12
A Direct Comparison of Two Nonviral Gene Therapy Vectors for Somatic Integration: In Vivo Evaluation of the Bacteriophage Integrase fc31 and the Sleeping Beauty Transposase Anja Ehrhardt, 1 Hui Xu, 1 Zan Huang, 1 Jeffrey A. Engler, 3 and Mark A. Kay 1,2, * 1 Departments of Pediatrics and 2 Genetics, School of Medicine, Stanford University, 300 Pasteur Drive, Stanford, CA 94305, USA 3 Department of Biochemistry and Molecular Genetics, Schools of Medicine and of Dentistry, University of Alabama at Birmingham, Birmingham, AL 35294, USA *To whom correspondence and reprint requests should be addressed. Fax: +1 650 498 6540. E-mail: [email protected]. Available online 26 February 2005 In this study we performed a head-to-head comparison of the integrase fC31 derived from a Streptomyces phage and the Sleeping Beauty (SB) transposase, a member of the TC1/mariner superfamily of transposable elements. Mouse liver was cotransfused with a vector containing our most robust human coagulation factor IX expression cassette and the appropriate recombinase recognition site and either a fC31- or a SB transposase-expressing vector. To analyze transgene persistence and to prove somatic integration in vivo we induced cell cycling of mouse hepatocytes and found that the transgene expression levels dropped by only 16 to 21% and 56 to 66% in mice that received fC31 and SB, respectively. Notably, no difference in the toxicity profile was detected in mice treated with either recombinase. Moreover we observed that with the integrase-mediated gene transfer, transgene expression levels were dependent on the remaining noncoding vector sequences, which also integrate into the host genome. Further analyses of a hot spot of integration after fC31-mediated integration revealed small chromosomal deletions at the target site and that the recombination process was not dependent on the orientation in which the fC31 recognition site attached to the pseudo-recognition sites in the host genome. Coupled together with ongoing improvements in both systems this study suggests that both nonviral vector systems will have important roles in achieving stable gene transfer in vivo. Key Words: Sleeping Beauty , bacteriophage integrase, fC31, nonviral, hemophilia, gene therapy INTRODUCTION Gene therapy approaches for genetic disorders require long- term and sustained transgene expression levels in vivo. Sustained transgene expression levels were obtained in viral-based and nonviral gene therapy approaches [1–9]. However, each gene delivery system displays advantages and disadvantages and the ideal gene therapy technology would result in life-long and therapeutic transgene expression levels in vivo without toxicity. Viral gene therapy vectors have been developed and were demon- strated to result in long-term phenotypic correction in mice and larger animal models for hemophilia B [7,10– 12]. However, major obstacles in viral gene therapy approaches remain. For example the immune response against the incoming viral particle needs to be dimin- ished and the labor-intensive and expensive production of viral vectors is, in contrast to nonviral vectors, a major challenge. Conversely, delivery limitations have dimin- ished the application of nonviral systems. To achieve prolonged transgene expression levels in vivo, nonviral vector systems for somatic integration were developed [5,6] and cis -acting DNA elements were optimized [13,14]. It was demonstrated that nonviral-based vectors with the ability to integrate into the host genome maintain stable transgene expression levels even during rapid cell cycling of mouse hepatocytes in vivo [6,15,16]. Two major nonviral vectors for somatic integration currently used in gene therapy approaches are based on the Sleeping Beauty (SB) transposase or the bacteriophage integrase fC31 from Streptomyces . DNA transposons are genetic elements that have the ability to move from one genetic location to another. SB transposase is a molecular reconstruction from the fish genome and belongs to the TC1/mariner family of transposable elements [17]. The SB recombination process is based on a cut-and-paste ARTICLE doi:10.1016/j.ymthe.2005.01.010 MOLECULAR THERAPY Vol. 11, No. 5, May 2005 695 Copyright C The American Society of Gene Therapy 1525-0016/$30.00

A direct comparison of two nonviral gene therapy vectors ...web.stanford.edu/group/markkaylab/publications/15851008.pdf · influence transgene expression levels in a gene therapy

  • Upload
    others

  • View
    3

  • Download
    0

Embed Size (px)

Citation preview

Page 1: A direct comparison of two nonviral gene therapy vectors ...web.stanford.edu/group/markkaylab/publications/15851008.pdf · influence transgene expression levels in a gene therapy

ARTICLEdoi:10.1016/j.ymthe.2005.01.010

A Direct Comparison of Two Nonviral Gene TherapyVectors for Somatic Integration: In Vivo Evaluation

of the Bacteriophage Integrase fc31 and theSleeping Beauty Transposase

Anja Ehrhardt,1 Hui Xu,1 Zan Huang,1 Jeffrey A. Engler,3 and Mark A. Kay1,2,*

1Departments of Pediatrics and 2Genetics, School of Medicine, Stanford University, 300 Pasteur Drive, Stanford, CA 94305, USA3Department of Biochemistry and Molecular Genetics, Schools of Medicine and of Dentistry, University of Alabama at Birmingham, Birmingham, AL 35294, USA

*To whom correspondence and reprint requests should be addressed. Fax: +1 650 498 6540. E-mail: [email protected].

Available online 26 February 2005

MOLECULA

Copyright C

1525-0016/$

In this study we performed a head-to-head comparison of the integrase fC31 derived from aStreptomyces phage and the Sleeping Beauty (SB) transposase, a member of the TC1/marinersuperfamily of transposable elements. Mouse liver was cotransfused with a vector containing ourmost robust human coagulation factor IX expression cassette and the appropriate recombinaserecognition site and either a fC31- or a SB transposase-expressing vector. To analyze transgenepersistence and to prove somatic integration in vivo we induced cell cycling of mouse hepatocytesand found that the transgene expression levels dropped by only 16 to 21% and 56 to 66% in micethat received fC31 and SB, respectively. Notably, no difference in the toxicity profile was detectedin mice treated with either recombinase. Moreover we observed that with the integrase-mediatedgene transfer, transgene expression levels were dependent on the remaining noncoding vectorsequences, which also integrate into the host genome. Further analyses of a hot spot of integrationafter fC31-mediated integration revealed small chromosomal deletions at the target site and thatthe recombination process was not dependent on the orientation in which the fC31 recognition siteattached to the pseudo-recognition sites in the host genome. Coupled together with ongoingimprovements in both systems this study suggests that both nonviral vector systems will haveimportant roles in achieving stable gene transfer in vivo.

R

Th

30

Key Words: Sleeping Beauty, bacteriophage integrase, fC31, nonviral, hemophilia, gene therapy

INTRODUCTION

Genetherapyapproachesforgeneticdisordersrequirelong-term and sustained transgene expression levels in vivo.Sustained transgene expression levels were obtained inviral-based and nonviral gene therapy approaches [1–9].However, each gene delivery system displays advantagesand disadvantages and the ideal gene therapy technologywould result in life-long and therapeutic transgeneexpression levels in vivo without toxicity. Viral genetherapy vectors have been developed and were demon-strated to result in long-term phenotypic correction inmice and larger animal models for hemophilia B [7,10–12]. However, major obstacles in viral gene therapyapproaches remain. For example the immune responseagainst the incoming viral particle needs to be dimin-ished and the labor-intensive and expensive productionof viral vectors is, in contrast to nonviral vectors, a major

THERAPY Vol. 11, No. 5, May 2005

e American Society of Gene Therapy

.00

challenge. Conversely, delivery limitations have dimin-ished the application of nonviral systems. To achieveprolonged transgene expression levels in vivo, nonviralvector systems for somatic integration were developed[5,6] and cis-acting DNA elements were optimized[13,14]. It was demonstrated that nonviral-based vectorswith the ability to integrate into the host genomemaintain stable transgene expression levels even duringrapid cell cycling of mouse hepatocytes in vivo [6,15,16].

