The genetic engineering of hematopoietic stem cells is the basis for potentially treating a large array of hereditary and acquired diseases, and stands as the paradigm for stem cell engineering in general. Recent clinical reports support the formidable promise of this approach but also highlight the limitations of the technologies used to date, which have on occasion resulted in clonal expansion, myelodysplasia, or leukemogenesis. New research directions, predicated on improved vector designs, targeted gene delivery or the therapeutic use of pluripotent stem cells, herald the advent of safer and more effective hematopoietic stem cell therapies that may transform medical practice. In this review, we place these recent advances in perspective, emphasizing the solutions emerging from a wave of new technologies and highlighting the challenges that lie ahead.

The safe engineering and engraftment of hematopoietic stem cells (HSCs) are the keys to treating a vast spectrum of genetic and acquired disorders that affect hematopoietic and other tissues. These include disorders of the immune system, such as severe combined immunodeficiency (SCID) syndromes and AIDS, the thalassemias, sickle cell anemia, metabolic disorders, including central nervous system pathologies, autoimmune diseases, and an array of hematologic malignancies, which could be treated with cancer-free autologous cells or prevented (eg, in the case of Fanconi anemia).1,2  The safe and effective engineering of HSCs thus represents one of the central goals of stem cell and gene therapies. Since the pioneering studies in adenosine deaminase (ADA) deficiency initiated at the National Institutes of Health in the early 1990s, nearly 100 patients have been treated with genetically modified CD34+ hematopoietic progenitors worldwide (Table 1). This slow adaptation reflects both the complexity of the biologic challenges posed by the ex vivo manipulation and genetic engineering of HSCs as well as the chilling impact of the first report of a leukemic transformation caused by a γ-retroviral vector in a boy with X-linked SCID (X-SCID).3  The series of leukemias that ensued (in 5 of 20 patients with X-SCID), which were later followed by similar adverse events in trials for chronic granulomatous disease and Wiscott-Aldrich syndrome, raised serious doubts as to the merits of this approach and undermined human and financial investments in this field over the past decade. However, these serious adverse events also spurred an enormous, collective investigation into the genotoxicity of gene delivery methodologies, resulting in tremendous progress in our understanding of retroviral vector integration and its impact on endogenous gene structure and function.4-6  Although these serious adverse events have resulted in discontinuation of the use of long terminal repeat (LTR)–driven γ-retroviral vectors for the genetic modification of HSCs, they provided a major impetus for developing novel approaches to genetically modify human cells.

Table 1

Patients treated with engineered HSCs

DiseaseVector typeLTR-drivenNo. of treated patients
ADA γRV 40 
Gaucher αRV 
X-SCID γRV/SIN-γRV +/− 20/3 
CGD γRV 
ALD SIN-LV 
WAS γRV/SIN-LV +/− 10/3 
β-thal SIN-LV − 
MLD SIN-LV − 
DiseaseVector typeLTR-drivenNo. of treated patients
ADA γRV 40 
Gaucher αRV 
X-SCID γRV/SIN-γRV +/− 20/3 
CGD γRV 
ALD SIN-LV 
WAS γRV/SIN-LV +/− 10/3 
β-thal SIN-LV − 
MLD SIN-LV − 

These new trends in HSC engineering broadly fall into 3 categories: improvements in the design of retroviral vectors, development of technologies for targeted gene delivery, and novel approaches made possible by the advent of patient-specific pluripotent stem cells. Some of these have recently entered the clinical arena and others are soon to follow. This review briefly summarizes the first 2 decades of HSC gene therapy, based on the use of first-generation (ie, LTR-driven) γ-retroviral vectors, and critically assesses future directions from the perspective of genetic engineering, examining the prospects, and challenges that lie ahead.

