1
|
Sehlin D, Roshanbin S, Zachrisson O, Ingelsson M, Syvänen S. A brain-penetrant bispecific antibody lowers oligomeric alpha-synuclein and activates microglia in a mouse model of alpha-synuclein pathology. Neurotherapeutics 2025; 22:e00510. [PMID: 39676023 PMCID: PMC12014296 DOI: 10.1016/j.neurot.2024.e00510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 12/04/2024] [Accepted: 12/04/2024] [Indexed: 12/17/2024] Open
Abstract
Parkinson's disease (PD) is characterized by a progressive loss of dopaminergic neurons, linked to aggregation of alpha-synuclein (αSYN) into Lewy bodies. Current treatments are symptomatic and do not halt or reverse the neurodegeneration. Immunotherapy targeting aggregated αSYN shows potential, but therapeutic efficacy is limited by poor brain penetration of antibodies. We developed a bispecific antibody, RmAb38E2-scFv8D3, based on αSYN oligomer selective RmAb38E2 fused to a transferrin receptor (TfR)-binding domain to enhance brain delivery. Both RmAb38E2 and RmAb38E2-scFv8D3 showed higher affinity for αSYN oligomers than for monomers or fibrils. In vivo, RmAb38E2-scFv8D3 exhibited higher brain and lower blood concentrations compared to RmAb38E2, suggesting a better brain uptake and reduced peripheral exposure for the bispecific antibody. Treatment over five days of 3-4 months old transgenic L61 mice, which overexpress human αSYN, with three doses of RmAb38E2-scFv8D3 reduced brain αSYN oligomer levels and increased microglial activation, as indicated by elevated soluble TREM2 levels. Treatment with the monospecific RmAb38E2, however, showed no significant effect compared to PBS. This study demonstrates that TfR-mediated delivery enhances the therapeutic potential of αSYN-targeted immunotherapy by resulting in a higher concentration and a more uniform distribution of antibodies in the brain. The use of bispecific antibodies offers a promising strategy to improve the efficacy of antibody therapies in PD and other α-synucleinopathies.
Collapse
Affiliation(s)
- Dag Sehlin
- Department of Public Health and Caring Sciences, Uppsala University, 751 85, Uppsala, Sweden
| | - Sahar Roshanbin
- Department of Public Health and Caring Sciences, Uppsala University, 751 85, Uppsala, Sweden
| | | | - Martin Ingelsson
- Department of Public Health and Caring Sciences, Uppsala University, 751 85, Uppsala, Sweden; Krembil Brain Institute, University Health Network, Toronto, Ontario, Canada; Tanz Centre for Research in Neurodegenerative Diseases, Departments of Medicine and Laboratory Medicine & Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Stina Syvänen
- Department of Public Health and Caring Sciences, Uppsala University, 751 85, Uppsala, Sweden.
| |
Collapse
|
2
|
Khoury N, Pizzo ME, Discenza CB, Joy D, Tatarakis D, Todorov MI, Negwer M, Ha C, De Melo GL, Sarrafha L, Simon MJ, Chan D, Chau R, Chew KS, Chow J, Clemens A, Robles-Colmenares Y, Dugas JC, Duque J, Kaltenecker D, Kane H, Leung A, Lozano E, Moshkforoush A, Roche E, Sandmann T, Tong M, Xa K, Zhou Y, Lewcock JW, Ertürk A, Thorne RG, Calvert MEK, Yu Zuchero YJ. Fc-engineered large molecules targeting blood-brain barrier transferrin receptor and CD98hc have distinct central nervous system and peripheral biodistribution. Nat Commun 2025; 16:1822. [PMID: 39979268 PMCID: PMC11842567 DOI: 10.1038/s41467-025-57108-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 02/10/2025] [Indexed: 02/22/2025] Open
Abstract
Blood brain barrier-crossing molecules targeting transferrin receptor (TfR) and CD98 heavy chain (CD98hc) are widely reported to promote enhanced brain delivery of therapeutics. Here, we provide a comprehensive and unbiased biodistribution characterization of TfR and CD98hc antibody transport vehicles (ATVTfR and ATVCD98hc) compared to control IgG. Mouse whole-body tissue clearing reveals distinct organ localization for each molecule. In the brain, ATVTfR and ATVCD98hc achieve enhanced exposure and parenchymal distribution even when brain exposures are matched between ATV and control IgG in bulk tissue. Using a combination of cell sorting and single-cell RNAseq, we reveal that control IgG is nearly absent from parenchymal cells and is distributed primarily to brain perivascular and leptomeningeal cells. In contrast, ATVTfR and ATVCD98hc exhibit broad and unique parenchymal cell-type distribution. Finally, we profile in detail brain region-specific biodistribution of ATVTfR in cynomolgus monkey brain and spinal cord. Taken together, this in-depth multiscale characterization will guide platform selection for therapeutic targets of interest.
Collapse
Affiliation(s)
- Nathalie Khoury
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Michelle E Pizzo
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Claire B Discenza
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - David Joy
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - David Tatarakis
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | | | | | - Connie Ha
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Gabrielly L De Melo
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Lily Sarrafha
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Matthew J Simon
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Darren Chan
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Roni Chau
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Kylie S Chew
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Johann Chow
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Allisa Clemens
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | | | - Jason C Dugas
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Joseph Duque
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | | | - Holly Kane
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Amy Leung
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Edwin Lozano
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Arash Moshkforoush
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Elysia Roche
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Thomas Sandmann
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Mabel Tong
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Kaitlin Xa
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Yinhan Zhou
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Joseph W Lewcock
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | | | - Robert G Thorne
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Meredith E K Calvert
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA.
| | - Y Joy Yu Zuchero
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA.
