SwePub
Sök i SwePub databas

  Utökad sökning

Träfflista för sökning "WFRF:(Hultqvist Greta) srt2:(2020-2024)"

Sökning: WFRF:(Hultqvist Greta) > (2020-2024)

  • Resultat 1-10 av 20
Sortera/gruppera träfflistan
   
NumreringReferensOmslagsbildHitta
1.
  • Banka, Vinay, et al. (författare)
  • Development of brain-penetrable antibody radioligands for in vivo PET imaging of amyloid-β and tau
  • 2023
  • Ingår i: Frontiers in nuclear medicine. - : Frontiers Media S.A.. - 2673-8880. ; 3
  • Tidskriftsartikel (refereegranskat)abstract
    • INTRODUCTION: Alzheimer's disease (AD) is characterized by the misfolding and aggregation of two major proteins: amyloid-beta (Aβ) and tau. Antibody-based PET radioligands are desirable due to their high specificity and affinity; however, antibody uptake in the brain is limited by the blood-brain barrier (BBB). Previously, we demonstrated that antibody transport across the BBB can be facilitated through interaction with the transferrin receptor (TfR), and the bispecific antibody-based PET ligands were capable of detecting Aβ aggregates via ex vivo imaging. Since tau accumulation in the brain is more closely correlated with neuronal death and cognition, we report here our strategies to prepare four F-18-labeled specifically engineered bispecific antibody probes for the selective detection of tau and Aβ aggregates to evaluate their feasibility and specificity, particularly for in vivo PET imaging.METHODS: We first created and evaluated (via both in vitro and ex vivo studies) four specifically engineered bispecific antibodies, by fusion of single-chain variable fragments (scFv) of a TfR antibody with either a full-size IgG antibody of Aβ or tau or with their respective scFv. Using [18F]SFB as the prosthetic group, all four 18F-labeled bispecific antibody probes were then prepared by conjugation of antibody and [18F]SFB in acetonitrile/0.1 M borate buffer solution (final pH ~ 8.5) with an incubation of 20 min at room temperature, followed by purification on a PD MiniTrap G-25 size exclusion gravity column.RESULTS: Based on both in vitro and ex vivo evaluation, the bispecific antibodies displayed much higher brain concentrations than the unmodified antibody, supporting our subsequent F18-radiolabeling. [18F]SFB was produced in high yields in 60 min (decay-corrected radiochemical yield (RCY) 46.7 ± 5.4) with radiochemical purities of >95%, confirmed by analytical high performance liquid chromatography (HPLC) and radio-TLC. Conjugation of [18F]SFB and bispecific antibodies showed a 65%-83% conversion efficiency with radiochemical purities of 95%-99% by radio-TLC.CONCLUSIONS: We successfully labeled four novel and specifically engineered bispecific antibodies with [18F]SFB under mild conditions with a high RCY and purities. This study provides strategies to create brain-penetrable F-18 radiolabeled antibody probes for the selective detection of tau and Aβ aggregates in the brain of transgenic AD mice via in vivo PET imaging.
  •  
2.
  • de la Rosa, Andrés (författare)
  • Design, expression, and analysis of antibody-based blood-brain barrier shuttles
  • 2023
  • Doktorsavhandling (övrigt vetenskapligt/konstnärligt)abstract
    • Antibody therapeutics, with their strong and highly selective target binding, are now used to treat various diseases. However, to enable their use to treat brain disorders, they must be delivered across the blood-brain barrier (BBB), as without active transport, only around 0.01% of intravenously injected doses reach the brain. Brain delivery can be done by BBB shuttles capable of binding receptors that naturally transport proteins, e.g., the Transferrin receptor (TfR). This thesis has studied strategies for designing TfR-binding shuttles and how to enhance the protein expression of antibody therapeutics. In Paper I, we shared our updated transient gene expression (TGE) protocol and developed a small-scale version to surmount the cost limitations of testing many conditions. Large variations of protein expression were observed for both protocols, prompting future studies investigating its cause(s). In paper II, we investigated if binding to the glycosaminoglycan heparan sulfate (HS) present at the BBB could improve brain delivery. Our results indicate that the BBB shuttle scFv8D3 is not dependent on the HS-binding sites identified, and adding new HS-binding sites did not enhance delivery. However, further studies are required due to HS's complexity and heterogeneity. Decreasing the TfR affinity of BBB shuttles has been shown to boost the delivery of therapeutic doses of high affinity anti-TfR antibodies, e.g., bivalent 