Gene Therapy Using Stem Cells - CSHL...

12
Gene Therapy Using Stem Cells Erin R. Burnight 1 , Luke A. Wiley 1 , Robert F. Mullins 1 , Edwin M. Stone 1,2 , and Budd A. Tucker 1 1 The Stephen A. Wynn Institute for Vision Research, Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, Iowa 52242 2 Howard Hughes Medical Institute, University of Iowa, Iowa City, Iowa 52242 Correspondence: [email protected] Viral-mediated gene augmentation therapy has recently shown success in restoring vision to patients with retinal degenerative disorders. Key to this success was the availability of animal models that accurately presented the human phenotype to test preclinical efficacyand safety. These exciting studies support the use of gene therapy in the treatment of devastating retinal degenerative diseases. In some cases, however, in vivo gene therapy for retinal degeneration would not be effective because the cell types targeted are no longer present. The develop- ment of somatic cell reprogramming methods provides an attractive source of autologous cells for transplantation and treatment of retinal degenerative disease. This article explores the development of gene therapy and patient-derived stem cells for the purpose of restoring vision to individuals suffering from inherited retinal degenerations. G ene therapy for the treatment of inherited disorders such as adrenoleukodystrophy (Cartier et al. 2009), adenosine deaminase-relat- ed severe combined immunodeficiency (Gaspar et al. 2011), and the childhood blindness Leber congenital amaurosis (LCA) (Bainbridge et al. 2008; Hauswirth et al. 2008; Maguire et al. 2009) have recently shown great promise. Inherent to the success of these clinical trials was the avail- ability of animal models to test therapeutic strat- egies. For instance, gene replacement with an adeno-associated virus (AAV) vector to treat RPE65-associated LCA (LCA2) was largely suc- cessful in both the Rpe65 2/ 2 mouse and the large Briard dog model of the disease (Ben- nicelli et al. 2008). In comparison to con- tralateral sham-injected eyes, electroretinogram (ERG) responses and visual acuity increased sig- nificantly in Rpe65 2/ 2 mouse eyes treated sub- retinally with an AAV-RPE65 vector. Similarly, subretinal AAV-RPE65 delivery resulted in sig- nificant reversal of nystagmus and visual deficits in RPE65 mutant dogs (Acland et al. 2001, 2005; Narfstro ¨m et al. 2003). Notably, ERG a-waves, indicative of photoreceptor responses, also in- creased dramatically (by 26%–60%) and per- sisted for the duration of the study following injection of the therapeutic gene transfer vector. The kinetics of the a-waveswere not significantly different between treated and phenotypically normal animals (Bennicelli et al. 2008). These promising findings paved the way for the devel- opment of AAV-mediated RPE65 gene replace- ment therapy for patients with LCA2. Editors: Eric A. Pierce, Richard H. Masland, and Joan W. Miller Additional Perspectives on Retinal Disorders: Genetic Approachesto Diagnosis and Treatment available at www.perspectivesinmedicine.org Copyright # 2015 Cold Spring Harbor Laboratory Press; all rights reserved; doi: 10.1101/cshperspect.a017434 Cite this article as Cold Spring Harb Perspect Med 2015;5:a017434 1 www.perspectivesinmedicine.org on October 10, 2020 - Published by Cold Spring Harbor Laboratory Press http://perspectivesinmedicine.cshlp.org/ Downloaded from

Transcript of Gene Therapy Using Stem Cells - CSHL...

Page 1: Gene Therapy Using Stem Cells - CSHL Pperspectivesinmedicine.cshlp.org/content/5/4/a017434.full.pdf · ( 6.5 kb coding sequence) require vectorsthat canaccommodatemorethanthe4.5 kbpackag-inglimitofAAV(Wuetal.2010).Tothisend,the

Gene Therapy Using Stem Cells

Erin R. Burnight1, Luke A. Wiley1, Robert F. Mullins1, Edwin M. Stone1,2, and Budd A. Tucker1

1The Stephen A. Wynn Institute for Vision Research, Department of Ophthalmology and Visual Sciences,University of Iowa, Iowa City, Iowa 52242

2Howard Hughes Medical Institute, University of Iowa, Iowa City, Iowa 52242

Correspondence: [email protected]

Viral-mediated gene augmentation therapy has recently shown success in restoring vision topatients with retinal degenerative disorders. Key to this success was the availability of animalmodels that accurately presented the human phenotype to test preclinical efficacyand safety.These exciting studies support the use of gene therapy in the treatment of devastating retinaldegenerative diseases. In some cases, however, in vivo gene therapy for retinal degenerationwould not be effective because the cell types targeted are no longer present. The develop-ment of somatic cell reprogramming methods provides an attractive source of autologouscells for transplantation and treatment of retinal degenerative disease. This article exploresthe development of gene therapy and patient-derived stem cells for the purpose of restoringvision to individuals suffering from inherited retinal degenerations.

Gene therapy for the treatment of inheriteddisorders such as adrenoleukodystrophy

(Cartier et al. 2009), adenosine deaminase-relat-ed severe combined immunodeficiency (Gasparet al. 2011), and the childhood blindness Lebercongenital amaurosis (LCA) (Bainbridge et al.2008; Hauswirth et al. 2008; Maguire et al. 2009)have recently shown great promise. Inherent tothe success of these clinical trials was the avail-ability of animal models to test therapeutic strat-egies. For instance, gene replacement with anadeno-associated virus (AAV) vector to treatRPE65-associated LCA (LCA2) was largely suc-cessful in both the Rpe652/2 mouse andthe large Briard dog model of the disease (Ben-nicelli et al. 2008). In comparison to con-tralateral sham-injected eyes, electroretinogram

(ERG) responses and visual acuity increased sig-nificantly in Rpe652/2 mouse eyes treated sub-retinally with an AAV-RPE65 vector. Similarly,subretinal AAV-RPE65 delivery resulted in sig-nificant reversal of nystagmus and visual deficitsin RPE65 mutant dogs (Acland et al. 2001, 2005;Narfstrom et al. 2003). Notably, ERG a-waves,indicative of photoreceptor responses, also in-creased dramatically (by 26%–60%) and per-sisted for the duration of the study followinginjection of the therapeutic gene transfer vector.The kinetics of the a-waves were not significantlydifferent between treated and phenotypicallynormal animals (Bennicelli et al. 2008). Thesepromising findings paved the way for the devel-opment of AAV-mediated RPE65 gene replace-ment therapy for patients with LCA2.

Editors: Eric A. Pierce, Richard H. Masland, and Joan W. Miller

Additional Perspectives on Retinal Disorders: Genetic Approaches to Diagnosis and Treatment available at

www.perspectivesinmedicine.org

Copyright # 2015 Cold Spring Harbor Laboratory Press; all rights reserved; doi: 10.1101/cshperspect.a017434

Cite this article as Cold Spring Harb Perspect Med 2015;5:a017434

1

ww

w.p

ersp

ecti

vesi

nm

edic

ine.

org

on October 10, 2020 - Published by Cold Spring Harbor Laboratory Presshttp://perspectivesinmedicine.cshlp.org/Downloaded from

Page 2: Gene Therapy Using Stem Cells - CSHL Pperspectivesinmedicine.cshlp.org/content/5/4/a017434.full.pdf · ( 6.5 kb coding sequence) require vectorsthat canaccommodatemorethanthe4.5 kbpackag-inglimitofAAV(Wuetal.2010).Tothisend,the

