Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 16 de 16
1.
Radiology ; 310(1): e231984, 2024 Jan.
Article En | MEDLINE | ID: mdl-38226877

Background The presence of gadolinium traces in the skin after administration of gadolinium-based contrast agents (GBCAs) raised safety concerns regarding a potential association with small fiber neuropathy (SFN). Purpose To investigate signs of SFN in rat foot pads by quantification of the intraepidermal nerve fiber density (IENFD) after multiple GBCA administrations and to evaluate gadolinium concentration, chemical species, and clearance. Materials and Methods Fifty rats received eight intravenous injections of either gadodiamide, gadobutrol, gadoterate, gadoteridol (8 × 0.6 mmol per kilogram of body weight), or saline (1.2 mL per kilogram of body weight), within 2 weeks and were sacrificed 5 days or 5 weeks after the last injection. IENFD was determined with protein gene product (PGP) 9.5 immunofluorescent staining and blinded and automated image analysis. The gadolinium and GBCA concentrations were measured with inductively coupled plasma mass spectrometry (ICP-MS), laser ablation ICP-MS, and matrix-assisted laser desorption/ionization mass spectrometry imaging (MALDI MSI). P values were calculated using linear contrasts of model analysis. Results The IENFD (measured as geometric mean [SD] and in number of nerve fibers per millimeter of epidermis) was not significantly altered after 5 days (saline, 8.4 [1.1]; gadobutrol, 9.7 [1.2]; gadoterate, 9.2 [1.2]; gadoteridol, 9.9 [1.3]; gadodiamide, 10.5 [1.2]) or 5 weeks (saline, 19.7 [1.4]; gadobutrol, 16.4 [1.6]; gadoterate, 14.3 [1.6]; gadoteridol, 22.2 [1.8]; gadodiamide, 17.9 [1.4]). Gadolinium skin concentrations were highest for gadodiamide after 5 days (16.0 nmol/g [1.1]) and 5 weeks (10.6 nmol/g [1.2], -33%). Macrocyclic agents were lower at 5 days (gadoteridol, 2.6 nmol/g [1.2]; gadobutrol, 2.7 nmol/g [1.1]; and gadoterate, 2.3 nmol/g [1.2]) and efficiently cleared after 5 weeks (gadoteridol, -95%; gadobutrol and gadoterate, -96%). The distribution of gadolinium and IENF did not visually overlap. For macrocyclic agents, gadolinium was found in sweat glands and confirmed to be intact chelate. Conclusion There were no signs of SFN in rat foot pads using multiple dosing regimens at two time points after administration of GBCAs. Macrocyclic GBCAs exhibited lower levels of gadolinium in the skin and were effectively eliminated within 5 weeks compared with linear gadodiamide, and intact macrocyclic GBCA was detected in sweat glands. © RSNA, 2024 Supplemental material is available for this article. See also the editorial by Clement in this issue.


Gadolinium DTPA , Gadolinium , Heterocyclic Compounds , Organometallic Compounds , Small Fiber Neuropathy , Animals , Rats , Contrast Media , Body Weight
2.
Invest Radiol ; 57(10): 629-638, 2022 10 01.
Article En | MEDLINE | ID: mdl-35703267

OBJECTIVES: The aim of this report was to characterize the key physicochemical, pharmacokinetic (PK), and magnetic resonance imaging (MRI) properties of gadoquatrane (BAY 1747846), a newly designed tetrameric, macrocyclic, extracellular gadolinium-based contrast agent (GBCA) with high relaxivity and stability. MATERIALS AND METHODS: The r1-relaxivities of the tetrameric gadoquatrane at 1.41 and 3.0 T were determined in human plasma and the nuclear magnetic relaxation dispersion profiles in water and plasma. The complex stability was analyzed in human serum over 21 days at pH 7.4 at 37°C and was compared with the linear GBCA gadodiamide and the macrocyclic GBCA (mGBCA) gadobutrol. In addition, zinc transmetallation assay was performed to investigate the kinetic inertness. Protein binding and the blood-to-plasma ratio were determined in vitro using rat and human plasma. The PK profile was evaluated in rats (up to 7 days postinjection). Magnetic resonance imaging properties were investigated using a glioblastoma (GS9L) rat model. RESULTS: The new chemical entity gadoquatrane is a macrocyclic tetrameric Gd complex with one inner sphere water molecule per Gd ( q = 1). Gadoquatrane showed high solubility in buffer (1.43 mol Gd/L, 10 mM Tris-HCl, pH 7.4), high hydrophilicity (logP -4.32 in 1-butanol/water), and negligible protein binding. The r1-relaxivity of gadoquatrane in human plasma per Gd of 11.8 mM -1 ·s -1 (corresponding to 47.2 mM -1 ·s -1 per molecule at 1.41 T at 37°C, pH 7.4) was more than 2-fold (8-fold per molecule) higher compared with established mGBCAs. Nuclear magnetic relaxation dispersion profiles confirmed the more than 2-fold higher r1-relaxivity in human plasma for the clinically relevant magnetic field strengths from 0.47 to 3.0 T. The complex stability of gadoquatrane at physiological conditions was very high. The observed Gd release after 21 days at 37°C in human serum was below the lower limit of quantification. Gadoquatrane showed no Gd 3+ release in the presence of zinc in the transmetallation assay. The PK profile (plasma elimination, biodistribution, recovery) was comparable to that of gadobutrol. In MRI, the quantitative evaluation of the tumor-to-brain contrast in the rat glioblastoma model showed significantly improved contrast enhancement using gadoquatrane compared with gadobutrol at the same Gd dose administered (0.1 mmol Gd/kg body weight). In comparison to gadoterate meglumine, similar contrast enhancement was reached with gadoquatrane with 75% less Gd dose. In terms of the molecule dose, this was reduced by 90% when compared with gadoterate meglumine. Because of its tetrameric structure and hence lower number of molecules per volume, all prepared formulations of gadoquatrane were iso-osmolar to blood. CONCLUSIONS: The tetrameric gadoquatrane is a novel, highly effective mGBCA for use in MRI. Gadoquatrane provides favorable physicochemical properties (high relaxivity and stability, negligible protein binding) while showing essentially the same PK profile (fast extracellular distribution, fast elimination via the kidneys in an unchanged form) to established mGBCAs on the market. Overall, gadoquatrane is an excellent candidate for further clinical development.