| |
Collapse
|
3
|
Wuensche TE, Pereira PM, Windhorst AD, Bjerregaard-Andersen K, Sotty F, Kallunki P, Jensen A, Bang-Andersen B, van Dongen GAMS, Beaino W, Vugts DJ. New prospects for 89Zr-immuno-PET in brain applications - Alpha-synucleinopathies. Nucl Med Biol 2025; 140-141:108969. [PMID: 39591759 DOI: 10.1016/j.nucmedbio.2024.108969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Revised: 10/24/2024] [Accepted: 11/06/2024] [Indexed: 11/28/2024]
Abstract
BACKGROUND Recently, 89Zr-immuno-PET imaging of therapeutic antibodies, actively transported over the blood-brain-barrier via transferrin-mediated transcytosis, was demonstrated using the chelator DFO*. In these studies, aducanumab targeting amyloid-beta was fused with a transferrin binding unit: a single-chain Fab fragment derived from 8D3 (scFab8D3). Alpha-synuclein is a hallmark protein of several neurodegenerative diseases such as Parkinson's Disease, Lewy-Body-Dementia, and Multiple System Atrophy. 89Zr-immuno-PET imaging of alpha-synuclein can be a valuable tool for image-guided drug development and assessment of target engagement. The goal of this study was to compare two currently used constructs of 8D3 for targeting potential, namely a single moiety of scFab8D3 fused to the alpha-synuclein antibody HLu-3 (HLu-3-scFab8D3) versus HLu-3 fused with two 8D3 single-chain variable fragments (HLu-3-(scFv8D3)2), by 89Zr-immuno-PET in an alpha-synuclein pre-formed fibril (PFF) deposition model. HLu-3 and the HIV-targeting B12-scFab8D3 were used as controls. The best-performing compound was further investigated in an animal model with predominantly intraneural target aggregation. METHODS Antibodies were conjugated with DFO* using DFO*-NCS and subsequently radiolabeled with 89Zr. Assessment of binding affinity was done by alpha-synuclein ELISA and with FACS analysis using mTfR1 expressing CHO-S cells. Radioimmunoconjugates were first evaluated in an extracellular alpha-synuclein deposition model established by intracranial injection of non-sonicated PFFs into the left striatum of C57Bl/6 WT mice, whereas saline was injected into the contralateral site as control. PET imaging was performed 1, 3, and 7 days post-injection, followed by ex vivo biodistribution, autoradiography and immunofluorescence analysis. Based on the results from these studies, the better-performing antibody candidate was tested similarly in an alpha-synuclein seeding model. The seeding model has intraneural alpha-synuclein aggregation and was established by intracranial injection of sonicated PFFs into both striata of F28tg mice, which overexpress human wild-type alpha-synuclein. Untreated F28tg and C57Bl/6 WT mice served as controls. RESULTS The radioimmunoconjugate was produced in sufficient radiochemical yields and purity. There was no impairment of binding affinity towards alpha-synuclein, and acceptable binding with negligible losses to mTfR1. PET imaging with [89Zr]Zr-HLu-3-scFab8D3 and [89Zr]Zr-HLu-3-(scFv8D3)2 in the deposition model showed uptake at the site of alpha-synuclein deposits. However, uptake was variable between mice. Reliable PET quantification was hampered due to the small deposition volume (~2 μL). Immunofluorescence revealed specific alpha-synuclein target engagement of both constructs with PFF deposits in the striatum, in contrast to the [89Zr]Zr-B12-scFab8D3 control. Unexpectedly, ex vivo autoradiography showed uptake in some controls ([89Zr]Zr-B12-scFab8D3 in the contralateral striatum without PFFs), potentially related to astrocyte activation at the injection sites. Ex vivo and PET brain uptake was higher for [89Zr]Zr-HLu-3-scFab8D3 when compared to [89Zr]Zr-HLu-3-(scFv8D3)2 and was therefore selected for further testing in the alpha-synuclein seeding model. No significant difference in in vivo and ex vivo brain uptake of [89Zr]Zr-HLu-3-scFab8D3 between PFF-injected F28tg, F28tg and C57Bl/6 mice was observed. Furthermore, ex vivo immunofluorescence and autoradiography showed no specific alpha-synuclein target engagement. CONCLUSIONS Successful target engagement of [89Zr]Zr-HLu-3-scFab8D3 and [89Zr]Zr-HLu-3-(scFv8D3)2 with alpha-synuclein was shown in a PFF deposition model. PET imaging showed variable results, and in vivo detection of the depositions was possible in some cases. Due to the better performance in the deposition model, [89Zr]Zr-HLu-3-scFab8D3 was further investigated in an alpha-synuclein seeding model with intraneural Lewy-body pathology, showing no difference between the control groups and PFF-seeded mice. Furthermore, immunostaining of seeded F28tg mice manifested sufficient intraneural alpha-synuclein pathology but no corresponding antibody accumulation. These results underscore the ongoing challenge of imaging intraneural inclusions via immuno-PET.
Collapse
Affiliation(s)
- Thomas E Wuensche
- Amsterdam UMC location Vrije Universiteit Amsterdam, Dept Radiology & Nuclear Medicine, De Boelelaan 1117, Amsterdam, the Netherlands.
| | - Pedro M Pereira
- Amsterdam UMC location Vrije Universiteit Amsterdam, Dept Radiology & Nuclear Medicine, De Boelelaan 1117, Amsterdam, the Netherlands
| | - Albert D Windhorst
- Amsterdam UMC location Vrije Universiteit Amsterdam, Dept Radiology & Nuclear Medicine, De Boelelaan 1117, Amsterdam, the Netherlands; Amsterdam Neuroscience, Brain Imaging, Amsterdam, the Netherlands
| | | | | | | | - Allan Jensen
- H. Lundbeck A/S, Ottiliavej 9, Valby 2500, Denmark
| | | | - Guus A M S van Dongen
- Amsterdam UMC location Vrije Universiteit Amsterdam, Dept Radiology & Nuclear Medicine, De Boelelaan 1117, Amsterdam, the Netherlands; Amsterdam Neuroscience, Brain Imaging, Amsterdam, the Netherlands
| | - Wissam Beaino
- Amsterdam UMC location Vrije Universiteit Amsterdam, Dept Radiology & Nuclear Medicine, De Boelelaan 1117, Amsterdam, the Netherlands; Amsterdam Neuroscience, Brain Imaging, Amsterdam, the Netherlands
| | - Danielle J Vugts
- Amsterdam UMC location Vrije Universiteit Amsterdam, Dept Radiology & Nuclear Medicine, De Boelelaan 1117, Amsterdam, the Netherlands; Amsterdam Neuroscience, Brain Imaging, Amsterdam, the Netherlands.
