Elsevier

Metabolic Engineering

Volume 7, Issues 5–6, September–November 2005, Pages 426-436
Metabolic Engineering

Detailed design and comparative analysis of protocols for optimized production of high-performance HIV-1-derived lentiviral particles

https://doi.org/10.1016/j.ymben.2005.06.006Get rights and content

Abstract

Transgenic HIV-1-derived lentiviral particles are at the forefront of current gene therapy and tissue engineering initiatives, which will require optimal protocols for large-scale production of clinical-grade therapeutic lentiviruses. Production of latest-generation self-inactivating lentiviral particles requires cotransfection of mammalian production cell lines with two helper plasmids along with the lentivector, whose transgene-encoding expression cassette is the only genetic information stably transduced into target chromosomes. Capitalizing on a recently designed lentiviral expression vector family, we conducted rigorous analysis of production-relevant parameters including transfection, cell density, media composition, temperature, relative (helper) vector concentrations and genetic configuration. Comparative analysis of lentiviral particle performance (VP) was based on the viral titer (reflecting the number of transduction-competent lentiviral particles) relative to the number of lentiviral particles produced (correlating with p24 production levels) (VP=titer/viral particle number). Optimal lentiviral production parameters, resulting in up to 132-fold greater VP compared to standard protocols, required (i) CaPO4-based transfection (ii) of helper plasmids and lentivector at a fixed concentration ratio (helper plasmid I:helper plasmid II:lentivector=1:1:2) (iii) into 1×105 human embryonic kidney cells/cm2 (HEK293-T) (iv) cultivated at 37 °C (v) in Advanced D-MEM medium supplemented with (vi) 2% fetal calf serum, (vii) and a culture additive containing 0.01 mM cholesterol, 0.01 mM egg's lecithin and 1× chemically defined lipid concentrate. (viii) Furthermore, constitutive transgene expression units placed in a forward polyadenylation site (pA)-free orientation relative to the lentivector backbone resulted in optimal transgene transduction/expression. Our studies suggest that detailed knowledge of lentivector design and the production of lentiviral particles will advance large-scale manufacturing of clinically relevant lentiviruses for future gene therapy applications.

Introduction

Owing to their efficient transduction of proliferating as well as mitotically inert cells in the absence of significant humoral immune responses, human immunodeficiency virus type 1 (HIV-1) lentiviral expression vectors have been designed for therapeutic interventions in (prototype) gene therapy scenarios (Crystal, 1995; Williams, 1995; Anderson, 2000; Somia and Verma, 2000; Kay et al., 2001; Nishikawa and Huang, 2001; Kang et al., 2002). Latest-generation HIV-1-based lentiviral transduction systems consist of multiply attenuated self-inactivating (SIN) viral genomes split among several (helper) plasmids harboring (i) gag (encoding major structural proteins), pol (coding for lentivirus-specific enzymes) and rev (a regulator of gag/pol expression and nuclear export of virus RNA), (ii) a vsv-g expression vector promoting pantropic transduction of pseudotyped lentiviral particles and (iii) the actual transgene(s)-encoding lentivector, which is the only genetic material transferred to the target cells (Dull et al., 1998). The lentivector harbors non-coding cis-acting elements, which manage encapsidation (extended packaging signal [ψ+], reverse transcription and integration (polypurine tracts [PPT, cPPT]; 5′ and 3′LTRs; Rev response element [RRE]). In order to prevent transcriptional interference with transgene expression and provirus-flanking chromosomal elements at the integration site, most lentivectors contain an enhancer-specific deletion in their 3′LTR, which renders the lentiviral particles SIN (Deglon et al., 2000). Standard lentiviral particle production requires cotransfection of two helper plasmids encoding a different pseudotyping envelope (Bartz and Vodicka, 1997; Mochizuki et al., 1998; Reiser et al., 2000; Beyer et al., 2002; Duisit et al., 2002; Kang et al., 2002) and complementing structural genes (De Palma and Naldini, 2002), along with the transgene-encoding lentivector, into human embryonic kidney cells (HEK293-T). Following transfection, lentiviral particles were produced and released into the culture supernatant, from which they were filtered for ready-to-use transduction batches (Naldini et al., 1996; Mitta et al., 2002; Koponen et al., 2003; Blesch, 2004). Stable helper cell lines, transgenic for constitutive complementation of structural genes and conditional expression of toxic vsv-g, have been considered as an alternative to triple transient transfections (Yu et al., 1996; Kaul et al., 1998; Kafri et al., 1999; Klages et al., 2000; Beyer et al., 2002). However, overall production performance did not always meet expectations (Rohll et al., 2002). Transient transfection thus remains the preferred technology for pilot production of lentiviral particles in a rapid, reliable and safe format (Naldini et al., 1996; Dull et al., 1998; Johnston et al., 1999; Morizono et al., 2001; D’Costa et al., 2003; Koponen et al., 2003; Mangeat et al., 2003; Mitta et al., 2004; Ventura et al., 2004). Although most established biopharmaceutical manufacturing scenarios use stable production cell lines (Zahn-Zabal et al., 2001), transient transfection is gathering momentum as a straightforward, consistent and cost-effective method for pilot production of biopharmaceuticals and recombinant viral particles (Zahn-Zabal et al., 2001; Derouazi et al., 2004; Maranga et al., 2004; Merten, 2004). There are a variety of protocols for the production of lentiviral particles which render a critical evaluation and comparative analysis from a bioengineering perspective impossible (Naldini et al., 1996; Zufferey et al., 1997; Mochizuki et al., 1998; Reiser et al., 2000; Zhao et al., 2002; Coleman et al., 2003; Condiotti et al., 2004). Furthermore, there is no generally accepted parameter describing overall virus performance. p24, the capsid protein encoded by gag (Oroszlan et al., 1979; Copeland et al., 1983; Coffin et al., 1997), is often quantified and provided as a measure correlating with virus titer. Yet, p24 levels fail to indicate the transduction quality of lentiviral particle preparations, since they only score the presence of this protein on the surface of lentiviral particles in a function-independent manner (2000 p24 proteins per lentiviral particle (Layne et al., 1992; Huang et al., 2001)). While virus titer generally correlates with transduction-competent lentiviral particles it fails to quantify the number of lentiviral particles generated during a production process.

