ArticlePDF Available

Gadolinium Retention, Brain T1 Hyperintensity, and Endogenous Metals: A Comparative Study of Macrocyclic Versus Linear Gadolinium Chelates in Renally Sensitized Rats

Authors:

Abstract and Figures

Objectives: This preclinical study was designed to compare gadolinium (Gd) brain uptake after repeated injections of a macrocyclic Gd-based contrast agent (GBCA) (gadoterate meglumine) or 2 linear GBCAs (L-GBCAs) (gadobenate dimeglumine or gadodiamide) on a translational model of moderate renal impairment in rats. Methods: The study was carried out in subtotally nephrectomized rats. Animals received 4 intravenous injections per week of GBCA (gadoterate meglumine, gadobenate dimeglumine, or gadodiamide) for 5 weeks, resulting in a cumulative dose of 12 mmol/kg, followed by a 1-month injection-free period. T1 hyperintensity in the deep cerebellar nuclei (DCNs) was investigated, and brain structures were carefully dissected to determine elemental Gd, iron (Fe), copper (Cu), and zinc (Zn) distribution by mass spectrometry. Urinary excretion of endogenous metals was also investigated soon after GBCA administration and several days later in order to assess a potential transmetalation phenomenon. Results: Unlike gadoterate, repeated injections of L-GBCAs gadobenate and gadodiamide both induced T1 hyperintensity in the DCNs. Fine dissection of cerebral and cerebellar structures demonstrated very low levels or absence of Gd after repeated injections of gadoterate, in contrast to the two L-GBCAs, for which the highest total Gd concentration was demonstrated in the DCNs (Gd concentration in DCNs after 4.5 weeks of injection-free period: 27.1 ± 6.5 nmol/g for gadodiamide [P < 0.01 vs saline and P < 0.05 vs gadoterate]; 12.0 ± 2.6 nmol/g for gadobenate [P < 0.09 vs saline]; compared with 1.4 ± 0.2 nmol/g for gadoterate [ns vs saline]). The distribution of Gd concentration among the various brain structures dissected was also well correlated with the Fe distribution in these structures. No difference in endogenous metal levels in brain structures was observed. However, injection of gadobenate or gadodiamide resulted in an increase in urinary Zn excretion (urinary Zn concentrations: 57.9 ± 20.5 nmol/mL with gadobenate [P < 0.01 vs gadoterate and saline] and 221.6 ± 83.3 nmol/L with gadodiamide [P < 0.0001 vs all other treatments] vs 8.1 ± 2.3 nmol/L with saline and 10.6 ± 4.8 nmol/L with gadoterate]). Conclusions: In a model of renally impaired rats, only traces of gadoterate meglumine were detected in the brain with no T1 hyperintensity of the DCNs, whereas marked Gd retention was observed in almost all brain areas after injections of the L-GBCAs, gadobenate dimeglumine and gadodiamide. Brain structures with higher Gd uptake corresponded to those structures containing more Fe. Urinary Zn excretion was significantly increased after a single injection of L-GBCAs.This is an open-access article distributed under the terms of the Creative Commons Attribution-Non Commercial-No Derivatives License 4.0 (CCBY-NC-ND), where it is permissible to download and share the work provided it is properly cited. The work cannot be changed in any way or used commercially without permission from the journal.
Content may be subject to copyright.
Gadolinium Retention, Brain T1 Hyperintensity,
and Endogenous Metals
A Comparative Study of Macrocyclic Versus Linear Gadolinium
Chelates in Renally Sensitized Rats
Marlène Rasschaert, MS,*†‡§||¶ Andréa Emerit, MS,* Nathalie Fretellier, PhD,* Cécile Factor, PhD,*
Philippe Robert, PhD,* Jean-Marc Idée, PharmD, MS,* and Claire Corot, PharmD, PhD*
Objectives: This preclinical study was designed to compare gadolinium (Gd)
brain uptake after repeated injections of a macrocyclic Gd-based contrast agent
(GBCA) (gadoterate meglumine) or 2 linear GBCAs (L-GBCAs) (gadobenate
dimeglumine or gadodiamide) on a translational model of moderate renal impair-
ment in rats.
Methods: The study was carried out in subtotally nephrectomized rats. Animals
received 4 intravenous injections per week of GBCA (gadoterate meglumine,
gadobenate dimeglumine, or gadodiamide) for 5 weeks, resulting in a cumulative
dose of 12 mmol/kg, followed by a 1-month injection-free period. T1 hyper-
intensity in the deep cerebellar nuclei (DCNs) was investigated, and brain struc-
tures were carefully dissected to determine elemental Gd, iron (Fe), copper (Cu),
and zinc (Zn) distribution by mass spectrometry. Urinary excretion of endoge-
nous metals was also investigated soon after GBCA administration and several
days later in order to assess a potential transmetalation phenomenon.
Results: Unlike gadoterate, repeated injections of L-GBCAs gadobenate and
gadodiamide both induced T1 hyperintensity in the DCNs. Fine dissection of ce-
rebral and cerebellar structures demonstrated very low levels or absenceof Gd af-
ter repeated injections of gadoterate, in contrast to the two L-GBCAs, for which
the highest total Gd concentration was demonstrated in the DCNs (Gd concentra-
tion in DCNs after 4.5 weeks of injection-free period: 27.1 ± 6.5 nmol/g for
gadodiamide [P< 0.01 vs saline and P< 0.05 vs gadoterate]; 12.0 ± 2.6 nmol/
g for gadobenate [P< 0.09 vs saline]; compared with 1.4 ± 0.2 nmol/g for
gadoterate [ns vs saline]). The distribution of Gd concentration among the various
brain structures dissected was also well correlated with the Fe distribution in these
structures. No difference in endogenous metal levels in brain structures was ob-
served. However, injection of gadobenate or gadodiamide resulted in an increase
in urinary Zn excretion (urinary Zn concentrations: 57.9 ± 20.5 nmol/mL with
gadobenate [P< 0.01 vs gadoterate and saline] and 221.6 ± 83.3 nmol/L with
gadodiamide [P< 0.0001 vs all other treatments] vs 8.1 ± 2.3 nmol/L with saline
and 10.6 ± 4.8 nmol/L with gadoterate]).
Conclusions: In a model of renally impaired rats, only traces of gadoterate
meglumine were detected in the brain with no T1 hyperintensity of the DCNs,
whereas marked Gd retention was observed in almost all brain areas after injec-
tions of the L-GBCAs, gadobenate dimeglumine and gadodiamide. Brain struc-
tures with higher Gd uptake corresponded to those structures containing more
Fe. Urinary Zn excretion was significantly increased after a single injection
of L-GBCAs.
Key Words: cerebellum, endogenous metals, gadobenate dimeglumine,
gadodiamide, gadolinium uptake, gadoterate meglumine,
magnetic resonance imaging, renal impairment
(Invest Radiol 2018;00: 0000)
The recent findings of gadolinium (Gd) accumulation in the brain
following repeated injections of Gd-based contrast agents (GBCAs)
have raised considerable interest in the scientific community, and this
accumulation could potentially represent a major concern for patients.
Health authorities, especially the US Food and Drug Administration
and the European Medicines Agency, have urged marketing authoriza-
tion holders to elucidate this phenomenon, which may require reevalu-
ation of the safety of GBCAs in everyday use.
It is generally accepted that Gd accumulation in tissues is much
higher with linear GBCAs (L-GBCAs) than macrocyclic agents and
is inversely proportional to their thermodynamic conditional stability
constant log Kcond, calculated for pH 7.4 and kinetic stabilities for
L-GBCAs and driven by the high kinetic stabilities (ie, very low disso-
ciation kinetics) of macrocyclic GBCAs (M-GBCAs),
1
as demonstrated
in nonclinical models
2,3
and in humans.
410
Gadolinium accumulation
in the central nervous system depends on the cumulative dose,
7
with
heterogeneous cerebral Gd distribution.
11,12
The detection of at-risk populations is of critical clinical impor-
tance, and recent clinical studies have focused on potentially sensitized
populations such as pediatric patients,
1315
newborns after potential ex-
posure by injection to the mother during pregnancy,
1618
or renally
impaired subjects.
19,20
Almost all preclinical studies are performed in rodent species,
which constitute a relevant translational model,
21,22
reproducing the
T1 effect and Gd accumulation in the deep cerebellar nuclei (DCNs).
A previous study demonstrated that moderate renal impairment
induced by subtotal nephrectomy constituted a sensitive model to study
brain Gd uptake in the rat. Indeed, it amplified the brain Gd retention
and T1 hyperintensity for an identical injected dose of gadodiamide
compared with animals with normal renal function.
23
Gadolinium up-
take was positively correlated with the severity of renal impairment.
24
We therefore decided to compare the behavior of different molecular
categories of GBCAs in this same model.
This model seems to be translationally and clinically relevant, as
the estimated prevalence of chronic kidney disease (CKD) in patients
70 years or older in the United States (estimated with the CKD-EPI
equation) is 46.8%,
24
and the vast majority of these patients present
stage 3 CKD, that is, moderate renal failure (glomerular filtration rate
between 30 and 59 mL/min per 1.73 m
2
).
24,25
Older people represent a population of major concern, in which
contrast-enhanced magnetic resonance imaging (MRI) examinations are
commonly performed. This population of patients may be repeatedly
Received for publication November 22, 2017; and accepted for publication, after revi-
sion, December 5, 2017.
From the *Guerbet Research and Innovation Department, Aulnay-sous-Bois; and
INSERM, U1196; Institut Curie, PSL Research University; §Université Paris
Sud; ||Université Paris-Saclay; and }CNRS, UMR 9187, Orsay, France.
Correspondence to: Marlène Rasschaert, MS, Guerbet Research and Innovation
Department, Guerbet, BP57400, 95943 Roissy CDG Cedex, France. E-mail:
marlene.rasschaert@guerbet-group.com.
Copyright© 2018 The Author(s). Published by Wolters Kluwer Health, Inc. This is an
open-access article distributed under the terms of the Creative Commons
Attribution-Non Commercial-No Derivatives License 4.0 (CCBY-NC-ND),
where it is permissible to download and share the work provided it is properly
cited. The work cannot be changed in any way or used commercially without per-
mission from the journal.
ISSN: 0020-99 96/18/00000000
DOI: 10.1097/RLI.0000000000000447
ORIGINAL ARTICLE
Investigative Radiology Volume 00, Number 00, Month 2018 www.investigativeradiology.com 1
exposed to linear Gd chelates, which are not contraindicated for pa-
tients with stage 3 CKD.
26,27
The purpose of this study was therefore to compare all categories
of GBCAs, that is, a linear ionic GBCA (gadobenate), a linear nonionic
GBCA (gadodiamide), and an M-GBCA (gadoterate), in a rat model of
moderate renal failure mimicking a substantial population of patients
exposed to GBCAs.
MATERIALS AND METHODS
All experimental procedures and animal care were carried out ac-
cording to French regulations and in compliance with European Direc-
tive 2010/63/EU on the protection of animals used for scientific
purposes. All experiments (administrations, imaging, image analyses,
and total Gd concentration measurements) were carried out blindly.
