Evaluating risks of insertional mutagenesis by DNA transposons in gene therapy

Perry B Hackett, David A Largaespada, Kirsten C Switzer, Laurence J N Cooper, Perry B Hackett, David A Largaespada, Kirsten C Switzer, Laurence J N Cooper

Abstract

Investigational therapy can be successfully undertaken using viral- and nonviral-mediated ex vivo gene transfer. Indeed, recent clinical trials have established the potential for genetically modified T cells to improve and restore health. Recently, the Sleeping Beauty (SB) transposon/transposase system has been applied in clinical trials to stably insert a chimeric antigen receptor (CAR) to redirect T-cell specificity. We discuss the context in which the SB system can be harnessed for gene therapy and describe the human application of SB-modified CAR(+) T cells. We have focused on theoretical issues relating to insertional mutagenesis in the context of human genomes that are naturally subjected to remobilization of transposons and the experimental evidence over the last decade of employing SB transposons for defining genes that induce cancer. These findings are put into the context of the use of SB transposons in the treatment of human disease.

Copyright © 2013 Mosby, Inc. All rights reserved.

Figures

Fig. 1. Genetic consequences of integration of…
Fig. 1. Genetic consequences of integration of therapeutic vectors into genomes
In this instance a transposon vector is illustrated, but the considerations apply to any vector system. A therapeutic transposon can integrate into four general categories of chromatin. Heterochromatin will suppress expression of the transgene – no gain-of-function (GOF) or loss-of-function (LOF) will occur. The most desirable integration events will be into intergenic regions where the Therapeutic Gene (TG) will be expressed. Integration into or proximal to a transcriptional regulatory region can have several outcomes including GOF of the transgenic cassette, as well as either enhancement or loss of expression of the neighboring gene (Gene X). As reviewed in the text, in some cases the transcriptional regulatory elements of the transgene can activate quiescent chromosomal genes. Integration of the transposon into a transcriptional unit may allow expression of the transgene but block expression of the host gene leading to a phenotypic loss of function due to blockage of the gene or alterations in splicing. Integration within some genes can also lead to a dominant gain-of-function (DGF) and/or production of a dominant-negative form (DNF) of the original Gene X.
Fig. 2. DNA transposition
Fig. 2. DNA transposition
DNA transposition, as exemplified by the Sleeping Beauty (SB) transposon system, is a cut-and-paste reaction in which a transposon containing an expression cassette with a therapeutic gene (TG) and its promoter (pentagon) is delivered to target cells wherein the transposon is cut out of the plasmid and inserted into a chromosome. The inverted terminal repeats (inverted set of double arrowheads) define a transposon. The second part of the SB system is SB transposase, which in this example is carried in a separate expression cassette that is on the same plasmid, but not in the transposon. The SB transposon will only integrate into TA-dinucleotide basepairs (about 200 million in mammalian genomes). 1) The plasmid is delivered into a cell by any of several means and proceeds through the nuclear membrane (dotted oval) by a poorly understood process. 2) The SB gene is expressed. 3) The transposase molecules enter the nucleus and bind to the transposon. 4) Four transposases work in concert to cut the transposon out of the plasmid and paste it (dotted lines) into an AT sequence in chromatin (tangled line). A plasmid excision product is left behind in this reaction (the transposon site is marked by an X). Integration into a chromosome can confer sustained expression of the gene of interest that is contained within the transposon.
Fig. 3. Potential consequences of remobilization of…
Fig. 3. Potential consequences of remobilization of SB transposons
The schematic illustrates the initial transposon integration site (orange) and subsequent hopping to other TA sites in chromatin, one of which might induce an adverse transforming event.
Fig. 4. The T2/onc transposon vector
Fig. 4. The T2/onc transposon vector
This SB vector contains elements designed to elicit either transcriptional activation (Mouse Stem Cell Virus 5'-LTR and splice donor [SD]) or inactivation (splice acceptors [SA] and polyadenylation signals [pA]). The inverted terminal repeats are indicated by the arrows labeled ITR
Figure 5
Figure 5
Using SB for cancer-gene screens in mice. Step 1. Breed SB transposon and transposase transgenes together. In some cases, arrangements for tissue-specific expression of the transposase will be made or specific cancer-predisposed backgrounds could be used. Step 2. Transposition in somatic cells causes random insertion mutations. A correctly designed transposon vector can cause gain or loss-of-function mutations. Step 3. Mice are aged for tumor development. Step 4. Tumors develop as a result of transposon-induced mutations. Step 5. Transposon insertions are cloned from tumor genomic DNA. Step 6. Clones are sequenced. Step 7. Insertion sites are mapped and annotated with respect to nearest genes. Those genes repeatedly mutated in multiple, independent tumors are designated as common insertion sites or CIS. Step 8. CIS can be analyzed to determine what genes and genetic pathways contribute to cancer. Cancer genetic fingerprints are obtained that can be characterized by networks of interacting cancer-gene mutations. These genes can be queried in relevant human cancers.

References

    1. Wagner EF, Covarrubias L, Stewart TA, Mintz B. Prenatal lethalities in mice homozygous for human growth hormone gene sequences integrated in the germ line. Cell. 1983;35:647–655.
    1. Hacein-Bey-Abina S, et al. Sustained correction of X-linked severe combined immunodeficiency by ex vivo gene therapy. New Eng J Med. 2002;346:1185–1193.
    1. Hacein-Bey-Abina S, et al. A serious adverse event after successful gene therapy for X-linked severe combined immunodeficiency. New Engl J Med. 2003;348:255–256.
    1. Hacein-Bey-Abina S, Hauer J, Lim A, Picard C, Wang GP, Berry CC, Martinache C, Rieux-Laucat F, Latour S, Belohradsky BH, Leiva L, Sorensen R, Debré M, Casanova JL, Blanche S, Durandy A, Bushman FD, Fischer A, Cavazzana-Calvo M. Efficacy of gene therapy for X-linked severe combined immunodeficiency. New Eng J Med. 2010;363:355–364.
