Skip to main content
Erschienen in: European Journal of Nuclear Medicine and Molecular Imaging 4/2011

Open Access 01.04.2011 | Original Article

Renal uptake of different radiolabelled peptides is mediated by megalin: SPECT and biodistribution studies in megalin-deficient mice

verfasst von: Erik Vegt, Marleen Melis, Annemarie Eek, Monique de Visser, Maarten Brom, Wim J. G. Oyen, Martin Gotthardt, Marion de Jong, Otto C. Boerman

Erschienen in: European Journal of Nuclear Medicine and Molecular Imaging | Ausgabe 4/2011

Abstract

Purpose

Radiolabelled peptides used for peptide receptor radionuclide therapy are excreted mainly via the kidneys and are partly reabsorbed and retained in the proximal tubular cells. The resulting high renal radiation dose can cause nephrotoxicity, limiting the maximum activity dose and the effectiveness of peptide receptor radionuclide therapy. The mechanisms of kidney reabsorption of these peptides are incompletely understood, but the scavenger receptor megalin has been shown to play a role in the reabsorption of 111In-octreotide. In this study, the role of megalin in the renal reabsorption of various relevant radiolabelled peptides was investigated.

Methods

Groups of kidney-specific megalin-deficient mice and wild-type mice were injected with 111In-labelled somatostatin, exendin, neurotensin or minigastrin analogues. Single photon emission computed tomographic (SPECT) images of the kidneys were acquired and analysed quantitatively, or the animals were killed 3 h after injection and the activity concentration in the kidneys was measured.

Results

Megalin-deficient mice showed significantly lower uptake of all studied radiolabelled peptides in the kidneys, ranging from 22% (111In-octreotide) to 65% (111In-exendin) of uptake in wild-type kidneys. Quantitative analysis of renal uptake by SPECT and ex vivo measurements showed a very good correlation.

Conclusion

Megalin is involved in the renal reabsorption of radiolabelled octreotide, octreotate, exendin, neurotensin and minigastrin. This knowledge may help in the design of strategies to reduce this reabsorption and the resulting nephrotoxicity in peptide receptor radionuclide therapy, enabling more effective therapy. Small-animal SPECT is an accurate tool, allowing in vivo quantification of renal uptake and serial measurements in individual mice.
Hinweise
Erik Vegt and Marleen Melis contributed equally to this work.

Introduction

In peptide receptor radionuclide therapy (PRRT), radiolabelled peptide analogues are used to target tumours expressing particular receptors, such as the somatostatin receptor. Most radiolabelled peptides are predominantly cleared from the body via the kidneys. Rapid clearance of the radiolabelled peptides from the blood and low retention in the kidneys minimize the radiation dose to normal tissues. However, part of the filtered load of these small proteins and peptides is reabsorbed from the ultrafiltrate in the proximal tubules. Evidence suggests that, after glomerular filtration, proteins and peptides in the ultrafiltrate bind to endocytic receptors at the luminal surface of proximal tubular cells and are internalized. Subsequently, the compounds are transferred to the lysosomes, where they are proteolytically degraded into amino acids [1]. These are transported back into the bloodstream. However, residualizing radiolabels (e.g. N-terminal amino acid chelate conjugates or lysine chelate conjugates) are trapped in the tubular cell lysosomes and can deliver high radiation doses to the kidney tubules and glomeruli [2]. Nephrotoxicity is dose-limiting in PRRT with somatostatin analogues such as 90Y-DOTA-Tyr3-octreotide [3, 4].
The receptors involved in the tubular reabsorption of peptides have not yet been completely characterized, but for various nonradiolabelled peptides the involvement of megalin has been shown [5]. Megalin is a multiligand receptor belonging to the LDL receptor family. The receptor contains four large cysteine-rich ligand-binding domains and is a high-capacity pathway for the reabsorption of different structurally nonrelated peptides and proteins such as albumin, vitamin D binding protein, β2-microglobulin and aprotinin [5, 6]. De Jong et al. recently reported that the renal uptake of 111In-octreotide is significantly lower in kidney-specific megalin-deficient mice than in their wild-type counterparts, implicating the involvement of megalin in the renal reabsorption of radiolabelled somatostatin analogues [7]. Many other radiolabelled peptides that are being studied for their potential in tumour imaging and PRRT display renal retention severalfold higher than octreotide. Examples include exendin and minigastrin, targeting glucagon-like peptide-1 and cholecystokinin2 receptors, respectively [8, 9]. It is unknown whether megalin is also involved in the renal uptake of these peptides.
In clinical PRRT with somatostatin analogues, the standard renoprotective regimen nowadays consists of coinfusion of basic amino acids, which are thought to interfere with the binding of somatostatin analogues to megalin or other endocytic receptors on the proximal tubular cells [10, 11]. However, Gotthardt et al. have shown that the renal uptake of 111In-minigastrin is not reduced significantly by coinfusion of basic amino acids [9], suggesting that the reabsorption of this peptide is mediated by other receptors. Knowledge about the molecular mechanisms of proximal tubular reabsorption of different radiolabelled peptides is important to devise new methods to reduce their renal retention, for example by selecting or designing more efficient inhibitors of their renal reabsorption, or by structurally modifying the peptides to reduce their binding to renal receptors.
In this study, the role of megalin in the renal retention of [111In-DTPA-d-Phe1]-octreotide (111In-octreotide), [111In-DTPA0-D-Phe1,Tyr3]-octreotate (111In-octreotate), [Lys40(111In-DTPA)]-exendin-3 (111In-exendin), [111In-DOTA-Glu1]-minigastrin (111In-minigastrin) and [111In-DTPA0]-neurotensin (111In-neurotensin) [613] was studied in mice. The renal uptake of the peptides was measured by single photon emission computed tomography (SPECT) imaging and by ex vivo measurements of kidney-specific megalin-deficient mice (megalinlox/lox; apoECre [12]) and wild-type mice. Since the level of renal uptake of radiopeptides may differ between female and male mice [13], both genders were imaged by SPECT.