8D3 antibodies. In Paper III, we applied the strategy to a monovalent single-chain fragment variable (scFv) of 8D3 (scFv8D3) based BBB shuttle. Our affinity mutants exhibited lowered TfR affinity, longer blood half-life, and higher brain concentration. Using our In-Cell BBB Trans assay, we concluded that the increased brain concentration is likely due to extended blood half-life. In paper IV, we fused the TfR ligand holo-transferrin to the TfR binding arms of the partly bivalent RmAb158-scFv8D3 antibody. Our results indicate that the TfR binding shifted from partly to fully bivalent, resulting in markedly decreased in vitro transcytosis. The potential transcytosis-promoting effect of the fused holoTf was absent and/or counteracted by the bivalent binding of the design. However, the strategy may still prove useful for monovalent TfR binders. In conclusion, monovalent and low-to-moderate affinity are likely beneficial binding properties for TfR-mediated brain delivery at therapeutic doses. However, whether it is possible to enhance brain delivery with HS-binding or holoTf-fusion requires further study.
  •  
3.
  • de la Rosa, Andres, et al. (författare)
  • Introducing or removing heparan sulfate binding sites does not alter brain uptake of the blood-brain barrier shuttle scFv8D3
  • 2022
  • Ingår i: Scientific Reports. - : Springer Nature. - 2045-2322. ; 12:1
  • Tidskriftsartikel (refereegranskat)abstract
    • The blood-brain barrier (BBB) greatly limits the delivery of protein-based drugs into the brain and is a major obstacle for the treatment of brain disorders. Targeting the transferrin receptor (TfR) is a strategy for transporting protein-based drugs into the brain, which can be utilized by using TfR-binding BBB transporters, such as the TfR-binding antibody 8D3. In this current study, we investigated if binding to heparan sulfate (HS) contributes to the brain uptake of a single chain fragment variable of 8D3 (scFv8D3). We designed and produced a scFv8D3 mutant, engineered with additional HS binding sites, HS(+)scFv8D3, to assess whether increased HS binding would improve brain uptake. Additionally, a mutant with a reduced number of HS binding sites, HS(-)scFv8D3, was also engineered to see if reducing the HS binding sites could also affect brain uptake. Heparin column chromatography showed that only the HS(+)scFv8D3 mutant bound HS in the experimental conditions. Ex vivo results showed that the brain uptake was unaffected by the introduction or removal of HS binding sites, which indicates that scFv8D3 is not dependent on the HS binding sites for brain uptake. Conversely, introducing HS binding sites to scFv8D3 decreased its renal excretion while removing them had the opposite effect.
  •  
4.
  • Gustavsson, Tobias, et al. (författare)
  • Long-term effects of immunotherapy with a brain penetrating Aβ antibody in a mouse model of Alzheimer's disease
  • 2023
  • Ingår i: Alzheimer's Research & Therapy. - : BioMed Central (BMC). - 1758-9193. ; 15:1
  • Tidskriftsartikel (refereegranskat)abstract
    • BackgroundBrain-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.MethodsAppNL−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.ResultsNeither 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.ConclusionsBoth 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.
  •  
5.
  • Meier, Silvio R., et al. (författare)
  • Pinpointing Brain TREM2 Levels in Two Mouse Models of Alzheimer's Disease
  • 2021
  • Ingår i: Molecular Imaging and Biology. - : Springer. - 1536-1632 .- 1860-2002. ; 23, s. 665-675
  • Tidskriftsartikel (refereegranskat)abstract
    • PURPOSE: The triggering receptor expressed on myeloid cells 2 (TREM2) is expressed by brain microglia. Microglial activation, as observed in Alzheimer's disease (AD) as well as in transgenic mice expressing human amyloid-beta, appears to increase soluble TREM2 (sTREM2) levels in CSF and brain. In this study, we used two different transgenic mouse models of AD pathology and investigated the potential of TREM2 to serve as an in vivo biomarker for microglial activation in AD.PROCEDURES: We designed and generated a bispecific antibody based on the TREM2-specific monoclonal antibody mAb1729, fused to a single-chain variable fragment of the transferrin receptor binding antibody 8D3. The 8D3-moiety enabled transcytosis of the whole bispecific antibody across the blood-brain barrier. The bispecific antibody was radiolabeled with I-125 (ex vivo) or I-124 (PET) and administered to transgenic AD and wild-type (WT) control mice. Radioligand retention in the brain of transgenic animals was compared to WT mice by