Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Medchemcomm ; 9(10): 1649-1662, 2018 Oct 01.
Article in English | MEDLINE | ID: mdl-30429970

ABSTRACT

The natural products colchicine and combretastatin A-4 (CA4) have provided inspiration for the discovery and development of a wide array of derivatives and analogues that inhibit tubulin polymerization through a binding interaction at the colchicine site on ß-tubulin. A water-soluble phosphate prodrug salt of CA4 (referred to as CA4P) has demonstrated the ability to selectively damage tumor-associated vasculature and ushered in a new class of developmental anticancer agents known as vascular disrupting agents (VDAs). Through a long-term program of structure activity relationship (SAR) driven inquiry, we discovered that the dihydronaphthalene molecular scaffold provided access to small-molecule inhibitors of tubulin polymerization. In particular, a dihydronaphthalene analogue bearing a pendant trimethoxy aryl ring (referred to as KGP03) and a similar aroyl ring (referred to as KGP413) were potent inhibitors of tubulin polymerization (IC50 = 1.0 and 1.2 µM, respectively) and displayed low nM cytotoxicity against human cancer cell lines. In order to enhance water-solubility for in vivo evaluation, the corresponding phosphate prodrug salts (KGP04 and KGP152, respectively) were synthesized. In a preliminary in vivo study in a SCID-BALB/c mouse model bearing the human breast tumor MDA-MB-231-luc, a 99% reduction in signal was observed with bioluminescence imaging (BLI) 4 h after IP administration of KGP152 (200 mg kg-1) indicating reduced tumor blood flow. In a separate study, disruption of tumor-associated blood flow in a Fischer rat bearing an A549-luc human lung tumor was observed by color Doppler ultrasound following administration of KGP04 (15 mg kg-1).

2.
Nucl Med Biol ; 43(7): 424-37, 2016 07.
Article in English | MEDLINE | ID: mdl-27179748

ABSTRACT

INTRODUCTION: Dysregulated MMP expression or activation is associated with several diseases. To study MMP activity in vivo by means of PET a radiolabeled MMP inhibitor (MMPI) functioning as radiotracer has been developed by our group based on the lead structure CGS 25966. MATERIALS AND METHODS: Aiming at the modification of the pharmacokinetics of this lipophilic model tracer a new class of MMPIs has been discovered, consisting of additional fluorinated hydrophilic substructures, such as mini-PEG and/or 1,2,3-triazole units. To identify the best candidate for further clinical applications, radiofluorinated compounds of each subgroup have been (radio) synthesized and evaluated regarding their biodistribution behavior and their metabolic stability. RESULTS: Radiosyntheses of different triazole based MMPIs could be realized using two step "click chemistry" procedures. Compared to lead structure [(18)F]FEtO-CGS 25966 ([(18)F]1e, log D(exp) =2.02, IC50=2-50nM) all selected candidates showed increased hydrophilicities and inhibition potencies (log D(exp) =0.23-1.25, IC50=0.006-6nM). Interestingly, despite different hydrophilicities most triazole based MMPIs showed no significant differences in their in vivo biodistribution behavior and were cleared predominantly via the hepatobiliary excretion route. Biostability and metabolism studies in vitro and in vivo revealed significant higher metabolic stability for the triazole moiety compared to the benzyl ring in the lead structure. Cleavage of ethylene glycol subunits of the mini-PEG chain led to a faster metabolism of mini-PEG containing MMPIs. CONCLUSION: The introduction of hydrophilic groups such as mini-PEG and 1,2,3-triazole units did not lead to a significant shift of the hepatobiliary elimination towards renal clearance. Particularly the introduction of mini-PEG chains led to an intense metabolic decomposition. Substitution of the benzyl moiety in lead structure 1e by a 1,2,3-trizole ring resulted in an increased metabolic stability. Therefore, the 1,2,3-triazole-1-yl-methyl substituted MMPI [(18)F]3a was found to be the most stable candidate in this series and should be chosen for further preclinical evaluation.