Animal Model and Administration Protocol
The study was carried out on 5/6th subtotally nephrectomized fe-
male Sprague-Dawley rats (SPF/OFA rats; Charles River, L'Arbresle,
France) aged 10 weeks and weighing 246 ± 15 g at the beginning of
the study. Subtotal nephrectomy was performed at Charles River Labo-
ratories: a first surgical procedure was performed when the rats were
6 weeks old to excise 1 kidney, and a second procedure was performed
1 week later to remove the 2 poles of the remaining kidney. After
2 weeks of recovery and acclimatization, the animals were randomized
(n = 10/group, except for the saline group, which included 9 rats). The
rats were housed 2 per cage, at an ambient temperature of 22°C ± 2°C,
hygrometry of 45% ± 10%, in a room with 12:12-hour light-dark cy-
cles. Rats had access to water and food ad libitum.
The animals received 20 injections of 0.6 mmol Gd/kg per in-
jection (1.2 mL/kg) of meglumine gadoterate (macrocyclic ionic
GBCA, Dotarem 500 mmol Gd/L [Guerbet, France], batches
14GD107A and 16GD091A), dimeglumine gadobenate (linear ionic
GBCA, MultiHance 500 mmol Gd/L [Bracco, Italy], batch SP6251H),
or gadodiamide (linear nonionic GBCA, Omniscan 500 mmol Gd/L
[GE Healthcare, Chalfont St Giles, United Kingdom], batch
12965458), and the control group received 0.9% saline solution
(CDM Lavoisier, Paris, France) (1.2 mL/kg). Intravenous injections
were performed in the tail once a day, 4 days a week for 5 weeks, un-
der isoflurane anesthesia (IsoFlo; Axience, Pantin, France). The 0.6-
mmol Gd/kg dose corresponds to the clinical dose (0.1 mmol Gd/kg)
adjusted to the body surface area of the rat according to the US Food
and Drug Administration guidelines.
28
A 4.5-week injection-free
period was observed after the last injection. The study protocol is
showninFigure1.
Creatinine Clearance
Endogenous creatinine clearance (CrCl) was determined from
plasma and 24-hour urine collected atthe beginning (ie, the week before
the first administration) of the study, after the administration period (the
week after the last administration), and at completion of the study (ie,
4 weeks after the last administration). Plasma and urine creatinine con-
centrations were assayed by an enzymatic technique on an Abbott Ar-
chitect ci8200 automated analyzer (Abbott, Rungis, France).
Magnetic Resonance Imaging Protocol
Magnetic resonance imaging procedures were performed once a
week, using a dedicated phased-array quadrature head coil in a gradient/
shims insert B-GA 12S HP (660 mT/m intensity and 4570 T/m/s max-
imum slew rate) on a 4.7 T preclinical magnet (Biospec 47/40; Bruker,
Ettlingen, Germany). The first MRI was performed before the first in-
jection, and subsequent MRI examinations were performed once a week
(just before the f irst injection of the week, ie, 72 hours after the last
injection of the previous week). An MRI examination consisted of a
T1-weighted 2-dimensional FLASH (fast low-angle shot) sequence
(repetition time/echo time, 50/1.78 milliseconds; 48 averages; in-
plane resolution, 164 164 μm
2
; slice thickness, 700 μm; acquisition
time 6 minutes 36 seconds), targeted exclusively on the cerebellum
(11 slices), and T1 mapping on the slice displaying the DCNs, using
a FAIR (flow-sensitive alternating inversion recovery)RARE (rapid
acquisition with relaxation enhancement) sequence with 8 inversion
times (0, 100, 200, 400, 600, 800, 1200, 2000 milliseconds; repetition
time/effective echo time, 36.9/2079.9 milliseconds; 4 averages; in-
plane resolution 164 164 μm
2
; slice thickness, 700 μm; and acqui-
sition time, 11 minutes 5 seconds).
FIGURE 1. Study protocol. Subtotally nephrectomized rats received 20 injections of 0.6 mmol/kg over 5 weeks (cumulative dose of 12 mmol Gd/kg).
Rasschaert et al Investigative Radiology Volume 00, Number 00, Month 2018
2www.investigativeradiology.com © 2018 Wolters Kluwer Health, Inc. All rights reserved.
Blood and Tissue Collection
On days 16, 30 (before the daily injection), 37, and 45 (before
the weekly MRI examination), rats were anesthetized with isoflurane,
and a 600-μL sublingual blood sample was drawn. Plasma was har-
vested after centrifugation and frozen at 20°C for subsequent determi-
nation of Gd concentrations. At completion of the study, on week 10
(day 66), the animals were anesthetized with 5% isoflurane in oxygen.
Sublingual venous blood was collected in heparinized tubes, and the
rats were subsequently killed by exsanguination via the abdominal
aorta. Venous plasma was collected after centrifugation and frozen at
20°C. For 4 to 6 rats per group (Fig. 1) (the other rats were used for
other experiments), the forebrain was carefully harvested, and the fol-
lowing structures: cortical forebrain, amygdala, olfactory bulbs, mid-
brain, hippocampus, hypothalamus, thalamus, and striatum, were
dissected.
29,30
The frozen cerebellum was sliced using a Brain Slicer
Matrix (Stoelting Co, Wood Dale, Ill), and the DCNs, cerebellum ex-
cept the DCNs (called cerebellum*), and brain stem were carefully
dissected. The DCNs was clearly distinguishable on 1-mm slices of
fresh tissue as shown in Figure 2. Tissues were then frozen at 20°C
for determination of total Gd concentrations.
Image Analysis
All image analyses were performed under blinded (for both test
groups and all time points) and randomized conditions. Both qualita-
tive and quantitative evaluations of the DCN T1 signal intensity
were performed.
Qualitative Analysis of MRI Scans
Qualitative evaluation of T1 signal enhancement in the DCNs
was performed under blinded conditions for the rat, group, and time
point. A 3-point scoring scale for the DCNs relative to adjacent areas
was applied: a score of 0 was attributed for no enhancement in the
DCNs, 1 for doubtful enhancement, and 2 for definite enhancement.
Quantitative Analysis of MRI Scans
Blinded quantitative analysis of signal intensity on randomized
images was performed by positioning regions of interest in the various
cerebellar structures: cerebellar parenchyma, brain stem, and left and
right DCNs. Signal intensity was calculated as the ratio of the DCNs with
the highest signal to the brain stem signal (DCN
max
-to-brain stem ratio).
R1 Mapping and Determination
R1 mapping was calculated from the FAIR-RARE acquisition on a
pixel-by-pixel basis using in-house software written in MATLAB (The
Mathworks Inc, Natick, Mass). The same regions of interest as those used
for the FLASH sequence were positioned, and the R1 value was extracted.
Determination of Tissue Total Gd, Iron, Copper, and
Zinc Concentrations
Total Gd concentrations in the various tissues collected were
determined by inductively coupled plasma mass spectrometry (ICP-
MS) (7700x; Agilent Technologies, Santa Clara, Calif ) after sample
mineralization in 65% nitric acid for 8 hours at a temperature of
80°C. The lower limits of quantification (LLOQs) of Gd were
0.02 nmol/mL in plasma, 0.14 nmol/g in DCNs, 0.03 nmol/g in hypo-
thalamus, and 0.02 nmol/g in other brain matrices. The LLOQs in
urine were 0.07 nmol/mL for Gd, 8 nmol/mL for iron (Fe), and
7 nmol/mL for copper (Cu) and zinc (Zn).
For calculation of means, SDs, and for statistical tests, values less
than the LLOQ were arbitrarily replaced by LLOQ, and values less than
the limit of detection were arbitrarily replaced by 0.
Urinary Excretion of Gd and Endogenous Metals
In addition to the 24-hour urine collection performed for deter-
mination of CrCl (before any injection [day 3], 4 days [day 36], and
1 month [day 64] after the last injection), rats were also placed in a me-
tabolism cage for 4 hours, starting 1 hour after the daily GBCA injec-
tion, at weeks 2 and 4 (days 11 and 25), in order to determine the
urinary excretion of endogenous metals (Fe, Zn, Cu) and total Gd fol-
lowing a GBCA injection.
Statistical Analysis
Values are shown as individual data, or mean ± SD. Dixon exclu-
sion test was used to exclude aberrant values at a 5% risk. Normality
was verified by the Shapiro-Wilk test.
31
A 2-way analysis of variance
with repeated measures was performed for CrCl and plasma Gd concen-
trations. When 1 or several parameters (time or group) were significant,
Tukey post hoc tests were applied to compare values for these parame-
ters. Two-way analysis of variance and Tukey post hoc tests were also
applied for quantification of T1 signal enhancement in DCNs, R1 map-
ping, and urinary excretion of metals. Kruskal-Wallis test and Dunn
post hoc test, when required, were used for simple comparison of total
Gd concentrations in the various brain tissues between groups. Pearson
correlation coefficient was calculated for correlations between metals.
Graphs present the values for all rats, but repeated-measure statistics
were performed only on values from rats that completed the study. A
significance level of 5% was adopted.
RESULTS
During the second MRI examination, 1 rat in the saline group died
of anesthesia, and 3 rats in the gadobenate group and 1 rat in the
gadodiamide group died during the study (2 rats in the gadobenate group
lost between 17% and 19% of body weight and were killed for ethical rea-
sons on days 21 and 52; the others were found dead, 1 in the gadodiamide
group on day 51 with a body weight loss of 23% and 1 in the gadobenate
group on day 57 with a body weight loss of 12%). Consequently, on com-
pletion of the study, the numbers of rats per group were 10 for gadoterate,
9 for gadodiamide, 8 for saline, and 7 for gadobenate.
Creatinine Clearance
Subtotal nephrectomy resulted in moderate renal impairment
(CrCl of tested rats was 0.21 ± 0.05 mL/min per 100 g prior to adminis-
tration of the test compounds). A transient improvement in renal function
was observed after the injection period (CrCl of 0.31 ± 0.09 mL/min per
100 g), independently of the group (P< 0.0001 for all groups). After the
injection-free period, CrCl decreased (P< 0.0001 for all groups vs post-
injection), reaching an intermediate value (CrCl of 0.25 ± 0.05 mL/min
FIGURE 2. Fresh cerebellar slices of 1 mm, starting (left) from the caudal extremity, obtained with the Brain Slicer Matrix. Coronal plane. Deep cerebellar
nuclei (red arrows) appear in light pink, surrounded by white matter.
Investigative Radiology Volume 00, Number 00, Month 2018 Comparative CNS Gd Uptake in Renally Impaired Rats
© 2018 Wolters Kluwer Health, Inc. All rights reserved. www.investigativeradiology.com 3
per 100 g) between the first 2 CrCl measurements. The rats in the
gadobenate and gadodiamide groups that died during the study had se-
verely impaired renal function: 0.09, 0.10, and 0.14 mL/min per 100 g
for the rats in the gadobenate group and 0.13 mL/min per 100 g for the
rat in the gadodiamide group at the last CrCl estimation.
Qualitative Evaluation of T1 Enhancement of DCNs on
T1-Weighted MRI Examinations
Typical images at week 10 are shown in Figure 3A. A progres-
sive and lasting increase in T1 enhancement of the DCNs compared
with surrounding areas was found in both the gadobenate and
gadodiamide groups from week 4, whereas no T1 enhancement was ob-
served with gadoterate or for the control group (score <0.5). The T1 en-
hancement effect was higher in the gadodiamide group than in the
gadobenate group (Fig. 3B).
Quantitative Evaluation of T1 Enhancement in the DCNs
Quantitatively, significant T1 enhancement was confirmed in the
gadodiamide group, from the third week of injections (P< 0.05) until
the end of the injection-free period (P< 0.01 vs the control group),
compared with the saline and gadoterate groups. The T1 enhancement
in the gadobenate group seemed to be intermediate (on completion of
the injection-free period, P= 0.074 compared with saline) (Fig. 4).
