kth.sePublications
Change search
Link to record
Permanent link

Direct link
Publications (10 of 86) Show all publications
Rönning, S., Thorell, H., Malm, M. & Rockberg, J. (2025). Automated plasmid purification can reducemanual labour and increase titres in AAV production. Human Gene Therapy, 36(3-4), E179-E180
Open this publication in new window or tab >>Automated plasmid purification can reducemanual labour and increase titres in AAV production
2025 (English)In: Human Gene Therapy, ISSN 1043-0342, E-ISSN 1557-7422, Vol. 36, no 3-4, p. E179-E180Article in journal, Meeting abstract (Other academic) Published
Place, publisher, year, edition, pages
Mary Ann Liebert, 2025
National Category
Medical Biotechnology (Focus on Cell Biology, (incl. Stem Cell Biology), Molecular Biology, Microbiology, Biochemistry or Biopharmacy)
Identifiers
urn:nbn:se:kth:diva-366120 (URN)001418811501111 ()
Note

QC 20250703

Available from: 2025-07-03 Created: 2025-07-03 Last updated: 2025-07-03Bibliographically approved
Masson, H. O., Di Giusto, P., Kuo, C. C., Malm, M., Lundqvist, M., Sivertsson, Å., . . . Lewis, N. E. (2025). Deciphering the determinants of recombinant protein expression across the human secretome. Proceedings of the National Academy of Sciences of the United States of America, 122(41)
Open this publication in new window or tab >>Deciphering the determinants of recombinant protein expression across the human secretome
Show others...
2025 (English)In: Proceedings of the National Academy of Sciences of the United States of America, ISSN 0027-8424, E-ISSN 1091-6490, Vol. 122, no 41Article in journal (Refereed) Published
Abstract [en]

Protein secretion is an essential process of mammalian cells. In biomanufacturing, this process can be optimized to enhance production yields and biotherapeutic quality. While cell line engineering and bioprocess optimization have yielded high protein titers for some recombinant proteins, many remain difficult to express. Here, we investigated factors influencing protein expression in Chinese hamster ovary (CHO) cells, expressing 2,135 Human Secretome Project proteins. While the abundance of mRNA from recombinant proteins explained less than 1% of observed variation in secretion titers, analysis of 218 biochemical and biophysical descriptors uncovered intrinsic protein features that account for ~15% of secretion variability, pinpointing key drivers such as molecular weight, cysteine content, and N-linked glycosylation, and establishing a roadmap for rational design of difficult-to-express proteins. We subsequently analyzed RNA-Seq data from 95 CHO cell cultures, each expressing a distinct recombinant protein, spanning a wide range of titers. Host cell transcriptomic signatures showed strong correlations with titer, thereby providing insights into cellular processes that covary with expression. Cells failing to produce proteins exhibited increased ubiquitin-mediated proteasomal degradation, including ER-associated degradation; whereas high-producing cells demonstrated enhanced lipid metabolism and a stronger response to oxidative stress, suggesting these factors may support successful recombinant protein productions. Together, using this resource, we quantified the contributions of various protein and cellular factors that correlate with the expression of diverse recombinant human proteins in a heterologous host, thereby providing insights for next-generation CHO cell engineering.

Place, publisher, year, edition, pages
Proceedings of the National Academy of Sciences, 2025
Keywords
Chinese hamster ovary cells, machine learning, protein secretion, recombinant protein, transcriptomics
National Category
Molecular Biology Bioprocess Technology Medical Biotechnology (Focus on Cell Biology, (incl. Stem Cell Biology), Molecular Biology, Microbiology, Biochemistry or Biopharmacy)
Identifiers
urn:nbn:se:kth:diva-372360 (URN)10.1073/pnas.2506036122 (DOI)41055974 (PubMedID)2-s2.0-105017946891 (Scopus ID)
Note

