1
|
Kumari N, Adhikari A, Bhagat S, Mishra AK, Tiwari AK. Benzoxazolone-based FITC-conjugated fluorescent probe for locating in-vivo expression level of translocator protein (TSPO) during lung inflammation. Mol Divers 2025:10.1007/s11030-025-11192-9. [PMID: 40259117 DOI: 10.1007/s11030-025-11192-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 04/07/2025] [Indexed: 04/23/2025]
Abstract
Translocator protein 18 kDa (TSPO) has been a salient target for probing and monitoring inflammation in the central nervous system (CNS) and peripheral systems. Leveraging our previously developed, TSPO specific, modified acetamidobenzoxazolone derivative, the present work describes the synthesis and development of an optical probe for lung inflammation imaging: 2-(3,6-dihydroxy-9H-xanthen-9-yl)-5-(3-(3-(2-(methyl(phenyl)amino)-2-oxoethyl)-2-oxo-2,3-dihydrobenzo[d]oxazol-5-yl)thioureido)benzoic acid (FITC-MBP). The FITC-MBP is prepared through facile methodology by conjugating MBP to fluorophore dye FITC. Spectral properties remained equivalent to FITC dye with absorption and emission wavelength at 486 and 520 nm, respectively. Cellular uptake studies established overexpression of TSPO in lipopolysaccharide (LPS)-induced inflammation in H1299 lung cells. Reduced mean fluorescence intensity (MFI) during blocking experiments with PK11195 in flow cytometry suggests the specificity of the fluorescent probe towards TSPO. In-vivo optical imaging analysis on LPS-induced lung-inflamed balb/c mice revealed major sequestration of FITC-MBP in the lungs compared to control at 25 min post-injection that significantly decreased on pretreatment with PK11195 due to competitive binding to TSPO. On ground of these findings, we believe the novel fluorescent probe (FITC-MBP) might be utilized to visualize the overexpressed TSPO.
Collapse
Affiliation(s)
- Neelam Kumari
- Division of Cyclotron and Radiopharmaceutical Sciences (DCRS), Institute of Nuclear Medicine and Allied Sciences (INMAS), Defence Research & Development Organization, Brig S K Mazumdar Road, Delhi, 110054, India
- Department of Chemistry, Sri Venkateswara College, University of Delhi, Delhi, India
| | - Anupriya Adhikari
- Department of Chemistry, Graphic Era Hill University, Dehradun, Uttarakhand, India
| | - Sunita Bhagat
- Organic Synthesis Research Laboratory, Department of Chemistry, A.R.S.D. College, University of Delhi, New Delhi, India
| | - Anil K Mishra
- Division of Cyclotron and Radiopharmaceutical Sciences (DCRS), Institute of Nuclear Medicine and Allied Sciences (INMAS), Defence Research & Development Organization, Brig S K Mazumdar Road, Delhi, 110054, India
| | - Anjani K Tiwari
- Division of Cyclotron and Radiopharmaceutical Sciences (DCRS), Institute of Nuclear Medicine and Allied Sciences (INMAS), Defence Research & Development Organization, Brig S K Mazumdar Road, Delhi, 110054, India.
- Department of Chemistry, Babasaheb Bhimrao Ambedkar University, Lucknow, Uttar Pradesh, India.
| |
Collapse
|
2
|
Rousseau C, Metz R, Kerdraon O, Ouldamer L, Boiffard F, Renaudeau K, Ferrer L, Vercouillie J, Doutriaux-Dumoulin I, Mouton A, Le Thiec M, Morel A, Rusu D, Santiago-Ribeiro MJ, Campion L, Arlicot N, Kraeber-Bodéré F. Pilot Feasibility Study: 18 F-DPA-714 PET/CT Macrophage Imaging in Triple-Negative Breast Cancers (EITHICS). Clin Nucl Med 2024; 49:701-708. [PMID: 38913962 DOI: 10.1097/rlu.0000000000005338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
ABSTRACT Tumor-associated macrophages are targets of interest in triple-negative breast cancer (TNBC). The translocator protein 18 kDa (TSPO) is a sensitive marker for macrophages and holds potential relevance in TNBC stratification. This pilot prospective study (EITHICS, NCT04320030) aimed to assess the potential of TSPO PET/CT imaging using 18 F-DPA-714 in primary TNBC, compared with immunohistochemistry, autoradiography, and TSPO polymorphism. PATIENTS AND METHODS Thirteen TNBC patients were included. They underwent TSPO genotyping (HAB, MAB, LAB), 18 F-FDG PET/CT, and breast MRI. Semiquantitative PET parameters were computed. VOIs were defined on the tumor lesion, healthy breast tissue, and pectoral muscle to obtain SUV, tumor-to-background ratio (TBR), and time-activity curves (TACs). Additionally, immunohistochemistry, 3 H-DPA-714, and 3 H-PK-11195 autoradiography were conducted. RESULTS The majority of TNBC tumors (11/13, 84%) had a preponderance of M2-polarized macrophages with a median proportion of 82% (range, 44%-94%). 18 F-DPA-714 PET/CT clearly identified TNBC tumors with an excellent TBR. Three distinct patterns of 18 F-DPA-714 TACs were identified, categorized as "above muscular," "equal to muscular," and "below muscular" with reference to the muscular background. For the "above muscular" group (2 HAB and 2 MAB), "equal muscular" group (3 HAB, 3 MAB, and 1 LAB), and "below muscular" group (1 LAB and 1 MAB), tumor TACs showed a 18 F-DPA-714 accumulation slope of 1.35, 0.62, and 0.22, respectively, and a median SUV mean of 4.02 (2.09-5.31), 1.66 (0.93-3.07), and 0.61 (0.43-1.02). CONCLUSIONS This study successfully demonstrated TNBC tumor targeting by 18 F-DPA-714 with an excellent TBR, allowing to stratify 3 patterns of uptake potentially influenced by the TSPO polymorphism status. Further studies in larger populations should be performed to evaluate the prognostic value of this new biomarker.
Collapse
Affiliation(s)
| | - Raphaël Metz
- From the ICO René Gauducheau, F-44800, Saint-Herblain, France
| | | | | | | | | | | | | | | | - Alexis Mouton
- From the ICO René Gauducheau, F-44800, Saint-Herblain, France
| | - Maelle Le Thiec
- From the ICO René Gauducheau, F-44800, Saint-Herblain, France
| | - Agnès Morel
- From the ICO René Gauducheau, F-44800, Saint-Herblain, France
| | - Daniela Rusu
- From the ICO René Gauducheau, F-44800, Saint-Herblain, France
| | | | | | | | | |
Collapse
|
3
|
Viviano M, Barresi E, Siméon FG, Costa B, Taliani S, Da Settimo F, Pike VW, Castellano S. Essential Principles and Recent Progress in the Development of TSPO PET Ligands for Neuroinflammation Imaging. Curr Med Chem 2022; 29:4862-4890. [PMID: 35352645 PMCID: PMC10080361 DOI: 10.2174/0929867329666220329204054] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 12/21/2021] [Accepted: 01/25/2022] [Indexed: 11/22/2022]
Abstract
The translocator protein 18kDa (TSPO) is expressed in the outer mitochondrial membrane and is implicated in several functions, including cholesterol transport and steroidogenesis. Under normal physiological conditions, TSPO is present in very low concentrations in the human brain but is markedly upregulated in response to brain injury and inflammation. This upregulation is strongly associated with activated microglia. Therefore, TSPO is particularly suited for assessing active gliosis associated with brain lesions following injury or disease. For over three decades, TSPO has been studied as a biomarker. Numerous radioligands for positron emission tomography (PET) that target TSPO have been developed for imaging inflammatory progression in the brain. Although [11C]PK11195, the prototypical first-generation PET radioligand, is still widely used for in vivo studies, mainly now as its single more potent R-enantiomer, it has severe limitations, including low sensitivity and poor amenability to quantification. Second-generation radioligands are characterized by higher TSPO specific signals but suffer from other drawbacks, such as sensitivity to the TSPO single nucleotide polymorphism (SNP) rs6971. Therefore, their applications in human studies have the burden of needing to genotype subjects. Consequently, recent efforts are focused on developing improved radioligands that combine the optimal features of the second generation with the ability to overcome the differences in binding affinities across the population. This review presents essential principles in the design and development of TSPO PET ligands and discusses prominent examples among the main chemotypes.