R1 Mapping
A trend toward an increase in R1 relaxation rate was observed af-
ter injections for gadobenate (P= 0.107 vs the control group), and a sig-
nificant increase in R1 relaxation rate was observed for gadodiamide
(P< 0.0001) compared with the control and gadoterate groups, whereas
the R1 relaxation rate in DCNs in the gadoterate group remained similar
to that of the saline group, regardless of the time point. R1 enhancement
was maintained after the injection-free period in the gadodiamide group
(P< 0.01 vs saline, P< 0.001 vs gadoterate) (Fig. 5).
Total Gd Concentrations Determined by ICP-MS in the
Dissected Brain Areas
The highest total Gd concentrations were observed with
gadodiamide in the DCNs, olfactory bulbs, striatum, thalamus, and cer-
ebellar parenchyma (Fig. 6).
Some of the dissected brain areas presented a high total Gd
concentration ratio for gadodiamide versus gadoterate (>7) and
FIGURE 3. A, Typical T1-weighted MR images (4.7 T) of all treated groups, at study completion (week 10). B, Qualitative scoring of T1 enhancement in
DCNs on weekly T1-weighted sequences (blinded). The DCN scores were attributed as follows: 0 = no T1 enhancement, 1 = doubtful T1 enhancement,
2 = definite T1 enhancement. All values are expressed as mean + SD.
FIGURE 4. Quantitative follow-up of T1 enhancement in the DCNs on
weekly T1-weighted sequences. T1 enhancement is described by the
DCNtobrain stem ratio of T1 signal intensity. First MRI after the injection
period is MRI 6. Gadodiamide versus saline and gadodiamide versus
gadoterate: P< 0.05 from the fourth MRI examination. All values are
expressed as mean + SD.
Rasschaert et al Investigative Radiology Volume 00, Number 00, Month 2018
4www.investigativeradiology.com © 2018 Wolters Kluwer Health, Inc. All rights reserved.
for gadodiamide versus gadobenate (>3.5), whereas this ratio was
close to 2 for the other structures. Statistical analyses comparing
the test groups for the structures more prone to store Gd are shown
in Figure 7.
No significant difference in terms of endogenous metal concen-
trations was observed between the test groups (data not shown). How-
ever, a good correlation was observed between tissue total Gd and
total Fe concentrations for the gadodiamide and gadobenate groups:
higher Fe concentrations in brain structures were correlated with higher
total Gd concentrations (Fig. 8, A and B). No correlation was observed
between Fe and total Gd concentrations in the gadoterate group
(Fig. 8C). Furthermore, total Gd concentration did not correlate with
tissue copper or Zn concentrations (Cu-Gd correlation: r= 0.6,
r=0.35,r= 0.53; and Zn-Gd correlation: r=0.15,r= 0.41, and
r=0.23, for gadodiamide, gadobenate, and gadoterate, respectively).
Plasma Total Gd Concentrations
After the injection period, plasma total Gd concentrations pro-
gressively decreased, but still remained above the lower limit of quanti-
fication on day 66, that is, 1 month after the last injection. Overall,
FIGURE 5. Relaxation rate R1 (s
1
) determined in the DCNs from T1 mapping sequences performed at the beginning of the study, after 5 weeks of
injections, and at completion of the study (4 weeks after the last injection). Circles: values for rats that did not complete the study. Individual values are
given, as well as mean ± SD. According to the Dixon exclusion test at a 5% risk, 1 value in gadobenate and gadodiamide groups at week 1 was excluded,
as well as 1 value in saline, gadobenate, and gadodiamide groups at week 10.
FIGURE 6. Total Gd concentrations determined by ICP-MS in the various dissected brain areas (cerebellum*: except for DCNs) 4½weeks after the last
injection (20*0.6 mmol Gd/kg body weight). Individual values are given, as well as mean ± SD.
Investigative Radiology Volume 00, Number 00, Month 2018 Comparative CNS Gd Uptake in Renally Impaired Rats
© 2018 Wolters Kluwer Health, Inc. All rights reserved. www.investigativeradiology.com 5
plasma Gd curves were similar between gadoterate and gadodiamide.
However, plasma total Gd concentrations in the gadobenate group were
significantly lower (P< 0.01 until day 30 compared with gadodiamide
and P< 0.05 until day 37 compared with gadoterate) (Fig. 9).
Urinary Excretion of Gd and Endogenous Metals
Immediately after injection ofGBCAs at days 11 and 25 (Fig. 1),
the urinary excretion of Gd was significantly increased (P<0.0001for
all vs saline) (Fig. 10). However, the Gd excretion was reduced by a fac-
tor of 3 to 4 in the case of gadobenate compared with the other GBCA-
treated groups (P< 0.0001). A nonsignificant increase in endogenous
urinary Fe excretion was observed on day 25 in the gadodiamide group
compared with the other 3 groups, whereas no difference in endogenous
urinary copper excretion was observed between the groups (group ef-
fect: P= 0.66) (Fig. 10).
At both days 11 and 25, a substantial increase in endogenous
urinary Zn excretion was observed after injection of gadobenate
(7-fold increase, P< 0.01 vs saline and gadoterate), and an even
greater increase was observed after injection of gadodiamide (27-
fold increase, P< 0.0001 vs other groups), whereas urinary Zn ex-
cretion after gadoterate was increased only 1.3-fold compared with
saline (not statistically significant).
DISCUSSION
Moderate renal impairment has been shown to represent a trans-
lational model of potentiation of brain Gd uptake, which isproportional
to baseline renal function.
23
Baseline CrCl measured at the beginning of
the study was 0.21 ± 0.05 mL/min per 100 g, comparable to the value
reported by Rasschaert et al
23
(0.19 ± 0.06 mL/min per 100 g), corre-
sponding to stage 3 CKD. The use of a nonclinical model of moderate
renal impairment would seem to be clinically relevant, because stage 3
CKD is very common in the elderly population, more prone to undergo
MRI examinations.
24,25
Three rats treated with gadobenate and 1 rat treated with
gadodiamide that were killed for ethicalreasons or that were found dead
presented the lowest CrCl values in their respective groups (CrCl values
between 0.09 and 0.15 mL/min per 100 g), suggesting that poor base-
line renal function compromises the safety of these L-GBCAs.
Qualitative and Quantitative Assessment of T1
Enhancement in the DCNs
T1 signal enhancement in the DCNs was monitored weekly by
T1-weighted MRI sequences on a 4.7 T nonclinical magnet. T1 signal
intensity in the DCNs was quantified with respect to brain stem signal
intensity, as classically assessed in both clinical and nonclinical studies,
although this area also accumulates Gd (total Gd concentrations of
1.4 ± 0.5 nmol/g for gadobenate, 3.6 ± 0.8 nmol/g for gadodiamide,
and 0.7 ± 0.09 nmol/g for gadoterate) and may consequently underesti-
mate T1 signal enhancement in the DCNs. Images were scored qualita-
tively under blinded conditions. Scoring of T1 signal intensity, starting
after 3 weeks of injections (ie, a cumulative dose of 7.2 mmol Gd/kg
body weight), revealed doubtful T1 signal enhancement in the DCNs
after gadobenate administrations and definite T1 signal enhancement
after gadodiamide administrations, whereas no effect was observed in
the gadoterate group. Analysis of the DCNtobrain stem ratio clearly
distinguished between the group treated with gadodiamide for 3 weeks
and the saline and gadoterate groups, and this ratio continued to
slightly increase, even during the injection-free period. Regarding
the gadobenate-treated group, intermediate T1 signal enhancement
was observed in the DCNs (mean, 55% ± 16% increase of the signal ra-
tio compared with the signal ratio observed with gadodiamide). No
FIGURE 7. Total Gd concentration in the main brain structures that accumulate Gd (cerebellum*: all parenchyma except for the DCNs). Individual values
are given, as well as mean ± SD. According to the Dixon exclusion test at a 5% risk, 1 saline-treated rat was excluded for DCNs, 1 saline and 1
gadoterate-treated rats for olfactory bulbs, and 1 gadoterate-treated rat for the cerebellum*(*P<0.05;**P<0.01;***P< 0.001).
Rasschaert et al Investigative Radiology Volume 00, Number 00, Month 2018
6www.investigativeradiology.com © 2018 Wolters Kluwer Health, Inc. All rights reserved.
effects on the T1 signal of the DCNs were demonstrated in the gadoterate
group, regardless of the time point.
R1 mapping confirmed T1 signal enhancement in the DCNs
with gadobenate and gadodiamide, even after the administration-free
period for gadodiamide (ie, at week 10).
Persistence of T1 signal enhancement throughout the study, de-
spite reports that Gd is partially cleared from the tissues,
11,32
suggests
a change in the form of Gd stored in brain tissues. The total Gd
concentration would be expected to be lower on day 66 than on day
45, but the T1 signal ratio actually increased or remained relatively sta-
ble, suggesting that residual Gd is transformed into a storage form that
enhances the T1 effect, or that Gd is cleared more rapidly from the brain
stem than from the DCNs. It can be hypothesized that Gd dissociated
from L-GBCAs progressively binds to (yet unidentified) macromole-
cules, as recently shown.
33
Gianolio et al
34
suggested that this form of
Gd could be responsible for the majority of the T1 signal enhancement.
Total Gd Concentrations in Brain Structures
The highest Gd concentration in the DCNs was observed
with the 2 L-GBCAs: 27.1 ± 6.5 nmol/g for gadodiamide and
12.0 ± 2.6 nmol/g for gadobenate, that is, 20- and 9-fold higher than
the total Gd concentration measured in the DCNs with gadoterate, re-
spectively. Furthermore, high Gd accumulation in the olfactory bulbs
was observed after administration of L-GBCAs (consistent with the re-
sults observed in mice),
11
and interestingly, the olfactory bulbs are also
the major area of storage of manganese after exposure reported in both
rodents (oral exposure)
35
and humans (respiratory exposure).
36
While total Gd concentration ratios for many brain structures
(amygdala, cortical forebrain, hippocampus, hypothalamus, brain stem,
midbrain) were equal to approximately 2 between gadodiamide and
gadobenate and between gadobenate and gadoterate, some structures
seemed to store higher proportions of Gd after gadodiamide injections
compared with gadobenate injections: olfactory bulb (ratio of 4.8 be-
tween gadodiamide and gadobenate), cerebellar cortex (ratio of 4.0),
and striatum (ratio of 3.4). As gadodiamide is the GBCA more prone
to dissociate in vivo and release free Gd
3+
,
33
we can speculate that these
structures are more likely to accumulate Gd in a dissociated form than
the other structures.
Total Gd concentrations observed in brain areas in our study were
1.5-2-fold higher than those reported by Kartamihardja et al,
11
but with
generally the same order of distribution between brain structures. These
authors studied the distribution and washout of gadodiamide and
gadoterate in renally impaired mice. These discrepancies in total Gd
concentration could be explained by differences in the species studied
(rat vs mouse), the renal impairment model (subtotal nephrectomy vs
electrocoagulation), the washout time in the present study, and the cu-
mulative dose (100 mmol Gd/kg in mice, which would be equivalent
to 50 mmol/kg for rats, after adjustment for body surface area, versus
12 mmol Gd/kg in our study).
