1
|
Chen Z, Haider A, Chen J, Xiao Z, Gobbi L, Honer M, Grether U, Arnold SE, Josephson L, Liang SH. The Repertoire of Small-Molecule PET Probes for Neuroinflammation Imaging: Challenges and Opportunities beyond TSPO. J Med Chem 2021; 64:17656-17689. [PMID: 34905377 PMCID: PMC9094091 DOI: 10.1021/acs.jmedchem.1c01571] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Neuroinflammation is an adaptive response of the central nervous system to diverse potentially injurious stimuli, which is closely associated with neurodegeneration and typically characterized by activation of microglia and astrocytes. As a noninvasive and translational molecular imaging tool, positron emission tomography (PET) could provide a better understanding of neuroinflammation and its role in neurodegenerative diseases. Ligands to translator protein (TSPO), a putative marker of neuroinflammation, have been the most commonly studied in this context, but they suffer from serious limitations. Herein we present a repertoire of different structural chemotypes and novel PET ligand design for classical and emerging neuroinflammatory targets beyond TSPO. We believe that this Perspective will support multidisciplinary collaborations in academic and industrial institutions working on neuroinflammation and facilitate the progress of neuroinflammation PET probe development for clinical use.
Collapse
Affiliation(s)
- Zhen Chen
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, MA, 02114, United States
| | - Ahmed Haider
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, MA, 02114, United States
| | - Jiahui Chen
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, MA, 02114, United States
| | - Zhiwei Xiao
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, MA, 02114, United States
| | - Luca Gobbi
- Pharma Research and Early Development, F. Hoffmann-La Roche Ltd, CH-4070 Basel, Switzerland
| | - Michael Honer
- Pharma Research and Early Development, F. Hoffmann-La Roche Ltd, CH-4070 Basel, Switzerland
| | - Uwe Grether
- Pharma Research and Early Development, F. Hoffmann-La Roche Ltd, CH-4070 Basel, Switzerland
| | - Steven E. Arnold
- Department of Neurology and the Massachusetts Alzheimer’s Disease Research Center, Massachusetts General Hospital, Harvard Medical School, 114 16th Street, Charlestown, Massachusetts 02129, USA
| | - Lee Josephson
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, MA, 02114, United States
| | - Steven H. Liang
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, MA, 02114, United States
| |
Collapse
|
2
|
Ghazanfari N, van Waarde A, Dierckx RAJO, Doorduin J, de Vries EFJ. Is cyclooxygenase-1 involved in neuroinflammation? J Neurosci Res 2021; 99:2976-2998. [PMID: 34346520 PMCID: PMC9542093 DOI: 10.1002/jnr.24934] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 05/08/2021] [Accepted: 07/13/2021] [Indexed: 12/15/2022]
Abstract
Purpose: Reactive microglia are an important hallmark of neuroinflammation. Reactive microglia release various inflammatory mediators, such as cytokines, chemokines, and prostaglandins, which are produced by enzymes like cyclooxygenases (COX). The inducible COX‐2 subtype has been associated with inflammation, whereas the constitutively expressed COX‐1 subtype is generally considered as a housekeeping enzyme. However, recent evidence suggests that COX‐1 can also be upregulated and may play a prominent role in the brain during neuroinflammation. In this review, we summarize the evidence that supports this involvement of COX‐1. Methods: Five databases were used to retrieve relevant studies that addressed COX‐1 in the context of neuroinflammation. The search resulted in 32 articles, describing in vitro, in vivo, post mortem, and in vivo imaging studies that specifically investigated the COX‐1 isoform under such conditions. Results: Reviewed literature generally indicated that the overexpression of COX‐1 was induced by an inflammatory stimulus, which resulted in an increased production of prostaglandin E2. The pharmacological inhibition of COX‐1 was shown to suppress the induction of inflammatory mediators like prostaglandin E2. Positron emission tomography (PET) imaging studies in animal models confirmed the overexpression of COX‐1 during neuroinflammation. The same imaging method, however, could not detect any upregulation of COX‐1 in patients with Alzheimer's disease. Conclusion: Taken together, studies in cultured cells and living rodents suggest that COX‐1 is involved in neuroinflammation. Most postmortem studies on human brains indicate that the concentration of COX‐1‐expressing microglial cells is increased near sites of inflammation. However, evidence for the involvement of COX‐1 in neuroinflammation in the living human brain is still largely lacking.
Collapse
Affiliation(s)
- Nafiseh Ghazanfari
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Aren van Waarde
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Rudi A J O Dierckx
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Janine Doorduin
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Erik F J de Vries
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| |
Collapse
|
3
|
Confinement fluorescence effect (CFE): Lighting up life by enhancing the absorbed photon energy utilization efficiency of fluorophores. Coord Chem Rev 2021. [DOI: 10.1016/j.ccr.2021.213979] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
|
4
|
Dagallier C, Avry F, Touchefeu Y, Buron F, Routier S, Chérel M, Arlicot N. Development of PET Radioligands Targeting COX-2 for Colorectal Cancer Staging, a Review of in vitro and Preclinical Imaging Studies. Front Med (Lausanne) 2021; 8:675209. [PMID: 34169083 PMCID: PMC8217454 DOI: 10.3389/fmed.2021.675209] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 05/03/2021] [Indexed: 12/29/2022] Open
Abstract
Colorectal cancer (CRC) is the second most common cause of cancer death, making early diagnosis a major public health challenge. The role of inflammation in tumorigenesis has been extensively explored, and among the identified markers of inflammation, cyclooxygenase-2 (COX-2) expression seems to be linked to lesions with a poor prognosis. Until now, COX-2 expression could only be accessed by invasive methods, mainly by biopsy. Imaging techniques such as functional Positron Emission Tomography (PET) could give access to in vivo COX-2 expression. This could make the staging of the disease more accurate and would be of particular interest in the exploration of the first metastatic stages. In this paper, we review recent progress in the development of COX-2 specific PET tracers by comparing the radioligands' characteristics and highlighting the obstacles that remain to be overcome in order to achieve the clinical development of such a radiotracer, and its evaluation in the management of CRC.
