kth.sePublications
Change search
Link to record
Permanent link

Direct link
Publications (10 of 30) Show all publications
Mortensen, A. C., Hofström, C., Persson, H., Dahllund, L., Frejd, F. Y. & Nestor, M. (2025). Affinity maturation and optimization of CD44v6-targeting antibodies for molecular radiotherapy. Nuclear Medicine and Biology, 144, Article ID 109012.
Open this publication in new window or tab >>Affinity maturation and optimization of CD44v6-targeting antibodies for molecular radiotherapy
Show others...
2025 (English)In: Nuclear Medicine and Biology, ISSN 0969-8051, E-ISSN 1872-9614, Vol. 144, article id 109012Article in journal (Refereed) Published
Abstract [en]

Aim: This study aimed to improve the efficacy of the CD44v6-targeting antibody UU-40 for molecular radiotherapy through affinity maturation and IgG subclass optimization. M&M: A panel of affinity-matured antibody candidates was generated and characterized as both human IgG4 and IgG1 with LALA mutations. Surface plasmon resonance and LigandTracer analyses identified several candidates with superior affinity and off-rates compared to the parental UU-40. Biodistribution studies in xenograft models using Lutetium-177 (177Lu)-labeled antibodies showed improved tumor retention for selected candidates, particularly UU-A-155. Species cross-reactivity assays confirmed binding to cynomolgus and rabbit v6-peptides, supporting future toxicity studies. Results: The IgG1 LALA format demonstrated reduced binding to Fc gamma receptors, potentially improving the safety profile. UU-A-155 emerged as the lead candidate for clinical translation, showing superior performance in both affinity and tumor retention. Our findings highlight the importance of comprehensive in vitro and in vivo assessments in antibody development, and provides valuable insights into optimizing antibody-based molecular radiotherapy.

Place, publisher, year, edition, pages
Elsevier BV, 2025
Keywords
CD44v6, Antibody maturation, Fc gamma-silencing, Molecular radiotherapy, IgG1 LALA, Epitope mapping
National Category
Immunology
Identifiers
urn:nbn:se:kth:diva-364241 (URN)10.1016/j.nucmedbio.2025.109012 (DOI)001476578600001 ()40253776 (PubMedID)2-s2.0-105002768929 (Scopus ID)
Note

QC 20250609

Available from: 2025-06-09 Created: 2025-06-09 Last updated: 2025-06-09Bibliographically approved
Mebrahtu, A., Laurén, I., Veerman, R., Akpinar, G. G., Lord, M., Kostakis, A., . . . Mangsbo, S. (2024). A bispecific CD40 agonistic antibody allowing for antibody-peptide conjugate formation to enable cancer-specific peptide delivery, resulting in improved T proliferation and anti-tumor immunity in mice. Nature Communications, 15(1), Article ID 9542.
Open this publication in new window or tab >>A bispecific CD40 agonistic antibody allowing for antibody-peptide conjugate formation to enable cancer-specific peptide delivery, resulting in improved T proliferation and anti-tumor immunity in mice
Show others...
2024 (English)In: Nature Communications, E-ISSN 2041-1723, Vol. 15, no 1, article id 9542Article in journal (Refereed) Published
Abstract [en]

Current antibody-based immunotherapy depends on tumor antigen shedding for proper T cell priming. Here we select a novel human CD40 agonistic drug candidate and generate a bispecific antibody, herein named BiA9*2_HF, that allows for rapid antibody-peptide conjugate formation. The format is designed to facilitate peptide antigen delivery to CD40 expressing cells combined with simultaneous CD40 agonistic activity. In vivo, the selected bispecific antibody BiA9*2_HF loaded with peptide cargos induces improved antigen-specific proliferation of CD8+ (10-15 fold) and CD4+ T cells (2-7 fold) over control in draining lymph nodes. In both virus-induced and neoantigen-based mouse tumor models, BiA9*2_HF demonstrates therapeutic efficacy and elevated safety profile, with complete tumor clearance, as well as measured abscopal impact on tumor growth. The BiA9*2_HF drug candidate can thus be utilized to tailor immunotherapeutics for cancer patients.