| |
Collapse
|
4
|
Ellerman DA. The Evolving Applications of Bispecific Antibodies: Reaping the Harvest of Early Sowing and Planting New Seeds. BioDrugs 2025; 39:75-102. [PMID: 39673023 DOI: 10.1007/s40259-024-00691-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/06/2024] [Indexed: 12/15/2024]
Abstract
After decades of gradual progress from conceptualization to early clinical trials (1960-2000), the therapeutic potential of bispecific antibodies (bisp Abs) is now being fully realized. Insights gained from both successful and unsuccessful trials are helping to identify which mechanisms of action, antibody formats, and targets prove most effective, and which may benefit from further refinement. While T-cell engagers remain the most commonly used class of bisp Abs, current efforts aim to increase their effectiveness by co-engaging costimulatory molecules, reducing escape mechanisms, and countering immunosuppression. Strategies to minimize cytokine release syndrome (CRS) are also actively under development. In addition, novel antibody formats that are selectively activated within tumors are an exciting area of research, as is the precise targeting of specific T-cell subsets. Beyond T cells, the recruitment of macrophages and dendritic cells is attracting increasing interest, with researchers exploring various macrophage receptors to promote phagocytosis or to carry out specialized functions, such as the immunologically silent clearance of amyloid-beta plaques in the brain. While bisp Abs targeting B cells are relatively limited, they are primarily aimed at inhibiting B-cell activity in autoimmune diseases. Another evolving application involves the forced interaction between proteins. Beyond the successful development of Hemlibra for hemophilia, bispecific antibodies that mimic cytokine activity are being explored. Additionally, the recruitment of cell surface ubiquitin ligases and other enzymes represents a novel and promising therapeutic strategy. In regard to antibody formats, some applications such as the combination of T-cell engagers with costimulatory molecules are driving the development of trispecific antibodies, at least in preclinical settings. However, the increasing structural complexity of multispecific antibodies often leads to more challenging development paths, and the number of multispecific antibodies in clinical trials remains low. The clinical success of certain applications and well-established production methods position this therapeutic class to expand its benefits into other therapeutic areas.
Collapse
Affiliation(s)
- Diego A Ellerman
- Antibody Engineering Department, Genentech Inc, South San Francisco, USA.
| |
Collapse
|
5
|
Wuensche TE, Stergiou N, Mes I, Verlaan M, Kooijman EJM, Windhorst AD, Jensen A, Asuni AA, Bang-Andersen B, van Dongen GAMS, Vugts DJ, Beaino W. Investigation of the Impact of the H310A FcRn Region Mutation on 89Zr-Immuno-PET Brain Imaging with a BBB-Shuttle Anti‑Amyloid Beta Antibody. Mol Imaging Biol 2024; 26:823-834. [PMID: 39093482 PMCID: PMC11436416 DOI: 10.1007/s11307-024-01931-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 05/27/2024] [Accepted: 06/19/2024] [Indexed: 08/04/2024]
Abstract
PURPOSE In the emerging field of antibody treatments for neurodegenerative diseases, reliable tools are needed to evaluate new therapeutics, diagnose and select patients, monitor disease progression, and assess therapy response. Immuno-PET combines the high affinity and exceptional specificity of monoclonal antibodies with the non-invasive imaging technique positron emission tomography (PET). Its application in neurodegenerative disease brain imaging has been limited due to the marginal uptake across the blood-brain barrier (BBB). The emergence of BBB-shuttle antibodies with enhanced uptake across the BBB extended immuno-PET to brain imaging. We recently reported about specific brain uptake of a bispecific aducanumab mTfR antibody in APP/PS1 TG mice using 89Zr-immuno-PET. However, a sufficient target-to-background ratio was reached at a relatively late scanning time point of 7 days post-injection. To investigate if a better target-to-background ratio could be achieved earlier, an aducanumab BBB-shuttle with a mutated Fc region for reduced FcRn affinity was evaluated. PROCEDURES AduH310A-8D3 and Adu-8D3 were modified with DFO*-NCS and subsequently radiolabeled with 89Zr. The potential influence of the H310A mutation, modification with DFO*-NCS, and subsequent radiolabeling on the in vitro binding to amyloid-beta and mTfR1 was investigated via amyloid-beta peptide ELISA and FACS analysis using mTfR1 transfected CHO-S cells. Blood kinetics, brain uptake, in vivo PET imaging and target engagement of radiolabeled AduH310A-8D3 were evaluated and compared to non-mutated Adu-8D3 in APP/PS1 TG mice and wild-type animals as controls. RESULTS Radiolabeling was performed with sufficient radiochemical yields and radiochemical purity. In vitro binding to amyloid-beta and mTfR1 showed no impairment. [89Zr]Zr-AduH310A-8D3 showed faster blood clearance and earlier differentiation of amyloid-beta-related brain uptake compared to [89Zr]Zr-Adu-8D3. However, only half of the brain uptake was observed for [89Zr]Zr-AduH310A-8D3. CONCLUSIONS Although a faster blood clearance of AduH310A-8D3 was observed, it was concluded that no beneficial effects for 89Zr-immuno-PET imaging of brain uptake were obtained.