Collapse
Affiliation(s)
- Caroline Dagallier
- Unité de Radiopharmacie, CHRU de Tours, Tours, France.,Inserm UMR1253, iBrain, Université de Tours, Tours, France
| | - François Avry
- Inserm UMR1253, iBrain, Université de Tours, Tours, France
| | - Yann Touchefeu
- CRCINA, INSERM, CNRS, Nantes University, Nantes, France.,Institut des Maladies de l'Appareil Digestif, University Hospital, Nantes, France
| | - Frédéric Buron
- ICOA, Université d'Orléans, UMR CNRS 7311, Orléans, France
| | | | - Michel Chérel
- CRCINA, INSERM, CNRS, Nantes University, Nantes, France
| | - Nicolas Arlicot
- Unité de Radiopharmacie, CHRU de Tours, Tours, France.,Inserm UMR1253, iBrain, Université de Tours, Tours, France.,INSERM CIC 1415, CHRU de Tours, Tours, France
| |
Collapse
|
5
|
Kumar JSD, Prabhakaran J, Molotkov A, Sattiraju A, Kim J, Doubrovin M, Mann JJ, Mintz A. Radiosynthesis and evaluation of [ 18F]FMTP, a COX-2 PET ligand. Pharmacol Rep 2020; 72:1433-1440. [PMID: 32632914 DOI: 10.1007/s43440-020-00124-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 06/22/2020] [Accepted: 06/25/2020] [Indexed: 01/07/2023]
Abstract
BACKGROUND The upregulation of cyclooxygenase-2 (COX-2) is involved in neuroinflammation associated with many neurological diseases as well as cancers of the brain. Outside the brain, inflammation and COX-2 induction contribute to the pathogenesis of pain, arthritis, acute allograft rejection, and in response to infections, tumors, autoimmune disorders, and injuries. Herein, we report the radiochemical synthesis and evaluation of [18F]6-fluoro-2-(4-(methylsulfonyl)phenyl)-N-(thiophen-2-ylmethyl)pyrimidin-4-amine ([18F]FMTP), a high-affinity COX-2 inhibitor, by cell uptake and PET imaging studies. METHODS The radiochemical synthesis of [18F]FMTP was optimized using chlorine to fluorine displacement method, by reacting [18F]fluoride/K222/K2CO3 with the precursor molecule. Cellular uptake studies of [18F]FMTP was performed in COX-2 positive BxPC3 and COX-2 negative PANC-1 cell lines with unlabeled FMTP as well as celecoxib to define specific binding agents. Dynamic microPET image acquisitionwas performed in anesthetized nude mice (n = 3), lipopolysaccharide (LPS) induced neuroinflammation mice (n = 4), and phosphate-buffered saline (PBS) administered control mice (n = 4) using a Trifoil microPET/CT for a scan period of 60 min. RESULTS A twofold higher binding of [18F]FMTP was found in COX-2 positive BxPC3 cells compared with COX-2 negative PANC-1 cells. The radioligand did not show specific binding to COX-2 negative PANC-1 cells. MicroPET imaging in wild-type mice indicated blood-brain barrier (BBB) penetration and fast washout of [18F]FMTP in the brain, likely due to the low constitutive COX-2 expression in the normal brain. In contrast, a ~ twofold higher uptake of the radioligand was found in LPS-induced mice brain than PBS treated control mice. CONCLUSIONS Specific binding to COX-2 in BxPC3 cell lines, BBB permeability, and increased brain uptake in neuroinflammation mice qualifies [18F]FMTP as a potential PET tracer for studying inflammation.
Collapse
Affiliation(s)
- J S Dileep Kumar
- Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, Manhattan, NY, USA.
| | - Jaya Prabhakaran
- Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, Manhattan, NY, USA.,Department of Psychiatry, Columbia University Medical Center, Manhattan, NY, USA
| | - Andrei Molotkov
- Department of Radiology, Columbia University Medical Center, Manhattan, NY, USA
| | - Anirudh Sattiraju
- Department of Radiology, Columbia University Medical Center, Manhattan, NY, USA
| | - Jongho Kim
- Department of Radiology, Columbia University Medical Center, Manhattan, NY, USA
| | - Mikhail Doubrovin
- Department of Radiology, Columbia University Medical Center, Manhattan, NY, USA
| | - J John Mann
- Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, Manhattan, NY, USA.,Department of Psychiatry, Columbia University Medical Center, Manhattan, NY, USA.,Department of Radiology, Columbia University Medical Center, Manhattan, NY, USA
| | - Akiva Mintz
- Department of Radiology, Columbia University Medical Center, Manhattan, NY, USA.
| |
Collapse
|
6
|
Yadav AK, Reinhardt CJ, Arango AS, Huff HC, Dong L, Malkowski MG, Das A, Tajkhorshid E, Chan J. An Activity-Based Sensing Approach for the Detection of Cyclooxygenase-2 in Live Cells. Angew Chem Int Ed Engl 2020; 59:3307-3314. [PMID: 31854058 PMCID: PMC7416425 DOI: 10.1002/anie.201914845] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Indexed: 01/05/2023]
Abstract
Cyclooxygenase-2 (COX-2) overexpression is prominent in inflammatory diseases, neurodegenerative disorders, and cancer. Directly monitoring COX-2 activity within its native environment poses an exciting approach to account for and illuminate the effect of the local environments on protein activity. Herein, we report the development of CoxFluor, the first activity-based sensing approach for monitoring COX-2 within live cells with confocal microscopy and flow cytometry. CoxFluor strategically links a natural substrate with a dye precursor to engage both the cyclooxygenase and peroxidase activities of COX-2. This catalyzes the release of resorufin and the natural product, as supported by molecular dynamics and ensemble docking. CoxFluor enabled the detection of oxygen-dependent changes in COX-2 activity that are independent of protein expression within live macrophage cells.
