Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Chantal Kwizera is active.

Publication


Featured researches published by Chantal Kwizera.


Journal of Medicinal Chemistry | 2009

Synthesis and Preclinical Evaluation of Novel PET Probes for P-Glycoprotein Function and Expression

Aren van Waarde; Nisha Kuzhuppilly Ramakrishnan; Anna A. Rybczynska; Philip H. Elsinga; Francesco Berardi; Johan R. de Jong; Chantal Kwizera; Roberto Perrone; Mariangela Cantore; Jurgen W. A. Sijbesma; Rudi Dierckx; Nicola Antonio Colabufo

UNLABELLED P-glycoprotein (P-gp) is an ATP-dependent efflux pump protecting the body against xenobiotics. A P-gp substrate (7) and an inhibitor (6) were labeled with (11)C, resulting in potential tracers of P-gp function and expression. METHODS 6 and 7 were labeled using (11)CH(3)I. (11)C-verapamil was prepared as published previously, using (11)C-methyl triflate. MicroPET scans (with arterial sampling) and biodistribution studies were performed in rats pretreated with saline, cyclosporin A (CsA, 50 mg/kg), or cold 6 (15 mg/kg). RESULTS The radiochemical yields of (11)C-6 and (11)C-7 were approximately 30% with a total synthesis time of 45 min. Cerebral distribution volumes (DV) of (11)C-6 (2.35 +/- 0.11) and (11)C-7 (1.86 +/- 0.15) in saline-treated rats were higher than of (11)C-verapamil (0.64 +/- 0.12). DVs of (11)C-7 and (11)C-verapamil were significantly increased by CsA (to 5.26 +/- 0.14 and 5.85 +/- 0.32, respectively). The DV of (11)C-6 was reduced by cold 6 (to 1.65 +/- 0.03). Its uptake was also reduced (up to 67%) in several peripheral organs that express P-gp. CONCLUSIONS (11)C-7 is a novel tracer of P-gp function with higher baseline uptake than (11)C-verapamil. Upregulation of P-gp function in response to treatment (which is hard to detect with (11)C-verapamil) may be detectable using (11)C-7 and PET. Because (11)C-6 shows specific binding in target organs, this compound is the first PET tracer allowing measurement of P-gp expression.


The Journal of Nuclear Medicine | 2011

Small-Animal PET Study of Adenosine A 1 Receptors in Rat Brain: Blocking Receptors and Raising Extracellular Adenosine

Soumen Paul; Shivashankar Khanapur; Anna A. Rybczynska; Chantal Kwizera; Jurgen W. A. Sijbesma; Kiichi Ishiwata; Antoon T. M. Willemsen; Philip H. Elsinga; Rudi Dierckx; Aren van Waarde

Activation of adenosine A1 receptors (A1R) in the brain causes sedation, reduces anxiety, inhibits seizures, and promotes neuroprotection. Cerebral A1R can be visualized using 8-dicyclopropylmethyl-1-11C-methyl-3-propyl-xanthine (11C-MPDX) and PET. This study aims to test whether 11C-MPDX can be used for quantitative studies of cerebral A1R in rodents. Methods: 11C-MPDX was injected (intravenously) into isoflurane-anesthetized male Wistar rats (300 g). A dynamic scan of the central nervous system was obtained, using a small-animal PET camera. A cannula in a femoral artery was used for blood sampling. Three groups of animals were studied: group 1, controls (saline-treated); group 2, animals pretreated with the A1R antagonist 8-cyclopentyl-1,3-dipropylxanthine (DPCPX, 1 mg, intraperitoneally); and group 3, animals pretreated (intraperitoneally) with a 20% solution of ethanol in saline (2 mL) plus the adenosine kinase inhibitor 4-amino-5-(3-bromophenyl)-7-(6-morpholino-pyridin-3-yl)pyrido[2,3-d] pyrimidine dihydrochloride (ABT-702) (1 mg). DPCPX is known to occupy cerebral A1R, whereas ethanol and ABT-702 increase extracellular adenosine. Results: In groups 1 and 3, the brain was clearly visualized. High uptake of 11C-MPDX was noted in striatum, hippocampus, and cerebellum. In group 2, tracer uptake was strongly suppressed and regional differences were abolished. The treatment of group 3 resulted in an unexpected 40%–45% increase of the cerebral uptake of radioactivity as indicated by increases of PET standardized uptake value, distribution volume from Logan plot, nondisplaceable binding potential from 2-tissue-compartment model fit, and standardized uptake value from a biodistribution study performed after the PET scan. The partition coefficient of the tracer (K1/k2 from the model fit) was not altered under the study conditions. Conclusion: 11C-MPDX shows a regional distribution in rat brain consistent with binding to A1R. Tracer binding is blocked by the selective A1R antagonist DPCPX. Pretreatment of animals with ethanol and adenosine kinase inhibitor increases 11C-MPDX uptake. This increase may reflect an increased availability of A1R after acute exposure to ethanol.


