Abstract
The high cost and complexity of manufacturing recombinant adeno-associated virus vectors continue to limit the broader application of gene therapies, which offer life-changing potential for individuals affected by genetic diseases. Although stable producer cell lines represent a scalable and cost-effective alternative to transient transfection methods, their development is often delayed by inefficient selection strategies and extended timelines. In this study, we present a novel application of the glutamine synthetase-based selection system - commonly used in CHO cells - to a HeLaS3-based rAAV production platform. By generating glutamine synthetase-knockout HeLaS3 cells via CRISPR-Cas9 and applying glutamine deprivation under serum-free conditions, we significantly streamlined the PCL generation process, reducing the timeline to approximately two months while maintaining rAAV productivity (>1x1011 vg/mL) and product quality (~70% full capsids). This work establishes a robust and scalable workflow for rAAV manufacturing, with the potential to enhance accessibility and reduce viral vector production costs for applications in gene therapy.
Data availability
All research data and methods presented in the main and supplementary figures are available from the lead contact upon reasonable request. Correspondence and requests for materials should be directed to J.M.E. (jose.escandell@ibet.pt).
References
Zwi-Dantsis, L., Mohamed, S., Massaro, G. & Moeendarbary, E. Adeno-associated virus vectors: principles, practices, and prospects in gene therapy. Viruses 17(2), 239; https://doi.org/10.3390/v17020239 (2025).
Liu, Z. et al. The clinical safety landscape for ocular AAV gene therapies: a systematic review and meta-analysis. iScience 28, 112265; https://doi.org/10.1016/j.isci.2025.112265 (2025).
Reid, C. A., Hörer, M. & Mandegar, M. A. Advancing AAV production with high-throughput screening and transcriptomics. Cell Gene. Ther. Insights 10, 821–840; https://doi.org/10.18609/CGTI.2024.095 (2024).
Merten, O.-W. Development of stable packaging and producer cell lines for the production of AAV vectors. Microorganisms 12, 384; https://doi.org/10.3390/microorganisms12020384 (2024).
Escandell, J. M. et al. Leveraging rAAV bioprocess understanding and next generation bioanalytics development. Curr. Opin. Biotechnol. 74, 271–277; https://doi.org/10.1016/j.copbio.2021.12.009 (2022).
Clark, K. R., Voulgaropoulou, F., Fraley, D. M. & Johnson, P. R. Cell lines for the production of recombinant adeno-associated virus. Hum. Gene. Ther. 6, 1329–1341; https://doi.org/10.1089/hum.1995.6.10-1329 (1995).
Escandell, J. et al. Towards a scalable bioprocess for rAAV production using a HeLa stable cell line. Biotechnol. Bioeng. 120, 2578–2587; https://doi.org/10.1002/bit.28394 (2023).
Martin, J. et al. Generation and characterization of adeno-associated virus producer cell lines for research and preclinical vector production. Hum. Gene. Ther. Methods 24, 253–269; https://doi.org/10.1089/hgtb.2013.046 (2013).
Escandell, J. M. et al. Development of a serum-free producer cell line generation process for scalable and efficient rAAV production for gene therapy applications. bioRxiv. https://doi.org/10.1101/2025.10.02.679972 (2025).
Yu, D. Y., Lee, S. Y. & Lee, G. M. Glutamine synthetase gene knockout-human embryonic kidney 293E cells for stable production of monoclonal antibodies. Biotechnol. Bioeng. 115, 1367–1372; https://doi.org/10.1002/bit.26552 (2018).
Chin, C. L. et al. A human expression system based on HEK293 for the stable production of recombinant erythropoietin. Sci. Rep. 9, 16768; https://doi.org/10.1038/s41598-019-53391-z (2019).
Fan, L., Frye, C. C. & Racher, A. J. The use of glutamine synthetase as a selection marker: recent advances in Chinese hamster ovary cell line generation processes. Pharm. Bioprocess 1, 487–502; https://doi.org/10.4155/PBP.13.56 (2013).
Srivastava, A., Mallela, K. M. G., Deorkar, N. & Brophy, G. Manufacturing challenges and rational formulation development for AAV viral vectors. J. Pharm. Sci. 110, 2609–2624; https://doi.org/10.1016/j.xphs.2021.03.024 (2021).