Collapse
Affiliation(s)
- Thomas E Wuensche
- Department Radiology & Nuclear Medicine, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands.
| | - Natascha Stergiou
- Department Radiology & Nuclear Medicine, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
| | - Iris Mes
- Department Radiology & Nuclear Medicine, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
| | - Mariska Verlaan
- Department Radiology & Nuclear Medicine, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
| | - Esther J M Kooijman
- Department Radiology & Nuclear Medicine, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
| | - Albert D Windhorst
- Department Radiology & Nuclear Medicine, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Brain Imaging, Amsterdam, The Netherlands
| | - Allan Jensen
- H. Lundbeck A/S, Ottiliavej 9, 2500, Valby, Denmark
| | | | | | - Guus A M S van Dongen
- Department Radiology & Nuclear Medicine, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Brain Imaging, Amsterdam, The Netherlands
| | - Danielle J Vugts
- Department Radiology & Nuclear Medicine, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Brain Imaging, Amsterdam, The Netherlands
| | - Wissam Beaino
- Department Radiology & Nuclear Medicine, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Brain Imaging, Amsterdam, The Netherlands
| |
Collapse
|
6
|
Yang Y, Zhang Z. α-Synuclein pathology from the body to the brain: so many seeds so close to the central soil. Neural Regen Res 2024; 19:1463-1472. [PMID: 38051888 PMCID: PMC10883481 DOI: 10.4103/1673-5374.387967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 08/24/2023] [Indexed: 12/07/2023] Open
Abstract
ABSTRACT α-Synuclein is a protein that mainly exists in the presynaptic terminals. Abnormal folding and accumulation of α-synuclein are found in several neurodegenerative diseases, including Parkinson's disease. Aggregated and highly phosphorylated α-synuclein constitutes the main component of Lewy bodies in the brain, the pathological hallmark of Parkinson's disease. For decades, much attention has been focused on the accumulation of α-synuclein in the brain parenchyma rather than considering Parkinson's disease as a systemic disease. Recent evidence demonstrates that, at least in some patients, the initial α-synuclein pathology originates in the peripheral organs and spreads to the brain. Injection of α-synuclein preformed fibrils into the gastrointestinal tract triggers the gut-to-brain propagation of α-synuclein pathology. However, whether α-synuclein pathology can occur spontaneously in peripheral organs independent of exogenous α-synuclein preformed fibrils or pathological α-synuclein leakage from the central nervous system remains under investigation. In this review, we aimed to summarize the role of peripheral α-synuclein pathology in the pathogenesis of Parkinson's disease. We also discuss the pathways by which α-synuclein pathology spreads from the body to the brain.
Collapse
Affiliation(s)
- Yunying Yang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Zhentao Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
- TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei Province, China
| |
Collapse
|
7
|
Schlein E, Rokka J, Odell LR, van den Broek SL, Herth MM, Battisti UM, Syvänen S, Sehlin D, Eriksson J. Synthesis and evaluation of fluorine-18 labelled tetrazines as pre-targeting imaging agents for PET. EJNMMI Radiopharm Chem 2024; 9:21. [PMID: 38446356 PMCID: PMC10917718 DOI: 10.1186/s41181-024-00250-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Accepted: 02/28/2024] [Indexed: 03/07/2024] Open
Abstract
BACKGROUND 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 physiological obstacle, we have previously developed bispecific antibody ligands that pass through the BBB via receptor-mediated transcytosis. While these radiolabelled ligands have high affinity and specificity, their long residence time in the blood and brain, typical for large molecules, poses another challenge for PET imaging. A viable solution could be a two-step pre-targeting 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 small 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 by using the inverse electron demand Diels-Alder reaction (IEDDA), with trans-cyclooctene (TCO) and tetrazine groups participating as reactants. In this study, two novel 18F-labelled tetrazines were synthesized and evaluated for their potential use as pre-targeting imaging agents, i.e., for their ability to rapidly enter the brain and, if unbound, to be efficiently cleared with minimal background retention. RESULTS 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 showed [18F]MeTz to be the more pharmacokinetically suitable agent, given its fast and homogenous distribution in the brain and rapid clearance. However, in terms of rection kinetics, H-tetrazines are advantageous, exhibiting faster reaction rates in IEDDA reactions with dienophiles like trans-cyclooctenes, making [18F]HTz potentially more beneficial for pre-targeting applications. CONCLUSION This study demonstrates a significant potential of [18F]MeTz and [18F]HTz as agents for pre-targeted PET brain imaging due to their efficient brain uptake, swift clearance and appropriate chemical stability.
Collapse
Affiliation(s)
- Eva Schlein
- Department of Public Health and Caring Sciences, Uppsala University, 751 85, Uppsala, Sweden
| | - Johanna Rokka
- Department of Public Health and Caring Sciences, Uppsala University, 751 85, Uppsala, Sweden
| | - Luke R Odell
- Department of Medicinal Chemistry, Uppsala University, 751 23, Uppsala, Sweden
| | | | - Matthias M Herth
- Department of Drug Design and Pharmacology, University of Copenhagen, 2100, Copenhagen, Denmark
- Department of Clinical Physiology, Nuclear Medicine & PET, Rigshospitalet Copenhagen University Hospital, Blegdamsvej 9, 2100, Copenhagen, Denmark
| | - Umberto M Battisti
- Department of Drug Design and Pharmacology, University of Copenhagen, 2100, Copenhagen, Denmark
| | - Stina Syvänen
- Department of Public Health and Caring Sciences, Uppsala University, 751 85, Uppsala, Sweden
| | - Dag Sehlin
- Department of Public Health and Caring Sciences, Uppsala University, 751 85, Uppsala, Sweden
| | - Jonas Eriksson
- Department of Medicinal Chemistry, Uppsala University, 751 23, Uppsala, Sweden.
- PET Centre, Uppsala University Hospital, 751 85, Uppsala, Sweden.
| |
Collapse
|
8
|
Faresjö R, Sjöström EO, Dallas T, Berglund MM, Eriksson J, Sehlin D, Syvänen S. Single domain antibody-scFv conjugate targeting amyloid β and TfR penetrates the blood-brain barrier and interacts with amyloid β. MAbs 2024; 16:2410968. [PMID: 39358860 PMCID: PMC11451328 DOI: 10.1080/19420862.2024.2410968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 09/05/2024] [Accepted: 09/25/2024] [Indexed: 10/04/2024] Open
Abstract
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.