Successes in HSC gene therapy have slowly but steadily accumulated over the past decade. Most early trials focused on severe monogenic immune deficiencies, including ADA deficiency, X-SCID, chronic granulomatous deficiency (CGD), and, more recently, Wiskott-Aldrich syndrome (WAS). These severe disorders were chosen in part because ubiquitous expression of the therapeutic protein (the ADA enzyme for ADA deficiency, the interleukin receptor common γ-chain for X-SCID, the gp91phox oxidase complex protein for CGD, and the WAS signaling integrator protein for WAS), in all hematopoietic cells, not just the defective cell types, was deemed to be acceptable, thus justifying the use of the “first-generation” vectors available in the early 1990s. These consist of recombinant replication-incompetent γ-retroviral genomes derived from murine leukemia viruses (MLVs), termed LXSN,7  MFG/SFG,8  and FMEV,9  which provide constitutive expression of the therapeutic cDNA driven by the viral enhancer/promoter present in the vector's 5′- and 3′-LTRs (Figure 1).

Figure 1

Retroviral vector designs under clinical evaluation. (A) LTR-driven γ-RV, exemplified by the MFG/SFG vector design used in X-SCID and WAS clinical trials. (B) SIN-γ-RV, exemplified by the SRS11 EFS vector design used in the X-SCID consortium trial. (C) Nonspecific SIN-LV, exemplified by the MND-ALD vector design used in the ALD trial. (D) Lineage-restricted SIN-LV, exemplified by the TNS9.3 vector for the treatment of β-thalassemia major. U3 E/P indicates retroviral enhancer/promoter from the LTR U3 region; PRE/WPRE (woodchuck hepatitis), posttranscriptional regulatory element; SIN, self-inactivating vector design (▿ represents U3 deletion); specificity: − indicates ubiquitous; and +, lineage-specific; LCR, locus control region; and HBB, human β-globin gene. Green represents retroviral enhancer/promoter elements; and red, mammalian enhancer/promoter elements.

Figure 1

Retroviral vector designs under clinical evaluation. (A) LTR-driven γ-RV, exemplified by the MFG/SFG vector design used in X-SCID and WAS clinical trials. (B) SIN-γ-RV, exemplified by the SRS11 EFS vector design used in the X-SCID consortium trial. (C) Nonspecific SIN-LV, exemplified by the MND-ALD vector design used in the ALD trial. (D) Lineage-restricted SIN-LV, exemplified by the TNS9.3 vector for the treatment of β-thalassemia major. U3 E/P indicates retroviral enhancer/promoter from the LTR U3 region; PRE/WPRE (woodchuck hepatitis), posttranscriptional regulatory element; SIN, self-inactivating vector design (▿ represents U3 deletion); specificity: − indicates ubiquitous; and +, lineage-specific; LCR, locus control region; and HBB, human β-globin gene. Green represents retroviral enhancer/promoter elements; and red, mammalian enhancer/promoter elements.

Close modal

In terms of therapeutic efficacy, the overall clinical results have been compelling. The majority of patients with SCID, ADA, and WAS showed dramatic improvements in their immune function, including improved T- and B-cell immunity, as well as restored natural killer cell function in X-SCID and regression of eczema and thrombocytopenia in WAS.10-14  Quality of life was improved for the majority of these patients.15  The outcome in CGD, a myeloid disorder in contrast to the aforementioned lymphoid syndromes, and where adults rather than children were treated, was less dramatic but still resulted in short-term regression or stabilization of intractable infections in the first 2 treated subjects.16 

Using LTR-driven expression in another class of retroviral vectors derived from HIV-1, promising clinical results were recently obtained in 2 children with adrenoleukodystrophy (ALD), a disease characterized by multifocal brain demyelination. The 2 first patients showed neuroradiologic improvement and stabilization of their declining cognitive functions.17  Although the observation period is still limited (∼ 3 years), these results bode very well for this class of vectors, which are currently entering the clinic for WAS and other metabolic disorders, including metachromatic leukodystrophy, and β-thalassemia (Table 1). Like the first-generation γ-retroviral vectors, the lentiviral vectors used in the ALD study express the protein nonspecifically in all hematopoietic lineages, including the myeloid cells that eventually reconstitute the brain microglia.18  Significantly, these vectors lack the duplicated full-length LTR that is characteristic of the early γ-RVs, although they still encode an LTR as their internal promoter (Figure 1).

Altogether, these studies support the feasibility of genetically engineering HSCs for use in an autologous setting and the notion that genetically engineered HSCs could provide substantial benefits to patients with a broad range of inherited and acquired disorders.

Just as the benefits of retroviral therapies were starting to be revealed, so were the shortcomings of gene transfer into stem cells. These include limitations of therapeutic efficacy and toxicities, especially genotoxicity. Some of these problems are inherent to stem cell harvest and cell culture, whereas others are disease- or vector-specific. A successful therapy requires polyclonal hematopoietic reconstitution by self-renewing HSCs, with a sufficient fraction of engrafting HSCs that harbor the vector and expression of the corrective genetic material on a per-cell basis that reaches over the thresholds required to therapeutically impact on the underlying disease over the long term.

Several obstacles of a quantitative nature can interfere with this objective. Effective retroviral transduction of HSCs requires having enough patient CD34+ cells and adequate vector stocks for their transduction. Although CD34+ cell collection from bone marrow or mobilized blood is usually satisfactory, it may be challenging in some conditions, such as Fanconi anemia19  and sickle cell anemia.20 

Current approaches to vector transduction require ex vivo culture of CD34+ cells in the presence of pro-survival cytokines, and even short-term culture results in decreased engraftment and ability to compete with endogenous HSCs, perhaps resulting from loss of self-renewal capacity or defects in homing.21,22  As a result, some degree of potentially toxic conditioning with chemotherapy or irradiation is required in clinical settings where corrected HSCs and their progeny do not have a competitive advantage.

Vector production to reach a sufficient titer may pose a challenge. Although adequate for most phase 1 or 2 studies, robust manufacturing to support larger trials has yet to be developed. Some specific vectors, such as the complex globin vectors23-25  or vectors containing the cHS4 insulator element,26,27  exhibit lower titers that pose a manufacturing challenge. More research on vector production is needed to advance the field and eventually meet commercial goals.

Sufficient and stable expression of vector-encoded transgenes is another category of concern because of the vagaries of position effects and the risk of transcriptional inactivation.28-30  Although well documented and extensively studied in murine models,30,31  vector silencing has been less investigated in clinical studies. It is noteworthy that the vast collections of integration sites documented in many trials10,11  do not provide information on vector expression. Furthermore, studies in SCID and WAS may blunt such an analysis because of the selective outgrowth of transgene-expressing cells. In the case of CGD, where such selective pressure on transgene expression does not apply, silencing and methylation of the vector's retroviral promoter were found in several clones as early as 5 months after therapy.32  On the other hand, very prolonged expression of marker genes, with no evidence for significant silencing, has been documented in early human clinical and nonhuman primate studies using both γ-retroviral and lentiviral vectors.33-35  Silencing may be more problematic in murine cells, which have evolved mechanisms for inactivation of the huge proviral load integrated into the murine endogenous genome, but occurs in human cells as well.

The toxicities associated with the γ-retroviral transduction of HSCs are the consequence of the semirandom pattern of retroviral integration and the presence of strong enhancers in the proviral LTR,36-38  resulting in obligatory insertional mutagenesis. Activation of proto-oncogenes in the genomic neighborhood is the most dreaded consequence. The direst outcome, frank leukemic transformation, has been dramatically illustrated in X-SCID and WAS, where so far 5 of 20 and 1 of 10 patients, respectively, have developed clonal T-cell leukemias.39,40  A surprising feature of these clonal transformations, all linked to the integration of an LTR-driven γ-retroviral vector in the vicinity of an oncogene, is the striking involvement in all but one case of the LMO-2 gene (Table 2).41,42  This gene is expressed in early hematopoietic progenitors (where it is therefore accessible to the retroviral pre-integration complex) and normally silenced on hematopoietic differentiation, unless the LTR prevents this from occurring,43  which may result in the activation of an HSC-like transcription profile in thymocytes and clonal expansion, eventually resulting in full malignant transformation with the acquisition over time of additional chromosomal abnormalities or point mutations in genes, such as Notch.44  Just as striking is the absence of such transformations in patients with ADA-SCID, who also harbor LTR-containing vectors in their T-cell precursors, including occasional integrations in the vicinity of LMO2.10  Marked expansion of a single corrected clone, persisting without malignant transformation for many years, has been documented in an early ADA-SCID gene therapy trial, suggesting that clonal expansion does not irrevocably progress to malignancy.45  The reason for such contrasting outcomes is still unclear and may have to do the nature of the disease, with less profound and rapid expansion of corrected T-cell precursors, or a unique interaction between the transgene and activated LMO2.46 