The Journal of Nuclear Medicine | 2013

Small-Animal PET with a σ-Ligand, 11C-SA4503, Detects Spontaneous Pituitary Tumors in Aged Rats

Nisha Kuzhuppilly Ramakrishnan; Anna A. Rybczynska; Anniek K. D. Visser; Krisztina Marosi; Csaba Nyakas; Chantal Kwizera; Jurgen W. A. Sijbesma; Philip H. Elsinga; Kiichi Ishiwata; Jan Pruim; Rudi Dierckx; Aren van Waarde

Pituitary tumors are often detected only after death or at late stages of the disease when they are macroadenomas with a low surgical cure rate. Spontaneous pituitary tumors occur in rats over 1 y of age. In an ongoing study of changes in σ-1 agonist binding related to aging, several of our rats developed such tumors. The aim of the current study was to assess the kinetics of 11C-SA4503 (11C-labeled 1-[2-(3,4-dimethoxyphenthyl)]-4-(3-phenylpropyl)-piperazine dihydrochloride) in tumor and brain and to evaluate the utility of this tracer in the detection of pituitary tumors. Methods: Small-animal PET scans of the brain region of male Wistar Hannover rats (age, 18–32 mo) were acquired using the σ-1 agonist tracer 11C-SA4503. The time-dependent uptake of 11C in the entire brain, tumor or normal pituitary, and thyroid was measured. A 2-tissue-compartment model was fitted to the PET data, using metabolite-corrected plasma radioactivity as the input function. Results: Pituitary tumors showed up as bright hot spots in the scans. The total distribution volume (VT) of the tracer was significantly higher in the tumor than in the normal pituitary. Surprisingly, a higher VT was also seen in the brain and thyroid tissue of animals with pituitary tumors than in healthy rats. The increase in VT in the brain and thyroid was not related to a change in nondisplaceable binding potential (BPND) but rather to an increase in the partition coefficient (K1/k2) of 11C-SA4503. The increase in VT in the tumor on the other hand was accompanied by a significant increase in BPND. Western blotting analysis indicated that pituitary tumors overexpressed σ-1 receptors. Conclusion: The overexpression of σ-1 receptors in spontaneous pituitary tumors is detected as an increase in uptake and BPND of 11C-SA4503. Therefore, this tracer may have promise for the detection of pituitary adenomas, using PET.


European Journal of Nuclear Medicine and Molecular Imaging | 2012

Population pharmacokinetics of cutamesine in rats using NONMEM, C-11-SA4503, and microPET

Nisha Kuzhuppilly Ramakrishnan; V. Pilla Reddy; Johannes H. Proost; Csaba Nyakas; Chantal Kwizera; Jurgen W. A. Sijbesma; Philippus Elsinga; K. Ishiwata; Rudi Dierckx; van Aren Waarde

C. Asferg, R. Møgelvang, A. Flyvbjerg, A. Frystyk, J.S. Jensen, J.L. Marott, M. Appleyard, P. Schnoh, G.B. Jensen, J. Jeppesen. Copenhagen University Hospital Glostrup, Department of Clinical Physiology and Nuclear Medicine, Glostrup, Denmark, Copenhagen University Hospital Gentofte, Department of Cardiology, Gentofte, Denmark, Aarhus University Hospital Aarhus, Medical Research Laboratories, Clinical Institute and Medical Department M, Aarhus, Denmark, Copenhagen University Hospital Bispebjerg, The Copenhagen City Heart Study, Bispebjerg, Denmark, Copenhagen University Hospital Hvidovre, Department of Cardiology, Hvidovre, Denmark, Copenhagen University Hospital Glostrup, Department of Medicine, Glostrup, Denmark, Faculty of Health Sciences, University of Aarhus, Aarhus, Denmark, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark


ChemMedChem | 2016

(11)C- and (18)F-Labeled Radioligands for P-Glycoprotein Imaging by Positron Emission Tomography.