28
Total Gd concentrations observed in the various brain structures
after administration of L-GBCAs were correlated with Fe concentra-
tion, but not with Zn and Cu concentrations. These results support the
possibility of Gd versus Fe transmetalation for L-GBCAs. The thermo-
dynamic constants are much higher for Fe
3+
(eg, for Fe-BOPTA:
FIGURE 8. Correlations between mean brain tissue total Gd and
Fe concentrations measured in brain areas for the various GBCAs
(cerebellum*: all cerebellar parenchyma except for the DCNs). A, Tissue
Fe versus total Gd concentrations in gadodiamide-treated rats. B, Tissue
Fe versus total Gd concentrations in gadobenate-treated rats. C, Tissue
Fe versus total Gd concentrations in gadoterate-treated rats.
FIGURE 9. Time course of plasma total Gd concentration over the
entire study. During the injection period (days 16 and 30), plasma was
collected just before the second injection of the week (24 hours after
the 9th injection and 24 hours after the 17th injection).
Investigative Radiology Volume 00, Number 00, Month 2018 Comparative CNS Gd Uptake in Renally Impaired Rats
© 2018 Wolters Kluwer Health, Inc. All rights reserved. www.investigativeradiology.com 7
log K
cond
= 23.4) than for Cu
2+
or Zn
2+
(log K
cond
of Cu-BOPTA = 17.3
and log K
cond
of Zn-BOPTA = 13.9),
1
and Fe concentrations are higher.
However, it should be noted that only the labile fraction of the Fe pool
(mainly in the Fe
2+
form) is susceptible to transmetalation. No differ-
ence in total endogenous metal concentrations in brain structures was
observed between the various groups, which can be explained by the
fact that ICP-MS is an elemental technique that does not take into ac-
count the labile fraction of the metals. Another possible explanation
for the Gd-Fe correlation could be that Gd and Fe access brain areas
such as the DCNs, olfactory bulb, or striatum via the same pathways.
Interestingly, it has been reported that the brain areas associated
with T1 signal enhancement after more than 35 administrations of
L-GBCAs to patients were the posterior thalamus, substantia nigra,
red nucleus, cerebellar peduncle, colliculi, dentate nucleus, and globus
pallidus,
37
that is, the brain structures associated with the highest
Fe concentrations.
38
Plasma Gd Concentration
A classic pharmacokinetic profile was observed for plasma total
Gd concentrations during the study. However, plasma Gd was still de-
tected 1 month after the last injection. Plasma total Gd concentrations
were lower following administration of gadobenate at all time points
compared with the other GBCA groups, which could be attributed to
specific biliary excretion of this GBCA related to its aromatic moiety,
especially in the context of renal impairment and in the rat species.
3941
This excretion pattern has been previously described in renally impaired
rats.
42
In the case of gadobenate, the rat model may underestimate the
Gd concentrations, because of a different pharmacokinetics profile
and excretion pathway in this species compared with the human spe-
cies. Indeed, only 3% to 5% of gadobenate is taken up by the hepato-
cytes in humans, while this phenomenon accounts for approximately
50% of the molecule in the rat with normal renal function.
39
Urinary Excretion of Gd and Endogenous Metals
Urinary excretion of Gd and endogenous metals (Fe, Zn, Cu)
was determined immediately after GBCA injection, at days 11 and 25
and on days 36 and 64, in a context of tissue Gd retention. As found
in the plasma Gd concentrations, Gd urinary excretion was reduced in
gadobenate in comparison with gadoterate and gadodiamide, because
of the specific hepatic excretion of this molecule. The Gd urinary excre-
tion is reduced by a factor of 3 to 4. In this model, the hepatic excretion
would then represent approximately 70% of gadobenate excretion. Re-
garding endogenous metals, a substantial increase in urinary Zn con-
centrations was observed immediately after injections of gadodiamide
and gadobenate. Urinary Zn excretion occurring immediately after
GBCA injection has been described in patients receiving a single ad-
ministration of the L-GBCA gadodiamide and, to a lesser extent,
gadopentetate, but not with gadoterate.
43
In the case of gadodiamide,
the excess free ligand caldiamide (5% wt/vol) can chelate endogenous
metals, and by extrapolating from data in humans, a significant propor-
tion of plasma total and labile Zn could be available for chelation com-
pared with plasma Fe or copper.
4346
The affinity constant (log K
therm
) for Zn-DTPA-BMA is 12.04 ver-
sus 7.17 for Ca-DTPA-BMA.
37
However, although the pharmaceutical
FIGURE 10. Urinary excretion of endogenous Zn, Fe, and Cu (measured by ICP-MS) at various time points (day 3: before injection period; day 36:
after the injection period; day 64: after injection-free period), and 1 to 5 hours after injection of GBCAs (at day 11 and day 25). Individual values
are given, as well as mean.
Rasschaert et al Investigative Radiology Volume 00, Number 00, Month 2018
8www.investigativeradiology.com © 2018 Wolters Kluwer Health, Inc. All rights reserved.
solution of gadobenate does not contain any added free ligand in the
pharmaceutical solution,
47
it induced a significant increase in urinary
excretion of endogenous Zn. Therefore, the most plausible explana-
tion for this phenomenon is a transmetalation phenomenon occurring
between Gd
3+
and Zn
2+
, possibly facilitated by the presence of ele-
ments capable of binding Gd
3+
, such as proteins and PO
4
3
, displacing
the equilibrium.
4851
Gadolinium versus Zn transmetalation may
therefore also be responsible for part of the Zn excretion observed
with gadodiamide.
Overall, clinical and nonclinical studies published over recent
years have clearly demonstrated that less thermodynamically and kinet-
ically stable GBCAs are associated with higher Gd accumulation in
brain and body tissues, with a tropism for certain structures, such as
the DCNs. Although all GBCAs enter cerebrospinal fluid and brain tis-
sue via the choroid plexus, M-GBCAs remain chelated and return to the
circulation to be subsequently eliminated in urine, whereas less stable
L-GBCAs rapidly dissociate in the tissues, and Gd is therefore trapped
in the brain. Gianolio et al
34
recently showed that, after 22 injections of
0.6 mmol/kg of gadodiamide in the rat, the tissues studied only 3 days
after the last injection mostly contained dissociated Gd (<20% of che-
lated Gd in the cerebellum and 4% in the cerebrum). In contrast, only
very low levels of the M-GBCA, gadoteridol, were observed and en-
tirely in its original chelated form. Frenzel et al
33
reported fairly similar
results, with a decreasing proportion of chelated Gd over time (3-day
injection-free period vs 24-day injection-free period).
According to one hypothesis of dechelation, chelated Gd could
form a ternary complex with PO
4
3
, which would then allow dechelation
of an intermediate state Gd
3+
(PO
4
3
) from the ligand.
5153
Once
dechelated, Gd either remains in this precipitated form or is immedi-
ately bound to macromolecules (thereby leading to T1 hyperintensity),
either peptides or proteins.
33,54
The coexistence or predominance of
these various forms would putatively lead to different levels and forms
of toxicity.
In conclusion, in a sensitive translational model of a common at-
risk population, only traces of Gd were observed in the brain following
injections of gadoterate, in contrast to the linear Gd chelates gadobenate
and gadodiamide. Gadolinium brain uptake from L-GBCAs is associ-
ated with T1 hyperintensity in the DCNs, which could be due to binding
of dissociated and soluble Gd derived from L-GBCAs to macromole-
cules. Furthermore, the global distribution of Gd after administration
of L-GBCAs (but not the macrocyclic gadoterate) in brain areas was
correlated with the local tissue distribution of Fe, which supports the
possibility of Gd versus Fe transmetalation or is a hint for the same
pathway to access the brain. The precise localization of Gd tissue stor-
age and identification of the Gd-binding macromolecules have yet to be
documented, as well as long-term putative neurotoxic effects associated
with repeated administration of L-GBCAs.
ACKNOWLEDGMENTS
The authors thank Evangeline M'Boumba, MS, for the Gd deter-
mination in tissues, and Anthony Saul, PhD, and Hélène Poenaru, for
reviewing the English language. The authors also thank Jean-Luc
Guerquin-Kern, PhD; Sergio Marco, PhD; and Jean-Pierre Laissy,
MD, PhD, for helpful discussions.
REFERENCES
1. Port M, Idée JM, Medina C, et al. Efficiency, thermodynamic and kinetic stability
of marketed gadolinium chelates and their possible clinical consequences: a critical
review. Biometals. 2008;21:469490.
2. Robert P, Violas X, Grand S, et al. Linear gadolinium-based contrast agents are as-
sociated with brain gadolinium retention in healthy rats. Invest Radiol.2016;51:
7382.
3. Jost G, Lenhard DC, Sieber MA, et al. Signal increase on unenhanced T1-weighted
images in the rat brain after repeated, extended doses of gadolinium-based con-
trast agents: comparison of linear and macrocyclic agents. Invest Radiol. 2016;
51:8389.
4. White GW, Gibby WA, Tweedle MF. Comparison of Gd(DTPA-BMA) (Omniscan)
versus Gd(HP-DO3A) (ProHance) relative to gadolinium retention in human
bone tissue by inductively coupled plasma mass spectroscopy. Invest Radiol.
2006;41:272278.
5. Errante Y, Cirimele V, Mallio CA, et al. Progressive increase of T1 signalintensity
of the dentate nucleus on unenhanced magnetic resonance images is associated
with cumulative dosesof intravenously administered gadodiamide in patientswith
normal ren al function, suggesting dechelation. Invest Radiol. 2014;49:6 85690.
6. Weberling LD, Kieslich PJ, Kickingereder P, et al. Increased signal intensity in the
dentate nucleus on unenhanced T1-weighted images after gadobenate dimeglumine
administration. Invest Radiol. 2015;50:743748.
7. McDonald RJ, McDonald JS, Kallmes DF, et al. Intracranial gadolinium deposi-
tion after contrast-enhanced MR imaging. Radiology. 2015;275:772782.
8. Radbruch A, Weberling LD, KieslichPJ, et al. Gadolinium retention in the dentate
nucleus andglobus pallidus is dependent on the class of contrast agent.Radiology.
2015;275:783791.
9. Kanda T, Ishii K, Kawaguchi H, et al. High signal intensity in the dentate nucleus
and globuspallidus on unenhanced T1-weighted MRimages: relationship with in-
creasing cumulative dose of a gadolinium-based contrast material. Radiology.
2014;270:834841.
10. Radbruch A, Weberling LD, Kieslich PJ, et al. Intraindividual analysis of signal
intensity changes in the dentate nucleus afterconsecutive serial applications of lin-
ear and macrocyclic gadolinium-based contrast agents. Invest Radiol. 2016;51:
683690.
11. Kartamihardja AA, Nakajima T, Kameo S, et al. Distribution and clearance of
retained gadolinium inthe brain: differences between linear and macrocyclic gad-
olinium based contrast agents in a mouse model. Br J Radiol. 2016;89:20160509.
12. Lohrke J, Frisk AL, Frenzel T, et al. Histology and gadolinium distribution in the
rodent brain after the administration of cumulative high doses of linear and mac-
rocyclic gadolinium-based contrast agents. Invest Radiol. 2017;52:324333.
13. Scala M, Koob M, de Buttet S, et al. A Pharmacokinetics, efficacy, and safety
study of gadoterate meglumine in pediatric subjects aged younger than 2 years.
Invest Radiol. 2018;53:7079.