isolation of brain tissue at 24 h or 72 h, or with in vivo PET at 24 h, 48 h, and 72 h. Intrabrain distribution of radiolabeled mAb1729-scFv8D3CL was further studied by autoradiography, while ELISA was used to determine TREM2 brain concentrations.RESULTS: Transgenic animals displayed higher total exposure, calculated as the AUC based on SUV determined at 24h, 48h, and 72h post injection, of PET radioligand [124I]mAb1729-scFv8D3CL than WT mice. However, differences were not evident in single time point PET images or SUVs. Ex vivo autoradiography confirmed higher radioligand concentrations in cortex and thalamus in transgenic mice compared to WT, and TREM2 levels in brain homogenates were considerably higher in transgenic mice compared to WT.CONCLUSION: Antibody-based radioligands, engineered to enter the brain, may serve as PET radioligands to follow changes of TREM2 in vivo, but antibody formats with faster systemic clearance to increase the specific signal in relation to that from blood in combination with antibodies showing higher affinity for TREM2 must be developed to further progress this technique for in vivo use.
  •  
6.
  • Meier, Silvio R., 1990- (författare)
  • Preclinical PET imaging and therapy of Alzheimer's disease
  • 2021
  • Doktorsavhandling (övrigt vetenskapligt/konstnärligt)abstract
    • The main histopathological hallmarks of Alzheimer’s disease are extracellular amyloid-beta (Aβ) plaques and intracellular neurofibrillary tangles, containing tau protein. Because of misfolded and aggregated proteins, activated microglia and astroglia react with a neuroinflammatory response, which may contribute to disease progression and severity. To date, there is no treatment available that stops the underlying mechanisms of the disease, but several new drug candidates entered clinical trials every year during the last decade. New treatments, aiming to clear Aβ from the brain parenchyma or to reduce Aβ production, are dependent on diagnostic tools to follow changes in brain Aβ pathology in vivo. The presence of brain amyloid, verified with positron emission tomography (PET), is a regularly used criterion for enrolling patients in clinical trials. However, current amyloid radioligands such as [11C]Pittsburgh Compound B ([11C]PiB) have some disadvantages, e.g. early saturation during disease progression and reduced binding to diffuse Aβ pathology. Currently available radioligands for imaging of neuroinflammation are also suboptimal. In this thesis, we investigated the potential of a brain-penetrating, bispecific Aβ antibody as a PET ligand to detect effects of treatment. In paper I and II, we demonstrated that this ligand can follow Aβ disease progression and that Aβ reduction due to treatment with a BACE-1 inhibitor can be quantified in a mouse model of AD. In paper II we also compared antibody-PET with [11C]PiB-PET and showed that the two ligands provided differing read-outs.In paper III we created and investigated an antibody-based radioligand against the triggering receptor expressed on myeloid cells 2. Compared to wild type mice, transgenic animals displayed higher total in vivo exposure, calculated as the area under the concentration curve based on PET at 24 h, 48 h and 72 h post injection. However, differences were not evident in single time point PET images.In paper IV we investigated brain delivery of a nanobody against GFAP with and without active transcytosis over the blood-brain barrier in vivo. Brain uptake with active transcytosis was two times higher. However, brain retention after 8 h, 24 h or 48 h did not differ between transgenic and wild type mice. In paper V we studied the potential of a hexavalent and bispecific antibody construct against soluble Aβ aggregates for PET or immunotherapy in vivo. Its brain retention increased with age when applied at tracer doses in genetically modified mice. However, when applied at therapeutic dose, it had no or very low impact on Aβ levels measured in brain homogenates. 
  •  
7.
  • Morrison, Jamie I., et al. (författare)
  • A single-chain fragment constant design enables easy production of a monovalent blood-brain barrier transporter and provides an improved brain uptake at elevated doses
  • 2023
  • Ingår i: Journal of Neurochemistry. - : John Wiley & Sons. - 0022-3042 .- 1471-4159. ; 165:3, s. 413-425
  • Tidskriftsartikel (refereegranskat)abstract