Materials and methods

Animals

The megalin-deficient mice used were megalinlox/lox; apoECre mice [12]. The principles of creating tissue-specific gene knockout models are described in detail elsewhere [14, 15]. In brief, in megalinlox/lox; apoECre mice, expression of the enzyme Cre recombinase is controlled by the apolipoprotein E (apoE) promoter, and is thus produced only in tissues where the apoE gene is transcribed. In these tissues, Cre recombinase excises sequences from the genome that are flanked by two loxP sequences, in this case the megalin gene [14]. Animals were bred locally using males heterozygous for the apoECre gene. Offspring expressing the apoECre gene were identified by means of polymerase chain reaction (PCR) analysis as described below. Animals not expressing the apoECre gene (megalinlox/lox mice) were used as the wild type in the biodistribution studies. In the SPECT studies, C57Bl/6 mice were used as the wild type.

PCR analysis

For PCR the primers CCCAAGAAGAGGAAGGTG (forward) and GCTGGCCCAAATGTTGCTG (reverse) were used. The reaction mixture consisted of approximately 50 ng mouse DNA in a total of 25 μl colourless PCR buffer (Go Taq Flexi reaction buffer, Promega) with 5 mM MgCl2 (Promega), 0.5 mM deoxyribonucleotide triphosphate mix (dNTP, Promega), 12.5 pmol forward primer, 12.5 pmol reverse primer and 2.5 IU Taq polymerase (Promega). This mixture was heated for 4 min at 95°C, followed by 32 cycles of 30 s at 95°C, 30 s at 56°C and 30 s at 72°C. After cycling, the temperature was maintained at 72°C for 10 min and subsequently lowered to 10°C. The DNA formed was analysed by agarose gel electrophoresis.

Radiolabelled compounds

The following peptide derivatives were studied: [DTPA-D-Phe1]-octreotide (Covidien) [16], [DTPA0-D-Phe1,Tyr3]-octreotate (Biosynthema) [17], [DTPA0]-neurotensin [613] (Biosynthema) [18], [Lys40-DTPA]-exendin-3 (exendin, Peptide Specialty Laboratories) [19], and [DOTA-Glu1]-minigastrin (Peptide Specialty Laboratories) [20]. Relevant properties of the peptides are summarized in Table 1.
Table 1
Characteristics of studied peptide analogues
Peptide analogue
Target receptor
Molecular weight (kDa)
Number of amino acid residues (positive/negative)
Charge at pH 7
111In-Octreotide
Somatostatin receptor2
1.5
8 (1+/0−)
+1
111In-Octreotate
Somatostatin receptor2
1.5
8 (1+/0−)
+1
111In-Exendin
Glucagon-like peptide-1
4.8
39 (4+/6−)
−2
111In-Minigastrin
Cholecystokinin2 and gastrin
2.1
13 (0+/7−)
−7
111In-Neurotensin
Neurotensin
1.3
8 (1+/0−)
+1
Peptides were labelled with 111InCl3 (20 MBq/nmol for SPECT studies, 10–80 MBq/nmol for ex vivo biodistribution studies) as described previously [21]. Labelling efficiency and radiochemical purity of the labelled peptides were determined by silica gel instant thin-layer chromatography and reverse-phase high-performance liquid chromatography. Radiochemical purity was >95% for all compounds.

SPECT studies

Megalin-deficient and wild-type mice (male and female, four to six per group) received an intravenous injection of 40 MBq (0.2 ml, 2 nmol) 111In-octreotide. The exact injected activity was determined by measuring the syringe in a dose calibrator before and after injection. A 24-min SPECT scan of the kidney region was acquired 3 and 24 h after injection with a four-headed helical NanoSPECT/CT system (Bioscan) using Nucline software (v 2.01, Mediso). Multipinhole mouse collimators with nine pinholes (1.4 mm diameter) per head were used, with a matrix of 256 × 256 and 24 projections (2 min per projection). During the scan the animals were anaesthetized with isoflurane/O2 and body temperature was maintained. This procedure was repeated in the same mice with 111In-octreotate, 111In-exendin, 111In-neurotensin and 111In-minigastrin consecutively at intervals of at least 3 weeks. Some of the animals developed signs of kidney damage at the end of this series of experiments, probably caused by the relatively high renal uptake of 111In-exendin, resulting in a high radiation dose to the kidneys [22]. Therefore, the 111In-exendin, 111In-neurotensin and 111In-minigastrin data were discarded and an extra experiment with these peptides was carried out in a new set of animals. To reduce the risk of kidney damage, the injected activity of 111In-minigastrin and 111In-exendin was reduced to 10 MBq, while the peptide dose was kept constant at 2 nmol. In this second series, SPECT scans were acquired only 3 h after injection.
SPECT scans were reconstructed iteratively using InVivoScope software (v 1.32, Bioscan) with medium noise reduction, a voxel size of 0.3 mm3 and standard reconstruction settings. The amount of radioactivity in a volume of interest drawn around the kidneys was quantified and expressed as percent of injected dose per gram tissue (%ID/g). To achieve accurate quantification, the camera was calibrated by scanning a 20-ml polypropylene tube mouse phantom filled with a known amount of 111In activity.
After the final SPECT scan, the animals were killed and the biodistribution of 111In-minigastrin and 111In-exendin was determined as described in the next section to confirm the accuracy of the SPECT measurements.

Ex vivo biodistribution studies

Animals (four to six per group) received an intravenous injection of 0.4 MBq (0.2 ml, 5–40 pmol) 111In-octreotide, 111In-exendin, 111In-neurotensin or 111In-minigastrin. For 111In-octreotide the experiment was performed with male and female mice; for the other peptides only female mice were used. The animals were killed 3 h after injection, and organs were dissected. The biodistribution of the 111In-labelled peptides was determined by weighing the organs and measuring the radioactivity in a gamma counter. Measured activity was expressed as %ID/g. The right kidney of each animal was cut in half. One half was snap-frozen in liquid nitrogen and processed for cryosectioning, the other half was processed for paraffin sectioning. Frozen 10-μm sections were mounted on glass slides for autoradiography. A phosphor imaging screen was exposed to the sections for 2 days and scanned using a BAS 1800-II phosphor imager (Fujifilm).