Subject(s)
Hydroxamic Acids/chemistry , Matrix Metalloproteinase Inhibitors/chemistry , Matrix Metalloproteinase Inhibitors/pharmacokinetics , Animals , Drug Stability , Humans , Isotope Labeling , Matrix Metalloproteinase Inhibitors/metabolism , Mice , Mice, Inbred C57BL , Positron-Emission Tomography , Structure-Activity Relationship , Tissue Distribution
3.
Mol Imaging Biol ; 16(5): 739-46, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24696183

ABSTRACT

PURPOSE: [(18) F]VM4-037 has been developed as a positron emission tomography (PET) imaging marker to detect carbonic anhydrase IX (CA-IX) overexpression and is being investigated for use as a surrogate marker for tissue hypoxia. The purpose of this study was to determine the biodistribution and estimate the radiation dose from [(18) F]VM4-037 using whole-body PET/CT scans in healthy human volunteers. PROCEDURES: Successive whole-body PET/CT scans were performed after intravenous injection of [(18) F]VM4-037 in four healthy humans. The radiotracer uptakes in different organs were determined from the analysis of the PET scans. Human radiation doses were estimated using OLINDA/EXM software. RESULTS: High uptake of [(18) F]VM4-037 was observed in the liver and kidneys, with little clearance of activity during the study period, with mean standardized uptake values of ~35 in liver and ~22 in kidneys at ~1 h after injection. The estimated effective dose was 28 ± 1 µSv/MBq and the absorbed doses for the kidneys and liver were 273 ± 31 and 240 ± 68 µGy/MBq, respectively, for the adult male phantom. Hence, the effective dose would be 10 ± 0.5 mSv for the anticipated injected activity of 370 MBq, and the kidney and liver doses would be 101 ± 11 and 89 ± 25 mGy, respectively. CONCLUSIONS: [(18) F]VM4-037 displayed very high uptake in the liver and kidneys with little clearance of activity during the study period, resulting in these organs receiving the highest radiation doses among all bodily organs. Though the effective dose and the organ doses are within the limits considered as safe, the enhanced uptake of [(18) F]VM4-037 in the kidneys and liver will make the compound unsuitable for imaging overexpression of CA-IX in those two organs. However, the tracer may be suitable for imaging overexpression of CA-IX in lesions in other regions of the body such as in the lungs or head and neck region.


Subject(s)
Antigens, Neoplasm/metabolism , Carbonic Anhydrases/metabolism , Dipeptides/pharmacokinetics , Healthy Volunteers , Positron-Emission Tomography , Radiometry , Sulfonamides/pharmacokinetics , Adult , Aged , Carbonic Anhydrase IX , Dipeptides/chemical synthesis , Dipeptides/chemistry , Female , Humans , Male , Middle Aged , Sulfonamides/chemical synthesis , Sulfonamides/chemistry , Tissue Distribution , Tomography, X-Ray Computed
4.
Mol Imaging Biol ; 15(6): 748-57, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23689985

ABSTRACT

PURPOSE: A novel caspase-3 substrate-based probe [(18)F]-CP18 was evaluated as an in vivo positron emission tomography (PET) imaging agent for monitoring apoptosis in tumors. METHODS: Uptake of [(18)F]-CP18 in cell assays and tumors was measured. Caspase-3/7 activities in cell lysates and tumor homogenates were determined. Autoradiography,Terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL), and cleaved caspase-3 immunostaining were performed on adjacent tumor sections to identify areas of apoptosis. RESULTS: The in vitro cell assays showed caspase-3-dependent uptake of [(18)F]-CP18 in tumor cells when treated with an apoptosis inducer. The in vivo microPET imaging signal of [(18)F]-CP18 in xenograft tumors correlated with the ex vivo caspase-3/7 activities in these tumors. Furthermore, tumor autoradiographies of [(18)F]-CP18 in tumor sections matched adjacent sections stained by TUNEL and caspase-3 immunohistochemistry (IHC). CONCLUSIONS: [(18)F]-CP18 demonstrated high affinity and selectivity for activated caspase-3 both in vitro and in vivo, and the results support [(18)F]-CP18 as a promising new PET imaging agent for apoptosis.


Subject(s)
Apoptosis , Glycopeptides/pharmacokinetics , Molecular Imaging/methods , Neoplasms/diagnostic imaging , Positron-Emission Tomography/methods , Radiopharmaceuticals/pharmacokinetics , Animals , Caspase 3/analysis , Caspase 3/metabolism , Caspase 7/analysis , Caspase 7/metabolism , Cell Line, Tumor , Glycopeptides/chemistry , Humans , Linear Models , Mice , Radiopharmaceuticals/chemistry , Tissue Distribution , Xenograft Model Antitumor Assays
5.
Mol Imaging Biol ; 15(6): 739-47, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23681757