    • The interest for developing antibody-driven therapeutic interventions has exponentially grown over the last few decades. Even though there have been promising leaps in the development of efficacious antibody therapies, problems revolving around production and site-directed delivery of these large macromolecules persist. This is especially pertinent when it comes to designing and producing antibodies to penetrate the blood-brain barrier (BBB) to tackle neurodegenerative diseases. One of the most effective approaches to alleviating this problem is to employ a "Trojan Horse " approach, using receptor-mediated transcytosis, such as those governed by the transferrin receptor (TfR)-mediated pathways, to deliver large protein payloads into the brain. Even though this method is effective, ideal limiting factors, related to how the antibody binds to the TfR, need to be elucidated to improve BBB penetrance. With this said, we have designed and produced a single-chain Fc antibody, conjugated to an scFv8D3 TfR binding motif, creating a single-chain monovalent BBB transporter (scFc-scFv8D3). This recombinant protein is easy to produce and purify, demonstrates monovalent binding to the TfR and is structurally stable at physiologically relevant temperatures. Using an in vitro BBB model system, we show a positive correlation between the concentration of administered antibody and transcytosis efficacy, with scFc-scFv8D3 demonstrating significantly higher transcytosis levels compared with scFv8D3-conjugated bivalent antibodies at elevated administered concentrations. Furthermore, in vivo studies recapitulate the in vitro results, with the scFc-scFv8D3 demonstrating an elevated brain uptake at higher therapeutic doses in wild-type mice, comparable with that of the scFv8D3-conjugated bivalent antibody control. In addition, the half-life of the single-chain monovalent BBB transporter is comparable with that of standard IgG antibodies, indicating that the scFc format does not exacerbate physiological degradation. Our results lead us to the conclusion that valency and affinity are important variables to consider when discerning optimal transport across the BBB using TfR-mediated transcytosis pathways. In addition, we believe the single-chain Fc antibody we have described, which can easily be manipulated to accommodate a bispecific target tactic, provides a simple and efficacious approach for delivering therapeutic payloads to the brain milieu.
  •  
8.
  • Morrison, Jamie, et al. (författare)
  • Standardized Preclinical In Vitro Blood-Brain Barrier Mouse Assay Validates Endocytosis-Dependent Antibody Transcytosis Using Transferrin-Receptor-Mediated Pathways
  • 2023
  • Ingår i: Molecular Pharmaceutics. - : American Chemical Society (ACS). - 1543-8384 .- 1543-8392. ; 20:3, s. 1564-1576
  • Tidskriftsartikel (refereegranskat)abstract
    • The presence of the blood-brain barrier (BBB) creates a nigh-on impenetrable obstacle for large macromolecular therapeutics that need to be delivered to the brain milieu to treat neurological disorders. To overcome this, one of the strategies used is to bypass the barrier with what is referred to as a "Trojan Horse" strategy, where therapeutics are designed to use endogenous receptor-mediated pathways to piggyback their way through the BBB. Even though in vivo methodologies are commonly used to test the efficacy of BBB-penetrating biologics, comparable in vitro BBB models are in high demand, as they benefit from being an isolated cellular system devoid of physiological factors that can on occasion mask the processes behind BBB transport via transcytosis. We have developed an in vitro BBB model (In-Cell BBB-Trans assay) based on the murine cEND cells that help delineate the ability of modified large bivalent IgG antibodies conjugated to the transferrin receptor binder scFv8D3 to cross an endothelial monolayer grown on porous cell culture inserts (PCIs). Following the administration of bivalent antibodies into the endothelial monolayer, a highly sensitive enzyme-linked immunosorbent assay (ELISA) is used to determine the concentration in the apical (blood) and basolateral (brain) chambers of the PCI system, allowing for the evaluation of apical recycling and basolateral transcytosis, respectively. Our results show that antibodies conjugated to scFv8D3 transcytose at considerably higher levels compared to unconjugated antibodies in the In-Cell BBB-Trans assay. Interestingly, we are able to show that these results mimic in vivo brain uptake studies using identical antibodies. In addition, we are able to transversely section PCI cultured cells, allowing for the identification of receptors and proteins that are likely involved in the transcytosis of the antibodies. Furthermore, studies using the In-Cell BBB-Trans assay revealed that transcytosis of the transferrin-receptor-targeting antibodies is dependent on endocytosis. In conclusion, we have designed a simple, reproducible In-Cell BBB-Trans assay based on murine cells that can be used to rapidly determine the BBB-penetrating capabilities of transferrin-receptor-targeting antibodies. We believe that the In-Cell BBB-Trans assay can be used as a powerful, preclinical screening platform for therapeutic neurological pathologies.