Two major nonviral vectors for somatic integrationcurrently used in gene therapy approaches are based onthe Sleeping Beauty (SB) transposase or the bacteriophageintegrase fC31 from Streptomyces. DNA transposons aregenetic elements that have the ability to move from onegenetic location to another. SB transposase is a molecularreconstruction from the fish genome and belongs to theTC1/mariner family of transposable elements [17]. The SBrecombination process is based on a cut-and-paste

695

Page 2: A direct comparison of two nonviral gene therapy vectors ...web.stanford.edu/group/markkaylab/publications/15851008.pdf · influence transgene expression levels in a gene therapy

ARTICLE doi:10.1016/j.ymthe.2005.01.010

mechanism and results in integration of the transposoninto the chromosomal target site [17,18] (Fig. 1). Afterresurrection of SB it became clear that this transposableelement is suitable for somatic gene therapy approaches.It was demonstrated that SB was functional in mamma-lian cells [19–21] and shown that this recombinaseresulted in persistent transgene expression levels in amouse model of hemophilia B [5]. Recent data suggestthat the target sites of integration after SB-mediatedrecombination show random genomic distribution andit is estimated that about 35% of transposition events arein genes [22]. This is in sharp contrast to integrating viralvectors such as lentivirus and adeno-associated virus(AAV), which predominantly integrate into active genes[23,24].

The bacteriophage integrase fC31 from Streptomyces isa recombinase with the potential to perform site-specificintegration into a host chromosome and it was previ-ously demonstrated that hot spots of integration in themammalian genome exist [6,25,26]. In contrast to otherbacterial-derived recombinases the phage integrase fC31is based on a unidirectional reaction in which two fC31molecules attach to the attB recognition site and the attPpseudo-sites in the mammalian genome (attPV) [27]. Therecombination process is based on a 2-bp staggered cut ofthe DNAs and rotation of the DNA targets by 1808 [27].The integration reaction results in integration of thetransgene flanked by two hybrid att sites, attL and attR(Fig. 1). After it was demonstrated that the phageintegrase fC31 is sufficient to perform site-specificintegration in human cells [25,28], the in vivo efficacyof this recombinase was tested. It was shown that fC31-mediated integration in mice results in sustained andtherapeutic levels of the coagulation factor IX [6].Another study utilized the fC31 recombinase for correc-tion of inherited human skin diseases [26,29].

The goal of this study was to compare directly the invitro and in vivo efficiency of the two recombinases SB andfC31 in the context of an identical expression cassette.We investigated the stability of transgene expression

FIG. 1. Shown schematically is the integration process

mediated by the bacteriophage fC31 (INT) and

Sleeping Beauty (SB) transposase. Left: Two fC31

molecules attach to the attB recognition site and the

attP pseudo-sites in the mammalian genome (attPV)and the substrate with the fC31 attachment site attB

attaches to the attP pseudo-sites. The recombination

process is based on a 2-bp staggered cut of the DNAs

and rotation of the DNA targets by 1808. The

integration reaction results in integration of the

transgene flanked by two hybrid att sites, attL and

attR. Right: SB binds to the inverted repeats (IR) and

integrates the transgene by a cut-and-paste mecha-

nism into the host chromosomal DNA. SB cuts the

transposon in which the transgene is flanked by the

IRs out of the donor plasmid and integrates it into TA

dinucleotide base pairs.

696

levels during rapid cell cycling in mouse liver andanalyzed the toxicity profile of both recombinases shortlyafter administration. Recently, we showed that bacterialbackbone sequences in nonviral vectors influence trans-gene expression levels in vivo [30]. Based on these findingswe addressed the question if bacterial DNA sequencesinfluence transgene expression levels in a gene therapyapproach based on fC31. In contrast to SB-mediatedtransposition, bacterial backbone sequences integrateinto the host genome. Including further ongoingimprovements of the SB transposase and the bacterio-phage fC31, we believe that both recombinases havegreat potential for nonviral gene therapy approaches.

RESULTS

A Direct Comparison of the Recombinases fC31 andSleeping Beauty Transposase in Vitro and in VivoIn an attempt to compare the integration efficiency invitro after phage integrase fC31- and SB transposase-mediated recombination, we performed a colony-formingassay in HeLa cells. All DNA constructs used for this studyare shown in Fig. 2A. Previous studies revealed that the SB-mediated integration efficiency in vitro depends on themolar ratios of the SB-encoding plasmid and the trans-poson due to a recently described overproduction-inhib-ition effect [41]. To use optimal conditions for SB-mediated recombination, we used different molar ratiosof the transposase-encoding DNA and the transposonDNA in a colony-forming assay. To ensure that the sameamount of total DNA was transfected we added theappropriate amount of bfillerQ DNA to the differentgroups. In concordance with previous studies[41] thisexperiment revealed that a transposon-to-transposaseratio of 5:1 resulted in highest transposition efficiency,with a 42-fold increase in colony-forming units comparedto groups that received the mutated version of the SBtransposase (Fig. 2B). Further reduction in the amount ofSB-encoding DNA by using a transposon-to-transposaseratio of 7.5:1 or 10:1 resulted in similar or slightly lower

MOLECULAR THERAPY Vol. 11, No. 5, May 2005

Copyright C The American Society of Gene Therap

y
Page 3: A direct comparison of two nonviral gene therapy vectors ...web.stanford.edu/group/markkaylab/publications/15851008.pdf · influence transgene expression levels in a gene therapy

FIG. 2. A direct in vitro comparison of the integration efficiency mediated by the bacteriophage integrase fC31 and the Sleeping Beauty transposase. (A) To

determine the integration efficiency the following DNA sequences were transfected into HeLa cells to perform a colony-forming assay. The fC31 (INT) cDNA,

mutated fC31 encoding sequence (mINT), SB transposase, and mutated SB transposase (mSB) were driven by the cytomegalovirus immediate-early promoter

(CMV-p). Two groups received a SB expression cassette in which the SB cDNA is driven by either the elongation factor a promoter (EF1a-p) or the

phosphoglycerate kinase promoter (PGK-p). The integrase fC31 and SB substrates conferring G418-resistant growth contained a neo-resistance gene (neo), the

p15 bacterial origin of replication (p15A), the SV40 promoter, and the Tn5 promoter. The recombinase fC31 attachment site attB and the inverted repeats (IR)

for SB-mediated recombination were added to the appropriate substrates. (B) In vitro integration efficiency after SB- and fC31-mediated integration. Shown is a

summary of counted colonies for each group after methylene blue staining. To use optimized conditions for the SB-mediated integration process transposon-to-

transposase (Tn:SB) ratios of 1:1, 1:5, and 5:1 were used. Two groups received DNA encoding the active SB cDNA driven by the EF1a promoter or the PGK

promoter. All groups received the same amount of total DNA and for a direct comparison of the fC31- and the SB-based recombination system the molar

amounts of the appropriate recombinase recognition site-containing plasmid were identical. Experiments were performed in triplicate. (C) Integration

efficiencies after SB-mediated integration using Tn:SB ratios of 5:1, 7.5:1, and 10:1.