QC 20251106

Available from: 2025-11-06 Created: 2025-11-06 Last updated: 2025-11-06Bibliographically approved
Möller, M., Jönsson, M., Lundqvist, M., Rockberg, J., Löfblom, J., Tegel, H. & Hober, S. (2025). Deep sequencing combined with high-throughput screening enables efficient development of a pH-dependent high-affinity binding domain targeting HER3. Protein Science, 34(8), Article ID e70247.
Open this publication in new window or tab >>Deep sequencing combined with high-throughput screening enables efficient development of a pH-dependent high-affinity binding domain targeting HER3
Show others...
2025 (English)In: Protein Science, ISSN 0961-8368, E-ISSN 1469-896X, Vol. 34, no 8, article id e70247Article in journal (Refereed) Published
Abstract [en]

In vitro methods for developing binding domains have been well-established for many years, owing to the cost-efficient synthesis of DNA and high-throughput selection and screening technologies. However, generating high-affinity binding domains often requires the development of focused maturation libraries for a second selection, which typically demands a detailed understanding of the binding surfaces from the initial selection, a process that can be time-consuming. In this study, we accelerated this process by using deep sequencing data from the first selection to guide the design of the maturation library. Additionally, we employed a high-throughput screening system using flow cytometry based on Escherichia coli display to identify conditional binding domains from the selection output. This approach enabled the development of a high-affinity binder targeting the cancer biomarker HER3, with a binding affinity of 3.3 nM at extracellular pH 7.4, 100 times higher than the first-generation binding domain. Notably, the binding domain features a pH-dependent release mechanism, enabling rapid release in slightly acidic environments (pH ≈6), which resemble endosomal conditions. When conjugated to the cytotoxin mertansine (DM1), the binding domain demonstrated specific cytotoxic activity against HER3-expressing cell lines, with an IC50 of 2–5 nM. The presented approach enables the efficient development of conditional binding domains which hold promise for therapeutic applications.

Place, publisher, year, edition, pages
Wiley, 2025
Keywords
calcium-regulated affinity, cancer, cell display screening, conditional targeting, deep sequencing, drug conjugate, endosomal release, HER3
National Category
Molecular Biology
Identifiers
urn:nbn:se:kth:diva-368575 (URN)10.1002/pro.70247 (DOI)001536267500001 ()40716110 (PubMedID)2-s2.0-105011861520 (Scopus ID)
Note

QC 20250820

Available from: 2025-08-20 Created: 2025-08-20 Last updated: 2025-10-21Bibliographically approved
Zhang, Y., Sundäng Peters, E., Daramola, O., Tucker, T. A., Rockberg, J., Hatton, D. & Chotteau, V. (2025). Intensification of rAAV Production Based on HEK293 Cell Transient Transfection. Biotechnology Journal, 20(6), Article ID e70020.
Open this publication in new window or tab >>Intensification of rAAV Production Based on HEK293 Cell Transient Transfection
Show others...
2025 (English)In: Biotechnology Journal, ISSN 1860-6768, E-ISSN 1860-7314, Vol. 20, no 6, article id e70020Article in journal (Refereed) Published
Abstract [en]

Recombinant adeno-associated virus (rAAV) vectors are widely used in gene therapies, but the rapidly increasing global demand has created a significant challenge for rAAV manufacturing, where production capacity remains a critical bottleneck. To address this, strategies to enhance production yields are urgently needed. This study presents an innovative approach to rAAV production using high cell density (HCD) stirred tank perfusion culture. rAAV1 and rAAV9 vectors carrying GFP cargo were used as models, with triple-plasmid transfection performed in suspension HEK293 cells at a high viable cell density of 50 million cells/mL in culture then maintained at >= 30 million cells/mL throughout production. Transfection and production parameters were first optimized in a 5 mL pseudo-perfusion spin tube screening system at HCD. A proof-of-concept was then demonstrated by scaling up to a 200 mL stirred tank bioreactor (STR) in perfusion mode. This intensified process achieved rAAV9 production levels per cell comparable to those observed in reference shake flask cultures at 1 million cells/mL. By implementing transfection at very HCD in a perfusion-based STR, this approach has the potential to significantly enhance rAAV volumetric production capacity, providing a promising solution to meet the growing demand for gene therapies.