    1. Hacein-Bey-Abina S, et al. LMO2-associated clonal T cell proliferation in two patients after gene therapy for SCID-X1. Science. 2003;302:415–419.
    1. McCormack MP, Forster A, Drynan L, Pannell R, Rabbitts TH. The LMO2 T-cell oncogene is activated via chromosomal translocations or retroviral insertion during gene therapy but has no mandatory role in normal T-cell development. Mol Cell Biol. 2003;23:9003–9013.
    1. Dave' UP, Jenkins NA, Copeland NG. Gene therapy insertional mutagenesis insights. Science. 2004;303:33.
    1. McCormack MP, Rabbitts TH. Activation of the T-cell oncogene LMO2 after gene therapy for X-linked severe combined immunodeficiency. New Eng J Med. 2004;350:913–922.
    1. Nam CH, Rabbitts TH. The role of LMO2 in development and in T cell leukemia after chromosomal translocation or retroviral insertion. Mol Ther. 2006;13:15–25.
    1. Woods NB, Bottero V, Schmidt M, von Kalle C, Verma IM. Gene therapy: therapeutic gene causing lymphoma. Nature. 2006;440:1123.
    1. Gaspar HB, et al. Long-term persistence of a polyclonal T cell repertoire after gene therapy fo rX-linked severe combined immunodeficiency. Sci Transl Med. 2011;3:97ra79.
    1. Ott MG, et al. Correction of X-linked chronic granulomatous disease by gene therapy, augmented by insertional activation of MDS1-EVI1, PRDM16 or SETBP1. Nat Med. 2006;5:401–409.
    1. Kang EM, Choi U, Theobald N, Linton G, Long Priel DA, Kuhns D, Malech HL. Retrovirus gene therapy for X-linked chronic granulomatous disease can achieve stable long-term correction of oxidase activity in peripheral blood neutrophils. Blood. 2010;115:783–791.
    1. Stein S, Ott MG, Schultze-Strasser S, Jauch A, Burwinkel B, Kinner A, Schmidt M, Krämer A, Schwäble J, Glimm H, Koehl U, Preiss C, Ball C, Martin H, Göhring G, Schwarzwaelder K, Hofmann WK, Karakaya K, Tchatchou S, Yang R, Reinecke P, Kühlcke K, Schlegelberger B, Thrasher AJ, Hoelzer D, Seger R, von Kalle C, Grez M. Genomic instability and myelodysplasia with monosomy 7 consequent to EVI1 activation after gene therapy for chronic granulomatous disease. Nature Med. 2010;16:198–204.
    1. Baum C, et al. Side effects of retroviral gene transfer into hematopoietic stem cells. Blood. 2003;101:2099–2114.
    1. Baum C, Fehse B. Mutagenesis by retroviral transgene insertion: risk assessment and potential alternatives. Curr Opin Mol Ther. 2003;5:458–462.
    1. Baum C, et al. Chance or necessity? Insertional mutagenesis in gene therapy and its consequences. Mol Ther. 2004;9:5–13.
    1. Kustikova O, et al. Clonal dominance of hematopoietic stem cells triggered by retroviral gene marking. Science. 2005;308:1171–1174.
    1. Muul L, et al. Persistence and expression of the adenosine deaminase gene for 12 years and immune reaction to gene transfer components: long-term results of the first clinical gene therapy trial. Blood. 2003;101:2563–2569.
    1. Schmidt M, et al. Clonality analysis after retroviral-mediated gene transfer to CD34+ cells from the cord blood of ADA-deficient SCID neonates. Nature Med. 2003;9:463–468.
    1. Aiuti A, Ficara F, Cattaneo F, Bordignon C, Roncarolo MG. Gene therapy for adenosine deaminase deficiency. Curr Opin Allergy Clin Immunol. 2004;3:461–466.
    1. Gaspar HB, et al. Successful reconstitution of immunity in ADA-SCID by stem cell gene therapy following cessation of PEG-ADA and use of mild preconditioning. Mol Ther. 2006;14:505–513.
    1. Aiuti A, et al. Multilineage hematopoietic reconstitution without clonal selection in ADA-SCID patients treated with stem cell gene therapy. J Clin Invest. 2007;117:2233–2240.
    1. Cassani B, et al. Integration of retroviral vectors induces minor changes in the transcriptional activity of T cells from ADA-SCID patients treated with gene therapy. Blood. 2009;114:3546–3556.
    1. Boztug K, Schmidt M, Schwarzer A, Banerjee PP, Díez IA, Dewey RA, Böhm M, Nowrouzi A, Ball CR, Glimm H, Naundorf S, Kühlcke K, Blasczyk R, Kondratenko I, Maródi L, Orange JS, von Kalle C, Klein C. Stem-cell gene therapy for the Wiskott-Aldrich Syndrome. New Eng J Med. 2010;363:1918–1927.
    1. Buonamici S, Chakraborty S, Senyuk V, Nucifora G. The role of EVI1 in normal and leukemic cells. Blood Cells Mol Dis. 2003;31:206–212.
    1. Buonamici S, et al. EVI1 induces myelodysplastic syndrome in mice. J Clin Invest. 2004;114:713–719.
    1. Li Z, et al. Murine leukemia induced by retroviral gene marking. Science. 2002;296:497.
    1. Biasco L, Baricordi C, Aiuti A. Retroviral integrations in gene therapy trials. Mol Ther. 2012;20:709–716.
    1. Gabriel R, Schmidt M, von Kalle C. Integration of retroviral vectors. Curr Opin Immunol. 2012;24:592–597.
    1. Scholler J, et al. Decade-long safety and function of retroviral-modified chimeric antigen receptor T cells. Sci Transl Med. 2012;4:132ra153.