ARTICLEdoi:10.1016/j.ymthe.2005.01.010

integration efficiencies, respectively (Fig. 2C). This resultconfirms the overproduction-inhibition effect of thetransposase protein during SB-mediated integration. Ourlaboratory recently found that the strength of thepromoter driving the expression of SB influences persis-tence of transgene expression levels in vivo [32]. Thus, wecompared the cytomegalovirus (CMV) immediate-earlypromoter as a strong promoter driving SB expression withthe less robust phosphoglycerate kinase promoter (PGK-p)and the elongation factor 1a promoter (EF1a-p). For thesegroups the transposon-to-transposase ratio was 1:1. Thiswas the same ratio we used for the colony-forming assay,

MOLECULAR THERAPY Vol. 11, No. 5, May 2005

Copyright C The American Society of Gene Therapy

which determined the in vitro efficacy of a fC31-encodingplasmid and the plasmid with the fC31 recognition site.We found that the PGK promoter, the EF1a promoter, andthe CMV promoter resulted in a 34-, 17-, and 21-foldincreased transposition efficiency, respectively, comparedto the inactive mutated SB transposase (mSB) controlgroup (Fig. 2B). For the fC31-based selection study, weused the substrates pCR-attB-nori and p11, in which theplasmid backbone was deleted. We observed an 18- and39-fold increase for pCR-attB-nori and pAttB-nori, respec-tively, compared to groups that received the mutated andinactive version of fC31 (Fig. 2B). This experiment

697

Page 4: A direct comparison of two nonviral gene therapy vectors ...web.stanford.edu/group/markkaylab/publications/15851008.pdf · influence transgene expression levels in a gene therapy

ARTICLE doi:10.1016/j.ymthe.2005.01.010

implies that the plasmid backbone with bacterial DNAsequences that also integrates into the host genomeduring fC31-mediated integration influences integrationefficiency in vitro. In summary we conclude that underoptimized conditions the integration efficiencies in vitrofor SB and fC31 are similar. SB showed 42-fold increasedtransposition efficiency above background and fC31resulted in a 39-fold increase in integration compared tothe mutant fC31 control group.

In an attempt to compare the in vivo efficacy of bothrecombinases, we compared our most robust humancoagulation factor IX (hFIX) expression cassette in thecontext of both the recombinases fC31 and SB. The hFIXexpression cassette utilized in this study was describedearlier [13,14] and contained the hFIX cDNA with part ofthe first intron of the hFIX gene driven by the human a1-antitrypsin promoter (hAAT-p) and two liver-specificenhancers (Fig. 3A). The in vivo comparison of bothrecombinases was based on a two-vector approach inwhich a nonviral vector containing the hFIX expressioncassette with the recombinase attachment site was

FIG. 3. In vivo properties of the Streptomyces-derived

bacteriophage fC31 and the Sleeping Beauty trans-

posase. The in vivo comparison was based on a two-

vector approach in which the recombinase and the

transgene expression cassette with the conferring

recombinase attachment site were co-injected. (A)

Shown are the DNA sequences transfused into mouse

liver. The human coagulation factor IX (hFIX) expres-

sion cassette contained the hFIX cDNA and part of the

first intron of the hFIX gene driven by the human a1-

antitrypsin promoter (hAAT-p) and two liver-specific

enhancers. The plasmids pCS-attB-hFIX and pTMCS-

hFIX with the hFIX expression cassette contained the

fC31 attachment site attB and the SB inverted repeats

(IR), respectively. The fC31 (INT) cDNA, mutated

fC31-encoding sequence (mINT), SB transposase,

and mutated SB transposase (mSB) were driven by

the CMV immediate-early promoter. (B) Vectors were

transfused into mouse liver (n = 10 per group, C57Bl/

6 mice) using a high-pressure tail vein injection

method. The plasmids pCS-attB-hFIX (20 Ag) and

pTMCS-hFIX (20 Ag) were co-injected with pCMV-INT

(20 Ag) or pCMV-mINT (20 Ag) and pCMV-SB (2.0 +

18.0 Ag pBS-P/P) or pCMV-mSB (2.0 + 18.0 Ag pBS-P/

P), respectively. A two-thirds partial hepatectomy was

performed 63 days postinjection. Transgene expres-

sion levels were monitored at the predicted time

points. Means F SD are shown. (C) The relative serum

hFIX concentrations for the bacteriophage integrase

fC31 and the mutated version of the fC31 recombi-

nase post-partial hepatectomy are shown. (D) The

relative transgene expression levels post-partial hep-

atectomy are shown for the groups that received the

plasmids pCMV-SB and pCMV-mSB.

698

codelivered with a second vector encoding the recombi-nase fC31 (n = 10 per group) or SB (n = 10 per group).Mouse livers of control groups received a mutated andinactive version of the phage integrase fC31 (mINT; n =10 per group) and the SB transposase (mSB; n = 10 pergroup). Although the hFIX expression levels started out athigher levels for the SB group (up to 20,000 ng/ml,normal level 5000 ng/ml), we detected similar serumlevels of hFIX 8 weeks postinjection for the fC31 (1867ng/ml) and the SB group (1489 ng/ml) (Fig. 3B). Toanalyze transgene persistence and to prove somaticintegration in vivo we induced cell cycling of mousehepatocytes by performing a surgical two-thirds partialhepatectomy 66 days postinjection. Eighty-four dayspost-partial hepatectomy we found that the transgeneexpression levels dropped by 16% (1565 ng/ml) and 66%(498 ng/ml) in mice that received the active fC31recombinase and the SB transposase, respectively (Figs.3C and 3D). In contrast to this observation, transgeneexpression dropped by N96% in both the mINT and themSB control groups.

MOLECULAR THERAPY Vol. 11, No. 5, May 2005

Copyright C The American Society of Gene Therapy

Page 5: A direct comparison of two nonviral gene therapy vectors ...web.stanford.edu/group/markkaylab/publications/15851008.pdf · influence transgene expression levels in a gene therapy

ARTICLEdoi:10.1016/j.ymthe.2005.01.010

In Vivo Efficacy of Both Recombinases in the Contextof the Identical Plasmid Backbone SequencesPrevious data from our laboratory revealed that DNAsequences contained in the plasmid backbone of anonviral vector can significantly influence the timecourse and level of transgene expression levels in vivo.Thus, in a further experiment, we compared the in vivoproperties of both recombinases when the transgeneexpression cassettes with the recombinase attachmentsites were delivered in the context of an identical plasmidbackbone. We co-infused nonviral vectors encoding therecombinases and a second vector with the appropriaterecombinase attachment site (Fig. 4A) into the livers ofC57Bl/6 mice (n = 10 per group) by high-pressure tail veininjection. Control groups received a vector expressing amutated version of the bacteriophage integrase fC31 orSB transposase. Expression levels increased and reachedpeak levels 3 weeks postinjection in all four groups (Fig.4B). Interestingly, at this time point, the fC31 groupshowed highest transgene expression, with serum hFIXconcentrations of up to 20,000 ng/ml (Fig. 4B). In sharpcontrast, we observed serum hFIX concentrations of up to1100, 1300, and 1100 ng/ml for the mutant fC31 group,the SB group, and the mutant SB group, respectively. Toinvestigate the integration efficiency, we induced cellcycling in mouse liver by performing a two-thirds partialhepatectomy (Fig. 3) 102 days after vector delivery. Oneday before partial hepatectomy, we measured serum hFIXlevels, and in mice that received the active integrase, wedetected serum hFIX concentrations of up to 11,900 ng/ml (Fig. 4B). In sharp contrast, in mice injected with themutated version of the phage integrase fC31 transgenewe observed expression levels of up to 290 ng/ml. Mouselivers that were infused with either the SB transposase orthe mutated SB recombinase and a hFIX expressioncassette flanked by the inverted repeats expressed serumhFIX concentrations of up to 3200 and 649 ng/ml,respectively, 102 days postinjection (Fig. 4B). Afterperforming a two-thirds surgical hepatectomy we moni-tored hFIX serum levels for an additional 122 days and, inconcordance with the first experiment (Fig. 3), we foundthat transgene expression levels dropped only 21% forthe bacteriophage fC31- and 54% for the SB transposase-treated animals (Fig. 4C). Control groups that receivedthe inactive version of either recombinase showed adecrease in hFIX serum concentrations of N95%.

In an attempt to analyze liver samples for hFIX-encoding vector genomes, we isolated genomic DNA frommouse liver pre- and post-partial hepatectomy and per-formed a Southern blot analysis. As illustrated in Fig. 5, wedetected a higher number of vector genomes in mouseliver isolated from animals that received the vector pBS-attB-hFIX and the active integrase fC31 compared themutant fC31 group. Post-partial hepatectomy and inconcordance with the hFIX expression levels, we detectedvector genomes in the fC31 group but not in the mutant

MOLECULAR THERAPY Vol. 11, No. 5, May 2005

Copyright C The American Society of Gene Therapy

fC31 group (Fig. 5B). In contrast to this finding, weobserved a similar amount of vector genomes in the SB andthe mutant SB transposase groups pre-partial hepatectomy(Fig. 5C). Although hFIX expression could still be detectedin mice that received the active SB transposase, 224 dayspostinjection we detected no vector genomes by theSouthern blot performed in this study. However, a moresensitive method might be required to detect the remain-ing vector genomes in the SB transposase group.

Plasmid Backbone Sequences Influence the in VivoPerformance of the fC31 Recombinase in a NonviralGene Therapy ApproachWe observed that transgene expression levels after fC31-mediated integration varied among the experiments inwhich different plasmids were used for the delivery of theattB site and the transgene expression cassette. Thus, in afurther step we wanted to address the question ofwhether bacterial sequences influence the in vivo per-formance of fC31 in a nonviral gene therapy approach.We injected C57Bl/6 mice (n = 10 per group) with eitherplasmid pCS-attB-hFIX or plasmid pBS-attB-hFIX (Figs.3A and 4A) and co-injected with the fC31-encodingvector pCMV-INT or pCMV-mINT, respectively. Thebacterial plasmid backbone pCS contains, in addition tothe plasmid backbone pBS, an expression cassette encod-ing the kanamycin resistance gene. We found that 8weeks postinjection the hFIX expression levels were up to7.6-fold higher with the pBS backbone compared to theidentical expression cassette in the pCS backbone (Fig. 6).We believe that there are at least two potential explan-ations for this finding: different plasmid backbonesequences influence the integration efficiency and/orplasmid backbone sequences affect transgene expressionlevels after fC31-mediated integration into the hostgenome. Our in vitro study presented in Fig. 2 B suggestedthat plasmid backbone sequences may influence integra-tion efficiencies after fC31-mediated integration. Takentogether these results suggest that the in vitro and in vivoperformance of the fC31 integrase is influenced by DNAsequences contained in the plasmid backbone.

Liver Toxicity Studies Revealed No SignificantDifferences for either the Phage Integrase fC31 or SbTo analyze the toxicity profile of both recombinases wemeasured liver enzymes as an indicator of acute toxicityafter injection of both fC31 and SB. We detected nosignificant differences in alanine aminotransferase activ-ity (SGPT) levels in mice treated with either recombinase 1day after injection (Fig. 7A). C57Bl/6 mice (n = 10 pergroup) in all four groups had values that averaged ataround 600 U/ml. We believe that these elevated SGPTlevels are a consequence of the high-pressure tail veininjection procedure as seen in previous studies [42]. In afurther step, we addressed the question if the integrasefC31 alone (Fig. 7B) or co-injected with the appropriate

699

Page 6: A direct comparison of two nonviral gene therapy vectors ...web.stanford.edu/group/markkaylab/publications/15851008.pdf · influence transgene expression levels in a gene therapy

FIG. 4. A direct in vivo comparison of the phage integrase fC31 system and the Sleeping Beauty transposase system in the context of the identical bacterial

backbone sequences. (A) DNA sequences injected into C57Bl/6 mice. The hFIX expression cassette with the corresponding recombinase recognition site

contained in the nonviral vectors pBS-TMCS-hFIX and pBS-attB-hFIX was identical to the one described for Fig. 1A. However, for this experiment both transgene

expression cassettes were ligated into the identical plasmid backbone pBluescript (pBS). (B) C57Bl/6 mice (n = 10 per group) were co-injected with either pBS-

TMCS-hFIX or pBS-attB-hFIX and the mutated or active version of INT or SB (pCMV-INT or pCMV-mINT and pCMV-SB or pCMV-mSB, respectively). Serum hFIX

concentrations were monitored by performing an hFIX ELISA. A two-thirds partial hepatectomy was conducted 102 days after partial hepatectomy for all groups.

Means F SD are shown. (C) The relative serum hFIX concentrations 1 day pre-partial hepatectomy compared to the transgene expression levels 16 weeks post-

partial hepatectomy are shown.

ARTICLE doi:10.1016/j.ymthe.2005.01.010

substrate (Fig. 7C) results in elevated liver enzymes afterinfusion into mouse liver. The time course revealed thatthere were no significant differences between the differentgroups into which the active integrase (INT), the inactiveintegrase (mINT), or the control plasmid (pBS-P/P) wasinjected (Fig. 7B). SGPT levels returned to normal (normalSGPT levels: 10–80 U/ml) 3 days after infusion. In additionwe found that there were no significant differences inSGPT levels in mice that received the fC31-encodingplasmid either alone or together with a substrate (Fig. 7C).Representative histological findings in liver samplesobtained from one mouse in each group are shown in

700

Fig. 7D. Liver histology sections revealed that the livermicrostructure remained normal in mice that receivedeither recombinase.

Characterization of the Integration Site Mpsl1, a HotSpot for Phage Integrase fC31-Mediated IntegrationRecently, the hot spot for fC31-mediated integration inthe mouse genome, mpsL1, was published [6]. Thisfinding motivated us to analyze further the integrationsite mpsL1 in the present study and to test whether theorientation in which the attB site attaches to the mpsL1site is important. Thus, we performed a PCR that

MOLECULAR THERAPY Vol. 11, No. 5, May 2005

Copyright C The American Society of Gene Therapy

Page 7: A direct comparison of two nonviral gene therapy vectors ...web.stanford.edu/group/markkaylab/publications/15851008.pdf · influence transgene expression levels in a gene therapy

FIG. 5. Southern blot analyses for hFIX-encoding vector genomes in mouse livers that received the fC31 or the SB recombinase. Genomic DNA from mouse liver

pre- and post-partial hepatectomy was isolated. Twenty micrograms of liver genomic DNA was digested with the restriction endonuclease HindIII, and a

Southern blot was performed, which was hybridized with a hFIX cDNA probe (A). (B) Detection of vector genomes in mouse liver, which received the plasmid

pCS-attB-hFIX and the fC31-encoding DNA sequence or a plasmid expressing the mutated fC31 cDNA. (C) Southern blot analyses of mouse liver co-infused

with the plasmid pTMCS-hFIX and an expression cassette encoding either the active SB or the inactive mutant SB.

ARTICLEdoi:10.1016/j.ymthe.2005.01.010

specifically recognized the mpsL1 integration site foreither orientation of the attB site. The strategy of this PCRapproach is schematically demonstrated in Fig. 8A andthe final PCR product of 280 bp for orientation I (Fig. 8A)is shown in Fig. 8B. A detailed characterization of eachintegration event at the mpsL1 site is shown in Fig. 8C. Insummary, we found deletions of up to 33 and 160 bp inthe attB site and the chromosome, respectively (Fig. 8D).In addition we found insertions into the target chromo-some of 6 and 175 bp, of which the 175-bp insertion was

FIG. 6. Transgene expression levels after bacteriophage integrase fC31-

mediated chromosomal integration in the context of different bacterial

backbone sequences. The plasmids pCS-attB-hFIX and pBS-attB-hFIX (Figs.

3A and 4A) were co-infused with either the fC31-encoding plasmid pCMV-

INT or the mutant fC31-expressing plasmid pCMV-mINT, respectively. Serum

hFIX concentrations were monitored by ELISA. Means F SD are shown.

MOLECULAR THERAPY Vol. 11, No. 5, May 2005

Copyright C The American Society of Gene Therapy

derived from the mouse DNA sequence from clone RP23-112C19 on chromosome 11. Sequence analysis of thesePCR products, which were received for both primercombinations, strongly suggested that the fC31-medi-ated recombination process was not dependent on theorientation in which the attB site attaches to the pseudo-recognition site in the host genome. This observation isschematically shown in Fig. 8E.

DISCUSSION

At the current time the SB transposase and the phageintegrase fC31 are available for nonviral gene therapyapproaches that are based on integration of the transgeneexpression cassette into the host genome. The presentstudy compares directly the in vitro and in vivo propertiesof the two recombinases fC31 and SB. Under optimizedconditions for chromosomal integration SB showed 42-fold increased transposition efficiency above backgroundand fC31 resulted in a 39-fold increase in integrationcompared to the mutant fC31 control group. This resultdemonstrates that the in vitro performance of both systemsis similar and that both systems will have important rolesin achieving stable gene transfer. However, there was aclear difference between the in vitro and in vivo perform-ance for both systems. The SB system seems to work betterin vitro than in vivo and the fC31 system supported higherlevels of transgene persistence and expression in vivocompared to the SB system. In addition the fC31 system

701

Page 8: A direct comparison of two nonviral gene therapy vectors ...web.stanford.edu/group/markkaylab/publications/15851008.pdf · influence transgene expression levels in a gene therapy

FIG. 7. Toxicity studies performed after injection of the bacteriophage integrase fC31 and the Sleeping Beauty recombinase. (A) C57Bl/6 mice (n = 10 per group)

were injected with the plasmid pCS-attB-hFIX or pTMCS-hFIX and co-injected with either pCMV-INT and pCMV-mINT or pCMV-SB and pCMV-mSB (Fig. 3A). The

average SGPT enzyme levels 24 h postinjection are shown. (B) SGPT levels after transfusion of mouse liver with the active integrase (INT), the inactive integrase

(mINT), and the control plasmid (pBS-P/P). (C) SGPT levels in mice that received either the fC31-encoding plasmid alone or together with a substrate. (D)

Histopathology of liver after hepatic infusion of either the recombinase fC31 or the SB transposase and the corresponding substrate. Representative histological

findings in liver samples were obtained from one mouse in each group. Hematoxylin and eosin staining of formalin-fixed liver sections from mice co-injected with

pCS-attB-hFIX and pCMV-INT or pCMV-mINT or co-injected with pTMCS-hFIX and pCMV-SB or pCMV-mSB are shown. Original magnification: �20.

ARTICLE doi:10.1016/j.ymthe.2005.01.010

seems to perform better in vivo than in vitro. We speculatethat fC31-mediated integration might be more efficientin quiescent cells in vivo compared to rapidly dividing cellsin vitro. The opposite might be true for SB-mediatedintegration, which might be more efficient in dividingcells in vitro compared to quiescent cells in vivo. Further-more, the integration efficiencies of both recombinasesmay be cell type dependent. In the current study bothrecombinases were compared only in HeLa cells in vitroand in mouse liver in vivo.

We found that after inducing cell cycling in mouseliver, transgene expression levels dropped by 16 or 21%and 66 or 56% in mice that received the active fC31

702

recombinase and the SB transposase, respectively. Incontrast hFIX expression levels dropped by N96% in boththe mutant fC31 and the mutant SB control groups. Inconcordance with these findings we observed a lesspronounced loss of vector genomes in mice that receivedfC31 compared to the SB group. Taken together theseresults may indicate that the mechanism of persistence oftransgene expression levels during mouse hepatocyte cellcycling is more robust in fC31 approaches compared toSB-based gene therapy approaches. One potential reasonfor this observation could be a higher integrationefficiency in mouse hepatocytes after fC31-mediatedrecombination compared to SB-mediated integration.

MOLECULAR THERAPY Vol. 11, No. 5, May 2005

Copyright C The American Society of Gene Therapy

Page 9: A direct comparison of two nonviral gene therapy vectors ...web.stanford.edu/group/markkaylab/publications/15851008.pdf · influence transgene expression levels in a gene therapy

FIG. 8. Characterization at the hot spot of integration after fC31-mediated recombination. To characterize the chromosomal effects we used a PCR strategy that

specifically detects the previously described integration site in mouse liver mpsL1. (A) Schematically shown is the PCR approach to test whether the orientation

(primer combination I and II) in which the attB site attaches to the mpsL1 (PV) integration site is important. The primer attB-F and the nested primer attB-F3 bind

in the attB site and the primers MpsL1 forw and MpsL1 rev bind on the opposite sites of the target site mpsL1 (PVarm). Primers are indicated by arrows. (B) The

final PCR products of which the sequences were analyzed. Shown is an example of the primer combination attB and MpsL1 forw for the first PCR and attB-F3 and

MpsL1 forw for the second PCR (primer combination I). A similar PCR was performed for the primer combination attB and MpsL1 rev and the second PCR was

performed with the primers attB-F3 and MpsL1 rev. (C) Detailed analyses of chromosomal effects at the fC31 integration site. Deletions in the attB arm and the

mpsL1 integration site are indicated. Insertions are underlined. (D) Summary of chromosomal deletions and insertions in the attB arm and the hot spot of

integration mpsL1. Note to 516: Mouse DNA sequence from clone RP23-112C19 on chromosome 11 (Accession No. AL606480). (E) Schematically shown is a

model that indicates that fC31-mediated integration of a transgene is independent of the orientation of the attB-containing vector relative to the pseudo-attP

site in the mouse genome.

ARTICLEdoi:10.1016/j.ymthe.2005.01.010

MOLECULAR THERAPY Vol. 11, No. 5, May 2005 703Copyright C The American Society of Gene Therapy

Page 10: A direct comparison of two nonviral gene therapy vectors ...web.stanford.edu/group/markkaylab/publications/15851008.pdf · influence transgene expression levels in a gene therapy

ARTICLE doi:10.1016/j.ymthe.2005.01.010

Studies in which marker genes are used to determine andquantify the integration efficiency in vivo for bothrecombinases may shed light on that question. It is alsopossible that the relative integration efficiencies in vivoare similar for the SB and the fC31 system but transgenesdelivered by the fC31 system display a better expressionprofile. Another potential explanation for the morerobust transgene expression levels observed with fC31may be the duration and amount of the fC31-expressingvector DNA in mouse hepatocytes. We injected 10-foldmore DNA of the fC31-encoding vector compared to theSB-expressing vector because in contrast to SB [41] fC31appears to have no overproduction-inhibition effect.Thus, the amount of DNA molecules expressing fC31 ismost likely significantly higher than the number of SB-encoding vector molecules, which might increase theintegration efficiency and thus transgene expressionlevels in vivo. In the present study we induced cell cyclingin mouse liver 2 to 3 months postinjection. During thetime prior to induction of cell cycle, episomal DNA mostlikely persists in quiescent cells. Previous studies showedthat up to 10 copies per hepatocyte are still present 10weeks postinjection [16]. Finally, it needs to be analyzedif transgenes delivered by the fC31 system have a morerobust mechanism to persist extrachromosomally com-pared to transgenes that were introduced into mousehepatocytes by the SB system.

We found microdeletions of up to 160 bp after fC31-mediated integration at the attB site and the previouslypublished chromosomal hot spot of integration in mouseliver, mpsL1 [6]. In concordance with our investigationmicrodeletions in the attB arm of the integration sitewere also detected in a previous in vitro study [25]. Insharp contrast to this phenomenon, chromosomal dele-tions after SB-mediated integration are rare. Character-ization of chromosomal integration sites of naturallyintegrating viral vectors, such as AAV, revealed thatdeletions of even larger than 200 bp can occur [23,43].However, the PCR strategy used in our study to charac-terize the integration sites is able to pick up deletionsand/or insertions of only up to ~200 bp. A morethorough study that characterizes the integration sitesafter fC31-mediated integration in detail will address thequestion if larger deletions at the chromosomal targetsite occur.

This study suggested that the in vivo performance afterfC31-mediated integration may be dependent on theremaining plasmid backbone sequences, which, in addi-tion to the transgene expression cassette, ultimatelyintegrate into the host genome (Fig. 5). We speculatethat there are several potential explanations for thisfinding. (1) DNA sequences contained in the plasmidbackbone influence the fC31-mediated integration effi-ciency and/or (2) plasmid backbone sequences influencetransgene expression levels after integration of thetransgene into the host genome and/or (3) plasmid

704

backbone sequences play a role in transgene persistenceand duration of transgene expression from episomalvector DNA. Our in vitro study presented in Fig. 2Bsuggested that plasmid backbone sequences may indeedplay an important role in integration efficiency. Thecolony-forming assay revealed a twofold increase inG418-resistant colonies if the plasmid backbone wascompletely deleted from the circular substrate forfC31-mediated integration. Previous studies demonstra-ted that bacterial backbone sequences have the potentialto influence significantly the duration of transgeneexpression levels in a nonviral gene therapy approach[30,44]. There is strong evidence that bacterial DNAsequences in a nonviral vector may play a role inepisomal transgene silencing [30]. We propose thatintegrated bacterial sequences may be involved insilencing of the transgene expression cassette or mayfunction as insulators. In addition low-level leakyexpression of bacterial DNA-encoded transcripts mayoccur, which may interfere with the transcriptionalproducts of the transgene expression cassette. Toimprove further nonviral integrating vectors based onfC31 it may be of interest to generate a vector devoid ofall bacterial sequences. Recent studies utilizing episomalminicircles that are devoid of bacterial sequences resultin significantly increased transgene expression levelscompared to nonviral vectors that still contain bacterialDNA backbone sequences [45].

The current study may indicate that a gene therapyapproach in vivo based on fC31 may be superior to agene therapy protocol that utilizes the wild-type SBtransposase. However, based on our in vitro study andrecent publications [32,41,46] it is extremely importantto point out that recent developments in the trans-poson technology may dramatically improve genetherapy approaches based on this type of recombinase.Recently, a hyperactive SB transposase was developed.Mutational analyses of the N-terminal DNA bindingdomain revealed hyperactive mutants with about asixfold increased activity in vivo [46]. Thus, a directcomparison of the hyperactive SB and the fC31recombinase would be of great interest. Moreover itwas recently demonstrated that the promoter strengthstrongly influences transposition efficiencies in vivo [32].In addition, more safety issues for fC31-mediatedrecombination in mammalian cells need be addressed.Careful studies about target site selection of fC31 andother members of this recombinase family need to beperformed and a detailed characterization of integrationsites in mammalian cells needs to be undertaken. Takentogether with ongoing improvements in both systemsincluding the recent development of transposase hyper-mutants, this study suggests that both nonviral vectorsystems will have an important role in achieving stablegene transfer in vivo but can be improved further withoptimization.

MOLECULAR THERAPY Vol. 11, No. 5, May 2005

Copyright C The American Society of Gene Therapy

Page 11: A direct comparison of two nonviral gene therapy vectors ...web.stanford.edu/group/markkaylab/publications/15851008.pdf · influence transgene expression levels in a gene therapy

ARTICLEdoi:10.1016/j.ymthe.2005.01.010

MATERIALS AND METHODS

DNA constructs. The plasmid pBS-ApoEHCR-hAATp-FIX-Int-bpA, con-

taining the hFIX expression cassette in which a hFIX minigene is driven

by the hAAT-p and two liver-specific enhancers, was previously described

[13,14]. The plasmids pCR-attB (pTA-attB) [6,28] and pT-MCS [5] were

described elsewhere. The SpeI fragment from pBS-ApoEHCR-hAATp-FIX-

Int-bpA containing the hFIX expression cassette was cloned into either

the SpeI site of pCR-attB or the SpeI site of pT-MCS, resulting in pCR-attB-

hFIX and pT-MCS-hFIX, respectively. The EcoRI fragment from pCR-attB

with the 308-bp fC31 recognition site was subcloned into the EcoRI site

of pBluescript K/S (pBS K/S; Stratagene) followed by ligation of the SpeI

fragment from pBS-ApoEHCR-hAATp-FIX-Int-bpA containing the hFIX

expression cassette blunt-ended into the EcoRV site, resulting in pBS-attB-

hFIX. The KpnI–XbaI fragment obtained from pT-MCS with the inverted

repeats (IRs) was subcloned into the EcoRV site of pBS S/K by blunt-end

ligation, resulting in pBS-TMCS. The SpeI fragment containing the DNA

sequences for the hFIX expression cassette was cloned blunt into the NsiI

and NotI sites of pBS-TMCS, resulting in pBS-TMCS-hFIX.

The plasmids pCR-attB-nori and pTMCS-Snori were used to determine

the integration efficiency in vitro by performing a colony-forming assay.

To produce the plasmid pCR-attB-nori the BsmI fragment from pAAV-

Snori [31] with the selection marker and the p15 bacterial origin of

replication was cloned blunt into the EcoRV site of pCR-attB. The plasmid

p11, which is deleted for the plasmid backbone, was based on pCR-attB-

nori and was generated by digesting pCR-attB-nori with SpeI and NotI

followed by a fill-in reaction to create blunt ends and religation. The

plasmid pTnori [5] was previously described and contained the identical

selection marker flanked by the SB IRs.