Glioblastoma , Organometallic Compounds , Animals , Contrast Media/pharmacokinetics , Gadolinium/pharmacokinetics , Humans , Magnetic Resonance Imaging/methods , Organometallic Compounds/pharmacokinetics , Rats , Tissue Distribution , Water , Zinc
3.
Radiology ; 290(2): 340-348, 2019 02.
Article En | MEDLINE | ID: mdl-30422091

Purpose To investigate the long-term course of MRI signal intensity (SI) changes and the presence of gadolinium in the rat brain during a 1-year period after multiple administrations of gadolinium-based contrast agents (GBCAs). Materials and Methods Rats received a linear GBCA (gadodiamide, gadopentetate dimeglumine, gadobenate dimeglumine), a macrocyclic GBCA (gadobutrol, gadoterate meglumine, gadoteridol), or saline. Animals received eight injections over 2 weeks (1.8 mmol/kg per injection). Brain MRI and gadolinium measurements were performed with inductively coupled plasma mass spectrometry (ICP-MS) and laser ablation ICP-MS 5, 26, and 52 weeks after administration. Results Animals that received linear GBCAs showed higher deep cerebellar nuclei (DCN)-to-brainstem SI ratios compared with the saline group (P < .001 at all time points). After 1 year, mean gadolinium concentrations in the cerebellum were 3.38 nmol/g (gadodiamide), 2.13 nmol/g (gadopentetate dimeglumine), and 1.91 nmol/g (gadobenate dimeglumine). For linear agents, laser ablation ICP-MS revealed gadolinium depositions in the cerebellar nuclei. For macrocyclic GBCAs, the DCN-to-brainstem SI ratios did not significantly differ from those in the saline group (P > .42) and the cerebellar gadolinium concentrations decreased between weeks 5 and 52, reaching 0.08 nmol/g (gadobutrol), 0.04 nmol/g (gadoterate meglumine), and 0.07 nmol/g (gadoteridol). The respective laser ablation ICP-MS analysis showed no gadolinium depositions. Conclusion Increased signal intensity in the deep cerebellar nuclei of rats persists for at least 1 year after administration of linear gadolinium-based contrast agents (GBCAs), in line with persistent brain gadolinium concentrations with no elimination after the initial 5-week period. The animals that received macrocyclic GBCAs showed an ongoing elimination of gadolinium from the brain during the entire observation period. © RSNA, 2018.


Cerebellar Nuclei , Contrast Media/pharmacokinetics , Gadolinium DTPA/pharmacokinetics , Gadolinium/pharmacokinetics , Animals , Brain/diagnostic imaging , Brain/metabolism , Brain Chemistry/drug effects , Brain Stem/chemistry , Brain Stem/metabolism , Cerebellar Nuclei/chemistry , Cerebellar Nuclei/metabolism , Magnetic Resonance Imaging , Male , Mass Spectrometry , Rats
4.
Invest Radiol ; 54(2): 76-82, 2019 02.
Article En | MEDLINE | ID: mdl-30358694

OBJECTIVE: This study investigated the potential effect of the chelating agent calcium trisodium pentetate (Ca-DTPA) on the urinary excretion of gadolinium and the subsequent elimination of gadolinium (Gd) in the brain after a single intravenous administration of either a linear (gadodiamide) or a macrocyclic (gadobutrol) Gd-based contrast agent in rats. MATERIALS AND METHODS: Rats received either a single injection of gadodiamide or gadobutrol (1.8 mmol/kg, each) or saline (n = 18 per group). Seven weeks after the injection, 6 animals of each group were killed before the treatment period. From the remaining 12 animals, 6 received either 3 intravenous injections of Ca-DTPA (180 µmol/kg) or saline. Urine was collected daily for 3 days after each infusion. Gadolinium measurements by ICP-MS were performed in urine and tissue samples. RESULTS: In animals that initially received the linear gadodiamide, Ca-DTPA infusion increased the urinary excretion of Gd by a factor of 10 (cumulative amount of 114 ± 21 nmol Gd vs 10 ± 4 nmol Gd after saline infusion, P ≤ 0.0001). In contrast, animals that received the macrocyclic gadobutrol exhibited a higher spontaneous urinary excretion of Gd (33 ± 12 nmol after saline infusion) and Ca-DTPA had no impact (30 ± 11 nmol Gd, P = 0.68).The urinary excretion of Gd was associated with Gd brain content. Seven weeks after the initial Gd-based contrast agent administration, a total amount of 0.74 ± 0.053 nmol Gd was quantified in the brain after administration of gadodiamide. The Gd brain burden was partially reduced at the end of the treatment period in the animals that were repeatedly infused with Ca-DTPA (0.56 ± 0.13 nmol Gd, P = 0.009) but not with saline (0.66 ± 0.081 nmol, P = 0.32). In contrast, the total amount of macrocyclic gadobutrol measured in the brain was lower (0.11 ± 0.029 nmol Gd) and still spontaneously cleared during the 3-week saline infusion period (0.057 ± 0.019 nmol Gd (P = 0.003). Gadolinium quantified in the brain after infusions with Ca-DTPA did not differ from saline-infused animals (0.049 ± 0.014 nmol Gd). CONCLUSIONS: Administration of the chelating agent Ca-DTPA 7 weeks after injection of linear gadodiamide induced relevant urinary Gd excretion. In parallel, the Gd amount in the brain tissue decreased. This indicates a dechelated pool among the chemical Gd forms present in the rat brain after linear gadodiamide administration that can be mobilized by chelation with Ca-DTPA. In contrast, Ca-DTPA did not mobilize Gd in animals that received macrocyclic gadobutrol, indicating that the Gd measured is intact gadobutrol.