Collapse
Affiliation(s)
- Monica Viviano
- Department of Pharmacy, University of Salerno, 84084 Fisciano (SA), Italy
| | | | - Fabrice G. Siméon
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - Barbara Costa
- Department of Pharmacy, University of Pisa, 56126, Pisa, Italy
| | - Sabrina Taliani
- Department of Pharmacy, University of Pisa, 56126, Pisa, Italy
| | | | - Victor W. Pike
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - Sabrina Castellano
- Department of Pharmacy, University of Salerno, 84084 Fisciano (SA), Italy
| |
Collapse
|
4
|
Wongso H, Yamasaki T, Kumata K, Ono M, Higuchi M, Zhang MR, Fulham MJ, Katsifis A, Keller PA. Design, Synthesis, and Biological Evaluation of Novel Fluorescent Probes Targeting the 18-kDa Translocator Protein. ChemMedChem 2021; 16:1902-1916. [PMID: 33631047 DOI: 10.1002/cmdc.202000984] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 02/22/2021] [Indexed: 12/20/2022]
Abstract
A series of fluorescent probes from the 6-chloro-2-phenylimidazo[1,2-a]pyridine-3-yl acetamides ligands featuring the 7-nitro-2-oxa-1,3-diazol-4-yl (NBD) moiety has been synthesized and biologically evaluated for their fluorescence properties and for their binding affinity to the 18-kDa translocator protein (TSPO). Spectroscopic studies including UV/Vis absorption and fluorescence measurements showed that the synthesized fluorescent probes exhibit favorable spectroscopic properties, especially in nonpolar environments. In vitro fluorescence staining in brain sections from lipopolysaccharide (LPS)-injected mice revealed partial colocalization of the probes with the TSPO. The TSPO binding affinity of the probes was measured on crude mitochondrial fractions separated from rat brain homogenates in a [11 C]PK11195 radioligand binding assay. All the new fluorescent probes demonstrated moderate to high binding affinity to the TSPO, with affinity (Ki ) values ranging from 0.58 nM to 3.28 μM. Taking these data together, we propose that the new fluorescent probes could be used to visualize the TSPO.
Collapse
Affiliation(s)
- Hendris Wongso
- School of Chemistry and Molecular Bioscience, and Molecular Horizons, University of Wollongong, Wollongong, NSW, 2522, Australia.,Center for Applied Nuclear Science and Technology, National Nuclear Energy Agency, Bandung, 40132, Indonesia
| | - Tomoteru Yamasaki
- Department of Advanced Nuclear Medicine Sciences, National Institute of Radiological Sciences, Chiba, 263-8555, Japan
| | - Katsushi Kumata
- Department of Advanced Nuclear Medicine Sciences, National Institute of Radiological Sciences, Chiba, 263-8555, Japan
| | - Maiko Ono
- Department of Functional Brain Imaging Research, National Institutes for Quantum and Radiological Science and Technology, Chiba, 263-8555, Japan
| | - Makoto Higuchi
- Department of Functional Brain Imaging Research, National Institutes for Quantum and Radiological Science and Technology, Chiba, 263-8555, Japan
| | - Ming-Rong Zhang
- Department of Advanced Nuclear Medicine Sciences, National Institute of Radiological Sciences, Chiba, 263-8555, Japan
| | - Michael J Fulham
- Department of PET and Nuclear Medicine, Royal Prince Alfred Hospital, Camperdown, NSW, 2050, Australia
| | - Andrew Katsifis
- Department of PET and Nuclear Medicine, Royal Prince Alfred Hospital, Camperdown, NSW, 2050, Australia
| | - Paul A Keller
- School of Chemistry and Molecular Bioscience, and Molecular Horizons, University of Wollongong, Wollongong, NSW, 2522, Australia
| |
Collapse
|
5
|
Cohen AS, Li J, Hight MR, McKinley E, Fu A, Payne A, Liu Y, Zhang D, Xie Q, Bai M, Ayers GD, Tantawy MN, Smith JA, Revetta F, Washington MK, Shi C, Merchant N, Manning HC. TSPO-targeted PET and Optical Probes for the Detection and Localization of Premalignant and Malignant Pancreatic Lesions. Clin Cancer Res 2020; 26:5914-5925. [PMID: 32933996 DOI: 10.1158/1078-0432.ccr-20-1214] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 07/24/2020] [Accepted: 09/10/2020] [Indexed: 12/28/2022]
Abstract
PURPOSE Pancreatic cancer is among the most aggressive malignancies and is rarely discovered early. However, pancreatic "incidentalomas," particularly cysts, are frequently identified in asymptomatic patients through anatomic imaging for unrelated causes. Accurate determination of the malignant potential of cystic lesions could lead to life-saving surgery or spare patients with indolent disease undue risk. Current risk assessment of pancreatic cysts requires invasive sampling, with attendant morbidity and sampling errors. Here, we sought to identify imaging biomarkers of high-risk pancreatic cancer precursor lesions. EXPERIMENTAL DESIGN Translocator protein (TSPO) expression, which is associated with cholesterol metabolism, was evaluated in premalignant and pancreatic cancer lesions from human and genetically engineered mouse (GEM) tissues. In vivo imaging was performed with [18F]V-1008, a TSPO-targeted PET agent, in two GEM models. For image-guided surgery (IGS), V-1520, a TSPO ligand for near-IR optical imaging based upon the V-1008 pharmacophore, was developed and evaluated. RESULTS TSPO was highly expressed in human and murine pancreatic cancer. Notably, TSPO expression was associated with high-grade, premalignant intraductal papillary mucinous neoplasms (IPMNs) and pancreatic intraepithelial neoplasia (PanIN) lesions. In GEM models, [18F]V-1008 exhibited robust uptake in early pancreatic cancer, detectable by PET. Furthermore, V-1520 localized to premalignant pancreatic lesions and advanced tumors enabling real-time IGS. CONCLUSIONS We anticipate that combined TSPO PET/IGS represents a translational approach for precision pancreatic cancer care through discrimination of high-risk indeterminate lesions and actionable surgery.