The plasmid pThAAT.PGK.SB.uni was described previously [32] and

contains an expression cassette for SB10 in which SB cDNA is driven by

the PGK promoter. The plasmid pHM5.lox.EF1a.SB10 encoding SB10

under the transcriptional control of the EF1a-p is based on the

previously described vector pHM5 [33,34]. The plasmids pCMV-SB,

expressing the SB transposase from the CMV immediate-early promoter,

and pCMV-mSB expressing an inactive version of the SB transposase,

into which an aspartate Y alanine (D244A) mutation was introduced

into the catalytic D,D(35)E domain, have been described [5]. The fC31

integrase expression plasmid pCMV-INT, in which the fC31 cDNA is

driven by the CMV promoter, has been used in a previous study [28]. The

plasmid pCMV-mINT, expressing an inactive version of the integrase

fC31, was a gracious gift from Michele P. Calos. This mutated version of

the phage integrase fC31, which contains an S12F mutation, was

described earlier [35].

In vitro studies. HeLa cells were cultured in DMEM supplemented with

10% fetal bovine serum, 100 units/ml penicillin–streptomycin, and 20

mM l-glutamine. To perform a colony-forming assay cells were seeded in

6-cm dishes and, at 50 to 80% confluency, transfected with the plasmid

DNA using Superfect (Gibco BRL). For all in vitro studies, the same amount

of total DNA was transfected and as bfillerQ DNA the plasmid pBS-P/P [2]

was used. The molar ratios of the recombinase-encoding plasmid and a

second plasmid with the recombinase recognition site were 1:1 for fC31

and 1:1, 1:5, and 5:1 (transposon-to-transposase ratio) for the SB trans-

posase. The transposon-to-transposase ratio for groups that received a

plasmid in which SB is driven by the PGK promoter or the EF1a promoter

was 1:1. For a direct comparison of the fC31- and the SB-mediated

integration efficiencies the same molar amounts of the appropriate

recombinase attachment site were transfected. We transfected 1 Ag of

the plasmid pTnori per 6-cm dish and the molar amounts of recombinase-

encoding DNA and plasmids with the recombinase attachment sites in

other groups were adjusted accordingly. Forty-eight hours after trans-

fection cells were split into 10-cm dishes at two different densities (3.8 �104 and 3.8 � 105 cells/10-cm dish). After 14 days of G418 selection (500

mg/ml) colonies were stained with methylene blue and counted.

Animal procedures. C57Bl/6 mice (ca. 8 weeks of age) were injected via

high-pressure tail vein injection as previously described [36,37]. In brief,

the plasmid DNA was diluted in 2.0 ml of a physiological 0.9% (w/v)

MOLECULAR THERAPY Vol. 11, No. 5, May 2005

Copyright C The American Society of Gene Therapy

NaCl solution and injected into the mouse tail vein over a period of 5–7

s. The plasmids pCS-attB-hFIX or pBS-attB-hFIX (20 Ag) and pTMCS-

hFIX or pBS-TMCS-hFIX (20 Ag) were co-injected with pCMV-INT (20

Ag) or pCMV-mINT (20 Ag) and pCMV-SB (2.0 + 18.0 Ag pBS-P/P [2]) or

pCMV-mSB (2.0 + 18.0 Ag pBS-P/P), respectively. To obtain serum

samples blood was collected by retro-orbital technique at the predicted

time points. Surgical two-thirds partial hepatectomies were performed

as described earlier [38].

Blood analyses. Mouse serum human FIX antigen levels were determined

by an enzyme-linked immunosorbent assay (ELISA) as previously

described [2,13]. The normal human serum level was 5000 ng/ml. SGPT

assays were performed by using a diagnostic kit for colorimetric

determination of SGPT (TECO Diagnostics, Anaheim, CA, USA; Cat. No.

A526-120).

Southern blot analysis. Genomic DNA from mouse liver was isolated by

using a previously described protocol [39]. In brief about 100 mg of mouse

liver was harvested and digested overnight with proteinase K. Genomic

DNA was extracted by a previously described salting-out procedure

followed by an ethanol precipitation. Twenty micrograms of genomic

mouse liver DNA was digested with the restriction enzyme nuclease

HindIII, run on a 0.8% agarose gel, and electrotransferred to a Hybond

membrane (Amersham). The blots were hybridized with an [a-32P]dCTP-

labeled cDNA hFIX probe (HindIII/EcoRI fragment from pAAVCM2), using

a random priming kit (Stratagene).

Detection and characterization of the mpsL1 integration sites in mouse

liver. Genomic DNA from mouse liver was isolated and a PCR with the

two mpsL1 integration site-specific primers, MpsL1 forward (5V-GTGGCA-

CATTCCTTAATCCC-3V) and MpsL1 reverse (5V-TGAGGAGGAGCCTTAG-

CAAC-3V), which were combined with the attB-specific primer AttB-F

(5V-TACCGTCGACGATGTAGGTCACGGTC-3V), was performed. This

PCR was followed by a second PCR in which the primer attB-F3

(5V-CGAAGCCGCGGTGCG-3V) was combined with either MpsL1 forward

or MpsL1 reverse. To determine the DNA sequences of the resulting PCR

products the DNA fragments were subcloned using a TA cloning kit

(Invitrogen) and sequenced.

Histology procedures. Liver sections were generated as described earlier

[40]. In brief, mouse liver was fixed in 10% paraformaldehyde and

embedded in paraffin. Tissue sections were generated and hematoxylin

and eosin staining was performed.

ACKNOWLEDGMENTS

The authors thank Michele P. Calos for providing the plasmids pCS-attB, pCMV-

INT, and pCMV-mINT. This work was supported by NIH Grant DK49022 to

M.A.K.

RECEIVED FOR PUBLICATION JULY 20, 2004; ACCEPTED JANUARY 9, 2005.

REFERENCES1. Schiedner, G., et al. (1998). Genomic DNA transfer with a high-capacity adenovirus

vector results in improved in vivo gene expression and decreased toxicity. Nat. Genet.

18: 180 – 183.

2. Ehrhardt, A., and Kay, M. A. (2002). A new adenoviral helper-dependent vector results

in long-term therapeutic levels of human coagulation factor IX at low doses in vivo.

Blood 99: 3923 – 3930.

3. Snyder, R. O., et al. (1997). Persistent and therapeutic concentrations of human factor

IX in mice after hepatic gene transfer of recombinant AAV vectors. Nat. Genet. 16:

270 – 276.

4. Park, F., Ohashi, K., and Kay, M. A. (2000). Therapeutic levels of human factor VIII and

IX using HIV-1-based lentiviral vectors in mouse liver. Blood 96: 1173 – 1176.

5. Yant, S. R., et al. (2000). Somatic integration and long-term transgene expression in

normal and haemophilic mice using a DNA transposon system. Nat. Genet. 25: 35 – 41.

6. Olivares, E. C., et al. (2002). Site-specific genomic integration produces therapeutic

factor IX levels in mice. Nat. Biotechnol. 20: 1124 – 1128.

7. Snyder, R. O., et al. (1999). Correction of hemophilia B in canine and murine models

using recombinant adeno-associated viral vectors. Nat. Med. 5: 64 – 70.

8. Kafri, T., et al. (1997). Sustained expression of genes delivered directly into liver and

muscle by lentiviral vectors. Nat. Genet. 17: 314 – 317.

705

Page 12: A direct comparison of two nonviral gene therapy vectors ...web.stanford.edu/group/markkaylab/publications/15851008.pdf · influence transgene expression levels in a gene therapy

ARTICLE doi:10.1016/j.ymthe.2005.01.010

9. Morral, N., et al. (1998). High doses of a helper-dependent adenoviral vector yield

supraphysiological levels of alpha1-antitrypsin with negligible toxicity. Hum. Gene Ther.

