH-(CH2)4-NH-5-[I-124/125]I-Nic ([I-124/125]6) and Ind-NH-(CH2)4-NH-5-[I-125]I-Iphth ([I-125]7) were prepared from the respective SnBu3-precursors (45-80% radiochemical yield; > 95% radiochemical purity). The cellular uptake of [I-125]5 and [I-125]6 correlated with COX-2 expression determined by SDS page/Western blot analysis. [I-125]5 was predominantly localized in the cell membrane while [I-125]6 was internalized and displayed a diffuse and favorable cytoplasmic distribution. In contrast, [I-125]7 showed only low uptake in COX-2 positive cells. Co-incubation with the COX-2 inhibitor Celecoxib led to an almost complete suppression of cellular uptake of [I-125]5 and [I-125]6. In vivo molecular imaging using positron emission tomography (PET) in SCID mice xenografted with COX-2+ (HT29) and COX-2- (HCT116) human colorectal carcinoma cells was performed for [I-124]6. HT29 xenografts displayed a significantly higher uptake than HCT-116 xenografts (5.6 ± 1.5 vs. 0.5 ± 0.1 kBq/g, P < 0.05) with an extraordinary high tumor to muscle ratio (50.3 ± 1.5). Immunohistological staining correlated with the imaging data. In conclusion, the novel radioiodinated indomethacin derivative ([I-124/125]6) could become a valuable tool for development of molecular imaging probes for visualization of COX-2 expressing tumors.
Collapse
Affiliation(s)
- Agnieszka Morgenroth
- Department for Nuclear Medicine, RWTH Aachen University Hospital, 52074, Aachen, Germany
| | - Andreas T J Vogg
- Department for Nuclear Medicine, RWTH Aachen University Hospital, 52074, Aachen, Germany
| | - Bernd Neumaier
- Institute of Radiochemistry and Experimental Molecular Imaging, University Clinic Cologne, 50937, Cologne, Germany.,Max Planck Institute of Metabolism Research, 50931, Cologne, Germany.,Institute of Neuroscience and Medicine, Nuclear Chemistry (INM-5), Research Center Jülich, 52425, Jülich, Germany
| | - Felix M Mottaghy
- Department for Nuclear Medicine, RWTH Aachen University Hospital, 52074, Aachen, Germany.,Department of Nuclear Medicine, Maastricht University Medical Center, 6229 HX, Maastricht, The Netherlands
| | - Boris D Zlatopolskiy
- Department for Nuclear Medicine, RWTH Aachen University Hospital, 52074, Aachen, Germany.,Institute of Radiochemistry and Experimental Molecular Imaging, University Clinic Cologne, 50937, Cologne, Germany.,Max Planck Institute of Metabolism Research, 50931, Cologne, Germany
| |
Collapse
|
20
|
Raji I, Yadudu F, Janeira E, Fathi S, Szymczak L, Kornacki JR, Komatsu K, Li JD, Mrksich M, Oyelere AK. Bifunctional conjugates with potent inhibitory activity towards cyclooxygenase and histone deacetylase. Bioorg Med Chem 2017; 25:1202-1218. [PMID: 28057407 PMCID: PMC5291751 DOI: 10.1016/j.bmc.2016.12.032] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Revised: 12/17/2016] [Accepted: 12/20/2016] [Indexed: 12/16/2022]
Abstract
We herein disclose a series of compounds with potent inhibitory activities towards histone deacetylases (HDAC) and cyclooxygenases (COX). These compounds potently inhibited the growth of cancer cell lines consistent with their anti-COX and anti-HDAC activities. While compound 2b showed comparable level of COX-2 selectivity as celecoxib, compound 11b outperformed indomethacin in terms of selectivity towards COX-2 relative to COX-1. An important observation with our lead compounds (2b, 8, 11b, and 17b) is their enhanced cytotoxicity towards androgen dependent prostate cancer cell line (LNCaP) relative to androgen independent prostate cancer cell line (DU-145). Interestingly, compounds 2b and 17b arrested the cell cycle progression of LNCaP in the S-phase, while compound 8 showed a G0/G1 arrest, similar to SAHA. Relative to SAHA, these compounds displayed tumor-selective cytotoxicity as they have low anti-proliferative activity towards healthy cells (VERO); an attribute that makes them attractive candidates for drug development.