14. Roberts DR, Chatterjee AR, Yazdani M, et al. Pediatric patients demonstrate pro-
gressive T1-weighted hyperintensity in the dentate nucleus following multiple
doses of g adoliniu m-based c ontrast a gent. AJNR Am J Neuroradiol.2016;37:
23402347.
15. Renz DM, Kümpel S, Böttcher J, et al. Comparison of unenhanced T1-weighted
signal intensities within the dentate nucleus and theglobus pallidus after serial ap-
plications of gadopentetate dimeglumine versus gadobutrol in a pediatric popula-
tion. Invest Radiol. 2018;53 :119127.
16. Prola-Netto J, Woods M, Roberts VHJ, et al. Gadolinium chelate safety in preg-
nancy: barely detectable gadolinium levels in the juvenile nonhuman primate
after in utero exposure [published online ahead of print September 4, 2017].
Radiology. 2017.
17. Erdene K, Nakajima T, Kameo S, et al. Organ retention of gadolinium in mother
and pup mice: effect of pregnancy and type of gadolinium-based contrast agents.
Jpn J Radiol. 2017;35 :568573.
18. Khairinisa MA, Takatsuru Y, Amano I, et al. The effect of perinatal gadolinium-
based contrast agents on adult mice behavior. Invest Radiol. 2018;53:1 10118.
19. Barbieri S, Schroeder C, Froehlich JM, et al. High signal intensity in dentate nu-
cleus and globus pallidus on unenhanced T1-weighted MR images in three pa-
tients with impaired renal function and vascular calcification. Contrast Media
Mol Imaging. 2016;11:2 45250.
20. Cao Y, Zhang Y, Shih G, et al. Effect of renal function on gadolinium-related sig-
nal increases on unenhanced T1-weighted brain magnetic resonance imaging. In-
vest Radiol. 2016;51 :677682.
21. Cenci MA, Whishaw IQ, Schallert T. Animal models of neurological deficits: how
relevant is the rat? Nat Rev Neurosci. 2002;3:574579.
22. Shah A, Garzon-Muvdi T, Mahajan R, et al. Animal models of neurological dis-
ease. Adv Exp Med Biol. 2010;671:2340.
23. Rasschaert M, Idée JM, Robert P, et al. Moderate renal failure accentuates T1 sig-
nal enhancement in the deep cerebellar nuclei of gadodiamide-treated rats. Invest
Radiol. 2017;52:255264.
24. Stevens LA, Viswanathan G, Weiner DE. Chronic kidney disease and end-stage
renal disease in the elderly population: current prevalence, future projections,
and clinical significanc e. Adv Chronic Kidney Dis. 2010;17:293301.
25. Duru OK, Vargas RB, Kermah D, et al. Highprevalence of stage 3 chronic kidney
disease in older adults despite normal serum creatinine. J Gen Intern Med.2009;
24:8692.
26. European Medicine Agency. Assessment report for gadolinium-containing con-
trast agents. Procedure No. EMEA/H/A-31/1097. July 1, 2010. Available at:
http://www.ema.europa.eu/docs/en_GB/document_library/Referrals_document/
gadolinium_31/WC500099538.pdf. Accessed 14 July, 2017.
Investigative Radiology Volume 00, Number 00, Month 2018 Comparative CNS Gd Uptake in Renally Impaired Rats
© 2018 Wolters Kluwer Health, Inc. All rights reserved. www.investigativeradiology.com 9
27. Food and DrugAdministration.FDA Drug Safety Communication:new warnings
for using gadolinium based contrast agents in patients with kidney dysfunction.
October 9, 2010. Available at: http://www.fda.gov/Drugs/DrugSafety/
ucm223966.htm. Accessed 14 July, 2017.
28. Food and Drug Administration. US Department of Health and Human Services.
Center for Drug Evaluation and Research. Guidance for Industry. Estimating the
maximum safestarting dose in initialclinical trials for therapeuticsin adult healthy
volunteers. July 6, 2005. Available at: http://www.fda.gov/downloads/Drugs//
Guidances/UCM078932.pdf. Accessed June 15, 2017.
29. Tissue prep and protein assay 2012. Available at: https://fr.scribd.com/document/
317128344/Tissue-Prep-and-Protein-Assay-2012. Accessed 5 December, 2017.
30. Spijker S. Dissection of rodent brain regions. In: Li KW, ed. Neuroproteomics
(Neuromethods, Volume 57). New York, NY: Humana Press; 2011:1326.
31. Shapiro SS, Wilk MB. An analysis of variance test for normality (complete sam-
ples). Biometrika. 1965;52:591611.
32. Smith AP, Marino M, Roberts J, et al. Clearance of gadolinium fromthe brain with
no pathologic effect after repeated administration of gadodiamide in healthy rats:
an analytical and histologic study. Radiology. 2017;282:743751.
33. Frenzel T, Apte C, Jost G, et al. Quantification and assessment of the chemical
form of residual gadolinium in the brain after repeated administration of
gadolinium-based contrast agents: comparative study in rats. Invest Radiol.
2017;52:396404.
34. Gianolio E, Bardini P, Arena F, et al. Gadolinium retention in the rat brain: assess-
ment of the amounts of insoluble gadolinium-containing species and intact gado-
linium complexes after repeated administration of gadolinium-based contrast
agents. Radiology. 2017;285:83 9849.
35. O'Neal SL, Hong L, Fu X, et al. Manganese accumulation in bone following
chronic exposure in rats: steady-state concentration and half-life in bone. Toxicol
Lett. 2014;229:93100.
36. Sen S, FlynnMR, Du G, et al. Manganeseaccumulation in theolfactory bulbs and
other brain regions of asymptomaticwelders. Toxicol Sci. 2011;121:160167.
37. Zhang Y, Cao Y, Shih GL, et al. Extent of signal hyperintensity on unenhanced
T1-weighted brain MR images after more than 35 administrations of linear
gadolinium-based contrast agents. Radiology. 2017;282:516525.
38. Hallgren B, Sourander P. The effect of age on the non-haemin iron in the human
brain. JNeurochem. 1 958;3:4151.
39. Lorusso V, Arbughi T, Tirone P, et al. Pharmacokinetics and tissue distribution
in animals of gadobenate ion, the magnetic resonance imaging contrast en-
hancing component of gadobenate dimeglumine 0.5 M solution for injection
(MultiHance). J Comput Assist Tomogr. 1999;23 suppl 1:S181S194.
40. Kirchin MA, Lorusso V, Pirovano G. Compensatory biliary and urinary excretion
of gadobenate ion after administration of gadobenate dimeglumine (MultiHance
(®)) in cases of impaired hepatic or renal function: a mechanism that may aid in
the prevention of nephrogenic systemic fibrosis? Br J Radiol. 2015;88 :20140526.
41. Ersoy H, Rybicki FJ. Biochemical safetyprofiles of gadolinium-based extracellu-
lar contrast agents and nephrogenic systemic fibrosis. J Magn Reson Imaging.
2007;26:11901197.
42. Fretellier N, Idée J, Bruneval P, et al. Hyperphosphataemia sensitizes renally im-
paired rats to the profibrotic effects of gadodiamide. Br J Pharmacol. 2012;165:
11511162.
43. Kimura J,Ishiguchi T, Matsuda J, et al. Human comparative study of zinc and cop-
per excretion via urine after administration of magnetic resonance imaging con-
trast agents. Radiat Med. 2005;23:322326.
44. Zalewski P, Truong-Tran A, Lincoln S, et al. Use of a zinc fluorophore to measure
labile pools of zinc in body fluids and cell-conditioned media. Biotechniques.
2006;40:509520.
45. Mills E, Dong XP, Wang F, et al. Mechanisms of brain iron transport: insight
into neurodegeneration and CNS disorders [review]. Future Med Chem. 2010;
2:5164.
46. Cabantchik ZI. Labile iron in cells and body fluids: physiology, pathology, and
pharmacology [review]. Front P ha rm ac ol. 2014;5: 45.
47. Idée JM, Port M, Robic C, et al. Role of thermodynamic and kinetic parameters in
gadolinium chelate stability. J Magn Reson Imaging. 2009;30:124 91258.
48. Tweedle MF, Hagan JJ, Kumar K, et al. Reaction of gadolinium chelates with en-
dogenously available ions. Magn Reson Imaging. 1991;9:409415.
49. Taupitz M, Stolzenburg N, Ebert M, et al. Gadolinium-containing magnetic reso-
nance contrast media: investigation on the possible transchelation of Gd
3+
to the
glycosaminoglycan heparin. Contrast Media Mol Imaging. 2013;8:108116.
50. Laurent S, Elst LV, Copoix F, et al. Stabilityof MRI paramagnetic contrast media:
a proton relaxometric protocol for transmetallation assessment. Invest Radiol.
2001;36:115122.
51. Idée JM, Fretellier N, Robic C, et al. The role of gadolinium chelatesin the mech-
anism of nephrogenic systemic fibrosis: a critical update. Crit Rev Toxicol.2014;
44:895913.
52. Baranyai Z, Pálinkás Z, Uggeri F, et al. Dissociation kinetics of open-chain and
macrocyclic gadolinium(III)-aminopolycarboxylate complexes related to mag-
netic resonance imaging: catalytic effect of endogenous ligands. Chemistry.
2012;18:1642616435.
53. Robic C, CatoenS, de Goltstein MC, et al.The role of phosphateon Omniscan(®)
dechelation: an in vitro relaxivity study at pH 7. Biometals. 2011;24:759768.
54. Knoepp F, Bettmer J, Fronius M. Gadolinium released by the linear gadolinium-
based contrast-agent Gd-DTPA decreases the activity of human epithelial Na
+
channels (ENaCs). Biochim Biophys Acta. 2017;1859:10401048.
Rasschaert et al Investigative Radiology Volume 00, Number 00, Month 2018
10 www.investigativeradiology.com © 2018 Wolters Kluwer Health, Inc. All rights reserved.
... Similarly, as observed by McDonald and McDonald (2020), the rapidly growing body of literature has refuted the postulation of GBCAs in-vivo maintained stability post intravenous administration and in excreted urine. Convincing clinical and laboratory pieces of evidence of retained Gd in tissues of virtually all patients administered GBCA injection, particularly the linear chelates, regardless of the state of their renal functions' status, indicates some degree of tissue retention of dechelated Gd (Penfield and Reilly, 2007)(Le Fur and Caravan, 2019) (Telgmann et al., 2012b) (Rasschaert et al., 2018). ...
... Generally, it was observed that the linear GBCAs have higher degradation susceptibility compared to the macrocyclic chelates (Aime, 2019;Aime and Caravan, 2009;Rasschaert et al., 2018;Runge, 2018;Sieber et al., 2008;Thomsen, 2017). The macrocyclic ligand structure roundly embedded the Gd atom like a cast-iron structure, while the linear chelates have a more flexible ligand bond with a tail-like structure partially securing the Gd atom as depicted in Fig. 2(a) Gadoterate meglumine (Dotarem)-Macrocyclic structure and Fig. 2(b) Gadopentetate dimeglumine, (Magnevist)-Linear structure respectively (Le Fur and Caravan, 2019). ...