We suggest that putting titer and lentiviral particles into relative perspective will enable a direct comparison of lentiviral particle production and transduction performance across platforms and protocols. Detailed multi-factor evaluation of different protocols for lentiviral particle production revealed critical parameters for optimal pilot production of high-quality recombinant lentiviral particles.

Section snippets

Materials and methods

Vector design: A complete list of plasmids used and constructed in this study is provided in Table 1.

Cell culture: Human embryonic kidney cells, transgenic for SV40 large T antigen [HEK293-T; (Mitta et al., 2002)], were typically cultivated in Dulbecco's modified Eagle's medium (DMEM; Invitrogen, Carlsbad, CA) supplemented with 10% heat-inactivated fetal calf serum (FCS; PAN Biotech GmbH, Aidenbach, Germany, Cat. No.: 3302-P231902, Lot No.: P231902), 100 U penicillin and 100 μg/ml streptomycin

Results

Impact of HEK293-T transfection on lentiviral particle production: In order to assess the impact of HEK293-T transfection on lentiviral particle production we produced pMF365 [(5′LTR-ψ+-oriSV40-cPPT-RRE-PhEF1α-EYFP-3′LTRΔU3; (Mitta et al., 2002)]-derived lentiviral particles using FuGENE6 and CaPO4-based HEK293-T transfection protocols. p24 was quantified in production culture supernatants and used to calculate the number of lentiviral particles. Lentiviral particle titers were determined by

Discussion

High-level, large-scale and cost-effective production of recombinant lentiviral particles will be essential for a clinical impact of gene-based therapies. Progress during the past decade has substantiated the potential of lentivirus-based transduction of therapeutic transgenes for prototype gene therapy scenarios (Deglon et al., 2000; Kordower et al., 2000; Mitta et al., 2002; Koponen et al., 2003; Blomer et al., 2004; Humeau et al., 2004; Mitta et al., 2004; Mitta et al., 2005). Despite

Acknowledgments

We thank Valeria Gonzalez-Nicolini, David Greber as well as Andrea Guarino and Alessandro Usseglio Viretta for critical comments and fruitful discussions about the manuscript. This work was supported by the Swiss National Science Foundation (Grant no. 631-065946), the Swiss State Secretariat for Education and Research within EC Framework 6 and an ETH scholarship granted to B.M.

References (62)

  • M. Kaul et al.

    Regulated lentiviral packaging cell line devoid of most viral cis-acting sequences

    Virology

    (1998)
  • N. Klages et al.

    A stable system for the high-titer production of multiply attenuated lentiviral vectors

    Mol. Ther.

    (2000)
  • S.P. Layne et al.

    Factors underlying spontaneous inactivation and susceptibility to neutralization of human immunodeficiency virus

    Virology

    (1992)
  • L. Maranga et al.

    Scale-up of virus-like particles production: effects of sparging, agitation and bioreactor scale on cell growth, infection kinetics and productivity

    J. Biotechnol.

    (2004)
  • J.B. Rohll et al.

    Design, production, safety, evaluation, and clinical applications of nonprimate lentiviral vectors

    Methods Enzymol.