Collapse
Affiliation(s)
- Idris Raji
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA 30332-0400, USA
| | - Fatima Yadudu
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA 30332-0400, USA
| | - Emily Janeira
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA
| | - Shaghayegh Fathi
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA 30332-0400, USA
| | - Lindsey Szymczak
- Departments of Chemistry and Biomedical Engineering, Northwestern University, 2145 Sheridan Road, Evanston, IL 60208-3113, USA
| | - James Richard Kornacki
- Departments of Chemistry and Biomedical Engineering, Northwestern University, 2145 Sheridan Road, Evanston, IL 60208-3113, USA
| | - Kensei Komatsu
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, USA
| | - Jian-Dong Li
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, USA
| | - Milan Mrksich
- Departments of Chemistry and Biomedical Engineering, Northwestern University, 2145 Sheridan Road, Evanston, IL 60208-3113, USA
| | - Adegboyega K Oyelere
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA 30332-0400, USA; Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332-0400, USA.
| |
Collapse
|
21
|
Tietz O, Dzandzi J, Bhardwaj A, Valliant JF, Wuest F. Design and synthesis of [ 125 I]Pyricoxib: A novel 125 I-labeled cyclooxygenase-2 (COX-2) inhibitors. Bioorg Med Chem Lett 2016; 26:1516-1520. [DOI: 10.1016/j.bmcl.2016.02.029] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Revised: 02/08/2016] [Accepted: 02/10/2016] [Indexed: 02/01/2023]
|
22
|
Frese J, Morgenroth A, Mertens ME, Koch S, Rongen L, Vogg ATJ, Zlatopolskiy BD, Neumaier B, Gesche VN, Lammers T, Schmitz-Rode T, Mela P, Jockenhoevel S, Mottaghy FM, Kiessling F. Nondestructive monitoring of tissue-engineered constructs. ACTA ACUST UNITED AC 2015; 59:165-75. [PMID: 24021591 DOI: 10.1515/bmt-2013-0029] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2013] [Accepted: 08/13/2013] [Indexed: 11/15/2022]
Abstract
Abstract Tissue engineering as a multidisciplinary field enables the development of living substitutes to replace, maintain, or restore diseased tissue and organs. Since the term was introduced in medicine in 1987, tissue engineering strategies have experienced significant progress. However, up to now, only a few substitutes were able to overcome the gap from bench to bedside and have been successfully approved for clinical use. Substantial donor variability makes it difficult to predict the quality of tissue-engineered constructs. It is essential to collect sufficient data to ensure that poor or immature constructs are not implanted into patients. The fulfillment of certain quality requirements, such as mechanical and structural properties, is crucial for a successful implantation. There is a clear need for new nondestructive and real-time online monitoring and evaluation methods for tissue-engineered constructs, which are applicable on the biomaterial, tissue, cellular, and subcellular levels. This paper reviews current established nondestructive techniques for implant monitoring including biochemical methods and noninvasive imaging.
Collapse
|
23
|
Uddin MJ, Elleman AV, Ghebreselasie K, Daniel CK, Crews BC, Nance KD, Huda T, Marnett LJ. Design of Fluorine-Containing 3,4-Diarylfuran-2(5H)-ones as Selective COX-1 Inhibitors. ACS Med Chem Lett 2014; 5:1254-8. [PMID: 25408841 DOI: 10.1021/ml500344j] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Accepted: 10/12/2014] [Indexed: 01/29/2023] Open
Abstract
We report the design and synthesis of fluorine-containing cyclooxygenase-1 (COX-1)-selective inhibitors to serve as prototypes for the development of a COX-1-targeted imaging agent. Deletion of the SO2CH3 group of rofecoxib switches the compound from a COX-2- to a COX-1-selective inhibitor, providing a 3,4-diarylfuran-2(5H)-one scaffold for structure-activity relationship studies of COX-1 inhibition. A wide range of fluorine-containing 3,4-diarylfuran-2(5H)-ones were designed, synthesized, and tested for their ability to selectively inhibit COX-1 in purified protein and human cancer cell assays. Compounds containing a fluoro-substituent on the C-3 phenyl ring and a methoxy-substituent on the C-4 phenyl ring of the 3,4-diarylfuran-2(5H)-one scaffold were the best COX-1-selective agents of those evaluated, exhibiting IC50s in the submicromolar range. These compounds provide the foundation for development of an agent to facilitate radiologic imaging of ovarian cancer expressing elevated levels of COX-1.