Table 2

Clonal expansion, myelodysplasias, and transformation

Patient/disease/transgeneRelevant vector sequences (references)Secondary effect (mo after treatment)Genomic insertion sites (transcript status)Other genetic alterations (mo after treatment)Reference(s)
P4/SCID-X1/γC MFG(B2), Moloney-MLV LTR (8,138T-ALL, mature T cell (30) LMO2 (↑) Translocation(6,13); CDKN2A deletion 3,139  
P5/SCID-X1/γC T-ALL, late cortical T cell (34LMO2 (↑) SIL-TAL microdeletion, trisomy 10, Notch mutation (1593F/S) 139  
P7/SCID-X1/γC T-ALL, late cortical T cell (68CCND2 (↑) CDKN2A deletion 42,139  
P10/SCID-X1/γC T-ALL, late cortical T cell (33LMO2 (↑), BMI1 (↑) Notch mutation (1707A/P) 42,139  
P1/X-CGD/gp91phox SFFV LTR (9,16Multiple predominant progenitor cell clones (5), subsequent oligoclonal hematopoiesis, monosomy 7 (21), MDS (27MDS1-EVI1 (↑), PRDM16 (=), SETBP1 (↑) CpG methylation in promoter of the viral LTR (9); CDKN2B and p15INK4B hypermethylation; phosphorylation of H2AX and DNA double-strand breaks (27) 16,32  
P2/X-CGD/gp91phox Multiple predominant progenitor cell clones (5), subsequent oligoclonal hematopoiesis, monosomy 7 (33), MDS (43MDS1-EVI1 (↑), PRDM16 (↑) CpG methylation in promoter of the viral LTR (15); CDKN2B and p15INK4B hypermethylation; phosphorylation of H2AX and DNA double-strand breaks (43) 16,32  
P8/SCID-X1/γC MFG, Moloney-MLV LTR (8,140T-ALL (24) LMO2 (↑) Notch1 mutation (gain-of-function, 1559R/P), CDKN2A deletion, TCRb/STIL-TAL1 translocation 41  
P2/Thalassemia/β(T87Q)-globin ΔU3 HIV LTR + 2xcHS4 insulators (24,141Dominant, myeloid-biased cell clone HMGA2 (↑) Vector rearrangement; transcriptional activation of HMGA2 in erythroid cells with increased expression of a truncated HMGA2 mRNA insensitive to degradation by let-7 micro-RNAs 24  
Patient/disease/transgeneRelevant vector sequences (references)Secondary effect (mo after treatment)Genomic insertion sites (transcript status)Other genetic alterations (mo after treatment)Reference(s)
P4/SCID-X1/γC MFG(B2), Moloney-MLV LTR (8,138T-ALL, mature T cell (30) LMO2 (↑) Translocation(6,13); CDKN2A deletion 3,139  
P5/SCID-X1/γC T-ALL, late cortical T cell (34LMO2 (↑) SIL-TAL microdeletion, trisomy 10, Notch mutation (1593F/S) 139  
P7/SCID-X1/γC T-ALL, late cortical T cell (68CCND2 (↑) CDKN2A deletion 42,139  
P10/SCID-X1/γC T-ALL, late cortical T cell (33LMO2 (↑), BMI1 (↑) Notch mutation (1707A/P) 42,139  
P1/X-CGD/gp91phox SFFV LTR (9,16Multiple predominant progenitor cell clones (5), subsequent oligoclonal hematopoiesis, monosomy 7 (21), MDS (27MDS1-EVI1 (↑), PRDM16 (=), SETBP1 (↑) CpG methylation in promoter of the viral LTR (9); CDKN2B and p15INK4B hypermethylation; phosphorylation of H2AX and DNA double-strand breaks (27) 16,32  
P2/X-CGD/gp91phox Multiple predominant progenitor cell clones (5), subsequent oligoclonal hematopoiesis, monosomy 7 (33), MDS (43MDS1-EVI1 (↑), PRDM16 (↑) CpG methylation in promoter of the viral LTR (15); CDKN2B and p15INK4B hypermethylation; phosphorylation of H2AX and DNA double-strand breaks (43) 16,32  
P8/SCID-X1/γC MFG, Moloney-MLV LTR (8,140T-ALL (24) LMO2 (↑) Notch1 mutation (gain-of-function, 1559R/P), CDKN2A deletion, TCRb/STIL-TAL1 translocation 41  
P2/Thalassemia/β(T87Q)-globin ΔU3 HIV LTR + 2xcHS4 insulators (24,141Dominant, myeloid-biased cell clone HMGA2 (↑) Vector rearrangement; transcriptional activation of HMGA2 in erythroid cells with increased expression of a truncated HMGA2 mRNA insensitive to degradation by let-7 micro-RNAs 24  