Place, publisher, year, edition, pages
Nature Research, 2024
National Category
Immunology in the medical area Biochemistry Molecular Biology Medical Biotechnology (with a focus on Cell Biology (including Stem Cell Biology), Molecular Biology, Microbiology, Biochemistry or Biopharmacy) Cancer and Oncology
Identifiers
urn:nbn:se:kth:diva-356688 (URN)10.1038/s41467-024-53839-5 (DOI)001348514000014 ()39500897 (PubMedID)2-s2.0-85208602407 (Scopus ID)
Note

QC 20241122

Available from: 2024-11-20 Created: 2024-11-20 Last updated: 2025-02-20Bibliographically approved
Thalén, N., Karlander, M., Lundqvist, M., Persson, H., Hofström, C., Turunen, S. P., . . . Rockberg, J. (2024). Mammalian cell display with automated oligo design and library assembly allows for rapid residue level conformational epitope mapping. Communications Biology, 7(1), Article ID 805.
Open this publication in new window or tab >>Mammalian cell display with automated oligo design and library assembly allows for rapid residue level conformational epitope mapping
Show others...
2024 (English)In: Communications Biology, E-ISSN 2399-3642, Vol. 7, no 1, article id 805Article in journal (Refereed) Published
Abstract [en]

Precise epitope determination of therapeutic antibodies is of great value as it allows for further comprehension of mechanism of action, therapeutic responsiveness prediction, avoidance of unwanted cross reactivity, and vaccine design. The golden standard for discontinuous epitope determination is the laborious X-ray crystallography method. Here, we present a combinatorial method for rapid mapping of discontinuous epitopes by mammalian antigen display, eliminating the need for protein expression and purification. The method is facilitated by automated workflows and tailored software for antigen analysis and oligonucleotide design. These oligos are used in automated mutagenesis to generate an antigen receptor library displayed on mammalian cells for direct binding analysis by flow cytometry. Through automated analysis of 33930 primers an optimized single condition cloning reaction was defined allowing for mutation of all surface-exposed residues of the receptor binding domain of SARS-CoV-2. All variants were functionally expressed, and two reference binders validated the method. Furthermore, epitopes of three novel therapeutic antibodies were successfully determined followed by evaluation of binding also towards SARS-CoV-2 Omicron BA.2. We find the method to be highly relevant for rapid construction of antigen libraries and determination of antibody epitopes, especially for the development of therapeutic interventions against novel pathogens.

Place, publisher, year, edition, pages
Springer Nature, 2024
National Category
Biochemistry Molecular Biology
Identifiers
urn:nbn:se:kth:diva-350701 (URN)10.1038/s42003-024-06508-8 (DOI)001262592300006 ()38961245 (PubMedID)2-s2.0-85197485847 (Scopus ID)
Note

QC 20240719

Available from: 2024-07-17 Created: 2024-07-17 Last updated: 2025-02-20Bibliographically approved
Sahi, M., Andersson, S., Mattsson, C., Dale, M., Kagiolglou, S., Hofström, C., . . . Fredolini, C. (2024). Profiling of Surface Protein Epitopes on Viral Particles by Multiplex Dual-Reporter Strategy. Journal of Visualized Experiments, 2024(203), Article ID e66230.
Open this publication in new window or tab >>Profiling of Surface Protein Epitopes on Viral Particles by Multiplex Dual-Reporter Strategy
Show others...
2024 (English)In: Journal of Visualized Experiments, E-ISSN 1940-087X, Vol. 2024, no 203, article id e66230Article in journal (Refereed) Published
Abstract [en]

