Skip to main content

ImmunoPET imaging of amyloid-beta in a rat model of Alzheimer’s disease with a bispecific, brain-penetrating fusion protein

Abstract

Background

Hijacking the transferrin receptor (TfR) is an effective strategy to transport amyloid-beta (Aβ) immuno-positron emission tomography (immunoPET) ligands across the blood–brain barrier (BBB). Such ligands are more sensitive and specific than small-molecule ligands at detecting Aβ pathology in mouse models of Alzheimer’s disease (AD). This study aimed to determine if this strategy would be as sensitive in rats and to assess how TfR affinity affects BBB transport of bispecific immunoPET radioligands.

Methods

Two affinity variants of the rat TfR antibody, OX26, were chemically conjugated to a F(ab′)2 fragment of the anti-Aβ antibody, bapineuzumab (Bapi), to generate two bispecific fusion proteins: OX265-F(ab′)2-Bapi and OX2676-F(ab′)2-Bapi. Pharmacokinetic analyses were performed 4 h and 70 h post-injection of radioiodinated fusion proteins in wild-type (WT) rats. [124I]I-OX265-F(ab′)2-Bapi was administered to TgF344-AD and WT rats for in vivo PET imaging. Ex vivo distribution of injected [124I]I-OX265-F(ab′)2-Bapi and Aβ pathology were assessed.

Results

More [125I]I-OX265-F(ab′)2-Bapi was taken up into the brain 4 h post-administration than [124I]I-OX2676-F(ab′)2-Bapi. [124I]I-OX265-F(ab′)2-Bapi PET visualized Aβ pathology with significantly higher signals in the TgF344-AD rats than in the WT littermates without Aβ pathology. The PET signals significantly correlated with Aβ levels in AD animals.

Conclusion

Affinity to TfR affects how efficiently a TfR-targeting bispecific fusion protein will cross the BBB, such that the higher-affinity bispecific fusion protein crossed the BBB more efficiently. Furthermore, bispecific immunoPET imaging of brain Aβ pathology using TfR-mediated transport provides good imaging contrast between TgF344-AD and WT rats, suggesting that this immunoPET strategy has the potential to be translated to higher species.

Background

Positron emission tomography (PET) is a useful diagnostic tool for neurodegenerative diseases to visualize and quantify aspects of pathology inside the brain in vivo. For example, visualization of amyloid-beta (Aβ) plaques in the brains of Alzheimer’s disease (AD) patients with [11C]Pittsburgh Compound B ([11C]PiB) or other fluorine-18 (18F)-labelled analogues increases the diagnostic confidence of AD [1]. However, these small-molecule tracers do not bind to soluble Aβ aggregates that are assumed to be the toxic species underlying early AD progression [2, 3] or to diffuse Aβ plaques such as those found in AD patients with the Arctic APP mutation [4]. Small-molecule tracers also perform poorly in discriminating between different proteins with similar fibrillary structures (e.g., Aβ vs alpha-synuclein, a protein which aggregates in Parkinson’s disease) [5].

Antibody-based tracers are highly specific to their targets, and were increasingly used for peripheral PET in the oncology field during the last decade [6, 7]. However, one of the main reasons that hinder the use of immunoPET radioligands for central nervous system (CNS) targets is their limited blood–brain barrier (BBB) passage. Only less than 0.05% of the injected antibody dose passes into the brain 2 h after injection, making it difficult to achieve adequate signal-to-noise ratios for in vivo imaging [8,9,10,11].

Antibodies can be engineered to utilize endogenous active transport systems at the BBB to enhance brain uptake, such as transferrin receptor (TfR)-mediated transcytosis [12,13,14]. Targeting the TfR is an effective strategy for increasing brain delivery of therapeutic antibodies and peptides in mice [10, 15,16,17,18,19], rats [20,21,22,23], non-human primates [24,25,26], and human subjects in some clinical trials [27]. This strategy has also proven successful for developing bispecific antibody-based radioligands for Aβ PET in AD mouse models [7, 9,10,11, 28,29,30,31,32,33].

The affinity of bispecific antibodies to TfR influences their BBB-crossing efficacy, and there is an optimal TfR affinity window at therapeutic doses [16, 18, 22, 24, 34,35,36,37]. Antibodies with too high affinity are not released on the abluminal side of brain endothelial cells, while antibodies with too low affinity bind to TfR insufficiently for transcytosis and remain in the blood [16, 18, 22, 24]. This relationship is potentially dose-related. At tracer doses, increasing the affinity directly correlates with higher brain uptake [16].

Bispecific antibody-based radioligands targeting both TfR and Aβ have been shown to detect Aβ pathology and discriminate between AD and WT mice at an earlier disease stage than [11C]PiB [28, 31, 32]. Moreover, a bispecific antibody-based radioligand is more sensitive than [11C]PiB at detecting reductions in Aβ levels in two AD mouse models after treatment with an Aβ-reducing BACE-1 inhibitor [7]. These studies indicate that the bispecific immunoPET radioligands can image more subtle changes in Aβ pathology than small-molecule tracers.

Given this background, the primary aim of this study was to evaluate if immunoPET imaging of Aβ aggregates is equally sensitive in rats as in mice. The second aim was to assess the role of TfR affinity in transport of immunoPET radioligands across the BBB, using two bispecific fusion proteins with different TfR-affinity, OX265-F(ab′)2-Bapi and OX2676-F(ab′)2-Bapi.

Materials and methods

Recombinant protein expression and purification

Two variants of the mouse anti-rat TfR (rTfR) antibody, OX26, were produced: OX265 and OX2676 [22, 38]. OX265 is the WT OX26 antibody and OX2676 has a single alanine mutation in the complementarity determining region 1 on the heavy chain variable region. Both OX26 variants were expressed as mouse IgG2c molecules. The extracellular domain of rTfR (L101 to F761) was also produced with flag and 10xHis tags attached to the C-terminus.

Expression vectors pcDNA3.4 were synthesized by GeneArt Elements (Invitrogen, Carlsbad, CA) and were transfected into the Expi293 Expression System following the manufacturer’s instructions (Life Technologies, Carlsbad, CA). Plasmid DNA (100 µg total; 1:1 ratio of heavy to light chain for antibodies) and ExpiFectamine 293 reagent in Opti-MEM® I medium were added to the Expi293F cells. Cells were incubated at 37 °C, 120 RPM, with 70% humidity and 8% CO2 for 20 h before ExpiFectamine 293 Transfection enhancer 1 and 2 were added. Four days later, cell supernatant was harvested for purification.

OX26 variants were purified on a HiTrap Protein G column (Cytiva, Uppsala, Sweden) with an ÄKTA Purifier system (Cytiva) and eluted with an increasing gradient of 0.7% acetic acid (HAc). rTfR was purified on a HisTrap Excel immobilized metal ion affinity chromatography column (Cytiva) with a binding buffer containing 20 mM Tris and 200 mM NaCl and eluted in the binding buffer with 500 mM imidazole. Following elution, all three proteins underwent buffer exchange to phosphate buffered saline (PBS) on a HiPrep 26/10 desalting column (Cytiva).

Biacore analysis of the OX26 affinity to rTfR

With the Biacore 8 K (Cytiva), anti-mouse IgG from the Mouse Antibody Capture Kit, type 2 (Cytiva) was immobilized on flow cells 1 and 2 of a CM5 chip (Cytiva) following kit instructions. Ten micrograms of OX265, OX2676, or a commercial control OX26 (LS-C43741, LSBio, Seattle, WA) were captured on flow cell 2. A 1-min regeneration step with 10 nM glycine-HCl (pH 1.7) on flow cell 1 succeeded the capture step to ensure no antibody was captured on the reference flow cell. A single cycle kinetics assay with 5 concentration steps from 3.2 to 2000 nM of rTfR for 2 min each was run over both flow cells, followed by 1 h of dissociation. All dilutions were done in HBS-EP + running buffer (0.01 M HEPES, 0.15 M NaCl, 3 mM EDTA and 0.05% v/v Surfactant P20, Cytiva) supplemented with 0.1% w/v bovine serum albumin (BSA). Data were analysed with Biacore Insight Evaluation 3.0.12.15655 (Cytiva) and was fitted with a 1:1 binding model.

F(ab′)2 fragmentation of bapineuzumab (Bapi)

Bapineuzumab (Bapi; Absolute Antibody, Oxford, UK), a humanized monoclonal antibody binding Aβ based on the murine antibody 3D6 and former clinical therapeutic candidate [39], was enzymatically cleaved into F(ab′)2 fragments (F(ab′)2-Bapi) with the cysteine protease IdeS in FragIT columns (Genovis AB, Lund, Sweden). IdeS produces a homogenous preparation of F(ab′)2 fragments by cleaving human IgG at a specific site below the hinge region. F(ab′)2 fragments were purified from Fc fragments and uncleaved antibodies with CaptureSelect Fc (multi-species) Affinity Resin (ThermoFisher Scientific, Stockholm, Sweden). The F(ab′)2 purity was assessed with SDS-PAGE under non-reducing conditions.

Chemical conjugation and purification of bispecific fusion proteins

Trans-cyclooctene (TCO)-functionalized OX265 or OX2676 was chemically conjugated to tetrazine-functionalized F(ab′)2-Bapi via an inverse-electron-demand Diels–Alder (IEDDA) reaction to produce two bispecific fusion proteins: OX265-F(ab′)2-Bapi and OX2676-F(ab′)2-Bapi.

To prepare for chemical conjugation, OX265 or OX2676 (2 mg/ml) was incubated with a 20-fold molar excess of axial TCO-NHS (Conju-Probe, LLC, San Diego, CA) and F(ab′)2-Bapi (4 mg/ml) with a 7-fold molar excess of Tetrazine-PEG5-NHS (Sigma-Aldrich, Stockholm, Sweden) in PBS with 30 mM carbonate buffer (pH 9.6) for 2.5 h, with shaking in darkness at room temperature (RT). After incubation, the buffer was exchanged to PBS with Zeba spin desalting columns 7 K MWCO to remove any free TCO-NHS or Tetrazine-PEG5-NHS. Modified OX26 affinity variants were incubated with 1.5-fold molar excess of modified F(ab′)2-Bapi, with shaking in darkness at RT. After 30 min, a 200-fold molar excess of methyltetrazine-amine HCl salt (Click Chemistry Tools, Scottsdale, AZ) was added to stop any unreacted TCOs from conjugating to unreacted tetrazine on F(ab′)2-Bapi and forming large multi-antibody complexes. Reactions were incubated for another 30 min in darkness before free tetrazine was removed with Zeba spin desalting columns 7k MWCO. Proteins were separated via gel filtration chromatography on a HiLoad® 26/600 Superdex® 200 prep grade column (Cytiva) using an ÄKTA Purifier system. Components of each fraction were assessed with SDS-PAGE. Approximately 2 µg of protein was mixed with LDL Sample Buffer (ThermoFisher Scientific), loaded onto a NuPAGE 3%–8% Tris-Acetate gel (ThermoFisher Scientific) and run at 150 V for 1 h with NuPAGE Tris-Acetate SDS Running Buffer (ThermoFisher Scientific). The gel was washed in water, fixed in 50% methanol and 7% HAc solution, and stained with GelCode Blue Stain Reagent (ThermoFisher Scientific).

Radiochemistry

OX265-F(ab′)2-Bapi and OX2676-F(ab′)2-Bapi were radiolabelled with iodine-124 (124I) or iodine-125 (125I) by the direct iodination Chloramine-T method (Table 1) [40].

Table 1 Radiochemical reaction yield and specific activity of radioligands

For 124I-labelling, 31.5 ± 9.9 MBq of 124I stock solution (Advanced Centre Oncology Macerata, Montecosaro, Italy) was pre-incubated with 33.3% v/v sodium iodine (NaI, 50 µM) for 15 min, and then neutralized with 0.5% HAc and 11.1% v/v 10xPBS. Then, 92.9 ± 57.2 µg of OX265-F(ab′)2-Bapi or 37.0 ± 0 µg of OX2676-F(ab′)2-Bapi and Chloramine-T (final concentration 0.1 mg/ml, Sigma-Aldrich) were added to the pre-incubated 124I solution. The reaction was quenched after 120 s by adding sodium metabisulfite (final concentration 0.2 mg/ml, Sigma-Aldrich).

For 125I-labelling, either 60 µg of OX265-F(ab′)2-Bapi or 23.4 µg of OX265-F(ab′)2-Bapi modified with Bolton-Hunter reagent, as described previously [28], was mixed with 11.1 ± 1.1 MBq of 125I stock solution (PerkinElmer Inc., Waltham, MA), Chloramine-T (final concentration 40 µg/ml) and PBS to a final volume of 110 µl. After incubating 90 s at room temperature, the reaction was stopped with sodium metabisulfite (final concentration 74 µg/ml).

Radiolabelled products were diluted to 500 µl with PBS, purified of free iodine with a disposable NAP-5 size exclusion column (Cytiva) and eluted in 1 ml of PBS. Radiolabelling was performed no more than 2 h prior to each in vivo study.

Quality control of radiolabelled bispecific fusion proteins

Sandwich ELISA was performed to determine the concentrations of bispecific fusion proteins after radiolabelling using plates coated with 2 nM anti-mouse-IgG (#AI-2000, Vector Laboratories Inc., Newark, CA). Indirect ELISA was performed to assess the potential effects of conjugation and radiolabelling on the binding of OX265-F(ab′)2-Bapi or OX2676-F(ab′)2-Bapi to rTfR and Aβ, using plates coated with 13.3 nM of rTfR or 50 nM of Aβ (Innovagen, Lund, Sweden), respectively.

ELISA assays were performed in 96-well half-area plates (Corning Inc., New York, NY). Plates were coated with respective proteins diluted in PBS overnight at 4 °C and then blocked with 1% BSA in PBS for 1 h. All further dilutions were made in ELISA incubation buffer (PBS with 0.1% BSA, 0.05% Tween and 0.15% Kathon). Control antibodies (OX265, OX2676 and Bapi) and the bispecific fusion proteins before and after radiolabelling were serially diluted from 50 nM to 3.2 pM, incubated overnight at 4 °C, washed and then detected with horseradish peroxidase (HRP)-coupled goat anti-mouse IgG-F(ab′)2 (1:2000, #115-035-006, Jackson ImmunoResearch Laboratories, West Grove, PA) or goat anti-human IgG-F(ab′)2 (1:2000, #109-036-006, Jackson ImmunoResearch Laboratories). Signals were developed with K blue aqueous TMB substrate (Neogen Corp., Lexington, KY), halted with 1 M H2SO4 and read with a spectrophotometer at 450 nm. The EC50 values were calculated from agonist concentration vs response curves with variable slope (four parameters) where the bottom was constrained to 0 in GraphPad Prism.

Animals

Animals were housed with ad libitum access to food and water in an approved animal facility at Uppsala University with controlled temperature and humidity. All procedures in this study were approved by the Uppsala County Animal Ethics board (5.8.18–20401/2020) following the rules and regulations of the Swedish Animal Welfare Agency and in compliance with the European Communities Council Directive of 22 September 2010 (2010/63/EU).

WT Fischer 344 rats (Janvier Labs, Le Genest-Saint Isle, France) were used in the pharmacokinetic studies, and TgF344-AD rats and WT littermates for PET scans. The TgF344-AD rats express human APP with the Swedish mutation (AβPP KM670/671NL) and human PSEN1 with exon 9 deletion (PS1-ΔE9). They begin displaying age-dependent Aβ plaque pathology at 6 months [41, 42] and do not show any sex differences in Aβ pathology load [41, 43].

Pharmacokinetic study

Three-month-old male WT rats were lightly sedated with isoflurane (Isoflurane Baxter®, Baxter Medical AB, Kista, Sweden) and injected in the tail vein with either [125I]I-OX265-F(ab′)2-Bapi or [124I]I-OX2676-F(ab′)2-Bapi (Table 2). The rats were anaesthetized with isoflurane 4 h or 70 h post-administration and a terminal blood sample was taken from the heart. The rats were then euthanized by transcardial perfusion with 130 ml of 0.9% NaCl in 8 min to clear the brain and organs of blood. The brain was isolated and dissected into olfactory bulbs, right hemisphere, left cortex, left midbrain and left cerebellum. Brain samples were immediately frozen on dry ice, except for left cortices used for capillary depletion. Lung, heart, liver, pancreas, spleen, kidney, femoral bone, femoral bone marrow, and skull were isolated and a urine sample was collected. Radioactivity from samples was measured with a γ-counter (2480 Wizard™, PerkinElmer Inc.). Concentrations of the bispecific fusion proteins were expressed as standardized uptake values (SUV) to account for variation in weight between animals.

Table 2 Number of rats (male/female), injected radioactivity, and dose for the pharmacokinetic (PK) and the PET studies

Blood pharmacokinetics

Blood samples (8 µl) were obtained from the tail vein at 1 h, 4 h, 24 h, and 48 h post-administration. Terminal blood was collected and plasma was separated from the blood cell pellet by centrifugation. Radioactivity was measured with a γ-counter to calculate blood concentration and the percent of free antibody in plasma.

Whole blood half-life was estimated with a non-linear regression two-phase decay model. The plateau was constrained to zero. Y0 was fixed to 14.3 based on the assumption that 100% of the injected dose enters the blood immediately after injection and the average rat blood volume is 7% of their body weight [44]. The fusion protein exposure, quantified as area under the curve (AUC), was calculated from the SUV blood curves.

Capillary depletion

Capillary depletion was performed on perfused left cortices of rats euthanized 4 h post-injection of [125I]I-OX265-F(ab′)2-Bapi or [124I]I-OX2676-F(ab′)2-Bapi. Immediately after transcardial perfusion, cortices were isolated, weighed and homogenized in 2 ml cold physiological buffer (10 mM HEPES, 141 mM NaCl, 4 mM KCl, 2.8 mM CaCl2, 1 mM MgSO4, 1 mM NaH2PO4, 10 mM D-glucose, pH 7.4) with 8 strokes in an ice-cold Dounce homogeniser. Then 4 ml of 30% Ficoll 400 (Sigma-Aldrich) was added, followed by an additional 2 strokes. The homogenate was centrifuged at 5200 g for 20 min at 4 °C resulting in two fractions: a capillary-enriched pellet and a parenchymal supernatant. Measured radioactivity for each fraction was normalized to the total activity of the homogenate.

PET/computed tomography (CT) imaging

Fourteen-month-old male and female TgF344-AD rats and WT littermates were lightly sedated and injected with [124I]I-OX265-F(ab′)2-Bapi in the tail vein (Table 2). The day before injection, animals were given water containing 0.5% NaI to reduce 124I-uptake in the thyroid. After injection, the concentration of NaI was reduced to 0.2% until scanning.

Three days post-injection, rats underwent PET and CT scans. The animals were anesthetized with 5% sevoflurane in 50% medical oxygen and 50% air, and placed on the gantry of a nanoScan® PET/MRI 3T system (Mediso Medical Imaging Systems, Budapest, Hungary) in a prone position for a 60–120-min PET scan (Field of view = 9.8 cm). A 5-min CT scan was taken with a nanoScan® SPECT/CT system (Mediso Medical Imaging Systems) which is compatible with PET/magnetic resonance image (MRI) and allows for correct image co-registration.

PET data were reconstructed using a Tera-TomoTM 3D algorithm (Mediso Medical Imaging Systems) with 4 iterations and 6 subsets. CT data were reconstructed using Filter Back Projection. Further PET and CT image processing was performed with Amide 1.0.4 [45]. CT and PET scans were manually aligned to a T2-weighted MRI-based rat brain atlas [46]. The following regions of interest were outlined in the MRI: caudate putamen, hippocampi, parietal cortex, occipital cortex, cerebellum and olfactory bulbs. PET data were quantified as mean radioactivity concentrations during the scan expressed as SUV.

Ex vivo analysis of TgF344-AD rats

Immediately after the CT scan, animals were euthanized and dissected as described above, except that brains were dissected into olfactory bulbs, right hemisphere, left cortex (left front half of the cortex), left hippocampus and left cerebellum. Brain samples were immediately frozen and the radioactivity in the collected tissues, blood and urine samples was measured with a γ-counter. Bispecific fusion protein concentrations were expressed as SUVex vivo to account for weight variations between animals.

Ex vivo autoradiography

Frozen hemispheres from PET/CT-scanned rats were cryosectioned (CM1850, Leica Biosystems, Nussloch, Germany) into 20-µm sagittal sections. Two sections from each animal were immediately exposed to a phosphor imaging plate (MS, MultiSensitive, PerkinElmer Inc.) for 7 days. The plates were scanned in a Typhoon phosphor imager (Cytiva) at 600 dots per inch. The resulting digital images were converted to a false colour scale (Royal) in ImageJ.

Aβ immunofluorescence

Sagittal brain slices were fixed in 4% paraformaldehyde and washed in PBS. Antigen retrieval was performed by boiling sections in 25 mM citrate buffer (pH 7.3) for 2 min and leaving them to cool to RT for 40 min. The sections were treated with 70% formic acid (FA) for 10 min, rinsed in milliQ water, washed in PBS and permeabilized in 0.4% Triton in PBS for 5 min. Primary antibody was added to the sections and incubated overnight at 4 °C with slow shaking. The next day, sections were washed in PBS. Secondary antibody was added and incubated for 1 h with shaking at RT, followed by three washes, before mounting with Vectashield Hard Set Mounting medium with DAPI (BioNordika, Solna, Sweden). Fluorescence images were acquired with a Zeiss Observer Z1 microscope (Carl Zeiss Imaging GmBH, Jena, Germany) and processed using ZEN software.

42 immunofluorescence was performed on sagittal brain slices from PET animals. The primary antibody was rabbit anti-human Aβ42 (#700,254, Invitrogen) diluted to 1 µg/ml in 0.1% Tween-20 in PBS. The secondary antibody was Alexa Fluor 488 goat anti-rabbit IgG (1:500 in PBS, #A11008, Invitrogen).

Overall Aβ immunofluorescence was performed on sagittal brain slices from 15-month-old TgF344-AD and WT rats. Sections were also blocked with Mouse on Mouse (M.O.M.) immunodetection kit (Vector Laboratories) according to kit instructions before permeabilization. The primary antibody was mAb3D6 (murine version of Bapi [47]) diluted to 4 µg/ml in M.O.M. diluent. The secondary antibody was Alexa Fluor 488 goat-anti-mouse IgG (1:500 in PBS, #A11029, Invitrogen).

Biochemical Aβ analysis

Brain Aβ aggregate concentrations in rats that underwent PET scanning were measured with sandwich ELISA as previously described [28]. Briefly, isolated cortex, hippocampus, and cerebellum were homogenized separately with 4 × 10 s spins at 5500 rpm in a Precellys® Evolution (Bertin Instruments, Montigny-le-Bretonneux, France) at a 1:5 weight-to-volume ratio in Tris-buffered saline (TBS) with Complete Protease Inhibitor Cocktail Tablets (Roche Diagnostics International AG, Rotkreuz, Switzerland). Samples were centrifuged at 16,000 g at 4 °C for 1 h. Supernatants were collected carefully. Pellets were homogenized in 70% FA at a weight-to-volume ratio of 1:5 and centrifuged at 16,000 g at 4 °C for 1 h. Again, supernatants were collected.

The concentration of soluble Aβ aggregates from each brain region was measured by coating a 96-well half-area plate with 145 ng/well of mAb3D6 overnight and blocking with 1% BSA in PBS for 1 h. Aβ protofibrils (BioArctic) were used as standard. TBS brain extracts were diluted 1:200 and incubated overnight at 4 °C, then detected with biotinylated 3D6 (0.5 µg/ml) and streptavidin-HRP (1:3000, Mabtech AB, Nacka Strand, Sweden). Signals were developed with K blue aqueous TMB substrate, stopped with 1 M H2SO4 and read with a spectrophotometer at 450 nm. In GraphPad Prism 9.1.0, a sigmoidal, 4PL (X is concentration) standard curve was plotted for the interpolation of sample concentrations. All dilutions were made in ELISA incubation buffer.

For total Aβ40 and Aβ42 concentrations, 96-well half-area plates were coated with 100 ng of polyclonal rabbit anti-human Aβ40 (custom production from Agrisera, Vännäs, Sweden) or monoclonal rabbit anti-human Aβ42 (#700,254, Invitrogen), respectively. The next day, plates were blocked with 1% BSA in PBS for 1 h. Aβ40 (Innovagen) and Aβ42 (Innovagen) were used as standards respectively. FA brain extracts were neutralized with 2 M Tris, diluted 1:10,000 and incubated overnight at 4 °C. The procedure continued as described above for the soluble Aβ aggregate ELISA.

Statistical analyses

Statistical analyses were performed in GraphPad Prism 9.1.0 (GraphPad Software, Inc., San Diego, CA). Results are reported as mean ± standard deviation. Statistical assessment was carried out by two-tailed t-test, one-way ANOVA with Tukey’s multiple comparisons test or two-way ANOVA with Šídák’s multiple comparisons test. Linear correlation was expressed by Pearson’s correlation coefficient.

Results

The KD of OX265 was 4.1 nM and the KD of OX2676 was 78.4 nM (Fig. 1a), which were consistent with literature [22]. The KD of the OX265 variant was also similar to that of the commercial control, OX26LSBio. Immunofluorescent staining with mAb3D6, the murine version of Bapi, illustrated that this antibody can bind to the Aβ pathology in TgF344-AD rats but not in WT tissues (Additional file 1: Fig. S1). Therefore, Bapi was a suitable anti-Aβ antibody for use in the bispecific fusion proteins. OX265 or OX2676 was chemically conjugated to F(ab′)2-Bapi to produce two bispecific fusion proteins: OX265-F(ab′)2-Bapi and OX2676-F(ab′)2-Bapi (Fig. 1b). The final bispecific fusion protein products contained OX265 or OX2676 conjugated to 1–3 F(ab′)2-Bapi, as identified by SDS-PAGE analysis (Fig. 1c, d). Compared to OX265, OX265-F(ab′)2-Bapi retained its binding to rTfR before and after 125I-labelling (Fig. 1e; Table 3). Conversely, OX2676-F(ab′)2-Bapi lost most of its binding to rTfR compared with OX2676. The EC50 of OX2676-F(ab′)2-Bapi, both before and after 124I-labelling, was unmeasurable in the given ELISA assay. Both bispecific fusion proteins retained their binding to Aβ before and after radiolabelling compared to the full Bapi IgG (Fig. 1f; Table 3).

Fig. 1
figure 1

Generation of bispecific fusion proteins with OX26 affinity variants. a Biacore analysis of affinity to rTfR for OX265, OX2676 and commercial OX26LSBio. b Schematic of the IEDDA reaction to generate OX26-F(ab′)2-Bapi bispecific fusion proteins. c, d SDS-PAGE analysis of OX265-F(ab′)2-Bapi (c) and OX2676-F(ab′)2-Bapi (d). Lanes 1, molecular weight ladder; 2, parental OX26 IgG; 3, F(ab′)2-Bapi; 4–5, preparative SEC fractions for the final pool of bispecific fusion protein. e, f ELISA analysis of OX265-F(ab′)2-Bapi and OX2676-F(ab′)2-Bapi before and after 125I-labelling binding to rTfR (e) and Aβ (f). RU, response unit; OD, optical density

Table 3 EC50 of antibody binding to rTfR or Aβ in direct ELISA

[125I]I-OX265-F(ab′)2-Bapi had higher brain uptake than [124I]I-OX2676-F(ab′)2-Bapi in WT rats

Four hours post-injection, the concentration of [125I]I-OX265-F(ab′)2-Bapi in WT rats was 1.3- to 9-fold higher than the concentration of [124I]I-OX2676-F(ab′)2-Bapi in the different brain regions measured (P < 0.01, Fig. 2a). Capillary depletion 4 h post-injection determined that the majority of the bispecific fusion protein was distributed to the parenchyma (Fig. 2b), with a slightly higher parenchymal portion for [124I]I-OX2676-F(ab′)2-Bapi (P = 0.03). The whole blood exposure, measured as the AUC, was similar between the bispecific fusion proteins (Fig. 2c), although [125I]I-OX265-F(ab′)2-Bapi had faster elimination than [124I]I-OX2676-F(ab′)2-Bapi in the fast distribution phase (Table 4). The brain-to-blood ratio for [125I]I-OX265-F(ab′)2-Bapi was significantly higher 70 h post-injection compared with 4 h post-injection (P = 0.024) while there was no difference between the two time points for [124I]I-OX2676-F(ab′)2-Bapi (P = 0.999; Fig. 2d). The brain-to-blood ratio 70 h post-administration was also significantly higher for [125I]I-OX265-F(ab′)2-Bapi compared to [124I]I-OX2676-F(ab′)2-Bapi (P = 0.004), indicating that [125I]I-OX265-F(ab′)2-Bapi entered the brain to a higher extent. The percent in plasma, indicative of the amount of free ligand in the blood, was significantly higher for [124I]I-OX2676-F(ab′)2-Bapi compared to [125I]I-OX265-F(ab′)2-Bapi at 4 h post-administration (P = 0.018, Fig. 2e). The peripheral biodistribution was similar for both fusion proteins at 4 h and 70 h post-administration. There was significantly more [125I]I-OX265-F(ab′)2-Bapi than [124I]I-OX2676-F(ab′)2-Bapi in the spleen at both time points (3.5-fold, P = 0.0002 at 4 h and 2-fold, P < 0.0001 at 70 h post-injection) and in the bone marrow at 70 h post-injection (3.3-fold, P < 0.0001) (Fig. 2f, g). Overall, the ex vivo studies showed that OX265-F(ab′)2-Bapi was a more favourable PET ligand candidate .