ANIMAL MODELS AND THERAPEUTICGENE TRANSFER

Although animal models are invaluable for in-vestigation of disease pathophysiology and eval-uation of gene transfer strategies, not all modelsfaithfully recapitulate the human phenotype.This is especially true when developing thera-peutic strategies for the treatment of inheritedmacular degenerations. For example, the studyof gene transfer for the treatment of age-relatedmacular degeneration (AMD), Stargardt dis-ease, and Best disease are hampered by the factthat the rod-dominant mouse retina lacks amacula, the central zone of the primate retinathat has evolved to subserve very fine visual acu-ity. Similarly, many disease models have a dras-tically different age of onset and disease progres-sion than is observed in the human condition.For instance, dominant Pro23His rhodopsin-associated retinitis pigmentosa typically causesrelatively late-onset disease in humans, with sig-nificant vision remaining until late in life. Thepig model of Pro23His rhodopsin-associatedretinitis pigmentosa has a retinal disease thatis significantly more aggressive than that seenin humans; as early as 3 mo of age, these ani-mals have extensive photoreceptor cell loss (Fig.1) (Ross et al. 2012). Similarly, in the rd16CEP290 mouse model of LCA, mice are bornwith a full complement of photoreceptor cells,but the very aggressive disease results in almostcomplete loss of photoreceptor cells by 3 wk ofage (Chang et al. 2006). The fact that a largeproportion of the photoreceptor cells are lostbefore the eyes open makes gene transfer ap-proaches for the treatment of this disease verydifficult.

Advances in pluripotent stem cell technolo-gies provide an exciting avenue for the treatmentof retinal degenerative disease. For instance, hu-man embryonic stem cell (ESC)-derived photo-receptor precursor cells transplanted into amouse model of retinal degeneration havebeen shown to migrate into and integrate withinthe degenerative retina, express rod and conephotoreceptor markers and restore electroreti-nal activity (Lamba et al. 2009). However, ESC-derived photoreceptor transplants are by defini-

tion allogeneic and would likely require life-longimmune modulation if used clinically (Preynat-Seauve et al. 2009; West et al. 2010). Like ESCs,patient-specific induced pluripotent stem cells(iPSCs) possess the ability to differentiate intophotoreceptor precursors which can integrateinto a diseased retina and give rise to new func-tional photoreceptor cells (Lamba et al. 2010;

Figure 1. The transgenic pig carrying a humanPro23His rhodopsin transgene displays drastically ac-celerated retinal degeneration. A–B: Histologicalanalysis of both wild-type (A) and Pro23His (B) pigretinal tissue at 6 mo of age. C–D: Cytochemicalanalysis of both wild-type (C) and Pro23His (D)pig retinal tissue at 6 mo of age, using wheat germagglutinin (WGA, red) and DAPI (blue).

E.R. Burnight et al.

2 Cite this article as Cold Spring Harb Perspect Med 2015;5:a017434

ww

w.p

ersp

ecti

vesi

nm

edic

ine.

org

on October 10, 2020 - Published by Cold Spring Harbor Laboratory Presshttp://perspectivesinmedicine.cshlp.org/Downloaded from

Page 3: Gene Therapy Using Stem Cells - CSHL Pperspectivesinmedicine.cshlp.org/content/5/4/a017434.full.pdf · ( 6.5 kb coding sequence) require vectorsthat canaccommodatemorethanthe4.5 kbpackag-inglimitofAAV(Wuetal.2010).Tothisend,the

Homma et al. 2013; Tucker et al. 2013b). How-ever, unlike ESCs, iPSCs can be autologous andthus have a greatly reduced risk of being rejectedby the host immune system.

Patient-derived iPSCs can be generated in4–6 wk via forced factor expression in a varietyof cell types. Takahashi and Yamanaka firstshowed this concept through retroviral-mediat-ed expression of the transcription factors OCT4,SOX2, KLF4, and cMYC in mouse embryonicfibroblasts (Takahashi and Yamanaka 2006)and human primary dermal fibroblasts (Taka-hashi et al. 2007). In addition to the “Yamanakafactors,” Yu and colleagues in the Thomson lab-oratory established that the factors OCT4,SOX2, LIN28, and NANOG were capable of gen-erating induced pluripotent stem cells frommesenchymal cells (Yu et al. 2007). Many celltypes have since been reprogrammed usingforced factor expression including dermal kera-tinocytes (Carey et al. 2009), neural progenitorcells (Kim et al. 2011), hepatocytes (Liu et al.2010), and peripheral blood cells (Loh et al.2009). Since Yamanaka’s seminal report, severalgroups showed reprogramming using many de-livery methods—both integrating and noninte-grating. Integrating lentiviral vectors are widelyused for delivery of reprogramming transgenes(Brambrink et al. 2008; Maherali et al. 2008).However, to avoid the insertional mutagenicrisk associated with integrating vector systems,investigators use adenovirus (Stadtfeld et al.2008), Sendai virus (Jin et al. 2012; Tuckeret al. 2013a), and transient plasmid delivery(Gonzalez et al. 2009; Si-Tayeb et al. 2010). Fora more detailed description of these and otherdelivery methods please refer to reviews (Malikand Rao 2013; Hu 2014).

Beyond being an attractive cell source forretinal transplantation, the patient-specific na-ture of the iPSC makes this technology veryattractive for disease modeling and investiga-tion of disease pathophysiology. That is, themolecular causes of clinical variations in diseasecan be studied by comparing cells derived frommildly and severely affected individuals. For ex-ample, fibroblast cultures from CEP290-associ-ated LCA patients with different genotypesshow cellular phenotypes of varying degrees of

severity (Burnight et al. 2014). iPSC-derivedphotoreceptors from each of these patients canbe used to study the molecular causes of thephenotypic variation observed. The efficacy ofnovel therapies can also be tested on cells of theactual patients who might be treated.

INDUCED PLURIPOTENT STEM CELLSAS PATIENT-SPECIFIC MODELS TO TESTEXOGENOUS GENE DELIVERY

There are many vector systems from which tochoose when developing a strategy for the treat-ment of an inherited retinal degenerative dis-ease. For in vivo gene transfer, there are severalobstacles to the effective delivery and expressionof therapeutic transgenes. In many cases, genedelivery methods must overcome host immuni-ty (Reichel et al. 1998; Ikeda et al. 2002; Li et al.2008). Moreover, certain cell and tissue typesare difficult to transduce in vivo, which is likelybecause of a combination of incompatible vec-tor tropism and limited diffusion away from theinjection site. Gene delivery to cells ex vivo (i.e.,“in a dish”), circumvents many of these limita-tions and shifts the scientific focus from thedelivery vehicle to the genetic cargo. This is ofparticular value when one wishes to chooseamong several possible promoters, evaluategene function, and determine the optimal levelof transgene expression without killing dozens,if not hundreds, of animals in the process.

One of the major considerations when de-termining which vector system to use is genesize. Small, nonstructural enzymes (i.e., MAK,RPE65, etc.) are often well-suited for expressionvia the AAV vector system (Bennicelli et al. 2008;Conlon et al. 2013; Molday et al. 2013; Dai et al.2014). Indeed, the recent success of clinical tri-als involving patients with LCA2 highlights theutility of the AAV vector for in vivo gene transferto the retina (Bainbridge et al. 2008; Hauswirthet al. 2008; Maguire et al. 2008). However, thenatural serotypes of AAV do not transduce hu-man pluripotent stem cells very efficiently. Thecommonly used AAV2 serotype transduces hu-man stem cells only marginally (10% at a mul-tiplicity of infection of 10,000 [Asuri et al.2012]). Asuri and colleagues (2012) addressed

Gene Therapy Using Stem Cells

Cite this article as Cold Spring Harb Perspect Med 2015;5:a017434 3

ww

w.p

ersp

ecti

vesi

nm

edic

ine.

org

on October 10, 2020 - Published by Cold Spring Harbor Laboratory Presshttp://perspectivesinmedicine.cshlp.org/Downloaded from

