Biomaterials 32 (2011) 9854e9865 Contents lists available at SciVerse ScienceDirect Biomaterials journal homepage: www.elsevier.com/locate/biomaterials The significance of plasmid DNA preparations contaminated with bacterial genomic DNA on inflammatory responses following delivery of lipoplexes to the murine lung Reto P. Bazzania, c, Ying Caib, Henry L. Hebelb, Stephen C. Hydea, c, Deborah R. Gilla, c, * a b c Gene Medicine Research Group, NDCLS, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DU, UK VGXI, Inc. 2700 Research Forest Dr., #180 The Woodlands, TX 77381, USA United Kingdom Cystic Fibrosis Gene Therapy Consortium, UK a r t i c l e i n f o a b s t r a c t Article history: Received 15 August 2011 Accepted 31 August 2011 Available online 23 September 2011 Non-viral gene transfer using plasmid DNA (pDNA) is generally acknowledged as safe and nonimmunogenic compared with the use of viral vectors. However, pre-clinical and clinical studies have shown that non-viral (lipoplex) gene transfer to the lung can provoke a mild, acute inflammatory response, which is thought to be, partly, due to unmethylated CG dinucleotides (CpGs) present in the pDNA sequence. Using a murine model of lung gene transfer, bronchoalveolar lavage fluid was collected following plasmid delivery and a range of inflammatory markers was analysed. The results showed that a Th1-related inflammatory cytokine response was present that was substantially reduced, though not abolished, by using CpG-free pDNA. The remaining minor level of inflammation was dependent on the quality of the pDNA preparation, specifically the quantity of contaminating bacterial genomic DNA, also a source of CpGs. Successful modification of a scalable plasmid manufacturing process, suitable for the production of clinical grade pDNA, produced highly pure plasmid preparations with reduced genomic DNA contamination. These studies help define the acceptable limit of genomic DNA contamination that will impact FDA/EMEA regulatory guidelines defining clinical grade purity of plasmid DNA for human use in gene therapy and vaccination studies. Ó 2011 Elsevier Ltd. All rights reserved. Keywords: Cytokine Bacterial genomic DNA Gene therapy Lung inflammation Lipoplex Plasmid 1. Introduction Plasmid DNA (pDNA) is being investigated for therapeutic use, including the development of strategies for gene replacement and vaccination in humans and animals. Plasmid DNA can be administered alone, often termed ’naked’ DNA delivery, or complexed with a cationic lipid (lipoplex), or polymer (polyplex) [1]. Non-viral gene transfer vectors are generally regarded as being safer and less toxic compared with viral vectors, and the absence of viral proteins predicts that non-viral vectors should be less immunogenic and thus can be administered repeatedly without generating an immune response. Gene replacement in the lung is being developed for treatment of Cystic Fibrosis (CF), a monogenic disease affecting greater than 70,000 patients worldwide [2], where the key factor affecting * Corresponding author. Gene Medicine Research Group, NDCLS, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DU, UK. Tel.: þ44 1865 221845; fax: þ44 1865 221834. E-mail address: [email protected] (D.R. Gill). 0142-9612/$ e see front matter Ó 2011 Elsevier Ltd. All rights reserved. doi:10.1016/j.biomaterials.2011.08.092 morbidity and mortality is the chronic destructive infection of the conducting airways, eventually leading to lung failure and premature death [3]. Plasmid DNA complexed with the cationic liposome GL67A, which was specifically formulated for aerosol delivery, has been delivered to the nose and lungs of CF patients. In one early trial, pDNA/GL67A was aerosolised to the lungs of CF patients and resulted in partial electrophysiological correction of the CF ion transport defect in the lung after a single aerosol dose [4]. However, lung delivery of the pDNA/GL67A lipoplex also resulted in an acute inflammatory response [4,5], similar to that observed in early preclinical studies in mouse models [6e9]. This inflammatory response was thought to be due in part to the plasmid containing unmethylated CG dinucleotides (CpGs) [10,11], the ligand for the Toll-like receptor (TLR)9 [12]. Following lipoplex delivery to the murine lung, inflammatory markers, such as IFNg and IL-12, can be detected in bronchoalveolar lavage (BAL) fluid [13], but are decreased if CpGs in the pDNA are methylated [6,14], reduced [11] or eliminated [10]. The UK CF Gene Therapy Consortium (UKCFGTC) [15] has developed a non-viral, pDNA/GL67A formulation, where all CpGs have been removed from the plasmid DNA, such that delivery R.P. Bazzani et al. / Biomaterials 32 (2011) 9854e9865 of the new CpG-free plasmid (pGM169) to the murine lung was inflammation-free [10]. Another observation from early lung gene therapy trials was that transgene expression was transient, lasting no more than 1 week [4,16e20]. Investigation of this phenomenon in animal models showed that loss of transgene expression did not correlate with a loss of plasmid DNA [21], but instead, was linked to the specific promoter used to control expression of the transgene [10,22,23]. Replacement of commonly used viral promoters with those from constitutively expressed ’housekeeping’ genes can increase the duration of expression. For example, the use of the human polyubiquitinC (UbC) promoter can lead to persistent transgene expression in the mouse lung when delivered in the form of naked DNA via electroporation [24] and following aerosol lung delivery [10]. Unfortunately, this rather successful promoter could not be incorporated into CpG-free plasmids, since the removal of CpGs from the UbC promoter sequences abolished promoter activity (unpublished data). However after testing a variety of CpG-free promoter/enhancer combinations, high-level persistent expression in the lung was achieved from a novel, CpGfree promoter based on the human cytomegalovirus enhancer and elongation factor 1 alpha promoter (hCEFI promoter) [10]. Thus the modification of plasmid DNA is an important area for the development of novel non-viral formulations. Plasmid sequences can be easily manipulated to customise the level and duration of transgene expression and exploited for treatment of different disease targets. Plasmids can vary in size, but the majority of vectors are too large for chemical synthesis that is economically feasible. When produced in non-pathogenic E. coli, plasmid DNA only constitutes 1e2% of the total bacterial cell mass, which means its separation from abundant impurities is challenging. Guidance from regulatory bodies such as the Food & Drug Administration [25] and the European Medicines Agency [26] recommends that in plasmid preparations for human use, bacterial host protein, RNA, and genomic DNA levels constitute preferably < 1% each and endotoxin levels < 40 endotoxin units (EU)/mg, although more stringent criteria may be required depending on the specific