Immunohistochemistry

Frozen 5-μm kidney sections were fixed in 4% formalin for 10 min. After rinsing with 0.05% polysorbate 80 in PBS, the sections were incubated with goat anti-rat megalin polyclonal antibody (SC-16478, Santa Cruz) 10 μg/ml in PBS with 5% BSA for 1 h at room temperature, followed by incubation with horseradish peroxidase-conjugated donkey anti-goat IgG, F(ab')2 (SC-3851, Santa Cruz) 1/100 for 30 min at room temperature. Peroxidase activity was visualized with diaminobenzidine (Powervision) and nuclei were counterstained using haematoxylin. Slides were dehydrated with ethanol and xylene and embedded in slide mounting fluid (Permount), after which they were studied microscopically. Megalin expression was scored visually by an independent, blinded observer on an arbitrary scale of 0 (negative) to 4 (all tubules positive).

Statistical analysis

Data are presented as mean values ± standard deviation. Renal uptake values were compared using Student’s t-test. For the SPECT studies of 111In-minigastrin and 111In-exendin (after which the mice were killed), the correlation between renal uptake measured on the SPECT images and uptake in the same mice measured ex vivo was determined. Spearman’s rank correlation coefficient was calculated with SPSS 16.0 (SPSS).

Results

Immunohistochemistry

Immunostaining revealed lower expression of megalin in the kidney cortex of the megalin-deficient mice as compared to the wild-type mice. The expression of megalin in the megalin-deficient kidneys varied considerably, ranging from almost absent (score 0) to moderate expression (score 2). Examples are shown in Fig. 1.

SPECT measurement of renal peptide uptake

As shown in Fig. 2, the kidneys were visualized very well on the SPECT scans acquired at 3 h after injection. The images indicated that the radioactivity mainly accumulated in the renal cortex.
The measured renal uptake values of the 111In-labelled peptides as derived from the SPECT images at 3 h after injection and the ratios of uptake between megalin-deficient mice and wild-type mice are presented in Table 2. The data are summarized together with the ex vivo biodistribution data in Fig. 3. 111In-exendin expressed the highest renal uptake: 371±35 %ID/g in female wild-type mice. The peptide with the lowest renal uptake (15±2.7 %ID/g in female wild-type mice) was 111In-neurotensin. In the SPECT studies, the renal retention of all 111In-labelled peptides was significantly lower in megalin-deficient mice than in wild-type mice, both in males and females. The effect was most prominent for 111In-neurotensin, for which the renal uptake in female megalin-deficient mice was only 23% of the uptake in wild-type mice (p < 0.0001). The least effect was observed with 111In-exendin, for which the uptake in female megalin-deficient mice was 62% of the uptake in wild-type mice (p = 0.003).
Table 2
Renal uptake of radiolabelled peptides on SPECT in wild-type and megalin-deficient mice
Peptide
Gender
Wild-type (%ID/g ± SD)
Megalin-deficient (%ID/g ± SD)
Ratio megalin-deficient/wild-type
3 h after injection
111In-Octreotide
Female
17±2.4
8.5±2.6
49% (p = 0.001)
Male
22±5.6
5.7±2.0
26% (p = 0.0003)
111In-Octreotate
Female
16±4.6
7.6±2.4
46% (p = 0.007)
Male
16±2.8
5.9±1.9
36% (p = 0.0001)
111In-Exendin
Female
371±35
230±58
62% (p = 0.003)
Male
328±51
171±34
52% (p = 0.001)
111In-Minigastrin
Female
89±10
33±8.9
37% (p < 0.0001)
Male
104±13
52±14
49% (p = 0.0003)
111In-Neurotensin
Female
15±2.7
3.6±1.18
23% (p < 0.0001)
Male
18±9.0
4.6±1.1
25% (p = 0.02)
24 h after injection
111In-Octreotide
Female
2.2±0.25
1.4±0.47
64% (p = 0.0001)
Male
Not measurable
Not measurable
111In-Octreotate
Female
5.3±0.97
2.5±0.71
47% (p = 0.001)
Male
1.6±0.37
0.44±0.20
28% (p = 0.0003)
111In-Exendin
Female
148±11
82±24
55% (p = 0.0005)
Male
142±10
69±19
49% (p = 0.0001)
The data measured 24 h after injection are presented in Table 2. The renal uptake of the studied radiolabelled peptides remained significantly lower in the megalin-deficient mice, both in females and in males. Overall, the uptake of 111In-octreotide and 111In-octreotate was significantly lower in male mice than in female mice: the retention of 111In-octreotide in males was too low to delineate the kidneys and the uptake of 111In-octreotate was more than threefold lower in males than in females. For 111In-exendin, no difference between the genders was observed.