9: 2709 – 2716.

10. Harding, T. C., et al. (2004). Intravenous administration of an AAV-2 vector for the

expression of factor IX in mice and a dog model of hemophilia B. Gene Ther. 11:

204 – 213.

11. Mount, J. D., et al. (2002). Sustained phenotypic correction of hemophilia B dogs with

a factor IX null mutation by liver-directed gene therapy. Blood 99: 2670 – 2676.

12. Herzog, R. W., et al. (1999). Long-term correction of canine hemophilia B by gene

transfer of blood coagulation factor IX mediated by adeno-associated viral vector. Nat.

Med. 5: 56 – 63.

13. Ehrhardt, A., et al. (2003). Optimization of cis-acting elements for gene expression

from nonviral vectors in vivo. Hum. Gene Ther. 14: 215 – 225.

14. Miao, C. H., et al. (2000). Inclusion of the hepatic locus control region, an intron, and

untranslated region increases and stabilizes hepatic factor IX gene expression in vivo

but not in vitro. Mol. Ther. 1: 522 – 532.

15. Nakai, H., et al. (2001). Extrachromosomal recombinant adeno-associated virus vector

genomes are primarily responsible for stable liver transduction in vivo. J. Virol. 75:

6969 – 6976.

16. Chen, Z. Y., et al. (2001). Linear DNAs concatemerize in vivo and result in sustained

transgene expression in mouse liver. Mol. Ther. 3: 403 – 410.

17. Ivics, Z., et al. (1997). Molecular reconstruction of Sleeping Beauty, a Tc1-like

transposon from fish, and its transposition in human cells. Cell 91: 501 – 510.

18. Izsvak, Z., et al. (2002). Involvement of a bifunctional, paired-like DNA-binding domain

and a transpositional enhancer in Sleeping Beauty transposition. J. Biol. Chem. 277:

34581 – 34588.

19. Belur, L. R., et al. (2003). Gene insertion and long-term expression in lung mediated by

the Sleeping Beauty transposon system. Mol. Ther. 8: 501 – 507.

20. Ortiz-Urda, S., et al. (2003). Sustainable correction of junctional epidermolysis bullosa

via transposon-mediated nonviral gene transfer. Gene Ther. 10: 1099 – 1104.

21. Luo, G., et al. (1998). Chromosomal transposition of a Tc1/mariner-like element in

mouse embryonic stem cells. Proc. Natl. Acad. Sci. USA 95: 10769 – 10773.

22. Vigdal, T. J., et al. (2002). Common physical properties of DNA affecting target site

selection of sleeping beauty and other Tc1/mariner transposable elements. J. Mol. Biol.

323: 441 – 452.

23. Nakai, H., et al. (2003). AAV serotype 2 vectors preferentially integrate into active

genes in mice. Nat. Genet. 34: 297 – 302.

24. Schroder, A. R., et al. (2002). HIV-1 integration in the human genome favors active

genes and local hotspots. Cell 110: 521 – 529.

25. Thyagarajan, B., et al. (2001). Site-specific genomic integration in mammalian cells

mediated by phage fC31 integrase. Mol. Cell. Biol. 21: 3926 – 3934.

26. Ortiz-Urda, S., et al. (2002). Stable nonviral genetic correction of inherited human skin

disease. Nat. Med. 8: 1166 – 1170.

27. Groth, A. C., and Calos, M. P. (2004). Phage integrases: biology and applications. J. Mol.

Biol. 335: 667 – 678.

706

28. Groth, A. C., et al. (2000). A phage integrase directs efficient site-specific integration in

human cells. Proc. Natl. Acad. Sci. USA 97: 5995 – 6000.

29. Ortiz-Urda, S., et al. (2003). fC31 integrase-mediated nonviral genetic correction of

junctional epidermolysis bullosa. Hum. Gene Ther. 14: 923 – 928.

30. Chen, Z. Y., et al. (2004). Silencing of episomal transgene expression by plasmid

bacterial DNA elements in vivo. Gene Ther. 11: 856 – 864.

31. Rutledge, E. A., and Russell, D. W. (1997). Adeno-associated virus vector integration

junctions. J. Virol. 71: 8429 – 8436.

32. Mikkelsen, J. G., et al. (2003). Helper-independent Sleeping Beauty transposon–

transposase vectors for efficient nonviral gene delivery and persistent gene expression

in vivo. Mol. Ther. 8: 654 – 665.

33. Mizuguchi, H., and Kay, M. A. (1998). Efficient construction of a recombinant

adenovirus vector by an improved in vitro ligation method. Hum. Gene Ther. 9:

2577 – 2583.

34. Mizuguchi, H., and Kay, M. A. (1999). A simple method for constructing E1- and E1/

E4-deleted recombinant adenoviral vectors. Hum. Gene Ther. 10: 2013 – 2017.

35. Thorpe, H. M., and Smith, M. C. (1998). In vitro site-specific integration of

bacteriophage DNA catalyzed by a recombinase of the resolvase/invertase family.

Proc. Natl. Acad. Sci. USA 95: 5505 – 5510.

36. Liu, F., Song, Y., and Liu, D. (1999). Hydrodynamics-based transfection in animals by

systemic administration of plasmid DNA. Gene Ther. 6: 1258 – 1266.

37. Zhang, G., Budker, V., and Wolff, J. A. (1999). High levels of foreign gene expression in

hepatocytes after tail vein injections of naked plasmid DNA. Hum. Gene Ther. 10:

1735 – 1737.

38. Ehrhardt, A., Xu, H., and Kay, M. A. (2003). Episomal persistence of recombinant

adenoviral vector genomes during the cell cycle in vivo. J. Virol. 77: 7689 – 7695.

39. Vrancken-Peeters, M. J. T. F. D., et al. (1996). A method for multiple portal vein

infusions in mice: quantitation of adenovirus-mediated hepatic gene transfer.

BioTechniques 20: 278 – 285.

40. Ehrhardt, A., et al. (2003). A gene-deleted adenoviral vector results in phenotypic

correction of canine hemophilia B without liver toxicity or thrombocytopenia. Blood

102: 2403 – 2411.

41. Geurts, A. M., et al. (2003). Gene transfer into genomes of human cells by the Sleeping

Beauty transposon system. Mol. Ther. 8: 108 – 117.

42. Miao, C. H., et al. (2001). Long-term and therapeutic-level hepatic gene expression of

human factor IX after naked plasmid transfer in vivo. Mol. Ther. 3: 947 – 957.

43. Miller, D. G., Rutledge, E. A., and Russell, D. W. (2002). Chromosomal effects of adeno-

associated virus vector integration. Nat. Genet. 30: 147 – 148.

44. Yew, N. S., et al. (2002). CpG-depleted plasmid DNA vectors with enhanced safety and

long-term gene expression in vivo. Mol. Ther. 5: 731 – 738.

45. Chen, Z. Y., et al. (2003). Minicircle DNA vectors devoid of bacterial DNA result in

persistent and high-level transgene expression in vivo. Mol. Ther. 8: 495 – 500.

46. Yant, S. R., et al. (2004). Mutational analysis of the N-terminal DNA-binding domain of

Sleeping Beauty transposase: critical residues for DNA binding and hyperactivity in

mammalian cells. Mol. Cell. Biol. 24: 9239 – 9247.

MOLECULAR THERAPY Vol. 11, No. 5, May 2005

Copyright C The American Society of Gene Therapy