Membrane proteins on enveloped viruses play an important role in many biological functions involving virus attachment to target cell receptors, fusion of viral particles to host cells, host-virus interactions, and disease pathogenesis. Furthermore, viral membrane proteins on virus particles and presented on host cell surfaces have proven to be excellent targets for antivirals and vaccines. Here, we describe a protocol to investigate surface proteins on intact severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) particles using the dual-reporter flow cytometric system. The assay exploits multiplex technology to obtain a triple detection of viral particles by three independent affinity reactions. Magnetic beads conjugated to recombinant human angiotensin-converting enzyme-2 (ACE2) were used to capture viral particles from the supernatant of cells infected with SARS-CoV-2. Then, two detection reagents labeled with R-phycoerythrin (PE) or Brilliant Violet 421 (BV421) were applied simultaneously. As a proof-of-concept, antibody fragments targeting different epitopes of the SARS CoV-2 surface protein Spike (S1) were used. The detection of viral particles by three independent affinity reactions provides strong specificity and confirms the capture of intact virus particles. Dose-dependency curves of SARS-CoV-2 infected cell supernatant were generated with replicate coefficient variances (mean/SD) ˂14%. Good assay performance in both channels confirmed that two virus surface target protein epitopes are detectable in parallel. The protocol described here could be applied for (i) high-multiplex, high-throughput profiling of surface proteins expressed on enveloped viruses; ii) detection of active intact viral particles; and (iii) assessment of specificity and affinity of antibodies and antiviral drugs for surface epitopes of viral antigens.The application can be potentially extended to any type of extracellular vesicles and bioparticles, exposing surface antigens in body fluids or other liquid matrices.

Place, publisher, year, edition, pages
MyJove Corporation, 2024
National Category
Biochemistry Molecular Biology
Identifiers
urn:nbn:se:kth:diva-343495 (URN)10.3791/66230 (DOI)38284526 (PubMedID)2-s2.0-85183777150 (Scopus ID)
Note

QC 20240215

Available from: 2024-02-15 Created: 2024-02-15 Last updated: 2025-02-20Bibliographically approved
Preger, C., Notarnicola, A., Hellström, C., Wigren, E., Fernandes-Cerqueira, C., Kvarnstrom, M., . . . Jakobsson, P.-J. (2023). Autoantigenic properties of the aminoacyl tRNA synthetase family in idiopathic inflammatory myopathies. Journal of Autoimmunity, 134, Article ID 102951.
Open this publication in new window or tab >>Autoantigenic properties of the aminoacyl tRNA synthetase family in idiopathic inflammatory myopathies
Show others...
2023 (English)In: Journal of Autoimmunity, ISSN 0896-8411, E-ISSN 1095-9157, Vol. 134, article id 102951Article in journal (Refereed) Published
Abstract [en]

Objectives: Autoantibodies are thought to play a key role in the pathogenesis of idiopathic inflammatory my-opathies (IIM). However, up to 40% of IIM patients, even those with clinical manifestations of anti-synthetase syndrome (ASSD), test seronegative to known myositis-specific autoantibodies. We hypothesized the existence of new potential autoantigens among human cytoplasmic aminoacyl tRNA synthetases (aaRS) in patients with IIM.Methods: Plasma samples from 217 patients with IIM according to 2017 EULAR/ACR criteria, including 50 pa-tients with ASSD, 165 without, and two with unknown ASSD status were identified retrospectively, as well as age and gender-matched sera from 156 population controls, and 219 disease controls. Patients with previously documented ASSD had to test positive for at least one of the five most common anti-aaRS autoantibodies (anti-Jo1,-PL7,-PL12,-EJ, and-OJ) and present with one or more of the following clinical manifestations: interstitial lung disease, myositis, arthritis, Raynaud's phenomenon, fever, or mechanic's hands. Demographics, laboratory, and clinical data of the IIM cohort (ASSD and non-ASSD) were compared. Samples were screened using a multiplex bead array assay for presence of autoantibodies against a panel of 117 recombinant protein variants, representing 33 myositis-related proteins, including all nineteen cytoplasmic aaRS. Prospectively collected clinical data for the IIM cohort were retrieved and compared between groups within the IIM cohort and correlated with the results of the autoantibody screening. Principal component analysis was used to analyze clinical manifestations between ASSD, non-ASSD groups, and individuals with novel anti-aaRS autoantibodies. Results: We identified reactivity towards 16 aaRS in 72 of the 217 IIM patients. Twelve patients displayed reactivity against nine novel aaRS. The novel autoantibody specificities were detected in four previously sero-negative patients for myositis-specific autoantibodies and eight with previously detected myositis-specific au-toantibodies. IIM individuals with novel anti-aaRS autoantibodies (n = 12) all had signs of myositis, and they had either muscle weakness and/or muscle enzyme elevation, 2/12 had mechanic's hands, 3/12 had interstitial lung disease, and 2/12 had arthritis. The individuals with novel anti-aaRS and a pathological muscle biopsy all presented widespread up-regulation of major histocompatibility complex class I. The reactivities against novel aaRS could be confirmed in ELISA and western blot. Using the multiplex bead array assay, we could confirm previously known reactivities to four of the most common aaRS (Jo1, PL12, PL7, and EJ (n = 45)) and identified patients positive for anti-Zo,-KS, and-HA (n = 10) that were not previously tested. A low frequency of anti-aaRS autoantibodies was also detected in controls.Conclusion: Our results suggest that most, if not all, cytoplasmic aaRS may become autoantigenic. Autoantibodies against new aaRS may be found in plasma of patients previously classified as seronegative with potential high clinical relevance.