Collapse
Affiliation(s)
- Md. Jashim Uddin
- A. B. Hancock, Jr., Memorial
Laboratory for Cancer Research, Department of Biochemistry, Chemistry
and Pharmacology, Vanderbilt Institute of Chemical Biology, Center
for Molecular Toxicology and Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Anna V. Elleman
- A. B. Hancock, Jr., Memorial
Laboratory for Cancer Research, Department of Biochemistry, Chemistry
and Pharmacology, Vanderbilt Institute of Chemical Biology, Center
for Molecular Toxicology and Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Kebreab Ghebreselasie
- A. B. Hancock, Jr., Memorial
Laboratory for Cancer Research, Department of Biochemistry, Chemistry
and Pharmacology, Vanderbilt Institute of Chemical Biology, Center
for Molecular Toxicology and Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Cristina K. Daniel
- A. B. Hancock, Jr., Memorial
Laboratory for Cancer Research, Department of Biochemistry, Chemistry
and Pharmacology, Vanderbilt Institute of Chemical Biology, Center
for Molecular Toxicology and Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Brenda C. Crews
- A. B. Hancock, Jr., Memorial
Laboratory for Cancer Research, Department of Biochemistry, Chemistry
and Pharmacology, Vanderbilt Institute of Chemical Biology, Center
for Molecular Toxicology and Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Kellie D. Nance
- A. B. Hancock, Jr., Memorial
Laboratory for Cancer Research, Department of Biochemistry, Chemistry
and Pharmacology, Vanderbilt Institute of Chemical Biology, Center
for Molecular Toxicology and Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Tamanna Huda
- A. B. Hancock, Jr., Memorial
Laboratory for Cancer Research, Department of Biochemistry, Chemistry
and Pharmacology, Vanderbilt Institute of Chemical Biology, Center
for Molecular Toxicology and Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Lawrence J. Marnett
- A. B. Hancock, Jr., Memorial
Laboratory for Cancer Research, Department of Biochemistry, Chemistry
and Pharmacology, Vanderbilt Institute of Chemical Biology, Center
for Molecular Toxicology and Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| |
Collapse
|
24
|
Pacelli A, Greenman J, Cawthorne C, Smith G. Imaging COX-2 expression in cancer using PET/SPECT radioligands: current status and future directions. J Labelled Comp Radiopharm 2014; 57:317-22. [PMID: 24470172 DOI: 10.1002/jlcr.3160] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Accepted: 10/29/2013] [Indexed: 12/21/2022]
Abstract
The role of cyclooxygenase (COX)-2 as a driving force in early tumourigenesis and the current interest in the combination of COX-2 inhibitors with standard therapy in clinical trials creates an urgent need to establish clinically relevant diagnostic tests for COX-2 expression. Molecular imaging using small-molecule probes radiolabelled for both positron emission tomography (PET) and single photon emission computed tomography (SPECT) offers the potential to meet this need, providing a minimally invasive readout for the whole disease burden. This review summarises current approaches to the radiolabelling of small-molecule COX-2 inhibitors and their analogues for PET and SPECT imaging, and gives an overview of their biological evaluation and likely success of clinical application.
Collapse
Affiliation(s)
- A Pacelli
- School of Biological, Biomedical and Environmental Sciences, University of Hull, Hull, HU6 7RX, UK
| | | | | | | |
Collapse
|
25
|
Mert S, Kasımoğulları R, İça T, Çolak F, Altun A, Ok S. Synthesis, structure-activity relationships, and in vitro antibacterial and antifungal activity evaluations of novel pyrazole carboxylic and dicarboxylic acid derivatives. Eur J Med Chem 2014; 78:86-96. [PMID: 24681068 DOI: 10.1016/j.ejmech.2014.03.033] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2014] [Revised: 03/09/2014] [Accepted: 03/12/2014] [Indexed: 11/25/2022]
Abstract
A series of pyrazole-3-carboxylic acid and pyrazole-3,4-dicarboxylic acid derivatives were synthesized, the structures were confirmed by their NMR ((1)H and (13)C) and FT-IR spectra, and elemental analyses. The antibacterial and antifungal activities of the compounds against five bacterial and five fungal pathogens were screened using modified agar well diffusion assay. Most of the molecules have inhibitory effects on both standard and clinical Candida albicans strains. However, only the molecules 8, 10, 21, and 22 demonstrate some inhibitory effects on Candida parapsilosis, Candida tropicalis, and Candida glabrata strains. The structure-antifungal activity relationships of the compounds on the C. albicans strains were investigated by electron-conformational method. The pharmacophores and antipharmacophores responsible for the inhibition and non-inhibition of the C. albicans strains were obtained by electronic and geometrical characteristics of the reactive fragments of the molecules. These fragments along with the associated parameters can be used in designing the future more potent antifungal agents. It has been shown that both the positions of electronegative atoms like F and O in the pyrazole substituents and the amount of the associated charges on such atoms are crucial in regulating the strength of antifungal activity for the C. albicans strain.