Page 4: Gene Therapy Using Stem Cells - CSHL Pperspectivesinmedicine.cshlp.org/content/5/4/a017434.full.pdf · ( 6.5 kb coding sequence) require vectorsthat canaccommodatemorethanthe4.5 kbpackag-inglimitofAAV(Wuetal.2010).Tothisend,the

this limitation via directed evolution of the cap-sid, which yielded threefold higher transduc-tion efficiency when transduced at an equalmultiplicity of infection. Another considerationwhen using AAV vectors in gene transfer studiesis the packaging limit. Large genes such asCEP290 (�8 kb coding sequence) or ABCA4(�6.5 kb coding sequence) require vectors thatcan accommodate more than the 4.5 kb packag-ing limit of AAV (Wu et al. 2010). To this end, thelentivirus is an attractive choice. The packaginglimit of lentivirus is �8210 kb (Balaggan andAli 2012) and can thus accommodate largertransgene expression cassettes. Moreover, len-tiviruses can be pseudotyped with a variety ofpantropic envelope glycoproteins (Calameet al. 2011)—the most common of which is theG glycoprotein of the vesicular stomatitis virus(VSVG)—and can transduce pluripotent stemcells well (Endo et al. 2007; Tucker et al. 2011).

We have employed lentiviral-mediated genetransfer to transduce pluripotent stem cell-de-rived photoreceptor precursor cultures frompatients with LCA and retinitis pigmentosa.Forexample, the most common cause of retinitispigmentosa in the Jewish population is a splice-altering insertion of an Alu element in the malegerm cell-associated kinase (MAK) gene. Ciliaformation assays showed that patient’s cells gen-erated elongated cilia when compared with phe-notypically normal cells (Tucker et al. 2011). Ontransductionwith VSVG-pseudotyped lentiviralvectors expressing MAK, cells formed signifi-cantlyshorterciliacompared with untransducedcultures. Thus, we achieved functional genecorrection in patient-specific, stem cell-derivedphotoreceptor precursors. For treatment of pa-tients with LCA, we successfully packaged cDNAencoding the large structural gene CEP290 intolentiviral vectors with the heterologous cyto-megalovirus (CMV) promoter driving trans-gene expression (Burnight et al. 2014).

Another important consideration in devel-oping gene transfer strategies to treat inheriteddisorders in vitro is the timing of the geneticcorrection. Is it more effective to perform genetransfer to the somatic cells (i.e., fibroblasts orkeratinocytes) before reprogramming them intoiPSC cultures, or to differentiate the cultures

first and then transduce them? Long-term geneexpression in immature cell types transducedwith g-retroviral and lentiviral vectors can besilenced because of epigenetic mechanismssuch as DNA methylation or histone modifica-tion of the heterologous promoter (Ellis 2005;Herbst et al. 2012). For example, transgenesilencing was observed in iPSCs derived from amurine model of tyrosinemia type 1 (FAH – / –)after transduction with a lentivirus expressingFAH (Wu et al. 2011). Only 20% of cells ex-pressed the transgene. Nonetheless, a smallpercentage of these genetically corrected iPSCsshowed full pluripotent potential, and viablemice were generated via tetracomplementation(Wu et al. 2011).

Because phenotypic correction is not as ef-ficient when transducing iPSCs (because oftransgene silencing), it may be more advanta-geous to correct somatic cells before reprogram-ming. For example, Raya and colleagues trans-duced fibroblasts from Fanconi anemia patientswith lentiviral vectors expressing FANCA orFANCD2. These cells, on reprogramming, ex-pressed the pluripotency markers OCT4,SOX2, NANOG, SSEA3, SSEA4, and TRA1-60,and were indistinguishable from human embry-onic stem cells and iPSCs from healthy individ-uals. Moreover, the genetically corrected iPSCsdifferentiated into phenotypically normal mye-loid and erythroid progenitors (Raya et al. 2009).

Introducing therapeutic transgenes after dif-ferentiation is also a viable option in some cases.We recently showed successful lentiviral genetransfer to differentiated photoreceptor precur-sor cultures from patients with CEP290-associ-ated LCA. Transduced photoreceptor precursorcells (identified by expression of retinal mark-ers OTX2, OPSN1SW, and ROM1) expressedCEP290 transcript and full-length protein after90 d of differentiation (Burnight et al. 2014).Furthermore, a cilia formation defect that wasidentified in serum-starved fibroblasts isolatedfrom a CEP290 patient carrying two hypomor-phic alleles was rescued following transductionwith the CEP290-expressing lentiviral vector de-scribed above (Burnight et al. 2014).

Effective gene therapy strategies must resultin therapeutic transgene expression in the ap-

E.R. Burnight et al.

4 Cite this article as Cold Spring Harb Perspect Med 2015;5:a017434

ww

w.p

ersp

ecti

vesi

nm

edic

ine.

org

on October 10, 2020 - Published by Cold Spring Harbor Laboratory Presshttp://perspectivesinmedicine.cshlp.org/Downloaded from

Page 5: Gene Therapy Using Stem Cells - CSHL Pperspectivesinmedicine.cshlp.org/content/5/4/a017434.full.pdf · ( 6.5 kb coding sequence) require vectorsthat canaccommodatemorethanthe4.5 kbpackag-inglimitofAAV(Wuetal.2010).Tothisend,the

propriate cell type and at the appropriate level.Transgene expression is commonly driven bystrong, ubiquitously expressing promoter ele-ments, such as CMV. However, overexpressionof large structural proteins such as CEP290 ormembers of the BBSome, a complex of proteinsassociated with Bardet-Biedl syndrome, maylead to cytotoxicity and cell death (Seo et al.2013; Burnight et al. 2014). For an inheritedretinal disease, it is important to direct the ap-propriate level of expression in the cell type spe-cific to the pathophysiology of the disease. Thus,developing a “molecular toolbox” of gene trans-fer reagents is of great interest. This array ofreagents would include promoters specific toheterologous cell or tissue types with varyingdegrees of expressivity and a series of viral vec-tors with a range of carrying capacities (Fig. 2).Indeed, work from our laboratory and othersindicates promoter elements from retina-specif-ic genes such as GRK1, IRBP, RHO, PDE, NRL,and RCVRN achieve photoreceptor-specific ex-pression both in vitro and in vivo (Nicoud et al.2007). However, it has yet to be shown whether

issues relating to the level of transgene expres-sion and cellular toxicity will remain. For somegenes, precise stoichiometry may require iden-tification and use of the endogenous promoterof the therapeutic gene. Conversely, a series ofcell type-specific promoters, such as those indi-cated above, with tunable expression elementsmay be useful. The ability to efficiently generateand test these reagents will almost certainly ben-efit from the ability to create disease-specific celltypes from affected individuals.

GENOME EDITING

As indicated above, successful genetic correc-tion in iPSCs was achieved through gene aug-mentation using exogenous vector systems.Therapeutic transgene expression in the retinapersists for long periods of time, reducing theneed for vector readministration (Bemelmanset al. 2008; Ikeda et al. 2009; Koirala et al.2013; Testa et al. 2013). However, integratingvectors such as those derived from g retroviruscarry the inherent risk of inducing insertional

Constitutive promoter

Expression level

EF1a

Vec

tor

HSV Rhok-Ush2A

Car

go s

ize

AAV

Lenti

PhotoreceptorRhoK, Rec, GNAT2

Cell type specific promoter

RPERPE65, BEST1, MITF

HSV

CMV

RhoK

Ush2A

Figure 2. Schematic diagram depicting a molecular toolbox for exogenous replacement of genes. Genes ofvarious sizes requiring various levels of expression and cell type selectivity.