Ex vivo measurement of renal uptake

The renal uptake of the 111In-labelled peptides measured ex vivo in megalin-deficient and wild-type mice is presented in Table 3 and summarized in Fig. 3. The renal uptake of 111In-octreotide, 111In-octreotate, 111In-minigastrin and 111In-neurotensin was significantly lower in the megalin-deficient mice than in the wild-type mice. The effect was most prominent for 111In-octreotide, for which the renal uptake in female megalin-deficient mice was only 22% of the uptake in wild-type mice (p = 0.0007). For 111In-exendin, no significant difference in renal uptake between the two groups was observed when a peptide dose of 5 pmol was used. However, when a higher peptide dose of 2 nmol was administered for SPECT imaging, a significant difference was measured, both on SPECT and ex vivo (see Table 3).
Table 3
Renal uptake of radiolabelled peptides measured ex vivo 3 h after injection in wild-type and megalin-deficient mice
Peptide
Gender
Wild-type (%ID/g ± SD)
Megalin-deficient (%ID/g ± SD)
Ratio megalin-deficient/wild-type
111In-Octreotide
Female
38±9.8
8.4±3.5
22% (p = 0.0007)
Male
25±12
7.9±2.7
32% (p = 0.03)
111In-Exendin 2 nmola
Female
331±33
215±34
65% (p = 0.001)
111In-Exendin 5 pmola
Female
225±35
183±39
81% (ns)
111In-Minigastrin
Female
66±5.9
17±9.1
26% (p < 0.0001)
111In-Neurotensin
Female
5.1±0.94
1.7±0.18
34% (p < 0.0001)
ns not significant.
a111In-Exendin was measured ex vivo in two experiments: after the final SPECT scan using 2 nmol peptide, and in a separate biodistribution experiment using 5 pmol peptide.
Autoradiography of the kidneys revealed a patchy distribution of the 111In-labelled peptides, mainly in the renal cortex (Fig. 4), and confirmed lower uptake in the kidneys of the megalin-deficient mice.
Biodistribution of 111In-labelled octreotide, octreotate, neurotensin and minigastrin in organs other than the kidneys did not differ significantly between the wild-type and megalin-deficient mice (data not shown). For the high dose of 111In-exendin (2 nmol), lung uptake in the wild-type animals was significantly higher than in the megalin-deficient mice (1.2±0.18 %ID/g vs. 0.23±0.037 %ID/g in females, and 0.98±0.039 %ID/g vs. 0.26±0.034 %ID/g in males; both p < 0.0001). However, in the animals that received the low dose of 5 pmol 111In-exendin, no difference in lung uptake was observed: in both the wild-type and the megalin-deficient mice, lung uptake was 11 %ID/g, much higher than in the animals that received 2 nmol of peptide.

Correlation between biodistribution and SPECT

The correlation between renal uptake values measured by biodistribution and SPECT was very good, as depicted in Fig. 5. Spearman’s rank correlation coefficient was 0.924 (r 2 = 0.85, p < 0.0005).

Discussion

In the present study we showed that the multiligand receptor megalin is involved in the proximal tubular reabsorption of 111In-octreotate, 111In-minigastrin, 111In-neurotensin and probably of 111In-exendin, and we confirmed megalin’s role in the reabsorption of 111In-octreotide: the renal uptake of these peptides in megalin-deficient mice was reduced to 23–65% of the uptake in wild-type mice. The remaining renal uptake of the radiolabelled peptides in the megalin-deficient mice may have been due to residual megalin expression. The knock-out of megalin expression in the kidneys of these megalinlox/lox; apoECre mice occurs in a mosaic pattern, with a considerable percentage of tubular cells expressing normal levels of megalin, presumably caused by insufficient expression of Cre recombinase. Leheste et al. initially reported that approximately 10% of the proximal tubular cells of these mice express normal levels of megalin [12], and Motoyoshi et al. reported residual megalin expression in 35–50% of proximal tubular cells [23]. Immunohistochemical staining of kidney sections in the present study confirmed residual megalin expression and considerable variation between individual mice. In addition to residual expression of megalin, part of the residual uptake may also be explained by the involvement of other uptake mechanisms in the reabsorption of these peptides, such as fluid phase endocytosis [24] or other receptors. Proximal tubular expression of ligand-specific receptors for somatostatin, glucagon-like peptide-1 and cholecystokinin2 has been described [2527]. Cubilin, another multiligand receptor, is dependent on other transmembrane proteins such as megalin and amnionless for its internalization [28, 29]. In megalin-deficient mice, the internalization of cubilin may therefore also be reduced.
The data obtained in the current study with 111In-exendin were paradoxical: significantly reduced renal uptake in megalin-deficient mice was measured when the animals received an imaging dose of 2 nmol, but no significant difference between the two groups was found at a biodistribution dose of 5 pmol 111In-exendin. This paradox may very well be caused by the residual megalin expression in the megalin-deficient mice. The residual megalin receptors may still be able to internalize much of the 5 pmol dose, while they are saturated by the 2 nmol dose. Even though proximal tubular endocytosis is regarded as a low-specificity, high-capacity pathway, it can be blocked competitively by excesses of ligands [30, 31]. The difference between 111In-exendin and the other peptides in this respect might be explained by a relatively high affinity of 111In-exendin for megalin and/or associated receptors such as cubilin. This may also account for the very high baseline renal uptake of this peptide (225–371 %ID/g) and suggests that megalin is indeed involved in the renal reabsorption of 111In-exendin.