Mariangela Cantore; Marcel Benadiba; Philip H. Elsinga; Chantal Kwizera; Rudi Dierckx; Nicola Antonio Colabufo; Gert Luurtsema

P‐Glycoprotein (P‐gp) is an efflux transporter widely expressed at the human blood–brain barrier. It is involved in xenobiotics efflux and in onset and progression of neurodegenerative disorders. For these reasons, there is great interest in the assessment of P‐gp expression and function by noninvasive techniques such as positron emission tomography (PET). Three radiolabeled aryloxazole derivatives: 2‐[2‐(2‐methyl‐(11C)‐5‐methoxyphenyl)oxazol‐4‐ylmethyl]‐6,7‐dimethoxy‐1,2,3,4‐tetrahydroisoquinoline ([11C]‐5); 2‐[2‐(2‐fluoromethyl‐(18F)‐5‐methoxyphenyl)oxazol‐4‐ylmethyl]‐6,7‐dimethoxy‐1,2,3,4‐tetra‐hydroisoquinoline ([18F]‐6); and 2‐[2‐(2‐fluoroethyl‐(18F)‐5‐methoxyphenyl)oxazol‐4‐ylmethyl]‐6,7‐dimethoxy‐1,2,3,4‐tetrahydroisoquinoline ([18F]‐7), were tested in several in vitro biological assays to assess the effect of the aryl substituent in terms of potency and mechanism of action toward P‐gp. Methyl derivative [11C]‐5 is a potent P‐gp substrate, whereas the corresponding fluoroethyl derivative [18F]‐7 is a P‐gp inhibitor. Fluoromethyl compound [18F]‐6 is classified as a non‐transported P‐gp substrate, because its efflux increases after cyclosporine A modulation. These studies revealed a promising substrate and inhibitor, [11C]‐5 and [18F]‐7, respectively, for in vivo imaging of P‐gp by using PET.


Nuclear Medicine and Biology | 2015

In vivo imaging of brain androgen receptors in rats: a ( 18 F)FDHT PET study

M. A. Khayum; Janine Doorduin; Ines Farinha Antunes; Chantal Kwizera; Rolf Zijlma; J. A. den Boer; Rudi Dierckx; E. F. J. de Vries

INTRODUCTION Steroid hormones like androgens play an important role in the development and maintenance of several brain functions. Androgens can act through androgen receptors (AR) in the brain. This study aims to demonstrate the feasibility of positron emission tomography (PET) with 16β-[(18)F]fluoro-5α-dihydrotestosterone ([(18)F]FDHT) to image AR expression in the brain. METHODS Male Wistar rats were either orchiectomized to inhibit endogenous androgen production or underwent sham-surgery. Fifteen days after surgery, rats were subjected to a 90-min dynamic [(18)F]FDHT PET scan with arterial blood sampling. In a subset of orchiectomized rats, 1mg/kg dihydrotestosterone was co-injected with the tracer in order to saturate the AR. Plasma samples were analyzed for the presence of radioactive metabolites by radio-TLC. Pharmacokinetic modeling was performed to quantify brain kinetics of the tracer. After the PET scan, the animals were terminated for ex-vivo biodistribution. RESULTS PET imaging and ex vivo biodistribution studies showed low [(18)F]FDHT uptake in all brain regions, except pituitary. [(18)F]FDHT uptake in the surrounding cranial bones was high and increased over time. [(18)F]FDHT was rapidly metabolized in rats. Metabolism was significantly faster in orchiectomized rats than in sham-orchiectomized rats. Quantitative analysis of PET data indicated substantial spill-over of activity from cranial bones into peripheral brain regions, which prevented further analysis of peripheral brain regions. Logan graphical analysis and kinetic modeling using 1- and 2-tissue compartment models showed reversible and homogenously distributed tracer uptake in central brain regions. [(18)F]FDHT uptake in the brain could not be blocked by endogenous androgens or administration of dihydrotestosterone. CONCLUSION The results of this study indicate that imaging of AR availability in rat brain with [(18)F]FDHT PET is not feasible. The low AR expression in the brain, the rapid metabolism of [(18)F]FDHT in rats and the poor brain penetration of the tracer likely contributed to the poor performance of [(18)F]FDHT PET in this study.