    1. van der Loo JC SW, Grassman E, Terwilliger A, Higashimoto T, Schambach A, Hacein-Bey-Abina S, Nordling DL, Cavazzana-Calvo M, Thrasher AJ, Williams DA, Reeves L, Malik P. Critical variables affecting clinical-grade production of the self-inactivating gamma-retroviral vector for the treatment of X-linked severe combined immunodeficiency. Gene Ther. 2012;19:872–876.
    1. June CH, Blazar BR, Riley JL. Engineering lymphocyte subsets: tools, trials and tribulations. Nature Rev Immunol. 2009;9:704–716.
    1. Schwarzwaelder K, Howe SJ, Schmidt M, Brugman MH, Deichmann A, Glimm H, Schmidt S, Prinz C, Wissler M, King DJ, Zhang F, Parsley KL, Gilmour KC, Sinclair J, Bayford J, Peraj R, Pike-Overzet K, Staal FJ, de Ridder D, Kinnon C, Abel U, Wagemaker G, Gaspar HB, Thrasher AJ, von Kalle C. Gammaretrovirus-mediated corrrectionof SCID-X1 is associated with skewed vector integration site distribution in vivo. J Clin Invest. 2007;117:2241–2249.
    1. Newrzela S, et al. Resistance of mature T cells to oncogene transformation. Blood. 2008;112:2278–2286.
    1. Cattoglio C, et al. High-definition mapping of retroviral integration sites defines the fate of allogeneic T cells after donor lymphocyte infusion. PLoS One. 2010;5:e15688.
    1. Wang GP, et al. Dynamics of gene-modified progenitor cells analyzed by tracking retroviral integration sites in a human SCID-X1 gene therapy trial. Blood. 2010;115:4356–4366.
    1. Deichmann A, et al. Insertion sites in engrafted cells cluster within a limited repertoire of genomic areas after gammaretroviral vector gene therapy. Mol Ther. 2011;19:2031–2039.
    1. Montini E, et al. The genotoxic potential of retroviral vectors is strongly modulated by vector design and integration site selection in a mouse model of HSC gene therapy. J Clin Invest. 2009;119:964–075.
    1. Kustikova O, Brugman M, Baum C. The genomic risk of somatic gene therapy. Semin Cancer Biol. 2010;20:269–278.
    1. Almarza E, et al. Correction of SCID-X1 using an enhancerless Vav promoter. Hum Gene Ther. 2011;22:263–270.
    1. Baum C. Parachuting in the epigenome: the biology of gene vector insertion profiles in the context of clinical trials. EMBO Mol Med. 2011;3:75–77.
    1. Wu C, Dunbar CE. Stem cell gene therapy: the risks of insertional mutagenesis and approaches to minimize genotoxicity. Front Med. 2011;5:356–371.
    1. Trobridge GD. Genotoxicity of retroviral hematopoietic stem cell gene therapy. Expert Opin Biol Ther. 2011;11:581–593.
    1. Huston MW, Brugman MH, Horsman S, Stubbs AP, van der Spek PJ, Wagemaker G. Comprehensive investigation of the parameters of viral integration site analysis and their effects on the gene annotations produced. Hum Gene Ther. 2012 in press.
    1. Corrigan-Curay J, Cohen-Haguenauer O, O'Reilly M, Ross SR, Fan H, Rosenberg N, Somia N, King N, Friedmann T, Dunbar C, Aiuti A, Naldini L, Baum C, von Kalle C, Kiem HP, Montini E, Bushman F, Sorrentino BP, Carrondo M, Malech H, Gahrton G, Shapiro R, Wolff L, Rosenthal E, Jambou R, Zaia J, Kohn DB. Challenges in vector and trial design using retroviral vectors for long-term gene correction in hematopoietic stem cell gene therapy. Mol Ther. 2012;20:1084–1094.
    1. Aiuti A, Bacchetta R, Seger R, Villa A, Cavazzana-Calvo M. Gene therapy for primary immunodeficiencies: Part 2. Curr Opin Immunol. 2012;24:1–7.
    1. Scobie L, et al. A novel model of SCID-X1 reconstitution reveals predisposition to retrovirus-induced lymphoma but no evidence of gammaC gene oncogenicity. Mol Ther. 2009;17:1031–1038.
    1. Deichmann A, et al. Vector integration is nonrandom and clustered and influences the fate of lymphopoiesis in SCID-X1 gene therapy. J Clin Invest. 2007;117:2225–2232.
    1. Gabriel R, et al. Comprehensive genomic access to vector integration in clinical gene therapy. Nature Med. 2009;15:1431–1436.
    1. Kolb AF, Coates CJ, Kaminski JM, Summers JB, Miller AD, D.J. S. Site-directed genome modification: nucleic acid and protein modules for targeted integration and gene correction. Trends Biotech. 2005;23:399–406.
    1. de Jong J, et al. Computational identification of insertional mutagenesis targets for cancer gene discovery. Nucl Acids Res. 2011;39:e105.
    1. March HN, et al. Insertional mutagenesis identifies multiple networks of cooperating genes driving intestinal tumorigenesis. Nature Genet. 2011;43:1202–1209.
    1. Dupuy AJ. Transposon-based screens for cancer gene discovery in mouse models. Semin Cancer Biol. 2010;20:261–268.
    1. Bergemann TL, et al. New methods for finding common insertion sites and co-occurring common insertion sites in transposon- and virus-based genetic screens. Nucl Acids Res. 2012;40:3822–3833.
    1. Howell VM. Sleeping Beauty - a mouse model for all cancers? Cancer Lett. 2012;317:1–8.
    1. Mann KM, Ward JM, Yew CC, Kovochich A, Dawson DW, Black MA, Brett BT, Sheetz T, E, Dupuy AJ, Australian Pancreatic Cancer Genome Initiative. Chang DK, Biankin AV, Waddell N, Kassahn KS, Grimmond SM, Rust AG, Adams DJ, Jenkins NA, Copeland NG. Sleeping Beauty mutagenesis reveals cooperating mutations and pathways in pancreatic adenocarcinoma. Proc Natl Acad Sci USA. 2012;109:5934–5941.