The renal uptake of the radiolabelled peptides at 3 h after injection was similar in male and female mice. However, at 24 h after injection the uptake of 111In-octreotide and 111In-octreotate was significantly lower in male mice, which has been reported previously [13]. This suggests that the proximal tubular processing and retention of 111In-labelled somatostatin analogues differ between male and female mice, leading to more rapid washout of radioactivity in males. However, the ratios of uptake between megalin-deficient and wild-type mice were comparable in both genders at both time-points, which suggests that the role of megalin in the reabsorption of radiopeptides is independent of gender influences.
Reduced proximal tubular reabsorption of radiolabelled peptides would be expected to cause increased excretion of the radiolabelled peptides in the urine without affecting plasma clearance, since glomerular filtration is not affected and reabsorbed radiolabelled peptides are not returned to the bloodstream. This was confirmed by our observation that the radioactivity concentrations of these radiolabelled peptides in the blood and in most organs other than the kidneys did not differ between megalin-deficient and wild-type mice. The one exception, lower uptake of 111In-exendin in the lungs of megalin-deficient mice, was only observed at the high peptide dose of 2 nmol. At the lower dose of 5 pmol, no difference between megalin-deficient and wild-type animals was observed, and lung uptake was approximately 5-fold to 20-fold higher than in the 2-nmol groups. The lower uptake at the higher peptide dose was most likely due to saturation of specific lung uptake, which has also been described recently by Brom et al. [19]. The lower lung uptake in the megalin-deficient mice in this group may indicate involvement of megalin in the pulmonary uptake of this peptide. Megalin is expressed in the lungs [32], and this high-capacity receptor may contribute significantly to peptide uptake at higher concentrations, when specific glucagon-like peptide-1 receptor mediated uptake (high affinity but lower capacity) is saturated. Although the megalinlox/lox; apoECre mice are described as kidney-specific megalin-deficient mice [12], apoE is also expressed in the lungs [33], which may cause reduced expression of megalin in the lungs of these animals. Possible lower expression of megalin in the lungs is unlikely to have affected the renal uptake of the studied peptides, as their concentrations in blood and other organs did not differ significantly between the megalin-deficient and wild-type mice.
In this study the renal uptake measured ex vivo and on SPECT images correlated very well, which confirms the previously reported accuracy of the small-animal SPECT system [34]. SPECT enables serial measurements in vivo and possibly the reuse of animals in multiple experiments. However, SPECT remains much less sensitive than ex vivo organ measurement, which means that only organs with a considerable uptake can reliably be imaged and relatively high radiation doses have to be administered. This can lead to complications: in the present study, mice that received a relatively high kidney radiation dose from 111In-exendin developed long-term kidney damage, which has been described and analysed in detail elsewhere [22]. In addition, high activity doses require administration of a relatively high peptide dose, which can influence a peptide’s pharmacokinetics and biodistribution [19, 35].
Our results indicate that megalin plays an important role in the renal reabsorption of these diverse 111In-labelled peptides. The variety of radiolabelled peptides and other ligands that are taken up via megalin suggests that it may also be involved in the renal reabsorption of other radiolabelled peptides. The knowledge that megalin is involved in the renal uptake of these radiolabelled peptides may help in the development of new strategies for the reduction of PRRT-induced nephrotoxicity. For example, the reduction in renal uptake of radiolabelled peptides by coadministration of compounds such as succinylated gelatin and albumin fragments [21, 3638], is probably due to their competitive binding to megalin. Lysine, a positively charged amino acid, is used to reduce nephrotoxicity of clinical somatostatin analogue PRRT [10]. However, Gotthardt et al. have shown that lysine does not reduce the renal uptake of negatively charged minigastrin analogues, whereas negatively charged polyglutamic acids do reduce uptake of 111In-minigastrin, but not of the positively charged 111In-octreotide [9]. These observations suggest that different mechanisms are involved in the uptake of these radiolabelled peptides and seems to contradict our present findings that megalin is involved in the renal uptake of both peptides. However, megalin contains four large binding domains [6], and lysine and polyglutamic acid may bind selectively to distinct regions of these domains, thereby only interfering with the binding of specific radiolabelled peptides. The peptide mixtures in succinylated gelatin and albumin fragments are more likely to block several of megalin’s binding regions, thereby interfering with the binding and uptake of several different radiolabelled peptides.
In conclusion, the multiligand receptor megalin plays an important role in the renal reabsorption of 111In-labelled octreotide, octreotate, minigastrin, exendin and neurotensin. This knowledge may be used in the development of new methods to reduce the renal retention of these peptides, thus reducing the risk of nephrotoxicity and improving the safety and effectiveness of PRRT.