Collapse
Affiliation(s)
- Samet Mert
- Department of Chemistry, Faculty of Arts and Sciences, Dumlupinar University, 43100 Kutahya, Turkey
| | - Rahmi Kasımoğulları
- Department of Chemistry, Faculty of Arts and Sciences, Dumlupinar University, 43100 Kutahya, Turkey
| | - Tuba İça
- Department of Biology, Faculty of Arts and Sciences, Dumlupinar University, 43100 Kutahya, Turkey
| | - Ferdağ Çolak
- Department of Biology, Faculty of Arts and Sciences, Dumlupinar University, 43100 Kutahya, Turkey
| | - Ahmet Altun
- Department of Physics, Fatih University, 34500 B. Cekmece, Istanbul, Turkey
| | - Salim Ok
- School of Earth Sciences, The Ohio State University, 43210 Columbus, OH, USA.
| |
Collapse
|
26
|
Bhardwaj A, Kaur J, Wuest F, Knaus EE. Fluorophore-labeled cyclooxygenase-2 inhibitors for the imaging of cyclooxygenase-2 overexpression in cancer: synthesis and biological studies. ChemMedChem 2013; 9:109-16, 240. [PMID: 24376205 DOI: 10.1002/cmdc.201300355] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2013] [Revised: 10/08/2013] [Indexed: 01/26/2023]
Abstract
A group of cyclooxygenase-2 (COX-2)-specific fluorescent cancer biomarkers were synthesized by linking the anti-inflammatory drugs ibuprofen, (S)-naproxen, and celecoxib to the 7-nitrobenzofurazan (NBD) fluorophore. In vitro COX-1/COX-2 inhibition studies indicated that all of these fluorescent conjugates are COX-2 inhibitors (IC₅₀ range: 0.19-23.0 μM) with an appreciable COX-2 selectivity index (SI≥4.3-444). In this study the celecoxib-NBD conjugate N-(2-((7-nitrobenzo[c][1,2,5]oxadiazol-4-yl)amino)ethyl)-4-(5-(p-tolyl)-3-(trifluoromethyl)-1H-pyrazol-1-yl)benzenesulfonamide, which displayed the highest COX-2 inhibitory potency and selectivity (COX-2 IC₅₀ =0.19 μM; SI=443.6), was identified as an impending COX-2-specific biomarker for the fluorescence imaging of cancer using a COX-2-expressing human colon cancer cell line (HCA-7).
Collapse
Affiliation(s)
- Atul Bhardwaj
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, T6G 2E1 (Canada); Department of Oncology, Cross Cancer Institute, University of Alberta, Edmonton, Alberta, T6G 1Z2 (Canada)
| | | | | | | |
Collapse
|
27
|
Laube M, Kniess T, Pietzsch J. Radiolabeled COX-2 inhibitors for non-invasive visualization of COX-2 expression and activity--a critical update. Molecules 2013; 18:6311-55. [PMID: 23760031 PMCID: PMC6269837 DOI: 10.3390/molecules18066311] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2013] [Revised: 05/16/2013] [Accepted: 05/24/2013] [Indexed: 01/21/2023] Open
Abstract
Cyclooxygenase-2 (COX-2) is a key player in inflammation. Its overexpression is directly associated with various inflammatory diseases and, additionally, with several processes of carcinogenesis. The development of new selective COX-2 inhibitors (COXIBs) for use in cancer treatment is in the focus of the medicinal chemistry research field. For this purpose, a set of methods is available to determine COX-2 expression and activity in vitro and ex vivo but it is still a problem to functionally characterize COX-2 in vivo. This review focusses on imaging agents targeting COX-2 which have been developed for positron emission tomography (PET) and single photon emission computed tomography (SPECT) since 2005. The literature reveals that different radiochemical methods are available to synthesize COXIBs radiolabeled with fluorine-18, carbon-11, and isotopes of radioiodine. Unfortunately, most of the compounds tested did not show sufficient stability in vivo due to de[18F]fluorination or de[11C]methylation or they failed to bind specifically in the target region. So, suitable stability in vivo, matching lipophilicity for the target compartment and both high affinity and selectivity for COX-2 were identified as prominent criteria for radiotracer development. Up to now, it is not clear what approach and which model is the most suited to evaluate COX-2 targeting imaging agents in vivo. However, for proof of principle it has been shown that some radiolabeled compounds can bind specifically in COX-2 overexpressing tissue which gives hope for future work in this field.