    1. van der Weyden L, et al. Loss of rassf1a synergizes with deregulated runx2 signaling in tumorigenesis. Cancer Res. 2012;72:3817–3827.
    1. Bergerson RJ, Collier LS, Sarver AL, Been RA, Lugthart S, Diers MD, Zuber J, Rappaport AR, Nixon MJ, Silverstein KA, Fan D, Lamblin AF, Wolff L, Kersey JH, Delwe l. R., Lowe SW, O'Sullivan MG, Kogan SC, Adams DJ, Largaespada DA. An insertional mutagenesis screen identifies genes that cooperate with Mll-AF9 in a murine leukemogenesis model. Blood. 2012;119:4512–4523.
    1. van der Weyden L, Arends MJ, Rust AG, Poulogiannis G, McIntyre RE, Adams DJ. Increased tumorigenesis associated with loss of the tumor suppressor gene Cadm. Mol Cell. 2012;11 in press.
    1. Oliveira-Cobucci RN, et al. Comparative incidence of cancer in HIV-AIDS patients and transplant recipients. Cancer Epidemiol. 2012;36:e69–73.
    1. Wang GP, et al. Analysis of lentiviral vector integration in HIV+ study subjects receiving autologous infusions of gene modified CD4+ T cells. Mol Ther. 2009;17:844–850.
    1. Cesana D, Squaldino J, Rudilosso L, Merella S, Naldini L, Montini E. Whole transcriptome characterization of aberrant splicing events induced by lentivirus vector integrations. J Clin Invest. 2012;122:1667–1676.
    1. Brady T, Bushman FD. Nondividing cells: a safer bet for integrating vectors? Mol Ther. 2011;19:640–641.
    1. Cattoglio C, Facchini G, Sartori D, Antonelli A, Miccio A, Cassani B, Schmidt M, von Kalle C, Howe S, Thrasher AJ, Aiuti A, Ferrari G, Recchia A, Mavilio F. Hot spots of retroviral integration in human CD34+ hematopoietic cells. Blood. 2007;110:1770–1778.
    1. Nowrouzi A, Glimm H, von Kalle C, Schmidt M. Retroviral vectors: post entry events and genomic alterations. Viruses. 2011;3:429–455.
    1. Biffi A, et al. Lentiviral vector common integration sites in preclinical models and a clinical trial reflect a benign integration bias and not oncogenic selection. Blood. 2012;117:5332–5339.
    1. Roth SL, Malani N, Bushman FD. Gammaretroviral integration into nucleosomal target DNA in vivo. J Virol. 2011;85:7393–7401.
    1. Hackett PB. Integrating DNA vectors for gene therapy. Mol Ther. 2007;15:10–12.
    1. Zhang Y, Satterlee A, Huang L. In vivo gene delivery by nonviral vectors: overcoming hurdles? Mol Ther. 2012;20:1298–1304.
    1. Hackett PB, Largaespada DA, Cooper LJN. A transposon and transposase system for human application. Mol Ther. 2010;18:674–683.
    1. Hackett CS, Geurts AM, Hackett PB. Predicting preferential DNA vector insertion sites: Implications for functional genomics and gene therapy. Genome Biol. 2007;8(Suppl. 1):S12.
    1. Consortium1000 GP A map of human genome variation from population-scale sequencing. Nature. 2010;467:1061–1073.
    1. ConsortiumENCODE P et al. An integrated encyclopedia of DNA elements in the human genome. Nature. 2012;489:57–74.
    1. Lander ES, et al. Initial sequencing and analysis of the human genome. Nature. 2001;409:860–921.
    1. Venter JC, et al. The sequence of the human genome. Science. 2001;291:1304–1351.
    1. Cordaux R. The human genome in the LINE of fire. Proc Natl Acad Sci USA. 2008;105:19033–19034.
    1. de Koning AP, Gu W, Castoe TA, Batzer MA, Pollock DD. Repetitive elements may comprise over two-thirds of the human genome. PLoS Genet. 2011;7:e1002384.
    1. Beck CR, et al. LINE-1 retrotransposition activity in human genomes. Cell. 2010;141:1159–1170.
    1. Brady T, et al. Integration target site selection by a resurrected human endogenous retrovirus. Genes Dev. 2009;23:633–642.
    1. Brouha B, et al. Hot L1s account for the bulk of retrotransposition in the human population. Proc Natl Acad Sci USA. 2003;100:5280–5288.
    1. Lee E, et al. Landscape of somatic retrotransposition in human cancers. Science. 2012;337:967–971.
    1. Solyom S, Ewing AD, Rahrmann EP, Doucet TT, Nelson HH, Burns MB, Harris RS, Sigmon DF, Casella A, Erlanger B, Wheelan S, Upton KR, Shukla R, Faulkner GJ, Largaespada DA, Kazazian HH. Extensive somatic L1 retrotransposition in colorectal tumors. Genome Res. 2012 in press.
    1. Dombroski BA, Mathias SL, Nanthakumar E, Scott AF, Kazazian HHJ. Isolation of an active human transposable element. Science. 1991;254:1805–1808.
    1. Moran JV, Holmes SE, Naas TP, DeBerardinis RJ, Boeke JD, Kazazian HHJ. High frequency retrotransposition in cultured mammalian cells. Cell. 1996;87:917–927.
    1. Kazazian HH, Goodier JL. LINE drive: retrotransposition and genome instability. Cell. 2002;110:277–280.
    1. Deininger PL, Moran JV, Batzer MA, Kazazian HHJ. Mobile elements and mammalian genome evolution. Curr Opin Genet Dev. 2003;13:651–658.