    (2002)
  • M. Zahn-Zabal et al.

    Development of stable cell lines for production or regulated expression using matrix attachment regions

    J. Biotechnol.

    (2001)
  • W.F. Anderson

    Gene therapy scores against cancer

    Nat. Med.

    (2000)
  • M. Azzouz et al.

    Lentiviral vectors for treating and modeling human CNS disorders

    J. Gene Med.

    (2004)
  • W.R. Beyer et al.

    Oncoretrovirus and lentivirus vectors pseudotyped with lymphocytic choriomeningitis virus glycoprotein: generation, concentration, and broad host range

    J. Virol.

    (2002)
  • U. Blomer et al.

    Shuttle of lentiviral vectors via transplanted cells in vivo

    Gene Ther.

    (2004)
  • N. Chazal et al.

    Virus entry, assembly, budding, and membrane rafts

    Microbiol. Mol. Biol. Rev.

    (2003)
  • J.M. Coffin et al.

    Retroviruses

    (1997)
  • J.E. Coleman et al.

    Efficient large-scale production and concentration of HIV-1-based lentiviral vectors for use in vivo

    Physiol. Genom.

    (2003)
  • R.G. Crystal

    Transfer of genes to humans: early lessons and obstacles to success

    Science

    (1995)
  • J. D’Costa et al.

    HIV-2 derived lentiviral vectors: gene transfer in Parkinson's and Fabry disease models in vitro

    J. Med. Virol.

    (2003)
  • N. Deglon et al.

    Self-inactivating lentiviral vectors with enhanced transgene expression as potential gene transfer system in Parkinson's disease

    Hum. Gene Ther.

    (2000)
  • M. Derouazi et al.

    Serum-free large-scale transient transfection of CHO cells

    Biotechnol. Bioeng.

    (2004)
  • T. Dull et al.

    A third-generation lentivirus vector with a conditional packaging system

    J. Virol.

    (1998)
  • C. Fux et al.

    Dual-regulated expression of C/EBP-alpha and BMP-2 enables differential differentiation of C2C12 cells into adipocytes and osteoblasts

    Nucl. Acids Res.

    (2004)
  • C. Fux et al.

    Streptogramin- and tetracycline-responsive dual regulated expression of p27(Kip1) sense and antisense enables positive and negative growth control of Chinese hamster ovary cells

    Nucl. Acids Res.

    (2001)
  • K. Holm et al.

    Human immunodeficiency virus type 1 assembly and lipid rafts: Pr55(gag) associates with membrane domains that are largely resistant to Brij98 but sensitive to Triton X-100

    J. Virol.

    (2003)
  • Cited by (49)

    • Targeted supplementation design for improved production and quality of enveloped viral particles in insect cell-baculovirus expression system

      2016, Journal of Biotechnology
      Citation Excerpt :

      Cholesterol was the main system booster, capable of improving cell specific yields of both baculovirus and enveloped VLPs. The manipulation of lipid and cholesterol metabolism has culminated in improved production of enveloped viral particles in several producer systems (Cervera et al., 2013; Chen et al., 2010; Mitta et al., 2005; Rodrigues et al., 2009). Cholesterol has an important role in membrane fluidity and rigidity, being invaluable for biogenesis and functionality (Bloch, 1983), as well as aiding in the stabilization of viral particles envelope, maturation and budding (Chan, 2010), thus being a major contributor to viral infectivity.

    • Influence of HEK293 metabolism on the production of viral vectors and vaccine

      2015, Vaccine
      Citation Excerpt :

      Nevertheless, feb-batch approach has provided some interesting indications for improvement of HEK293 viral production processes. First, although serum-free media are desired in viral production, addition of foetal bovine serum (FBS) or calf serum (CS) is a well-known strategy to increase viral yields [66,78,80,84]. It is now evident that lipids usually contained in serum have to be supplemented for successful viral production in serum-free media, especially for production of enveloped viruses such as lentivirus and retrovirus [85].

    • Construction and expression of lentiviral vectors encoding recombinant mouse CREBZF in NIH 3T3 cells

      2014, Plasmid
      Citation Excerpt :

      The mixture of DNA and Turbofect was added to the medium very gently and was incubated overnight at 37 °C in a CO2 incubator. The medium was changed after 16 h. Conditioned medium, which consisted of Advanced DMEM medium, 2% FBS, 0.01 mM cholesterol, 0.01 mM egg lecithin and 1 × chemically defined lipid concentrate (Mitta et al., 2005), was harvested after an additional 48 h, purified by low-speed centrifugation, filtered through a 0.45 µm PVDF filter (Millipore) and stored at −80 °C. Before transduction, 5 × 103 HEK 293T cells were seeded into each well.

    View all citing articles on Scopus
    View full text