Collapse
Affiliation(s)
- Rebecca Faresjö
- Department of Public Health and Caring Sciences, Uppsala University, Uppsala, Sweden
| | - Elisabet O. Sjöström
- Department of Public Health and Caring Sciences, Uppsala University, Uppsala, Sweden
- Key2Brain AB, Solna, Sweden
| | - Tiffany Dallas
- Department of Public Health and Caring Sciences, Uppsala University, Uppsala, Sweden
- Key2Brain AB, Solna, Sweden
| | | | - Jonas Eriksson
- PET Centre, Uppsala University Hospital, Uppsala, Sweden
- Department of Medicinal Chemistry, Uppsala University, Sweden
| | - Dag Sehlin
- Department of Public Health and Caring Sciences, Uppsala University, Uppsala, Sweden
| | - Stina Syvänen
- Department of Public Health and Caring Sciences, Uppsala University, Uppsala, Sweden
| |
Collapse
|
9
|
Lan HR, Chen M, Yao SY, Chen JX, Jin KT. Bispecific antibodies revolutionizing breast cancer treatment: a comprehensive overview. Front Immunol 2023; 14:1266450. [PMID: 38111570 PMCID: PMC10725925 DOI: 10.3389/fimmu.2023.1266450] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 11/16/2023] [Indexed: 12/20/2023] Open
Abstract
Breast cancer (BCa) is known as a complex and prevalent disease requiring the development of novel anticancer therapeutic approaches. Bispecific antibodies (BsAbs) have emerged as a favorable strategy for BCa treatment due to their unique ability to target two different antigens simultaneously. By targeting tumor-associated antigens (TAAs) on cancer cells, engaging immune effector cells, or blocking critical signaling pathways, BsAbs offer enhanced tumor specificity and immune system involvement, improving anti-cancer activity. Preclinical and clinical studies have demonstrated the potential of BsAbs in BCa. For example, BsAbs targeting human epidermal growth factor receptor 2 (HER2) have shown the ability to redirect immune cells to HER2-positive BCa cells, resulting in effective tumor cell killing. Moreover, targeting the PD-1/PD-L1 pathway by BsAbs has demonstrated promising outcomes in overcoming immunosuppression and enhancing immune-mediated tumor clearance. Combining BsAbs with existing therapeutic approaches, such as chemotherapy, targeted therapies, or immune checkpoint inhibitors (ICIs), has also revealed synergistic effects in preclinical models and early clinical trials, emphasizing the usefulness and potential of BsAbs in BCa treatment. This review summarizes the latest evidence about BsAbs in treating BCa and the challenges and opportunities of their use in BCa.
Collapse
Affiliation(s)
- Huan-Rong Lan
- Department of Surgical Oncology, Hangzhou Cancer Hospital, Hangzhou, Zhejiang, China
| | - Min Chen
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Shi-Ya Yao
- Department of Colorectal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, China
| | - Jun-Xia Chen
- Department of Gynecology, Shaoxing People’s Hospital, Shaoxing, Zhejiang, China
| | - Ke-Tao Jin
- Department of Colorectal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, China
| |
Collapse
|
10
|
Pardridge WM. Treatment of Parkinson's disease with biologics that penetrate the blood-brain barrier via receptor-mediated transport. Front Aging Neurosci 2023; 15:1276376. [PMID: 38035276 PMCID: PMC10682952 DOI: 10.3389/fnagi.2023.1276376] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 10/27/2023] [Indexed: 12/02/2023] Open
Abstract
Parkinson's disease (PD) is characterized by neurodegeneration of nigral-striatal neurons in parallel with the formation of intra-neuronal α-synuclein aggregates, and these processes are exacerbated by neuro-inflammation. All 3 components of PD pathology are potentially treatable with biologics. Neurotrophins, such as glial derived neurotrophic factor or erythropoietin, can promote neural repair. Therapeutic antibodies can lead to disaggregation of α-synuclein neuronal inclusions. Decoy receptors can block the activity of pro-inflammatory cytokines in brain. However, these biologic drugs do not cross the blood-brain barrier (BBB). Biologics can be made transportable through the BBB following the re-engineering of the biologic as an IgG fusion protein, where the IgG domain targets an endogenous receptor-mediated transcytosis (RMT) system within the BBB, such as the insulin receptor or transferrin receptor. The receptor-specific antibody domain of the fusion protein acts as a molecular Trojan horse to ferry the biologic into brain via the BBB RMT pathway. This review describes the re-engineering of all 3 classes of biologics (neurotrophins, decoy receptor, therapeutic antibodies) for BBB delivery and treatment of PD. Targeting the RMT pathway at the BBB also enables non-viral gene therapy of PD using lipid nanoparticles (LNP) encapsulated with plasmid DNA encoding therapeutic genes. The surface of the lipid nanoparticle is conjugated with a receptor-specific IgG that triggers RMT of the LNP across the BBB in vivo.
Collapse
|
11
|
Pardridge WM. Receptor-mediated drug delivery of bispecific therapeutic antibodies through the blood-brain barrier. FRONTIERS IN DRUG DELIVERY 2023; 3:1227816. [PMID: 37583474 PMCID: PMC10426772 DOI: 10.3389/fddev.2023.1227816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 08/17/2023]
Abstract
Therapeutic antibody drug development is a rapidly growing sector of the pharmaceutical industry. However, antibody drug development for the brain is a technical challenge, and therapeutic antibodies for the central nervous system account for ~3% of all such agents. The principal obstacle to antibody drug development for brain or spinal cord is the lack of transport of large molecule biologics across the blood-brain barrier (BBB). Therapeutic antibodies can be made transportable through the blood-brain barrier by the re-engineering of the therapeutic antibody as a BBB-penetrating bispecific antibody (BSA). One arm of the BSA is the therapeutic antibody and the other arm of the BSA is a transporting antibody. The transporting antibody targets an exofacial epitope on a BBB receptor, and this enables receptor-mediated transcytosis (RMT) of the BSA across the BBB. Following BBB transport, the therapeutic antibody then engages the target receptor in brain. RMT systems at the BBB that are potential conduits to the brain include the insulin receptor (IR), the transferrin receptor (TfR), the insulin-like growth factor receptor (IGFR) and the leptin receptor. Therapeutic antibodies have been re-engineered as BSAs that target the insulin receptor, TfR, or IGFR RMT systems at the BBB for the treatment of Alzheimer's disease and Parkinson's disease.