... The in vivo displacement of Gd 3+ from its medical chelating agents (despite its high stability constants) by these substituents metal ions has been reported by several authors (Port et al., 2008). The competitive tendencies of several endogenous cations substituents (Fe 3+ , Zn 2+ , Cu 2+ and Ca 2+ ) with the Gd 3+ cation for the chelating ligand have also been reported by Telgmann et al. (2012aTelgmann et al. ( , 2012b in their study which investigate the consequence of potential iron (Fe 3+ ) transmetalation in blood plasma samples containing Magnevist and Dotarem in the presence of parenteral Fe(III) (Aime and Caravan, 2009;Rasschaert et al., 2018). The observed lack of / incomplete removal of GBCAs shows that the tertiary and conventional treated wastewater still contains highly stable Gd. ...
Article
The current toxicity concerns of gadolinium-based contrast agents (GBCAs) have birthed the need to regulate and restrict, in some instances, its clinical administration. However, its tolerable concentration levels in the water sector have not been set. Therefore, the detection and speedy increase of the anthropogenic Gd-GBCAs in the various water bodies, including those serving as the source of drinking water for adults and children, is perturbing. Nevertheless, the strongly canvassed risk-benefit considerations and superior uniqueness of GBCAs compared to the other ferromagnetic metals guarantees its continuous future administration for Magnetic resonance imaging (MRI) investigations regardless of the toxicity concerns. Furthermore, findings have shown that both the advanced and conventional treatment processes of wastewater do not satisfactorily remove GBCAs but rather risk transforming the chelated GBCAs to their free ionic metal (Gd ³⁺) through inadvertent degradation processes. This unintentional water processing-induced leads to the pathway for unintentional human intake of its free Gd transformative product and exposure to its probable ecotoxicity and several reported inimical effects on human health such as; digestive symptoms, twitching or weakness, cognitive flu, persistent skin diseases, body pains, acute renal and non-renal adverse reactions, chronic skin, and eyes changes. Hence, this work proposed an economical and manageable remediation technique for the potential remediation of Gd-GBCAs in wastewater, while a precautionary limit for Gd in public water and commercial drinks is advocated.
... Given that severe renal impairment resulting in reduced renal elimination and prolonged elevated blood GBCA levels is considered a primary factor leading to NSF in humans given certain linear GBCAs, several studies have looked at the impact of subtotal renal failure on Gd retention and/or T1 signal hyperintensity in deep cerebellar nuclei in animals [19][20][21]. These studies have shown that renal impairment potentiates Gd retention in deep cerebellar nuclei and that greater Gd retention and T1 signal hyperintensity are seen with linear GBCAs. ...
... To date, studies to evaluate Gd retention in rodent models of renal failure have looked at only linear GBCAs [20] or compared a solitary macrocyclic GBCA (gadoterate) with one [19] or two [21] linear GBCAs. Moreover, no studies have yet looked at Gd retention in renally compromised animals at multiple post-treatment timepoints. ...
Article
Full-text available
Background: Gd levels are higher in tissues of animals with compromised renal function, but studies to compare levels after exposure to different macrocyclic gadolinium-based contrast agents (GBCAs) are lacking. We compared Gd levels in tissues of subtotally nephrectomised (SN) rats after repeated exposure to macrocyclic GBCAs. Methods: Sprague-Dawley SN male rats (19 per group) received 16 injections of gadoteridol, gadobutrol, or gadoterate meglumine at 0.6 mmol Gd/kg 4 times/weeks over 4 weeks. A control group of healthy male rats (n = 10) received gadoteridol at the same dosage. Plasma urea and creatinine levels were monitored. Blood, cerebrum, cerebellum, liver, femur, kidney(s), skin and peripheral nerves were harvested for Gd determination by inductively coupled plasma-mass spectrometry at 28 and 56 days after the end of treatment. Results: Plasma urea and creatinine levels were roughly twofold higher in SN rats than in healthy rats at all timepoints. At day 28, Gd levels in the peripheral nerves of gadobutrol- or gadoterate-treated SN animals were 5.4 or 7.2 times higher than in gadoteridol-treated animals (p < 0.001). Higher Gd levels after administration of gadobutrol or gadoterate versus gadoteridol were also determined in kidneys (p ≤ 0.002), cerebrum (p ≤ 0.001), cerebellum (p ≤ 0.003), skin (p ≥ 0.244), liver (p ≥ 0.053), and femur (p ≥ 0.271). At day 56, lower Gd levels were determined both in SN and healthy rats for all GBCAs and tissues, except the femur. Conclusions: Gd tissue levels were lower following gadoteridol exposure than following gadobutrol or gadoterate exposure.
... That being said, MT is mainly used as a marker of myelination in white matter, while the predilection sites of gadolinium retention are mainly gray matter rich in metalloproteins. [53][54][55] This would likely reduce any potential confounding effects of MS pathology. ...
Article
Full-text available
Background and Purpose: Evidence of brain gadolinium retention has affected gadolinium-based contrast agent usage. It is, however, unclear towhat extentmacrocyclic agents are retained andwhether their in vivo detectionmay necessitate nonconventional MRI. Magnetization transfer (MT) could prove suitable to detect gadolinium-related signal changes since dechelated gadolinium ions bind to macromolecules. Therefore, this study aimed to investigate associations of prior gadolinium administrations with MT and T1 signal abnormalities. Methods: A cohort of 23 persons with multiple sclerosis (MS) (18 females, 5 males, 57 8.0 years) with multiple past gadolinium administrations (median 6, range 3-12) and 23 age- and sex-matched healthy controls underwent 1.5 Tesla MRI with MT, T1- weighted 2-dimensional spin echo, and T1-weighted 3-dimensional gradient echo. The signal intensity index was assessed byMRI in gadolinium retention predilection sites. Results: There were dose-dependent associations of the globus pallidus signal on gradient echo (r = .55, p < .001) and spin echo (r = .38, p = .013) T1-weighted imaging, but not on MT. Relative to controls, MS patients had higher signal intensity index in the dentate nucleus on T1-weighted gradient echo (1.037 0.040 vs. 1.016 0.023, p = .04) with a similar trend in the globus pallidus on T1-weighted spin echo (1.091 0.034 vs. 1.076 0.014, p = .06).MT detected no group differences. Conclusions: Conventional T1-weighted imaging provided dose-dependent associations with gadolinium administrations in MS, while these could not be detected with 2-dimensional MT. Future studies could explore newer MT techniques like 3D and inhomogenous MT. Notably, these associations were identified with conventional MRI even though most patients had not received gadolinium administrations in the preceding 9 years, suggestive of long-term retention.
... These results suggest that lower gadolinium concentrations than customary at 1.5 and 3 T may be sufficient for stenosis grading at UHF-CMR. In terms of patient safety, the application of reduced contrast agent concentrations without loss of image information is beneficial [33][34][35]. These findings are in concordance with previously published brain UHF-MRI studies [6,36]. ...
Article
Full-text available
Background To investigate the effects of B1-shimming and radiofrequency (RF) parallel transmission (pTX) on the visualization and quantification of the degree of stenosis in a coronary artery phantom using 7 Tesla (7 T) magnetic resonance imaging (MRI).Methods Stenosis phantoms with different grades of stenosis (0%, 20%, 40%, 60%, 80%, and 100%; 5 mm inner vessel diameter) were produced using 3D printing (clear resin). Phantoms were imaged with four different concentrations of diluted Gd-DOTA representing established arterial concentrations after intravenous injection in humans. Samples were centrally positioned in a thorax phantom of 30 cm diameter filled with a custom-made liquid featuring dielectric properties of muscle tissue. MRI was performed on a 7 T whole-body system. 2D-gradient-echo sequences were acquired with an 8-channel transmit 16-channel receive (8 Tx / 16 Rx) cardiac array prototype coil with and without pTX mode. Measurements were compared to those obtained with identical scan parameters using a commercially available 1 Tx / 16 Rx single transmit coil (sTX). To assess reproducibility, measurements (n = 15) were repeated at different horizontal angles with respect to the B0-field.ResultsB1-shimming and pTX markedly improved flip angle homogeneity across the thorax phantom yielding a distinctly increased signal-to-noise ratio (SNR) averaged over a whole slice relative to non-manipulated RF fields. Images without B1-shimming showed shading artifacts due to local B1+-field inhomogeneities, which hampered stenosis quantification in severe cases. In contrast, B1-shimming and pTX provided superior image homogeneity. Compared with a conventional sTX coil higher grade stenoses (60% and 80%) were graded significantly (p
... One potential reaction partner for gadolinium is the ironstoring protein ferritin, which has been found to interact with gadolinium (31) and result in the same SEC chromatographic retention as the gadolinium-binding macromolecules in brain tissue extracts (33). As also suggested by previous studies (34)(35)(36), it is likely that gadodiamide enters the brain rapidly after injection and partially dechelates due to its limited kinetic stability. The binding of gadolinium to macromolecules or transmetallation reactions can subsequently form insoluble permanent gadolinium depositions, impeding its elimination with time. ...
Article
Background Safety concerns caused by gadolinium retention call for the development of high-relaxivity gadolinium-based contrast agents (GBCAs) allowing minimal dosing. Purpose To investigate brain gadolinium retention in healthy rats after exposure to gadopiclenol (Elucirem, Guerbet; macrocyclic GBCA) compared with gadobutrol (Gadovist or Gadavist, Bayer; macrocyclic GBCA) and gadodiamide (Omniscan, GE Healthcare; linear GBCA) over 1 year. Materials and Methods In this study conducted between May 2018 and April 2020, 9-week-old healthy Sprague Dawley rats received five injections of either gadopiclenol, gadobutrol, or gadodiamide (2.4 mmol of gadolinium per kilogram of body weight for each), or saline (control animals) over a period of 5 weeks. Rats were randomly assigned to different groups (six female and six male rats per group). MRI examinations were performed before euthanasia at 1, 3, 5, or 12 months after the last injection. Brains were sampled to determine the total gadolinium content via inductively coupled plasma mass spectrometry (ICP-MS), to characterize gadolinium species with size exclusion chromatography (SEC)-ICP-MS, and to perform elemental mapping with laser ablation (LA)-ICP-MS. Mann-Whitney tests were performed on pairwise comparisons of the same time points. Results For both macrocyclic agents, no T1 signal hyperintensities were observed in the cerebellum, and approximately 80% of gadolinium washout was found between 1 month (gadobutrol, 0.30 nmol/g; gadopiclenol, 0.37 nmol/g) and 12 months (gadobutrol, 0.062 nmol/g; gadopiclenol, 0.078 nmol/g). After 12 months, only low-molecular-weight gadolinium species were detected in the soluble fraction. Gadodiamide led to significantly higher gadolinium concentrations after 1 month in the cerebellum (gadodiamide, 2.65 nmol/g; P < .001 vs both macrocyclics) combined with only 15% washout after 12 months (gadodiamide, 2.25 nmol/g) and with gadolinium detected bound to macromolecules. Elemental bioimaging enabled visualization of gadolinium deposition patterns colocalized with iron. Conclusion Gadopiclenol and gadobutrol demonstrated similar in vivo distribution and washout of gadolinium in the healthy rat brain, markedly differing from gadodiamide up to 12 months after the last injection. © RSNA, 2022 Online supplemental material is available for this article.
... Several studies reported on Zn depletion due to transmetallation with Gd and resulting zincuria after injections of linear GBCAs. 31,76,77 Thus, increased Zn content in EAE brains may lead to elevated transmetallation rates with administered Gd and therefore facilitate its retention. ...