Collapse
Affiliation(s)
- Allison S Cohen
- Vanderbilt Center for Molecular Probes, Vanderbilt University Medical Center, Nashville, Tennessee.,Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Jun Li
- Vanderbilt Center for Molecular Probes, Vanderbilt University Medical Center, Nashville, Tennessee.,Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Matthew R Hight
- Vanderbilt Center for Molecular Probes, Vanderbilt University Medical Center, Nashville, Tennessee.,Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Eliot McKinley
- Vanderbilt Center for Molecular Probes, Vanderbilt University Medical Center, Nashville, Tennessee.,Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, Tennessee.,Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee
| | - Allie Fu
- Vanderbilt Center for Molecular Probes, Vanderbilt University Medical Center, Nashville, Tennessee.,Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Adria Payne
- Vanderbilt Center for Molecular Probes, Vanderbilt University Medical Center, Nashville, Tennessee.,Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Yang Liu
- Vanderbilt Center for Molecular Probes, Vanderbilt University Medical Center, Nashville, Tennessee.,Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Dawei Zhang
- Vanderbilt Center for Molecular Probes, Vanderbilt University Medical Center, Nashville, Tennessee.,Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Qing Xie
- Vanderbilt Center for Molecular Probes, Vanderbilt University Medical Center, Nashville, Tennessee.,Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Mingfeng Bai
- Vanderbilt Center for Molecular Probes, Vanderbilt University Medical Center, Nashville, Tennessee.,Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, Tennessee.,Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, Tennessee.,Vanderbilt Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Gregory D Ayers
- Vanderbilt Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee.,Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Mohammed Noor Tantawy
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, Tennessee.,Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Jarrod A Smith
- Vanderbilt University Center for Structural Biology, Vanderbilt University, Nashville, Tennessee
| | - Frank Revetta
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - M Kay Washington
- Vanderbilt Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee.,Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Chanjuan Shi
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Nipun Merchant
- Vanderbilt Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - H Charles Manning
- Vanderbilt Center for Molecular Probes, Vanderbilt University Medical Center, Nashville, Tennessee. .,Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, Tennessee.,Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, Tennessee.,Vanderbilt Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee
| |
Collapse
|
6
|
Buck JR, Saleh S, Claus T, Lovly C, Hight MR, Nickels ML, Noor Tantawy M, Charles Manning H. N-[ 18F]-Fluoroacetylcrizotinib: A potentially potent and selective PET tracer for molecular imaging of non-small cell lung cancer. Bioorg Med Chem Lett 2020; 30:127257. [PMID: 32631505 PMCID: PMC7357882 DOI: 10.1016/j.bmcl.2020.127257] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 05/08/2020] [Accepted: 05/09/2020] [Indexed: 12/24/2022]
Abstract
N-[18F]fluoroacetylcrizotinib, a fluorine-18 labeled derivative of the first FDA approved tyrosine kinase inhibitor (TKI) for the treatment of Anaplastic lymphoma kinase (ALK)-rearranged non-small cell lung cancer (NSCLC), crizotinib, was successfully synthesized for use in positron emission tomography (PET). Sequential in vitro biological evaluation of fluoracetylcrizotinib and in vivo biodistribution studies of [18F]fluoroacetylcrizotinib demonstrated that the biological activity of the parent compound remained unchanged, with potent ALK kinase inhibition and effective tumor growth inhibition. These results show that [18F]fluoroacetylcrizotinib has the potential to be a promising PET ligand for use in NSCLC imaging. The utility of PET in this context provides a non-invasive, quantifiable method to inform on the pharmacokinetics of an ALK-inhibitor such as crizotinib prior to a clinical trial, as well as during a trial in the event of acquired drug resistance.
Collapse
Affiliation(s)
- Jason R Buck
- Vanderbilt Center for Molecular Probes, United States; Vanderbilt University, Institute of Imaging Science, United States; Vanderbilt University Medical Center, United States
| | - Samir Saleh
- Vanderbilt Center for Molecular Probes, United States; Vanderbilt University, Institute of Imaging Science, United States; Vanderbilt University Medical Center, United States
| | - Trey Claus
- Vanderbilt Center for Molecular Probes, United States; Vanderbilt University, Institute of Imaging Science, United States; Vanderbilt University Medical Center, United States
| | - Christine Lovly
- Vanderbilt University Medical Center, United States; Vanderbilt Ingram Cancer Center, United States; Department of Hematology and Oncology, Vanderbilt University Medical Center, United States
| | - Matthew R Hight
- Vanderbilt Center for Molecular Probes, United States; Vanderbilt University, Institute of Imaging Science, United States; Vanderbilt University Medical Center, United States
| | - Michael L Nickels
- Vanderbilt Center for Molecular Probes, United States; Vanderbilt University, Institute of Imaging Science, United States; Vanderbilt University Medical Center, United States; Mallinckrodt Institute of Radiology, Washington University School of Medicine, United States
| | - M Noor Tantawy
- Vanderbilt University, Institute of Imaging Science, United States; Vanderbilt University Medical Center, United States
| | - H Charles Manning
- Vanderbilt Center for Molecular Probes, United States; Vanderbilt University, Institute of Imaging Science, United States; Vanderbilt University Medical Center, United States; Department of Radiology, Vanderbilt University Medical Center, United States.
| |
Collapse
|
7
|
Tang D, Li J, Nickels ML, Huang G, Cohen AS, Manning HC. Preclinical Evaluation of a Novel TSPO PET Ligand 2-(7-Butyl-2-(4-(2-[ 18F]Fluoroethoxy)phenyl)-5-Methylpyrazolo[1,5-a]Pyrimidin-3-yl)-N,N-Diethylacetamide ( 18F-VUIIS1018A) to Image Glioma. Mol Imaging Biol 2019; 21:113-121. [PMID: 29869061 PMCID: PMC6953265 DOI: 10.1007/s11307-018-1198-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
PURPOSE There is an urgent need for the development of novel positron emission tomography (PET) tracers for glioma imaging. In this study, we developed a novel PET probe ([18F]VUIIS1018A) by targeting translocator protein (TSPO), an imaging biomarker for glioma. The purpose of this preclinical study was to evaluate this novel TSPO probe for glioma imaging. PROCEDURES In this study, we synthesized [19F]VUIIS1018A and the precursor for radiosynthesis of [18F]VUIIS1018A. TSPO binding affinity was confirmed using a radioligand competitive binding assay in C6 glioma cell lysate. Further, dynamic imaging studies were performed in rats using a microPET system. These studies include displacement and blocking studies for ligand reversibility and specificity evaluation, and compartment modeling of PET data for pharmacokinetic parameter measurement using metabolite-corrected arterial input functions and PMOD. RESULTS Compared to previously reported TSPO tracers including [18F]VUIIS1008 and [18F]DPA-714, the novel tracer [18F]VUIIS1018A demonstrated higher binding affinity and BPND. Pretreatment with the cold analog [19F]VUIIS1018A could partially block tumor accumulation of this novel tracer. Further, compartment modeling of this novel tracer also exhibited a greater tumor-to-background ratio, a higher tumor binding potential and a lower brain binding potential when compared with other TSPO probes, such as [18F]DPA-714 and [18F]VUIIS1008. CONCLUSIONS These studies illustrate that [18F]VUIIS1018A can serve as a promising TSPO PET tracer for glioma imaging and potentially imaging of other solid tumors.
Collapse
Affiliation(s)
- Dewei Tang
- Center for Molecular Imaging, Shanghai University of Medicine & Health Sciences, Shanghai, China
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Pudong New District, Shanghai, 200127, China
| | - Jun Li
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Center for Molecular Probes (CMP), Vanderbilt University Medical School, 1161 21st Ave. S., AA 1105 MCN, Nashville, TN, 37232-2310, USA
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Michael L Nickels
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Center for Molecular Probes (CMP), Vanderbilt University Medical School, 1161 21st Ave. S., AA 1105 MCN, Nashville, TN, 37232-2310, USA
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Gang Huang
- Center for Molecular Imaging, Shanghai University of Medicine & Health Sciences, Shanghai, China
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Pudong New District, Shanghai, 200127, China
| | - Allison S Cohen
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Center for Molecular Probes (CMP), Vanderbilt University Medical School, 1161 21st Ave. S., AA 1105 MCN, Nashville, TN, 37232-2310, USA
| | - H Charles Manning
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN, USA.
- Vanderbilt Center for Molecular Probes (CMP), Vanderbilt University Medical School, 1161 21st Ave. S., AA 1105 MCN, Nashville, TN, 37232-2310, USA.
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, USA.
- Program in Chemical and Physical Biology, Vanderbilt University Medical Center, Nashville, TN, USA.
- Vanderbilt Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA.
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA.