Gene Therapy Using Stem Cells

Cite this article as Cold Spring Harb Perspect Med 2015;5:a017434 5

ww

w.p

ersp

ecti

vesi

nm

edic

ine.

org

on October 10, 2020 - Published by Cold Spring Harbor Laboratory Presshttp://perspectivesinmedicine.cshlp.org/Downloaded from

Page 6: Gene Therapy Using Stem Cells - CSHL Pperspectivesinmedicine.cshlp.org/content/5/4/a017434.full.pdf · ( 6.5 kb coding sequence) require vectorsthat canaccommodatemorethanthe4.5 kbpackag-inglimitofAAV(Wuetal.2010).Tothisend,the

mutagenesis (Hacein-Bey-Abina et al. 2003). Aswas shown by Hacein-Bey-Abina et al., somepatients who received ex vivo gene augmenta-tion via a g-retroviral vector developed a T cell-like leukemia as a result of vector insertion andproto-oncogene activation in treated cells. Thisadverse event highlights the mutagenic risk in-volved with employing integrating vectors. Fur-thermore, given the fact that precise regulationof gene expression may be required, the abilityto genetically modify defective genes in vitroprovides an attractive strategy for both the cre-ation and the repair of disease-causing muta-tions. Development of patient-specific autolo-gous cells for transplantation would greatlybenefit from such a strategy.

The use of genome editing tools such as zincfinger nucleases (ZFNs), transcription activa-tor-like effector nucleases (TALENs), and therecently reported clustered regularly inter-spersed short tandem repeat (CRISPR)/Cassystem is increasing. By editing the mutationdirectly through double-strand break inductionby a genome editing reagent and subsequent ho-mology-directed repair, one can take advantageof the endogenous expression control elements(i.e., promoters, enhancers, and repressors).This approach addresses some of the more seri-ous limitations of gene replacement such as in-appropriate spatial expression or incorrect levelsof expression from heterologous promotersas well as the possibility of activation or inacti-vation of deleterious genes from vector inte-gration.

In nature, transcription activator-like (TAL)effector proteins are transcription factors usedby the plant pathogen Xanthamonas to evadehost immune mechanisms (Gu et al. 2005; Kayet al. 2007; Sugio et al. 2007). The DNA target-ing domain in the TAL effector protein consistsof tandem repeats of �34 amino acids, at thecenter of which is a highly variable pair of aminoacids that confer nucleotide specificity (Schor-nack et al. 2006). The DNA recognition “code”was recently deciphered (Boch et al. 2009; Mos-cou and Bogdanove 2009), allowing TAL effec-tors to be engineered to recognize specific sitesin the genome. Complexing the engineered TALeffectors with the nuclease domain of the type-

II restriction endonuclease FokI can inducedouble-strand breaks at specific sites at or nearmutations in patient-specific iPSCs. When co-delivered with a donor cassette containing a cor-rected sequence, homology-directed repair canresult in genetic correction of these cells.

Recently Hockemeyer and colleagues (2011)targeted three endogenous loci, PPP1RP12C,OCT4, and PITX3, with engineered TALENs.TALEN expression constructs and donor cas-settes were electroporated into both humanESCs and iPSCs. The researchers reported tar-geting efficiencies of 50%–100% depending onthe locus. To assess binding specificity of thePPP1RP12C TALEN pair, Hockmeyer et al. in-terrogated the 19 top-ranked, maximal-likeli-hood potential off-target sites, as determinedusing systematic evolution of ligands by expo-nential enrichment (SELEX)-derived base-fre-quency matrices (Perez et al. 2008). NextGenIllumina sequencing revealed insertions or de-letions at off-target sites at a very low rate(0.01%), indicating the TALENs were quite spe-cific. Southern analysis at all three sites indicat-ed modification at the intended site only, con-firming these results (Hockemeyer et al. 2011).This valuable series of experiments providessupport for the feasibility of successful genomemodification in pluripotent stem cells, and sug-gests a powerful method for employing geneticrepair in patient-specific, iPSC-derived photo-receptor precursor cells for treatment of inher-ited retinal degenerations. That said, beforeclinical use, it will be important to assess off-targeting in cells treated with genome editingreagents to prevent unwanted outcomes. Dou-ble-strand breaks are induced via heterodimeri-zation of the FokI nuclease domains from twoengineered TALENs targeted to half-recognitionsequences at the intended locus. An unintendedoutcome of TALEN expression is the formationof homodimers, from which off-target cleavagecan result. Thus, reengineering the FokI catalyt-ic domain such that TALEN pairs form obligateheterodimers reduces the frequency of off-targetcleavage (Doyon et al. 2011; Hockemeyer et al.2011; Ramalingam et al. 2011).

Recently, the CRISPR/Cas system has beenintroduced (Cong et al. 2013; Jinek et al. 2013;

E.R. Burnight et al.

6 Cite this article as Cold Spring Harb Perspect Med 2015;5:a017434

ww

w.p

ersp

ecti

vesi

nm

edic

ine.

org

on October 10, 2020 - Published by Cold Spring Harbor Laboratory Presshttp://perspectivesinmedicine.cshlp.org/Downloaded from

Page 7: Gene Therapy Using Stem Cells - CSHL Pperspectivesinmedicine.cshlp.org/content/5/4/a017434.full.pdf · ( 6.5 kb coding sequence) require vectorsthat canaccommodatemorethanthe4.5 kbpackag-inglimitofAAV(Wuetal.2010).Tothisend,the

Mali et al. 2013) and offers several advantagesfor genome editing in iPSCs. First, CRISPR re-agents are easier to design and work more pre-dictably because their DNA targeting is basedon RNA hybridization instead of the zinc-fingerprotein binding of TALENs. The guide RNA ofthe CRISPR system consists of an NGG se-quence (protospacer adjacent motif, or PAM)downstream from the 20-nucleotide target(Cho et al. 2013; Cong et al. 2013; Mali et al.2013). One can generate a CRISPR targetingreagent by simply synthesizing these guide se-quences as complementary oligonucleotidesand ligating them into a bicistronic vector thatalso contains a Cas9 nuclease optimized for ex-pression in human cells (Cong et al. 2013; Ranet al. 2013b).

Recently, Mali and colleagues (2013) showedsuccessful genome editing in human iPSCs us-ing the CRISPR/Cas system. These researchersdeveloped humanized CRISPR/Cas reagentstargeting the endogenous AAVS1 locus. Theyshowed homology-directed repair using a dou-ble-stranded DNA donor cassette in 2%–4%of fibroblast-derived iPSCs generated fromparticipants in the Personal Genome Project(PGP).

As with TALEN and ZFN technology, thepossibility of toxicity from off-target double-stranded breaks must also be addressed whenemploying the CRISPR/Cas system (Fu et al.2013). Targeting via CRISPR/Cas can toleratemispairing of the 20-nucleotide guide RNA(Cong et al. 2013; Jinek et al. 2013), which cre-ates the potential for double-stranded breaks atunintended genomic loci. To address this issue,Cong et al. (2013) mutated a single amino acidin the catalytic domain of the Cas9 nuclease,thereby limiting the enzyme’s activity to sin-gle-strand “nicks.” These “nicks” can be re-paired by the endogenous base excision repairpathway (Lindahl 1993). By using guide RNAswith a mutant Cas9 “nickase,” targeted to se-quences on opposite strands, one can increaseediting specificity and significantly reduce off-target toxicity. For example, Ran and colleagues(2013a) recently showed efficient modifica-tion (.40%) at three loci in human cells usingpaired nickases targeting sequences separated

by four to 20 bases on opposite strands of ge-nomic DNA. Moreover, using deep sequencinganalysis of off targets, these investigatorsshowed a 200- to 1500-fold increase in specific-ity when compared with wild-type Cas9. Ranand colleagues extended these studies in vivoby coinjecting guide RNAs targeting theMecp2 locus and either mutant or wild-typeCas9 mRNA into single-cell mouse zygotes.These experiments yielded .80% modifica-tion at the intended locus. A previous studyalso supported in vivo genome editing withthe CRISPR/Cas system. Wang et al. injectedwild-type Cas9 and guide RNAs targeting theTet1 and Tet2 loci as well as oligo repair tem-plates into zygotes and generated mice withmodification of both genes at a rate of 80%(Wang et al. 2013). This elegant set of experi-ments show the utility of employing nickases toincrease specificity and safety of the valuablegenome editing CRISPR/Cas technology forboth in vitro and in vivo studies (Ran et al.2013a; Wang et al. 2013). To treat patientswho have molecularly confirmed retinal degen-erative disorders, CRISPR/Cas reagents can bedelivered to patient-derived iPSCs to correctdisease-causing mutations before differentiat-ing into photoreceptor precursor cells for trans-plantation. For in vivo therapeutic use, the ad-dition of inducible promoters to allow transientexpression of the guide RNAs may be desirableto further reduce off-target effects. ShouldCRISPR/Cas technology prove safe and effec-tive for in vivo studies, many of the challengesassociated with delivering large transgenes andregulating their level of expression may be over-come.

CONCLUDING REMARKS

In the not-too-distant past, the prognosis forpatients affected with an inherited retinal disor-der was grim. Patients were often told that therewas nothing that could be performed and thattheir fate was irreversible blindness. Fortunately,with developments in stem cell and gene trans-fer technologies there is now realistic hope fortreating many of these individuals. The abilityto accurately identify a patient’s disease-causing

Gene Therapy Using Stem Cells

Cite this article as Cold Spring Harb Perspect Med 2015;5:a017434 7

ww

w.p

ersp

ecti

vesi

nm

edic

ine.

org

on October 10, 2020 - Published by Cold Spring Harbor Laboratory Presshttp://perspectivesinmedicine.cshlp.org/Downloaded from

Page 8: Gene Therapy Using Stem Cells - CSHL Pperspectivesinmedicine.cshlp.org/content/5/4/a017434.full.pdf · ( 6.5 kb coding sequence) require vectorsthat canaccommodatemorethanthe4.5 kbpackag-inglimitofAAV(Wuetal.2010).Tothisend,the

mutations is a prerequisite to the developmentof both gene-based and autologous cell-basedtherapies. As depicted in Figure 3, one can en-vision a strategy in which high-quality genetictesting, coupled with in vitro patient-specific,stem cell-based model systems, will allow forthe rapid development of gene- and cell-basedtreatments. This process would begin by collect-ing blood and skin samples from the affectedpatient. DNA isolated from blood would besubjected to genetic screening to identify po-tential disease-causing mutations, and skin-derived fibroblasts or keratinocytes would beharvested for generation of iPSCs. To confirmpathogenic mutations and elucidate pathophys-iologic mechanisms, iPSC-derived photorecep-tor precursor cells, generated via directed differ-

entiation, could be subjected to a variety ofpathway-specific analyses. Disease-specific phe-notypes identified in these studies would beused to test and confirm the efficacy of genetransfer and genome editing approaches. Genetransfer vectors shown to be effective at correct-ing the disease phenotype would then be used totreat patients’ remaining photoreceptor cells.For patients with extensive photoreceptor cellloss, an autologous cell replacement strategyusing patient-specific photoreceptor precur-sor cells genetically corrected via CRISPR- orTALEN-based genome editing would also beemployed. This approach could be used to treatinherited retinal disorders, regardless of theclinical stage and prevalence of the disease,and regardless of the size of the causative gene.

Sample collection

Blood

Sequencing

Genomic correction ofpatient specific cells

Test efficacy ofgene therapeutics

AAVLenti

HSV

Interrogate pathophysiology

Gene- and cell-based therapies

iPSCgeneration

Skin

Figure 3. Schematic diagram depicting the proposed pipeline from gene discovery to treatment of inheritedretinal degenerative disease.

E.R. Burnight et al.

8 Cite this article as Cold Spring Harb Perspect Med 2015;5:a017434

ww

w.p

ersp

ecti

vesi

nm

edic

ine.

org

on October 10, 2020 - Published by Cold Spring Harbor Laboratory Presshttp://perspectivesinmedicine.cshlp.org/Downloaded from

Page 9: Gene Therapy Using Stem Cells - CSHL Pperspectivesinmedicine.cshlp.org/content/5/4/a017434.full.pdf · ( 6.5 kb coding sequence) require vectorsthat canaccommodatemorethanthe4.5 kbpackag-inglimitofAAV(Wuetal.2010).Tothisend,the

ACKNOWLEDGMENTS

This work was supported by National Institutesof Health (NIH) Directors New InnovatorAward 1-DP2-OD007483-01, NEI EY017451,F32 EY022834, Howard Hughes Medical Insti-tute (HHMI), Foundation Fighting Blindness,Stephen A. Wynn Foundation, Grousbeck Fam-ily Foundation, and Leo, Jacques & MarionHauser Family Vision Restoration Fund.

REFERENCES

Acland GM, Aguirre GD, Ray J, Zhang Q, Aleman TS,Cideciyan AV, Pearce-Kelling SE, Anand V, Zeng Y, Ma-guire AM, et al. 2001. Gene therapy restores vision in acanine model of childhood blindness. Nat Genet 28: 92–95.

Acland GM, Aguirre GD, Bennett J, Aleman TS, CideciyanAV, Bennicelli J, Dejneka NS, Pearce-Kelling SE, MaguireAM, Palczewski K, et al. 2005. Long-term restoration ofrod and cone vision by single dose rAAV-mediated genetransfer to the retina in a canine model of childhoodblindness. Mol Ther 12: 1072–1082.

Asuri P, Bartel MA, Vazin T, Jang J-H, Wong TB, Schaffer DV.2012. Directed evolution of adeno-associated virus forenhanced gene delivery and gene targeting in humanpluripotent stem cells. Mol Ther 20: 329–338.

Bainbridge JWB, Smith AJ, Barker SS, Robbie S, HendersonR, Balaggan K, Viswanathan A, Holder GE, Stockman A,Tyler N, et al. 2008. Effect of gene therapy on visual func-tion in Leber’s congenital amaurosis. N Engl J Med 358:2231–2239.

Balaggan KS, Ali RR. 2012. Ocular gene delivery using len-tiviral vectors. Gene Ther 19: 145–153.

Bemelmans A-P, Kostic C, Cachafeiro M, Crippa SV, WannerD, Tekaya M, Wenzel A, Arsenijevic Y. 2008. Lentiviralgene transfer-mediated cone vision restoration inRPE65 knockout mice. Adv Exp Med Biol 613: 89–95.

Bennicelli J, Wright JF, Komaromy A, Jacobs JB, Hauck B,Zelenaia O, Mingozzi F, Hui D, Chung D, Rex TS, et al.2008. Reversal of blindness in animal models of Lebercongenital amaurosis using optimized AAV2-mediatedgene transfer. Mol Ther 16: 458–465.

Boch J, Scholze H, Schornack S, Landgraf A, Hahn S, Kay S,Lahaye T, Nickstadt A, Bonas U. 2009. Breaking the codeof DNA binding specificity of TAL-type III effectors.Science 326: 1509–1512.

Brambrink T, Foreman R, Welstead GG, Lengner CJ, WernigM, Suh H, Jaenisch R. 2008. Sequential expression ofpluripotency markers during direct reprogramming ofmouse somatic cells. Cell Stem Cell 2: 151–159.

Burnight ER, Wiley LA, Drack AV, Braun TA, Anfinson KR,Kaalberg EE, Halder JA, Affatigato LM, Mullins RF, StoneEM, et al. 2014. CEP290 gene transfer rescues Lebercongenital amaurosis cellular phenotype. Gene Ther 21:662–672.