application. Large scale manufacture of clinical grade pDNA still faces several obstacles [27]. A low shear cell lysis process is critical, as this determines the achievable concentration and purity of the final product. Although non-chromatographic techniques such as selective precipitation or aqueous two-phase partitioning have been studied, anion exchange (AEX), hydrophobic interaction chromatography (HIC), and size exclusion chromatography are three major workhorses in plasmid purification. These allow for the exploitation of plasmid specific characteristics such as charge density, hydrophobicity and size. High purity is an ultimate goal, but other factors such as cost, flexibility, broad utility and 9855 robustness are just as valuable for establishing a feasible manufacturing process. In this study, we investigated the inflammatory consequences of delivering lipoplexes to the lungs of wild type (WT) and TLR9deficient (TLR9/) mice. This involved measuring a broad spectrum of inflammatory markers induced in response to delivery of first generation plasmid DNA containing large numbers of CpGs (CpG-rich) and comparing the results with those obtained following delivery of lipoplexes prepared using CpG-free plasmid DNA. In addition we investigated how scalable processes suitable for manufacturing plasmid DNA for human use could be improved to produce high purity plasmid and reduce inflammation following in vivo delivery. 2. Materials and methods 2.1. Animals Wild type female BALB/c mice aged 6e12 weeks (at point of procedure performance) were purchased from Harlan Laboratories UK (Loughborough, UK) or Biomedical Service Unit (University of Oxford, Oxford, UK). Two breeding pairs of TLR9/ mice were purchased from Oriental BioService Inc. (Kyoto, Japan). Mice were housed in accordance with UK Home Office ethical and welfare guidelines and fed on standard chow and water ad libitum. All procedures were carried out under UK Home Office approved project and personal licenses for performing experiments on animals under the terms of the Animals (Scientific Procedures) Act 1986. 2.2. Delivery of pDNA by intranasal instillation Mice were anaesthetised with methoxyfluorane (Mallinckrodt Veterinary Inc., Mundelein, IL, USA) or isoflurane (University of Oxford Vet Services, Oxford, UK). Lipoplexes were formed using pDNA and GL67A (1:2:0.05 M ratio of GL67:DOPE:DMPE-PEG5000); GL67 was a gift from Genzyme Corporation (Framingham, MA, USA), DOPE:DMPE-PEG5000 was purchased from Avanti Polar Lipids Inc. (Alabaster, AL, USA) and formulated by OctoPlus (Leiden, NL). A volume of 100 ml pDNA/GL67A complexes, containing 80 mg pDNA, at a pDNA:GL67A mM ratio of 2.4:0.6, was delivered to the lung via intranasal instillation as described previously [28]. 2.3. Harvesting of bronchoalveolar lavage fluid Mice were euthanised by cervical dislocation at 24 h following treatment. For analysis of BAL fluid cytokines, the trachea was exposed by blunt dissection. A small tear was made in the trachea and a cannula was inserted. Lungs were washed three times with 1 ml BAL fluid solution (1 PBS, 0.1% w/v BSA, 0.05 mM EDTA). Cells from BAL fluid were concentrated by centrifugation. The supernatant was collected to determine cytokine levels. 2.4. Plasmid DNA Plasmid descriptions are in Table 1. Plasmid pCIKLux [22], contains the firefly luciferase transgene driven by the human CMV immediate-early promoter and enhancer and contains 317 CpGs, thus also referred to as CpG-rich (Table 1). Plasmid pCIKLux was prepared using an EndoFree Plasmid Mega Kit (Qiagen, Crawley, UK). The CpG-free plasmid pGM169 contains the synthetic optimised cystic fibrosis transmembrane conductance regulator (CFTR) transgene (called soCFTR2 [10]) driven by the human CMV enhancer, human elongation factor 1a promoter and Table 1 Characteristics of plasmid preparations. Summary of plasmid DNA preparations used in this study and the quantity of bacterial gDNA measured, pre-DNase treatment and postDNase treatment where applicable. The CpG-rich plasmid was made using an EndoFree Plasmid Mega Kit (Qiagen), whereas CpG-free plasmid preparations were manufactured by commercial suppliers. Endotoxin levels were <5 endotoxin units (EU)/mg for all preparations used. CMV, cytomegalovirus; Lux, firefly luciferase; soCFTR2, synthetic optimised cystic fibrosis transmembrane conductance regulator [10]; N/A, not applicable; hCEFI, human CMV enhancer human elongation factor 1a [10], CpG, CG dinucleotide; SEM, standard error of the mean. Name Plasmid CpG-content pCIKL ux-LB pGM169-A2 pGM169-P1 pGM169-P2 pGM169-V15 CpGCpGCpGCpGCpG- a b c richb freec freec freec freec Promoter Transgene % bacterial gDNA SEMa CMV hCEFI hCEFI hCEFI hCEFI Lux soCFTR2 soCFTR2 soCFTR2 soCFTR2 1.680 4.468 4.5 0.306 0.077 The quantity of bacterial gDNA per pDNA preparation is shown as a percentage of the total DNA. CpG-rich: pDNA construct containing 317 CpGs. CpG-free: pDNA construct containing no CpGs. 0.058 0.164 0.146 0.008 0.003 % bacterial gDNA SEMa after DNase treatment N/A N/A 0.425 0.019 0.009 0.0005 N/A 9856 R.P. Bazzani et al. / Biomaterials 32 (2011) 9854e9865 R.P. Bazzani et al. / Biomaterials 32 (2011) 9854e9865 enhancer (hCEFI [10]). Preparations of pGM169 were supplied by Aldevron (Fargo, ND, USA), PlasmidFactory (Bielefeld, Germany) or VGXI (The Woodlands, TX, USA) as indicated in the text. Plasmid DNA preparation pGM169-V15 was manufactured to clinical grade standard compared with others that resulted from research-grade processes. All plasmid preparations contained < 5 EU/mg endotoxin. 2.5. DNase treatment of pDNA Plasmid-SafeTM ATP-Dependent DNase (Epicentre Biotechnologies, Madison, WI, USA) was used according to the manufacturer’s instructions to remove residual bacterial gDNA from plasmid preparations. Following DNase treatment, plasmid preparations were re-purified using the EndoFree Plasmid Mega Kit (Qiagen). In brief, 1/12 volume of endotoxin removal buffer was added to the reaction mix, followed by dilution with 10 volumes of equilibration buffer. The mixture was incubated on ice for 30 min, before transferred to a Qiagen-tip 2500 plasmid purification column. The remainder of the purification process was performed according to the manufacturer’s instructions. 9857 2.9. Bio-Plex The levels of 23 cytokines (CCL2 (MCP-1), CCL3 (MIP-1a), CCL4 (MIP-1b), CCL5 (RANTES), CCL11 (Eotaxin), CXCL1 (KC), G-CSF, GM-CSF, IL-1a, IL-1b, IL-2, IL-3, IL-4, IL-5, IL-6, IL-9, IL-10, IL-12p40, IL-12p70, IL-13, IL-17, IFNg and TNFa) in BAL fluid were determined using a Bio-Plex multiplex cytokine assay (Bio-Rad Ltd., Hemel Hempstead, UK) according to manufacturer’s instructions. Samples were analysed using a Bio-Plex 100 or a Bio-Plex 200 system with Bio-Plex Manager software v5.0 (Bio-Rad). 2.10. Statistics Multiple groups were analysed using one-way ANOVA followed by Dunnett’s multiple comparison test. Pairs of interest determined in advance were analysed using one-way ANOVA followed by Bonferroni post-test correction. Data were considered to be statistically significant different when p < 0.05 (*p < 0.05, **p < 0.01 and ***p < 0.001). GraphPad Prism software v5 (GraphPad, San Diego, CA) was used for the analyses. 2.6. Agarose gel electrophoresis 3. Results Agarose gels consisted of 1.0% agarose (Invitrogen, Paisley, UK), 500 ng/ml ethidium bromide in 1 Tris-acetate-EDTA (TAE) buffer. Samples were mixed with 10 loading dye in a final volume of 10 ml and electrophoresed at 80e120 V for 1e2 h. For DNA size estimation, 500 ng of a 12 kb DNA ladder (Stratagene, Cambridge, UK) and 500 ng of a supercoiled DNA ladder (range 2e10 kb) (Promega, Southampton UK) were electrophoresed alongside. Plasmid DNA preparations in Fig. 6 were analysed with ready-to-use 1% SeaKemÒ Gold Reliant agarose gels (Lonza, Rockland, ME, USA). Prior to loading, high-salt samples were dialysed against water on 0.025 mm MF-Millipore membrane filters (Billerica, MA, USA) for 40e50 min and mixed with 5 loading dye (Hercules, CA, USA) to a final volume of 10 ml. Electrophoresis was conducted in TAE buffer, at 100 V for 80 min. Supercoiled DNA ladder (Invitrogen, Carlsbad, CA, USA) was used at 0.25 mg/lane. Gels were stained by SYBR green I nucleic acid gel stain (Invitrogen, USA) for 40 min and visualised with a Storm 840 imaging system (GE Healthcare, Piscataway, NJ, USA) at a resolution of 200 mm pixel size and using the fluorescence scanning mode. 2.7. Quantification of bacterial gDNA by TaqMan Absolute levels of bacterial gDNA were quantified by a GT115-specific quantitative PCR (qPCR) assay (GT115 sequence gift of Dr. Eric Perouzel, InvivoGen).The forward (F), reverse (R) primer and probe (P) sequences being: F: 50 -TGT GAG CGT CGC AGA ACA TT-30 ; R: 50 -TTT TAG CAA CGT ACT ATC CAC TCA-30 and P: 50 VIC-CAT TGA CGC AGG TGA ATC GGA CGC-TAMRA-30 . Each 10 ml PCR reaction consisted of 4 ml DNA sample (40 ng total DNA), 1 TaqMan Universal Mastermix (Applied Biosystems, Warrington, UK), 300 nM forward primer, 300 nM reverse primer and 100 nM probe. PCR plates were heat-sealed with a clear seal and placed in an 7900 HT Fast Real-Time PCR System (Applied Biosystems). Cycling conditions were: 2 min at 50 C and then 10 min at 95 C followed by 40 cycles of 15 s at 95 C and 1 min at 60 C. Nuclease-free water was included as a no template control. The number of bacterial gDNA copies was calculated from standard curves prepared from E. coli GT115 gDNA and expressed as percentage gDNA of total amount of DNA. 2.8. Scalable manufacturing of highly pure plasmid DNA The general plasmid DNA manufacturing process [29] was modified for production of pGM169 with very low levels of gDNA. A two-step purification process, involving anion exchange (AEX) followed by hydrophobic interaction chromatography (HIC), is utilised to make products for human applications. AEX employed Mustang Q (Pall, Pensacola, FL, USA) membrane capsules. Butyl 650-M resin (Tosoh Bioscience LLC, Montgomeryville, PA, USA) was packed in BPGÔ columns (GE Healthcare) for the HIC step (process A). For this study, a new technique of thiophilic interaction chromatography (TIC) was explored to replace (process B), or supplement HIC (process C). A XK50/20 column (GE healthcare) was packed with resin PlasmidSelect Xtra (GE healthercare) to a height of 14 cm, operating at a linear flow rate of 100 cm/h. The load of AEX product was pre-conditioned with 3 M ammonium sulphate to 245 mS/cm before loading to the TIC column (process B), but the pre-conditioning was omitted when TIC was added in succession to HIC (process C). Solutions used for column equilibration, wash and elution were 2.0 M, 1.9 M and 1.7 M ammonium sulphate, respectively. Water for injection (WFI) and 0.5 M NaOH were employed to strip and sanitise column/resin prior to and after each use. 3.1. Inflammatory cytokine levels in BAL fluid To investigate the inflammatory consequences of delivering lipoplexes to the lung, mice were intranasally instilled with CpGrich pCIKLux/GL67A lipoplexes, CpG-free pGM169/GL67A lipoplexes, the gene transfer agent (GTA) lipid GL67A alone, or water for injection (WFI). The BAL fluid was harvested 24 h post treatment and analysed using a multiplex cytokine assay (Bio-Plex) for a broad spectrum of 23 inflammatory markers. Six of the 23 analytes (IL-2, IL-3, IL-4, IL-9, IL-13 and CCL11) showed no significant differences between any treatment (p > 0.05; Suppl. Fig. 1). For one analyte (CXCL1) all treatment groups that contained GL67A were significantly higher than WFI treatment (p < 0.001; Fig. 1). The remaining sixteen analytes (TNFa, IFNg, IL-12p40, IL-12p70, IL-1a, IL-1b, IL-6, IL-5, IL-10, IL-17, CCL2, CCL3, CCL4, CCL5, G-CSF and GM-CSF) showed significantly (p < 0.05; Fig. 1) reduced levels in BAL fluid following treatment with CpG-free lipoplexes compared with CpG-rich lipoplexes, confirming our initial observations [10] that depletion of CpGs from pDNA results in a reduced host inflammatory reaction. However, for ten of these 16 analytes (IFNg, IL-12p40, IL-12p70, IL-1a, IL-5, IL-6, CCL2, CCL4, CCL5 and G-CSF), a modest residual response to CpGfree lipoplexes was noted such that levels in response to CpG-free lipolpex treatment were greater (p < 0.05) than for WFI treatment. For six of these ten analytes (IFNg, IL-12p40, IL-6, CCL2, CCL4 and CCL5) CpG-free lipoplex effects were also marginally greater than for GL67A treatment alone. Importantly, all plasmid preparations used contained endotoxin levels of <5 EU/mg (Table 1), 8 times lower than the level approved by the FDA for a drug used for intratracheal administration [25], thus the effect of endotoxins was likely to be negligible in these experiments. Taken together, these data suggest that in this model of lung gene transfer pDNA per se, independent of its CpG content, may be inflammatory, or that additional components of the plasmid preparation may need to be considered. Interestingly, the difference between CpG-associated cytokines IFNg, IL-12p40 and IL-6 after delivery of CpG-free lipoplexes and delivery of lipid alone suggested that the plasmid preparations contained a source of unmethylated CpGs leading to the observed residual inflammatory response. Fig. 1. Levels of 17 inflammatory response markers in BAL fluid following nasal instillation of lipoplexes. Female BALB/c mice (n ¼ 6 per group) were treated with 100 ml CpG-rich or CpG-free GL67A lipoplexes, lipid GL67A alone (Lipid only) or WFI via nasal instillation. BAL fluid was collected 24 h post-delivery and cytokine levels of TNFa (A), IFNg (B), IL-12p40 (C), IL-12p70 (D), IL-1a (E), IL-1b (F), IL-5 (G), IL-6 (H), IL-10 (I), IL-17 (J), CCL2 (K), CCL3 (L), CCL4 (M), CCL5 (N), CXCL1 (O), G-CSF (P) and GM-CSF (Q) were determined by Bio-Plex. Data shown are mean SEM. Assay sensitivity limits (- - -) were 297.22 pg/ml (A), 1.61 pg/ml (B), 4.73 pg/ml (C), 3.06 pg/ml (D), 1.46 pg/ml (E), 14.12 pg/ml (F), 4.89 pg/ml (G), 3.06 pg/ml (H), 5.18 pg/ml (I), 6.39 pg/ml (J), 102.02 pg/ml (K), 578.24 pg/ml (L), 31.72 pg/ml (M) 35.45 pg/ml (N), 1.94 pg/ml (O), 7.56 (P) and 22.38 pg/ml (Q). Statistical differences were analysed by one-way ANOVA/Dunnett to unlabelled comparator: ***p < 0.001, **p < 0.01, *p < 0.05 and ns: non-significant. Fig. 2. Residual inflammatory response in TLR9/ mice following instillation of CpG-free lipoplexes. Female BALB/c wild type (wt) mice (light grey) (n ¼ 5 to 6 per group), or female BALB/c TLR9-deficient (TLR9/) mice (dark grey) (n ¼ 5 to 6 per group) were treated with 100 ml CpG-free lipoplexes or WFI via nasal instillation. BAL fluid was collected 24 h post-delivery and levels of TNFa (A), IFNg (B), IL-12p40 (C), IL-6 (D), CCL2 (E), CCL3 (F), CCL4 (G), CCL5 (H), CXCL1 (I) and G-CSF (J) were determined by Bio-plex. Data shown are mean SEM. Assay sensitivity limits (- - -) were 12.7 pg/ml (A), 6.06 pg/ml (B), 5.67 pg/ml (C), 1.16 pg/ml (D), 35.56 pg/ml (E), 9.89 pg/ml (F), 5.65 pg/ml (G), 0.91 pg/ml (H), 3.47 pg/ml (I) and 2.93 pg/ml (J). No graphical representation of assay sensitivity limits < 1 pg/ml. Statistical differences between treatments within a genotype were analysed by one-way ANOVA/Dunnett to unlabelled comparator: ***p < 0.001; **p < 0.01; *p < 0.05 and ns: non-significant. A two-way ANOVA/Bonferroni statistical test was used to analyse differences between the same treatment in two different genotypes:]*p < 0.05. WT: wild type; TLR9/: TLR9-deficient. Fig. 3. Inflammatory markers in BAL fluid vary depending on CpG-free plasmid preparation. Female BALB/c mice (n ¼ 5e6 per group) were treated with 100 ml CpG-rich or CpG-free GL67A lipoplexes, or WFI via nasal instillation. CpG-free plasmid from three commercial suppliers was used. One supplier provided a clinical grade preparation (pGM169-V15). One supplier provided two differently manufactured plasmid preparations (pGM169-P1 and pGM169-P2). BAL fluid was collected 24 h post-delivery and levels of TNFa (A), IFNg (B), IL-12p40 (C), IL-6 (D), CCL2 (E), CCL3 (F), CCL4 (G), CCL5 (H), CXCL1 (I) and G-CSF (J) were determined by Bio-Plex. Data shown are mean SEM. Assay sensitivity limits (- - -) were 3.46 pg/ml (A), 1.56 pg/ml (B), 0.69 pg/ml (C), 1.32 pg/ml (D), 22.26 pg/ml (E), 42.37 pg/ml (F), 5.79 pg/ml (G) 2.53 pg/ml (H), 1.6 pg/ml (I) and 1.62 (J). No graphical representation of assay sensitivity limits < 1 pg/ml. Statistical differences between any treatment group and WFI control group were analysed by one-way ANOVA/Dunnett, whereas one-way ANOVA/Bonferroni was used to analyse the selected preparations from the same supplier: *** and p < 0.001, and ns: non-significant. Fig. 4. Decreased levels of inflammatory markers in BAL fluid following reduction of bacterial genomic DNA in plasmid DNA preparations. Female BALB/c mice (n ¼ 5e6 per group) were treated with 100 ml CpG-free GL67A lipoplexes, or WFI via nasal instillation. The CpG-free plasmid DNA preparation was pGM169-P1 and was treated with either PlasmidSafeÔ ATP-dependent DNase (Epicentre Biotechnologies) (0.43% gDNA), or a mock treatment without enzyme (4.5% gDNA). BAL fluid was collected 24 h post-delivery and levels of TNFa (A), IFNg (B), IL-12p40 (C), IL-6 (D), CCL2 (E), CCL3 (F), CCL4 (G), CCL5 (H), CXCL1 (I) and G-CSF (J) were determined by Bio-Plex. Data shown are mean SEM. Assay sensitivity limits (- - -) were 1.35 pg/ml (A), 12.51 pg/ml (B), 2.1 pg/ml (C), 0.48 pg/ml (D), 6.47 pg/ml (E), 13.73 pg/ml (F), 2.43 pg/ml (G) 0.09 pg/ml (H), 0.31 pg/ml (I) and 0.31 (J). No graphical representation of assay sensitivity limits < 1 pg/ml. Statistical differences were analysed by one-way ANOVA/Dunnett to unlabelled comparator: ***p < 0.001; *p < 0.05 and ns: nonsignificant. R.P. Bazzani et al. / Biomaterials 32 (2011) 9854e9865 9861 Fig. 5. Overview of plasmid manufacture process. Plasmid DNA production begins with batch mode fermentation to high cell density and high plasmid yields, followed by continuous centrifugation to harvest cells. Cells are frozen prior to downstream processing. Alkaline lysis allows low shear mixing of the viscous lysate at a large scale. Removal of solid flocculent from plasmid solution is realised with a series of filtration steps. Clarified lysate goes through the purification process (step I, II and optional III), desalting/ concentration with ultrafiltration/diafiltration and final aseptic filtration and fill; M: Motor. To determine if the residual response was due to the presence of unmethylated CpGs, lipoplexes were delivered to the lungs of TLR9/ mice, deficient in TLR9 required for detection of unmethylated CpGs. A subset of the 23 analytes, representative of the inflammatory response, is presented throughout Figs. 2e4. As shown in 1. 2, in WT mice, six of the ten analytes (IL-12p40, IL-6, CCL2, CCL5, CXCL1 and G-CSF) showed increased levels in BAL fluid after treatment with CpG-free lipoplexes compared with WFI. Similarly in TLR9/ mice, levels of IL-12p40, IL-6, CCL2, CCL4, CCL5 and G-CSF were significantly increased following treatment with CpG-free lipoplexes compared with WFI. Of the three CpG-associated cytokines IFNg, IL12-p40 and IL-6, only IL-12p40 showed a significant (p < 0.05; 2.6-fold) reduction in the TLR9/ mice following intranasal instillation of CpG-free lipoplexes compared with WT mice (Fig. 2C). IFNg and IL-6 appeared reduced in TLR9/ mice compared with WT mice (3.9-fold and 2.2- fold, respectively), but did not reach significance (p > 0.05; Fig. 2B and D). These results suggest that the residual inflammatory cytokine response observed following intranasal instillation of CpG-free lipoplexes may in part be due to a CpG-dependent response. We hypothesised that bacterial gDNA present in plasmid preparations was considered a possible source of unmethylated CG dinucleotides. 3.2. Inflammatory cytokines associated with CpG-free plasmid Four research or clinical grade preparations of the CpG-free plasmid pGM169 (termed pGM169-A2, -P1, -P2 and -V15; Table 1) were obtained from three different suppliers and their bacterial gDNA content was evaluated. As expected, agarose gel electrophoresis was not sufficiently sensitive to reveal the presence of sheared fragments of bacterial gDNA in pDNA preparations (data not shown), therefore a quantitative PCR (qPCR) assay for gDNA was developed. The qPCR assay used was highly sensitive with a detection limit of 4 copies per reaction and was reproducible such that results obtained for a specific sample were comparable when analysed in separate laboratories using independent tests (data not shown). The qPCR assay was used to measure the percentage bacterial gDNA present and showed that this varied widely between 0.077% 0.003e4.5% 0.15 in the preparations tested; the clinical grade preparation (pGM169-V15) showing the lowest levels of %gDNA measured (Table 1). Interestingly, although plasmid preparations pGM169-P1 and pGM169-P2 were manufactured by the same supplier, production of pGM169-P2 included a step for specific removal of bacterial chromosomal DNA, and this appeared to reduce bacterial gDNA contamination by w15-fold (Table 1). Next, the potential for each of these plasmid preparations to elicit an inflammatory response was investigated. Results showed that lower levels of contaminating bacterial gDNA appeared to correlate with lower levels of CpG-associated cytokines IFNg and IL-12p40 (Fig. 3B and C). Furthermore, the additional gDNA removal step for manufacturing pGM169-P2 compared with pGM169-P1 correlated with significantly reduced cytokine levels (p < 0.001) (Fig. 3B and C). 