ABSTRACT

PURPOSE: We identified and validated [(18)F]-CP18, a DEVD (the caspase 3 substrate recognition motif) containing substrate-based compound as an imaging tracer for caspase-3 activity in apoptotic cells. PROCEDURES: CP18 was radiolabeled with fluorine-18 using click chemistry. The affinity and selectivity of CP18 for caspase-3 were evaluated in vitro. The biodistribution and metabolism pattern of [(18)F]-CP18 were assessed in vivo. [(18)F]-CP18 positron emission tomography (PET) scans were performed in a dexamethasone-induced thymic apoptosis mouse model. After imaging, the mice were sacrificed, and individual organs were collected, measured in a gamma counter, and tested for caspase-3 activity. RESULTS: In vitro enzymatic caspase-3 assay demonstrated specific cleavage of CP18. In vivo, [(18)F]-CP18 is predominantly cleared through the kidneys and urine, and is rapidly eliminated from the bloodstream. There was a sixfold increase in caspase activity and a fourfold increase of [(18)F]-CP18 retention in the dexamethasone-induced thymus of treated versus control mice. CONCLUSIONS: We report the use [(18)F]-CP18 as a PET tracer for imaging apoptosis. Our data support further development of this tracer for clinical PET applications.


Subject(s)
Apoptosis , Glycopeptides/pharmacokinetics , Molecular Imaging/methods , Positron-Emission Tomography/methods , Analysis of Variance , Animals , Caspase 3/metabolism , Cell Line, Tumor , Dexamethasone/adverse effects , Glycopeptides/chemistry , Humans , Linear Models , Mice , Radiopharmaceuticals/chemistry , Radiopharmaceuticals/pharmacokinetics , Thymus Gland/chemistry , Thymus Gland/drug effects , Tissue Distribution
6.
Alzheimers Dement ; 9(6): 666-76, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23411393

ABSTRACT

OBJECTIVE: We wished to develop a highly selective positron emission tomography (PET) imaging agent targeting PHF-tau in human Alzheimer's disease (AD) brains. METHODS: To screen potential tau binders, human AD brain sections were used as a source of native paired helical filament (PHF)-tau and Aß rather than synthetic tau aggregates or Aß fibrils generated in vitro to measure the affinity and selectivity of [(18)F]T807 to tau and Aß. Brain uptake and biodistribution of [(18)F]T807 in mice were also tested. RESULTS: In vitro autoradiography results show that [(18)F]T807 exhibits strong binding to PHF-tau-positive human brain sections. A dissociation constant (Kd) of [(18)F]T807 (14.6 nM) was measured using brain sections from the frontal lobe of AD patients. A comparison of autoradiography and double immunohistochemical staining of PHF-tau and Aß on adjacent sections demonstrated that [(18)F]T807 binding colocalized with immunoreactive PHF-tau pathology, but did not highlight Aß plaques. In vivo studies in mice demonstrated that [(18)F]T807 was able to cross the blood-brain barrier and washed out quickly. CONCLUSIONS: [(18)F]T807 demonstrates high affinity and selectivity to PHF-tau as well as favorable in vivo properties, making this a promising candidate as an imaging agent for AD.


Subject(s)
Alzheimer Disease/diagnosis , Brain/diagnostic imaging , Fluorine Radioisotopes , tau Proteins/chemistry , tau Proteins/drug effects , Aged , Aged, 80 and over , Amyloid beta-Peptides/metabolism , Animals , Autoradiography , Brain/pathology , Case-Control Studies , Female , Humans , Male , Mice , Mice, Transgenic , Middle Aged , Positron-Emission Tomography , Protein Binding/drug effects , Protein Binding/genetics , Tissue Distribution , tau Proteins/genetics
7.
ChemMedChem ; 8(1): 43-8, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23208787

ABSTRACT

CA II makes a good PET: Discovering positron emission tomography (PET) probes with high target affinities is challenging. PET probe discovery using in situ click chemistry uses (19) F-bearing fragments as (18) F surrogates. This ensures that the lead hits and PET probes have equivalent chemical or biological characteristics, making PET probe discovery predictable and reliable.


Subject(s)
Carbonic Anhydrase II/analysis , Click Chemistry/methods , Fluorine Radioisotopes/analysis , Positron-Emission Tomography/methods , Animals , Erythrocytes/enzymology , Humans , Mice , Mice, SCID
8.
J Alzheimers Dis ; 31(3): 601-12, 2012.
Article in English | MEDLINE | ID: mdl-22683529

ABSTRACT

Senile plaques and neurofibrillary tangles are prominent neuropathological hallmarks in Alzheimer's disease and are considered to be targets for therapeutic intervention as well as biomarkers for diagnostic in vivo imaging agents. While there are a number of amyloid-ß positron emission tomography (PET) tracers currently in different stages of clinical development and commercialization, there have been very few reports on imaging agents selectively targeting tau aggregates. In search of [18F]-PET tracers that possess great binding affinity and selectivity toward tau tangles, we tested more than 900 compounds utilizing a unique screening process. A competitive autoradiography assay was set up to test compounds for binding to native tau tangles and amyloid-ß plaques on human brain tissue sections. In our in vitro assays, the 18F labeled compound [18F]-T808 displayed a high level of binding affinity and good selectivity for tau aggregates over amyloid-ß plaques. [18F]-T808 showed rapid uptake and washout in rodent brains. Our in vitro and preclinical in vivo studies suggest that [18F]-T808 possesses suitable properties and characteristics to be a specific and selective PET probe for imaging of paired helical filament tau in human brains.