Bioorganic & Medicinal Chemistry | 2017

Structure-activity relationship study towards non-peptidic positron emission tomography (PET) radiotracer for gastrin releasing peptide receptors: Development of [18F] (S)-3-(1H-indol-3-yl)-N-[1-[5-(2-fluoroethoxy)pyridin-2-yl]cyclohexylmethyl]-2-methyl-2-[3-(4-nitrophenyl)ureido]propionamide

Enza Lacivita; Ermelinda Lucente; Chantal Kwizera; Ines Farinha Antunes; Mauro Niso; Paola De Giorgio; Roberto Perrone; Nicola Antonio Colabufo; Philip H. Elsinga; Marcello Leopoldo

Gastrin-releasing peptide receptors (GRP-Rs, also known as bombesin 2 receptors) are overexpressed in a variety of human cancers, including prostate cancer, and therefore they represent a promising target for in vivo imaging of tumors using positron emission tomography (PET). Structural modifications of the non-peptidic GRP-R antagonist PD-176252 ((S)-1a) led to the identification of the fluorinated analog (S)-3-(1H-indol-3-yl)-N-[1-[5-(2-fluoroethoxy)pyridin-2-yl]cyclohexylmethyl]-2-methyl-2-[3-(4-nitrophenyl)ureido]propionamide ((S)-1m) that showed high affinity and antagonistic properties for GRP-R. This antagonist was stable in rat plasma and towards microsomal oxidative metabolism in vitro. (S)-1m was successfully radiolabeled with fluorine-18 through a conventional radiochemistry procedure. [18F](S)-1m showed high affinity and displaceable interaction for GRP-Rs in PC3 cells in vitro.


Psychopharmacology | 2015

Pridopidine selectively occupies sigma-1 rather than dopamine D2 receptors at behaviorally active doses

Kristoffer Sahlholm; Jurgen W. A. Sijbesma; Bram Maas; Chantal Kwizera; Daniel Marcellino; Nisha Kuzhuppilly Ramakrishnan; Rudi A. J. O. Dierckx; Philip H. Elsinga; Aren van Waarde


European Journal of Nuclear Medicine and Molecular Imaging | 2012

Dose-dependent sigma-1 receptor occupancy by donepezil in rat brain can be assessed with C-11-SA4503 and microPET

Nisha Kuzhuppilly Ramakrishnan; Anniek K. D. Visser; Chantal Kwizera; Jurgen W. A. Sijbesma; Philippus Elsinga; Csaba Nyakas; K. Ishiwata; Rudi A. J. O. Dierckx; van Aren Waarde


Molecular Imaging and Biology | 2016

Novel Approach to Repeated Arterial Blood Sampling in Small Animal PET: Application in a Test-Retest Study with the Adenosine A1 Receptor Ligand [11C]MPDX

Jurgen W. A. Sijbesma; Xiaoyun Zhou; David Vállez García; Martin C. Houwertjes; Janine Doorduin; Chantal Kwizera; Bram Maas; Peter Meerlo; Rudi Dierckx; Riemer H. J. A. Slart; Philip H. Elsinga; Aren van Waarde

Collaboration


Dive into the Chantal Kwizera's collaboration.

Top Co-Authors

Avatar

Jurgen W. A. Sijbesma

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar

Nisha Kuzhuppilly Ramakrishnan

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar

Rudi Dierckx

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar

Aren van Waarde

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar

Philip H. Elsinga

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar

Philippus Elsinga

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar

Rudi A. J. O. Dierckx

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Anna A. Rybczynska

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge