Logo: to the web site of Uppsala University

uu.sePublications from Uppsala University
Change search
Link to record
Permanent link

Direct link
Publications (10 of 89) Show all publications
Kanellopoulos, P., Lundmark, F., Abouzayed, A., Balestri, L. ., Olaniran, E., Obeid, K., . . . Orlova, A. (2025). Synthesis and preclinical evaluation of gastrin releasing peptide receptor antagonist [18F]MeTz-PEG2-RM26 for positron emission tomography. EJNMMI Radiopharmacy and Chemistry, 10(1), Article ID 14.
Open this publication in new window or tab >>Synthesis and preclinical evaluation of gastrin releasing peptide receptor antagonist [18F]MeTz-PEG2-RM26 for positron emission tomography
Show others...
2025 (English)In: EJNMMI Radiopharmacy and Chemistry, E-ISSN 2365-421X, Vol. 10, no 1, article id 14Article in journal (Refereed) Published
Abstract [en]

Background: The gastrin-releasing peptide receptor (GRPR) is overexpressed in the majority of primary prostate cancer lesions, with persistent expression in lymph nodes and bone metastases, making it a legitimate molecular target for diagnostic imaging and staging. This study presents the synthesis and preclinical evaluation of [18F]MeTz-PEG2-RM26, a GRPR antagonist which utilises the Inverse Electron Demand Diels-Alder (IEDDA) reaction for 18F-labelling. This click-chemistry approach allows for site-specific incorporation of fluorine-18 under mild conditions, preserving the peptide's structural integrity and biological activity. Receptor specificity and affinity of [18F]MeTz-PEG2-RM26 were evaluated in vitro using GRPR-expressing PC-3 cells. Furthermore, the biodistribution profile of [18F]MeTz-PEG2-RM26 was assessed in NMRI mice and its tumour-targeting capability was investigated in mice bearing PC-3 xenografts.

Results: The labelling of TCO-PEG2-RM26 precursor involved three steps: (1) synthesis of an 18F-labelled activated ester on a quaternary methyl ammonium (QMA) cartridge, (2) conjugation of the labelled ester to a tetrazine amine, and (3) attachment to TCO-PEG2-RM26 via an IEDDA click reaction. This production method of [18F]MeTz-PEG2-RM26 afforded a high apparent molar activity of 3.5-4.3 GBq/mu mol and radiochemical purity exceeding 98%, with 43-70 MBq activity incorporation, while the entire synthesis was completed within 75 min. Both in vitro and in vivo studies confirmed the specific binding of [18F]MeTz-PEG2-RM26 to GRPR, with a significant reduction in activity uptake observed upon receptor saturation. The radioligand exhibited rapid blood clearance and minimal bone uptake, confirming the stability of the fluorine-carbon bond. However, high hepatic uptake (12-13% IA/g at 1 h post-injection) indicated predominant hepatobiliary excretion. Receptor-mediated uptake was observed in the tumours and pancreatic tissue, although the overall activity uptake in tumours was low, likely due to the rapid hepatobiliary clearance of [18F]MeTz-PEG2-RM26.

Conclusions:  These findings demonstrate the effectiveness of the IEDDA click reaction for fluorine-18 labelling of GRPR-targeting PET tracers. Future studies should focus on increasing the hydrophilicity of the imaging probe to improve the targeting properties and biodistribution profile of the radioligand.

Place, publisher, year, edition, pages
Springer Nature, 2025
Keywords
Fluorine-18, TCO, Tetrazine, IEDDA click chemistry, GRPR, Bombesin, PET, Prostate cancer
National Category
Radiology and Medical Imaging Medicinal Chemistry Cancer and Oncology Pharmaceutical Sciences
Identifiers
urn:nbn:se:uu:diva-553823 (URN)10.1186/s41181-025-00336-9 (DOI)001453646400001 ()40138074 (PubMedID)2-s2.0-105000840760 (Scopus ID)
Available from: 2025-04-07 Created: 2025-04-07 Last updated: 2025-04-07Bibliographically approved
Pagnon de la Vega, M., Syvänen, S., Giedraitis, V., Hooley, M., Konstantinidis, E., Meier, S. R., . . . Sehlin, D. (2024). Altered amyloid-β structure markedly reduces gliosis in the brain of mice harboring the Uppsala APP deletion. Acta neuropathologica communications, 12(1), Article ID 22.
Open this publication in new window or tab >>Altered amyloid-β structure markedly reduces gliosis in the brain of mice harboring the Uppsala APP deletion
Show others...
2024 (English)In: Acta neuropathologica communications, E-ISSN 2051-5960, Vol. 12, no 1, article id 22Article in journal (Refereed) Published
Abstract [en]

Deposition of amyloid beta (Aβ) into plaques is a major hallmark of Alzheimer’s disease (AD). Different amyloid precursor protein (APP) mutations cause early-onset AD by altering the production or aggregation properties of Aβ. We recently identified the Uppsala APP mutation (APPUpp), which causes Aβ pathology by a triple mechanism: increased β-secretase and altered α-secretase APP cleavage, leading to increased formation of a unique Aβ conformer that rapidly aggregates and deposits in the brain. The aim of this study was to further explore the effects of APPUpp in a transgenic mouse model (tg-UppSwe), expressing human APP with the APPUpp mutation together with the APPSwe mutation. Aβ pathology was studied in tg-UppSwe brains at different ages, using ELISA and immunohistochemistry. In vivo PET imaging with three different PET radioligands was conducted in aged tg-UppSwe mice and two other mouse models; tg-ArcSwe and tg-Swe. Finally, glial responses to Aβ pathology were studied in cell culture models and mouse brain tissue, using ELISA and immunohistochemistry. Tg-UppSwe mice displayed increased β-secretase cleavage and suppressed α-secretase cleavage, resulting in AβUpp42 dominated diffuse plaque pathology appearing from the age of 5–6 months. The γ-secretase cleavage was not affected. Contrary to tg-ArcSwe and tg-Swe mice, tg-UppSwe mice were [11C]PiB-PET negative. Antibody-based PET with the 3D6 ligand visualized Aβ pathology in all models, whereas the Aβ protofibril selective mAb158 ligand did not give any signals in tg-UppSwe mice. Moreover, unlike the other two models, tg-UppSwe mice displayed a very faint glial response to the Aβ pathology. The tg-UppSwe mouse model thus recapitulates several pathological features of the Uppsala APP mutation carriers. The presumed unique structural features of AβUpp42 aggregates were found to affect their interaction with anti-Aβ antibodies and profoundly modify the Aβ-mediated glial response, which may be important aspects to consider for further development of AD therapies.

Place, publisher, year, edition, pages
BioMed Central (BMC), 2024
Keywords
Alzheimer's disease (AD), Amyloid precursor protein (APP), Amyloid-beta (A beta), PET imaging, Microglia, Astrocytes, Immunotherapy
National Category
Neurosciences Neurology
Identifiers
urn:nbn:se:uu:diva-523728 (URN)10.1186/s40478-024-01734-x (DOI)001158145500001 ()38317196 (PubMedID)
Funder
Knut and Alice Wallenberg FoundationUppsala UniversitySwedish Research Council, 2016‑02120Swedish Research Council, 2021‑01083Swedish Research Council, 2021‑03524AlzheimerfondenThe Swedish Brain FoundationTorsten Söderbergs stiftelseÅhlén-stiftelsenMagnus Bergvall FoundationStiftelsen Gamla TjänarinnorGun och Bertil Stohnes StiftelseKonung Gustaf V:s och Drottning Victorias FrimurarestiftelseStiftelsen Sigurd och Elsa Goljes minne
Note

De två sista författarna delar sistaförfattarskapet

Available from: 2024-02-26 Created: 2024-02-26 Last updated: 2024-02-26Bibliographically approved
Xiong, M., Dahlén, A., Roshanbin, S., Wik, E., Aguilar, X., Eriksson, J., . . . Syvänen, S. (2024). Antibody engagement with amyloid‐beta does not inhibit [11C]PiB binding for PET imaging. Journal of Neurochemistry, 168(9), 2601-2610
Open this publication in new window or tab >>Antibody engagement with amyloid‐beta does not inhibit [11C]PiB binding for PET imaging
Show others...
2024 (English)In: Journal of Neurochemistry, ISSN 0022-3042, E-ISSN 1471-4159, Vol. 168, no 9, p. 2601-2610Article in journal (Refereed) Published
Abstract [en]

The elimination of amyloid-beta (Aβ) plaques in Alzheimer's disease patients after treatment with anti-Aβ antibodies such as lecanemab and aducanumab is supported by a substantially decreased signal in amyloid positron emission tomography (PET) imaging. However, this decreased PET signal has not been matched by a similar substantial effect on cognitive function. There may be several reasons for this, including short treatment duration and advanced disease stages among the patients. However, one aspect that has not been investigated, and the subject of this study, is whether antibody engagement with amyloid plaques inhibits the binding of amyloid-PET ligands, leading to a false impression of Aβ removal from the brain. In the present study, tg-ArcSwe mice received three injections of RmAb158, the murine version of lecanemab or phosphate-buffered saline (PBS) before the administration of the amyloid-PET radioligand [11C]PiB, followed by isolation of brain tissue. Autoradiography showed that RmAb158- and PBS-treated mice displayed similar [11C]PiB binding. Moreover, the total Aβ1–40 levels, representing the major Aβ species of plaques in the tg-ArcSwe model, as well as soluble triggering receptor on myeloid cells 2 (sTREM2) levels, were similar in both groups. Interestingly, the concentration of soluble Aβ aggregates was decreased in the RmAb158-treated group, along with a small but significant decrease in the total Aβ1–42 levels. In conclusion, this study indicates that the binding of [11C]PiB to Aβ accurately mirrors the load of Aβ plaques in the brain, aligning with how amyloid-PET is interpreted in clinical studies of anti-Aβ antibodies. However, early treatment effects on soluble Aβ aggregates and Aβ1–42 levels were not detected. 

Place, publisher, year, edition, pages
John Wiley & Sons, 2024
National Category
Neurosciences
Identifiers
urn:nbn:se:uu:diva-546137 (URN)10.1111/jnc.16127 (DOI)001216124100001 ()
Funder
Gun och Bertil Stohnes StiftelseKonung Gustaf V:s och Drottning Victorias FrimurarestiftelseStiftelsen Gamla TjänarinnorMagnus Bergvall FoundationÅhlén-stiftelsenThe Swedish Brain FoundationAlzheimerfondenSwedish Research Council, 2021‐01083Swedish Research Council, 2021‐03524
Available from: 2025-01-07 Created: 2025-01-07 Last updated: 2025-01-16Bibliographically approved
Wegrzyniak, O., Lechi, F., Mitran, B., Cheung, P., Bitzios, A., Persson, J., . . . Eriksson, O. (2024). Non-invasive PET imaging of liver fibrogenesis using a RESCA-conjugated Affibody molecule. iScience, 27(5), Article ID 109688.
Open this publication in new window or tab >>Non-invasive PET imaging of liver fibrogenesis using a RESCA-conjugated Affibody molecule
Show others...
2024 (English)In: iScience, E-ISSN 2589-0042, Vol. 27, no 5, article id 109688Article in journal (Refereed) Published
Abstract [en]

Non-invasive assessment of fibrogenic activity, rather than fibrotic scars, could significantly improve the management of fibrotic diseases and the development of anti-fibrotic drugs. This study explores the potential of an Affibody molecule (Z09591) labeled with the Al(18)F-restrained complexing agent (RESCA) method as a tracer for the non-invasive detection of fibrogenic cells. Z09591 was functionalized with the RESCA chelator for direct labeling with [18F]AlF. 18 F]AlF. In vivo positron emission tomography/magnetic resonance imaging scans on U-87 tumor-bearing mice exhibited high selectivity of the resulting radiotracer, [18F]AlF-RESCA-Z09591, 18 F]AlF-RESCA-Z09591, for platelet-derived growth factor receptor b (PDGFRb), b ), with minimal non-specific background uptake. Evaluation in a mouse model with carbon tetrachloride-induced fibrotic liver followed by a disease regression phase, revealed the radiotracer's high affinity and specificity for fibrogenic cells in fibrotic livers (standardized uptake value [SUV] 0.43 +/- 0.05), with uptake decreasing during recovery (SUV 0.29 +/- 0.03) (p p < 0.0001). [18F]AlF-RESCA-Z09591 18 F]AlF-RESCA-Z09591 accurately detects PDGFRb, b, offering noninvasive assessment of fibrogenic cells and promising applications in precise liver fibrogenesis diagnosis, potentially contributing significantly to anti-fibrotic drug development.

Place, publisher, year, edition, pages
Elsevier, 2024
National Category
Radiology, Nuclear Medicine and Medical Imaging
Identifiers
urn:nbn:se:uu:diva-537082 (URN)10.1016/j.isci.2024.109688 (DOI)001288013000001 ()38660405 (PubMedID)
Funder
Swedish Research Council, 2020-02312Swedish Child Diabetes FoundationDiabetesfonden
Available from: 2024-08-29 Created: 2024-08-29 Last updated: 2025-06-23Bibliographically approved
Lechi, F., Eriksson, J., Odell, L. R., Wegrzyniak, O., Lofblom, J., Frejd, F., . . . Eriksson, O. (2024). Optimized method for fluorine-18 radiolabeling of Affibody molecules using RESCA. EJNMMI Radiopharmacy and Chemistry, 9(1), Article ID 73.
Open this publication in new window or tab >>Optimized method for fluorine-18 radiolabeling of Affibody molecules using RESCA
Show others...
2024 (English)In: EJNMMI Radiopharmacy and Chemistry, E-ISSN 2365-421X, Vol. 9, no 1, article id 73Article in journal (Refereed) Published
Abstract [en]

Background: In recent years, the interest in Al[18F]F as a labeling agent for Positron Emission Tomography (PET) radiotracers has risen, as it allows for fast and efficient fluorine-18 labeling by harnessing chelation chemistry. The introduction of Restrained Complexing Agent (RESCA) as a chelator has also shown that chelator-based radiolabeling reactions can be performed in mild conditions, making the radiolabeling process attractively more facile than most conventional radiofluorination methods. The aim of the study was to establish optimized conditions for Al[18F]F labeling of Affibody molecules using RESCA as a complexing agent, using Z09591 and Z0185, two Affibody proteins targeting PDGFR beta and TNF alpha, respectively, as model compounds.

Results: The Al[18F]F labeling of RESCA-conjugated Z09591 was tested at different temperatures (rt to 60 degrees C) and with varying reaction times (12 to 60 min), and optimal conditions were then implemented on RESCA-Z0185. The optimized synthesis method was: 1.5-2.5 GBq of cyclotron produced fluorine-18 were trapped on a QMA cartridge and eluted with saline solution to react with 12 nmol of AlCl3 and form Al[18F]F. The respective RESCA-conjugated Affibody molecule (14 nmol) in NaOAc solution was added to the Al[18F]F solution and left to react at 60 degrees C for 12 min. The mixture was purified on a NAP5 size exclusion column and then analyzed by HPLC. The entire process took approximately 35 min, was highly reproducible, indicating the efficiency and reliability of the method. The labeled compounds demonstrated retained biological function for their respective targets after purification.

Conclusions: We present a general and optimized method for Al[18F]F labeling of RESCA-conjugated Affibody molecules, which can be widely applied to this class of peptide-based imaging agents. Moreover, radiochemical yields were improved when the labeling was conducted at 37 degrees C or above. In vitro and in vivo assessment of the respective tracers was promising, showing retained binding capacity as well as moderate defluorination, which is usually regarded as a potential downside for RESCA-conjugated tracers.

Place, publisher, year, edition, pages
Springer Nature, 2024
Keywords
RESCA, Radiochemistry, Aluminium fluoride, PET imaging, Affibody molecule, Peptide
National Category
Radiology, Nuclear Medicine and Medical Imaging
Identifiers
urn:nbn:se:uu:diva-542252 (URN)10.1186/s41181-024-00304-9 (DOI)001342165900001 ()39460878 (PubMedID)
Available from: 2024-11-27 Created: 2024-11-27 Last updated: 2025-06-23Bibliographically approved
Faresjö, R., Sjöström, E. O., Dallas, T., Berglund, M. M., Eriksson, J., Sehlin, D. & Syvänen, S. (2024). Single domain antibody-scFv conjugate targeting amyloid β and TfR penetrates the blood–brain barrier and interacts with amyloid β. mAbs, 16(1), Article ID 2410968.
Open this publication in new window or tab >>Single domain antibody-scFv conjugate targeting amyloid β and TfR penetrates the blood–brain barrier and interacts with amyloid β
Show others...
2024 (English)In: mAbs, ISSN 1942-0862, E-ISSN 1942-0870, Vol. 16, no 1, article id 2410968Article in journal (Refereed) Published
Abstract [en]

Neurodegenerative diseases such as Alzheimer’s disease (AD) pose substantial challenges to patients and health-care systems, particularly in countries with aging populations. Immunotherapies, including the marketed antibodies lecanemab (Leqembi®) and donanemab (KisunlaTM), offer promise but face hurdles due to limited delivery across the blood–brain barrier (BBB). This limitation necessitates high doses, resulting in increased costs and a higher risk of side effects. This study explores transferrin receptor (TfR)-binding camelid single-domain antibodies (VHHs) for facilitated brain delivery. We developed and evaluated fusion proteins (FPs) combining VHHs with human IgG Fc domains or single-chain variable fragments (scFvs) of the anti-amyloid-beta (Aβ) antibody 3D6. In vitro assessments showed varying affinities of the FPs for TfR. In vivo evaluations indicated that specific VHH-Fc and VHH-scFv fusions reached significant brain concentrations, emphasizing the importance of optimal TfR binding affinities. The VHH-scFv fusions were further investigated in mouse models with Aβ pathology, showing higher retention compared to wild-type mice without Aβ pathology. Our findings suggest that these novel VHH-based FPs hold potential for therapeutic and diagnostic applications in AD, providing a strategy to overcome BBB limitations and enhance brain targeting of antibody-based treatments. Furthermore, our results suggest that a given bispecific TfR-binding fusion format has a window of “optimal” affinity where parenchymal delivery is adequate, while blood pharmacokinetics aligns with the desired application of the fusion protein.

Place, publisher, year, edition, pages
Taylor & Francis, 2024
Keywords
Blood-brain barrier, brain delivery, camelid antibody, fusion protein, transferrin receptor, VHH
National Category
Neurosciences
Identifiers
urn:nbn:se:uu:diva-540646 (URN)10.1080/19420862.2024.2410968 (DOI)001326899800001 ()39358860 (PubMedID)
Funder
Swedish Research Council, 2021- 01083Swedish Research Council, 2021-03524AlzheimerfondenHans-Gabriel och Alice Trolle-Wachtmeisters stiftelse för medicinsk forskningVinnovaThe Swedish Brain FoundationTorsten Söderbergs stiftelseÅhlén-stiftelsenStiftelsen Gamla TjänarinnorGun och Bertil Stohnes StiftelseMagnus Bergvall FoundationKonung Gustaf V:s och Drottning Victorias FrimurarestiftelseÅke Wiberg Foundation
Available from: 2024-10-22 Created: 2024-10-22 Last updated: 2024-10-22Bibliographically approved
Schlein, E., Rokka, J., Odell, L. R., Lopes van den Broek, S., Herth, M. M., Battisti, U. M., . . . Eriksson, J. (2024). Synthesis and evaluation of fluorine-18 labelled tetrazines as pre-targeting imaging agents for PET. EJNMMI Radiopharmacy and Chemistry, 9(1), Article ID 21.
Open this publication in new window or tab >>Synthesis and evaluation of fluorine-18 labelled tetrazines as pre-targeting imaging agents for PET
Show others...
2024 (English)In: EJNMMI Radiopharmacy and Chemistry, E-ISSN 2365-421X, Vol. 9, no 1, article id 21Article in journal (Refereed) Published
Abstract [en]