Calame M, Cachafeiro M, Philippe S, Schouwey K, TekayaM, Wanner D, Sarkis C, Kostic C, Arsenijevic Y. 2011.

Retinal degeneration progression changes lentiviral vec-tor cell targeting in the retina. PLoS ONE 6: e23782.

Carey BW, Markoulaki S, Hanna J, Saha K, Gao Q, Mitali-pova M, Jaenisch R. 2009. Reprogramming of murineand human somatic cells using a single polycistronic vec-tor. Proc Natl Acad Sci 106: 157–162.

Cartier N, Hacein-Bey-Abina S, Bartholomae CC, Veres G,Schmidt M, Kutschera I, Vidaud M, Abel U, Dal-CortivoL, Caccavelli L, et al. 2009. Hematopoietic stem cell genetherapy with a lentiviral vector in X-linked adrenoleuko-dystrophy. Science 326: 818–823.

Chang B, Khanna H, Hawes N, Jimeno D, He S, Lillo C,Parapuram SK, Cheng H, Scott A, Hurd RE, et al. 2006.In-frame deletion in a novel centrosomal/ciliary proteinCEP290/NPHP6 perturbs its interaction with RPGR andresults in early-onset retinal degeneration in the rd16mouse. Hum Mol Genet 15: 1847–1857.

Cho SW, Kim S, Kim JM, Kim J-S. 2013. Targeted genomeengineering in human cells with the Cas9 RNA-guidedendonuclease. Nat Biotechnol 31: 230–232.

Cong L, Ran FA, Cox D, Lin S, Barretto R, Habib N, Hsu PD,Wu X, Jiang W, Marraffini LA, et al. 2013. Multiplexgenome engineering using CRISPR/Cas systems. Science339: 819–823.

Conlon TJ, Deng W-T, Erger K, Cossette T, Pang J-J, Ryals R,Clement N, Cleaver B, McDoom I, Boye SE, et al. 2013.Preclinical potency and safety studies of an AAV2-medi-ated gene therapy vector for the treatment of MERTKassociated retinitis pigmentosa. Hum Gene Ther ClinDev 24: 23–28.

Dai X, Han J, Qi Y, Zhang H, Xiang L, Lv J, Li J, Deng W-T,Chang B, Hauswirth WW, et al. 2014. AAV-mediated ly-sophosphatidylcholine acyltransferase 1 (Lpcat1) gene re-placement therapy rescues retinal degeneration in rd11mice. Invest Ophthalmol Vis Sci 55: 1724–1734.

Doyon Y, Vo TD, Mendel MC, Greenberg SG, Wang J, XiaDF, Miller JC, Urnov FD, Gregory PD, Holmes MC. 2011.Enhancing zinc-finger-nuclease activity with improvedobligate heterodimeric architectures. Nat Methods 8:74–79.

Ellis J. 2005. Silencing and variegation of g retrovirus andlentivirus vectors. Hum Gene Ther 16: 1241–1246.

Endo M, Zoltick PW, Chung DC, Bennett J, Radu A, Mu-varak N, Flake AW. 2007. Gene transfer to ocular stemcells by early gestational intraamniotic injection of lenti-viral vector. Mol Ther 15: 579–587.

Fu Y, Foden JA, Khayter C, Maeder ML, Reyon D, Joung JK,Sander JD. 2013. High-frequency off-target mutagenesisinduced by CRISPR-Cas nucleases in human cells. NatBiotechnol 31: 822–826.

Gaspar HB, Cooray S, Gilmour KC, Parsley KL, Zhang F,Adams S, Bjorkegren E, Bayford J, Brown L, Davies EG,et al. 2011. Hematopoietic stem cell gene therapy foradenosine deaminase-deficient severe combined immu-nodeficiency leads to long-term immunological recoveryand metabolic correction. Sci Transl Med 3: p97ra80.

Gonzalez F, Barragan Monasterio M, Tiscornia G, Montser-rat Pulido N, Vassena R, Batlle Morera L, Rodrıguez-PizaI, Izpisua Belmonte JC. 2009. Generation of mouse-in-duced pluripotent stem cells by transient expression of asingle nonviral polycistronic vector. Proc Natl Acad Sci106: 8918–8922.

Gene Therapy Using Stem Cells

Cite this article as Cold Spring Harb Perspect Med 2015;5:a017434 9

ww

w.p

ersp

ecti

vesi

nm

edic

ine.

org

on October 10, 2020 - Published by Cold Spring Harbor Laboratory Presshttp://perspectivesinmedicine.cshlp.org/Downloaded from

Page 10: Gene Therapy Using Stem Cells - CSHL Pperspectivesinmedicine.cshlp.org/content/5/4/a017434.full.pdf · ( 6.5 kb coding sequence) require vectorsthat canaccommodatemorethanthe4.5 kbpackag-inglimitofAAV(Wuetal.2010).Tothisend,the

Gu K, Yang B, Tian D, Wu L, Wang D, Sreekala C, Yang F,Chu Z, Wang G-L, White FF, et al. 2005. R gene expressioninduced by a type-III effector triggers disease resistance inrice. Nature 435: 1122–1125.

Hacein-Bey-Abina S, Kalle Von C, Schmidt M, McCormackMP, Wulffraat N, Leboulch P, Lim A, Osborne CS, Paw-liuk R, Morillon E, et al. 2003. LMO2-associated clonalT cell proliferation in two patients after gene therapy forSCID-X1. Science 302: 415–419.

Hauswirth WW, Aleman TS, Kaushal S, Cideciyan AV,Schwartz SB, Wang L, Conlon TJ, Boye SL, Flotte TR,Byrne BJ, et al. 2008. Treatment of Leber congenital am-aurosis due to RPE65 mutations by ocular subretinal in-jection of adeno-associated virus gene vector: Short-termresults of a phase I trial. Hum Gene Ther 19: 979–990.

Herbst F, Ball CR, Tuorto F, Nowrouzi A, Wang W, Zavidij O,Dieter SM, Fessler S, van der Hoeven F, Kloz U, et al. 2012.Extensive methylation of promoter sequences silenceslentiviral transgene expression during stem cell differen-tiation in vivo. Mol Ther 20: 1014–1021.

Hockemeyer D, Wang H, Kiani S, Lai CS, Gao Q, Cassady JP,Cost GJ, Zhang L, Santiago Y, Miller JC, et al. 2011. Ge-netic engineering of human pluripotent cells using TALEnucleases. Nat Biotechnol 29: 731–734.

Homma K, Okamoto S, Mandai M, Gotoh N, RajasimhaHK, Chang Y-S, Chen S, Li W, Cogliati T, Swaroop A, et al.2013. Developing rods transplanted into the degeneratingretina of Crx-knockout mice exhibit neural activity sim-ilar to native photoreceptors. Stem Cells 31: 1149–1159.

Hu K. 2014. All roads lead to induced pluripotent stem cells:The technologies of iPSC generation. Stem Cells Dev 23:1285–1300.

Ikeda Y, Yonemitsu Y, Sakamoto T, Ishibashi T, Ueno H, KatoA, Nagai Y, Fukumura M, Inomata H, Hasegawa M, et al.2002. Recombinant Sendai virus-mediated gene transferinto adult rat retinal tissue: Efficient gene transfer by briefexposure. Exp Eye Res 75: 39–48.

Ikeda Y, Yonemitsu Y, Miyazaki M, Kohno R-I, Murakami Y,Murata T, Tabata T, Ueda Y, Ono F, Suzuki T, et al. 2009.Stable retinal gene expression in nonhuman primates viasubretinal injection of SIVagm-based lentiviral vectors.Hum Gene Ther 20: 573–579.