Collapse
Affiliation(s)
- Markus Laube
- Department Radiopharmaceutical and Chemical Biology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Bautzner Landstrasse 400, 01328 Dresden, Germany; E-Mails: (T.K.); (J.P.)
- Department of Chemistry and Food Chemistry, Technische Universität Dresden, 01062 Dresden, Germany
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +49-351-260-2810; Fax: +49-351-260-2915
| | - Torsten Kniess
- Department Radiopharmaceutical and Chemical Biology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Bautzner Landstrasse 400, 01328 Dresden, Germany; E-Mails: (T.K.); (J.P.)
| | - Jens Pietzsch
- Department Radiopharmaceutical and Chemical Biology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Bautzner Landstrasse 400, 01328 Dresden, Germany; E-Mails: (T.K.); (J.P.)
- Department of Chemistry and Food Chemistry, Technische Universität Dresden, 01062 Dresden, Germany
| |
Collapse
|
28
|
Uddin MJ, Crews BC, Ghebreselasie K, Marnett LJ. Design, synthesis, and structure-activity relationship studies of fluorescent inhibitors of cycloxygenase-2 as targeted optical imaging agents. Bioconjug Chem 2013; 24:712-23. [PMID: 23488616 PMCID: PMC3630741 DOI: 10.1021/bc300693w] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Cycloxygenase-2 (COX-2) is an attractive target for molecular imaging because it is an inducible enzyme that is expressed in response to inflammatory and proliferative stimuli. Recently, we reported that conjugation of indomethacin with carboxy-X-rhodamine dyes results in the formation of effective, targeted, optical imaging agents able to detect COX-2 in inflammatory tissues and premalignant and malignant tumors (Uddin et al. Cancer Res. 2010, 70, 3618-3627). The present paper summarizes the details of the structure-activity relationship (SAR) studies performed for lead optimization of these dyes. A wide range of fluorescent conjugates were designed and synthesized, and each of them was tested for the ability to selectively inhibit COX-2 as the purified protein and in human cancer cells. The SAR study revealed that indomethacin conjugates are the best COX-2-targeted agents compared to the other carboxylic acid-containing nonsteroidal anti-inflammatory drugs (NSAIDs) or COX-2-selective inhibitors (COXIBs). An n-butyldiamide linker is optimal for tethering bulky fluorescent functionalities onto the NSAID or COXIB cores. The activity of conjugates also depends on the size, shape, and electronic properties of the organic fluorophores. These reagents are taken up by COX-2-expressing cells in culture, and the uptake is blocked by pretreatment with a COX inhibitor. In in vivo settings, these reagents become highly enriched in COX-2-expressing tumors compared to surrounding normal tissue, and they accumulate selectively in COX-2-expressing tumors as compared with COX-2-negative tumors grown in mice. Thus, COX-2-targeted fluorescent inhibitors are useful for preclinical and clinical detection of lesions containing elevated levels of COX-2.