Collapse
Affiliation(s)
- Anuj K Yadav
- Department of Chemistry, Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Christopher J Reinhardt
- Department of Chemistry, Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Andres S Arango
- Center for Biophysics and Quantitative Biology, Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Hannah C Huff
- Department of Chemistry, Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Liang Dong
- Department of Structural Biology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, 14203, USA
| | - Michael G Malkowski
- Department of Structural Biology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, 14203, USA
| | - Aditi Das
- Center for Biophysics and Quantitative Biology, Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Emad Tajkhorshid
- Department of Chemistry, Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Center for Biophysics and Quantitative Biology, Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Jefferson Chan
- Department of Chemistry, Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| |
Collapse
|
7
|
Yadav AK, Reinhardt CJ, Arango AS, Huff HC, Dong L, Malkowski MG, Das A, Tajkhorshid E, Chan J. An Activity‐Based Sensing Approach for the Detection of Cyclooxygenase‐2 in Live Cells. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.201914845] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Affiliation(s)
- Anuj K. Yadav
- Department of Chemistry Beckman Institute for Advanced Science and Technology University of Illinois at Urbana-Champaign Urbana IL 61801 USA
| | - Christopher J. Reinhardt
- Department of Chemistry Beckman Institute for Advanced Science and Technology University of Illinois at Urbana-Champaign Urbana IL 61801 USA
| | - Andres S. Arango
- Center for Biophysics and Quantitative Biology Department of Biochemistry University of Illinois at Urbana-Champaign Urbana IL 61801 USA
| | - Hannah C. Huff
- Department of Chemistry Beckman Institute for Advanced Science and Technology University of Illinois at Urbana-Champaign Urbana IL 61801 USA
| | - Liang Dong
- Department of Structural Biology Jacobs School of Medicine and Biomedical Sciences University at Buffalo Buffalo NY 14203 USA
| | - Michael G. Malkowski
- Department of Structural Biology Jacobs School of Medicine and Biomedical Sciences University at Buffalo Buffalo NY 14203 USA
| | - Aditi Das
- Center for Biophysics and Quantitative Biology Department of Biochemistry University of Illinois at Urbana-Champaign Urbana IL 61801 USA
- Department of Comparative Biosciences University of Illinois at Urbana-Champaign Urbana IL 61801 USA
| | - Emad Tajkhorshid
- Department of Chemistry Beckman Institute for Advanced Science and Technology University of Illinois at Urbana-Champaign Urbana IL 61801 USA
- Center for Biophysics and Quantitative Biology Department of Biochemistry University of Illinois at Urbana-Champaign Urbana IL 61801 USA
| | - Jefferson Chan
- Department of Chemistry Beckman Institute for Advanced Science and Technology University of Illinois at Urbana-Champaign Urbana IL 61801 USA
| |
Collapse
|
8
|
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
|
9
|
Carpinelli A, Rainone P, Belloli S, Reale A, Cappelli A, Germano G, Murtaj V, Coliva A, Di Grigoli G, Valeri A, Gilardi MC, Gianolli L, Anzini M, Moresco RM. Radiosynthesis and Preclinical Evaluation of 11C-VA426, a Cyclooxygenase-2 Selective Ligand. CONTRAST MEDIA & MOLECULAR IMAGING 2019; 2019:5823261. [PMID: 31598112 PMCID: PMC6778861 DOI: 10.1155/2019/5823261] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 05/21/2019] [Accepted: 06/18/2019] [Indexed: 12/14/2022]
Abstract
Cyclooxygenase-2 (COX-2) is involved in the inflammatory response, and its recurrent overexpression in cancers as well as in neurodegenerative disorders has made it an important target for therapy. For this reason, noninvasive imaging of COX-2 expression may represent an important diagnostic tool. In this work, a COX-2 inhibitor analogue, VA426 [1-(4-fluorophenyl)-3-(2-methoxyethyl)-2-methyl-5-(4-(methylsulfonil)phenyl)-1H-pyrrole], was synthesized and radiolabelled with the 11C radioisotope. The ex vivo biodistribution profile of 11C-VA426 was evaluated in the brain and periphery of healthy rats and mice and in brain and periphery of inflammation models, based on the administration of LPS. 11C-VA426 synthesis with the tBuOK base showed optimal radiochemical yield (15 ± 2%) based on triflate activity, molar activity (range 37-148 GBq/μmol), and radiochemical purity (>95%). Ex vivo biodistribution studies showed a fast uptake of radioactivity but a rapid washout, except in regions expressing COX-2 (lungs, liver, and kidney) both in rats and in mice, with maximum values at 30 and 10 minutes p.i., respectively. LPS administration did not show significant effect on radioactivity accumulation. Celecoxib competition experiments performed in rats and mice treated with LPS produced a general target unrelated reduction of radioactivity concentration in all peripheral tissues and brain areas examined. Finally, in agreement with the negative results obtained from biodistribution experiments, radiometabolites analysis revealed that 11C-VA426 is highly unstable in vivo. This study indicates that the compound 11C-VA426 is not currently suitable to be used as radiopharmaceutical for PET imaging. This family of compounds needs further implementation in order to improve in vivo stability.
Collapse
Affiliation(s)
- Assunta Carpinelli
- Institute of Molecular Bioimaging and Physiology of CNR, 20090 Segrate, Italy
- PET and Nuclear Medicine Unit, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Paolo Rainone
- PET and Nuclear Medicine Unit, San Raffaele Scientific Institute, 20132 Milan, Italy
- Doctorate School of Molecular and Translational Medicine, University of Milan, 20122 Milan, Italy
| | - Sara Belloli
- Institute of Molecular Bioimaging and Physiology of CNR, 20090 Segrate, Italy
- PET and Nuclear Medicine Unit, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Annalisa Reale
- Department of Biotechnology, Chemistry, and Pharmacy, DoE 2018–2022, University of Siena, 53100 Siena, Italy
| | - Andrea Cappelli
- Department of Biotechnology, Chemistry, and Pharmacy, DoE 2018–2022, University of Siena, 53100 Siena, Italy
| | - Giuliani Germano
- Department of Biotechnology, Chemistry, and Pharmacy, DoE 2018–2022, University of Siena, 53100 Siena, Italy
| | - Valentina Murtaj
- Institute of Molecular Bioimaging and Physiology of CNR, 20090 Segrate, Italy
- PET and Nuclear Medicine Unit, San Raffaele Scientific Institute, 20132 Milan, Italy
- PhD Program in Neuroscience, Department of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
| | - Angela Coliva
- PET and Nuclear Medicine Unit, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Giuseppe Di Grigoli
- Institute of Molecular Bioimaging and Physiology of CNR, 20090 Segrate, Italy
- PET and Nuclear Medicine Unit, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Angela Valeri
- Department of Biotechnology, Chemistry, and Pharmacy, DoE 2018–2022, University of Siena, 53100 Siena, Italy
| | - Maria Carla Gilardi
- Institute of Molecular Bioimaging and Physiology of CNR, 20090 Segrate, Italy
| | - Luigi Gianolli
- PET and Nuclear Medicine Unit, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Maurizio Anzini
- Department of Biotechnology, Chemistry, and Pharmacy, DoE 2018–2022, University of Siena, 53100 Siena, Italy
| | - Rosa Maria Moresco
- Institute of Molecular Bioimaging and Physiology of CNR, 20090 Segrate, Italy
- PET and Nuclear Medicine Unit, San Raffaele Scientific Institute, 20132 Milan, Italy
- Department of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
| |
Collapse
|
10
|
Cortes-Salva MY, Shrestha S, Singh P, Morse CL, Jenko KJ, Montero Santamaria JA, Zoghbi SS, Innis RB, Pike VW. 2-(4-Methylsulfonylphenyl)pyrimidines as Prospective Radioligands for Imaging Cyclooxygenase-2 with PET-Synthesis, Triage, and Radiolabeling. Molecules 2018; 23:molecules23112850. [PMID: 30400142 PMCID: PMC6278313 DOI: 10.3390/molecules23112850] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 10/26/2018] [Accepted: 10/27/2018] [Indexed: 11/23/2022] Open
Abstract
Cyclooxygenase 2 (COX-2) is an inducible enzyme responsible for the conversion of arachidonic acid into the prostaglandins, PGG2 and PGH2. Expression of this enzyme increases in inflammation. Therefore, the development of probes for imaging COX-2 with positron emission tomography (PET) has gained interest because they could be useful for the study of inflammation in vivo, and for aiding anti-inflammatory drug development targeting COX-2. Nonetheless, effective PET radioligands are still lacking. We synthesized eleven COX-2 inhibitors based on a 2(4-methylsulfonylphenyl)pyrimidine core from which we selected three as prospective PET radioligands based on desirable factors, such as high inhibitory potency for COX-2, very low inhibitory potency for COX-1, moderate lipophilicity, and amenability to labeling with a positron-emitter. These inhibitors, namely 6-methoxy-2-(4-(methylsulfonyl)phenyl-N-(thiophen-2ylmethyl)pyrimidin-4-amine (17), the 6-fluoromethyl analogue (20), and the 6-(2-fluoroethoxy) analogue (27), were labeled in useful yields and with high molar activities by treating the 6-hydroxy analogue (26) with [11C]iodomethane, [18F]2-fluorobromoethane, and [d2-18F]fluorobromomethane, respectively. [11C]17, [18F]20, and [d2-18F]27 were readily purified with HPLC and formulated for intravenous injection. These methods allow these radioligands to be produced for comparative evaluation as PET radioligands for measuring COX-2 in healthy rhesus monkey and for assessing their abilities to detect inflammation.