- Department of Neurosurgery, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
8
|
Tang D, Li J, Buck JR, Tantawy MN, Xia Y, Harp JM, Nickels ML, Meiler J, Manning HC. Evaluation of TSPO PET Ligands [ 18F]VUIIS1009A and [ 18F]VUIIS1009B: Tracers for Cancer Imaging. Mol Imaging Biol 2018; 19:578-588. [PMID: 27853987 DOI: 10.1007/s11307-016-1027-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
PURPOSE Positron emission tomography (PET) ligands targeting translocator protein (TSPO) are potential imaging diagnostics of cancer. In this study, we report two novel, high-affinity TSPO PET ligands that are 5,7 regioisomers, [18F]VUIIS1009A ([18F]3A) and [18F]VUIIS1009B ([18F]3B), and their initial in vitro and in vivo evaluation in healthy mice and glioma-bearing rats. PROCEDURES VUIIS1009A/B was synthesized and confirmed by X-ray crystallography. Interactions between TSPO binding pocket and novel ligands were evaluated and compared with contemporary TSPO ligands using 2D 1H-15N heteronuclear single quantum coherence (HSQC) spectroscopy. In vivo biodistribution of [18F]VUIIS1009A and [18F]VUIIS1009B was carried out in healthy mice with and without radioligand displacement. Dynamic PET imaging data were acquired simultaneously with [18F]VUIIS1009A/B injections in glioma-bearing rats, with binding reversibility and specificity evaluated by radioligand displacement. In vivo radiometabolite analysis was performed using radio-TLC, and quantitative analysis of PET data was performed using metabolite-corrected arterial input functions. Imaging was validated with histology and immunohistochemistry. RESULTS Both VUIIS1009A (3A) and VUIIS1009B (3B) were found to exhibit exceptional binding affinity to TSPO, with observed IC50 values against PK11195 approximately 500-fold lower than DPA-714. However, HSQC NMR suggested that VUIIS1009A and VUIIS1009B share a common binding pocket within mammalian TSPO (mTSPO) as DPA-714 and to a lesser extent, PK11195. [18F]VUIIS1009A ([18F]3A) and [18F]VUIIS1009B ([18F]3B) exhibited similar biodistribution in healthy mice. In rats bearing C6 gliomas, both [18F]VUIIS1009A and [18F]VUIIS1009B exhibited greater binding potential (k 3/k 4)in tumor tissue compared to [18F]DPA-714. Interestingly, [18F]VUIIS1009B exhibited significantly greater tumor uptake (V T) than [18F]VUIIS1009A, which was attributed primarily to greater plasma-to-tumor extraction efficiency. CONCLUSIONS The novel PET ligand [18F]VUIIS1009B exhibits promising characteristics for imaging glioma; its superiority over [18F]VUIIS1009A, a regioisomer, appears to be primarily due to improved plasma extraction efficiency. Continued evaluation of [18F]VUIIS1009B as a high-affinity TSPO PET ligand for precision medicine appears warranted.
Collapse
Affiliation(s)
- Dewei Tang
- Vanderbilt University Institute of Imaging Science (VUIIS), Vanderbilt University Medical Center, Nashville, TN, 37232, USA.,Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, 37232, USA.,Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China.,Shanghai Key Laboratory for Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China
| | - Jun Li
- Vanderbilt University Institute of Imaging Science (VUIIS), Vanderbilt University Medical Center, Nashville, TN, 37232, USA.,Interdisciplinary Materials Science Program, Department of Physics and Astronomy, Vanderbilt University, Nashville, TN, 37240, USA
| | - Jason R Buck
- Vanderbilt University Institute of Imaging Science (VUIIS), Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Mohamed Noor Tantawy
- Vanderbilt University Institute of Imaging Science (VUIIS), Vanderbilt University Medical Center, Nashville, TN, 37232, USA.,Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Yan Xia
- Center for Structural Biology (CSB), Vanderbilt University, Nashville, TN, 37205, USA.,Department of Chemistry, Vanderbilt University, Nashville, TN, 37235, USA
| | - Joel M Harp
- Department of Biochemistry, Vanderbilt University Medical Center, Nashville, TN, 37232, USA.,Department of Biological Sciences, Vanderbilt University, Nashville, TN, 37235, USA
| | - Michael L Nickels
- Vanderbilt University Institute of Imaging Science (VUIIS), Vanderbilt University Medical Center, Nashville, TN, 37232, USA.,Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Jens Meiler
- Center for Structural Biology (CSB), Vanderbilt University, Nashville, TN, 37205, USA.,Department of Chemistry, Vanderbilt University, Nashville, TN, 37235, USA.,Vanderbilt Institute of Chemical Biology (VICB), Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - H Charles Manning
- Vanderbilt University Institute of Imaging Science (VUIIS), Vanderbilt University Medical Center, Nashville, TN, 37232, USA. .,Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, 37232, USA. .,Department of Chemistry, Vanderbilt University, Nashville, TN, 37235, USA. .,Program in Chemical and Physical Biology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA. .,Vanderbilt-Ingram Cancer Center (VICC), Vanderbilt University Medical Center, Nashville, TN, 37232, USA. .,Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37232, USA. .,Department of Neurosurgery, Vanderbilt University Medical Center, Nashville, TN, 37232, USA.
| |
Collapse
|
9
|
Li J, Smith JA, Dawson ES, Fu A, Nickels ML, Schulte ML, Manning HC. Optimized Translocator Protein Ligand for Optical Molecular Imaging and Screening. Bioconjug Chem 2017; 28:1016-1023. [PMID: 28156095 PMCID: PMC12097374 DOI: 10.1021/acs.bioconjchem.6b00711] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Translocator protein (TSPO) is a validated target for molecular imaging of a variety of human diseases and disorders. Given its involvement in cholesterol metabolism, TSPO expression is commonly elevated in solid tumors, including glioma, colorectal cancer, and breast cancer. TSPO ligands capable of detection by optical imaging are useful molecular tracers for a variety of purposes that range from quantitative biology to drug discovery. Leveraging our prior optimization of the pyrazolopyrimidine TSPO ligand scaffold for cancer imaging, we report herein a new generation of TSPO tracers with superior binding affinity and suitability for optical imaging and screening. In total, seven candidate TSPO tracers were synthesized and vetted in this study; the most promising tracer identified (29, Kd = 0.19 nM) was the result of conjugating a high-affinity TSPO ligand to a fluorophore used routinely in biological sciences (FITC) via a functional carbon linker of optimal length. Computational modeling suggested that an n-alkyl linker of eight carbons in length allows for positioning of the bulky fluorophore distal to the ligand binding domain and toward the solvent interface, minimizing potential ligand-protein interference. Probe 29 was found to be highly suitable for in vitro imaging of live TSPO-expressing cells and could be deployed as a ligand screening and discovery tool. Competitive inhibition of probe 29 quantified by fluorescence and 3H-PK11195 quantified by traditional radiometric detection resulted in equivalent affinity data for two previously reported TSPO ligands. This study introduces the utility of TSPO ligand 29 for in vitro imaging and screening and provides a structural basis for the development of future TSPO imaging ligands bearing bulky signaling moieties.
