Skip to main content

Phase I/II Manufacture of Lentiviral Vectors Under GMP in an Academic Setting

  • Protocol
  • First Online:
Chimeric Antigen Receptor T Cells

Abstract

In clinical gene transfer applications, lentiviral vectors (LV) have rapidly become the primary means to achieve permanent and stable expression of a gene of interest or alteration of gene expression in target cells. This status can be attributed primarily to the ability of the LV to (1) transduce dividing as well as quiescent cells, (2) restrict or expand tropism through envelope pseudo-typing, and (3) regulate gene expression within different cell lineages through internal promoter selection. Recent progress in viral vector design such as the elimination of unnecessary viral elements, split packaging, and self-inactivating vectors has established a significant safety profile for these vectors. The level of GMP compliance required for the manufacture of LV is dependent upon their intended use, stage of drug product development, and country where the vector will be used as the different regulatory authorities who oversee the clinical usage of such products may have different requirements. As such, successful GMP manufacture of LV requires a combination of diverse factors including: regulatory expertise, compliant facilities, validated and calibrated equipments, starting materials of the highest quality, trained production personnel, scientifically robust production processes, and a quality by design approach. More importantly, oversight throughout manufacturing by an independent Quality Assurance Unit who has the authority to reject or approve the materials is required. We describe here the GMP manufacture of LV at our facility using a four plasmid system where 293T cells from an approved Master Cell Bank (MCB) are transiently transfected using polyethylenimine (PEI). Following transfection, the media is changed and Benzonase added to digest residual plasmid DNA. Two harvests of crude supernatant are collected and then clarified by filtration. The clarified supernatant is purified and concentrated by anion exchange chromatography and tangential flow filtration. The final product is then diafiltered directly into the sponsor defined final formulation buffer and aseptically filled.

This is a preview of subscription content, log in via an institution to check access.

Access this chapter

Subscribe and save

Springer+ Basic
$34.99 /Month
  • Get 10 units per month
  • Download Article/Chapter or eBook
  • 1 Unit = 1 Article or 1 Chapter
  • Cancel anytime
Subscribe now

Buy Now

Protocol
USD 49.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD 79.99
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD 99.99
Price excludes VAT (USA)
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info
Hardcover Book
USD 129.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

Similar content being viewed by others

References

  1. Milone MC, O'Doherty U (2018) Clinical use of lentiviral vectors. Leukemia 32(7):1529–1541

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Coffin J (1997) In: Hughes S, Varmus H (eds) Retroviruses. Cold Spring Harbor Laboratory Press, New York

    Google Scholar 

  3. Rosenberg S, Aebersold P, Cornetta K et al (1990) Gene transfer into humans--immunotherapy of patients with advanced melanoma, using tumor-infiltrating lymphocytes modified by retroviral gene transduction. N Engl J Med 323:570–578

    Article  CAS  PubMed  Google Scholar 

  4. Blaese R, Culver K, Miller A et al (1995) T lymphocyte-directed gene therapy for ADA–SCID: initial trial results after 4 years. Science 270:475–480

    Article  CAS  PubMed  Google Scholar 

  5. Kohn D, Weinberg K, Nolta J et al (1995) Engraftment of gene-modified umbilical cord blood cells in neonates with adenosine deaminase deficiency. Nat Med 1:1017–1023

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Cavazzana-Calvo M, Hacein-Bey S, de Saint Basile G et al (2000) Gene therapy of human severe combined immunodeficiency (SCID)-X1 disease. Science 288:669–672

    Article  CAS  PubMed  Google Scholar 

  7. Aiuti A, Slavin S, Aker M et al (2002) Correction of ADA-SCID by stem cell gene therapy combined with nonmyeloablative conditioning. Science 296:2410–2413

    Article  CAS  PubMed  Google Scholar 

  8. Hacein-Bey-Abina S, Von Kalle C, Schmidt M et al (2003) LMO2-associated clonal T cell proliferation in two patients after gene therapy for SCID-X1. Science 302:415–419

    Article  CAS  PubMed  Google Scholar 

  9. Bushman F (2007) Retroviral integration and human gene therapy. J Clin Invest 117(8):2083–2086

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Cattoglio C, Pellin D, Rizzi E et al (2010) High definition mapping of retroviral integration sites identifies active regulatory elements in human multipotent hematopoietic progenitors. Blood 116:5507–5517