The brain is a challenging target for antibody-based positron emission tomography (immunoPET) imaging due to the restricted access of antibody-based ligands through the blood-brain barrier (BBB). To overcome this challenge we have previously developed bispecific antibody ligands that pass through the BBB via receptor-mediated transcytosis. These ligands, when radiolabelled, can be used for brain imaging with high affinity and specificity, but their long residence time in the blood and brain can be challenging for their use as PET radioligands. This could be solved by using a two-step approach which involves the administration of a tagged antibody that accumulates at the target site in the brain and then clears from the blood, followed by administration of a radiolabelled molecule, with fast kinetics. This radiolabelled molecule can couple to the tagged antibody and thereby make the antibody localisation visible by PET imaging. The in vivo linkage can be achieved using the inverse electron demand Diels-Alder reaction (IEDDA), with trans-cyclooctene (TCO) and tetrazine groups participating as reactants.

In this study, two 18F-labelled tetrazines were synthesized and evaluated for their potential as agents for pre-targeted imaging, i.e. for their ability to rapidly enter the brain and then, if non-bound, be sufficiently cleared with low background retention. The two compounds, a methyl tetrazine [18F]MeTz and an H-tetrazine [18F]HTz were radiolabelled using a two-step procedure via [18F]F-Py-TFP synthesized on solid support followed by amidation with amine-bearing tetrazines, resulting in radiochemical yields of 24% and 22%, respectively, and a radiochemical purity of > 96%. In vivo PET imaging was performed to assess their suitability for in vivo pre-targeting. Time-activity curves from PET-scans revealed that the [18F]MeTz had the most favourable profile for an imaging agent for pre-targeting, due to its fast and homogenous brain distribution and rapid clearance from the brain.

Place, publisher, year, edition, pages
Springer Nature, 2024
Keywords
Inverse electron demand Diels−Alder reaction IEDDA, pre-targeting, tetrazine, Alzheimer’s disease, bio-orthogonal, Fluorine-18
National Category
Radiology, Nuclear Medicine and Medical Imaging
Identifiers
urn:nbn:se:uu:diva-499057 (URN)10.1186/s41181-024-00250-6 (DOI)001176805900001 ()38446356 (PubMedID)
Funder
Swedish Research CouncilAlzheimerfondenTorsten Söderbergs stiftelseThe Swedish Brain FoundationHedlund foundationÅhlén-stiftelsenKonung Gustaf V:s och Drottning Victorias FrimurarestiftelseMagnus Bergvall FoundationGun och Bertil Stohnes StiftelseStiftelsen Gamla TjänarinnorEU, Horizon 2020, 813528
Available from: 2023-03-22 Created: 2023-03-22 Last updated: 2024-04-02Bibliographically approved
Cheung, P., Amin, M. A., Zhang, B., Lechi, F., Korsgren, O., Eriksson, J., . . . Eriksson, O. (2023). [18F]MK-7246 for Positron Emission Tomography Imaging of the Beta-Cell Surface Marker GPR44. Pharmaceutics, 15(2), Article ID 499.
Open this publication in new window or tab >>[18F]MK-7246 for Positron Emission Tomography Imaging of the Beta-Cell Surface Marker GPR44
Show others...
2023 (English)In: Pharmaceutics, E-ISSN 1999-4923, Vol. 15, no 2, article id 499Article in journal (Refereed) Published
Abstract [en]