Collapse
Affiliation(s)
- Michelle Y Cortes-Salva
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Drive, Bethesda, MD 20892, USA.
| | - Stal Shrestha
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Drive, Bethesda, MD 20892, USA.
| | - Prachi Singh
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Drive, Bethesda, MD 20892, USA.
| | - Cheryl L Morse
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Drive, Bethesda, MD 20892, USA.
| | - Kimberly J Jenko
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Drive, Bethesda, MD 20892, USA.
| | - Jose A Montero Santamaria
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Drive, Bethesda, MD 20892, USA.
| | - Sami S Zoghbi
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Drive, Bethesda, MD 20892, USA.
| | - Robert B Innis
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Drive, Bethesda, MD 20892, USA.
| | - Victor W Pike
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Drive, Bethesda, MD 20892, USA.
| |
Collapse
|
11
|
Ren SZ, Wang ZC, Zhu XH, Zhu D, Li Z, Shen FQ, Duan YT, Cao H, Zhao J, Zhu HL. Design and biological evaluation of novel hybrids of 1, 5-diarylpyrazole and Chrysin for selective COX-2 inhibition. Bioorg Med Chem 2018; 26:4264-4275. [DOI: 10.1016/j.bmc.2018.07.022] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 06/28/2018] [Accepted: 07/12/2018] [Indexed: 12/20/2022]
|
12
|
Kniess T, Laube M, Steinbach J. “Hydrous 18 F-fluoroethylation” – Leaving off the azeotropic drying. Appl Radiat Isot 2017; 127:260-268. [DOI: 10.1016/j.apradiso.2017.06.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 06/07/2017] [Accepted: 06/12/2017] [Indexed: 12/16/2022]
|
13
|
Gassner C, Neuber C, Laube M, Bergmann R, Kniess T, Pietzsch J. Development of a18F-labeled Diaryl-Substituted Dihydropyrrolo[3,2,1-hi]indole as Potential Probe for Functional Imaging of Cyclooxygenase-2 with PET. ChemistrySelect 2016. [DOI: 10.1002/slct.201601618] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Cemena Gassner
- Helmholtz-Zentrum Dresden-Rossendorf; Department of Radiopharmaceutical and Chemical Biology, Institute of Radiopharmaceutical Cancer Research; Bautzner Landstrasse 400 01328 Dresden Germany
- Department of Chemistry and Food Chemistry; Technische Universität Dresden; 01062 Dresden Germany
| | - Christin Neuber
- Helmholtz-Zentrum Dresden-Rossendorf; Department of Radiopharmaceutical and Chemical Biology, Institute of Radiopharmaceutical Cancer Research; Bautzner Landstrasse 400 01328 Dresden Germany
| | - Markus Laube
- Helmholtz-Zentrum Dresden-Rossendorf; Department of Radiopharmaceutical and Chemical Biology, Institute of Radiopharmaceutical Cancer Research; Bautzner Landstrasse 400 01328 Dresden Germany
| | - Ralf Bergmann
- Helmholtz-Zentrum Dresden-Rossendorf; Department of Radiopharmaceutical and Chemical Biology, Institute of Radiopharmaceutical Cancer Research; Bautzner Landstrasse 400 01328 Dresden Germany
| | - Torsten Kniess
- Helmholtz-Zentrum Dresden-Rossendorf; Department of Radiopharmaceutical and Chemical Biology, Institute of Radiopharmaceutical Cancer Research; Bautzner Landstrasse 400 01328 Dresden Germany
| | - Jens Pietzsch
- Helmholtz-Zentrum Dresden-Rossendorf; Department of Radiopharmaceutical and Chemical Biology, Institute of Radiopharmaceutical Cancer Research; Bautzner Landstrasse 400 01328 Dresden Germany
- Department of Chemistry and Food Chemistry; Technische Universität Dresden; 01062 Dresden Germany
| |
Collapse
|
14
|
Laube M, Kniess T, Pietzsch J. Development of Antioxidant COX-2 Inhibitors as Radioprotective Agents for Radiation Therapy-A Hypothesis-Driven Review. Antioxidants (Basel) 2016; 5:antiox5020014. [PMID: 27104573 PMCID: PMC4931535 DOI: 10.3390/antiox5020014] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 04/08/2016] [Accepted: 04/11/2016] [Indexed: 12/12/2022] Open
Abstract
Radiation therapy (RT) evolved to be a primary treatment modality for cancer patients. Unfortunately, the cure or relief of symptoms is still accompanied by radiation-induced side effects with severe acute and late pathophysiological consequences. Inhibitors of cyclooxygenase-2 (COX-2) are potentially useful in this regard because radioprotection of normal tissue and/or radiosensitizing effects on tumor tissue have been described for several compounds of this structurally diverse class. This review aims to substantiate the hypothesis that antioxidant COX-2 inhibitors are promising radioprotectants because of intercepting radiation-induced oxidative stress and inflammation in normal tissue, especially the vascular system. For this, literature reporting on COX inhibitors exerting radioprotective and/or radiosensitizing action as well as on antioxidant COX inhibitors will be reviewed comprehensively with the aim to find cross-points of both and, by that, stimulate further research in the field of radioprotective agents.
Collapse
Affiliation(s)
- Markus Laube
- Department of Radiopharmaceutical and Chemical Biology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Bautzner Landstrasse 400, Dresden D-01328, Germany.
| | - Torsten Kniess
- Department of Radiopharmaceutical and Chemical Biology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Bautzner Landstrasse 400, Dresden D-01328, Germany.
| | - Jens Pietzsch
- Department of Radiopharmaceutical and Chemical Biology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Bautzner Landstrasse 400, Dresden D-01328, Germany.