Jin Z-B, Okamoto S, Xiang P, Takahashi M. 2012. Integra-tion-free induced pluripotent stem cells derived fromretinitis pigmentosa patient for disease modeling. StemCells Transl Med 1: 503–509.

Jinek M, East A, Cheng A, Lin S, Ma E, Doudna J. 2013.RNA-programmed genome editing in human cells. eLife2: e00471.

Kay S, Hahn S, Marois E, Hause G, Bonas U. 2007. A bacte-rial effector acts as a plant transcription factor and in-duces a cell size regulator. Science 318: 648–651.

Kim KS, Lee HJ, Jeong HS, Li J, Teng YD, Sidman RL, SnyderEY, Kim SU. 2011. Self-renewal induced efficiently, safely,and effective therapeutically with one regulatable gene ina human somatic progenitor cell. Proc Natl Acad Sci 108:4876–4881.

Koirala A, Conley SM, Makkia R, Liu Z, Cooper MJ, SparrowJR, Naash MI. 2013. Persistence of non-viral vector me-diated RPE65 expression: Case for viability as a genetransfer therapy for RPE-based diseases. J Control Release172: 745–752.

Lamba DA, Gust J, Reh TA. 2009. Transplantation of humanembryonic stem cell-derived photoreceptors restoressome visual function in Crx-deficient mice. Cell StemCell 4: 73–79.

Lamba DA, McUsic A, Hirata RK, Wang P-R, Russell D, RehTA. 2010. Generation, purification and transplantation ofphotoreceptors derived from human induced pluripo-tent stem cells. PLoS ONE 5: e8763.

Li Q, Miller R, Han P-Y, Pang J, Dinculescu A, Chiodo V,Hauswirth WW. 2008. Intraocular route of AAV2 vectoradministration defines humoral immune response andtherapeutic potential. Mol Vis 14: 1760–1769.

Lindahl T. 1993. Instability and decay of the primary struc-ture of DNA. Nature 362: 709–715.

Liu H, Ye Z, Kim Y, Sharkis S, Jang Y-Y. 2010. Generation ofendoderm-derived human induced pluripotent stemcells from primary hepatocytes. Hepatology 51: 1810–1819.

Loh Y-H, Agarwal S, Park I-H, Urbach A, Huo H, HeffnerGC, Kim K, Miller JD, Ng K, Daley GQ. 2009. Generationof induced pluripotent stem cells from human blood.Blood 113: 5476–5479.

Maguire AM, Simonelli F, Pierce EA, Pugh EN, Mingozzi F,Bennicelli J, Banfi S, Marshall KA, Testa F, Surace EM, etal. 2008. Safety and efficacy of gene transfer for Leber’scongenital amaurosis. N Engl J Med 358: 2240–2248.

Maguire AM, High KA, Auricchio A, Wright JF, Pierce EA,Testa F, Mingozzi F, Bennicelli JL, Ying G-S, Rossi S, et al.2009. Age-dependent effects of RPE65 gene therapy forLeber’s congenital amaurosis: A phase 1 dose-escalationtrial. Lancet 374: 1597–1605.

Maherali N, Ahfeldt T, Rigamonti A, Utikal J, Cowan C,Hochedlinger K. 2008. A high-efficiency system for thegeneration and study of human induced pluripotent stemcells. Cell Stem Cell 3: 340–345.

Mali P, Yang L, Esvelt KM, Aach J, Guell M, DiCarlo JE,Norville JE, Church GM. 2013. RNA-guided human ge-nome engineering via Cas9. Science 339: 823–826.

Malik N, Rao MS. 2013. A review of the methods for humaniPSC derivation. Methods Mol Biol 997: 23–33.

Molday LL, Djajadi H, Yan P, Szczygiel L, Boye SL, ChiodoVA, Gregory-Evans K, Sarunic MV, Hauswirth WW, Mol-day RS. 2013. RD3 gene delivery restores guanylate cy-clase localization and rescues photoreceptors in the Rd3mouse model of Leber congenital amaurosis 12. HumMol Genet 22: 3894–3905.

Moscou MJ, Bogdanove AJ. 2009. A simple cipher governsDNA recognition by TAL effectors. Science 326: 1501.

Narfstrom K, Katz ML, Bragadottir R, Seeliger M, BoulangerA, Redmond TM, Caro L, Lai C-M, Rakoczy PE. 2003.Functional and structural recovery of the retina after genetherapy in the RPE65 null mutation dog. Invest Ophthal-mol Vis Sci 44: 1663–1672.

Nicoud M, Kong J, Iqball S, Kan O, Naylor S, Gouras P,Allikmets R, Binley K. 2007. Development of photore-ceptor-specific promoters and their utility to investigateEIAV lentiviral vector mediated gene transfer to photo-receptors. J Gene Med 9: 1015–1023.

Perez EE, Wang J, Miller JC, Jouvenot Y, Kim KA, Liu O,Wang N, Lee G, Bartsevich VV, Lee Y-L, et al. 2008. Es-tablishment of HIV-1 resistance in CD4þ T cells by ge-

E.R. Burnight et al.

10 Cite this article as Cold Spring Harb Perspect Med 2015;5:a017434

ww

w.p

ersp

ecti

vesi

nm

edic

ine.

org

on October 10, 2020 - Published by Cold Spring Harbor Laboratory Presshttp://perspectivesinmedicine.cshlp.org/Downloaded from

Page 11: Gene Therapy Using Stem Cells - CSHL Pperspectivesinmedicine.cshlp.org/content/5/4/a017434.full.pdf · ( 6.5 kb coding sequence) require vectorsthat canaccommodatemorethanthe4.5 kbpackag-inglimitofAAV(Wuetal.2010).Tothisend,the

nome editing using zinc-finger nucleases. Nat Biotechnol26: 808–816.

Preynat-Seauve O, de Rham C, Tirefort D, Ferrari-Lacraz S,Krause K-H, Villard J. 2009. Neural progenitors derivedfrom human embryonic stem cells are targeted by allo-geneic Tand natural killer cells. J Cell Mol Med 13: 3556–3569.

Ramalingam S, Kandavelou K, Rajenderan R, Chandrase-garan S. 2011. Creating designed zinc-finger nucleaseswith minimal cytotoxicity. J Mol Biol 405: 630–641.

Ran FA, Hsu PD, Lin C-Y, Gootenberg JS, Konermann S,Trevino AE, Scott DA, Inoue A, Matoba S, Zhang Y, et al.2013a. Double nicking by RNA-guided CRISPR Cas9 forenhanced genome editing specificity. Cell 154: 1380–1389.

Ran FA, Hsu PD, Wright J, Agarwala V, Scott DA, Zhang F.2013b. Genome engineering using the CRISPR-Cas9 sys-tem. Nat Protoc 8: 2281–2308.

Raya A, Rodrıguez-Piza I, Guenechea G, Vassena R, NavarroS, Barrero MJ, Consiglio A, Castella M, Rıo P, Sleep E, etal. 2009. Disease-corrected haematopoietic progenitorsfrom Fanconi anaemia induced pluripotent stem cells.Nature 460: 53–59.

Reichel MB, Ali RR, Thrasher AJ, Hunt DM, BhattacharyaSS, Baker D. 1998. Immune responses limit adenovirallymediated gene expression in the adult mouse eye. GeneTher 5: 1038–1046.

Ross JW, Fernandez de Castro JP, Zhao J, Samuel M, WaltersE, Rios C, Bray-Ward P, Jones BW, Marc RE, Wang W, etal. 2012. Generation of an inbred miniature pig model ofretinitis pigmentosa. Invest Ophthalmol Vis Sci 53: 501–507.

Schornack S, Meyer A, Romer P, Jordan T, Lahaye T. 2006.Gene-for-gene-mediated recognition of nuclear-targetedAvrBs3-like bacterial effector proteins. J Plant Physiol163: 256–272.