The progressive loss of beta-cell mass is a hallmark of diabetes and has been suggested as a complementary approach to studying the progression of diabetes in contrast to the beta-cell function. Non-invasive nuclear medicinal imaging techniques such as Positron Emission Tomography using radiation emitting tracers have thus been suggested as more viable methodologies to visualize and quantify the beta-cell mass with sufficient sensitivity. The transmembrane G protein-coupled receptor GPR44 has been identified as a biomarker for monitoring beta-cell mass. MK-7246 is a GPR44 antagonist that selectively binds to GPR44 with high affinity and good pharmacokinetic properties. Here, we present the synthesis of MK-7246, radiolabeled with the positron emitter fluorine-18 for preclinical evaluation using cell lines, mice, rats and human pancreatic cells. Here, we have described a synthesis and radiolabeling method for producing [18F]MK-7246 and its precursor compound. Preclinical assessments demonstrated the strong affinity and selectivity of [18F]MK-7246 towards GPR44. Additionally, [18F]MK-7246 exhibited excellent metabolic stability, a fast clearance profile from blood and tissues, qualifying it as a promising radioactive probe for GPR44-directed PET imaging.

Place, publisher, year, edition, pages
MDPI, 2023
Keywords
18F-labelling, CRTH2, GPR44, PET, beta-cell mass, diabetes, islet imaging
National Category
Endocrinology and Diabetes Medicinal Chemistry
Identifiers
urn:nbn:se:uu:diva-497911 (URN)10.3390/pharmaceutics15020499 (DOI)000940959200001 ()36839820 (PubMedID)
Funder
Insamlingsstiftelsen Diabetes Wellness, 2409Swedish Child Diabetes FoundationDiabetesfondenEXODIAB - Excellence of Diabetes Research in SwedenScience for Life Laboratory, SciLifeLabSwedish Research Council, 2020-0231Swedish Research Council, 2019-0145Novo NordiskErnfors FoundationGöran Gustafsson Foundation for promotion of scientific research at Uppala University and Royal Institute of Technology
Note

De tre första författarna delar förstaförfattarskapet

De tre sista författarna delar sistaförfattarskapet

Available from: 2023-03-05 Created: 2023-03-05 Last updated: 2024-07-04Bibliographically approved
Wegrzyniak, O., Zhang, B., Rokka, J., Rosestedt, M., Mitran, B., Cheung, P., . . . Eriksson, O. (2023). Imaging of fibrogenesis in the liver by [18F]TZ-Z09591, an Affibody molecule targeting platelet derived growth factor receptor β. EJNMMI Radiopharmacy and Chemistry, 8, Article ID 23.
Open this publication in new window or tab >>Imaging of fibrogenesis in the liver by [18F]TZ-Z09591, an Affibody molecule targeting platelet derived growth factor receptor β
Show others...
2023 (English)In: EJNMMI Radiopharmacy and Chemistry, E-ISSN 2365-421X, Vol. 8, article id 23Article in journal (Refereed) Published
Abstract [en]

Background: Platelet-derived growth factor receptor beta (PDGFR beta) is a receptor overexpressed on activated hepatic stellate cells (aHSCs). Positron emission tomography (PET) imaging of PDGFR beta could potentially allow the quantification of fibrogenesis in fibrotic livers. This study aims to evaluate a fluorine-18 radiolabeled Affibody molecule ([F-18]TZ-Z09591) as a PET tracer for imaging liver fibrogenesis.

Results: In vitro specificity studies demonstrated that the trans-Cyclooctenes (TCO) conjugated Z09591 Affibody molecule had a picomolar affinity for human PDGFR beta. Biodistribution performed on healthy rats showed rapid clearance of [F-18]TZ-Z09591 through the kidneys and low liver background uptake. Autoradiography (ARG) studies on fibrotic livers from mice or humans correlated with histopathology results. Ex vivo biodistribution and ARG revealed that [F-18]TZ-Z09591 binding in the liver was increased in fibrotic livers (p = 0.02) and corresponded to binding in fibrotic scars.

Conclusions: Our study highlights [F-18]TZ-Z09591 as a specific tracer for fibrogenic cells in the fibrotic liver, thus offering the potential to assess fibrogenesis clearly.