- Department of Chemistry and Food Chemistry, Technische Universität Dresden, Dresden D-01062, Germany.
| |
Collapse
|
15
|
Erfani M, Sharifzadeh S, Doroudi A, Shafiei M. Labeling and evaluation of99mTc-tricarbonyl-meloxicam as a preferential COX-2 inhibitor for inflammation imaging. J Labelled Comp Radiopharm 2016; 59:284-90. [DOI: 10.1002/jlcr.3396] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Revised: 02/14/2016] [Accepted: 03/03/2016] [Indexed: 11/10/2022]
Affiliation(s)
- Mostafa Erfani
- Radiation Application Research School; Nuclear Science and Technology Research Institute (NSTRI); P.O. Box: 14395-836, Tehran Iran
| | - Somayeh Sharifzadeh
- School of Pharmacy; Ahvaz Jundishapur University of Medical Sciences; P.O. Box: 61357-33184 Ahvaz Iran
| | - Alireza Doroudi
- School of Pharmacy; Ahvaz Jundishapur University of Medical Sciences; P.O. Box: 61357-33184 Ahvaz Iran
| | - Mohammad Shafiei
- Radiation Application Research School; Nuclear Science and Technology Research Institute (NSTRI); P.O. Box: 14395-836, Tehran Iran
| |
Collapse
|
16
|
Roughan JV, Bertrand HG, Isles HM. Meloxicam prevents COX-2-mediated post-surgical inflammation but not pain following laparotomy in mice. Eur J Pain 2016; 20:231-40. [PMID: 25908253 PMCID: PMC4728739 DOI: 10.1002/ejp.712] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/13/2015] [Indexed: 12/12/2022]
Abstract
BACKGROUND Inflammation is thought to be a major contributor to post-surgical pain, so non-steroidal anti-inflammatory drugs (NSAIDs) are commonly used analgesics. However, compared to rats, considerably less is known as to how successfully these prevent pain in mice. METHODS A fluorescent COX-2 selective probe was used for the first time to evaluate the post-surgical anti-inflammatory effects of meloxicam, and automated behaviour analyses (HomeCageScan; HCS), the Mouse Grimace Scale (MGS) and body weight changes to assess its pain-preventative properties. Groups of 8-9 BALB/c mice were subcutaneously injected with saline (0.3 mL) or meloxicam at (1, 5 or 20 mg/kg) 1 h before a 1.5-cm midline laparotomy. The probe or a control dye (2 mg/kg) was injected intravenously 3 h later. Imaging was used to quantify inflammation at 7, 24 and 48 h following surgery. HCS data and MGS scores were respectively obtained from video recordings and photographs before surgery and 24 h later. RESULTS Post-surgical inflammation was dose dependently reduced by meloxicam; with 5 or 20 mg/kg being most effective compared to saline. However, all mice lost weight, MGS scores increased and behavioural activity was reduced by surgery for at least 24 h with no perceivable beneficial effect of meloxicam on any of these potentially pain-associated changes. CONCLUSIONS Although meloxicam prevented inflammation, even large doses did not prevent post-laparotomy pain possibly arising due to a range of factors, including, but not limited to inflammation. MGS scoring can be applied by very naïve assessors and so should be effective for cage-side use.
Collapse
Affiliation(s)
- John V. Roughan
- Comparative Biology Centre, The Medical School, University of Newcastle, Newcastle upon Tyne, NE2 4HH, UK
| | - Henri G.M.J. Bertrand
- Comparative Biology Centre, The Medical School, University of Newcastle, Newcastle upon Tyne, NE2 4HH, UK
| | - Hannah M. Isles
- Comparative Biology Centre, The Medical School, University of Newcastle, Newcastle upon Tyne, NE2 4HH, UK
| |
Collapse
|
17
|
Qiu HY, Wang PF, Li Z, Ma JT, Wang XM, Yang YH, Zhu HL. Synthesis of dihydropyrazole sulphonamide derivatives that act as anti-cancer agents through COX-2 inhibition. Pharmacol Res 2015; 104:86-96. [PMID: 26723906 DOI: 10.1016/j.phrs.2015.12.025] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2015] [Revised: 12/20/2015] [Accepted: 12/20/2015] [Indexed: 12/13/2022]
Abstract
COX-2 has long been exploited in the treatment of inflammation and relief of pain; however, research increasingly suggests COX-2 inhibitors might possess potential benefits to thwart tumour processes. In the present study, we designed a series of novel COX-2 inhibitors based on analysis of known inhibitors combined with an in silico scaffold modification strategy. A docking simulation combined with a primary screen in vitro were performed to filter for the lead compound, which was then substituted, synthesized and evaluated by a variety of bioassays. Derivative 4d was identified as a potent COX-2 enzyme inhibitor and exerted an anticancer effect through COX-2 inhibition. Further investigation confirmed that 4d could induce A549 cell apoptosis and arrest the cell cycle at the G2/M phase. Moreover, treatment with 4d reduced A549 cell adhesive ability and COX-2 expression. The morphological variation of treated cells was also visualized by confocal microscopy. Overall, the biological profile of 4d suggests that this compound may be developed as a potential anticancer agent.
Collapse
Affiliation(s)
- Han-Yue Qiu
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210046, People's Republic of China
| | - Peng-Fei Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210046, People's Republic of China
| | - Zhen Li
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210046, People's Republic of China
| | - Jun-Ting Ma
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210046, People's Republic of China
| | - Xiao-Ming Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210046, People's Republic of China.
| | - Yong-Hua Yang
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210046, People's Republic of China.
| | - Hai-Liang Zhu
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210046, People's Republic of China.
| |
Collapse
|
18
|
Wang B, Fan J, Wang X, Zhu H, Wang J, Mu H, Peng X. A Nile blue based infrared fluorescent probe: imaging tumors that over-express cyclooxygenase-2. Chem Commun (Camb) 2015; 51:792-5. [DOI: 10.1039/c4cc08915d] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The free Niblue-C6-IMC exists in a folded conformation where fluorescence is quenched, and when it binds to COX-2 in the Golgi apparatus of cancer cells, it is forced to adopt the unfolded state, and then fluorescence is turned on. Niblue-C6-IMC was proved to specifically target COX-2 by native polyacrylamide gel electrophoresis analysis.