    Article  CAS  PubMed  Google Scholar 

  11. Raper SE, Chirmule N, Lee FS et al (2003) Fatal systemic inflammatory response syndrome in a ornithine transcarbamylase deficient patient following adenoviral gene transfer. Mol Genet Metab 80:148–158

    Article  CAS  PubMed  Google Scholar 

  12. Schröder A, Shinn P, Chen H et al (2002) HIV-1 integration in the human genome favors active genes and local hotspots. Cell 110(4):521–529

    Article  PubMed  Google Scholar 

  13. Mitchell R, Beitzel B, Schroder A et al (2004) Retroviral DNA integration: ASLV, HIV, and MLV show distinct target site preferences. PLoS Biol 2(8):E234

    Article  PubMed  PubMed Central  Google Scholar 

  14. https://www.cancer.gov/about-cancer/treatment/clinical-trials/intervention/C120309. Accessed 1 Nov 2018

  15. Schambach A, Swaney W, van der Loo J (2009) Design and production of retro- and lentiviral vectors for gene expression in hematopoietic cells. Methods Mol Biol 506:191–205

    Article  CAS  PubMed  Google Scholar 

  16. Dull T, Zufferey R, Kelly M et al (1998) A third-generation lentivirus vector with a conditional packaging system. J Virol 72(11):8463–8471

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Modlich U, Bohne J, Schmidt M et al (2006) Cell-culture assays reveal the importance of retroviral vector design for insertional genotoxicity. Blood 108:2545–2553

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Gándara C, Affleck V, Stoll EA (2018) Manufacture of third-generation Lentivirus for preclinical use, with process development considerations for translation to good manufacturing practice. Hum Gene Ther Methods 29(1):1–15

    Article  PubMed  PubMed Central  Google Scholar 

  19. Gama-Norton L, Botezatu L, Herrmann S et al (2011) Lentivirus production is influenced by SV40 large T-antigen and chromosomal integration of the vector in HEK293 cells. Hum Gene Ther 22:1269–1279

    Article  CAS  PubMed  Google Scholar 

  20. Valkama AJ, Leinonen HM, Lipponen EM et al (2018) Optimization of lentiviral vector production for scale-up in fixed-bed bioreactor. Gene Ther 25(1):39–46

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Merten OW, Hebben M, Bovolenta C (2016) Production of lentiviral vectors. Mol Ther Methods Clin Dev 3:16017. https://doi.org/10.1038/mtm.2016.17

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Tomás HA, Rodrigues AF, Carrondo MJT, Coroadinha AS (2018) LentiPro26: novel stable cell lines for constitutive lentiviral vector production. Sci Rep 8(1):5271. https://doi.org/10.1038/s41598-018-23593-y

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Slepushkin V, Chang N, Cohen R et al (2003) Large-scale purification of a lentiviral vector by size exclusion chromatography or mustang Q ion exchange chromatography. Bioprocess J 2(5):89–95

    Article  Google Scholar 

  24. Leath A, Cornetta K (2012) Developing novel lentiviral vectors into clinical products. Methods Enzymol 507:89–108

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgments

We are grateful to Prof. H. Trent Spencer, Department of Pediatrics, School of Medicine, Emory University, Atlanta GA, for his critical review of this book chapter. Our facility is generously supported by the Cincinnati Children’s Hospital Research Foundation and we would like to acknowledge the hard work and dedication of the entire staff of the Translational Core Laboratories at Cincinnati Children’s Hospital; without it, our contributions to the gene therapy field would not be possible.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Anindya Dasgupta .

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2020 Springer Science+Business Media, LLC, part of Springer Nature

About this protocol

Check for updates. Verify currency and authenticity via CrossMark

Cite this protocol

Dasgupta, A. et al. (2020). Phase I/II Manufacture of Lentiviral Vectors Under GMP in an Academic Setting. In: Swiech, K., Malmegrim, K., Picanço-Castro, V. (eds) Chimeric Antigen Receptor T Cells. Methods in Molecular Biology, vol 2086. Humana, New York, NY. https://doi.org/10.1007/978-1-0716-0146-4_3

Download citation

  • DOI: https://doi.org/10.1007/978-1-0716-0146-4_3

  • Published:

  • Publisher Name: Humana, New York, NY

  • Print ISBN: 978-1-0716-0145-7

  • Online ISBN: 978-1-0716-0146-4

  • eBook Packages: Springer Protocols

Publish with us

Policies and ethics