Article
Full-text available
Objectives: Using a murine model of multiple sclerosis, we previously showed that repeated administration of gadopentetate dimeglumine led to retention of gadolinium (Gd) within cerebellar structures and that this process was enhanced with inflammation. This study aimed to compare the kinetics and retention profiles of Gd in inflamed and healthy brains after application of the macrocyclic Gd-based contrast agent (GBCA) gadobutrol or the linear GBCA gadopentetate. Moreover, potential Gd-induced neurotoxicity was investigated in living hippocampal slices ex vivo. Materials and methods: Mice at peak of experimental autoimmune encephalomyelitis (EAE; n = 29) and healthy control mice (HC; n = 24) were exposed to a cumulative dose of 20 mmol/kg bodyweight of either gadopentetate dimeglumine or gadobutrol (8 injections of 2.5 mmol/kg over 10 days). Magnetic resonance imaging (7 T) was performed at baseline as well as at day 1, 10, and 40 post final injection (pfi) of GBCAs. Mice were sacrificed after magnetic resonance imaging and brain and blood Gd content was assessed by laser ablation-inductively coupled plasma (ICP)-mass spectrometry (MS) and ICP-MS, respectively. In addition, using chronic organotypic hippocampal slice cultures, Gd-induced neurotoxicity was addressed in living brain tissue ex vivo, both under control or inflammatory (tumor necrosis factor α [TNF-α] at 50 ng/μL) conditions. Results: Neuroinflammation promoted a significant decrease in T1 relaxation times after multiple injections of both GBCAs as shown by quantitative T1 mapping of EAE brains compared with HC. This corresponded to higher Gd retention within the EAE brains at 1, 10, and 40 days pfi as determined by laser ablation-ICP-MS. In inflamed cerebellum, in particular in the deep cerebellar nuclei (CN), elevated Gd retention was observed until day 40 after last gadopentetate application (CN: EAE vs HC, 55.06 ± 0.16 μM vs 30.44 ± 4.43 μM). In contrast, gadobutrol application led to a rather diffuse Gd content in the inflamed brains, which strongly diminished until day 40 (CN: EAE vs HC, 0.38 ± 0.08 μM vs 0.17 ± 0.03 μM). The analysis of cytotoxic effects of both GBCAs using living brain tissue revealed an elevated cell death rate after incubation with gadopentetate but not gadobutrol at 50 mM. The cytotoxic effect due to gadopentetate increased in the presence of the inflammatory mediator TNF-α (with vs without TNF-α, 3.15% ± 1.18% vs 2.17% ± 1.14%; P = 0.0345). Conclusions: In the EAE model, neuroinflammation promoted increased Gd retention in the brain for both GBCAs. Whereas in the inflamed brains, efficient clearance of macrocyclic gadobutrol during the investigated time period was observed, the Gd retention after application of linear gadopentetate persisted over the entire observational period. Gadopentetate but not gadubutrol appeared to be neurotoxic in an ex vivo paradigm of neuronal inflammation.
... [9][10][11] This process named transmetallation is well studied using various scientific approaches including NMR relaxometry. [9,[12][13][14][15][16][17] However, it is now established that all GBCAs can lead to Gd 3 + ion deposition in the human body, but the interaction of dissociated Gd 3 + ions with endogenous substances in the human body is still under-investigated. [18,19] Understanding the related mechanisms is a persisting task at the interface between chemistry, biology, and contrast agent design to ensure optimum safety of future formulations. ...
Article
Full-text available
Glycosaminoglycans (GAGs) are highly negatively charged macromolecules with a large cation binding capacity, but their interaction potential with exogeneous Gd³⁺ ions is under‐investigated. These might be released from chelates used as Gadolinium‐based contrast agents (GBCAs) for clinical MR imaging due to transmetallation with endogenous cations like Zn²⁺. Recent studies have quantified how an endogenous GAG sequesters released Gd³⁺ ions and impacts the thermodynamic and kinetic stability of some GBCAs. In this study, we investigate and compare the chelation ability of two important GAGs (heparin and chondroitin sulfate), as well as the homopolysaccharides dextran and dextran sulfate that are used as models for alternative macromolecular chelators. Our combined approach of MRI‐based relaxometry and isothermal titration calorimetry shows that the chelation process of Gd³⁺ into GAGs is not just a long‐range electrostatic interaction as proposed for the Manning model, but presumably a site‐specific binding. Furthermore, our results highlight the crucial role of sulfate groups in this process and indicate that the potential of a specific GAG to engage in this mechanism increases with its degree of sulfation. The transchelation of Gd³⁺ ions from GBCAs to sulfated GAGs should thus be considered as one possible explanation for the observed long‐term deposition of Gd³⁺ in vivo and related observations of long‐term signal enhancements on T1‐weighted MR images.
Article
Full-text available
Objectives After the administration of gadolinium-based contrast agents (GBCAs), residual gadolinium (Gd) has been detected in a few distinct morphological structures of the central nervous system (CNS). However, a systematic, comprehensive, and quantitative analysis of the spatial Gd distribution in the entire brain is not yet available. The first aim of this study is to provide this analysis in healthy rats after administration of high GBCA doses. The second aim is to assess the spatial distributions and possible Gd colocalizations of endogenous iron (Fe), manganese (Mn), and phosphorus (P). In addition, the presence of Gd in proximity to blood vessels was assessed by immunohistochemistry. Materials and Methods Male rats were randomly assigned to 3 groups (n = 3/group): saline (control), gadodiamide (linear GBCA), and gadobutrol (macrocyclic GBCA) with cumulative Gd doses of 14.4 mmol/kg of body mass. Five weeks after the last administration, the brains were collected and cryosectioned. The spatial distributions of Gd, Fe, Mn, and P were analyzed in a total of 130 sections, each covering the brain in 1 of the 3 perpendicular anatomical orientations, using laser ablation coupled with inductively coupled plasma mass spectrometry. Quantitative spatial element maps were generated, and the concentrations of Gd, Fe, and Mn were measured in 31 regions of interest covering various distinct CNS structures. Correlation analyses were performed to test for possible colocalization of Gd, Fe, and Mn. The spatial proximity of Gd and blood vessels was studied using metal-tagged antibodies against von Willebrand factor with laser ablation coupled with inductively coupled plasma mass spectrometry. Results After administration of linear gadodiamide, high Gd concentrations were measured in many distinct structures of the gray matter. This involved structures previously reported to retain Gd after linear GBCA, such as the deep cerebellar nuclei or the globus pallidus, but also structures that had not been reported so far including the dorsal subiculum, the retrosplenial cortex, the superior olivary complex, and the inferior colliculus. The analysis in all 3 orientations allowed the localization of Gd in specific subregions and layers of certain structures, such as the hippocampus and the primary somatosensory cortex. After macrocyclic gadobutrol, the Gd tissue concentration was significantly lower than after gadodiamide. Correlation analyses of region of interest concentrations of Gd, Fe, and Mn revealed no significant colocalization of Gd with endogenous Fe or Mn in rats exposed to either GBCA. Immunohistochemistry revealed a colocalization of Gd traces with vascular endothelium in the deep cerebellar nuclei after gadobutrol, whereas the majority of Gd was found outside the vasculature after gadodiamide. Conclusions In rats exposed to gadodiamide but not in rats exposed to gadobutrol, high Gd concentrations were measured in various distinct CNS structures, and structures not previously reported were identified to contain Gd, including specific subregions and layers with different cytoarchitecture and function. Knowledge of these distinct spatial patterns may pave the way for tailored functional neurological testing. Signs for the localization of the remaining Gd in the vascular endothelium were prominent for gadobutrol but not gadodiamide. The results also indicate that local transmetalation with endogenous Fe or Mn is unlikely to explain the spatial patterns of Gd deposition in the brain, which argues against a general role of these metals in local transmetalation and release of Gd ions in the CNS.
Article
Full-text available
Focused ultrasound (FUS) can be used to open the blood-brain barrier (BBB), and MRI with contrast agents can detect that opening. However, repeated use of gadolinium-based contrast agents (GBCAs) presents safety concerns to patients. This study is the first to propose the idea of modeling a volume transfer constant (Ktrans) through deep learning to reduce the dosage of contrast agents. The goal of the study is not only to reconstruct artificial intelligence (AI) derived Ktrans images but to also enhance the intensity with low dosage contrast agent T1 weighted MRI scans. We successfully validated this idea through a previous state-of-the-art temporal network algorithm, which focused on extracting time domain features at the voxel level. Then we used a Spatiotemporal Network (ST-Net), composed of a spatiotemporal convolutional neural network (CNN)-based deep learning architecture with the addition of a three-dimensional CNN encoder, to improve the model performance. We tested the ST-Net model on ten datasets of FUS-induced BBB-openings aquired from different sides of the mouse brain. ST-Net successfully detected and enhanced BBB-opening signals without sacrificing spatial domain information. ST-Net was shown to be a promising method of reducing the need of contrast agents for modeling BBB-opening K-trans maps from time-series Dynamic Contrast-Enhanced Magnetic Resonance Imaging (DCE-MRI) scans.
Article
Full-text available
Objectives: The aim of this study was to examine the effects of perinatal exposure to gadolinium (Gd)-based contrast agents (GBCAs) on the behavior of adulthood offspring. Materials and methods: Pregnant Balb/C mice (n = 5 per group) were intravenously injected with gadoterate meglumine (Magnescope, macrocyclic GBCA), gadodiamide (Omniscan, linear GBCA), or vehicle from pregnancy day 15 to 19, corresponding to embryonic day 15 to 19 of the fetus, at 2 mmol/kg body weight per day. Brain samples from dams and pups were collected on postpartum day 28. The total Gd concentration was quantified by inductively coupled plasma-mass spectrometry (dams, n = 3; gadoterate meglumine-treated pups group, n = 9; and gadodiamide-treated pups group, n = 10). Behavioral testing of offspring was started on postpartum day 70 (control group, n = 22; gadoterate meglumine-treated group, n = 23; and gadodiamide-treated group, n = 20). Results: Higher levels of Gd retention were observed in dams and pups in the gadodiamide-treated group. Perinatal exposure to GBCAs caused anxiety-like behavior, disrupted motor coordination, impaired memory function, stimulated tactile sensitivity, and decreased muscle strength, particularly in the gadodiamide-treated group. Conclusions: In the present study, we showed that Gd was transferred to pups and was retained in their brain during postnatal development. Gadolinium retention may lead to impaired brain development. These findings indicate that the use of GBCAs in pregnant women should be avoided because it may have adverse effects on the fetus, particularly on brain development.