Collapse
Affiliation(s)
- Benhua Wang
- State Key Laboratory of Fine Chemicals
- Dalian University of Technology
- High-tech District
- China
| | - Jiangli Fan
- State Key Laboratory of Fine Chemicals
- Dalian University of Technology
- High-tech District
- China
| | - Xianwu Wang
- Department School of Life Science and Biotechnology
- Dalian University of Technology
- Ganjingzi District
- China
| | - Hao Zhu
- State Key Laboratory of Fine Chemicals
- Dalian University of Technology
- High-tech District
- China
| | - Jingyun Wang
- Department School of Life Science and Biotechnology
- Dalian University of Technology
- Ganjingzi District
- China
| | - Huiying Mu
- State Key Laboratory of Fine Chemicals
- Dalian University of Technology
- High-tech District
- China
| | - Xiaojun Peng
- State Key Laboratory of Fine Chemicals
- Dalian University of Technology
- High-tech District
- China
| |
Collapse
|
19
|
Zhang H, Fan J, Wang K, Li J, Wang C, Nie Y, Jiang T, Mu H, Peng X, Jiang K. Highly Sensitive Naphthalene-Based Two-Photon Fluorescent Probe for in Situ Real-Time Bioimaging of Ultratrace Cyclooxygenase-2 in Living Biosystems. Anal Chem 2014; 86:9131-8. [DOI: 10.1021/ac501944y] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Hua Zhang
- School
of Chemistry and Chemical Engineering, Henan Normal University, 46
Jianshe Road, Muye Zone, Xinxiang 453007, P.R. China
| | - Jiangli Fan
- State
Key Laboratory of Fine Chemicals, Dalian University of Technology, 2 Linggong Road, Hi-Tech Zone, Dalian 116024, P.R. China
| | - Kui Wang
- School
of Chemistry and Chemical Engineering, Henan Normal University, 46
Jianshe Road, Muye Zone, Xinxiang 453007, P.R. China
| | - Jing Li
- School
of Chemistry and Chemical Engineering, Henan Normal University, 46
Jianshe Road, Muye Zone, Xinxiang 453007, P.R. China
| | - Caixia Wang
- School
of Chemistry and Chemical Engineering, Henan Normal University, 46
Jianshe Road, Muye Zone, Xinxiang 453007, P.R. China
| | - Yamin Nie
- School
of Chemistry and Chemical Engineering, Henan Normal University, 46
Jianshe Road, Muye Zone, Xinxiang 453007, P.R. China
| | - Tao Jiang
- School
of Chemistry and Chemical Engineering, Henan Normal University, 46
Jianshe Road, Muye Zone, Xinxiang 453007, P.R. China
| | - Huiying Mu
- State
Key Laboratory of Fine Chemicals, Dalian University of Technology, 2 Linggong Road, Hi-Tech Zone, Dalian 116024, P.R. China
| | - Xiaojun Peng
- State
Key Laboratory of Fine Chemicals, Dalian University of Technology, 2 Linggong Road, Hi-Tech Zone, Dalian 116024, P.R. China
| | - Kai Jiang
- School
of Chemistry and Chemical Engineering, Henan Normal University, 46
Jianshe Road, Muye Zone, Xinxiang 453007, P.R. China
| |
Collapse
|
20
|
Szalóki G, Pozzo JL. Synthesis of Symmetrical and Nonsymmetrical Bisthienylcyclopentenes. Chemistry 2013; 19:11124-32. [DOI: 10.1002/chem.201301645] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
21
|
Wu C, Li F, Niu G, Chen X. PET imaging of inflammation biomarkers. Theranostics 2013; 3:448-66. [PMID: 23843893 PMCID: PMC3706689 DOI: 10.7150/thno.6592] [Citation(s) in RCA: 143] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2013] [Accepted: 05/24/2013] [Indexed: 01/04/2023] Open
Abstract
Inflammation plays a significant role in many disease processes. Development in molecular imaging in recent years provides new insight into the diagnosis and treatment evaluation of various inflammatory diseases and diseases involving inflammatory process. Positron emission tomography using (18)F-FDG has been successfully applied in clinical oncology and neurology and in the inflammation realm. In addition to glucose metabolism, a variety of targets for inflammation imaging are being discovered and utilized, some of which are considered superior to FDG for imaging inflammation. This review summarizes the potential inflammation imaging targets and corresponding PET tracers, and the applications of PET in major inflammatory diseases and tumor associated inflammation. Also, the current attempt in differentiating inflammation from tumor using PET is also discussed.
Collapse
|
22
|
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
|
23
|
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
|
24
|
Chu XP, Zhou QF, Zhao S, Ge FF, Fu M, Chen JP, Lu T. Synthesis and biological evaluation of 3-amino-2-pyrones as selective cyclooxygenase-1 (COX-1) inhibitors. CHINESE CHEM LETT 2013. [DOI: 10.1016/j.cclet.2013.01.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
|
25
|
Tietz O, Sharma SK, Kaur J, Way J, Marshall A, Wuest M, Wuest F. Synthesis of three 18F-labelled cyclooxygenase-2 (COX-2) inhibitors based on a pyrimidine scaffold. Org Biomol Chem 2013; 11:8052-64. [DOI: 10.1039/c3ob41935e] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
26
|
Mamat C, Mosch B, Neuber C, Köckerling M, Bergmann R, Pietzsch J. Fluorine-18 Radiolabeling and Radiopharmacological Characterization of a Benzodioxolylpyrimidine-based Radiotracer Targeting the Receptor Tyrosine Kinase EphB4. ChemMedChem 2012; 7:1991-2003. [DOI: 10.1002/cmdc.201200264] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2012] [Revised: 08/29/2012] [Indexed: 12/24/2022]
|
27
|
Kniess T, Laube M, Bergmann R, Sehn F, Graf F, Steinbach J, Wuest F, Pietzsch J. Radiosynthesis of a ¹⁸F-labeled 2,3-diarylsubstituted indole via McMurry coupling for functional characterization of cyclooxygenase-2 (COX-2) in vitro and in vivo. Bioorg Med Chem 2012; 20:3410-21. [PMID: 22560838 DOI: 10.1016/j.bmc.2012.04.022] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2012] [Revised: 04/04/2012] [Accepted: 04/10/2012] [Indexed: 11/18/2022]
Abstract
The radiosynthesis of 3-(4-[(18)F]fluorophenyl)-2-(4-methylsulfonylphenyl)-1H-indole [(18)F]-3 as potential PET radiotracer for functional characterization of cyclooxygenase-2 (COX-2) in vitro and in vivo is described. [(18)F]-3 was prepared by McMurry cyclization of a (18)F-labeled intermediate with low valent titanium and zinc via a two-step procedure in a remote controlled synthesizer unit including HPLC purification and solid phase extraction. In this way [(18)F]-3 was synthesized in 80 min synthesis time in 10% total decay corrected yield from [(18)F]fluoride in radiochemical purity >98% and a specific activity of 74-91 GBq/μmol (EOS). [(18)F]-3 was evaluated in vitro using pro-inflammatory stimulated THP-1 and COX-2 expressing tumor cell lines (FaDu, A2058, HT-29), where the radiotracer uptake was shown to be consistent with up regulated COX-2 expression. The stability of [(18)F]-3 was determined by incubation in rat whole blood and plasma in vitro and by metabolite analysis of arterial blood samples in vivo, showing with 75% of original compound after 60 min an acceptable high metabolic stability. However, no substantial tumor accumulation of [(18)F]-3 could be observed by dynamic small animal PET studies on HT-29 tumor-bearing mice in vivo. This may be due to the only moderate COX-1/COX-2 selectivity of 3 as demonstrated by both cellular and enzymatic cyclooxygenase inhibition assay in vitro. Nevertheless, the new approach first using McMurry cyclization in (18)F-chemistry gives access to (18)F-labeled diarylsubstituted heterocycles that hold promise as radiolabeled COX-2 inhibitors.