Collapse
Affiliation(s)
- Md Jashim Uddin
- A. B. Hancock, Jr., Memorial Laboratory for Cancer Research, Department of Biochemistry, Chemistry and Pharmacology, Vanderbilt Institute of Chemical Biology, Center for Molecular Toxicology and Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN 37232-0146, USA
| | | | | | | |
Collapse
|
29
|
Cekanova M, Uddin MJ, Bartges JW, Callens A, Legendre AM, Rathore K, Wright L, Carter A, Marnett LJ. Molecular imaging of cyclooxygenase-2 in canine transitional cell carcinomas in vitro and in vivo. Cancer Prev Res (Phila) 2013; 6:466-76. [PMID: 23531445 DOI: 10.1158/1940-6207.capr-12-0358] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The enzyme COX-2 is induced at high levels in tumors but not in surrounding normal tissues, which makes it an attractive target for molecular imaging of cancer. We evaluated the ability of novel optical imaging agent, fluorocoxib A to detect urinary bladder canine transitional cell carcinomas (K9TCC). Here, we show that fluorocoxib A uptake overlapped with COX-2 expression in primary K9TCC cells in vitro. Using subcutaneously implanted primary K9TCC in athymic mice, we show specific uptake of fluorocoxib A by COX-2-expressing K9TCC xenograft tumors in vivo. Fluorocoxib A uptake by COX-2-expressing xenograft tumors was blocked by 70% (P < 0.005) when pretreated with the COX-2 selective inhibitor, celecoxib (10 mg/kg), 4 hours before intravenous administration of fluorocoxib A (1 mg/kg). Fluorocoxib A was taken up by COX-2-expressing tumors but not by COX-2-negative human UMUC-3 xenograft tumors. UMUC-3 xenograft tumors with no expression of COX-2 showed no uptake of fluorocoxib A. In addition, fluorocoxib A uptake was evaluated in five dogs diagnosed with TCC. Fluorocoxib A specifically detected COX-2-expressing K9TCC during cystoscopy in vivo but was not detected in normal urothelium. Taken together, our findings show that fluorocoxib A selectively bound to COX-2-expressing primary K9TCC cells in vitro, COX-2-expressing K9TCC xenografts tumors in nude mice, and heterogeneous canine TCC during cystoscopy in vivo. Spontaneous cancers in companion animals offer a unique translational model for evaluation of novel imaging and therapeutic agents using primary cancer cells in vitro and in heterogeneous cancers in vivo.
Collapse
Affiliation(s)
- Maria Cekanova
- Department of Small Animal Clinical Sciences, The University of Tennessee, College of Veterinary Medicine, 2407 River Drive A122, Knoxville, TN 37996, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Huang HL, Yeh CN, Lee WY, Huang YC, Chang KW, Lin KJ, Tien SF, Su WC, Yang CH, Chen JT, Lin WJ, Fan SS, Yu CS. [123I]Iodooctyl fenbufen amide as a SPECT tracer for imaging tumors that over-express COX enzymes. Biomaterials 2013; 34:3355-65. [PMID: 23384791 DOI: 10.1016/j.biomaterials.2013.01.050] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Accepted: 01/09/2013] [Indexed: 11/27/2022]
Abstract
This study is concerned with the development of an agent for single photon emission computer tomography (SPECT) for imaging inflammation and tumor progression. [(123)I]Iodooctyl fenbufen amide ([(123)I]IOFA) was prepared from the precursor N-octyl-4-oxo-4-(4'-(trimethylstannyl)biphenyl-4-yl)butanamide with a radiochemical yield of 15%, specific activity of 37 GBq/μmol, and radiochemical purity of 95%. Analysis of the binding of [(123)I]IOFA to COX-1 and COX-2 enzymes by using HPLC and a gel filtration column showed a selectivity ratio of 1:1.3. An assay for the competitive inhibition of substrate transfer showed that IOFA exhibited a comparable IC(50) value compared to fenbufen. In the normal rat liver, a lower level and homogeneous pattern of [(123)I]IOFA radioactivity was observed by SPECT. In contrast, in the rat liver with thioacetamide-induced cholangiocarcinoma, a higher uptake and heterogeneous pattern of [(123)I]IOFA radioactivity was seen as hot spots in tumor lesions by SPECT imaging. Importantly, elevated COX-1 and COX-2 expressions from immunostaining were found in the bile ducts of tumor rats but not of normal rats. Therefore, [(123)I]IOFA was found to exhibit the potential for imaging tumors that over-express COX.