Article
Full-text available
Objectives: The primary objective of this study was to investigate the pharmacokinetic profile of gadoterate meglumine in pediatric patients younger than 2 years; the secondary objectives were to document its efficacy and safety. Material and methods: This was a Phase IV open-label, prospective study conducted in 9 centers (4 countries). Forty-five patients younger than 2 years with normal estimated glomerular filtration rate and scheduled to undergo routine gadolinium-enhanced magnetic resonance imaging (MRI) of any organ were included and received a single intravenous injection of gadoterate meglumine (0.1 mmol/kg). To perform the population pharmacokinetics analysis, 3 blood samples per subject were drawn during 3 time windows at time points allocated by randomization. Results: Gadoterate meglumine concentrations were best fitted using a 2-compartmental model with linear elimination from central compartment. The median total clearance adjusted to body weight was estimated at 0.06 L/h per kg and increased with estimated glomerular filtration rate according to a power model. The median volume of distribution at steady state (Vss) adjusted to body weight was estimated at 0.047 L/kg. Estimated median terminal half-life (t1/2β) was 1.35 h, and the median systemic exposure (area under the curve) was 1591 μmol h/L. Efficacy was assessed by comparing precontrast +postcontrast images to precontrast images in a subset of 28 subjects who underwent an MRI examination of brain, spine, and associated tissues. A total of 28 lesions were identified and analyzed in 15 subjects with precontrast images versus 30 lesions in 16 subjects with precontrast + postcontrast images. Lesion visualization was improved with a mean (SD) increase in scores at subject level of 0.7 (1.0) for lesion border delineation, 0.9 (1.6) for internal morphology, and 3.1 (3.2) for contrast enhancement. Twenty-six adverse events occurred postinjection in 13 subjects (28.9%), including 3 serious reported in 1 subject (2.2%). One subject (2.2%) experienced 1 rash of moderate intensity considered as related to gadoterate meglumine. Conclusions: The pharmacokinetic profile of gadoterate meglumine after a single intravenous injection of 0.1 mmol/kg was appropriately described in newborns and infants younger than 2 years, for whom no dose adjustment is required. The improved efficacy of gadoterate meglumine for contrast-enhanced MRI examination of brain, spine, and associated tissues, as well as its good safety profile, was also demonstrated in this population.This is an open-access article distributed under the terms of the Creative Commons Attribution-Non Commercial-No Derivatives License 4.0 (CCBY-NC-ND), where it is permissible to download and share the work provided it is properly cited. The work cannot be changed in any way or used commercially without permission from the journal.
Article
Full-text available
Purpose: To investigate the effect of pregnancy and type of gadolinium (Gd)-based contrast agents (GBCAs) on organ retention of Gd in mother and pup mice after maternal administration. Materials and methods: Gd-DTPA-BMA (gadodiamide) or Gd-DOTA (gadoterate dimeglumine) was administered (2.0 mmol/kg of maternal weight) four times to pregnant Balb/c mice from gestational day 16-19, respectively. At 28 days after birth, they were euthanized and their organs (blood, brain, liver, kidney, spleen, and bone) were removed for the measurement of Gd by inductively coupled plasma mass spectrometry. Results: Gd retention in maternal organs was generally lower than that in the organs of non-pregnant mice in both Gd-DTPA-BMA and Gd-DOTA groups. Significantly higher Gd retention was observed in the organs of pups whose mothers were administered Gd-DTPA-BMA as compared to those whose mothers were administered Gd-DOTA. Tissue-to-muscle ratio in the brains of pups was higher than that of mothers. Conclusion: We demonstrated in utero transplacental Gd retention in pups. In various organs in both mothers and pups, Gd retention was consistently higher for Gd-DTPA-BMA than Gd-DOTA administration. Pregnancy affected Gd retention in many maternal organs.
Article
Full-text available
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.This is an open-access article distributed under the terms of the Creative Commons Attribution-Non Commercial-No Derivatives License 4.0 (CCBY-NC-ND), where it is permissible to download and share the work provided it is properly cited. The work cannot be changed in any way or used commercially without permission from the journal.
Article
Full-text available
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.This is an open-access article distributed under the terms of the Creative Commons Attribution-Non Commercial-No Derivatives License 4.0 (CCBY-NC-ND), where it is permissible to download and share the work provided it is properly cited. The work cannot be changed in any way or used commercially without permission from the journal.
Article
Objective: The aim of this study was to evaluate and compare changes in T1-weighted signal intensity (SI) within the dentate nucleus (DN) and globus pallidus (GP) in a pediatric population after serial applications of the linear gadolinium-based magnetic resonance contrast medium gadopentetate dimeglumine and the more stable macrocyclic agent gadobutrol. Materials and methods: Institutional review board approval was obtained. Two similar pediatric patient cohorts who underwent at least 3 serial contrast-enhanced magnetic resonance imaging (MRI) examinations with sole application of gadopentetate dimeglumine or gadobutrol were analyzed. All MRI examinations were performed on a 1.5 T system acquiring unenhanced T1-weighted spin echo sequences, which were evaluated on the baseline MRI and after the contrast medium administrations. For analysis of SI changes in the DN, the ratios of the DN to the pons (P) and to the middle cerebellar peduncle (MCP) were assessed. The GP was compared with the thalamus (TH) by dividing the SIs between GP and TH (GP-to-TH ratio). Results: Twenty-eight patients (13 boys, 15 girls; mean age, 8.4 ± 6.8 years) who received at least 3 applications of gadopentetate dimeglumine and 25 patients (13 boys, 12 girls; mean age, 9.7 ± 5.4 years) with 3 or more gadobutrol injections were included. After 3 administrations of gadopentetate dimeglumine, the T1-weighted SI ratios significantly increased: mean difference value of 0.036 ± 0.031 (DN-to-P; P < 0.001), 0.034 ± 0.032 (DN-to-MCP; P < 0.001), and 0.025 ± 0.025 (GP-to-TH; P = 0.001). In a subanalysis of 12 patients with more than 3 injections of gadopentetate dimeglumine, the mean differences of the SI ratios were slightly higher: 0.043 ± 0.032 (DN-to-P; P = 0.001), 0.041 ± 0.035 (DN-to-MCP; P = 0.002), and 0.028 ± 0.025 (GP-to-TH; P = 0.003). In contrast, gadobutrol did not show a significant influence on the SI ratios, neither after 3 nor after more than 3 applications. Conclusions: The T1-weighted SI increase within the DN and GP after serial administrations of the linear contrast medium gadopentetate dimeglumine, but not after serial applications of the macrocyclic agent gadobutrol, found in a pediatric population, is consistent with results published for adult patients. The clinical impact of the intracranial T1-hyperintensities is currently unclear. However, in accordance with the recent decision of the Pharmacovigilance and Risk Assessment Committee of the European Medicines Agency, intravenous macrocyclic agents should be preferred and MR contrast media should be used with caution and awareness of the pediatric brain development in children and adolescents.
Article
Purpose To determine whether gadolinium remains in juvenile nonhuman primate tissue after maternal exposure to intravenous gadoteridol during pregnancy. Materials and Methods Gravid rhesus macaques and their offspring (n = 10) were maintained, as approved by the institutional animal care and utilization committee. They were prospectively studied as part of a pre-existing ongoing research protocol to evaluate the effects of maternal malnutrition on placental and fetal development. On gestational days 85 and 135, they underwent placental magnetic resonance imaging after intravenous gadoteridol administration. Amniocentesis was performed on day 135 prior to administration of the second dose of gadoteridol. After delivery, the offspring were followed for 7 months. Tissue samples from eight different organs and from blood were harvested from each juvenile macaque. Gadolinium levels were measured by using inductively coupled plasma mass spectrometry. Results Gadolinium concentration in the amniotic fluid was 0.028 × 10(-5) %ID/g (percentage injected dose per gram of tissue) 50 days after administration of one gadoteridol dose. Gadolinium was most consistently detected in the femur (mean, 2.5 × 10(-5) %ID/g; range, [0.81-4.1] × 10(-5) %ID/g) and liver (mean, 0.15 × 10(-5) %ID/g; range, [0-0.26] × 10(-5) %ID/g). Levels were undetectable in the remaining sampled tissues, with the exception of one juvenile skin sample (0.07 × 10(-5) %ID/g), one juvenile spleen sample (0.039 × 10(-5) %ID/g), and one juvenile brain (0.095 × 10(-5) %ID/g) and kidney (0.13 × 10(-5) %ID/g) sample. Conclusion The presence of gadoteridol in the amniotic fluid after maternal injection enables confirmation that it crosses the placenta. Extremely low levels of gadolinium are found in juvenile macaque tissues after in utero exposure to two doses of gadoteridol, indicating that a very small amount of gadolinium persists after delivery. (©) RSNA, 2017.
Article
Purpose To evaluate the speciation of gadolinium-containing species after multiple administrations of the gadolinium-based contrast agents (GBCAs) gadodiamide and gadoteridol and to quantify the amount of intact gadolinium complexes and insoluble gadolinium-containing species. Materials and Methods A total dose of 13.2 mmol per kilogram of body weight of each GBCA was administered in healthy Wistar rats over a period of 8 weeks. Three days after the final administration, rats were sacrificed, and the brains were excised and divided into three portions. Each portion of brain homogenate was divided into two parts, one for determination of the total gadolinium concentration with inductively coupled plasma mass spectrometry and one for determination of the amount of intact GBCA and gadolinium-containing insoluble species. Relaxometric measurements of gadodiamide and gadolinium trichloride in the presence of polysialic acid were also performed. Results The mean total gadolinium concentrations for gadodiamide and gadoteridol, respectively, were 0.317 μg/g ± 0.060 (standard deviation) and 0.048 μg/g ± 0.004 in the cortex, 0.418 μg/g ± 0.078 and 0.051 μg/g ± 0.009 in the subcortical brain, and 0.781 μg/g ± 0.079 and 0.061 μg/g ± 0.012 in the cerebellum. Gadoteridol comprised 100% of the gadolinium species found in rats treated with gadoteridol. In rats treated with gadodiamide, the largest part of gadolinium retained in brain tissue was insoluble species. In the cerebellum, the amount of intact gadodiamide accounts for 18.2% ± 10.6 of the total gadolinium found therein. The mass balance found for gadolinium implies the occurrence of other soluble gadolinium-containing species (approximately 30%). The relaxivity of the gadolinium polysialic acid species formed in vitro was 97.8 mM/sec at 1.5 T and 298 K. Conclusion Gadoteridol was far less retained, and the entire detected gadolinium was intact soluble GBCA, while gadodiamide yielded both soluble and insoluble gadolinium-containing species, with insoluble species dominating. (©) RSNA, 2017 Online supplemental material is available for this article.
Article
Background: Gadolinium-based-contrast-agents (GBCAs) are used for magnetic-resonance-imaging and associated with renal and cardiovascular adverse reactions caused by released Gd(3+) ions. Gd(3+) is also a modulator of mechano-gated ion channels, including the epithelial Na(+) channel (ENaC) that is expressed in kidney epithelium and the vasculature. ENaC is important for salt-/water homeostasis and blood pressure regulation and a likely target of released Gd(3+) from GBCAs causing the above-mentioned adverse reactions. Therefore this study examined the effect of Gd(3+) and GBCAs on ENaC's activity. Methods: Human αβγENaC was expressed in Xenopus laevis oocytes and exposed to Gd(3+), linear (Gd-DTPA, Magnevist) or cyclic (Dotarem) GBCAs. Transmembrane ion-currents (IM) were recorded by the two-electrode-voltage-clamp technique and Gd(3+)-release by Gd-DTPA was confirmed by inductively coupled plasma-mass spectrometry. Results: Gd(3+) exerts biphasic effects on ENaC's activity: ≤0.3mmol/l decreased IM which was preventable by DEPC (modifies histidines). Strikingly Gd(3+)≥0.4mmol/l increased IM and this effect was prevented by cysteine-modifying MTSEA. Linear Gd-DTPA and Magnevist mimicked the effect of ≤0.3mmol/l Gd(3+), whereas the chelator DTPA showed no effect. Gd(3+) and Gd-DTPA increased the IC50 for amiloride, but did not affect ENaC's self-inhibition. Interestingly, cyclic Gd-DOTA (Dotarem) increased IM to a similar extent as its chelator DOTA, suggesting that the chelator rather than released Gd(3+) is responsible for this effect. Conclusion: These results confirm Gd(3+)-release from linear Gd-DTPA and indicate that the released Gd(3+) amount is sufficient to interfere with ENaC's activity to provide putative explanations for GBCA-related adverse effects.