Collapse
Affiliation(s)
- Jun Li
- Interdisciplinary Materials Science Program, Vanderbilt University, Nashville, Tennessee 37232, United States
- Vanderbilt University Institute of Imaging Science (VUIIS), Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
- Center for Molecular Probes, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
| | - Jarrod A. Smith
- Vanderbilt University Center for Structural Biology, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Eric S. Dawson
- Vanderbilt University Center for Structural Biology, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Allie Fu
- Vanderbilt University Institute of Imaging Science (VUIIS), Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
- Center for Molecular Probes, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
| | - Michael L. Nickels
- Vanderbilt University Institute of Imaging Science (VUIIS), Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
- Center for Molecular Probes, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
| | - Michael L. Schulte
- Vanderbilt University Institute of Imaging Science (VUIIS), Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
- Center for Molecular Probes, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
| | - H. Charles Manning
- Interdisciplinary Materials Science Program, Vanderbilt University, Nashville, Tennessee 37232, United States
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37232, United States
- Vanderbilt University Institute of Imaging Science (VUIIS), Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
- Center for Molecular Probes, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
- Department of Biochemistry, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
- Vanderbilt-Ingram Cancer Center (VICC), Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
- Department of Neurosurgery, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
| |
Collapse
|
10
|
Evaluation of PET Imaging Performance of the TSPO Radioligand [18F]DPA-714 in Mouse and Rat Models of Cancer and Inflammation. Mol Imaging Biol 2016; 18:127-34. [PMID: 26194010 PMCID: PMC4722075 DOI: 10.1007/s11307-015-0877-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Purpose Many radioligands have been explored for imaging the 18-kDa translocator protein (TSPO), a diagnostic and therapeutic target for inflammation and cancer. Here, we investigated the TSPO radioligand [18F]DPA-714 for positron emission tomography (PET) imaging of cancer and inflammation. Procedures [18F]DPA-714 PET imaging was performed in 8 mouse and rat models of breast and brain cancer and 4 mouse and rat models of muscular and bowel inflammation. Results [18F]DPA-714 showed different uptake levels in healthy organs and malignant tissues of mice and rats. Although high and displaceable [18F]DPA-714 binding is observed ex vivo, TSPO-positive PET imaging of peripheral lesions of cancer and inflammation in mice did not show significant lesion-to-background signal ratios. Slower [18F]DPA-714 metabolism and muscle clearance in mice compared to rats may explain the elevated background signal in peripheral organs in this species. Conclusion Although TSPO is an evolutionary conserved protein, inter- and intra-species differences call for further exploration of the pharmacological parameters of TSPO radioligands. Electronic supplementary material The online version of this article (doi:10.1007/s11307-015-0877-x) contains supplementary material, which is available to authorized users.
Collapse
|
11
|
Zhang S, Yang L, Ling X, Shao P, Wang X, Edwards WB, Bai M. Tumor mitochondria-targeted photodynamic therapy with a translocator protein (TSPO)-specific photosensitizer. Acta Biomater 2015; 28:160-170. [PMID: 26432436 DOI: 10.1016/j.actbio.2015.09.033] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Revised: 09/10/2015] [Accepted: 09/28/2015] [Indexed: 12/20/2022]
Abstract
Photodynamic therapy (PDT) has been proven to be a minimally invasive and effective therapeutic strategy for cancer treatment. It can be used alone or as a complement to conventional cancer treatments, such as surgical debulking and chemotherapy. The mitochondrion is an attractive target for developing novel PDT agents, as it produces energy for cells and regulates apoptosis. Current strategy of mitochondria targeting is mainly focused on utilizing cationic photosensitizers that bind to the negatively charged mitochondria membrane. However, such an approach is lack of selectivity of tumor cells. To minimize the damage on healthy tissues and improve therapeutic efficacy, an alternative targeting strategy with high tumor specificity is in critical need. Herein, we report a tumor mitochondria-specific PDT agent, IR700DX-6T, which targets the 18kDa mitochondrial translocator protein (TSPO). IR700DX-6T induced apoptotic cell death in TSPO-positive breast cancer cells (MDA-MB-231) but not TSPO-negative breast cancer cells (MCF-7). In vivo PDT study suggested that IR700DX-6T-mediated PDT significantly inhibited the growth of MDA-MB-231 tumors in a target-specific manner. These combined data suggest that this new TSPO-targeted photosensitizer has great potential in cancer treatment. STATEMENT OF SIGNIFICANCE Photodynamic therapy (PDT) is an effective and minimally invasive therapeutic technique for treating cancers. Mitochondrion is an attractive target for developing novel PDT agents, as it produces energy to cells and regulates apoptosis. Current mitochondria targeted photosensitizers (PSs) are based on cationic molecules, which interact with the negatively charged mitochondria membrane. However, such PSs are not specific for cancerous cells, which may result in unwanted side effects. In this study, we developed a tumor mitochondria-targeted PS, IR700DX-6T, which binds to translocator protein (TSPO). This agent effectively induced apoptosis in TSPO-positive cancer cells and significantly inhibited tumor growth in TSPO-positive tumor-bearing mice. These combined data suggest that IR700DX-6T could become a powerful tool in the treatment of multiple cancers that upregulate TSPO.
Collapse
Affiliation(s)
- Shaojuan Zhang
- Molecular Imaging Laboratory, Department of Radiology, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Ling Yang
- Molecular Imaging Laboratory, Department of Radiology, University of Pittsburgh, Pittsburgh, PA 15219, USA
- Department of Cellular and Genetic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Xiaoxi Ling
- Molecular Imaging Laboratory, Department of Radiology, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Pin Shao
- Molecular Imaging Laboratory, Department of Radiology, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Xiaolei Wang
- Molecular Imaging Laboratory, Department of Radiology, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - W Barry Edwards
- Molecular Imaging Laboratory, Department of Radiology, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Mingfeng Bai
- Molecular Imaging Laboratory, Department of Radiology, University of Pittsburgh, Pittsburgh, PA 15219, USA
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15261, USA
- University of Pittsburgh Cancer Institute, Pittsburgh, PA 15232, USA
| |
Collapse
|
12
|
The translocator protein as a potential molecular target for improved treatment efficacy in photodynamic therapy. Future Med Chem 2015; 6:775-92. [PMID: 24941872 DOI: 10.4155/fmc.14.37] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Since its serendipitous discovery over 30 years ago, the translocator protein (18 kDa) has been demonstrated to play an important role in a multitude of critical biological processes. Although implemented as a novel therapeutic and diagnostic tool for a variety of disease states, its most promising role is as a molecular target for anticancer treatments such as photodynamic therapy (PDT). This review gives an overview of the attempts made by researchers to design porphyrin-based photosensitizers for use as anticancer therapeutics in PDT as well as improved imaging agents for diagnostic purposes. With a better understanding of the structure and function of the translocator protein, the synthesis of porphyrins for use in PDT with optimum binding affinities will become ever more possible.
Collapse
|
13
|
Tang D, Nickels ML, Tantawy MN, Buck JR, Manning HC. Preclinical imaging evaluation of novel TSPO-PET ligand 2-(5,7-Diethyl-2-(4-(2-[(18)F]fluoroethoxy)phenyl)pyrazolo[1,5-a]pyrimidin-3-yl)-N,N-diethylacetamide ([ (18)F]VUIIS1008) in glioma. Mol Imaging Biol 2014; 16:813-20. [PMID: 24845529 PMCID: PMC4372299 DOI: 10.1007/s11307-014-0743-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
PURPOSE Translocator protein (TSPO) concentrations are elevated in glioma, suggesting a role for TSPO positron emission tomography (PET) imaging in this setting. In preclinical PET studies, we evaluated a novel, high-affinity TSPO PET ligand, [(18)F]VUIIS1008, in healthy mice and glioma-bearing rats. PROCEDURES Dynamic PET data were acquired simultaneously with [(18)F]VUIIS1008 injection, with binding reversibility and specificity evaluated in vivo by non-radioactive ligand displacement or blocking. Compartmental analysis of PET data was performed using metabolite-corrected arterial input functions. Imaging was validated with histology and immunohistochemistry. RESULTS [(18)F]VUIIS1008 exhibited rapid uptake in TSPO-rich organs. PET ligand uptake was displaceable with non-radioactive VUIIS1008 or PBR06 in mice. Tumor accumulation of [(18)F]VUIIS1008 was blocked by pretreatment with VUIIS1008 in rats. [(18)F]VUIIS1008 exhibited improved tumor-to-background ratio and higher binding potential in tumors compared to a structurally similar pyrazolopyrimidine TSPO ligand, [(18)F]DPA-714. CONCLUSIONS The PET ligand [(18)F]VUIIS1008 exhibits promising characteristics as a tracer for imaging glioma. Further translational studies appear warranted.