Chelating Agents/pharmacology , Contrast Media/pharmacokinetics , Gadolinium DTPA/pharmacokinetics , Gadolinium/metabolism , Organometallic Compounds/pharmacokinetics , Pentetic Acid/pharmacology , Animals , Brain/metabolism , Contrast Media/administration & dosage , Gadolinium/urine , Gadolinium DTPA/administration & dosage , Male , Organometallic Compounds/administration & dosage , Rats , Rats, Wistar
5.
Invest Radiol ; 53(5): 278-285, 2018 05.
Article En | MEDLINE | ID: mdl-29319556

OBJECTIVE: The aim of this retrospective study was to determine the gadolinium (Gd) concentration in different brain areas in a pig cohort that received repeated administration of Gd-based contrast agents (GBCAs) at standard doses over several years, comparable with a clinical setting. MATERIAL AND METHODS: Brain tissue was collected from 13 Göttingen mini pigs that had received repeated intravenous injections of gadopentetate dimeglumine (Gd-DTPA; Magnevist) and/or gadobutrol (Gadovist). The animals have been included in several preclinical imaging studies since 2008 and received cumulative Gd doses ranging from 7 to 129 mmol per animal over an extended period. Two animals with no history of administration of GBCA were included as controls. Brain autopsies were performed not earlier than 8 and not later than 38 months after the last GBCA application. Tissues from multiple brain areas including cerebellar and cerebral deep nuclei, cerebellar and cerebral cortex, and pons were analyzed for Gd using inductively coupled plasma mass spectrometry. RESULTS: Of the 13 animals, 8 received up to 48 injections of gadobutrol and Gd-DTPA and 5 received up to 29 injections of gadobutrol only. In animals that had received both Gd-DTPA and gadobutrol, a median (interquartile range) Gd concentration of 1.0 nmol/g tissue (0.44-1.42) was measured in the cerebellar nuclei and 0.53 nmol/g (0.29-0.62) in the globus pallidus. The Gd concentration in these areas in gadobutrol-only animals was 50-fold lower with median concentrations of 0.02 nmol/g (0.01-0.02) for cerebellar nuclei and 0.01 nmol/g (0.01-0.01) for globus pallidus and was comparable with control animals with no GBCA history. Accordingly, in animals that received both GBCAs, the amount of residual Gd correlated with the administered dose of Gd-DTPA (P ≤ 0.002) but not with the total Gd dose, consisting of Gd-DTPA and gadobutrol. The Gd concentration in cortical tissue and in the pons was very low (≤0.07 nmol/g tissue) in all animals analyzed. CONCLUSION: Multiple exposure to macrocyclic gadobutrol is not associated with Gd deposition in brain tissue of healthy pigs. A single additional administration of linear Gd-DTPA is sufficient for Gd accumulation in the nucleus dentatus and globus pallidus, underlining the importance of obtaining a complete GBCA history in clinical studies.


Cerebellar Nuclei/metabolism , Contrast Media/pharmacokinetics , Gadolinium DTPA/pharmacokinetics , Globus Pallidus/metabolism , Organometallic Compounds/pharmacokinetics , Animals , Contrast Media/metabolism , Female , Gadolinium DTPA/metabolism , Humans , Male , Models, Animal , Organometallic Compounds/metabolism , Retrospective Studies , Swine
6.
J Nucl Med ; 58(7): 1094-1099, 2017 07.
Article En | MEDLINE | ID: mdl-28302764