Collapse
|
12
|
Rodger AT, ALNasser M, Carter WG. Are Therapies That Target α-Synuclein Effective at Halting Parkinson's Disease Progression? A Systematic Review. Int J Mol Sci 2023; 24:11022. [PMID: 37446200 PMCID: PMC10341763 DOI: 10.3390/ijms241311022] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 06/29/2023] [Accepted: 06/29/2023] [Indexed: 07/15/2023] Open
Abstract
There are currently no pharmacological treatments available that completely halt or reverse the progression of Parkinson's Disease (PD). Hence, there is an unmet need for neuroprotective therapies. Lewy bodies are a neuropathological hallmark of PD and contain aggregated α-synuclein (α-syn) which is thought to be neurotoxic and therefore a suitable target for therapeutic interventions. To investigate this further, a systematic review was undertaken to evaluate whether anti-α-syn therapies are effective at preventing PD progression in preclinical in vivo models of PD and via current human clinical trials. An electronic literature search was performed using MEDLINE and EMBASE (Ovid), PubMed, the Web of Science Core Collection, and Cochrane databases to collate clinical evidence that investigated the targeting of α-syn. Novel preclinical anti-α-syn therapeutics provided a significant reduction of α-syn aggregations. Biochemical and immunohistochemical analysis of rodent brain tissue demonstrated that treatments reduced α-syn-associated pathology and rescued dopaminergic neuronal loss. Some of the clinical studies did not provide endpoints since they had not yet been completed or were terminated before completion. Completed clinical trials displayed significant tolerability and efficacy at reducing α-syn in patients with PD with minimal adverse effects. Collectively, this review highlights the capacity of anti-α-syn therapies to reduce the accumulation of α-syn in both preclinical and clinical trials. Hence, there is potential and optimism to target α-syn with further clinical trials to restrict dopaminergic neuronal loss and PD progression and/or provide prophylactic protection to avoid the onset of α-syn-induced PD.
Collapse
Affiliation(s)
- Abbie T. Rodger
- School of Medicine, University of Nottingham, Royal Derby Hospital Centre, Derby DE22 3DT, UK; (A.T.R.); (M.A.)
| | - Maryam ALNasser
- School of Medicine, University of Nottingham, Royal Derby Hospital Centre, Derby DE22 3DT, UK; (A.T.R.); (M.A.)
- Department of Biological Sciences, College of Science, King Faisal University, P.O. Box 400, Al-Ahsa 31982, Saudi Arabia
| | - Wayne G. Carter
- School of Medicine, University of Nottingham, Royal Derby Hospital Centre, Derby DE22 3DT, UK; (A.T.R.); (M.A.)
| |
Collapse
|
13
|
Düchs M, Blazevic D, Rechtsteiner P, Kenny C, Lamla T, Low S, Savistchenko J, Neumann M, Melki R, Schönberger T, Stierstorfer B, Wyatt D, Igney F, Ciossek T. AAV-mediated expression of a new conformational anti-aggregated α-synuclein antibody prolongs survival in a genetic model of α-synucleinopathies. NPJ Parkinsons Dis 2023; 9:91. [PMID: 37322068 DOI: 10.1038/s41531-023-00542-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 06/05/2023] [Indexed: 06/17/2023] Open
Abstract
Prion-like transmission of pathology in α-synucleinopathies like Parkinson's disease or multiple system atrophy is increasingly recognized as one potential mechanism to address disease progression. Active and passive immunotherapies targeting insoluble, aggregated α-synuclein are already being actively explored in the clinic with mixed outcomes so far. Here, we report the identification of 306C7B3, a highly selective, aggregate-specific α-synuclein antibody with picomolar affinity devoid of binding to the monomeric, physiologic protein. 306C7B3 binding is Ser129-phosphorylation independent and shows high affinity to several different aggregated α-synuclein polymorphs, increasing the likelihood that it can also bind to the pathological seeds assumed to drive disease progression in patients. In support of this, highly selective binding to pathological aggregates in postmortem brains of MSA patients was demonstrated, with no staining in samples from other human neurodegenerative diseases. To achieve CNS exposure of 306C7B3, an adeno-associated virus (AAV) based approach driving expression of the secreted antibody within the brain of (Thy-1)-[A30P]-hα-synuclein mice was used. Widespread central transduction after intrastriatal inoculation was ensured by using the AAV2HBKO serotype, with transduction being spread to areas far away from the inoculation site. Treatment of (Thy-1)-[A30P]-hα-synuclein mice at the age of 12 months demonstrated significantly increased survival, with 306C7B3 concentration reaching 3.9 nM in the cerebrospinal fluid. These results suggest that AAV-mediated expression of 306C7B3, targeting extracellular, presumably disease-propagating aggregates of α-synuclein, has great potential as a disease-modifying therapy for α-synucleinopathies as it ensures CNS exposure of the antibody, thereby mitigating the selective permeability of the blood-brain barrier.
Collapse
Affiliation(s)
- Matthias Düchs
- Boehringer Ingelheim Pharma GmbH & Co KG, Biberach an der Riss, Germany
| | - Dragica Blazevic
- Boehringer Ingelheim Pharma GmbH & Co KG, Biberach an der Riss, Germany
| | | | | | - Thorsten Lamla
- Boehringer Ingelheim Pharma GmbH & Co KG, Biberach an der Riss, Germany
| | - Sarah Low
- Boehringer Ingelheim USA, Ridgefield, CT, USA
| | | | - Manuela Neumann
- Molecular Neuropathology of Neurodegenerative Diseases, German Center for Neurodegenerative Diseases, Tübingen, Germany
- Department of Neuropathology, University Hospital of Tübingen, Tübingen, Germany
| | - Ronald Melki
- Institut Francois Jacob (MIRCen), CEA, CNRS, Fontenay-aux-Roses, France
| | - Tanja Schönberger
- Boehringer Ingelheim Pharma GmbH & Co KG, Biberach an der Riss, Germany
| | | | - David Wyatt
- Boehringer Ingelheim Pharma GmbH & Co KG, Biberach an der Riss, Germany
| | - Frederik Igney
- Boehringer Ingelheim Pharma GmbH & Co KG, Biberach an der Riss, Germany
| | - Thomas Ciossek
- Boehringer Ingelheim Pharma GmbH & Co KG, Biberach an der Riss, Germany.