    1. Kazazian HH. Mobile elements: drivers of genome evolution. Science. 2004;303:1626–1632.
    1. Cordaux R, Batzer MA. The impact of retrotransposons on human genome structure. Nature Rev Genet. 2009;10:691–703.
    1. Ewing AD, Kazazian HHJ. Whole-genome resequencing allows detection of many rare LINE-1 insertion alleles in humans. Genome Res. 2011;21:985–990.
    1. Burns KH, Boeke JD. Human transposon tectonics. Cell. 2012;149:740–752.
    1. Coufal NG, Garcia-Perez JL, Peng GE, Yeo GW, Mu Y, Lovci MT, Morell M, O'Shea KS, Moran JV, Gage FH. L1 retrotransposition in human neural progenitor cells. Nature. 2009;460:1127–1131.
    1. Baillie JK, Barnett MW, Upton KR, Gerhardt DJ, Richmond TA, De Sapio F, Brennan PM, Rizzu P, Smith S, Fell M, Talbot RT, Gustincich S, Freeman TC, Mattick JS, Hume DA, Heutink P, Carninci P, Jeddeloh JA, Faulkner GJ. Somatic retrotransposition alters the genetic landscape of the human brain. Nature. 2011;479:534–537.
    1. Muotri AR, et al. L1 retrotransposition in neurons is modulated by MeCP2. Nature. 2010;443:443–446.
    1. Huang CR, Schneider AM, Lu Y, Niranjan T, Shen P, Robinson MA, Steranka JP, Valle D, Civin CI, Wang T, Wheelan SJ, Ji H, Boeke JD, Burns KH. Mobile interspersed repeats are major structural variants in the human genome. Cell. 2010;141:1171–1182.
    1. Iskow RC, et al. Natural mutagenesis of human genomes by endogenous retrotransposons. Cell. 2010;141:1253–1261.
    1. Garcia-Perez JL, Morell M, Scheys JO, Kulpa DA, Morell S, Carter CC, Hammer GD, Collins KL, O'Shea KS, Menendez P, Moran JV. Epigenetic silencing of engineered L1 retrotransposition events in human embryonic carcinoma cells. Nature. 2010;466:769–773.
    1. Doerfler W. Impact of foreign DNA integration on tumor biology and on evolution via epigenetic alterations. Epigenomics. 2012;4:41–49.
    1. Bao J, Yan W. Male germline control of transposable elements. Biol Reprod. 2012;86:162, 161–114.
    1. Wagstaff BJ, Hedges DJ, Derbes RS, Campos-Sanchez R, Chiaromonte F, Makova KD, Roy-Engel AM. Rescuing Alu: Recovery of new inserts shows LINE-1 preserves Alu activity through A-tail expansion. PLoS Genet. 2012;8:e1002842.
    1. Zhang Y, Maksakova IA, Gagnier L, van de Lagemaat LN, Mager DL. Genome-wide assessments reveal extremely high levels of polymorphism of two active families of mouse endogenous retroviral elements. PLoS Genet. 2008;4:e1000007.
    1. Liu G, Aronovich EL, Cui Z, Whitley CB, Hackett PB. Excision of Sleeping Beauty transposons: parameters and applications to gene therapy. J Gene Med. 2004;6:574–583.
    1. Plasterk RHA, Izsvák Z, Ivics Z. Resident aliens: the Tc1/mariner superfamily of transposable elements. Trends Genet. 1999;15:326–332.
    1. Ivics Z, Hackett PB, Plasterk RH, Izsvak Z. Molecular reconstruction of Sleeping Beauty, a Tc1-like transposon from fish, and its transposition in human cells. Cell. 1997;91:501–510.
    1. Yant SR, Meuse L, Chiu W, Ivics Z, Izsvak Z, Kay MA. Somatic integration and long-term transgene expression in normal and haemophilic mice using a DNA transposon system. Nat Genet. 2000;25:35–41.
    1. Geurts AM, et al. Gene transfer into genomes of human cells by the Sleeping Beauty transposon system. Mol Ther. 2003;8:108–117.
    1. Zayed H, Izsvak Z, Walisko O, Ivics Z. Development of hyperactive Sleeping Beauty transposon vectors by mutational analysis. Mol Ther. 2004;9:292–304.
    1. Yant SR, Park J, Huang Y, Mikkelsen JG, Kay MA. 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. 2004;24:9239–9247.
    1. Mátés L, et al. Molecular evolution of a novel hyperactive Sleeping Beauty transposase enables robust stable gene transfer in vertebrates. Nat Genet. 2009;41:753–761.
    1. Ivics Z, Kaufman CD, Zayed H, Miskey C, Walisko O, Izsvak Z. The Sleeping Beauty transposable element: evolution, regulation and genetic applications. Curr Issues Mol Biol. 2004;6:43–55.
    1. Yant SR, et al. Nonrandom insertion site preferences for the SB transposon in vitro and in vivo. Mol Ther. 2004;9:S309.
    1. Liu G, et al. Target-site preference for Sleeping Beauty transposons. J Mol Biol. 2005;346:161–173.
    1. Geurts AM, et al. DNA structural patterns influence integration site preferences for mobile elements. Nucl Acids Res. 2006;34:2803–2811.
    1. Berry C, Hannenhalli S, Leipzig J, Bushman FD. Selection of target sites for mobile DNA integration in the human genome. PLoS Comp Biol. 2006;2:e157.
    1. Ohlfest JR, et al. Phenotypic correction and long-term expression of factor VIII in hemophilic mice by immunotolerization and nonviral gene transfer using the Sleeping Beauty transposon system. Blood. 2005;105:2691–2698.
    1. Liu L, Mah C, Fletcher BS. Sustained FVIII expression and phenotypic correction of hemophilia A in neonatal mice. Mol Ther. 2006;13:1006–1015.