Thromboembolic diseases such as myocardial infarction, stroke, transient ischemic attacks, and pulmonary embolism are major causes of morbidity and mortality worldwide. Glycoprotein IIb/IIIa (GPIIb/IIIa) is the key receptor involved in platelet aggregation and is a validated target for therapeutic approaches and diagnostic imaging. The aim of this study was to develop and characterize a specific small-molecule tracer for PET imaging that binds with high affinity to GPIIb/IIIa receptors and has suitable pharmacokinetic properties to overcome limitations of previous approaches. Methods: Binding of 18F-GP1 to GPIIb/IIIa receptors was investigated in competition binding assays and autoradiography using a fresh cardiac thrombus from an explanted human heart. The clot-to-blood ratio for 18F-GP1 was investigated by an in vitro blood flow model. Biodistribution and thrombus detection was investigated in cynomolgus monkeys after insertion of a roughened catheter into either the vena cava or the aorta. Results:18F-GP1 is an 18F-labeled small molecule for PET imaging of thrombi. The half maximal inhibitory concentration of 18F-GP1 to GPIIb/IIIa was 20 nM. 18F-GP1 bound to thrombi with a mean clot-to-blood ratio of 95. Binding was specific and can be displaced by excess nonradioactive derivative. Binding was not affected by anticoagulants such as aspirin or heparin. 18F-GP1 showed rapid blood clearance and a low background after intravenous injection in cynomolgus monkeys. Small arterial, venous thrombi, thrombotic depositions on damaged endothelial surface, and small cerebral emboli were detected in vivo by PET imaging. Conclusions:18F-GP1 binds specifically with high affinity to the GPIIb/IIIa receptor involved in platelet aggregation. Because of its favorable preclinical characteristics, 18F-GP1 is currently being investigated in a human clinical study.


Glutamine/analogs & derivatives , Laurates/pharmacokinetics , Molecular Imaging/methods , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Positron-Emission Tomography/methods , Thrombosis/diagnostic imaging , Thrombosis/metabolism , Animals , Female , Fluorine Radioisotopes/chemistry , Fluorine Radioisotopes/pharmacokinetics , Glutamine/pharmacokinetics , Humans , Isotope Labeling/methods , Macaca fascicularis , Radiopharmaceuticals/chemical synthesis , Radiopharmaceuticals/pharmacokinetics , Reproducibility of Results , Sensitivity and Specificity
7.
Invest Radiol ; 52(6): 324-333, 2017 06.
Article En | MEDLINE | ID: mdl-28323657

OBJECTIVES: Retrospective studies in patients with primary brain tumors or other central nervous system pathologies as well as postmortem studies have suggested that gadolinium (Gd) deposition occurs in the dentate nucleus (DN) and globus pallidus (GP) after multiple administrations of primarily linear Gd-based contrast agents (GBCAs). However, this deposition has not been associated with any adverse effects or histopathological alterations. The aim of this preclinical study was to systematically examine differences between linear and macrocyclic GBCAs in their potential to induce changes in brain and skin histology including Gd distribution in high spatial resolution. MATERIALS AND METHODS: Fifty male Wistar-Han rats were randomly allocated into control (saline, n = 10 rats) and 4 GBCA groups (linear GBCAs: gadodiamide and gadopentetate dimeglumine, macrocyclic GBCAs: gadobutrol and gadoteridol; n = 10 rats per group). The animals received 20 daily intravenous injections at a dose of 2.5 mmol Gd/kg body weight. Eight weeks after the last GBCA administration, the animals were killed, and the brain and skin samples were histopathologically assessed (hematoxylin and eosin; cresyl violet [Nissl]) and by immunohistochemistry. The Gd concentration in the skin, bone, brain, and skeletal muscle samples were analyzed using inductively coupled plasma mass spectroscopy (ICP-MS, n = 4). The spatial Gd distribution in the brain and skin samples was analyzed in cryosections using laser ablation coupled with ICP-MS (LA-ICP-MS, n = 3). For the ultra-high resolution of Gd distribution, brain sections of rats injected with gadodiamide or saline (n = 1) were assessed by scanning electron microscopy coupled to energy dispersive x-ray spectroscopy and transmission electron microscopy, respectively. RESULTS: No histological changes were observed in the brain. In contrast, 4 of 10 animals in the gadodiamide group but none of the animals in other groups showed macroscopic and histological nephrogenic systemic fibrosis-like skin lesions. The Gd concentrations observed in the skin/brain samples (in nanomole Gd per gram of tissue) for each agent were as follows: gadodiamide: 1472 ± 115/11.1 ± 5.1, gadopentetate dimeglumine: 80.8 ± 6.2/13.1 ± 7.3, gadobutrol: 1.1 ± 0.5/0.7 ± 0.4, and gadoteridol: 1.7 ± 0.8/0.5 ± 0.2. The average detected residual Gd concentration in the brain was approximately 15-fold higher for linear than for macrocyclic GBCAs. The highest amounts of Gd found in brain corresponded to less than 0.0002% of the injected dose per gram of tissue. Using LA-ICP-MS, high Gd concentrations in the deep cerebellar nuclei and in the granular layer of the cerebellar cortex were detected only for linear gadodiamide and gadopentetate dimeglumine but not for gadoteridol or gadobutrol. The energy dispersive x-ray spectroscopy analysis revealed Gd-containing spots in the skin of animals administered gadodiamide and gadopentetate dimeglumine. Transmission electron microscopy revealed several Gd-containing spots in the region of the dentate nuclei in the brain of 1 animal injected with gadodiamide. CONCLUSIONS: After repeated high dosing, nephrogenic systemic fibrosis-like macroscopic and histopathological lesions of the skin were observed only in some of the gadodiamide-treated animals. No histopathological findings were detected in the rodent brain. The administration of linear GBCAs was associated with significantly higher Gd concentrations in the brain and skin compared with macrocyclic GBCA administration. The results of LA-ICP-MS demonstrated local accumulation of Gd within the deep cerebellar nuclei and the granular layer only after the administration of linear agents. In summary, the detected low Gd concentrations in the skin and brain were well correlated with the higher kinetic stability of macrocyclic GBCA.