Place, publisher, year, edition, pages
Wiley, 2025
Keywords
bioreactor, high cell density, PEI, perfusion, process intensification, rAAV, recombinant adeno-associated viral vectors, triple plasmid transient transfection
National Category
Medical Biotechnology
Identifiers
urn:nbn:se:kth:diva-367884 (URN)10.1002/biot.70020 (DOI)001505069200001 ()40491022 (PubMedID)2-s2.0-105007845889 (Scopus ID)
Note

QC 20250804

Available from: 2025-08-04 Created: 2025-08-04 Last updated: 2025-08-04Bibliographically approved
Thorell, H., Karlander, M., Thalén, N., Fresk, M., Villacañas González, M. d., Jung, T., . . . Rockberg, J. (2025). Selective DNA delivery through amodular AAV platform utilising Affibody binding domains. Human Gene Therapy, 36(3-4), E211-E212
Open this publication in new window or tab >>Selective DNA delivery through amodular AAV platform utilising Affibody binding domains
Show others...
2025 (English)In: Human Gene Therapy, ISSN 1043-0342, E-ISSN 1557-7422, Vol. 36, no 3-4, p. E211-E212Article in journal, Meeting abstract (Other academic) Published
Place, publisher, year, edition, pages
Mary Ann Liebert, 2025
National Category
Medical Biotechnology (Focus on Cell Biology, (incl. Stem Cell Biology), Molecular Biology, Microbiology, Biochemistry or Biopharmacy)
Identifiers
urn:nbn:se:kth:diva-366121 (URN)001418811501198 ()
Note

QC 20250707

Available from: 2025-07-07 Created: 2025-07-07 Last updated: 2025-07-07Bibliographically approved
Esnal, M. R., Arrías, P. N., Rockberg, J. & Malm, M. (2025). Understanding recombinant AAV production kinetics in HEK293 cells. Human Gene Therapy, 36(3-4), E189-E189
Open this publication in new window or tab >>Understanding recombinant AAV production kinetics in HEK293 cells
2025 (English)In: Human Gene Therapy, ISSN 1043-0342, E-ISSN 1557-7422, Vol. 36, no 3-4, p. E189-E189Article in journal, Meeting abstract (Other academic) Published
Place, publisher, year, edition, pages
Mary Ann Liebert, 2025
National Category
Medical Biotechnology (Focus on Cell Biology, (incl. Stem Cell Biology), Molecular Biology, Microbiology, Biochemistry or Biopharmacy)
Identifiers
urn:nbn:se:kth:diva-366119 (URN)001418811501138 ()
Note

QC 20250707

Available from: 2025-07-07 Created: 2025-07-07 Last updated: 2025-07-07Bibliographically 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
Mebrahtu, A., Aniander, G., Mega, A., Moradi, M., Thalén, N., Gudmundsdotter, L., . . . Rockberg, J. (2024). Co-culture platform for tuning of cancer receptor density allows for evaluation of bispecific immune cell engagers. New Biotechnology, 79, 120-126
Open this publication in new window or tab >>Co-culture platform for tuning of cancer receptor density allows for evaluation of bispecific immune cell engagers
Show others...
2024 (English)In: New Biotechnology, ISSN 1871-6784, E-ISSN 1876-4347, Vol. 79, p. 120-126Article in journal (Refereed) Published
Abstract [en]