    1. Aronovich EL, McIvor RS, Hackett PB. The Sleeping Beauty transposon system: a non-viral vector for gene therapy. Hum Mol Genet. 2011;20(R1):14–20.
    1. Bell JB, Aronovich EL, Schreifels JM, Beadnell TC, Hackett PB. Duration of expression of Sleeping Beauty transposase in mouse liver following hydrodynamic DNA delivery. Mol Ther. 2010;18:1792–1802.
    1. Aravin AA, Hannon GJ, Brennecke J. The Piwi-piRNA pathway provides an adaptive defense in the transposon arms race. Science. 2007;318:761–764.
    1. Fedoroff NV. Transposable elements, epigenetics, and genome evolution. Science. 2012;338:758–767.
    1. Nesmelova IV, Hackett PB. DDE Transposases: structural similarity and diversity. Adv Drug Del Rev. 2010;62:1187–1195.
    1. Montaño SP, Pigli YZ, Rice PA. The Mu transpososome structure sheds light on DDE recombinase evolution. Nature. 2012;491:413–417.
    1. Wilber AC, Frandsen JL, Geurts AM, Largaespada DA, P.B. H, McIvor RS. RNA as a source of transposase for Sleeping Beauty-mediated gene insertion and expression in somatic cells and tissues. Mol Ther. 2006;13:625–630.
    1. Urschitz J, et al. Helper-independent piggyBac plasmids for gene delivery approaches: strategies for avoiding potential genotoxic effects. Proc Natl Acad Sci USA. 2010;107:8117–8122.
    1. Hastie ND, Bishop JO. The expression of three abundance classes of messenger RNA in mouse tissues. Cell. 1976;9:761–774.
    1. Rushforth AM, Anderson P. Splicing removes the Caenorhabditis elegans transposon Tc1 from most mutant pre-mRNAs. Mol Cell Biol. 1996;16:422–429.
    1. Hodgkinson A, Chen Y, Eyre-Walker A. The large-scale distribution of somatic mutations in cancer genomes. Hum Mutat. 2012;33:136–143.
    1. Schuster-Böckler B, Lehner B. Chromatin organization is a major influence on regional mutation rates in human cancer cells. Nature. 2012;488:504–507.
    1. Djebali S, et al. Landscape of transcription in human cells. Nature. 2012;489:101–108.
    1. Cheng C, Alexander R, Min R, Leng J, Yip KY, Rozowsky J, Yan KK, Dong X, Djebali S, Ruan Y, Davis CA, Carninci P, Lassman T, Gingeras TR, Guigó R, Birney E, Weng Z, Snyder M, Gerstein M. Understanding transcriptional regulation by integrative analysis of transcription factor binding data. Genome Res. 2012;22:1658–1667.
    1. Sanyal A, Lajoie BR, Jain G, Dekker J. The long-range interaction landscape of gene promoters. Nature. 2012;489:109–113.
    1. Thurman RE, et al. The accessiblechromatin landscape of the human genome. Nature. 2012;489:75–82.
    1. Zhang J, Markus J, J B, Paul T, Wolff L. Three murine leukemia virus integration regions within 100kb upstream of c-myb are proximal to the 5' regulatory region of the gene through DNA looping. J Virol. 2012 in press.
    1. Poleshko A, et al. Identification of cellular proteins that maintain retroviral epigenetic silencing: evidence for an antiviral response. J Virol. 2008;82:2313–2323.
    1. Alkan C, Coe BP, Eichler EE. Genome structural variation discovery and genotyping. Nature Rev Genet. 2011;12:363–376.
    1. Marth GT, et al. The functional spectrum of low-frequency coding variation. Genome Biol. 2011;12:R84.
    1. Durbin RM, et al. A map of human genome variation from population-scale sequencing. Nature. 2010;467:1061–1073.
    1. Pelak K, et al. The characterization of twenty sequenced human genomes. PLoS Genet. 2010;6:e1001111.
    1. MacArthur DG, et al. A systematic survey of loss-of-function variants in human protein-coding genes. Science. 2012;335:823–828.
    1. Wada T, Candotti F. Somatic mosaicism in primary immune deficiencies. Curr Opin Allergy Clin Immunol. 2008:8.
    1. Abyzov A, Urban AE, Snyder M, Gerstein M. CNVnator: an approach to discover, genotype, and characterize typical and atypical CNVs from family and population genome sequencing. Genome Res. 2011;21:974–984.
    1. O'Huallachain M, Karczewski KJ, Weissman SM, Urban AE, Snyder MP. Extensive genetic variation in somatic human tissues. Proc Natl Acad Sci USA. 2012;109:18018–18023.
    1. Kidd JM, et al. A human genome structural variation sequencing resource reveals insights into mutational mechanisms. Cell. 2010;143:837–847.
    1. Boyle AP, et al. Annotation of functional variation in personal genomes using RegulomeDB. Genome Res. 2012;22:1790–1797.
    1. Eide D, Anderson P. Insertion and excision of Caenorhabditis elegans transposable element Tc1. Mol Cell Biol. 1988;8:737–746.
    1. Ellis J. Silencing and variegation of gammaretrovirus and lentivirus vectors. Hum Gene Ther. 2005;16:1241–1246.
    1. Blumenstiel JP. Evolutionary dynamics of transposable elements in a small RNA world. Trends Genet. 2011;27:23–31.
    1. Siomi MC, Sato K, Pezic D, Aravin AA. PIWI-interacting small RNAs: the vanguard of genome defence. Nature Rev Mol Cell Biol. 2011;12:246–258.
    1. Garrison BS, Yant SR, Mikkelsen JG, Kay MA. Post-integrative gene silencing within the Sleeping Beauty transposition system. Mol Cell Biol. 2007;27:8824–8833.
    1. Rauschhuber C, Ehrhardt A. RNA interference is responsible for reduction of transgene expression after Sleeping Beauty transposase mediated somatic integration. PLoS One. 2012 in press.
    1. Chalberg TW, et al. Integration specificity of phage phiC31 integrase in the human genome. J Mol Biol. 2006;357:28–48.
    1. Ehrhardt A, Engler JA, Xu H, Kay MA. Molecular analysis of chromosomal rearrangements in mammalian cells after phiC31-mediated integration. Human Gene Ther. 2006;17:1077–1094.
    1. Largaespada DA. Transposon mutagenesis in mice. Methods Mol Biol. 2009;530:379–390.
    1. Copeland NG, Jenkins NA. Harnessing transposons for cancer gene discovery. Nat Rev Genet. 2010;10:696–706.
    1. Mikkers H, Berns A. Retroviral insertional mutagenesis: tagging cancer pathways. Adv Cancer Res. 2003;88:53–99.
    1. Jonkers J, Berns A. Retroviral insertional mutagenesis as a strategy to identify cancer genes. Biochim Biophys Acta. 1996;1287:29–57.
    1. Lohuizen Mv, Berns A. Tumorigenesis by slow transforming retroviruses--an update. Biochim Biophys Acta. 1990;1032(2–3):213–235.
    1. Largaespada DA. Genetic heterogeneity in acute myeloid leukemia: maximizing information flow from MuLV mutagenesis studies. Leukemia. 2000;14:1174–1184.
    1. Lu M, Zhang N, Maruyama M, Hawley RG, Ho AD. Retrovirus-mediated gene expression in hematopoietic cells correlates inversely with growth factor stimulation. Hum Gene Ther. 1996;7:2263–2271.
    1. Hawley RG, Lieu FH, Fong AZ, Hawley TS. Versatile retroviral vectors for potential use in gene therapy. Gene Ther. 1994;1:136–138.
    1. Cherry SR, Biniszkiewicz D, van Parijs L, Baltimore D, Jaenisch R. Retroviral expression in embryonic stem cells and hematopoietic stem cells. Mol Cell Biol. 2000;20:7419–7426.
    1. Vassiliou GS, et al. Mutant nucleophosmin and cooperating pathways drive leukemia initiation and progression in mice. Nature Genet. 2011;43:470–475.
    1. Dupuy AJ, et al. A modified Sleeping Beauty transposon system that can be used to model a wide variety of human cancers in mice. Cancer Res. 2009;69:8150–8156.
    1. Collier LS, Carlson CM, Ravimohan S, Dupuy AJ, Largaespada DA. Cancer gene discovery in solid tumors using transposon-based somatic mutagenesis in the mouse. Nature. 2005;436:272–276.
    1. Dupuy AJ, Akagi K, Largaespada DA, Copeland NG, Jenkins NA. Mammalian mutagenesis using a highly mobile somatic Sleeping Beauty transposon system. Nature. 2005;436:221–226.
    1. Collier LS, et al. Whole-body Sleeping Beauty mutagenesis can cause penetrant leukemia/lymphoma and rare high-grade glioma without associated embryonic lethality. Cancer Res. 2009;69:8429–8437.
    1. Starr TK, et al. A transposon-based genetic screen in mice identifies genes altered in colorectal cancer. Science. 2009;323:1747–1750.
    1. Starr TK, Scott PM, Marsh BM, Zhao L, Than BL, O'Sullivan MG, Sarver AL, Dupuy AJ, Largaespada DA, Cormier RT. A Sleeping Beauty transposon-mediated screen identifies murine susceptibility genes for adenomatous polyposis coli (Apc)-dependent intestinal tumorigenesis. Proc Natl Acad Sci USA. 2011;108:5765–5770.
    1. Keng VW, et al. A conditional transposon-based insertional mutagenesis screen for genes associated with mouse hepatocellular carcinoma. Nature Biotech. 2009;27:264–274.
    1. Pérez-Mancera PA, Rust AG, van der Weyden L, Kristiansen G, Li A, Sarver AL, Silverstein KA, Grützmann R, Aust D, Rümmele P, Knösel T, Herd C, Stemple DL, Kettleborough R, Brosnan JA, Li A, Morgan R, Knight S, Yu J, Stegeman S, Collier LS, ten Hoeve JJ, de Ridder J, Klein AP, Goggins M, Hruban RH, Chang DK, Biankin AV, Grimmond SM, Australian Pancreatic Cancer Genome Initiative. Wessels LF, Wood SA, Iacobuzio-Donahue CA, Pilarsky C, Largaespada DA, Adams DJ, Tuveson DA. The deubiquitinase USP9X suppresses pancreatic ductal adenocarcinoma. Nature. 2012;486:266–270.
    1. Dotti JB, Cooper LJN. Redirecting T-cell specificity by introducing a tumor-specific chimeric antigen receptor. Blood. 2010;116:1035–1044.
    1. Brentjens R, Yeh R, Bernal Y, Riviere I, Sadelain M. Treatment of chronic lymphocytic leukemia with genetically targeted autologous T cells: Case report of an unforeseen adverse event in a Phase I clinical trial. Mol Ther. 2010;18:666–668.
    1. Brentjens RJ, et al. Safety and persistence of adoptively transferred autologous CD19-targeted T cells in patients with relapsed or chemotherapy refractory B-cell leukemias. Blood. 2011;118:4817–4828.
    1. Kalos M, et al. T cells with chimeric antigen receptors have potent antitumor effects and can establish memory in patients with advanced leukemia. Sci Transl Med. 2011;3:95ra73.
    1. Kochenderfer JN, et al. B-cell depletion and remissions of malignancy along with cytokine-associated toxicity in a clinical trial of anti-CD19 chimeric-antigen-receptor-transduced T cells. Blood. 2012;119:2709–2720.
    1. Porter DL, Levine BL, Kalos M, Bagg A, June CH. Chimeric antigen receptor-modified T cells in chronic lympholid leukemia. New Eng J Med. 2011;365:725–733.