Place, publisher, year, edition, pages
Elsevier BV, 2023
Keywords
Autoantibodies, Anti-synthetase syndrome, anti-aaRS, Idiopathic inflammatory myopathies, Aminoacyl tRNA synthetases
National Category
Clinical Medicine
Identifiers
urn:nbn:se:kth:diva-323097 (URN)10.1016/j.jaut.2022.102951 (DOI)000897623600003 ()36470210 (PubMedID)2-s2.0-85143314752 (Scopus ID)
Note

QC 20230118

Available from: 2023-01-18 Created: 2023-01-18 Last updated: 2025-02-18Bibliographically approved
Sarén, T., Saronio, G., Marti Torrell, P., Zhu, X., Thelander, J., Andersson, Y., . . . Essand, M. (2023). Complementarity-determining region clustering may cause CAR-T cell dysfunction. Nature Communications, 14(1), 4732
Open this publication in new window or tab >>Complementarity-determining region clustering may cause CAR-T cell dysfunction
Show others...
2023 (English)In: Nature Communications, E-ISSN 2041-1723, Vol. 14, no 1, p. 4732-Article in journal (Refereed) Published
Abstract [en]

Chimeric antigen receptor (CAR)-T cell therapy is rapidly advancing as cancer treatment, however, designing an optimal CAR remains challenging. A single-chain variable fragment (scFv) is generally used as CAR targeting moiety, wherein the complementarity-determining regions (CDRs) define its specificity. We report here that the CDR loops can cause CAR clustering, leading to antigen-independent tonic signalling and subsequent CAR-T cell dysfunction. We show via CARs incorporating scFvs with identical framework and varying CDR sequences that CARs may cluster on the T cell surface, which leads to antigen-independent CAR-T cell activation, characterized by increased cell size and interferon (IFN)-γ secretion. This results in CAR-T cell exhaustion, activation-induced cell death and reduced responsiveness to target-antigen-expressing tumour cells. CDR mutagenesis confirms that the CAR-clustering is mediated by CDR-loops. In summary, antigen-independent tonic signalling can be induced by CDR-mediated CAR clustering, which could not be predicted from the scFv sequences, but could be tested for by evaluating the activity of unstimulated CAR-T cells.

Place, publisher, year, edition, pages
Springer Nature, 2023
National Category
Immunology in the medical area
Identifiers
urn:nbn:se:kth:diva-334796 (URN)10.1038/s41467-023-40303-z (DOI)001050351300037 ()37563127 (PubMedID)2-s2.0-85167700249 (Scopus ID)
Note

QC 20240326

Available from: 2023-09-01 Created: 2023-09-01 Last updated: 2024-04-05Bibliographically approved
Hueting, D. A., Schriever, K., Sun, R., Vlachiotis, S., Zuo, F., Du, L., . . . Syrén, P.-O. (2023). Design, structure and plasma binding of ancestral β-CoV scaffold antigens. Nature Communications, 14(1), Article ID 6527.
Open this publication in new window or tab >>Design, structure and plasma binding of ancestral β-CoV scaffold antigens
Show others...
2023 (English)In: Nature Communications, E-ISSN 2041-1723, Vol. 14, no 1, article id 6527Article in journal (Refereed) Published
Abstract [en]