Subject(s)
Fluorine Radioisotopes , Positron-Emission Tomography/methods , Radiopharmaceuticals , tau Proteins , Amyloid beta-Peptides/metabolism , Animals , Autoradiography/methods , Fluorine Radioisotopes/metabolism , Humans , Mice , Mice, Inbred ICR , Plaque, Amyloid/diagnostic imaging , Plaque, Amyloid/metabolism , Protein Binding/physiology , Radiopharmaceuticals/chemistry , Radiopharmaceuticals/metabolism , Rats , Rats, Sprague-Dawley , tau Proteins/metabolism
9.
J Nucl Med ; 53(5): 787-95, 2012 May.
Article in English | MEDLINE | ID: mdl-22499613

ABSTRACT

UNLABELLED: 2-((2S,5R,8S,11S)-5-benzyl-8-(4-((2S,3R,4R,5R,6S)-6-((2-(4-(3-(18)F-fluoropropyl)-1H-1,2,3-triazol-1-yl)acetamido)methyl)-3,4,5-trihydroxytetrahydro-2H-pyran-2-carboxamido)butyl)-11-(3-guanidinopropyl)-3,6,9,12,15-pentaoxo-1,4,7,10,13-pentaazacyclopentadecan-2-yl)acetic acid ((18)F-RGD-K5) has been developed as an α(v)ß(3) integrin marker for PET. The purpose of this study was to determine the biodistribution and estimate the radiation dose from (18)F-RGD-K5 using whole-body PET/CT scans in monkeys and humans. METHODS: Successive whole-body PET/CT scans were obtained after intravenous injection of (18)F-RGD-K5 in 3 rhesus monkeys (167 ± 19 MBq) and 4 healthy humans (583 ± 78 MBq). In humans, blood samples were collected between the PET/CT scans, and stability of (18)F-RGD-K5 was assessed. Urine was also collected between the scans, to determine the total activity excreted in urine. The PET scans were analyzed to determine the radiotracer uptake in different organs. OLINDA/EXM software was used to calculate human radiation doses based on human and monkey biodistributions. RESULTS: (18)F-RGD-K5 was metabolically stable in human blood up to 90 min after injection, and it cleared rapidly from the blood pool, with a 12-min half-time. For both monkeys and humans, increased (18)F-RGD-K5 uptake was observed in the kidneys, bladder, liver, and gallbladder, with mean standardized uptake values at 1 h after injection for humans being approximately 20, 50, 4, and 10, respectively. For human biodistribution data, the calculated effective dose was 31 ± 1 µSv/MBq, and the urinary bladder wall had the highest absorbed dose at 376 ± 19 µGy/MBq using the 4.8-h bladder-voiding model. With the 1-h voiding model, these doses reduced to 15 ± 1 µSv/MBq for the effective dose and 103 ± 4 µGy/MBq for the absorbed dose in the urinary bladder wall. For a typical injected activity of 555 MBq, the effective dose would be 17.2 ± 0.6 mSv for the 4.8-h model, reducing to 8.3 ± 0.4 mSv for the 1-h model. For monkey biodistribution data, the effective dose to humans would be 22.2 ± 2.4 mSv for the 4.8-h model and 12.8 ± 0.2 mSv for the 1-h model. CONCLUSION: The biodistribution profile of (18)F-RGD-K5 in monkeys and humans was similar, with increased uptake in the bladder, liver, and kidneys. There was rapid clearance of (18)F-RGD-K5 through the renal system. The urinary bladder wall received the highest radiation dose and was deemed the critical organ. Both whole-body effective dose and bladder dose can be reduced by more frequent voiding. (18)F-RGD-K5 can be used safely for imaging α(v)ß(3) integrin expression in humans.


Subject(s)
Integrin alphaVbeta3/metabolism , Multimodal Imaging , Oligopeptides/pharmacokinetics , Peptides, Cyclic/pharmacokinetics , Positron-Emission Tomography , Tomography, X-Ray Computed , Whole Body Imaging , Adult , Aged , Animals , Biomarkers/metabolism , Female , Humans , Macaca mulatta , Male , Middle Aged , Oligopeptides/metabolism , Peptides, Cyclic/metabolism , Radiometry
SELECTION OF CITATIONS
SEARCH DETAIL
...