Collapse
Affiliation(s)
- Dewei Tang
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, Tennessee 37232
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, Tennessee 37232
| | - Michael L. Nickels
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, Tennessee 37232
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, Tennessee 37232
| | - M. Noor Tantawy
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, Tennessee 37232
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, Tennessee 37232
| | - Jason R. Buck
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, Tennessee 37232
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, Tennessee 37232
| | - H. Charles Manning
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, Tennessee 37232
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, Tennessee 37232
- Program in Chemical and Physical Biology, Vanderbilt University Medical Center, Nashville, Tennessee 37232
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee 37232
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37232
- Department of Neurosurgery, Vanderbilt University Medical Center, Nashville, Tennessee 37232
- Vanderbilt Institute of Chemical Biology, Vanderbilt University Medical Center, Nashville, Tennessee 37232
| |
Collapse
|
14
|
Cheung YY, Nickels ML, Tang D, Buck JR, Manning HC. Facile synthesis of SSR180575 and discovery of 7-chloro-N,N,5-trimethyl-4-oxo-3(6-[(18)F]fluoropyridin-2-yl)-3,5-dihydro-4H-pyridazino[4,5-b]indole-1-acetamide, a potent pyridazinoindole ligand for PET imaging of TSPO in cancer. Bioorg Med Chem Lett 2014; 24:4466-4471. [PMID: 25172419 PMCID: PMC4163096 DOI: 10.1016/j.bmcl.2014.07.091] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Revised: 07/29/2014] [Accepted: 07/31/2014] [Indexed: 10/24/2022]
Abstract
A novel synthesis of the translocator protein (TSPO) ligand 7-chloro-N,N,5-trimethyl-4-oxo-3-phenyl-3,5-dihydro-4H-pyridazino[4,5-b]indole-1-acetamide (SSR180575, 3) was achieved in four steps from commercially available starting materials. Focused structure-activity relationship development about the pyridazinoindole ring at the N3 position led to the discovery of 7-chloro-N,N,5-trimethyl-4-oxo-3(6-fluoropyridin-2-yl)-3,5-dihydro-4H-pyridazino[4,5-b]indole-1-acetamide (14), a novel ligand of comparable affinity. Radiolabeling with fluorine-18 ((18)F) yielded 7-chloro-N,N,5-trimethyl-4-oxo-3(6-[(18)F]fluoropyridin-2-yl)-3,5-dihydro-4H-pyridazino[4,5-b]indole-1-acetamide ([(18)F]-14) in high radiochemical yield and specific activity. In vivo studies of [(18)F]-14 revealed this agent as a promising probe for molecular imaging of glioma.
Collapse
Affiliation(s)
- Yiu-Yin Cheung
- Vanderbilt University Institute of Imaging Science (VUIIS), Vanderbilt University Medical Center, Nashville, TN 37232, United States
| | - Michael L Nickels
- Vanderbilt University Institute of Imaging Science (VUIIS), Vanderbilt University Medical Center, Nashville, TN 37232, United States; Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN 37232, United States
| | - Dewei Tang
- Vanderbilt University Institute of Imaging Science (VUIIS), Vanderbilt University Medical Center, Nashville, TN 37232, United States; Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN 37232, United States
| | - Jason R Buck
- Vanderbilt University Institute of Imaging Science (VUIIS), Vanderbilt University Medical Center, Nashville, TN 37232, United States; Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN 37232, United States
| | - H Charles Manning
- Vanderbilt University Institute of Imaging Science (VUIIS), Vanderbilt University Medical Center, Nashville, TN 37232, United States; Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN 37232, United States; Program in Chemical and Physical Biology, Vanderbilt University Medical Center, Nashville, TN 37232, United States; Vanderbilt-Ingram Cancer Center (VICC), Vanderbilt University Medical Center, Nashville, TN 37232, United States; Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37232, United States; Department of Neurosurgery, Vanderbilt University Medical Center, Nashville, TN 37232, United States
| |
Collapse
|
15
|
O'Brien ER, Kersemans V, Tredwell M, Checa B, Serres S, Soto MS, Gouverneur V, Leppert D, Anthony DC, Sibson NR. Glial activation in the early stages of brain metastasis: TSPO as a diagnostic biomarker. J Nucl Med 2014; 55:275-80. [PMID: 24434290 DOI: 10.2967/jnumed.113.127449] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
UNLABELLED Metastatic spread of cancer cells to the brain is associated with high mortality, primarily because current diagnostic tools identify only well-advanced metastases. Brain metastases have been shown to induce a robust glial response, including both astrocyte and microglial activation. On the basis of these findings, we hypothesized that this stromal response may provide a sensitive biomarker of tumor burden, in particular through the use of SPECT/PET imaging agents targeting the translocator protein (TSPO) that is upregulated on activated glia. Our goals, therefore, were first to determine the spatial and temporal profile of glial activation during early metastasis growth in vivo and second to assess the potential of the radiolabeled TSPO ligand (123)I-DPA-713 for early detection of brain metastases. METHODS Metastatic mouse mammary carcinoma 4T1-green fluorescent protein cells were injected either intracerebrally or intracardially into female BALB/c mice to induce brain metastases. Astrocyte and microglial activation was assessed immunohistochemically over a 28-d period, together with immunofluorescence detection of TSPO upregulation. Subsequently, SPECT imaging and autoradiography were used to determine in vivo binding of (123)I-DPA-713 at metastatic sites. RESULTS Dynamic astrocyte and microglial activation was evident throughout the early stages of tumor growth, with the extent of astrocyte activation correlating significantly with tumor size (P < 0.0001). Microglial activation appeared to increase more rapidly than astrocyte activation at the earlier time points, but by later time points the extent of activation was comparable between the glial cell types. Upregulation of TSPO expression was found on both glial populations. Both autoradiographic and in vivo SPECT data showed strong positive binding of (123)I-DPA-713 in the intracerebrally induced model of brain metastasis, which was significantly greater than that observed in controls (P < 0.05). (123)I-DPA-713 binding was also evident autoradiographically in the intracardially induced model of brain metastasis but with lower sensitivity because of smaller tumor size (∼ 100-μm diameter vs. ∼ 600-μm diameter in the intracerebral model). CONCLUSION These data suggest that the glial response to brain metastasis may provide a sensitive biomarker of tumor burden, with a tumor detection threshold lying between 100 and 600 μm in diameter. This approach could enable substantially earlier detection of brain metastases than the current clinical approach of gadolinium-enhanced MR imaging.
Collapse
Affiliation(s)
- Emma R O'Brien
- CR-United Kingdom/MRC Gray Institute for Radiation Oncology and Biology, Department of Oncology, University of Oxford, Churchill Hospital, Oxford, United Kingdom
| | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Samuelson LE, Anderson BM, Bai M, Dukes MJ, Hunt CR, Casey JD, Han Z, Papadopoulos V, Bornhop DJ. A self-internalizing mitochondrial TSPO targeting imaging probe for fluorescence, MRI and EM. RSC Adv 2014; 4:9003-9011. [PMID: 32051760 DOI: 10.1039/c3ra47161f] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Advances in probes for cellular imaging have driven discoveries in biology and medicine. Primarily, antibodies and small molecules have been made for contrast enhancement of specific proteins. The development of new dendrimer-based tools offers opportunities to tune cellular internalization and targeting, image multiple modalities in the same molecule and explore therapeutics. The translocator protein (TSPO) offers an ideal target to develop dendrimer tools because it is well characterized and implicated in a number of disease states. The TSPO-targeted dendrimers reported here, primarily ClPhIQ-PAMAM-Gd-Liss, are cell membrane permeable nanoparticles that enable labeling of TSPO and provide contrast in fluorescence, electron microscopy and magnetic resonance imaging. The molecular binding affinity for TSPO was found to be 0.51 μM, 3 times greater than the monomeric agents previously demonstrated in our laboratory. The relaxivity per Gd3+ of the ClPhIQ23-PAMAM-Gd18 dendrimer was 7.7 and 8.0 mM-1 s-1 for r 1 and r 2 respectively, approximately double that of the clinically used monomeric Gd3+ chelates. In vitro studies confirmed molecular selectively for labeling TSPO in the mitochondria of C6 rat glioma and MDA-MB-231 cell lines. Fluorescence co-registration with Mitotracker Green® and increased contrast of osmium-staining in electron microscopy confirmed mitochondrial labeling of these TSPO-targeted agents. Taken collectively these experiments demonstrate the versatility of conjugation of our PAMAM dendrimeric chemistry to allow multi-modality agents to be prepared. These agents target organelles and use complementary imaging modalities in vitro, potentially allowing disease mechanism studies with high sensitivity and high resolution techniques.