Collapse
Affiliation(s)
- Torsten Kniess
- Institute of Radiopharmacy, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Sharma SK, Al-Hourani BJ, Wuest M, Mane JY, Tuszynski J, Baracos V, Suresh M, Wuest F. Synthesis and evaluation of fluorobenzoylated di- and tripeptides as inhibitors of cyclooxygenase-2 (COX-2). Bioorg Med Chem 2012; 20:2221-6. [PMID: 22386983 DOI: 10.1016/j.bmc.2012.02.021] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2011] [Revised: 01/28/2012] [Accepted: 02/07/2012] [Indexed: 10/28/2022]
Abstract
A series of fluorobenzoylated di- and tripeptides as potential leads for the development of molecular probes for imaging of COX-2 expression was prepared according to standard Fmoc-based solid-phase peptide synthesis. All peptides were assessed for their COX-2 inhibitory potency and selectivity profile in a fluorescence-based COX binding assay. Within the series of 15 peptides tested, cysteine-containing peptides numbered 7, 8, 11 and 12, respectively, were the most potent COX-2 inhibitors possessing IC(50) values ranging from 5 to 85 μM. Fluorobenzoylated tripeptides 7 and 8 displayed some COX-2 selectivity (COX-2 selectivity index 2.1 and 1.6), whereas fluorobenzoylated dipeptides 11 and 12 were shown not to be COX-2 selective. Fluorbenzoylated tripeptide FB-Phe-Cys-Ser-OH was further used in molecular modeling docking studies to determine the binding mode within the active site of the COX-2 enzyme.
Collapse
|
29
|
Mohamed Eissa AAH, Soliman GAEH, Khataibeh MH. Design, Synthesis and Anti-inflammatory Activity of Structurally Simple Anthranilic Acid Congeners Devoid of Ulcerogenic Side Effects. Chem Pharm Bull (Tokyo) 2012; 60:1290-300. [DOI: 10.1248/cpb.c12-00516] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
30
|
Yamamoto Y, Toyohara J, Ishiwata K, Sano K, Yamamoto F, Mukai T, Maeda M. ¹¹C-labeled analogs of indomethacin esters and amides for brain cyclooxygenase-2 imaging: radiosynthesis, in vitro evaluation and in vivo characteristics in mice. Chem Pharm Bull (Tokyo) 2011; 59:938-46. [PMID: 21804236 DOI: 10.1248/cpb.59.938] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
There is great potential in the use of positron emission tomography (PET) and suitable radiotracers for the study of cyclooxygenase type 2 (COX-2) enzyme in living subjects. In the present study, we prepared and evaluated five ¹¹C-labeled ester and amide analogs derived from indomethacin as potential PET imaging agents for the in vivo visualization of the brain COX-2 enzyme. Five ¹¹C-labeled COX-2 inhibitors, with different lipophilicities and moderate COX-2 inhibitory activity, were prepared by treatment of the corresponding O-desmethyl precursors with [¹¹C]methyl triflate and purified by HPLC (radiochemical yields of 55-71%, radiochemical purity of >93%, and the specific activities of 22-331 GBq/µmol). In mice, radioactivity in the brain for all radiotracers was low, with very low brain-to-blood ratios. A clear inverse relationship was observed between brain uptake at 1 min postinjection and the lipophilicity (experimental log P₇.₄) of the studied ¹¹C-radiotracers. Pretreatment of mice with cyclosporine A to block P-glycoproteins caused a significant increase in brain uptake of radioactivity following injection of the ¹¹C-radiotracer compared to control. HPLC analysis showed that each radiotracer was rapidly metabolized, and a few metabolites, which were more polar than the original radiotracers, were found in both plasma and brain. No specific binding of the tracers towards the COX-2 enzyme in the brain was clearly revealed by in vivo blocking study. Further structural refinement of the tracer agent is necessary for better enhancement of brain uptake and for sufficient metabolic stability.
Collapse
Affiliation(s)
- Yumi Yamamoto
- Graduate School of Pharmaceutical Sciences, Kyushu University, Japan
| | | | | | | | | | | | | |
Collapse
|
31
|
Lu X, Zhang H, Li X, Chen G, Li QS, Luo Y, Ruan BF, Chen XW, Zhu HL. Design, synthesis and biological evaluation of pyridine acyl sulfonamide derivatives as novel COX-2 inhibitors. Bioorg Med Chem 2011; 19:6827-32. [DOI: 10.1016/j.bmc.2011.09.034] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2011] [Revised: 09/18/2011] [Accepted: 09/19/2011] [Indexed: 10/17/2022]
|
32
|
Animal tumor models for PET in drug development. Ann Nucl Med 2011; 25:717-31. [DOI: 10.1007/s12149-011-0531-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2011] [Accepted: 08/16/2011] [Indexed: 10/17/2022]
|
33
|
Gao M, Wang M, Miller KD, Zheng QH. Synthesis and preliminary in vitro biological evaluation of new carbon-11-labeled celecoxib derivatives as candidate PET tracers for imaging of COX-2 expression in cancer. Eur J Med Chem 2011; 46:4760-7. [PMID: 21640445 DOI: 10.1016/j.ejmech.2011.05.024] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2011] [Revised: 05/02/2011] [Accepted: 05/10/2011] [Indexed: 12/25/2022]
Abstract
The enzyme cyclooxygenase-2 (COX-2) is overexpressed in a variety of malignant tumors. This study was designed to develop new radiotracers for imaging of COX-2 in cancer using biomedical imaging technique positron emission tomography (PET). Carbon-11-labeled celecoxib derivatives, [(11)C]4a-c and [(11)C]8a-d, were prepared by O-[(11)C] methylation of their corresponding precursors using [(11)C]CH(3)OTf under basic conditions and isolated by a simplified solid-phase extraction (SPE) method in 52 ± 2% (n = 5) and 57 ± 3% (n = 5) radiochemical yields based on [(11)C]CO(2) and decay corrected to end of bombardment (EOB). The overall synthesis time from EOB was 23 min, the radiochemical purity was >99%, and the specific activity at end of synthesis (EOS) was 277.5 ± 92.5 GBq/μmol (n = 5). The IC(50) values to block COX-2 for known compounds celecoxib (4d), 4a and 4c were 40, 290 and 8 nM, respectively, and preliminary findings from in vitro biological assay indicated that the synthesized new compounds 4b and 8a-d display similar strong inhibitory effectiveness in the MDA-MB-435 human cancer cell line in comparison with the parent compound 4d. These results encourage further in vivo evaluation of carbon-11-labeled celecoxib derivatives as new potential PET radiotracers for imaging of COX-2 expression in cancer.
Collapse
Affiliation(s)
- Mingzhang Gao
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, 1345 West 16th Street, L3-208, Indianapolis, IN 46202, USA
| | | | | | | |
Collapse
|
34
|
Uddin MJ, Crews BC, Blobaum AL, Kingsley PJ, Gorden DL, McIntyre JO, Matrisian LM, Subbaramaiah K, Dannenberg AJ, Piston DW, Marnett LJ. Selective visualization of cyclooxygenase-2 in inflammation and cancer by targeted fluorescent imaging agents. Cancer Res 2010; 70:3618-27. [PMID: 20430759 PMCID: PMC2864539 DOI: 10.1158/0008-5472.can-09-2664] [Citation(s) in RCA: 142] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Effective diagnosis of inflammation and cancer by molecular imaging is challenging because of interference from nonselective accumulation of the contrast agents in normal tissues. Here, we report a series of novel fluorescence imaging agents that efficiently target cyclooxygenase-2 (COX-2), which is normally absent from cells, but is found at high levels in inflammatory lesions and in many premalignant and malignant tumors. After either i.p. or i.v. injection, these reagents become highly enriched in inflamed or tumor tissue compared with normal tissue and this accumulation provides sufficient signal for in vivo fluorescence imaging. Further, we show that only the intact parent compound is found in the region of interest. COX-2-specific delivery was unambiguously confirmed using animals bearing targeted deletions of COX-2 and by blocking the COX-2 active site with high-affinity inhibitors in both in vitro and in vivo models. Because of their high specificity, contrast, and detectability, these fluorocoxibs are ideal candidates for detection of inflammatory lesions or early-stage COX-2-expressing human cancers, such as those in the esophagus, oropharynx, and colon.