Collapse
Affiliation(s)
- Ho-Lien Huang
- Department of Biomedical Engineering and Environmental Sciences, National Tsing-Hua University, Hsinchu 300, Taiwan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Laube M, Neumann W, Scholz M, Lönnecke P, Crews B, Marnett LJ, Pietzsch J, Kniess T, Hey-Hawkins E. 2-Carbaborane-3-phenyl-1H-indoles--synthesis via McMurry reaction and cyclooxygenase (COX) inhibition activity. ChemMedChem 2013; 8:329-35. [PMID: 23303738 DOI: 10.1002/cmdc.201200455] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2012] [Revised: 12/03/2012] [Indexed: 01/03/2023]
Abstract
Cyclooxygenase-2 (COX-2) inhibitors have been the focus of medicinal chemistry efforts for years, and many compounds that exhibit high selectivity and affinity have been developed. As carbaboranes represent interesting pharmacophores as phenyl mimetics in drug development, this paper presents the synthesis of carbaboranyl derivatives of COX-2-selective 2,3-disubstituted indoles. Despite the lability of carbaboranes under reducing conditions, 2-carbaborane-3-phenyl-1H-indoles could be synthesized by McMurry cyclization of the corresponding amides. Whereas the meta-carbaboranyl-substituted derivatives lacked COX inhibitory activity, an ortho-carbaboranyl analogue was active, but showed a selectivity shift toward COX-1.
Collapse
Affiliation(s)
- Markus Laube
- Institut für Radiopharmazie, Helmholtz-Zentrum Dresden-Rossendorf, Bautzner Landstrasse 400, 01328 Dresden, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Bhardwaj A, Kaur J, Sharma SK, Huang Z, Wuest F, Knaus EE. Hybrid fluorescent conjugates of COX-2 inhibitors: Search for a COX-2 isozyme imaging cancer biomarker. Bioorg Med Chem Lett 2013. [DOI: 10.1016/j.bmcl.2012.10.131] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
33
|
Cekanova M, Uddin MJ, Legendre AM, Galyon G, Bartges JW, Callens A, Martin-Jimenez T, Marnett LJ. Single-dose safety and pharmacokinetic evaluation of fluorocoxib A: pilot study of novel cyclooxygenase-2-targeted optical imaging agent in a canine model. JOURNAL OF BIOMEDICAL OPTICS 2012; 17:116002. [PMID: 23117797 PMCID: PMC3484194 DOI: 10.1117/1.jbo.17.11.116002] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
We evaluated preclinical single-dose safety, pharmacokinetic properties, and specific uptake of the new optical imaging agent fluorocoxib A in dogs. Fluorocoxib A, N-[(5-carboxy-X-rhodaminyl)but-4-yl]-2-[1-(4-chlorobenzoyl)-5-methoxy-2-methyl-1H-indol-3-yl]acetamide, selectively binds and inhibits the cyclooxygenase-2 (COX-2) enzyme, which is overexpressed in many cancers. Safety pilot studies were performed in research dogs following intravenous (i.v.) administration of 0.1 and 1 mg/kg fluorocoxib A. Blood and urine samples collected three days after administration of each dose of fluorocoxib A revealed no evidence of toxicity, and no clinically relevant adverse events were noted on physical examination of exposed dogs over that time period. Pharmacokinetic parameters were assessed in additional research dogs from plasma collected at several time points after i.v. administration of fluorocoxib A using high-performance liquid chromatography analysis. The pharmacokinetic studies using 1 mg/kg showed a peak of fluorocoxib A (92±28 ng/ml) in plasma collected at 0.5 h. Tumor specific uptake of fluorocoxib A was demonstrated using a dog diagnosed with colorectal cancer expressing COX-2. Our data support the safe single-dose administration and in vivo efficacy of fluorocoxib A, suggesting a high potential for successful translation to clinical use as an imaging agent for improved tumor detection in humans.
Collapse
Affiliation(s)
- Maria Cekanova
- The University of Tennessee, College of Veterinary Medicine, Department of Small Animal Clinical Sciences, Knoxville, Tennessee 37996, USA.
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Marnett LJ. Inflammation and cancer: chemical approaches to mechanisms, imaging, and treatment. J Org Chem 2012; 77:5224-38. [PMID: 22515568 PMCID: PMC3375764 DOI: 10.1021/jo300214d] [Citation(s) in RCA: 228] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2012] [Indexed: 11/30/2022]
Abstract
The inflammatory response represents a first line of defense against invading pathogens and is important to human health. Chronic inflammation contributes to the etiology of multiple diseases, especially those associated with aging, such as cancer and cardiovascular disease. The chemistry of the inflammatory response is complex and involves the generation of highly reactive oxidants and electrophiles designed to kill the pathogen as well as the release of small molecule and protein mediators of intercellular signaling, chemotaxis, vasoconstriction, and wound-healing. Oxidation of unsaturated fatty acids--either nonenzymatic or enzymatic--contributes to the inflammatory response and associated cellular pathologies. The current perspective summarizes our research on unsaturated fatty acid oxidation in the context of inflammation and cancer. In addition to understanding the consequences of DNA and protein modification by lipid electrophiles, our research has focused on the development of molecularly targeted agents to image and treat cancer.