Fig. 2
figure 2

Ex vivo biodistribution of [125I]I-OX265-F(ab′)2-Bapi or [124I]I-OX2676-F(ab′)2-Bapi in WT rats. a Concentration (SUV) of [125I]I-OX265-F(ab′)2-Bapi or [124I]I-OX2676-F(ab′)2-Bapi in WT rat brain regions 4 h and 70 h post-administration. b Bispecific fusion protein distribution in brain capillaries and parenchyma 4 h post-administration. c Whole blood elimination curves over 3 days post-administration. Curve fit based on a two-phase decay non-linear regression model. d, e Brain-to-blood ratio (d) and percent in plasma (e) 4 h and 70 h post-administration. f, g Peripheral ex vivo biodistribution (SUV) of [125I]I-OX265-F(ab′)2-Bapi or [124I]I-OX2676-F(ab′)2-Bapi in WT rats 4 h (f) and 70 h (g) post-administration. *P ≤ 0.05, **P ≤ 0.01, ***P ≤ 0.001, ****P ≤ 0.0001

Table 4 Whole blood half-lives and area under the curve (AUC) for bispecific fusion protein variants

[124I]I-OX265-F(ab′)2-Bapi PET visualized Aβ pathology in TgF344-AD rats and correlated with ex vivo brain distribution of [124I]I-OX265-F(ab′)2-Bapi

PET scans of TgF344-AD rats showed increased signal in the brain compared with baseline signals in the brains of WT rats, specifically in the cortex and cerebellum (Fig. 3a). Quantification of PET confirmed that TgF344-AD rats had 2- to 4-fold higher radioligand retention than WT rats, depending on the brain region (P < 0.01, Fig. 3b). Furthermore, ex vivo analysis on perfused brains showed significantly higher [124I]I-OX265-F(ab′)2-Bapi retention in all brain regions in TgF344-AD rats compared with WT rats (18- to 32-fold, P < 0.01, Fig. 3c). The PET signal quantification significantly correlated with ex vivo [124I]I-OX265-F(ab′)2-Bapi concentrations in TgF344-AD rat brains (r2 = 0.810, P < 0.0001, Fig. 3d). The brain-to-blood ratio was also significantly higher in TgF344-AD rats than WT rats (P = 0.0003, Fig. 3e). The peripheral distribution of [124I]I-OX265-F(ab′)2-Bapi was similar between TgF344-AD and WT rats (Fig. 3f), except that the WT animals had higher radioactivity concentrations in the spleen (1.2-fold, P = 0.0095) and urine (2.1-fold, P < 0.0001) compared with TgF344-AD rats. Finally, the blood pharmacokinetic profile was not affected by genotype. The TgF344-AD and WT rats had similar [124I]I-OX265-F(ab′)2-Bapi elimination curves in whole blood over 72 h post-administration (Additional file 1: Fig. S2a) and the percent in plasma at 72 h post-administration did not differ significantly (P = 0.76, Additional file 1: Fig. S2b).

Fig. 3
figure 3

[124I]I-OX265-F(ab′)2-Bapi PET images and quantification. a Representative in vivo PET images from TgF344-AD and WT rats 3 days post-administration of [124I]I-OX265-F(ab′)2-Bapi. b PET image-based quantification of [124I]I-OX265-F(ab′)2-Bapi distribution (SUV) in WT and TgF344-AD rat brains. c Ex vivo [124I]I-OX265-F(ab′)2-Bapi retention (SUV) in WT and TgF344-AD rat brains. d Pearson’s correlation between PET-based quantification (SUVPET) and ex vivo retention (SUVex vivo) of [124I]I-OX265-F(ab′)2-Bapi in four brain regions from TgF344-AD rats. e Brain-to-blood ratio in TgF344-AD and WT rats. f Peripheral ex vivo biodistribution (SUV) of [124I]I-OX265-F(ab′)2-Bapi in TgF344-AD and WT rats 3 days post-administration. **P ≤ 0.01, ***P ≤ 0.001, ****P ≤ 0.0001

[124I]I-OX265-F(ab′)2-Bapi PET correlated with Aβ pathology

Ex vivo autoradiography performed on perfused brain sections showed increased signals in the cortex and cerebellum of TgF344-AD rats (Fig. 4). Sagittal sections stained for Aβ42 illustrated that TgF344-AD rats had abundant Aβ42 pathology in the cortex, caudate putamen, hippocampus and cerebellum, displaying both dense and diffuse Aβ deposits (Fig. 4). On the contrary, there was no Aβ42 pathology in WT rat brains.

Fig. 4
figure 4

Representative sagittal images of ex vivo autoradiography and Aβ42 immunofluorescence from TgF344-AD and WT rats after [124I]I-OX265-F(ab′)2-Bapi PET

ELISA analysis of brain homogenates revealed that Aβ levels did not differ significantly among cortex, hippocampus and cerebellum in TgF344-AD rats for total Aβ40, total Aβ42 or soluble Aβ aggregates (Fig. 5a). The PET signal quantification correlated with concentrations of total Aβ40 (P < 0.05) and Aβ42 (P < 0.001) but not with soluble Aβ aggregates (Fig. 5b).

Fig. 5
figure 5

a ELISA quantification of total Aβ40, total Aβ42 and soluble Aβ aggregates in brain regions from TgF344-AD rats. b Pearson’s correlation between PET-based quantification (SUVPET) and Aβ concentrations

Discussion

To investigate the role TfR affinity plays in transport of bispecific immunoPET ligands across the BBB, two bispecific fusion protein variants were produced. In vivo pharmacokinetic studies indicated that 72 h was a good time point for PET imaging since there was little immunoPET ligand signal left in the brains or blood of WT rats, suggesting good contrast for PET imaging. This time corresponded with immunoPET imaging of Aβ in two different transgenic AD mouse models [7, 28]. Furthermore, we found that at 4 h post-administration, there was significantly more [125I]I-OX265-F(ab′)2-Bapi in all brain areas compared with [124I]I-OX2676-F(ab′)2-Bapi. These results contradict previous findings that the OX2676 IgG enters the brain significantly better than the OX265 IgG [22, 36]. One explanation for the higher BBB transcytosis of [125I]I-OX265-F(ab′)2-Bapi is that the conjugation reaction may have an effect on the affinity of the parent antibody to rTfR. After conjugation, OX265-F(ab′)2-Bapi retained a similar rTfR affinity as the parental OX265 IgG. Unexpectedly, the in vitro affinity of OX2676 to rTfR substantially decreased after conjugation, which likely led to the lower brain uptake of [124I]I-OX2676-F(ab′)2-Bapi due to the poor interaction with endogenous TfR [18, 22, 35, 36]. This hypothesis is supported by the higher percent in plasma at 4 h, the longer half-life in the fast distribution phase in whole blood and the lower concentration in the spleen compared with [125I]I-OX265-F(ab′)2-Bapi, all of which indicate insufficient binding to TfR in vivo.