Seo S, Mulins RF, Dumitrescu AV, Bhattarai S, Gratie D,Wang K, Stone E, Sheffield VC, Drack AV. 2013. Subretinalgene therapy of mice with Bardet-Biedl syndrome type 1.Invest Ophthalmol Vis Sci 54: 6118–6132.

Si-Tayeb K, Noto FK, Sepac A, Sedlic F, Bosnjak ZJ, LoughJW, Duncan SA. 2010. Generation of human inducedpluripotent stem cells by simple transient transfectionof plasmid DNA encoding reprogramming factors.BMC Dev Biol 10: 81.

Stadtfeld M, Nagaya M, Utikal J, Weir G, Hochedlinger K.2008. Induced pluripotent stem cells generated withoutviral integration. Science 322: 945–949.

Sugio A, Yang B, Zhu T, White FF. 2007. Two type III effectorgenes of Xanthomonas oryzae pv. oryzae control the in-

duction of the host genes OsTFIIAg1 and OsTFX1 duringbacterial blight of rice. Proc Natl Acad Sci 104: 10720–10725.

Takahashi K, Yamanaka S. 2006. Induction of pluripotentstem cells from mouse embryonic and adult fibroblastcultures by defined factors. Cell 126: 663–676.

Takahashi K, Tanabe K, Ohnuki M, Narita M, Ichisaka T,Tomoda K, Yamanaka S. 2007. Induction of pluripotentstem cells from adult human fibroblasts by defined fac-tors. Cell 131: 861–872.

Testa F, Maguire AM, Rossi S, Pierce EA, Melillo P, MarshallK, Banfi S, Surace EM, Sun J, Acerra C, et al. 2013. Three-year follow-up after unilateral subretinal delivery of ad-eno-associated virus in patients with Leber congenitalamaurosis type 2. Ophthalmology 120: 1283–1291.

Tucker BA, Scheetz TE, Mullins RF, DeLuca AP, HoffmannJM, Johnston RM, Jacobson SG, Sheffield VC, Stone EM.2011. Exome sequencing and analysis of induced plurip-otent stem cells identify the cilia-related gene male germcell-associated kinase (MAK) as a cause of retinitis pig-mentosa. Proc Natl Acad Sci 108: E569–E576.

Tucker BA, Anfinson KR, Mullins RF, Stone EM, Young MJ.2013a. Use of a synthetic xeno-free culture substrate forinduced pluripotent stem cell induction and retinal dif-ferentiation. Stem Cells Transl Med 2: 16–24.

Tucker BA, Mullins RF, Streb LM, Anfinson K, Eyestone ME,Kaalberg E, Riker MJ, Drack AV, Braun TA, Stone EM.2013b. Patient-specific iPSC-derived photoreceptor pre-cursor cells as a means to investigate retinitis pigmentosa.eLife 2: e00824.

Wang H, Yang H, Shivalila CS, Dawlaty MM, Cheng AW,Zhang F, Jaenisch R. 2013. One-step generation of micecarrying mutations in multiple genes by CRISPR/Cas-mediated genome engineering. Cell 153: 910–918.

West EL, Pearson RA, Barker SE, Luhmann UFO, MaclarenRE, Barber AC, Duran Y, Smith AJ, Sowden JC, Ali RR.2010. Long-term survival of photoreceptors transplantedinto the adult murine neural retina requires immunemodulation. Stem Cells 28: 1997–2007.

Wu Z, Yang H, Colosi P. 2010. Effect of genome size on AAVvector packaging. Mol Ther 18: 80–86.

Wu G, Liu N, Rittelmeyer I, Sharma AD, Sgodda M, ZaehresH, Bleidissel M, Greber B, Gentile L, Han DW, et al. 2011.Generation of healthy mice from gene-corrected disease-specific induced pluripotent stem cells. PLoS Biol 9:e1001099.

Yu J, Vodyanik MA, Smuga-Otto K, Antosiewicz-Bourget J,Frane JL, Tian S, Nie J, Jonsdottir GA, Ruotti V, Stewart R,et al. 2007. Induced pluripotent stem cell lines derivedfrom human somatic cells. Science 318: 1917–1920.

Gene Therapy Using Stem Cells

Cite this article as Cold Spring Harb Perspect Med 2015;5:a017434 11

ww

w.p

ersp

ecti

vesi

nm

edic

ine.

org

on October 10, 2020 - Published by Cold Spring Harbor Laboratory Presshttp://perspectivesinmedicine.cshlp.org/Downloaded from

Page 12: Gene Therapy Using Stem Cells - CSHL Pperspectivesinmedicine.cshlp.org/content/5/4/a017434.full.pdf · ( 6.5 kb coding sequence) require vectorsthat canaccommodatemorethanthe4.5 kbpackag-inglimitofAAV(Wuetal.2010).Tothisend,the

November 13, 20142015; doi: 10.1101/cshperspect.a017434 originally published onlineCold Spring Harb Perspect Med 

 Erin R. Burnight, Luke A. Wiley, Robert F. Mullins, Edwin M. Stone and Budd A. Tucker Gene Therapy Using Stem Cells

Subject Collection Retinal Disorders: Genetic Approaches to Diagnosis and Treatment

Degenerations in Systemic DiseaseA Review of Secondary Photoreceptor

Naveen Mysore, Jamie Koenekoop, Shen Li, et al.

What Is Next for Retinal Gene Therapy?Luk H. Vandenberghe

Dominant Retinitis PigmentosaGenes and Mutations Causing Autosomal

SullivanStephen P. Daiger, Sara J. Bowne and Lori S.

and Efficacy Gene Therapy Trials: SafetyRPE65The Status of

Eric A. Pierce and Jean Bennett

Retinal Biology and DiseaseRNA-Seq: Improving Our Understanding of

Sousa, et al.Michael H. Farkas, Elizabeth D. Au, Maria E.

RetinoschisisStudies: Gene Therapy for X-Linked Convergence of Human Genetics and Animal

Ronald A. Bush, Lisa L. Wei and Paul A. Sieving

Macular DegenerationDifferential Gene Expression in Age-Related

Denise J. Morgan and Margaret M. DeAngelisMacular DegenerationGene Therapies for Neovascular Age-Related

ScariaPeter Pechan, Samuel Wadsworth and Abraham

Optic Nerve: Clinical ApplicationsGenetics of Primary Inherited Disorders of the

Keri F. Allen, Eric D. Gaier and Janey L. WiggsMYO7AUsher Syndrome Caused by Mutations in

Gene Therapy for the Retinal Degeneration of

Vanda S. Lopes and David S. WilliamsGenetic Modifiers and Oligogenic Inheritance

Maria Kousi and Nicholas Katsanis-Associated DiseasesABCA4Gene Therapy of

AllikmetsAlberto Auricchio, Ivana Trapani and Rando

Inherited Retinal DegenerationsStem Cells as Tools for Studying the Genetics of

et al.Luke A. Wiley, Erin R. Burnight, Robert F. Mullins,

Gene Therapy Using Stem Cells

et al.Erin R. Burnight, Luke A. Wiley, Robert F. Mullins,

RPGRIP1in Leber Congenital Amaurosis Caused by Mutations

Tiansen LiAdeno-Associated Viral (AAV) VectorGene Therapy for Choroideremia Using an

MacLarenAlun R. Barnard, Markus Groppe and Robert E.

http://perspectivesinmedicine.cshlp.org/cgi/collection/ For additional articles in this collection, see

Copyright © 2015 Cold Spring Harbor Laboratory Press; all rights reserved

on October 10, 2020 - Published by Cold Spring Harbor Laboratory Presshttp://perspectivesinmedicine.cshlp.org/Downloaded from