Brain/drug effects , Brain/metabolism , Contrast Media/pharmacokinetics , Gadolinium/pharmacokinetics , Skin/drug effects , Animals , Bone and Bones/drug effects , Bone and Bones/metabolism , Brain/ultrastructure , Contrast Media/administration & dosage , Contrast Media/adverse effects , Dose-Response Relationship, Drug , Gadolinium/administration & dosage , Gadolinium/adverse effects , Gadolinium DTPA/administration & dosage , Gadolinium DTPA/adverse effects , Gadolinium DTPA/pharmacokinetics , Heterocyclic Compounds/administration & dosage , Heterocyclic Compounds/adverse effects , Heterocyclic Compounds/pharmacokinetics , Injections, Intravenous , Male , Mass Spectrometry , Models, Animal , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Organometallic Compounds/administration & dosage , Organometallic Compounds/adverse effects , Organometallic Compounds/pharmacokinetics , Rats , Rats, Wistar , Retrospective Studies , Rodentia , Skin/ultrastructure
8.
Invest Radiol ; 52(7): 396-404, 2017 07.
Article En | MEDLINE | ID: mdl-28125438

OBJECTIVE: Multiple clinical and preclinical studies have reported a signal intensity increase and the presence of gadolinium (Gd) in the brain after repeated administration of Gd-based contrast agents (GBCAs). This bioanalytical study in rat brain tissue was initiated to investigate whether the residual Gd is present as intact GBCA or in other chemical forms by using tissue fractionation and chromatography. MATERIALS AND METHODS: Rats were divided randomly in 6 groups of 10 animals each. They received 10 daily injections of 2.5 mmol/kg bodyweight of 1 of 5 different GBCAs: linear GBCAs such as gadodiamide (Omniscan; GE Healthcare), gadopentetate dimeglumine (Gd-DTPA, Magnevist; Bayer), or gadobenate dimeglumine (Multihance; Bracco) and macrocyclic GBCAs such as gadobutrol (Gadovist; Bayer) and gadoterate meglumine (Gd-DOTA, Dotarem; Guerbet) or saline. On days 3 and 24 after the last injection (p.i.), 5 randomly chosen animals of each group were killed by exsanguination, and their brains were excised and divided into cerebrum, pons, and cerebellum. The brain sections were homogenized by sonication in ice-cold buffer at pH 7.4. Soluble and insoluble fractions were separated by centrifugation, and the soluble fractions were further separated by gel permeation chromatography (GPC). The Gd concentration in all tissue fractions and in the GPC eluate was measured by inductively coupled plasma-mass spectrometry. In a recovery control experiment, all GBCAs were spiked to blank brain tissue and more than 94% recovery of Gd in the tissue fractions was demonstrated. RESULTS: Only traces of the administered Gd were found in the rat brain tissue on day 3 and day 24 p.i. In the animals treated with macrocyclic GBCAs, Gd was found only in the soluble brain fraction and was present solely as low molecular weight molecules, most likely the intact GBCA. In the animals treated with linear GBCAs Gd was found to a large extent in the insoluble tissue fraction. The Gd concentration in the soluble fraction was comparable to the macrocyclic agents. According to GPC, a smaller portion of the Gd in the soluble fraction of the linear GBCAs groups was bound to macromolecules larger than 250 to 300 kDa. The nature of the Gd-containing macromolecules and the insoluble species were not determined, but they appeared to be saturable with Gd. The excretion of the soluble Gd species in the linear and macrocyclic GBCA groups was still ongoing between days 3 and 24 p.i. This was also observed for the macromolecular Gd species in the linear GBCA groups, but at a slower rate. CONCLUSIONS: The residual Gd found in the rat brain after repeated administration of all 3 linear GBCAs was present in at least 3 distinctive forms-soluble small molecules, including the intact GBCA, soluble macromolecules, and to a large extent in insoluble form. The latter 2 are most likely responsible for the prolonged signal intensity enhancement in brain structures observed in magnetic resonance imaging. No relevant differences between the 3 linear GBCAs were observed. The Gd concentrations in the brain after administration of macrocyclic GBCAs are lower, and the Gd is only present in soluble small molecules, which were slowly excreted. This underlines the crucial importance of the kinetic inertness of macrocyclic agents in the prevention of potential retention of Gd in the brain compared with the 3 linear, kinetically less restricted GBCAs.


Brain/metabolism , Contrast Media/pharmacokinetics , Gadolinium DTPA/pharmacokinetics , Meglumine/analogs & derivatives , Organometallic Compounds/pharmacokinetics , Animals , Chemical Fractionation/methods , Chromatography, Gel/methods , Contrast Media/administration & dosage , Gadolinium DTPA/administration & dosage , Meglumine/administration & dosage , Meglumine/pharmacokinetics , Models, Animal , Organometallic Compounds/administration & dosage , Rats , Rats, Wistar , Sodium Chloride/administration & dosage
9.
Eur Radiol ; 27(7): 2877-2885, 2017 Jul.
Article En | MEDLINE | ID: mdl-27832312