Dosage has also been suggested to influence the effect of TfR affinity on BBB transcytosis, such that higher-affinity binders enter the brain better at tracer doses [9, 16]. This dose-dependency hypothesis could explain the better brain uptake of [125I]I-OX265-F(ab′)2-Bapi since the max dose used here was 0.12 mg/kg, while previous reports with these OX26 variants used therapeutic doses of 10–20 mg/kg [22, 36]. Further studies are necessary to determine the extent to which dosing influences the effects of affinity, specifically for bispecific immunoPET radioligands.

ImmunoPET ligands with TfR-mediated brain uptake have proven more sensitive in detecting Aβ pathology than small-molecule tracers in mice [7, 28, 31, 32]. Previous PET analyses of the TgF344-AD rat model with the amyloid ligands [18F]FDDNP in 15-month-old rats and [18F]Florbetaben in 18-month-old rats both measured small differences in brain pathology between AD and WT rats (1.07- and 1.25-fold increases in signal, respectively) [41, 42]. ImmunoPET with [124I]I-OX265-F(ab′)2-Bapi in slightly younger TgF344-AD rats (14-month-old) showed 2- to 4-fold higher signals than WT littermates, depending on brain region. Taken together, the data presented here indicate that immunoPET ligands may be more sensitive than small-molecule tracers in rats as well.

In TgF344-AD animals, the brain-region SUVs also correlated with the ex vivo levels of [124I]I-OX265-F(ab′)2-Bapi and with the total Aβ40 and Aβ42 levels detected by ELISA. Furthermore, the increased cortical and cerebellar PET signal corresponded with increased ex vivo autoradiography signal and abundant Aβ42 staining in these brain regions in TgF344-AD rats. These results suggest that the PET signal originated from radiolabelled bispecific fusion protein specifically binding to and around Aβ deposits in TgF344-AD rat brains. The high cortical, hippocampal and dorsal striatal (caudate and putamen) signals seen in the immunoPET scans, autoradiography and Aβ42 immunofluorescence correspond with descriptions of pathology in literature [41,42,43]. Cerebellar pathology has been reported to develop later than cortical and hippocampal pathology in TgF344-AD rats [41, 43]. Here, we noticed a strong PET signal in the cerebellum, which corresponds with the high concentration of Aβ in the brain tissue. One explanation for this strong cerebellar PET signal in comparison to previous PET studies [42] could be that, due to the overexpression of Aβ42 in this model and later development of cerebellar pathology, the cerebellum may contain proportionally more diffuse Aβ plaques than other brain regions. Diffuse deposits represent an earlier form of Aβ plaques [48] that are readily detected with [124I]I-OX265-F(ab′)2-Bapi but lack the amyloid core that is detected by traditional Aβ tracers.

Another difference from previous findings [28, 30, 32] is that in this study the PET signals did not correlate with the levels of soluble Aβ aggregates within AD animal groups. Furthermore, the correlations with total Aβ40 and Aβ42 were driven by one AD rat that had higher pathology than the others. This ligand clearly differentiated between AD and WT rats but the lack of strong correlations to Aβ pathology may be because the animals were at the same disease stage. In future studies, the ability of this radioligand to detect pathology changes at different ages and after therapeutic interventions should be explored.

One limitation in this study was the production of bispecific fusion proteins with chemical conjugation. The IEDDA reaction resulted in a heterogeneous product with OX26 IgG conjugated to 1–3 F(ab′)2-Bapi fragments. Further, the two moieties (OX26 IgG and F(ab′)2-Bapi) were conjugated randomly at different sites. This method is unsuitable to produce a clinically applicable bispecific immunoPET radioligand. An alternative would be to use a site-specific conjugation method, such as Sortase A catalysed chemo-enzymatic reaction [49].

Conclusion

In conclusion, we have shown that TfR affinity influences the BBB passage of bispecific immunoPET ligands. However, it remains unclear whether the dose of the bispecific antibody influences the effect of affinity. Finally, we have demonstrated that the TfR-mediated transport of an immunoPET radioligand enables sensitive imaging of brain Aβ pathology in a rat model of AD, expanding its use over mice. Therefore, this strategy for delivery of immunoPET ligands to the CNS could eventually be translated from bench to bedside given the development of suitable human-specific TfR-binders.

Availability of data and materials

The datasets generated and/or analysed during the current study are available from the corresponding author on reasonable request.

Abbreviations

[11C]PiB:

[11C]Pittsburgh Compound B

Aβ:

Amyloid-beta

AD:

Alzheimer’s disease

AUC:

Area under the curve

Bapi:

Bapineuzumab

BBB:

Blood–brain barrier

BSA:

Bovine serum albumin

CNS:

Central nervous system

CT:

Computed tomography

FA:

Formic acid

HAc:

Acetic acid

HRP:

Horseradish peroxidase

IEDDA:

Inverse electron demand Diels–Alder

MRI:

Magnetic resonance image

OD:

Optical density

PET:

Positron emission tomography

PK:

Pharmacokinetic

RT:

Room temperature

rTfR:

Rat transferrin receptor

SUV:

Standardized uptake value

TCO:

Trans-cyclooctene

TfR:

Transferrin receptor

References

  1. Ossenkoppele R, Jansen WJ, Rabinovici GD, Knol DL, van der Flier WM, van Berckel BNM, et al. Prevalence of amyloid PET positivity in dementia syndromes: a meta-analysis. JAMA. 2015;313(19):1939–49.

    Article  Google Scholar 

  2. Lambert MP, Barlow AK, Chromy BA, Edwards C, Freed R, Liosatos M, et al. Diffusible, nonfibrillar ligands derived from Aβ1–42 are potent central nervous system neurotoxins. Proc Natl Acad Sci U S A. 1998;95:6448–53.

    Article  CAS  Google Scholar 

  3. McLean CA, Cherny RA, Fraser FW, Fuller SJ, Smith MJ, Beyreuther K, et al. Soluble pool of Aβ amyloid as a determinant of severity of neurodegeneration in Alzheimer’s disease. Ann Neurol. 1999;46(6):860–6.

    Article  CAS  Google Scholar 

  4. Schöll M, Wall A, Thordardottir S, Ferreira D, Bogdanovic N, Långström B, et al. Low PiB PET retention in presence of pathologic CSF biomarkers in Arctic APP mutation carriers. Neurology. 2012;79(3):229–36.

    Article  Google Scholar 

  5. Sehlin D, Syvänen S. Engineered antibodies: new possibilities for brain PET? Eur J Nucl Med Mol Imaging. 2019;11.

  6. Wei W, Rosenkrans ZT, Liu J, Huang G, Luo QY, Cai W. ImmunoPET: concept, design, and applications. Chem Rev. 2020;120(8):3787–851.

    Article  CAS  Google Scholar 

  7. Meier SR, Sehlin D, Roshanbin S, Lim Falk V, Saito T, Saido TC, et al. 11 C-PIB and 124I-antibody PET provide differing estimates of brain amyloid-beta after therapeutic intervention. J Nucl Med. 2022;63(2):302-9.