  •  
9.
  • Napoleone, Antonino, et al. (författare)
  • Fed-batch production assessment of a tetravalent bispecific antibody : A case study on piggyBac stably transfected HEK293 cells
  • 2021
  • Ingår i: New Biotechnology. - : Elsevier BV. - 1871-6784 .- 1876-4347. ; 65, s. 9-19
  • Tidskriftsartikel (refereegranskat)abstract
    • The transition from preclinical biological drug development into clinical trials requires an efficient upscaling process. In this context, bispecific antibody drugs are particularly challenging due to their propensity to form aggregates and generally produce low titers. Here, the upscaling process for a tetravalent bispecific antibody expressed by a piggyBac transposon-mediated stable HEK293 cell pool has been evaluated. The project was performed as a case study at Testa Center, a non-GMP facility for scale-up testing of biologics in Sweden, and encompassed media adaptation strategies, fed-batch optimization and a novel antibody purification technology. The cell pool was adapted to different culture media for evaluation in terms of cell viability and titers compared to its original Expi293 Expression Medium. These parameters were assessed in both sequential stepwise adaption and direct media exchanges. By this, a more affordable medium was identified that did not require stepwise adaptation and with similar titers and viability as in the Expi293 Expression Medium. Fed-batch optimizations resulted in culture densities reaching up to 20 x 106 viable cells/mL with over 90 % viability 12 days postinoculum, and antibody titers three times higher than corresponding batch cultures. By implementing a novel high-speed protein A fiber technology (Fibro PrismA) with a capture residence time of only 7.5 s, 8 L of supernatant could be purified in 4.5 h without compromising the purity, structural integrity and function of the bispecific antibody. Results from this study related to medium adaptation and design of fed-batch protocols will be highly beneficial during the forthcoming scale-up of this therapeutic antibody.
  •  
10.
  • Pagnon de la Vega, María, et al. (författare)
  • Altered amyloid-β structure markedly reduces gliosis in the brain of mice harboring the Uppsala APP deletion
  • 2024
  • Ingår i: Acta neuropathologica communications. - : BioMed Central (BMC). - 2051-5960. ; 12:1
  • Tidskriftsartikel (refereegranskat)abstract
    • 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.
  •  
Skapa referenser, mejla, bekava och länka
  • Resultat 1-10 av 20
Typ av publikation
tidskriftsartikel (17)
doktorsavhandling (3)
Typ av innehåll
refereegranskat (17)
övrigt vetenskapligt/konstnärligt (3)
Författare/redaktör
Hultqvist, Greta, 19 ... (18)
Syvänen, Stina (13)
Sehlin, Dag, 1976- (11)
Rofo, Fadi (10)
Metzendorf, Nicole G ... (8)
Petrovic, Alex (5)
visa fler...
Gustavsson, Tobias (4)
Ingelsson, Martin (3)
Morrison, Jamie (3)
Lannfelt, Lars (2)
Nilsson, Per (2)
Bergström, Joakim (2)
Erlandsson, Anna (2)
Yilmaz, Canan Ugur (2)
Mangsbo, Sara, 1981- (2)
Eriksson, Jonas (2)
Xiong, Mengfei (2)
de la Rosa, Andrés (2)
Lord, Martin (1)
Andrén, Per E., Prof ... (1)
Falk, Ronny (1)
Aguilar, Ximena (1)
Olsson, Anders (1)
Ekmark-Lewén, Sara (1)
Andersson, Andreas (1)
Andersson, Oskar (1)
Andersson, Ken G. (1)
O’Callaghan, Paul (1)
Persson, Helena (1)
Banka, Vinay (1)
Kelleher, Andrew (1)
Sigurdsson, Einar M (1)
Ding, Yu-Shin (1)
Buijs, Jos (1)
Hall, Martin (1)
Giedraitis, Vilmanta ... (1)
Beretta, Chiara (1)
Zachrisson, Olof (1)
Masliah, Eliezer (1)
Nilsson, Lars N G (1)
Dahllund, Leif (1)
Walsh, Frank S (1)
Bondza, Sina (1)
Rokka, Johanna (1)
Hultqvist, Greta, Do ... (1)
O'Callaghan, Paul, D ... (1)
Metzendorf, Nicole G ... (1)
Moos, Torben, Profes ... (1)
Faresjö, Rebecca, 19 ... (1)
Lauren, Ida (1)
visa färre...
Lärosäte
Uppsala universitet (20)
Kungliga Tekniska Högskolan (1)
Karolinska Institutet (1)
Språk
Engelska (20)
Forskningsämne (UKÄ/SCB)
Medicin och hälsovetenskap (17)
Naturvetenskap (3)

År

Kungliga biblioteket hanterar dina personuppgifter i enlighet med EU:s dataskyddsförordning (2018), GDPR. Läs mer om hur det funkar här.
Så här hanterar KB dina uppgifter vid användning av denna tjänst.

 
pil uppåt Stäng

Kopiera och spara länken för att återkomma till aktuell vy