OBJECTIVE: Signal hyperintensity on unenhanced MRI in certain brain regions has been reported after multiple administrations of some, but not all, gadolinium-based contrast agents (GBCAs). One potential initial pathway of GBCA entry into the brain, infiltration from blood into the cerebrospinal fluid (CSF), was systematically evaluated in this preclinical study. METHODS: GBCA infiltration and distribution in the CSF were investigated in healthy rats using repeated fluid-attenuated MRI up to 4 h after high-dose (1.8 mmol/kg) administration of six marketed and one experimental GBCA. Additionally, gadolinium measurements in CSF, blood and brain tissue samples (after 24 h) were performed using inductively coupled plasma mass spectrometry. RESULTS: Enhanced MRI signals in the CSF spaces with similar distribution kinetics were observed for all GBCAs. No substantial differences in the gadolinium concentrations among the marketed GBCAs were found in the CSF, blood or brain tissue. After 4.5 h, the concentration in the CSF was clearly higher than in blood but was almost completely cleared and lower than the brain tissue concentration after 24 h. CONCLUSIONS: In contrast to the brain signal hyperintensities, no differences in penetration and distribution into the CSF of healthy rats exist among the marketed GBCAs. KEY POINTS: • Gadolinium-based contrast agents can cross the blood-CSF barrier. • Fluid-attenuated MRI shows GBCA distribution with CSF flow. • GBCA structure and physicochemical properties do not impact CSF penetration and distribution. • GBCA clearance from CSF was almost complete within 24 h in rats. • CSF is a potential pathway of GBCA entry into the brain.


Blood-Brain Barrier/physiology , Brain/diagnostic imaging , Cerebrospinal Fluid/metabolism , Gadolinium/pharmacokinetics , Magnetic Resonance Imaging/methods , Animals , Brain/metabolism , Contrast Media/pharmacokinetics , Male , Mass Spectrometry , Models, Animal , Rats , Rats, Wistar , Reference Values
10.
Adv Ther ; 33(1): 1-28, 2016 Jan.
Article En | MEDLINE | ID: mdl-26809251

UNLABELLED: In 1988, the first contrast agent specifically designed for magnetic resonance imaging (MRI), gadopentetate dimeglumine (Magnevist(®)), became available for clinical use. Since then, a plethora of studies have investigated the potential of MRI contrast agents for diagnostic imaging across the body, including the central nervous system, heart and circulation, breast, lungs, the gastrointestinal, genitourinary, musculoskeletal and lymphatic systems, and even the skin. Today, after 25 years of contrast-enhanced (CE-) MRI in clinical practice, the utility of this diagnostic imaging modality has expanded beyond initial expectations to become an essential tool for disease diagnosis and management worldwide. CE-MRI continues to evolve, with new techniques, advanced technologies, and novel contrast agents bringing exciting opportunities for more sensitive, targeted imaging and improved patient management, along with associated clinical challenges. This review aims to provide an overview on the history of MRI and contrast media development, to highlight certain key advances in the clinical development of CE-MRI, to outline current technical trends and clinical challenges, and to suggest some important future perspectives. FUNDING: Bayer HealthCare.


Contrast Media/administration & dosage , Magnetic Resonance Imaging/methods , Humans
11.
Invest Radiol ; 51(2): 83-9, 2016 Feb.
Article En | MEDLINE | ID: mdl-26606548

OBJECTIVES: In this prospective preclinical study, we evaluated T1-weighted signal intensity in the deep cerebellar nuclei (CN) and globus pallidus (GP) up to 24 days after repeated administration of linear and macrocyclic gadolinium-based contrast agents (GBCAs) using homologous imaging and evaluation methods as in the recently published retrospective clinical studies. In a second part of the study, cerebrospinal fluid (CSF) spaces were evaluated for contrast enhancement by fluid-attenuated magnetic resonance imaging (MRI). MATERIALS AND METHODS: Sixty adult male Wistar-Han rats were randomly divided into a control and 5 GBCA groups (n = 10 per group). The administered GBCAs were gadodiamide, gadopentetate dimeglumine, and gadobenate dimeglumine (linear GBCAs) as well as gadobutrol and gadoterate meglumine (macrocyclic GBCAs) and saline (control). Over a period of 2 weeks, the animals received 10 intravenous injections at a dose of 2.5 mmol Gd/kg body weight, each on 5 consecutive days per week. Before GBCA administration, as well as 3 and 24 days after the last injection, a whole-brain MRI was performed using a standard T1-weighted 3-dimensional turbo spin echo sequence on a clinical 1.5 T scanner. The ratios of signal intensities in deep CN to pons (CN/Po) and GP to thalamus (GP/Th) were determined. For the evaluation of the CSF spaces, 18 additional rats were randomly divided into 6 groups (n = 3 per group) that received the same GBCAs as in the first part of the study. After MR cisternography for anatomical reference, a fluid-attenuated inversion recovery sequence was performed before and 1 minute after intravenous injection of a dose of 1 mmol Gd/kg body weight GBCA or saline. RESULTS: A significantly increased signal intensity ratio of CN/Po was observed 3 and 24 days after the last injection of gadodiamide and gadobenate dimeglumine. No significant changes were observed between the 2 time points. Gadopentetate dimeglumine injection led to a moderately elevated but statistically not significant CN/Po signal intensity ratio. No increased CN/Po signal intensity ratios were determined in the MRI scans of rats that received macrocyclic GBCAs gadobutrol and gadoterate meglumine or saline. The ratio of signal intensity in GP/Th was not elevated in any group injected with GBCAs or saline. Enhanced signal intensities of CSF spaces were observed in the postcontrast fluid-attenuated inversion recovery images of all animals receiving GBCAs but not for saline. CONCLUSIONS: In this animal study in rats, increased signal intensity in the CN was found up to 24 days after multiple, extended doses of linear GBCAs. However, in contrast to clinical reports, the signal enhancement in the GP was not reproduced, demonstrating the limitations of this animal experiment. The elevated signal intensities remained persistent over the entire observation period. In contrast, no changes of signal intensities in either the CN or the GP were observed for macrocyclic GBCAs. However, all GBCAs investigated were able to pass the blood-CSF barrier in rats to a certain, not yet quantified extent.