Place, publisher, year, edition, pages
Springer NatureSpringer Nature, 2023
Keywords
PET imaging, Platelet derived growth factor receptor, Hepatic stellate cells, Fibrogenesis, Liver fibrosis
National Category
Radiology, Nuclear Medicine and Medical Imaging
Identifiers
urn:nbn:se:uu:diva-514754 (URN)10.1186/s41181-023-00210-6 (DOI)001075551000001 ()37733133 (PubMedID)
Funder
Magnus Bergvall FoundationEuropean Foundation for the Study of DiabetesSwedish Research Council, 2020-02312Swedish Child Diabetes Foundation
Available from: 2023-10-26 Created: 2023-10-26 Last updated: 2024-12-03Bibliographically approved
Cheung, P., Thorngren, J., Zhang, B., Vasylovska, S., Lechi, F., Persson, J., . . . Eriksson, O. (2023). Preclinical evaluation of Affibody molecule for PET imaging of human pancreatic islets derived from stem cells.. EJNMMI Research, 13(1), 107, Article ID 107.
Open this publication in new window or tab >>Preclinical evaluation of Affibody molecule for PET imaging of human pancreatic islets derived from stem cells.
Show others...
2023 (English)In: EJNMMI Research, E-ISSN 2191-219X, EJNMMI research, ISSN 2191-219X, Vol. 13, no 1, p. 107-, article id 107Article in journal (Refereed) Published
Abstract [en]

BackgroundBeta-cell replacement methods such as transplantation of isolated donor islets have been proposed as a curative treatment of type 1 diabetes, but widespread application is challenging due to shortages of donor tissue and the need for continuous immunosuppressive treatments. Stem-cell-derived islets have been suggested as an alternative source of beta cells, but face transplantation protocols optimization difficulties, mainly due to a lack of available methods and markers to directly monitor grafts survival, as well as their localization and function. Molecular imaging techniques and particularly positron emission tomography has been suggested as a tool for monitoring the fate of islets after clinical transplantation. The integral membrane protein DGCR2 has been demonstrated to be a potential pancreatic islet biomarker, with specific expression on insulin-positive human embryonic stem-cell-derived pancreatic progenitor cells. The candidate Affibody molecule ZDGCR2:AM106 was radiolabeled with fluorine-18 using a novel click chemistry-based approach. The resulting positron emission tomography tracer [18F]ZDGCR2:AM106 was evaluated for binding to recombinant human DGCR2 and cryosections of stem-cell-derived islets, as well as in vivo using an immune-deficient mouse model transplanted with stem-cell-derived islets. Biodistribution of the [18F]ZDGCR2:AM106 was also assessed in healthy rats and pigs.

Results[18F]ZDGCR2:AM106 was successfully synthesized with high radiochemical purity and yield via a pretargeting approach. [18F]ZDGCR2:AM106 retained binding to recombinant human DCGR2 as well as to cryosectioned stem-cell-derived islets, but in vivo binding to native pancreatic tissue in both rat and pig was low. However, in vivo uptake of [18F]ZDGCR2:AM106 in stem-cell-derived islets transplanted in the immunodeficient mice was observed, albeit only within the early imaging frames after injection of the radiotracer.

ConclusionTargeting of DGCR2 is a promising approach for in vivo detection of stem-cell-derived islets grafts by molecular imaging. The synthesis of [18F]ZDGCR2:AM106 was successfully performed via a pretargeting method to label a site-specific covalently bonded fluorine-18 to the Affibody molecule. However, the rapid washout of [18F]ZDGCR2:AM106 from the stem-cell-derived islets graft indicates that dissociation kinetics can be improved. Further studies using alternative binders of similar classes with improved binding potential are warranted.

Place, publisher, year, edition, pages
Springer, 2023
Keywords
Affibody molecule, DGCR2, Diabetes, Fluorine-18 chemistry, PET, Stem cells
National Category
Endocrinology and Diabetes
Identifiers
urn:nbn:se:uu:diva-518407 (URN)10.1186/s13550-023-01057-3 (DOI)001125115500001 ()38100042 (PubMedID)
Available from: 2023-12-19 Created: 2023-12-19 Last updated: 2024-03-20Bibliographically approved
Organisations
Identifiers
ORCID iD: ORCID iD iconorcid.org/0000-0003-0241-092X

Search in DiVA

Show all publications