    1. Bonini C, et al. Safety of retroviral gene marking with a truncated NGF receptor. Nature Med. 2003;9:367–369.
    1. Singh H, et al. Redirecting specificity of T-cell populations for CD19 using the Sleeping Beauty system. Cancer Res. 2008;68:2961–2971.
    1. Manuri PV, et al. piggyBac transposon/transposase system to generate CD19-specific T cells for the treatment of B-lineage malignancies. Hum Gene Ther. 2010;21:427–437.
    1. Singh H, et al. Reprogramming CD19-specific T cells with IL-21 signaling can improve adoptive immunotherapy of B-lineage malignancies. Cancer Res. 2011;71:3516–3527.
    1. Lamers CH, et al. Treatment of metastatic renal cell carcinoma with autologous T-lymphocytes genetically retargeted against carbonic anhydrase IX: first clinical experience. J Clin Oncol. 2006;24:e20–22.
    1. Morgan RA, Yang JC, Kitano M, Dudley ME, Laurencot CM, S/A. R. Case report of a serious adverse event following the administration of T cells transduced with a chimeric antigen receptor recognizing ERBB2. Mol Ther. 2010;18:843–851.
    1. Montini E, et al. Hematopoietic stem cell gene transfer in a tumor-prone mouse model uncovers low genotoxicity of lentiviral vector integration. Nature Biotech. 2006;24:687–696.
    1. Modlich U, et al. Leukemias following retroviral transfer of multidrug resistance 1 (MDR1) are driven by combinatorial insertional mutagenesis. Blood. 2005;105:4235–4246.
    1. Jensen MC, et al. Human T lymphocyte genetic modification with naked DNA. Mol Ther. 2000;1:49–55.
    1. Cooper LJ, et al. T lymphocytes for autologous cellular therapy for lymphoma. Cytotherapy. 2006;8:105–117.
    1. Maiti SN, et al. Sleeping Beauty system to redirect T-cell specificity for human applications. J Immunol. in press.
    1. Jena B, Dotti G, Cooper LJ. Redirecting T-cell specificity by introducing a tumor-specific chimeric antigen receptor. Blood. 2010;116:1035–1044.
    1. Kebriaei P, et al. Infusing CD19-directed T cells to augment disease control in patients undergoing autologous hematopoietic stem-cell transplantation for advanced B-lymphoid malignancies. Hum Gene Ther. 2012;23:444–450.
    1. Sadelain M, Papapetrou EP, Bushman FD. Safe harbours for the integration of new DNA in the human genome. Nature Rev Cancer. 2011;12:51–58.
    1. Papapetrou EP, et al. Genomic safe harbors permit high beta-globin transgene expression in thalassemia induced pluirpotent stem cells. Nature Biotech. 2011;29:73–78.
    1. Porteus MH, Baltimore D. Chimeric nucleases stimulate gene targeting in human cells. Science. 2003;300:763.
    1. Urnov FD, et al. Highly efficient endogenous human gene correction using designed zinc-finger nucleases. Nature. 2005;435:646–651.
    1. Urnov FD, Rebar EJ, Holmes MC, Zhang HS, Gregory PD. Genome editing with engineered zinc finger nucleases. Nature Rev Genet. 2010;11:636–646.
    1. Lombardo A, et al. Gene editing in human stem cells using zinc finger nucleases and integrase-defective lentiviral vector delivery. Nature Biotech. 2007;25:1298–1306.
    1. Perez EE, et al. Establishment of HIV-1 resistance in CD4+ T cells by genome editing using zinc-finger nucleases. Nature Biotech. 2008;26:808–816.
    1. Holt N, Wang J, Kim K, Friedman G, Wang X, Taupin V, Crooks GM, Kohn DB, Gregory PD, Holmes MC, Cannon PM. Human hematopoietic stem progenitor cells modified by zinc-finger nucleases targeted to CCR5 control HIV-1 in vivo. Nature Biotech. 2012;28:839–847.
    1. Ivics Z, Katzer A, Stuwe EE, Fiedler D, Knespel S, Izsvak Z. Targeted Sleeping Beauty transposition in human cells. Mol Ther. 2007;15:1137–1144.
    1. Yant SR, Huang Y, Akache B, Kay MA. Site-directed transposon integration in human cells. Nucl Acids Res. 2007;35:e50.
    1. Voigt K, et al. Retargeting Sleeping Beauty transposon insertions by engineered zinc finger DNA-binding domains. Mol Ther. 2012;20:1852–1862.
    1. Ammar I, et al. Retargeting transposon insertions by the adeno-associated virus Rep protein. Nucl Acids Res. 2012;40:6693–6712.
    1. Wang Z, Lisowski L, Finegold MJ, Nakai H, Kay MA, Grompe M. AAV vectors containing rDNA homology display increased chromosomal integration and transgene persistence. Mol Ther. 2012;20:1902–1911.
    1. Lisowski L, et al. Ribosomal DNA integrating rAAV-rDNA vectors allow for stable transgene expression. Mol Ther. 2012;20:1912–1923.
    1. Bogdanove AJ, Voytas DF. TAL efffectors: customizable proteins for DNA targeting. Science. 2011;333:1843–1844.
    1. Carlson DF, Fahrenkrug SC, Hackett PB. Targeting DNA with fingers and TALENs. Mol Ther Nuc Acids. 2012;1:e3.
    1. Torikai H, et al. A foundation for universal T-cell based immunotherapy: T cells engineered to express a CD19-specific chimeric-antigen-receptor and eliminate expression of endogenous TCR. Blood. 2012;119:5697–5705.
    1. Nakazawa Y, et al. Optimization of the PiggyBac transposon system for the sustained genetic modification of human T lymphocytes. J Immunother. 2009;32:826–836.
    1. Li H, et al. In vivo genome editing restores haemostasis in a mouse model of haemophilia. Nature. 2011;475:217–221.

Source: PubMed

3
Se inscrever