Brain/anatomy & histology , Diffusion Magnetic Resonance Imaging/methods , Gadolinium/administration & dosage , Image Enhancement/methods , Animals , Contrast Media/administration & dosage , Dose-Response Relationship, Drug , Drug Administration Schedule , Macrocyclic Compounds/administration & dosage , Male , Rats , Rats, Wistar , Reproducibility of Results , Sensitivity and Specificity
12.
Invest Radiol ; 50(9): 671-8, 2015 Sep.
Article En | MEDLINE | ID: mdl-26207928

Over the last 120 years, the extensive advances in medical imaging allowed enhanced diagnosis and therapy of many diseases and thereby improved the quality of life of many patient generations. From the beginning, all technical solutions and imaging procedures were combined with dedicated pharmaceutical developments of contrast media, to further enhance the visualization of morphology and physiology. This symbiosis of imaging hardware and contrast media development was of high importance for the development of modern clinical radiology. Today, all available clinically approved contrast media fulfill the highest requirements for clinical safety and efficacy. All new concepts to increase the efficacy of contrast media have also to consider the high clinical safety standards and cost of goods of current marketed contrast media. Nevertheless, diagnostic imaging will contribute significantly to the progresses in medicine, and new contrast media developments are mandatory to address the medical needs of the future.


Contrast Media , Image Enhancement/methods , Magnetic Resonance Imaging/methods , Magnetic Resonance Imaging/trends , Radiography/methods , Radiography/trends , Humans , Tomography, X-Ray Computed/methods , Tomography, X-Ray Computed/trends
13.
Article En | MEDLINE | ID: mdl-25266498

When diagnosing cancer, assessing the nodal stage is tremendously important in determining the patient's prognosis. Computed tomography (CT) and magnetic resonance (MR) imaging (MRI) assessments of the regional lymph node (LN) size and shape are currently used for the initial nodal staging in clinical settings, although this approach has a rather low sensitivity, and biopsy often leads to restaging of the LNs. Acknowledging the great medical need to accurately stage LNs, scientists and clinicians have been working since the late 1980s on MR contrast agents that provide more reliable staging results. Different types of molecules (i.e., iron oxide nanoparticles and Gd-based contrast agent) have shown promising LN accumulation and imaging results, but no clinically approved, dedicated LN staging contrast agent is currently available. The literature describes a mechanism of contrast agent accumulation in the LNs that considers some but not all published experimental evidence. However, confidence in the mechanism of LN accumulation is a prerequisite for the directed synthesis of compounds for accurate and sensitive LN staging. To improve our understanding of the LN contrast agent accumulation mechanism, we reviewed the published data on the enrichment of colloidal MR contrast agent candidates in LNs, and we suggest an extended mechanism for contrast agent enrichment in LNs. For further clarification, physiology and results from drug targeting studies are considered where applicable.


Contrast Media , Lymph Nodes/pathology , Magnetic Resonance Imaging , Animals , Colloids , Contrast Media/adverse effects , Extravasation of Diagnostic and Therapeutic Materials/etiology , Humans
14.
Int J Nanomedicine ; 7: 4447-58, 2012.
Article En | MEDLINE | ID: mdl-22927759

PURPOSE: Magnetic resonance imaging (MRI), one of the most powerful imaging techniques available, usually requires the use of an on-demand designed contrast agent to fully exploit its potential. The blood kinetics of the contrast agent represent an important factor that needs to be considered depending on the objective of the medical examination. For particulate contrast agents, such as superparamagnetic iron oxide nanoparticles (SPIOs), the key parameters are particle size and characteristics of the coating material. In this study we analyzed the effect of these two properties independently and systematically on the magnetic behavior and blood half-life of SPIOs. METHODS: Eleven different SPIOs were synthesized for this study. In the first set (a), seven carboxydextran (CDX)-coated SPIOs of different sizes (19-86 nm) were obtained by fractionating a broadly size-distributed CDX-SPIO. The second set (b) contained three SPIOs of identical size (50 nm) that were stabilized with different coating materials, polyacrylic acid (PAA), poly-ethylene glycol, and starch. Furthermore, small PAA-SPIOs (20 nm) were synthesized to gain a global insight into the effects of particle size vs coating characteristics. Saturation magnetization and proton relaxivity were determined to represent the magnetic and imaging properties. The blood half-life was analyzed in rats using MRI, time-domain nuclear magnetic resonance, and inductively coupled plasma optical emission spectrometry. RESULTS: By changing the particle size without modifying any other parameters, the relaxivity r(2) increased with increasing mean particle diameter. However, the blood half-life was shorter for larger particles. The effect of the coating material on magnetic properties was less pronounced, but it had a strong influence on blood kinetics depending on the ionic character of the coating material. CONCLUSION: In this report we systematically demonstrated that both particle size and coating material influence blood kinetics and magnetic properties of SPIO independently. These data provide key information for the selection of a contrast agent for a defined application and are additionally valuable for other nano areas, such as hyperthermia, drug delivery, and nanotoxicology.