| |
Collapse
|
14
|
Gustavsson T, Metzendorf NG, Wik E, Roshanbin S, Julku U, Chourlia A, Nilsson P, Andersson KG, Laudon H, Hultqvist G, Syvänen S, Sehlin D. Long-term effects of immunotherapy with a brain penetrating Aβ antibody in a mouse model of Alzheimer's disease. Alzheimers Res Ther 2023; 15:90. [PMID: 37131196 PMCID: PMC10152635 DOI: 10.1186/s13195-023-01236-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 04/23/2023] [Indexed: 05/04/2023]
Abstract
BACKGROUND Brain-directed immunotherapy is a promising strategy to target amyloid-β (Aβ) deposits in Alzheimer's disease (AD). In the present study, we compared the therapeutic efficacy of the Aβ protofibril targeting antibody RmAb158 with its bispecific variant RmAb158-scFv8D3, which enters the brain by transferrin receptor-mediated transcytosis. METHODS AppNL-G-F knock-in mice received RmAb158, RmAb158-scFv8D3, or PBS in three treatment regimens. First, to assess the acute therapeutic effect, a single antibody dose was given to 5 months old AppNL-G-F mice, with evaluation after 3 days. Second, to assess the antibodies' ability to halt the progression of Aβ pathology, 3 months old AppNL-G-F mice received three doses during a week, with evaluation after 2 months. Reduction of RmAb158-scFv8D3 immunogenicity was explored by introducing mutations in the antibody or by depletion of CD4+ T cells. Third, to study the effects of chronic treatment, 7-month-old AppNL-G-F mice were CD4+ T cell depleted and treated with weekly antibody injections for 8 weeks, including a final diagnostic dose of [125I]RmAb158-scFv8D3, to determine its brain uptake ex vivo. Soluble Aβ aggregates and total Aβ42 were quantified with ELISA and immunostaining. RESULTS Neither RmAb158-scFv8D3 nor RmAb158 reduced soluble Aβ protofibrils or insoluble Aβ1-42 after a single injection treatment. After three successive injections, Aβ1-42 was reduced in mice treated with RmAb158, with a similar trend in RmAb158-scFv8D3-treated mice. Bispecific antibody immunogenicity was somewhat reduced by directed mutations, but CD4+ T cell depletion was used for long-term therapy. CD4+ T cell-depleted mice, chronically treated with RmAb158-scFv8D3, showed a dose-dependent increase in blood concentration of the diagnostic [125I]RmAb158-scFv8D3, while concentration was low in plasma and brain. Chronic treatment did not affect soluble Aβ aggregates, but a reduction in total Aβ42 was seen in the cortex of mice treated with both antibodies. CONCLUSIONS Both RmAb158 and its bispecific variant RmAb158-scFv8D3 achieved positive effects of long-term treatment. Despite its ability to efficiently enter the brain, the benefit of using the bispecific antibody in chronic treatment was limited by its reduced plasma exposure, which may be a result of interactions with TfR or the immune system. Future research will focus in new antibody formats to further improve Aβ immunotherapy.
Collapse
Affiliation(s)
- Tobias Gustavsson
- Department of Public Health and Caring Sciences, Uppsala University, Uppsala, Sweden
| | | | - Elin Wik
- Department of Public Health and Caring Sciences, Uppsala University, Uppsala, Sweden
| | - Sahar Roshanbin
- Department of Public Health and Caring Sciences, Uppsala University, Uppsala, Sweden
| | - Ulrika Julku
- Department of Public Health and Caring Sciences, Uppsala University, Uppsala, Sweden
| | | | - Per Nilsson
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, Stockholm, Sweden
| | | | | | | | - Stina Syvänen
- Department of Public Health and Caring Sciences, Uppsala University, Uppsala, Sweden
| | - Dag Sehlin
- Department of Public Health and Caring Sciences, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
15
|
Haider A, Elghazawy NH, Dawoud A, Gebhard C, Wichmann T, Sippl W, Hoener M, Arenas E, Liang SH. Translational molecular imaging and drug development in Parkinson's disease. Mol Neurodegener 2023; 18:11. [PMID: 36759912 PMCID: PMC9912681 DOI: 10.1186/s13024-023-00600-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 01/23/2023] [Indexed: 02/11/2023] Open
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disorder that primarily affects elderly people and constitutes a major source of disability worldwide. Notably, the neuropathological hallmarks of PD include nigrostriatal loss and the formation of intracellular inclusion bodies containing misfolded α-synuclein protein aggregates. Cardinal motor symptoms, which include tremor, rigidity and bradykinesia, can effectively be managed with dopaminergic therapy for years following symptom onset. Nonetheless, patients ultimately develop symptoms that no longer fully respond to dopaminergic treatment. Attempts to discover disease-modifying agents have increasingly been supported by translational molecular imaging concepts, targeting the most prominent pathological hallmark of PD, α-synuclein accumulation, as well as other molecular pathways that contribute to the pathophysiology of PD. Indeed, molecular imaging modalities such as positron emission tomography (PET) and single-photon emission computed tomography (SPECT) can be leveraged to study parkinsonism not only in animal models but also in living patients. For instance, mitochondrial dysfunction can be assessed with probes that target the mitochondrial complex I (MC-I), while nigrostriatal degeneration is typically evaluated with probes designed to non-invasively quantify dopaminergic nerve loss. In addition to dopaminergic imaging, serotonin transporter and N-methyl-D-aspartate (NMDA) receptor probes are increasingly used as research tools to better understand the complexity of neurotransmitter dysregulation in PD. Non-invasive quantification of neuroinflammatory processes is mainly conducted by targeting the translocator protein 18 kDa (TSPO) on activated microglia using established imaging agents. Despite the overwhelming involvement of the brain and brainstem, the pathophysiology of PD is not restricted to the central nervous system (CNS). In fact, PD also affects various peripheral organs such as the heart and gastrointestinal tract - primarily via autonomic dysfunction. As such, research into peripheral biomarkers has taken advantage of cardiac autonomic denervation in PD, allowing the differential diagnosis between PD and multiple system atrophy with probes that visualize sympathetic nerve terminals in the myocardium. Further, α-synuclein has recently gained attention as a potential peripheral biomarker in PD. This review discusses breakthrough discoveries that have led to the contemporary molecular concepts of PD pathophysiology and how they can be harnessed to develop effective imaging probes and therapeutic agents. Further, we will shed light on potential future trends, thereby focusing on potential novel diagnostic tracers and disease-modifying therapeutic interventions.