Cancer immunotherapy, where a patient's immune system is harnessed to eradicate cancer cells selectively, is a leading strategy for cancer treatment. However, successes with immune checkpoint inhibitors (ICI) are hampered by reported systemic and organ-specific toxicities and by two-thirds of the patients being non-responders or subsequently acquiring resistance to approved ICIs. Hence substantial efforts are invested in discovering novel targeted immunotherapies aimed at reduced side-effects and improved potency. One way is utilizing the dual targeting feature of bispecific antibodies, which have made them increasingly popular for cancer immunotherapy. Easy and predictive screening methods for activation ranking of candidate drugs in tumor contra non-tumor environments are however lacking. Herein, we present a cell-based assay mimicking the tumor microenvironment by co-culturing B cells with engineered human embryonic kidney 293 T cells (HEK293T), presenting a controllable density of platelet-derived growth factor receptor β (PDGFRβ). A target density panel with three different surface protein levels on HEK293T cells was established by genetic constructs carrying regulatory elements limiting RNA translation of PDGFRβ. We employed a bispecific antibody-affibody construct called an AffiMab capable of binding PDGFRβ on cancer cells and CD40 expressed by B cells as a model. Specific activation of CD40-mediated signaling of immune cells was demonstrated with the two highest receptor-expressing cell lines, Level 2/3 and Level 4, while low-to-none in the low-expressing cell lines. The concept of receptor tuning and the presented co-culture protocol may be of general utility for assessing and developing novel bi-specific antibodies for immuno-oncology applications.

Place, publisher, year, edition, pages
Elsevier BV, 2024
Keywords
Expression tuning, Receptor density, Regulatory elements, Screening platform
National Category
Cancer and Oncology Immunology in the medical area
Identifiers
urn:nbn:se:kth:diva-342383 (URN)10.1016/j.nbt.2023.12.012 (DOI)38159596 (PubMedID)2-s2.0-85181763897 (Scopus ID)
Note

QC 20240122

Available from: 2024-01-17 Created: 2024-01-17 Last updated: 2024-05-21Bibliographically 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
Thalén, N., Moradi, M., Lundqvist, M., Rodhe, J., Andersson, M., Bidkhori, G., . . . Rockberg, J. (2024). Tuning of CHO secretional machinery improve activity of secreted therapeutic sulfatase 150-fold. Metabolic engineering, 81, 157-166
Open this publication in new window or tab >>Tuning of CHO secretional machinery improve activity of secreted therapeutic sulfatase 150-fold
Show others...
2024 (English)In: Metabolic engineering, ISSN 1096-7176, E-ISSN 1096-7184, Vol. 81, p. 157-166Article in journal (Refereed) Published
Abstract [en]

Rare diseases are, despite their name, collectively common and millions of people are affected daily of conditions where treatment often is unavailable. Sulfatases are a large family of activating enzymes related to several of these diseases. Heritable genetic variations in sulfatases may lead to impaired activity and a reduced macromolecular breakdown within the lysosome, with several severe and lethal conditions as a consequence. While therapeutic options are scarce, treatment for some sulfatase deficiencies by recombinant enzyme replacement are available. The recombinant production of such sulfatases suffers greatly from both low product activity and yield, further limiting accessibility for patient groups. To mitigate the low product activity, we have investigated cellular properties through computational evaluation of cultures with varying media conditions and comparison of two CHO clones with different levels of one active sulfatase variant. Transcriptome analysis identified 18 genes in secretory pathways correlating with increased sulfatase production. Experimental validation by upregulation of a set of three key genes improved the specific enzymatic activity at varying degree up to 150-fold in another sulfatase variant, broadcasting general production benefits. We also identified a correlation between product mRNA levels and sulfatase activity that generated an increase in sulfatase activity when expressed with a weaker promoter. Furthermore, we suggest that our proposed workflow for resolving bottlenecks in cellular machineries, to be useful for improvements of cell factories for other biologics as well.

Place, publisher, year, edition, pages
Elsevier BV, 2024
Keywords
CHO, Sulfatase, Systems biology, Transcriptomics
National Category
Medical Biotechnology (with a focus on Cell Biology (including Stem Cell Biology), Molecular Biology, Microbiology, Biochemistry or Biopharmacy)
Identifiers
urn:nbn:se:kth:diva-341758 (URN)10.1016/j.ymben.2023.12.003 (DOI)001138624600001 ()38081506 (PubMedID)2-s2.0-85179839715 (Scopus ID)
Note

QC 20240102

Available from: 2024-01-02 Created: 2024-01-02 Last updated: 2024-01-22Bibliographically approved
Organisations
Identifiers
ORCID iD: ORCID iD iconorcid.org/0000-0002-9977-5724

Search in DiVA

Show all publications