We report the application of ancestral sequence reconstruction on coronavirus spike protein, resulting in stable and highly soluble ancestral scaffold antigens (AnSAs). The AnSAs interact with plasma of patients recovered from COVID-19 but do not bind to the human angiotensin-converting enzyme 2 (ACE2) receptor. Cryo-EM analysis of the AnSAs yield high resolution structures (2.6-2.8 angstrom) indicating a closed pre-fusion conformation in which all three receptor-binding domains (RBDs) are facing downwards. The structures reveal an intricate hydrogen-bonding network mediated by well-resolved loops, both within and across monomers, tethering the N-terminal domain and RBD together. We show that AnSA-5 can induce and boost a broad-spectrum immune response against the wild-type RBD as well as circulating variants of concern in an immune organoid model derived from tonsils. Finally, we highlight how AnSAs are potent scaffolds by replacing the ancestral RBD with the wild-type sequence, which restores ACE2 binding and increases the interaction with convalescent plasma. Development of vaccines remains challenging because viral antigens can be unstable or aggregate. Here, authors present ancestral sequence reconstruction as a method to generate stable and soluble antigens using exclusively available sequence information.

Place, publisher, year, edition, pages
Springer Nature, 2023
National Category
Biochemistry Molecular Biology
Identifiers
urn:nbn:se:kth:diva-343082 (URN)10.1038/s41467-023-42200-x (DOI)001142518400009 ()37845250 (PubMedID)2-s2.0-85174288771 (Scopus ID)
Note

QC 20240206

Available from: 2024-02-06 Created: 2024-02-06 Last updated: 2025-02-20Bibliographically approved
Mortensen, A. C., Berglund, H., Segerström, L., Walle, M., Hofström, C., Persson, H., . . . Nestor, M. (2023). Selection, characterization and in vivo evaluation of novel CD44v6-targeting antibodies for targeted molecular radiotherapy. Scientific Reports, 13(1), Article ID 20648.
Open this publication in new window or tab >>Selection, characterization and in vivo evaluation of novel CD44v6-targeting antibodies for targeted molecular radiotherapy
Show others...
2023 (English)In: Scientific Reports, E-ISSN 2045-2322, Vol. 13, no 1, article id 20648Article in journal (Refereed) Published
Abstract [en]

Molecular radiotherapy combines the advantages of systemic administration of highly specific antibodies or peptides and the localized potency of ionizing radiation. A potential target for molecular radiotherapy is the cell surface antigen CD44v6, which is overexpressed in numerous cancers, with limited expression in normal tissues. The aim of the present study was to generate and characterize a panel of human anti-CD44v6 antibodies and identify a suitable candidate for future use in molecular radiotherapy of CD44v6-expressing cancers. Binders were first isolated from large synthetic phage display libraries containing human scFv and Fab antibody fragments. The antibodies were extensively analyzed through in vitro investigations of binding kinetics, affinity, off-target binding, and cell binding. Lead candidates were further subjected to in vivo biodistribution studies in mice bearing anaplastic thyroid cancer xenografts that express high levels of CD44v6. Additionally, antigen-dependent tumor uptake of the lead candidate was verified in additional xenograft models with varying levels of target expression. Interestingly, although only small differences were observed among the top antibody candidates in vitro, significant differences in tumor uptake and retention were uncovered in in vivo experiments. A high-affinity anti-CD44v6 lead drug candidate was identified, mAb UU-40, which exhibited favorable target binding properties and in vivo distribution. In conclusion, a panel of human anti-CD44v6 antibodies was successfully generated and characterized in this study. Through comprehensive evaluation, mAb UU-40 was identified as a promising lead candidate for future molecular radiotherapy of CD44v6-expressing cancers due to its high affinity, excellent target binding properties, and desirable in vivo distribution characteristics.

Place, publisher, year, edition, pages
Springer Nature, 2023
National Category
Cancer and Oncology Radiology, Nuclear Medicine and Medical Imaging
Identifiers
urn:nbn:se:kth:diva-340841 (URN)10.1038/s41598-023-47891-2 (DOI)001136085000078 ()38001360 (PubMedID)2-s2.0-85177659141 (Scopus ID)
Note