  8. Magnusson K, Sehlin D, Syvänen S, Svedberg MM, Philipson O, Söderberg L, et al. Specific uptake of an amyloid-β protofibril-binding antibody-tracer in AβPP transgenic mouse brain. J Alzheimers Dis. 2013;37(1):29–40.

    Article  CAS  Google Scholar 

  9. Hultqvist G, Syvänen S, Fang XT, Lannfelt L, Sehlin D. Bivalent brain shuttle increases antibody uptake by monovalent binding to the transferrin receptor. Theranostics. 2017;7(2):308–18.

    Article  CAS  Google Scholar 

  10. Syvänen S, Hultqvist G, Gustavsson T, Gumucio A, Laudon H, Söderberg L, et al. Efficient clearance of Aβ protofibrils in AβPP-transgenic mice treated with a brain-penetrating bifunctional antibody. Alzheimers Res Ther. 2018;10(1):1–10.

    Google Scholar 

  11. Sehlin D, Stocki P, Gustavsson T, Hultqvist G, Walsh FS, Rutkowski JL, et al. Brain delivery of biologics using a cross-species reactive transferrin receptor 1 VNAR shuttle. FASEB J. 2020;34(10):13272–83.

    Article  CAS  Google Scholar 

  12. Stanimirovic DB, Sandhu JK, Costain WJ. Emerging technologies for delivery of biotherapeutics and gene therapy across the blood–brain barrier. BioDrugs. 2018;32(6):547–59.

    Article  CAS  Google Scholar 

  13. Johnsen KB, Burkhart A, Thomsen LB, Andresen TL, Moos T. Targeting the transferrin receptor for brain drug delivery. Prog Neurobiol. 2019;181:101665.

    Article  CAS  Google Scholar 

  14. Zhao Z, Zlokovic BV. Therapeutic TVs for crossing barriers in the brain. Cell. 2020;182(2):267–9.

    Article  CAS  Google Scholar 

  15. Boado RJ, Zhou QH, Lu JZ, Hui EKW, Pardridge WM. Pharmacokinetics and brain uptake of a genetically engineered bifunctional fusion antibody targeting the mouse transferrin receptor. Mol Pharm. 2010;7(1):237–44.

    Article  CAS  Google Scholar 

  16. Yu YJ, Zhang Y, Kenrick M, Hoyte K, Luk W, Lu Y, et al. Boosting brain uptake of a therapeutic antibody by reducing its affinity for a transcytosis target. Sci Transl Med. 2011;3(84):1–9.

    Article  Google Scholar 

  17. Niewoehner J, Bohrmann B, Collin L, Urich E, Sade H, Maier P, et al. Increased brain penetration and potency of a therapeutic antibody using a monovalent molecular shuttle. Neuron. 2014;81(1):49–60.

    Article  CAS  Google Scholar 

  18. Webster CI, Hatcher J, Burrell M, Thom G, Thornton P, Gurrell I, et al. Enhanced delivery of IL-1 receptor antagonist to the central nervous system as a novel anti-Transferrin receptor-IL-1RA fusion reverses neuropathic mechanical hypersensitivity. Pain. 2017;158(4):660–8.

    Article  CAS  Google Scholar 

  19. Rofo F, Yilmaz CU, Metzendorf N, Gustavsson T, Beretta C, Erlandsson A, et al. Enhanced neprilysin-mediated degradation of hippocampal Aβ42 with a somatostatin peptide that enters the brain. Theranostics. 2020;11(2):789–804.

    Article  Google Scholar 

  20. Friden PM, Walus LR, Musso GF, Taylor MA, Malfroy B, Starzyk RM. Anti-transferrin receptor antibody and antibody-drug conjugates cross the blood-brain barrier. Proc Natl Acad Sci U S A. 1991;88(11):4771–5.

    Article  CAS  Google Scholar 

  21. Wu D, Pardridge WM. Neuroprotection with noninvasive neurotrophin delivery to the brain. Proc Natl Acad Sci U S A. 1999;96(1):254–9.

    Article  CAS  Google Scholar 

  22. Thom G, Burrell M, Haqqani AS, Yogi A, Lessard E, Brunette E, et al. Enhanced delivery of galanin conjugates to the brain through bioengineering of the anti-transferrin receptor antibody OX26. Mol Pharm. 2018;15(4):1420–31.

    Article  CAS  Google Scholar 

  23. Campos CR, Kemble AM, Niewoehner J, Freskgård PO, Urich E. Brain shuttle neprilysin reduces central amyloid-β levels. PLoS One. 2020;15(3):1–12.

    Article  Google Scholar 

  24. Yu YJ, Atwal JK, Zhang Y, Tong RK, Wildsmith KR, Tan C, et al. Therapeutic bispecific antibodies cross the blood-brain barrier in nonhuman primates. Sci Transl Med. 2014;6(261):1–11.

    Article  Google Scholar 

  25. Sonoda H, Morimoto H, Yoden E, Koshimura Y, Kinoshita M, Golovina G, et al. A blood-brain-barrier-penetrating anti-human transferrin receptor antibody fusion protein for neuronopathic mucopolysaccharidosis II. Mol Ther. 2018;26(5):1366–74.

    Article  CAS  Google Scholar 

  26. Kariolis MS, Wells RC, Getz JA, Kwan W, Mahon CS, Tong R, et al. Brain delivery of therapeutic proteins using an Fc fragment blood-brain barrier transport vehicle in mice and monkeys. Sci Transl Med. 2020;12(545):1–14.

    Article  Google Scholar 

  27. Terstappen GC, Meyer AH, Bell RD, Zhang W. Strategies for delivering therapeutics across the blood–brain barrier. Nat Rev Drug Discov. 2021;20(5):362–83.

    Article  CAS  Google Scholar 

  28. Sehlin D, Fang XT, Cato L, Antoni G, Lannfelt L, Syvänen S. Antibody-based PET imaging of amyloid beta in mouse models of Alzheimer’s disease. Nat Commun. 2016;7:1–11.

    Article  Google Scholar 

  29. Sehlin D, Fang XT, Meier SR, Jansson M, Syvänen S. Pharmacokinetics, biodistribution and brain retention of a bispecific antibody-based PET radioligand for imaging of amyloid-β. Sci Rep. 2017;7(1):1–9.

    Article  CAS  Google Scholar 

  30. Syvänen S, Fang XT, Hultqvist G, Meier SR, Lannfelt L, Sehlin D. A bispecific Tribody PET radioligand for visualization of amyloid-beta protofibrils—a new concept for neuroimaging. NeuroImage. 2017;148:55–63.

    Article  Google Scholar 

  31. Meier SR, Syvänen S, Hultqvist G, Fang XT, Roshanbin S, Lannfelt L, et al. Antibody-based in vivo PET imaging detects amyloid-ß reduction in Alzheimer transgenic mice after BACE-1 inhibition. J Nucl Med. 2018;59(12):1901–6.

    Article  Google Scholar 

  32. Fang XT, Hultqvist G, Meier SR, Antoni G, Sehlin D, Syvänen S. High detection sensitivity with antibody-based PET radioligand for amyloid beta in brain. NeuroImage. 2019;184(October):881–8.

    Article  Google Scholar 

  33. Syvänen S, Fang XT, Faresjö R, Rokka J, Lannfelt L, Olberg DE, et al. Fluorine-18-labeled antibody ligands for PET imaging of amyloid-β in brain. ACS Chem Neurosci. 2020;11(24):4460–8.