Contrast Media/chemistry , Contrast Media/pharmacokinetics , Iron/blood , Iron/chemistry , Magnetite Nanoparticles/chemistry , Oxides/blood , Oxides/chemistry , Acrylic Resins/chemistry , Animals , Cell Line , Half-Life , Iron/pharmacokinetics , Magnetic Resonance Imaging , Male , Mice , Oxides/pharmacokinetics , Particle Size , Polyethylene Glycols/chemistry , Rats , Rats, Wistar , Starch/chemistry , Structure-Activity Relationship , Surface Properties
15.
Invest Radiol ; 47(7): 383-91, 2012 Jul.
Article En | MEDLINE | ID: mdl-22659596

OBJECTIVE: Tumor imaging via molecular magnetic resonance imaging (MRI) that uses specific superparamagnetic iron oxide particles (SPIOs) has been addressed in the literature several times in the last 20 years. To our knowledge, none of the reported approaches is currently used for routine clinical diagnostic evaluation, nor are any in clinical development. This raises questions as to whether SPIO-enhanced molecular MRI is sensitive and specific enough for use in clinical practice. The aim of our preclinical study was to investigate the minimum requirements for obtaining sensitive molecular MRI for use in tumor evaluations under optimal conditions. The well-vascularized F9 teratocarcinoma tumor model, which exhibits high levels of the highly accessible target CD105 (endoglin), was used to compare the accumulation and visualization of target-specific SPIOs by MRI. MATERIAL AND METHODS: Superparamagnetic iron oxide particles were optimized in the following ways: (a) proton relaxivity was increased for higher imaging sensitivity, (b) a coating material was used for optimal loading density of the αCD105 antibody, and (c) binding activity to the target CD105 was increased. Binding activity and specificity were confirmed in vitro using enzyme-linked immunosorbent assay and in vivo using pharmacokinetic and biodistribution studies of 11 F9 teratoma-bearing mice together with micro-autoradiography. CD105 target expression was determined using immunohistochemistry and quantitative enzyme-linked immunosorbent assay. The transverse relaxation rate R2* was quantified by 3.0-T MRI in the tumors, kidneys, and muscles before and up to 60 minutes after injection in 11 mice. The use of [Fe]-labeled SPIOs for all in vivo experiments allowed for the direct correlation of the imaging results with SPIO accumulation. RESULTS: High-relaxivity αCD105-polyacrylic acid-SPIOs (r2 up to 440 L mmol Fe s) with strong binding activity accumulated specifically in tumors (1.4% injected dose/g) and kidneys (4.1% injected dose/g) in a manner dependent on the target concentration. The accumulation occurred within the first 3 minutes after injection. Visualization of specific SPIOs was accomplished with MRI. In contrast to the successful use of MRI in all examined kidneys (mean ± SEM ΔR2*, 61 ± 11 s), only 6 of 11 tumors (mean ± SEM ΔR2*, 15 ± 7 s) showed a clear signal when compared with the control even though optimal conditions were used. CONCLUSION: The accumulation of CD105-specific SPIOs in F9 mouse teratomas was robust. However, visualization of the specifically accumulated SPIOs by MRI was not reliable because of its limited signal detection sensitivity. We postulate that it will be challenging to improve the imaging properties of targeted SPIOs further. Therefore, molecular MRI by targeted SPIOs is currently not suitable for clinical tumor imaging using routinely applicable sequences and field strength.


Ferric Compounds , Intracellular Signaling Peptides and Proteins , Magnetic Resonance Imaging/methods , Molecular Imaging/methods , Nanoparticles , Neoplasms/diagnosis , Animals , Contrast Media , Disease Models, Animal , Endoglin , Enzyme-Linked Immunosorbent Assay , Mice , Neoplasms/pathology , Rats , Rats, Wistar , Sensitivity and Specificity , Statistics, Nonparametric
16.
Nanomedicine (Lond) ; 3(4): 437-52, 2008 Aug.
Article En | MEDLINE | ID: mdl-18694306

AIMS: The further development of diagnostic and therapeutic nanomedicines in research and their translation into clinical practice require appropriate characterization methods to ensure a reproducible quality and performance. However, many methods are insufficient for a detailed analysis of the particle size. The primary aim of the present work is to evaluate the application of asymmetrical flow-field-flow-fractionation (AF4) coupled with multiangle laser and dynamic light scattering (DLS) for the characterization of superparamagnetic iron oxide (SPIO) particles. METHODS: Eight carboxydextran-coated SPIO samples with different mean particle sizes, as determined by DLS, are investigated by means of an adequate AF4 separation method. RESULTS: In this work, we show that, with increasing sample particle size, as measured by DLS, the hydrodynamic and gyration radii obtained by the AF4 method increase respectively. We demonstrate that the applied AF4 method is able to separate nanoparticles of different sizes effectively, with superior reproducibility (relative standard deviation: <3%) and high accuracy (relative standard deviation: <10%). Furthermore, important characterization parameters that will affect the in vivo performance; namely, the shape factors and polydispersity indices, of all eight samples are presented. CONCLUSION: The work describes the application of AF4/DLS/multiangle laser light scattering as a highly useful method for characterization of SPIO particles, enabling valuable information to be accessed in addition to that obtained by transmission-electron microscopy and DLS in batch mode.


Fractionation, Field Flow/methods , Iron/analysis , Nanoparticles/analysis , Oxides/analysis , Dextrans , Ferrosoferric Oxide , Light , Magnetite Nanoparticles , Nanotechnology/methods , Reproducibility of Results , Scattering, Radiation
...