Collapse
Affiliation(s)
- Lynn E Samuelson
- Department of Chemistry, The Vanderbilt Institute for Chemical Biology and Vanderbilt-Ingram Cancer Center, Vanderbilt University, VU Station B 351822 Nashville, Tennessee 37235-1822, USA
| | - Bernard M Anderson
- Department of Chemistry, The Vanderbilt Institute for Chemical Biology and Vanderbilt-Ingram Cancer Center, Vanderbilt University, VU Station B 351822 Nashville, Tennessee 37235-1822, USA
| | - Mingfeng Bai
- Department of Chemistry, The Vanderbilt Institute for Chemical Biology and Vanderbilt-Ingram Cancer Center, Vanderbilt University, VU Station B 351822 Nashville, Tennessee 37235-1822, USA
| | - Madeline J Dukes
- Department of Chemistry, The Vanderbilt Institute for Chemical Biology and Vanderbilt-Ingram Cancer Center, Vanderbilt University, VU Station B 351822 Nashville, Tennessee 37235-1822, USA
| | - Colette R Hunt
- Department of Chemistry, The Vanderbilt Institute for Chemical Biology and Vanderbilt-Ingram Cancer Center, Vanderbilt University, VU Station B 351822 Nashville, Tennessee 37235-1822, USA
| | - Jonathon D Casey
- Department of Chemistry, The Vanderbilt Institute for Chemical Biology and Vanderbilt-Ingram Cancer Center, Vanderbilt University, VU Station B 351822 Nashville, Tennessee 37235-1822, USA
| | - Zeqiu Han
- Department of Biochemistry &Molecular and Ceilular Biology, Georgetown University Medical Center, BSB Room 315, 3900 Reservoir Road NW, Washington, DC 20057, USA
| | - Vassilios Papadopoulos
- The Research Institute of the McGill University Health Centre, Departments of Medicine, Biochemistry, and Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - Darryl J Bornhop
- Department of Chemistry, The Vanderbilt Institute for Chemical Biology and Vanderbilt-Ingram Cancer Center, Vanderbilt University, VU Station B 351822 Nashville, Tennessee 37235-1822, USA
| |
Collapse
|
17
|
Trapani A, Palazzo C, de Candia M, Lasorsa FM, Trapani G. Targeting of the Translocator Protein 18 kDa (TSPO): A Valuable Approach for Nuclear and Optical Imaging of Activated Microglia. Bioconjug Chem 2013; 24:1415-28. [DOI: 10.1021/bc300666f] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Adriana Trapani
- Department of Pharmacy and Drug
Sciences, University of Bari, Bari, 70125,
Italy
| | - Claudio Palazzo
- Department of Pharmacy and Drug
Sciences, University of Bari, Bari, 70125,
Italy
| | - Modesto de Candia
- Department of Pharmacy and Drug
Sciences, University of Bari, Bari, 70125,
Italy
| | | | - Giuseppe Trapani
- Department of Pharmacy and Drug
Sciences, University of Bari, Bari, 70125,
Italy
| |
Collapse
|
18
|
Kalk NJ, Owen DR, Tyacke RJ, Reynolds R, Rabiner EA, Lingford-Hughes AR, Parker CA. Are prescribed benzodiazepines likely to affect the availability of the 18 kDa translocator protein (TSPO) in PET studies? Synapse 2013; 67:909-12. [PMID: 23666806 DOI: 10.1002/syn.21681] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2012] [Revised: 04/13/2013] [Accepted: 05/03/2013] [Indexed: 11/07/2022]
Affiliation(s)
- N J Kalk
- Imperial College London, Centre for Neuropsychopharmacology, London, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
19
|
Tang D, McKinley ET, Hight MR, Uddin MI, Harp JM, Fu A, Nickels ML, Buck JR, Manning HC. Synthesis and structure-activity relationships of 5,6,7-substituted pyrazolopyrimidines: discovery of a novel TSPO PET ligand for cancer imaging. J Med Chem 2013; 56:3429-33. [PMID: 23521048 PMCID: PMC3648642 DOI: 10.1021/jm4001874] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Focused library synthesis and structure-activity relationship development of 5,6,7-substituted pyrazolopyrimidines led to the discovery of 2-(5,7-diethyl-2-(4-(2-fluoroethoxy)phenyl)pyrazolo[1,5-a]pyrimidin-3-yl)-N,N-diethylacetamide (6b), a novel translocator protein (TSPO) ligand exhibiting a 36-fold enhancement in affinity compared to another pyrazolopyrimidine-based TSPO ligand, 6a (DPA-714). Radiolabeling with fluorine-18 ((18)F) facilitated production of 2-(5,7-diethyl-2-(4-(2-[(18)F]fluoroethoxy)phenyl)pyrazolo[1,5-a]pyrimidin-3-yl)-N,N-diethylacetamide ((18)F-6b) in high radiochemical yield and specific activity. In vivo studies of (18)F-6b were performed which illuminated this agent as an improved probe for molecular imaging of TSPO-expressing cancers.