Acknowledgments

We would like to express our thanks to Thomas E. Willnow and his group for kindly providing the megalin-deficient mice. Furthermore, we would like to thank Saskia Berndsen, Bianca Lemmers-van de Weem, Kitty Lemmens-Hermans, Erik de Blois, Jan de Swart, Suzanne Reneman, Cathelijne Frielink, Janneke Molkenboer-Kuenen, Lieke Joosten, Gabie de Jong, Gerben Franssen, Rafke Schoffelen and René te Morsche for their invaluable help with the animal experiments.

Conflicts of interest

None.

Open Access

This article is distributed under the terms of the Creative Commons Attribution Noncommercial License which permits any noncommercial use, distribution, and reproduction in any medium, provided the original author(s) and source are credited.
Open AccessThis is an open access article distributed under the terms of the Creative Commons Attribution Noncommercial License (https://​creativecommons.​org/​licenses/​by-nc/​2.​0), which permits any noncommercial use, distribution, and reproduction in any medium, provided the original author(s) and source are credited.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Radiologie

Kombi-Abonnement

Mit e.Med Radiologie erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Radiologie, den Premium-Inhalten der radiologischen Fachzeitschriften, inklusive einer gedruckten Radiologie-Zeitschrift Ihrer Wahl.

Literatur
1.
Zurück zum Zitat Christensen EI, Gburek J. Protein reabsorption in renal proximal tubule-function and dysfunction in kidney pathophysiology. Pediatr Nephrol. 2004;19:714–21.PubMedCrossRef Christensen EI, Gburek J. Protein reabsorption in renal proximal tubule-function and dysfunction in kidney pathophysiology. Pediatr Nephrol. 2004;19:714–21.PubMedCrossRef
2.
Zurück zum Zitat Behr TM, Goldenberg DM, Becker W. Reducing the renal uptake of radiolabeled antibody fragments and peptides for diagnosis and therapy: present status, future prospects and limitations. Eur J Nucl Med. 1998;25:201–12.PubMedCrossRef Behr TM, Goldenberg DM, Becker W. Reducing the renal uptake of radiolabeled antibody fragments and peptides for diagnosis and therapy: present status, future prospects and limitations. Eur J Nucl Med. 1998;25:201–12.PubMedCrossRef
3.
Zurück zum Zitat Otte A, Herrmann R, Heppeler A, et al. Yttrium-90 DOTATOC: first clinical results. Eur J Nucl Med. 1999;26:1439–47.PubMedCrossRef Otte A, Herrmann R, Heppeler A, et al. Yttrium-90 DOTATOC: first clinical results. Eur J Nucl Med. 1999;26:1439–47.PubMedCrossRef
4.
Zurück zum Zitat Valkema R, Pauwels SA, Kvols LK, et al. Long-term follow-up of renal function after peptide receptor radiation therapy with (90)Y-DOTA(0), Tyr(3)-octreotide and (177)Lu-DOTA(0), Tyr(3)-octreotate. J Nucl Med. 2005;46 Suppl 1:83S–91S.PubMed Valkema R, Pauwels SA, Kvols LK, et al. Long-term follow-up of renal function after peptide receptor radiation therapy with (90)Y-DOTA(0), Tyr(3)-octreotide and (177)Lu-DOTA(0), Tyr(3)-octreotate. J Nucl Med. 2005;46 Suppl 1:83S–91S.PubMed
5.
Zurück zum Zitat Christensen EI, Birn H. Megalin and cubilin: multifunctional endocytic receptors. Nat Rev Mol Cell Biol. 2002;3:256–66.PubMed Christensen EI, Birn H. Megalin and cubilin: multifunctional endocytic receptors. Nat Rev Mol Cell Biol. 2002;3:256–66.PubMed
6.
Zurück zum Zitat Orlando RA, Exner M, Czekay RP, et al. Identification of the second cluster of ligand-binding repeats in megalin as a site for receptor-ligand interactions. Proc Natl Acad Sci U S A. 1997;94:2368–73.PubMedCrossRef Orlando RA, Exner M, Czekay RP, et al. Identification of the second cluster of ligand-binding repeats in megalin as a site for receptor-ligand interactions. Proc Natl Acad Sci U S A. 1997;94:2368–73.PubMedCrossRef
7.
Zurück zum Zitat De Jong M, Barone R, Krenning E, et al. Megalin is essential for renal proximal tubule reabsorption of (111)In-DTPA-octreotide. J Nucl Med. 2005;46:1696–700.PubMed De Jong M, Barone R, Krenning E, et al. Megalin is essential for renal proximal tubule reabsorption of (111)In-DTPA-octreotide. J Nucl Med. 2005;46:1696–700.PubMed
8.
Zurück zum Zitat Behr TM, Behe M, Angerstein C, et al. Cholecystokinin-B/gastrin receptor binding peptides: preclinical development and evaluation of their diagnostic and therapeutic potential. Clin Cancer Res. 1999;5:3124s–38s.PubMed Behr TM, Behe M, Angerstein C, et al. Cholecystokinin-B/gastrin receptor binding peptides: preclinical development and evaluation of their diagnostic and therapeutic potential. Clin Cancer Res. 1999;5:3124s–38s.PubMed
9.
Zurück zum Zitat Gotthardt M, Van Eerd-Vismale J, Oyen WJ, et al. Indication for different mechanisms of kidney uptake of radiolabeled peptides. J Nucl Med. 2007;48:596–601.PubMedCrossRef Gotthardt M, Van Eerd-Vismale J, Oyen WJ, et al. Indication for different mechanisms of kidney uptake of radiolabeled peptides. J Nucl Med. 2007;48:596–601.PubMedCrossRef
10.
Zurück zum Zitat Rolleman EJ, Valkema R, De Jong M, Kooij PP, Krenning EP. Safe and effective inhibition of renal uptake of radiolabelled octreotide by a combination of lysine and arginine. Eur J Nucl Med Mol Imaging. 2003;30:9–15.PubMedCrossRef Rolleman EJ, Valkema R, De Jong M, Kooij PP, Krenning EP. Safe and effective inhibition of renal uptake of radiolabelled octreotide by a combination of lysine and arginine. Eur J Nucl Med Mol Imaging. 2003;30:9–15.PubMedCrossRef
11.
Zurück zum Zitat Rolleman EJ, Melis M, Valkema R, Boerman OC, Krenning EP, De Jong M. Kidney protection during peptide receptor radionuclide therapy with somatostatin analogues. Eur J Nucl Med Mol Imaging. 2010;37:1018–31.PubMedCrossRef Rolleman EJ, Melis M, Valkema R, Boerman OC, Krenning EP, De Jong M. Kidney protection during peptide receptor radionuclide therapy with somatostatin analogues. Eur J Nucl Med Mol Imaging. 2010;37:1018–31.PubMedCrossRef
12.
Zurück zum Zitat Leheste JR, Melsen F, Wellner M, et al. Hypocalcemia and osteopathy in mice with kidney-specific megalin gene defect. FASEB J. 2003;17:247–9.PubMed Leheste JR, Melsen F, Wellner M, et al. Hypocalcemia and osteopathy in mice with kidney-specific megalin gene defect. FASEB J. 2003;17:247–9.PubMed
13.
Zurück zum Zitat Melis M, Krenning EP, Bernard BF, De Visser M, Rolleman E, De Jong M. Renal uptake and retention of radiolabeled somatostatin, bombesin, neurotensin, minigastrin and CCK analogues: species and gender differences. Nucl Med Biol. 2007;34:633–41.PubMedCrossRef Melis M, Krenning EP, Bernard BF, De Visser M, Rolleman E, De Jong M. Renal uptake and retention of radiolabeled somatostatin, bombesin, neurotensin, minigastrin and CCK analogues: species and gender differences. Nucl Med Biol. 2007;34:633–41.PubMedCrossRef
14.
Zurück zum Zitat Kos CH. Cre/loxP system for generating tissue-specific knockout mouse models. Nutr Rev. 2004;62:243–6.PubMed Kos CH. Cre/loxP system for generating tissue-specific knockout mouse models. Nutr Rev. 2004;62:243–6.PubMed
15.
Zurück zum Zitat Wu F. Conditional targeting in the kidney. Nephron Physiol. 2007;107:10–6.CrossRef Wu F. Conditional targeting in the kidney. Nephron Physiol. 2007;107:10–6.CrossRef
16.
Zurück zum Zitat Bakker WH, Albert R, Bruns C, et al. [111In-DTPA-D-Phe1]-octreotide, a potential radiopharmaceutical for imaging of somatostatin receptor-positive tumors: synthesis, radiolabeling and in vitro validation. Life Sci. 1991;49:1583–91.PubMedCrossRef Bakker WH, Albert R, Bruns C, et al. [111In-DTPA-D-Phe1]-octreotide, a potential radiopharmaceutical for imaging of somatostatin receptor-positive tumors: synthesis, radiolabeling and in vitro validation. Life Sci. 1991;49:1583–91.PubMedCrossRef
17.
Zurück zum Zitat De Jong M, Breeman WA, Bakker WH, et al. Comparison of (111)In-labeled somatostatin analogues for tumor scintigraphy and radionuclide therapy. Cancer Res. 1998;58:437–41.PubMed De Jong M, Breeman WA, Bakker WH, et al. Comparison of (111)In-labeled somatostatin analogues for tumor scintigraphy and radionuclide therapy. Cancer Res. 1998;58:437–41.PubMed
18.
Zurück zum Zitat De Visser M, Janssen PJ, Srinivasan A, et al. Stabilised 111In-labelled DTPA- and DOTA-conjugated neurotensin analogues for imaging and therapy of exocrine pancreatic cancer. Eur J Nucl Med Mol Imaging. 2003;30:1134–9.PubMedCrossRef De Visser M, Janssen PJ, Srinivasan A, et al. Stabilised 111In-labelled DTPA- and DOTA-conjugated neurotensin analogues for imaging and therapy of exocrine pancreatic cancer. Eur J Nucl Med Mol Imaging. 2003;30:1134–9.PubMedCrossRef
19.
Zurück zum Zitat Brom M, Oyen WJ, Joosten L, Gotthardt M, Boerman OC. (68)Ga-labelled exendin-3, a new agent for the detection of insulinomas with PET. Eur J Nucl Med Mol Imaging. 2010;37:1345–55.PubMedCrossRef Brom M, Oyen WJ, Joosten L, Gotthardt M, Boerman OC. (68)Ga-labelled exendin-3, a new agent for the detection of insulinomas with PET. Eur J Nucl Med Mol Imaging. 2010;37:1345–55.PubMedCrossRef
20.
Zurück zum Zitat Behe M, Becker W, Gotthardt M, Angerstein C, Behr TM. Improved kinetic stability of DTPA-dGlu as compared with conventional monofunctional DTPA in chelating indium and yttrium: preclinical and initial clinical evaluation of radiometal labelled minigastrin derivatives. Eur J Nucl Med Mol Imaging. 2003;30:1140–6.PubMedCrossRef Behe M, Becker W, Gotthardt M, Angerstein C, Behr TM. Improved kinetic stability of DTPA-dGlu as compared with conventional monofunctional DTPA in chelating indium and yttrium: preclinical and initial clinical evaluation of radiometal labelled minigastrin derivatives. Eur J Nucl Med Mol Imaging. 2003;30:1140–6.PubMedCrossRef
21.
Zurück zum Zitat Vegt E, Eek A, Oyen WJ, De Jong M, Gotthardt M, Boerman OC. Albumin-derived peptides efficiently reduce renal uptake of radiolabelled peptides. Eur J Nucl Med Mol Imaging. 2010;37:226–34.PubMedCrossRef Vegt E, Eek A, Oyen WJ, De Jong M, Gotthardt M, Boerman OC. Albumin-derived peptides efficiently reduce renal uptake of radiolabelled peptides. Eur J Nucl Med Mol Imaging. 2010;37:226–34.PubMedCrossRef
22.
Zurück zum Zitat Melis M, Vegt E, Konijnenberg MW, et al. Nephrotoxicity in mice after repeated imaging using 111In-labeled peptides. J Nucl Med. 2010;51:973–7.PubMedCrossRef Melis M, Vegt E, Konijnenberg MW, et al. Nephrotoxicity in mice after repeated imaging using 111In-labeled peptides. J Nucl Med. 2010;51:973–7.PubMedCrossRef
23.
Zurück zum Zitat Motoyoshi Y, Matsusaka T, Saito A, et al. Megalin contributes to the early injury of proximal tubule cells during nonselective proteinuria. Kidney Int. 2008;74:1262–9.PubMedCrossRef Motoyoshi Y, Matsusaka T, Saito A, et al. Megalin contributes to the early injury of proximal tubule cells during nonselective proteinuria. Kidney Int. 2008;74:1262–9.PubMedCrossRef
24.
Zurück zum Zitat Barone R, Van der Smissen P, Devuyst O, et al. Endocytosis of the somatostatin analogue, octreotide, by the proximal tubule-derived opossum kidney (OK) cell line. Kidney Int. 2005;67:969–76.PubMedCrossRef Barone R, Van der Smissen P, Devuyst O, et al. Endocytosis of the somatostatin analogue, octreotide, by the proximal tubule-derived opossum kidney (OK) cell line. Kidney Int. 2005;67:969–76.PubMedCrossRef
25.
Zurück zum Zitat Bates CM, Kegg H, Petrevski C, Grady S. Expression of somatostatin receptors 3, 4, and 5 in mouse kidney proximal tubules. Kidney Int. 2003;63:53–63.PubMedCrossRef Bates CM, Kegg H, Petrevski C, Grady S. Expression of somatostatin receptors 3, 4, and 5 in mouse kidney proximal tubules. Kidney Int. 2003;63:53–63.PubMedCrossRef
26.
Zurück zum Zitat Schlatter P, Beglinger C, Drewe J, Gutmann H. Glucagon-like peptide 1 receptor expression in primary porcine proximal tubular cells. Regul Pept. 2007;141:120–8.PubMedCrossRef Schlatter P, Beglinger C, Drewe J, Gutmann H. Glucagon-like peptide 1 receptor expression in primary porcine proximal tubular cells. Regul Pept. 2007;141:120–8.PubMedCrossRef
27.
Zurück zum Zitat de Weerth A, Jonas L, Schade R, et al. Gastrin/cholecystokinin type B receptors in the kidney: molecular, pharmacological, functional characterization, and localization. Eur J Clin Invest. 1998;28:592–601.PubMedCrossRef de Weerth A, Jonas L, Schade R, et al. Gastrin/cholecystokinin type B receptors in the kidney: molecular, pharmacological, functional characterization, and localization. Eur J Clin Invest. 1998;28:592–601.PubMedCrossRef
28.
Zurück zum Zitat Coudroy G, Gburek J, Kozyraki R, et al. Contribution of cubilin and amnionless to processing and membrane targeting of cubilin-amnionless complex. J Am Soc Nephrol. 2005;16:2330–7.PubMedCrossRef Coudroy G, Gburek J, Kozyraki R, et al. Contribution of cubilin and amnionless to processing and membrane targeting of cubilin-amnionless complex. J Am Soc Nephrol. 2005;16:2330–7.PubMedCrossRef
29.
Zurück zum Zitat Hammad SM, Barth JL, Knaak C, Argraves WS. Megalin acts in concert with cubilin to mediate endocytosis of high density lipoproteins. J Biol Chem. 2000;275:12003–8.PubMedCrossRef Hammad SM, Barth JL, Knaak C, Argraves WS. Megalin acts in concert with cubilin to mediate endocytosis of high density lipoproteins. J Biol Chem. 2000;275:12003–8.PubMedCrossRef
30.
Zurück zum Zitat ten Dam MAGJ, Branten AJW, Klasen IS, Wetzels JFM. The gelatin-derived plasma substitute Gelofusine causes low-molecular-weight proteinuria by decreasing tubular protein reabsorption. J Crit Care. 2001;16:115–20.PubMedCrossRef ten Dam MAGJ, Branten AJW, Klasen IS, Wetzels JFM. The gelatin-derived plasma substitute Gelofusine causes low-molecular-weight proteinuria by decreasing tubular protein reabsorption. J Crit Care. 2001;16:115–20.PubMedCrossRef
31.
Zurück zum Zitat Mogensen CE, Sølling. Studies on renal tubular protein reabsorption: partial and near complete inhibition by certain amino acids. Scand J Clin Lab Invest. 1977;37:477–86.PubMedCrossRef Mogensen CE, Sølling. Studies on renal tubular protein reabsorption: partial and near complete inhibition by certain amino acids. Scand J Clin Lab Invest. 1977;37:477–86.PubMedCrossRef
32.
Zurück zum Zitat Zheng G, Bachinsky DR, Stamenkovic I, et al. Organ distribution in rats of two members of the low-density lipoprotein receptor gene family, gp330 and LRP/alpha 2MR, and the receptor-associated protein (RAP). J Histochem Cytochem. 1994;42:531–42.PubMed Zheng G, Bachinsky DR, Stamenkovic I, et al. Organ distribution in rats of two members of the low-density lipoprotein receptor gene family, gp330 and LRP/alpha 2MR, and the receptor-associated protein (RAP). J Histochem Cytochem. 1994;42:531–42.PubMed
33.
Zurück zum Zitat Srivastava RA, Bhasin N, Srivastava N. Apolipoprotein E gene expression in various tissues of mouse and regulation by estrogen. Biochem Mol Biol Int. 1996;38:91–101.PubMed Srivastava RA, Bhasin N, Srivastava N. Apolipoprotein E gene expression in various tissues of mouse and regulation by estrogen. Biochem Mol Biol Int. 1996;38:91–101.PubMed
34.
Zurück zum Zitat Forrer F, Valkema R, Bernard B, et al. In vivo radionuclide uptake quantification using a multi-pinhole SPECT system to predict renal function in small animals. Eur J Nucl Med Mol Imaging. 2006;33:1214–7.PubMedCrossRef Forrer F, Valkema R, Bernard B, et al. In vivo radionuclide uptake quantification using a multi-pinhole SPECT system to predict renal function in small animals. Eur J Nucl Med Mol Imaging. 2006;33:1214–7.PubMedCrossRef
35.
Zurück zum Zitat De Jong M, Breeman WA, Bernard BF, et al. Tumour uptake of the radiolabelled somatostatin analogue [DOTA0, TYR3]octreotide is dependent on the peptide amount. Eur J Nucl Med. 1999;26:693–8.PubMedCrossRef De Jong M, Breeman WA, Bernard BF, et al. Tumour uptake of the radiolabelled somatostatin analogue [DOTA0, TYR3]octreotide is dependent on the peptide amount. Eur J Nucl Med. 1999;26:693–8.PubMedCrossRef
36.
Zurück zum Zitat Van Eerd JE, Vegt E, Wetzels JF, et al. Gelatin-based plasma expander effectively reduces renal uptake of 111In-octreotide in mice and rats. J Nucl Med. 2006;47:528–33.PubMed Van Eerd JE, Vegt E, Wetzels JF, et al. Gelatin-based plasma expander effectively reduces renal uptake of 111In-octreotide in mice and rats. J Nucl Med. 2006;47:528–33.PubMed
37.
Zurück zum Zitat Vegt E, Wetzels JF, Russel FG, et al. Renal uptake of radiolabeled octreotide in human subjects is efficiently inhibited by succinylated gelatin. J Nucl Med. 2006;47:432–6.PubMed Vegt E, Wetzels JF, Russel FG, et al. Renal uptake of radiolabeled octreotide in human subjects is efficiently inhibited by succinylated gelatin. J Nucl Med. 2006;47:432–6.PubMed
38.
Zurück zum Zitat Vegt E, Van Eerd JE, Eek A, et al. Reducing renal uptake of radiolabeled peptides using albumin fragments. J Nucl Med. 2008;49:1506–11.PubMedCrossRef Vegt E, Van Eerd JE, Eek A, et al. Reducing renal uptake of radiolabeled peptides using albumin fragments. J Nucl Med. 2008;49:1506–11.PubMedCrossRef
Metadaten
Titel
Renal uptake of different radiolabelled peptides is mediated by megalin: SPECT and biodistribution studies in megalin-deficient mice
verfasst von
Erik Vegt
Marleen Melis
Annemarie Eek
Monique de Visser
Maarten Brom
Wim J. G. Oyen
Martin Gotthardt
Marion de Jong
Otto C. Boerman
Publikationsdatum
01.04.2011
Verlag
Springer-Verlag
Erschienen in
European Journal of Nuclear Medicine and Molecular Imaging / Ausgabe 4/2011
Print ISSN: 1619-7070
Elektronische ISSN: 1619-7089
DOI
https://doi.org/10.1007/s00259-010-1685-9

Weitere Artikel der Ausgabe 4/2011

European Journal of Nuclear Medicine and Molecular Imaging 4/2011 Zur Ausgabe