Collapse
Affiliation(s)
- Md. Jashim Uddin
- A.B. Hancock Jr. Memorial Laboratory for Cancer Research, Departments of Biochemistry, Chemistry, and Pharmacology, Vanderbilt Institute of Chemical Biology, Center in Molecular Toxicology, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville TN 37232-0146
| | - Brenda C. Crews
- A.B. Hancock Jr. Memorial Laboratory for Cancer Research, Departments of Biochemistry, Chemistry, and Pharmacology, Vanderbilt Institute of Chemical Biology, Center in Molecular Toxicology, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville TN 37232-0146
| | - Anna L. Blobaum
- A.B. Hancock Jr. Memorial Laboratory for Cancer Research, Departments of Biochemistry, Chemistry, and Pharmacology, Vanderbilt Institute of Chemical Biology, Center in Molecular Toxicology, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville TN 37232-0146
| | - Philip J. Kingsley
- A.B. Hancock Jr. Memorial Laboratory for Cancer Research, Departments of Biochemistry, Chemistry, and Pharmacology, Vanderbilt Institute of Chemical Biology, Center in Molecular Toxicology, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville TN 37232-0146
| | - D. Lee Gorden
- Department of Cancer Biology, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville TN 37232-6840
| | - J. Oliver McIntyre
- Department of Cancer Biology, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville TN 37232-6840
| | - Lynn M. Matrisian
- Department of Cancer Biology, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville TN 37232-6840
| | - Kotha Subbaramaiah
- Department of Medicine, Weil Medical College of Cornell University, New York NY 10021
| | - Andrew J. Dannenberg
- Department of Medicine, Weil Medical College of Cornell University, New York NY 10021
| | - David W. Piston
- Departments of Molecular Physiology and Biophysics and Physics and Astronomy, Vanderbilt University School of Medicine, Nashville TN 37232-0615
| | - Lawrence J. Marnett
- A.B. Hancock Jr. Memorial Laboratory for Cancer Research, Departments of Biochemistry, Chemistry, and Pharmacology, Vanderbilt Institute of Chemical Biology, Center in Molecular Toxicology, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville TN 37232-0146
| |
Collapse
|
35
|
General Method for the11C-Labeling of 2-Arylpropionic Acids and Their Esters: Construction of a PET Tracer Library for a Study of Biological Events Involved in COXs Expression. Chemistry 2010; 16:4250-8. [DOI: 10.1002/chem.200903044] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
36
|
Kuge Y, Obokata N, Kimura H, Katada Y, Temma T, Sugimoto Y, Aita K, Seki KI, Tamaki N, Saji H. Synthesis and evaluation of a radioiodinated lumiracoxib derivative for the imaging of cyclooxygenase-2 expression. Nucl Med Biol 2009; 36:869-76. [PMID: 19875043 DOI: 10.1016/j.nucmedbio.2009.07.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2009] [Revised: 07/14/2009] [Accepted: 07/26/2009] [Indexed: 12/25/2022]
Abstract
INTRODUCTION Despite extensive attempts to develop cyclooxygenase (COX)-2 imaging radiotracers, no suitable positron emission tomography (PET)/single photon emission computed tomography (SPECT) tracers are currently available for in vivo imaging of COX-2 expression. The aims of this study were to synthesize and evaluate a radioiodinated derivative of lumiracoxib, 2-[(2-fluoro-6-iodophenyl)-amino]-5-methylphenylacetic acid (FIMA), which is structurally distinct from other drugs in the class and has weakly acidic properties, as a SPECT tracer for imaging COX-2 expression. METHODS The COX inhibitory potency was assessed by measuring COX-catalyzed oxidation with hydrogen peroxide. Cell uptake characteristics of (125)I-FIMA were assessed in control and linterfero/interferon-gamma-stimulated macrophages. The biodistribution of (125)I-FIMA was determined by the ex vivo tissue counting method in rats. RESULTS The COX-2 inhibitory potency of FIMA (IC(50)=2.46 microM) was higher than that of indomethacin (IC(50)=20.9 microM) and was comparable to lumiracoxib (IC(50)=0.77 microM) and diclofenac (IC(50)=0.98 microM). The IC(50) ratio (COX-1/COX-2=182) indicated FIMA has a high isoform selectivity for COX-2. (125)I-FIMA showed a significantly higher accumulation in COX-2 induced macrophages than in control macrophages, which decreased with nonradioactive FIMA in a concentration dependent manner. The biodistribution study showed rapid clearance of (125)I-FIMA from the blood and most organs including the liver and kidneys. No significant in vivo deiodination was observed with radioiodinated FIMA. CONCLUSIONS FIMA showed high inhibitory potency and selectivity for COX-2. Radioiodinated FIMA showed specific accumulation into COX-2 induced macrophages, no significant in vivo deiodination and rapid blood clearance. Radioiodinated FIMA deserves further investigation as a SPECT radiopharmaceutical for imaging COX-2 expression.
Collapse
Affiliation(s)
- Yuji Kuge
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Abstract
Cyclooxygenase (COX; prostaglandin G/H synthase, EC 1.14.99.1) catalyzes the first two steps in the biosynthesis of prostaglandins (PGs). The two COX isoforms COX-1 and COX-2 are the targets of the widely used nonsteroidal anti-inflammatory drugs, indicating a role for these enzymes in pain, fever, inflammation, and tumorigenesis. The ubiquitous constitutive expression of COX-1 and inducible expression of COX-2 have led to the widely held belief that COX-1 produces homeostatic PGs, while PGs produced by COX-2 are primarily pathophysiological. However, recent discoveries call this paradigm into question and reveal as yet underappreciated functions for both enzymes. This review focuses on some of these new insights.
Collapse
|
38
|
Wuest F, Tang X, Kniess T, Pietzsch J, Suresh M. Synthesis and cyclooxygenase inhibition of various (aryl-1,2,3-triazole-1-yl)-methanesulfonylphenyl derivatives. Bioorg Med Chem 2009; 17:1146-51. [DOI: 10.1016/j.bmc.2008.12.032] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2008] [Revised: 12/12/2008] [Accepted: 12/14/2008] [Indexed: 11/29/2022]
|