Collapse
Affiliation(s)
- Achi Haider
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, MA, 02114, USA.
- Department of Radiology and Imaging Sciences, Emory University, 101 Woodruff Circle, Atlanta, GA, 30322, USA.
| | - Nehal H Elghazawy
- Biochemistry Department, Faculty of Pharmacy and Biotechnology, German University in Cairo, Main Entrance of Al-Tagamoa Al-Khames, Cairo, 11835, Egypt
- Molecular Genetics Research Team (MGRT), Pharmaceutical Biology Department, Faculty of Pharmacy and Biotechnology, German University in Cairo, Main Entrance of Al-Tagamoa Al-Khames, Cairo, 11835, Egypt
| | - Alyaa Dawoud
- Biochemistry Department, Faculty of Pharmacy and Biotechnology, German University in Cairo, Main Entrance of Al-Tagamoa Al-Khames, Cairo, 11835, Egypt
- Molecular Genetics Research Team (MGRT), Pharmaceutical Biology Department, Faculty of Pharmacy and Biotechnology, German University in Cairo, Main Entrance of Al-Tagamoa Al-Khames, Cairo, 11835, Egypt
| | - Catherine Gebhard
- Department of Nuclear Medicine, University Hospital Zurich, Raemistrasse 100, 8091, Zurich, Switzerland
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | - Thomas Wichmann
- Department of Neurology/School of Medicine, Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
| | - Wolfgang Sippl
- Institute of Pharmacy, Department of Medicinal Chemistry, Martin-Luther-University Halle-Wittenberg, W.-Langenbeck-Str. 4, 06120, Halle, Germany
| | - Marius Hoener
- Neuroscience and Rare Diseases Discovery and Translational Area, Roche Innovation Center Basel, F. Hoffmann-La Roche, Grenzacherstrasse 124, 4070, Basel, Switzerland
| | - Ernest Arenas
- Karolinska Institutet, MBB, Molecular Neurobiology, Stockholm, Sweden
| | - Steven H Liang
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, MA, 02114, USA.
- Department of Radiology and Imaging Sciences, Emory University, 101 Woodruff Circle, Atlanta, GA, 30322, USA.
| |
Collapse
|
16
|
Petersen I, Ali MI, Petrovic A, Ytterberg AJ, Staxäng K, Hodik M, Rofo F, Bondza S, Hultqvist G. Multivalent design of the monoclonal SynO2 antibody improves binding strength to soluble α-Synuclein aggregates. MAbs 2023; 15:2256668. [PMID: 37737124 PMCID: PMC10519360 DOI: 10.1080/19420862.2023.2256668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 09/05/2023] [Indexed: 09/23/2023] Open
Abstract
Soluble aggregates are reported to be the most neurotoxic species of α-Synuclein (αSyn) in Parkinson's disease (PD) and hence are a promising target for diagnosis and treatment of PD. However, the predominantly intracellular location of αSyn limits its accessibility, especially for antibody-based molecules and prompts the need for exceptionally strong soluble αSyn aggregate binders to enhance their sensitivity and efficacy for targeting the extracellular αSyn pool. In this study, we have created the multivalent antibodies TetraSynO2 and HexaSynO2, derived from the αSyn oligomer-specific antibody SynO2, to increase avidity binding to soluble αSyn aggregate species through more binding sites in close proximity. The multivalency was achieved through recombinant fusion of single-chain variable fragments of SynO2 to the antibodies' original N-termini. Our ELISA results indicated a 20-fold increased binding strength of the multivalent formats to αSyn aggregates, while binding to αSyn monomers and unspecific binding to amyloid β protofibrils remained low. Kinetic analysis using LigandTracer revealed that only 80% of SynO2 bound bivalently to soluble αSyn aggregates, whereas the proportion of TetraSynO2 and HexaSynO2 binding bi- or multivalently to soluble αSyn aggregates was increased to ~ 95% and 100%, respectively. The overall improved binding strength of TetraSynO2 and HexaSynO2 implies great potential for immunotherapeutic and diagnostic applications with targets of limited accessibility, like extracellular αSyn aggregates. The ability of the multivalent antibodies to bind a wider range of αSyn aggregate species, which are not targetable by conventional bivalent antibodies, thus could allow for an earlier and more effective intervention in the progression of PD.
Collapse
Affiliation(s)
- Inga Petersen
- Department of Pharmacy, Uppsala University, Uppsala, Sweden
| | | | - Alex Petrovic
- Department of Pharmacy, Uppsala University, Uppsala, Sweden
| | - Anders Jimmy Ytterberg
- Department of Pharmacy, SciLifeLab Drug Discovery and Development, Uppsala University, Uppsala, Sweden
| | - Karin Staxäng
- TEM Laboratory, BioVis Platform, Uppsala University, Uppsala, Sweden
| | - Monika Hodik
- TEM Laboratory, BioVis Platform, Uppsala University, Uppsala, Sweden
| | - Fadi Rofo
- Department of Pharmacy, Uppsala University, Uppsala, Sweden
| | - Sina Bondza
- Ridgeview Instruments AB, Uppsala, Sweden
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | | |
Collapse
|
17
|
Pardridge WM. Advanced Blood-Brain Barrier Drug Delivery. Pharmaceutics 2022; 15:pharmaceutics15010093. [PMID: 36678722 PMCID: PMC9866552 DOI: 10.3390/pharmaceutics15010093] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 12/06/2022] [Indexed: 12/29/2022] Open
Abstract
This Special Issue of Pharmaceutics, "Advanced Blood-Brain Barrier Drug Delivery," comprises 16 articles or reviews, which cover a cross-section of brain drug delivery for either small-molecule or large-molecule therapeutics [...].
Collapse
Affiliation(s)
- William M Pardridge
- Department of Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA
| |
Collapse
|