3.3. Bacterial gDNA contamination in pDNA We hypothesised that bacterial gDNA contaminating plasmid preparations, was responsible, at least in part, for the residual inflammatory response observed in the mouse lung following intranasal instillation of CpG-free lipoplexes. To confirm this, two plasmid preparations (pGM169-P1 and pGM169-P2) were treated with Plasmid-SafeTM ATP-Dependent DNase (Epicentre Biotechnologies), an exonuclease that specifically targets linear double stranded DNA such as bacterial gDNA, but with no observed enzymatic activity on closed circular dsDNA or supercoiled (SC) DNA comprising the majority of pDNA. Agarose gel electrophoresis was used to confirm that enzymatic treatment did not grossly affect the predominantly supercoiled conformation of the plasmid (Suppl. Fig. 2), and qPCR was used to measure gDNA. Table 1 shows that enzymatic treatment reduced gDNA by at least 10-fold for each plasmid. Next, the enzymatically-treated and mock-treated plasmids were complexed with GL67A lipid and intranasally instilled into mice, so that the inflammatory response could be determined. The pGM169-P1 preparation, where bacterial gDNA was reduced w10-fold from 4.5 to 0.43%, (Table 1) resulted in significantly (p < 0.05) reduced levels of seven of the ten analytes (TNFa, IFNg, IL-12p40, IL-6, CCL2, CCL3 and G-CSF; Fig. 4). However, for some of the markers (IL-12p40, IL-6, CCL2, CCL4, CCL5, CXCL1, G-CSF) the residual levels were still significantly (p < 0.05) higher than the WFI control group, despite the reduction in bacterial gDNA. Delivery of the enzymatically-treated plasmid preparation pGM169-P2, where bacterial gDNA was reduced w33-fold from 0.3 to 0.009% (Table 1), resulted in no significant (p > 0.05) decrease for any of the ten analytes investigated (TNFa, IFNg, IL-12p40, IL-6, CCL2, CCL3, CCL4, CCL5, CXCL1 and G-CSF) (data not shown). In addition, seven of the ten analytes (IFNg, IL-12p40, IL-6, CCL2, CCL3, CCL4 and CCL5) were significantly (p < 0.05) increased compared with animals treated 9862 R.P. Bazzani et al. / Biomaterials 32 (2011) 9854e9865 Fig. 6. Agarose gel electrophoresis of samples of pGM169 processed by AEX, HIC and/or TIC. Following purification, samples of plasmid pGM169 were analysed by agarose gel electrophoresis. Bands represent genomic DNA (gDNA), open circular (OC) pDNA, supercoiled (SC) pDNA and RNA. (A): HIC process samples of Lot#1 (Process A, AEX þ HIC). Lane 1, SC plasmid ladder (Invitrogen); Lane 2, Load; Lane 3, flowthrough (FT) initial; Lane 4, FT middle; Lane 5, FT end; Lane 6, wash; Lane 7, Eluate; Lane 8, WFI strip. (B): TIC process samples of Lot#2 (Process B, AEX þ TIC). Lane 1, SC plasmid ladder; Lane 2, Load; Lane 3, FT middle; Lane 4, FT end; Lane 5, wash; Lane 6, Eluate; Lane 7, Eluate end; Lane 8, WFI strip. (C): HIC process samples of Lot#3 (Process C, AEX þ HIC þ TIC). Lane 1, SC plasmid ladder; Lane 2, Load; Lane 3, FT initial; Lane 4, FT middle; Lane 5, FT end; Lane 6, wash; Lane 7, Eluate; Lane 8, WFI strip. (D): TIC process samples of Lot#3 (Process C, AEX þ HIC þ TIC). Lane 1, SC plasmid ladder; Lane 2, Load; Lane 3, FT initial; Lane 4, FT middle; Lane 5, FT end; Lane 6, Eluate; Lane 7, Eluate end; Lane 8, WFI strip. with lipid alone (Fig. 4). Together, these results suggest that the quality of plasmid preparations, at least with respect to contaminating levels of bacterial gDNA, should be improved and might be of importance in minimising unwanted inflammatory side effects if used for therapeutic purpose. 3.4. Large scale pDNA production Preparation of pDNA with total yields in the region of 2e10 mg per column (e.g. Large-Scale Plasmid DNA Purification Kits) is routine in the laboratory, but can be more efficiently manufactured to higher purity by commercial suppliers, particularly at larger scales; and is essential when producing plasmid for therapeutic purposes. However, the purity of pDNA preparations seemed to depend on the manufacturing process used (Table 1). Therefore, we wished to develop a process for manufacturing clinical grade pDNA, where we could reliably achieve low levels of bacterial gDNA. Fig. 5 outlines the main steps used in the manufacture of clinical grade pGM169 plasmid by one commercial supplier, VGXI. Following a 10 L scale batch fermentation, cells were processed by alkaline lysis, and plasmid was isolated and purified with either two or three chromatographic steps. Anion exchange membrane chromatography was effective in primary capture of pDNA and major reduction of impurities. However, different polishing steps following AEX, HIC and/or TIC, were explored to achieve maximum gDNA removal. Thus, three robust purification processes were developed: (A) AEX þ HIC; (B) AEX þ TIC; (C) AEX þ HIC þ TIC. The CpG-free plasmid, pGM169 [10], was generated using each process and the purity of each final product was compared (Table 2). Process A resulted in high %SC and low levels of impurities (Table 2). In this process, the HIC step load conditions were R.P. Bazzani et al. / Biomaterials 32 (2011) 9854e9865 9863 Table 2 Comparison of final drug product specifications following the use of different purification processes. Three different purification processes (two 2-step processes (A and B) and one 3-step process (C)) were tested. The resulting filtered drug product was analysed using quality control release assays. The purification processes were tested following a medium scale fermentation of 10 L, yielding 100e400 mg of final drug product. AEX: anion exchange membrane chromatography, HIC: hydrophobic interaction chromatography, TIC: thiophilic interaction chromatography, EU: endotxin units, SC: supercoiled, and OC: open circular. Process Process A: AEX þ HIC Process B: AEX þ TIC Process C: AEX þ HIC þ TIC Concentration (mg/ml) Purity (A260/280) Restriction Appearance 5.618 2.016 Conform to standards Clear colourless solution with no visible particulates 0.06 0.125 0.5 0.26 97.4 1.2 1.4 5.606 2.025 Conform to standards Clear colourless solution with no visible particulates 0.06 0.362 0.1 0.24 97.2 1.3 1.5 5.563 2.042 Conform to standards Clear colourless solution with no visible particulates 0.06 0.204 0.26 0.003 97.7 0.8 1.6 Host cell protein (%) Endotoxin (EU/mg) Host cell RNA (%) Host cell genomic DNA (%) Forms SC (%) OC (%) Other (%) optimised to achieve pDNA isoform separation, with SC pDNA binding to the resin while open circular (OC) and trace multimeric pDNA forms along with gDNA (fragmented to w 20 kb during alkaline lysis and subsequent processing) passing through the column in the non-absorbed flowthrough (FT) phase (Fig. 6A; lanes 3e5). Single-stranded RNA exhibited much higher hydrophobicity than SC pDNA and was only recovered from the HIC column during WFI column strip/regeneration step (Fig. 6A; lane 8). Host protein and endotoxin predominantly co-eluted with RNA in the WFI column strip/regeneration step (data not shown). A typical lot of pGM169 produced by this process (Fig. 6A; lane 7) contained 0.26% residual gDNA as determined by qPCR (Table 2). In an attempt to reduce residual gDNA levels, we developed Process B e an alternative two-step chromatography process, where HIC is replaced by TIC. Process B demonstrated a similar impurity profile to Process A (Table 2). TIC, like HIC in Process A, allowed flowthrough removal of OC pDNA along with gDNA (Fig. 6B; lanes 3 and 4). A typical lot of pGM169 produced by this process (Fig. 6B; lanes 6 and 7) was highly enriched for SC pDNA (Table 2). However, residual gDNA levels achieved with Process B (0.24%) were broadly similar to levels achieved with Process A (Table 2). We hypothesised that a three-step chromatographic process would be required to substantially reduce levels of residual gDNA in the final product. Consequently, we developed Process C, combining AEX, HIC and TIC. Comparable with the two-step processes, OC pDNA and gDNA removal was facilitated in the flowthrough of both, the HIC (Fig. 6C; lanes 3e5) and TIC (Fig. 6D; lanes 3e5) steps. In a typical lot of pGM169 produced by this process, this resulted in a modest reduction in residual OC pDNA levels and a substantial w100-fold reduction in residual gDNA levels in the final product (Table 2). These data confirm that by process modification, clinical grade pDNA for human use can be generated with extremely low levels (0.003%) of contaminating bacterial gDNA. 4. Discussion Pre-clinical and clinical studies have documented an acute inflammatory response to lung delivery of lipoplexes [4e6,10,14], which may limit their therapeutic use. This inflammatory response, characterised by increased pro-inflammatory cytokine levels and an influx of white blood cells (WBC) to the lung, can be measured in BAL fluid from the lungs of treated mice [10]. Similar to mice, a WBC influx, dominated by neutrophils, has been reported in sheep following lipoplex delivery to the lung [30]. In this present study, a highly sensitive, multiplex assay was used to further characterise the response by measuring a broad spectrum of 23 markers of Th1 and Th2 immune responses and leukocyte trafficking and maturation (Fig. 1 and Suppl. Fig. 1). The results obtained were consistent with published reports of a Th1-biased inflammatory cytokine response triggered by unmethylated CpGs [6,10,11,14]. In addition, the inflammatory response was accompanied by increased levels of CC-chemokines, also associated with a type I (Th1-related) inflammation mediated by monocytes/ macrophages [31], and increased CXCL1, the murine functional homologue of the strong neutrophil attractant human CXCL8 (IL-8). Furthermore, the observed Th1-biased inflammatory response is in line with the use of unmethylated CpG oligodeoxynucleotides as an adjuvant for immunisation [32]. In an attempt to mitigate the observed acute inflammatory response following lung delivery of GL67A lipoplexes for CF gene therapy [4,5], the UKCFGTC [15] developed plasmid pGM169, which is capable of persistent CFTR expression in the mouse lung, and is entirely devoid of CpGs such that inflammation in the mouse lung, as judged by three cytokines analysed by ELISA, was reduced to background (naïve) levels [10]. However, with the development of the more sensitive and extensive assays used in these studies, further characterisation of the response to lipoplex delivery has revealed that even when using a CpG-free plasmid, a low-grade, residual inflammatory response remains (Fig. 1). To investigate this further, CpG-free lipoplexes were delivered via intranasal instillation to the lungs of mice deficient in the TLR9 (TLR9/) receptor for unmethylated CpGs [12]. Results indicated the presence of a TLR9-independent inflammatory response to lipoplex delivery (Fig. 2), consistent with reports that TLR9 signalling was only partially responsible for toxic responses following systemic delivery of lipoplexes [33]. Possible contaminants of plasmid preparations were considered. Bacterial lipopolysaccharide fragments, also known as endotoxins, can readily contaminate plasmid preparations and cause inflammation. However, all plasmid preparations used in this study had endotoxin levels of <5 EU/mg, at least 8 times lower than recommended by regulatory bodies [25]. Furthermore, studies have shown previously that routine endotoxin contamination of plasmid preparations is unlikely to contribute substantially towards the inflammatory response elicited by lipoplexes [6]. Results presented here showed that part of the inflammatory response could be due to the lipid component of the gene transfer agent, since delivery of GL67A alone resulted in a slight inflammatory response (Fig. 1 and Suppl. Fig. 1). Intriguingly, levels of CpG-associated cytokines IFNg, IL-6 and IL-12p40 were further increased when lipid was combined with CpG-free pDNA. This, together with the observation that these cytokines showed a > 2-fold reduction in their levels in TLR9/ mice, indicated the presence of an additional source of 9864 R.P. Bazzani et al. / Biomaterials 32 (2011) 9854e9865 unmethylated CpGs in plasmid preparations, most likely contaminating bacterial gDNA. In support of this, cytokine levels, (especially IFNg, IL-6 and IL-12p40) were found to vary between plasmid preparations (Fig. 3), and correlated with the levels of bacterial gDNA in the plasmid preparation used (Table 1). This hypothesis was confirmed by enzymatic digestion of bacterial gDNA in plasmid preparations prior to use in vivo and the concomitant reduction in pro-inflammatory cytokines (Fig. 4). Our observations, that plasmid preparations can induce a variable inflammatory response in vivo, are in line with a recent report of toxicity following hydrodynamic limb vein injection of naked plasmid DNA into rodent limb muscles [34]. Some of the pDNA preparations caused muscle necrosis whilst others did not; the results were dependent on the batch of pDNA used and correlated with the presence of bacterial gDNA, such that 5% of bacterial gDNA in plasmid preparations was sufficient to cause muscle damage [34]. The authors recommended that the amount of contaminating bacterial gDNA present in a plasmid preparation should be reported along with the experimental results. Unfortunately, since sheared fragments of bacterial gDNA in a plasmid preparation are variable in size, they are difficult to visualise and quantify using routine agarose gel electrophoresis, thus qPCR assays are required to accurately report residual low levels of gDNA. When we used our qPCR assay to measure gDNA levels in research-grade plasmid DNA prepared in our laboratory, we found that these were generally low (pCIKLux; Table 1). The %gDNA present in commercially prepared research-grade plasmid preparations was similar, except for pGM169-P2 where an additional processing step was included resulting in lower levels (Table 1). Importantly, it is possible for occasional plasmid preparations to contain > 5% gDNA (data not shown), which could contribute to toxicity when used in vivo. Irrespective of the procedures included to reduce bacterial gDNA in research-grade plasmid preparations, those plasmids ultimately intended for therapeutic use required a scalable, reproducible, production process. We investigated the feasibility of modifying a clinical plasmid manufacturing process to produce plasmid with minimal bacterial gDNA contamination. The two-step plasmid purification process, using AEX þ HIC (Process A), has been used to produce cGMP plasmid at 1e50 g scale for over 80 different plasmids, generating high SC% (95%) product and purity exceeding the recommendations set by the FDA [25] and other regulatory agencies. However, the levels of impurities in the final product can vary, typically correlating inversely with specific plasmid yields (data not shown). A high copy number plasmid can easily be produced with levels of RNA 0.1%, protein 0.1%, endotoxin 0.1 EU/mg and gDNA 0.01% (where “” designates result lower than assay limit of detection). Recent human papillomavirus and influenza DNA vaccines that used pUC derived backbone were manufactured with the two step process (AEX þ HIC) under cGMP regulations, all showing undetectable levels of impurities at a pDNA concentration of up to 10 mg/ml [35]. However, CpG-free plasmid pGM169, derived from R6K vectors [36] exhibited lower copy number and reduced yields, even under optimised fermentation conditions. Fragments of gDNA generated by shear during processing, share similar physical-chemical characteristics to SC pDNA and are most difficult to eliminate. The AEX step can reduce bulk protein, RNA and endotoxin, but is less effective at reducing fragmented gDNA that co-elutes with plasmid. A process of AEX þ HIC or AEX þ TIC is sufficient to reduce gDNA to < 1%, or lower with typical high yield plasmids. When initial plasmid yield is low, the ratio of %gDNA in the AEX eluate becomes relatively high, thus resolution with subsequent HIC or TIC is limited. In this study, two-step processes reduced bacterial gDNA to w0.2%; whereas combining them in a three-step process further reduced residual gDNA levels w100 fold (Table 2). However, one must take into consideration that such low level bacterial gDNA was obtained at the cost of a 10e20% loss of plasmid product. The extra processing step will reduce overall downstream yield and an increased fermentation scale will be required to achieve the desired quantity of final product. Eventually, the need for increased purity has to be balanced with the increased costs involved. Whilst 5% gDNA was toxic for delivery to rodent muscle, preparations with <1.3% gDNA did not cause significant muscle damage [34]. In this study, gDNA at contaminating levels of w4.5% correlated with a CpG-dependent inflammatory response (Figs. 3 and 4 and Table 1), which was reduced although still measureable when bacterial gDNA was reduced to < 0.43% (Fig. 4). The authors believe that the acceptable range of %gDNA for a plasmid preparation is reasonable at 1% for most applications and possibly as low as 0.1% for therapeutic purposes. After minimising the CpG-dependent inflammatory response by using CpG-free plasmid DNA and reducing the %gDNA contamination, the data indicated there was an additional CpGindependent and TLR9-independent cause of low level inflammation following intranasal instillation of lipolexes to the mouse lung. This additional mechanism could be due to the lipid component of the complex, which could potentially mask any auxiliary reduction in inflammatory cytokine levels that could be achieved by further improving the purity of plasmid preparations. In addition, TLR9-independent systems that recognise ’foreign’ DNA in the cytoplasm such as the inflammasomes [37] or the DNAdependent activator of IFN-regulatory factors (DAI) [38] might also play a role. For example, cell culture studies using non-TLR9expressing Glomerular endothelial cells transfected with CpGfree lipoplexes showed production of cytokines and chemokines, such as IL-6, CCL2, CCL5 and type I IFNs, in a MyD88-independent cytosolic DNA recognition pathway [39]. A hallmark of TLR9/ MyD88-independent DNA recognition pathways is the resulting expression of Type I IFNs [40], which were not amongst the inflammatory analytes studied herein. It would therefore be appropriate to investigate alternative inflammatory response markers and/or pathways associated with cytoplasmic DNA recognition, such as type I IFNs, and establish if their levels change upon treatment with CpG-rich lipoplex compared with CpG-free lipoplex or WFI. 5. Conclusion Removal of CpGs from plasmid expression vectors minimises the inflammatory response to plasmid/lipid complexes in the mouse lung and may help mitigate the acute inflammatory response in clinical studies. However, a low-level CpG-dependent response is still detectable due to contaminating bacterial gDNA present in plasmid preparations. Our results demonstrate that scalable manufacturing processes can be adapted to further reduce contaminating levels of gDNA in plasmid preparations, but the cost of additional processing must be considered. Acknowledgement We would like to thank Dr. Martin Schleef from PlasmidFactory for providing us with two different batches of pGM169, G. NuñezAlonso for helping with BAL fluid collection and Dr. I. Pringle for critically reading the manuscript. This work was funded by a studentship from the UK Cystic Fibrosis Trust via the UK Cystic Fibrosis Gene Therapy Consortium (http://www.cfgenetherapy. org.uk). R.P. Bazzani et al. / Biomaterials 32 (2011) 9854e9865 Appendix. Supplementary material Supplementary information associated with this article can be found, in the online version, at doi:10.1016/j.biomaterials.2011. 08.092. References [1] Gao X, Kim KS, Liu D. Nonviral gene delivery: what we know and what is next. AAPS J 2007;9:E92e104. [2] Griesenbach U, Alton EW. Current status and future directions of gene and cell therapy for cystic fibrosis. BioDrugs 2011;25:77e88. [3] Welsh MJ, Ramsey BW, Accurso F, Cutting GR. Cystic fibrosis. In: Scriver CR, Beaudet AL, Sly WS, Valle D, editors. The Metabolic and Molecular Bases of Inherited Diseases. 8th ed. New York: McGraw-Hill; 2001. p. 5121e88. [4] Alton EW, Stern M, Farley R, Jaffe A, Chadwick SL, Phillips J, et al. Cationic lipid-mediated CFTR gene transfer to the lungs and nose of patients with cystic fibrosis: a double-blind placebo-controlled trial. Lancet 1999;353: 947e54. [5] Ruiz FE, Clancy JP, Perricone MA, Bebok Z, Hong JS, Cheng SH, et al. A clinical inflammatory syndrome attributable to aerosolized lipid-DNA administration in cystic fibrosis. Hum Gene Ther 2001;12:751e61. [6] Freimark BD, Blezinger HP, Florack VJ, Nordstrom JL, Long SD, Deshpande DS, et al. Cationic lipids enhance cytokine and cell influx levels in the lung following administration of plasmid: cationic lipid complexes. J Immunol 1998;160:4580e6. [7] Li S, Wu SP, Whitmore M, Loeffert EJ, Wang L, Watkins SC, et al. Effect of immune response on gene transfer to the lung via systemic administration of cationic lipidic vectors. Am J Physiol 1999;276:L796e804. [8] Norman J, Denham W, Denham D, Yang J, Carter G, Abouhamze A, et al. Liposome-mediated, nonviral gene transfer induces a systemic inflammatory response which can exacerbate pre-existing inflammation. Gene Ther 2000;7: 1425e30. [9] Scheule RK, St George JA, Bagley RG, Marshall J, Kaplan JM, Akita GY, et al. Basis of pulmonary toxicity associated with cationic lipid-mediated gene transfer to the mammalian lung. Hum Gene Ther 1997;8:689e707. [10] Hyde SC, Pringle IA, Abdullah S, Lawton AE, Davies LA, Varathalingam A, et al. CpG-free plasmids confer reduced inflammation and sustained pulmonary gene expression. Nat Biotechnol 2008;26:549e51. [11] Yew NS, Zhao H, Wu IH, Song A, Tousignant JD, Przybylska M, et al. Reduced inflammatory response to plasmid DNA vectors by elimination and inhibition of immunostimulatory CpG motifs. Mol Ther 2000;1:255e62. [12] Hemmi H, Takeuchi O, Kawai T, Kaisho T, Sato S, Sanjo H, et al. A Toll-like receptor recognizes bacterial DNA. Nature 2000;408:740e5. [13] Abdullah S. Studies on the persistence of transgene expression in the murine airways. Oxford: University of Oxford; 2005. [14] Yew NS, Wang KX, Przybylska M, Bagley RG, Stedman M, Marshall J, et al. Contribution of plasmid DNA to inflammation in the lung after administration of cationic lipid:pDNA complexes. Hum Gene Ther 1999;10:223e34. [15] UKCFGTC. The UK Cystic Fibrosis Gene Therapy Consortium - http://www. cfgenetherapy.org.uk/clinical.html. [16] Zabner J, Cheng SH, Meeker D, Launspach J, Balfour R, Perricone MA, et al. Comparison of DNA-lipid complexes and DNA alone for gene transfer to cystic fibrosis airway epithelia in vivo. J Clin Invest 1997;100:1529e37. [17] Caplen NJ, Alton EW, Middleton PG, Dorin JR, Stevenson BJ, Gao X, et al. Liposome-mediated CFTR gene transfer to the nasal epithelium of patients with cystic fibrosis. Nat Med 1995;1:39e46. [18] Gill DR, Southern KW, Mofford KA, Seddon T, Huang L, Sorgi F, et al. A placebocontrolled study of liposome-mediated gene transfer to the nasal epithelium of patients with cystic fibrosis. Gene Ther 1997;4:199e209. [19] Porteous DJ, Dorin JR, McLachlan G, Davidson-Smith H, Davidson H, Stevenson BJ, et al. Evidence for safety and efficacy of DOTAP cationic lipo- [20] [21] [22] [23] [24] [25] [26] [27] [28] [29] [30] [31] [32] [33] [34] [35] [36] [37] [38] [39] [40] 9865 some mediated CFTR gene transfer to the nasal epithelium of patients with cystic fibrosis. Gene Ther 1997;4:210e8. Hyde SC, Southern KW, Gileadi U, Fitzjohn EM, Mofford KA, Waddell BE, et al. Repeat administration of DNA/liposomes to the nasal epithelium of patients with cystic fibrosis. Gene Ther 2000;7:1156e65. Pringle IA, Raman S, Sharp WW, Cheng SH, Hyde SC, Gill DR. Detection of plasmid DNA vectors following gene transfer to the murine airways. Gene Ther 2005;12:1206e14. Gill DR, Smyth SE, Goddard CA, Pringle IA, Higgins CF, Colledge WH, et al. Increased persistence of lung gene expression using plasmids containing the ubiquitin C or elongation factor 1alpha promoter. Gene Ther 2001;8: 1539e46. Poulsen TT, Pedersen N, Juel H, Poulsen HS. A chimeric fusion of the hASH1 and EZH2 promoters mediates high and specific reporter and suicide gene expression and cytotoxicity in small cell lung cancer cells. Cancer Gene Ther 2008;15:563e75. Pringle IA, McLachlan G, Collie DD, Sumner-Jones SG, Lawton AE, Tennant P, et al. Electroporation enhances reporter gene expression following delivery of naked plasmid DNA to the lung. J Gene Med 2007;9:369e80. FDA. Guidance for industry: consideration of plasmid DNA vaccines for infectious disease indications, http://www.fda.gov/downloads/BiologicsBloodVaccines/ GuidanceComplianceRegulatoryInformation/Guidances/Vaccines/ ucm091968pdf; 2007. EMEA. Note for guidance on the quality, preclinical and clinical aspects of gene trasnfer medicinal products; 2001. Cai Y, Rodriguez S, Hebel H. DNA vaccine manufacture: scale and quality. Expert Rev Vaccines 2009;8:1277e91. Lee ER, Marshall J, Siegel CS, Jiang C, Yew NS, Nichols MR, et al. Detailed analysis of structures and formulations of cationic lipids for efficient gene transfer to the lung. Hum Gene Ther 1996;7:1701e17. Hebel H, Attra H, Khan A, Draghia-Akli R. Successful parallel development and integration of a plasmid-based biologic, container/closure system and electrokinetic delivery device. Vaccine 2006;24:4607e14. Emerson M, Renwick L, Tate S, Rhind S, Milne E, Painter HA, et al. Transfection efficiency and toxicity following delivery of naked plasmid DNA and cationic lipid-DNA complexes to ovine lung segments. Mol Ther 2003;8:646e53. Mantovani A, Sica A, Sozzani S, Allavena P, Vecchi A, Locati M. The chemokine system in diverse forms of macrophage activation and polarization. Trends Immunol 2004;25:677e86. Krieg AM. CpG motifs in bacterial DNA and their immune effects. Annu Rev Immunol 2002;20:709e60. Zhao H, Wu I-H, Hemmi H, Akira S, Cheng SH, Scheule RK, et al. Role of the toll-like 9 receptor in the acute inflammatory response to nonviral vectors. Mol Ther 2003;7:S209. Wooddell CI, Subbotin VM, Sebestyen MG, Griffin JB, Zhang G, Schleef M, et al. Muscle damage after delivery of naked plasmid DNA into skeletal muscles is batch dependent. Hum Gene Ther 2011;22:225e35. Cai Y, Rodriguez S, Rameswaran R, Draghia-Akli R, Juba Jr RJ, Hebel H. Production of pharmaceutical-grade plasmids at high concentration and high supercoiled percentage. Vaccine 2010;28:2046e52. Wu F, Levchenko I, Filutowicz M. A DNA segment conferring stable maintenance on R6K gamma-origin core replicons. J Bacteriol 1995;177:6338e45. Muruve DA, Petrilli V, Zaiss AK, White LR, Clark SA, Ross PJ, et al. The inflammasome recognizes cytosolic microbial and host DNA and triggers an innate immune response. Nature 2008;452:103e7. Takaoka A, Wang Z, Choi MK, Yanai H, Negishi H, Ban T, et al. DAI (DLM-1/ ZBP1) is a cytosolic DNA sensor and an activator of innate immune response. Nature 2007;448:501e5. Hagele H, Allam R, Pawar RD, Reichel CA, Krombach F, Anders HJ. Doublestranded DNA activates glomerular endothelial cells and enhances albumin permeability via a toll-like receptor-independent cytosolic DNA recognition pathway. Am J Pathol 2009;175:1896e904. Takaoka A, Taniguchi T. Cytosolic DNA recognition for triggering innate immune responses. Adv Drug Deliv Rev 2008;60:847e57.
© Copyright 2025 Paperzz