Collapse
Affiliation(s)
- Dewei Tang
- Vanderbilt University Institute of Imaging Science (VUIIS), Vanderbilt University Medical Center, Nashville, TN
- Program in Chemical and Physical Biology, Vanderbilt University Medical Center, Nashville, TN
| | - Eliot T. McKinley
- Vanderbilt University Institute of Imaging Science (VUIIS), Vanderbilt University Medical Center, Nashville, TN
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN
| | - Matthew R. Hight
- Vanderbilt University Institute of Imaging Science (VUIIS), Vanderbilt University Medical Center, Nashville, TN
- Interdisciplinary Materials Science Program, Department of Physics & Astronomy, Vanderbilt University, Nashville, TN
| | - Md. Imam Uddin
- Vanderbilt University Institute of Imaging Science (VUIIS), Vanderbilt University Medical Center, Nashville, TN
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN
| | - Joel M. Harp
- Department of Biochemistry, Vanderbilt University Medical Center, Nashville, TN
- Department of Biological Sciences, Vanderbilt University, Nashville, TN
| | - Allie Fu
- Vanderbilt University Institute of Imaging Science (VUIIS), Vanderbilt University Medical Center, Nashville, TN
| | - Michael L. Nickels
- Vanderbilt University Institute of Imaging Science (VUIIS), Vanderbilt University Medical Center, Nashville, TN
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN
| | - Jason R. Buck
- Vanderbilt University Institute of Imaging Science (VUIIS), Vanderbilt University Medical Center, Nashville, TN
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN
| | - H. Charles Manning
- Vanderbilt University Institute of Imaging Science (VUIIS), Vanderbilt University Medical Center, Nashville, TN
- Program in Chemical and Physical Biology, Vanderbilt University Medical Center, Nashville, TN
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN
- Vanderbilt Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN
- Department of Neurosurgery, Vanderbilt University Medical Center, Nashville, TN
| |
Collapse
|
20
|
Vasdev N, Green DE, Vines DC, McLarty K, McCormick PN, Moran MD, Houle S, Wilson AA, Reilly RM. Positron-Emission Tomography Imaging of the TSPO with [18F]FEPPA in a Preclinical Breast Cancer Model. Cancer Biother Radiopharm 2013; 28:254-9. [DOI: 10.1089/cbr.2012.1196] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Neil Vasdev
- PET Centre, Centre for Addiction and Mental Health, Toronto, Ontario
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - David E. Green
- STTARR Innovation Centre, Radiation Medicine Program, Princess Margaret Hospital, Toronto, Ontario, Canada
| | - Douglass C. Vines
- STTARR Innovation Centre, Radiation Medicine Program, Princess Margaret Hospital, Toronto, Ontario, Canada
- Department of Radiation Oncology, University of Toronto, Toronto, Ontario, Canada
| | - Kristin McLarty
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, Ontario, Canada
| | - Patrick N. McCormick
- PET Centre, Centre for Addiction and Mental Health, Toronto, Ontario
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Matthew D. Moran
- PET Centre, Centre for Addiction and Mental Health, Toronto, Ontario
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Sylvain Houle
- PET Centre, Centre for Addiction and Mental Health, Toronto, Ontario
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Alan A. Wilson
- PET Centre, Centre for Addiction and Mental Health, Toronto, Ontario
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Raymond M. Reilly
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, Ontario, Canada
- Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
- Department of Medical Imaging, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
21
|
Proulx ST, Luciani P, Alitalo A, Mumprecht V, Christiansen AJ, Huggenberger R, Leroux JC, Detmar M. Non-invasive dynamic near-infrared imaging and quantification of vascular leakage in vivo. Angiogenesis 2013; 16:525-40. [PMID: 23325334 DOI: 10.1007/s10456-013-9332-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2012] [Accepted: 01/03/2013] [Indexed: 12/28/2022]
Abstract
Preclinical vascular research has been hindered by a lack of methods that can sensitively image and quantify vascular perfusion and leakage in vivo. In this study, we have developed dynamic near-infrared imaging methods to repeatedly visualize and quantify vascular leakage in mouse skin in vivo, and we have applied these methods to transgenic mice with overexpression of vascular endothelial growth factors VEGF-A or -C. Near-infrared dye conjugates were developed to identify a suitable vascular tracer that had a prolonged circulation lifetime and slow leakage into normal tissue after intravenous injection. Dynamic simultaneous imaging of ear skin and a large blood vessel in the leg enabled determination of the intravascular signal (blood volume fraction) from the tissue signal shortly after injection and quantifications of vascular leakage into the extravascular tissue over time. This method allowed for the sensitive detection of increased blood vascularity and leakage rates in K14-VEGF-A transgenic mice and also reliably measured inflammation-induced changes of vascularity and leakage over time in the same mice. Measurements after injection of recombinant VEGF-A surprisingly revealed increased blood vascular leakage and lymphatic clearance in K14-VEGF-C transgenic mice which have an expanded cutaneous lymphatic vessel network, potentially indicating unanticipated effects of lymphatic drainage on vascular leakage. Increased vascular leakage was also detected in subcutaneous tumors, confirming that the method can also be applied to deeper tissues. This new imaging method might facilitate longitudinal investigations of the in vivo effects of drug candidates, including angiogenesis inhibitors, in preclinical disease models.
Collapse
Affiliation(s)
- Steven T Proulx
- Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology, ETH Zurich, Wolfgang-Pauli-Str. 10, HCI H303, 8093 Zurich, Switzerland
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Wyatt SK, Manning HC, Bai M, Ehtesham M, Mapara KY, Thompson RC, Bornhop DJ. Preclinical molecular imaging of the translocator protein (TSPO) in a metastases model based on breast cancer xenografts propagated in the murine brain. Curr Mol Med 2012; 12:458-66. [PMID: 22348613 DOI: 10.2174/156652412800163361] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2011] [Revised: 01/09/2012] [Accepted: 02/02/2012] [Indexed: 01/14/2023]
Abstract
Previous studies have demonstrated the feasibility of translocator protein (TSPO) imaging to visualize and quantify human breast adenocarcinoma (MDA-MB-231) cells in vivo using a TSPO-targeted near-infrared (NIR) probe (NIR-conPK11195). This study aimed to extend the use of the TSPO-targeted probe to a more biologically relevant and clinically important tumor microenvironment as well as to assess our ability to longitudinally detect the presence and progression of breast cancer cells in the brain. The in vivo biodistribution and accumulation of NIR-conPK11195 and free (unconjugated) NIR dye were quantitatively evaluated in intracranial MDA-MB-231-bearing mice and non-tumor-bearing control mice longitudinally once a week from two to five weeks post-inoculation. The in vivo time-activity curves illustrate distinct clearance profiles for NIR-conPK11195 and free NIR dye, resulting in preferential accumulation of the TSPO-targeted probe in the intracranial tumor bearing hemisphere (TBH) with significant tumor contrast over normal muscle tissue (p < 0.005 at five weeks; p < 0.01 at four weeks). In addition, the TSPO-labeled TBHs demonstrated significant contrast over the TBHs of mice injected with free NIR dye (p < 0.001 at four and five weeks) as well as over the TSPO-labeled non-tumor-bearing hemispheres (NTBHs) of control mice (p < 0.005 at four and five weeks). Overall, TSPO-targeted molecular imaging appears useful for visualizing and quantifying breast cancer xenografts propagated in the murine brain and may assist in preclinical detection, diagnosis and monitoring of metastatic disease as well as drug discovery. Furthermore, these results indicate it should be possible to perform TSPO-imaging of breast cancer cells in the brain using radiolabeled TSPO-targeted agents, particularly in light of the fact that [11C]-labeled TSPO probes such as [11C]-PK 11195 have been successfully used to image gliomas in the clinic.
Collapse
Affiliation(s)
- Shelby K Wyatt
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | | | | | | | | | | |
Collapse
|
23
|
Fully automated synthesis of PET TSPO radioligands [11C]DAA1106 and [18F]FEDAA1106. Appl Radiat Isot 2012; 70:965-73. [DOI: 10.1016/j.apradiso.2012.03.011] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2011] [Revised: 02/09/2012] [Accepted: 03/05/2012] [Indexed: 11/17/2022]
|
24
|
|
25
|
Hight MR, Nolting DD, McKinley ET, Lander AD, Wyatt SK, Gonyea M, Zhao P, Manning HC. Multispectral fluorescence imaging to assess pH in biological specimens. JOURNAL OF BIOMEDICAL OPTICS 2011; 16:016007. [PMID: 21280913 PMCID: PMC3041815 DOI: 10.1117/1.3533264] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2010] [Revised: 11/01/2010] [Accepted: 11/17/2010] [Indexed: 05/29/2023]
Abstract
Simple, quantitative assays to measure pH in tissue could improve the study of complicated biological processes and diseases such as cancer. We evaluated multispectral fluorescence imaging (MSFI) to quantify extracellular pH (pHe) in dye-perfused, surgically-resected tumor specimens with commercially available instrumentation. Utilizing a water-soluble organic dye with pH-dependent fluorescence emission (SNARF-4F), we used standard fluorimetry to quantitatively assess the emission properties of the dye as a function of pH. By conducting these studies within the spectroscopic constraints imposed by the appropriate imaging filter set supplied with the imaging system, we determined that correction of the fluorescence emission of deprotonated dye was necessary for accurate determination of pH due to suboptimal excitation. Subsequently, employing a fluorimetry-derived correction factor (CF), MSFI data sets of aqueous dye solutions and tissuelike phantoms could be spectrally unmixed to accurately quantify equilibrium concentrations of protonated (HA) and deprotonated (A-) dye and thus determine solution pH. Finally, we explored the feasibility of MSFI for high-resolution pHe mapping of human colorectal cancer cell-line xenografts. Data presented suggest that MSFI is suitable for quantitative determination of pHe in ex vivo dye-perfused tissue, potentially enabling measurement of pH across a variety of preclinical models of disease.
Collapse
Affiliation(s)
- Matthew R Hight
- Vanderbilt University, Department of Physics, Nashville, Tennessee 37221, USA
| | | | | | | | | | | | | | | |
Collapse
|