QC 20231218

Available from: 2023-12-18 Created: 2023-12-18 Last updated: 2024-02-06Bibliographically approved
Preger, C., Notarnicola, A., Hellström, C., Wigren, E., Lundberg, I. E., Jakobsson, P. J., . . . Gräslund, S. (2022). Abundant autoantibody isotypes in idiopathic inflammatory myopathies. Annals of the Rheumatic Diseases, 81(Suppl 1), 242.2-243
Open this publication in new window or tab >>Abundant autoantibody isotypes in idiopathic inflammatory myopathies
Show others...
2022 (English)In: Annals of the Rheumatic Diseases, ISSN 0003-4967, E-ISSN 1468-2060, Vol. 81, no Suppl 1, p. 242.2-243Article in journal, Meeting abstract (Other academic) Published
Place, publisher, year, edition, pages
BMJ, 2022
National Category
Clinical Medicine
Identifiers
urn:nbn:se:kth:diva-321038 (URN)10.1136/annrheumdis-2022-eular.1980 (DOI)000850279000370 ()
Note

QC 20230404

Available from: 2022-11-04 Created: 2022-11-04 Last updated: 2025-02-18Bibliographically approved
Eltahir, M., Laurén, I., Lord, M., Chourlia, A., Dahllund, L., Olsson, A., . . . Mangsbo, S. M. (2022). An Adaptable Antibody-Based Platform for Flexible Synthetic Peptide Delivery Built on Agonistic CD40 Antibodies. Advanced Therapeutics, 5(7), Article ID 2200008.
Open this publication in new window or tab >>An Adaptable Antibody-Based Platform for Flexible Synthetic Peptide Delivery Built on Agonistic CD40 Antibodies
Show others...
2022 (English)In: Advanced Therapeutics, E-ISSN 2366-3987, Vol. 5, no 7, article id 2200008Article in journal (Refereed) Published
Abstract [en]

The agonistic potentials of therapeutic anti-CD40 antibodies have been profiled in relation to antibody isotype and epitope specificity. Still, clinical impact relies on a well-balanced clinical efficacy versus target-mediated toxicity. As CD40-mediated immune activation must rely on a combination of stimulation of antigen-presenting cells (APCs) alongside antigen presentation, for efficient T cell priming, alternative approaches to improve the therapeutic outcome of CD40-targeting strategies should focus on providing optimal antigen presentation together with CD40 stimulation. Herein, a bispecific antibody targeting CD40 as a means to deliver cargo (i.e., synthetic peptides) into APCs through a non-covalent, high-affinity interaction between the antibody and the cargo peptide, further referred to as the Adaptable Drug Affinity Conjugate (ADAC) technology, has been developed. The ADAC platform demonstrated a target-specific CD4+ and CD8+ T cell expansion in vitro and significantly improved peptide-specific CD8+ T cell proliferation in vivo. In addition, the strategy dramatically improved the in vitro and in vivo half-life of the synthetic peptides. Future applications of ADAC involve pandemic preparedness to viral genetic drift as well as neoepitope vaccination strategies where the bispecific antibody is an off-the-shelf product, and the peptide antigen is synthesized based on next-generation sequencing data mining. 

Place, publisher, year, edition, pages
Wiley, 2022
Keywords
Antibody Drug Affinity Conjugate (ADAC), cancer vaccine, cargo delivery, CD40, immunotherapy, multivalent antibodies, synthetic peptides, antibody conjugate, bispecific antibody, CD40 ligand monoclonal antibody, Fc receptor, immunoglobulin G1, immunoglobulin G2, interleukin 12p40, interleukin 12p70, synthetic peptide, adult, animal cell, animal tissue, antigen binding, antigen presenting cell, Article, binding affinity, CD4+ T lymphocyte, CD8+ T lymphocyte, cell differentiation, cell expansion, controlled study, drug degradation, drug delivery system, drug half life, drug tissue level, embryo, female, genetic drift, high throughput sequencing, human, human cell, in vitro study, in vivo study, internalization (cell), lymphocyte proliferation, male, monocyte, mouse, natural killer cell, nonhuman, pandemic, peripheral blood mononuclear cell, protein stability, T lymphocyte activation, vaccination
National Category
Immunology in the medical area
Identifiers
urn:nbn:se:kth:diva-324571 (URN)10.1002/adtp.202200008 (DOI)000810335300001 ()2-s2.0-85131734589 (Scopus ID)
Note

QC 20230308

Available from: 2023-03-08 Created: 2023-03-08 Last updated: 2023-03-08Bibliographically approved
Organisations
Identifiers
ORCID iD: ORCID iD iconorcid.org/0000-0002-2965-0395

Search in DiVA

Show all publications