Xue, W. et al. Adeno-associated virus perfusion enhanced expression: a commercially scalable, high titer, high quality producer cell line process. Mol. Ther. Methods Clin. Dev. 32, 101266; https://doi.org/10.1016/j.omtm.2024.101266 (2024).
Prasad, A. et al. Low-dose exposure to phytosynthesized gold nanoparticles combined with glutamine deprivation enhances cell death in the cancer cell line HeLa via oxidative stress-mediated mitochondrial dysfunction and G0/G1 cell cycle arrest. Nanoscale 14, 10399–10417; https://doi.org/10.1039/D2NR02150A (2022).
Reitzer, L. J., Wice, B. M. & Kennell, D. Evidence that glutamine, not sugar, is the major energy source for cultured HeLa cells. J. Biol. Chem. 254, 2669–76 (1979).
Srila, W. et al. Glutamine synthetase (GS) knockout (KO) using CRISPR/Cpf1 diversely enhances selection efficiency of CHO cells expressing therapeutic antibodies. Sci. Rep. 13, 10473; https://doi.org/10.1038/s41598-023-37288-6 (2023).
Fu, Q., Polanco, A., Lee, Y. S. & Yoon, S. Critical challenges and advances in recombinant adeno-associated virus (rAAV) biomanufacturing. Biotechnol. Bioeng. 120, 2601–2621; https://doi.org/10.1002/bit.28412 (2023).
Destro, F. et al. The state of technological advancement to address challenges in the manufacture of rAAV gene therapies. Biotechnol. Adv. https://doi.org/10.1016/j.biotechadv.2024.108433 (2024).
Lorek, J. K., Isaksson, M. & Nilsson, B. Chromatography in downstream processing of recombinant adeno-associated viruses: a review of current and future practises. Biotechnol. Bioeng. https://doi.org/10.1002/bit.28932 (2025).
FDA. Manufacturing changes and comparability for human cellular and gene therapy products; draft guidance for industry. http://www.regulations.gov. (2023).
Fernandes, S., Guerra, J., Ferreira, M. V. & Coroadinha, A. S. Deciphering key adenoviral elements in the production of recombinant adeno-associated virus vectors. Hum Gene Ther 36; https://doi.org/10.1089/hum.2024.265 (2025).
Acknowledgements
The author(s) gratefully acknowledge the members of the laboratory for their insightful discussions and constructive feedback throughout the development of this work.
Funding
This work was supported by the Research Unit UID/04462: iNOVA4Health – Programme in Translational Medicine, financially supported by Fundação para a Ciência e Tecnologia / Ministério da Educação, Ciência e Inovação (PTDC_BTM_ORG_1383_2020), the Associate Laboratory LS4FUTURE (LA/P/0087/2020) and the AAVscreen iBETXplore Grant. J.M.E is funded by Stimulus of Scientific Employment, Individual Support program (2020. 01216.CEECIND) and F.M. by PhD fellowship 2022.11494.BD from FCT. I.R.S. appreciate support by the FWF PhD Program Grant https://doi.org/10.55776/W1224 “Biotechnology of Proteins – BioTop”.
Author information
Authors and Affiliations
Contributions
M.A writing – original draft, investigation, methodology, visualization, and formal analysis. F.M., investigation, methodology, visualization, and formal analysis. I.R.S investigation, methodology, and formal analysis. P.M.A, supervision and funding acquisition P.G.A. supervision, project supervision, project administration, funding acquisition. J.M.E., writing – original draft, project conceptualization, supervision, project supervision, investigation, funding acquisition. All authors: writing – review & editing.
Corresponding author
Ethics declarations
Competing interests
The authors declare no competing interests.
Additional information
Publisher’s note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Supplementary Information
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.
About this article
Cite this article
Antunes, M., Moura, F., Sebastian, I.R. et al. Streamlined rAAV HeLaS3 producer cell line generation via GS selection. Sci Rep (2026). https://doi.org/10.1038/s41598-025-34826-2
Received:
Accepted:
Published:
DOI: https://doi.org/10.1038/s41598-025-34826-2