Collapse
Affiliation(s)
- Lawrence J Marnett
- A.B. Hancock Jr. Memorial Laboratory for Cancer Research, Department of Biochemistry, Vanderbilt Institute of Chemical Biology, Center in Molecular Toxicology, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee 37232-0146, USA.
| |
Collapse
|
35
|
Abstract
Development of imaging agents that can be used broadly for early detection of neoplasia at various tissue sites and at various stages of disease and that also can assess states of minimal residual disease would have tremendous utility in the diagnosis and management of cancer. In a series of articles culminating with a report in this issue of the journal (beginning on page 1536), Uddin and colleagues show their ability to systemically target the enzyme COX-2 with imaging probes that will serve as agents for early detection, risk assessment, prognosis, and intervention outcome measures. These probes will enable the detection and localization of regions of inflammation and a wide variety of premalignant lesions and cancers, with utility in monitoring the effects of cancer prevention and therapy.
Collapse
Affiliation(s)
- David A Ostrov
- Department of Pediatrics, University of Florida College of Medicine, Gainesville, USA
| | | |
Collapse
|
36
|
Abstract
Lung cancer prevention and early detection, which have fallen on hard times for more than the past 20 years, seem to have turned a corner toward better times ahead. Exciting new results of randomized controlled trials that targeted the arachidonic acid pathway, including a celecoxib trial reported by Mao and colleagues in this issue of the journal (beginning on page 984) and a trial of the prostacyclin analog iloprost, complement recently reported 20%-30% lung cancer mortality reductions, either with aspirin in targeting the arachidonic acid pathway or with computed tomography screening. The new results show encouraging activity personalized to former smokers and/or people expressing predictive biomarkers. These trials and technological advances in molecular profiling and imaging herald substantial clinical advances on the horizon of this field.
Collapse
Affiliation(s)
- Fadlo R Khuri
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, 1365 C Clifton Road, NE, Atlanta, GA 30322, USA.
| |
Collapse
|
37
|
Uddin MJ, Crews BC, Ghebreselasie K, Huda I, Kingsley PJ, Ansari MS, Tantawy MN, Reese J, Marnett LJ. Fluorinated COX-2 inhibitors as agents in PET imaging of inflammation and cancer. Cancer Prev Res (Phila) 2011; 4:1536-45. [PMID: 21900596 DOI: 10.1158/1940-6207.capr-11-0120] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
COX-2 is a major contributor to the inflammatory response and cancer progression so it is an important target for prevention and therapy. COX-2 is absent or expressed at low levels in most epithelial cells but is found at high levels in inflammatory lesions, and many premalignant and malignant tumors. Thus, it is an attractive target for molecular imaging. We report a series of novel fluorinated imaging agents, derived from indomethacin or celecoxib that selectively inhibit COX-2. The most promising lead, compound 7, was a fluorinated derivative of celecoxib. Kinetic analysis revealed that this fluorinated compound is a slow, tight-binding inhibitor of COX-2 and exhibits minimal inhibitory activity against COX-1. Efficient incorporation of (18)F into compound 7 by radiochemical synthesis and intravenous injection provided sufficient signal for in vivo positron emission tomography (PET) imaging. Selective uptake of (18)F-7 was observed in inflamed rat paws compared with the noninflamed contralateral paws and uptake was blocked by pretreatment with the COX-2 inhibitor, celecoxib. Uptake of (18)F-7 was not observed when inflammation was induced in COX-2-null mice. In nude mice bearing both a COX-2-expressing human tumor xenograft (1483) and a COX-2-negative xenograft (HCT116), (18)F-7 selectively accumulated in the COX-2-expressing tumor. Accumulation was blocked by pretreatment of the animals with celecoxib. The in vitro and in vivo properties of compound 7 suggest it will be a useful probe for early detection of cancer and for evaluation of the COX-2 status of premalignant and malignant tumors.
Collapse
Affiliation(s)
- Md Jashim Uddin
- A.B. Hancock, Jr., Memorial Laboratory for Cancer Research, Department of Biochemistry, Chemistry and Pharmacology, Vanderbilt Institute of Chemical Biology, Center for Molecular Toxicology, TN, USA
| | | | | | | | | | | | | | | | | |
Collapse
|