    Article  Google Scholar 

  34. Bien-Ly N, Yu YJ, Bumbaca D, Elstrott J, Boswell CA, Zhang Y, et al. Transferrin receptor (TfR) trafficking determines brain uptake of TfR antibody affinity variants. J Exp Med. 2014;211(2):233–44.

    Article  CAS  Google Scholar 

  35. Haqqani AS, Thom G, Burrell M, Delaney CE, Brunette E, Baumann E, et al. Intracellular sorting and transcytosis of the rat transferrin receptor antibody OX26 across the blood–brain barrier in vitro is dependent on its binding affinity. J Neurochem. 2018;146(6):735–52.

    Article  CAS  Google Scholar 

  36. Chang HY, Wu S, Li Y, Zhang W, Burrell M, Webster CI, et al. Brain pharmacokinetics of anti-transferrin receptor antibody affinity variants in rats determined using microdialysis. MAbs. 2021;13(1).

  37. Faresjö R, Bonvicini G, Fang XT, Aguilar X, Sehlin D, Syvänen S. Brain pharmacokinetics of two BBB penetrating bispecific antibodies of different size. Fluids Barriers CNS. 2021;18(1):1–15.

    Article  Google Scholar 

  38. Jefferies WA, Brandon MR, Hunt SV, Williams AF, Gatter KC, Mason DY. Transferrin receptor on endothelium of brain capillaries. Nature. 1984;312(5990):162–3.

    Article  CAS  Google Scholar 

  39. Abushouk AI, Elmaraezy A, Aglan A, Salama R, Fouda S, Fouda R, et al. Bapineuzumab for mild to moderate Alzheimer’s disease: a meta-analysis of randomized controlled trials. BMC Neurol. 2017;17(1):1–13.

    Article  Google Scholar 

  40. Greenwood FC, Hunter WM, Glover JS. The preparation of I-131-labelled human growth hormone of high specific. Biochem J. 1963;89:114–23.

    Article  CAS  Google Scholar 

  41. Cohen RM, Rezai-Zadeh K, Weitz TM, Rentsendorj A, Gate D, Spivak I, et al. A transgenic Alzheimer rat with plaques, tau pathology, behavioral impairment, oligomeric Aβ, and frank neuronal loss. J Neurosci. 2013;33(15):6245–56.

    Article  CAS  Google Scholar 

  42. Chaney AM, Lopez-Picon FR, Serrière S, Wang R, Bochicchio D, Webb SD, et al. Prodromal neuroinflammatory, cholinergic and metabolite dysfunction detected by PET and MRS in the TgF344-AD transgenic rat model of AD: A collaborative multi-modal study. Theranostics. 2021;11(14):6644–67.

    Article  CAS  Google Scholar 

  43. Saré RM, Cooke SK, Krych L, Zerfas PM, Cohen RM, Smith CB. Behavioral phenotype in the TgF344-AD rat model of Alzheimer’s disease. Front Neurosci. 2020;14:601.

    Article  Google Scholar 

  44. Argent NB, Liles J, Rodham D, Clayton CB, Wilkinson R, Baylis PH. A new method for measuring the blood volume of the rat using 113mlndium as a tracer. Lab Anim. 1994;28(2):172–5.

    Article  CAS  Google Scholar 

  45. Loening AM, Gambhir SS. AMIDE: A free software tool for multimodality medical image analysis. Mol Imaging. 2003;2(3):131–7.

    Article  Google Scholar 

  46. Buiter HJC, van Velden FHP, Leysen JE, Fisher A, Windhorst AD, Lammertsma AA, et al. Reproducible analysis of rat brain PET studies using an additional [18F]NaF scan and an MR-based ROI template. Int J Mol Imaging. 2012;2012:58071:1–10.

    Article  Google Scholar 

  47. Gustafsson S, Gustavsson T, Roshanbin S, Hultqvist G, Hammarlund-Udenaes M, Sehlin D, et al. Blood-brain barrier integrity in a mouse model of Alzheimer’s disease with or without acute 3D6 immunotherapy. Neuropharm. 2018;143:1–9.

    Article  CAS  Google Scholar 

  48. Michno W, Nyström S, Wehrli P, Lashley T, Brinkmalm G, Guerard L, et al. Pyroglutamation of amyloid-βx-42 (Aβx-42) followed by Aβ1–40 deposition underlies plaque polymorphism in progressing Alzheimer’s disease pathology. J Biol Chem. 2019;294(17):6719–32.

    Article  CAS  Google Scholar 

  49. Tsukiji S, Nagamune T. Sortase-mediated ligation: a gift from gram-positive bacteria to protein engineering. ChemBioChem. 2009;10(5):787–98.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank Rebecca Faresjö and Martin Larhammar for experimental support. The molecular imaging work in this study was performed at the SciLifeLab Pilot Facility for Preclinical PET-MRI, a Swedish nationally available imaging platform at Uppsala University, Sweden, financed by the Knut and Alice Wallenberg Foundation.

Funding

Open access funding provided by Uppsala University. This project received funding from the European Union’s Horizon 2020 research and innovation programme under the Marie Skłodowska-Curie Grant Agreement No. 813528, the Swedish Research Council (2017-02413, 2018-02715, 2021-1083 and 2021-03524), the Swedish Innovation Agency (2019-00106), Alzheimerfonden, Hjärnfonden, Hedlunds stiftelse, Torsten Söderbergs stiftelse, Åhlenstiftelsen, Stiftelsen för gamla tjänarinnor, Stohnes stiftelse, Magnus Bergvalls stiftelse, Konug Gustaf V:s och Drottning Victorias frimuarestiftelse, Åke Wibergs stiftelse and Turku University Hospital. The funding organizations did not take part in designing the study, in collecting, analysing, or interpreting the data, or in writing the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

GB, DS, KGA and SS contributed to the study conception and design. MHS and FLP provided the TgF344 AD and WT littermate rats. GB performed material preparation. GB, DS and SS performed data collection. GB performed data analysis and wrote the first draft of the manuscript. All authors commented on previous versions of the manuscript and have read and approved the final manuscript.

Corresponding author

Correspondence to Dag Sehlin.

Ethics declarations

Ethics approval and consent to participate

All animal experiments described in this study were approved by the Uppsala County Animal Ethics board (5.8.18–20401/2020), following the rules and regulations of the Swedish Animal Welfare Agency and complied with the European Communities Council Directive of 22 September 2010 (2010/63/EU).

Consent for publication

Not applicable.

Competing interests

GB and KGA are employees of BioArctic AB, Sweden. The other authors declare that they have no competing interests.

Supplementary Information

Additional file 1. Fig S1

. Representative sagittal images of Aβ pathology visualized with RmAb3D6, the murine version of Bapi, in 15-month-old TgF344-AD and WT rats. Fig S2. Ex vivo blood pharmacokinetics of [124I]I-OX265-F(ab′)2-Bapi in TgF344-AD and WT rats

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Bonvicini, G., Syvänen, S., Andersson, K.G. et al. ImmunoPET imaging of amyloid-beta in a rat model of Alzheimer’s disease with a bispecific, brain-penetrating fusion protein. Transl Neurodegener 11, 55 (2022). https://doi.org/10.1186/s40035-022-00324-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s40035-022-00324-y

Keywords