The anticipation that malignant transformation after HSC gene therapy would be limited to SCID patients receiving cells engineered to overexpression, a growth-promoting gene, such as the common γ-cytokine receptor and resulting from a unique interaction with insertions activating LMO2, was quashed by subsequent reports indicating that insertional mutagenesis after HSC gene transfer using γ-retroviral was a universal risk. The lack of events in the earlier clinical trials and large animal studies may have resulted from exceedingly low HSC gene transfer efficiency and lack of prolonged follow-up. Nonhuman primates and serially transplanted mice receiving HSCs transduced with γ-retroviral vectors carrying only marker genes developed clonal over-representation or overt myeloid and lymphoid leukemias, with LTR activation of a stereotypical group of proto-oncogenes, most strikingly MDS1/EVI1.47-50  Two patients with CGD receiving corrected CD34+ cells developed first clonal expansion of myeloid cells with vector insertions activating the MDS1/EVI1 or the related PRDM16 gene loci, and then clonal myelodysplasia and marrow failure, with acquisition of an additional monosomy 7 abnormality in the malignant clone in both patients16,32  (Table 2).

The use of LTR-driven γ-retroviral vectors to modify HSCs is thus all but over (with the possible exception of ADA deficiency, which has been remarkably devoid of malignant complications to date). New approaches are needed. The next paths to HSC gene therapy point in 3 directions, pursuing either new designs of randomly integrating viral vectors, targeted gene delivery strategies, or the use of reprogrammed stem cells. Each one of these avenues shows great promise but also distinctive, real challenges.

New retroviral vector types and vector designs, using a “self-inactivating” (SIN) vector modification of γ-retroviral vectors,51  lentiviral vectors,52  and lineage-restricted vectors,23  are now entering the clinic (Table 1). The first fundamental alteration to the first-generation design (Figure 1A) was the elimination of strong promoter/enhancer elements in integrated proviral LTRs via deletion of the LTR enhancer/promoter region from the 3′ end of the vector, which on proviral integration replaces the 5′ LTR. This SIN design then requires the incorporation of an internal promoter to drive transgene expression (Figure 1B). Hardly a new technique,51  this γ-retroviral vector design is now in use in an X-SCID clinical trial (Table 1). Because of concerns regarding recombination with endogenous HIV, this SIN vector design has been adopted from the get-go in later vectors derived from HIV-152  and foamy viruses53  (Figure 1C).

In addition, HIV-derived vectors may possess a safety advantage over those derived from MLV because of their natural propensity to integrate all along transcription units without preference for promoter regions, in contrast to LTR-driven MLV-derived vectors, as documented both in cell lines and in predictive large animal models.36-38,54-56  These differences in integration patterns have now been verified in human clinical trials via large-scale insertion site analyses.11,17,57,58  New studies using SIN γ-retroviral vectors will be interesting to compare in this regard. Common integration sites detected after HSC lentiviral transduction and transplantation are located throughout large genomic regions and appear to result from integration biases associated with the additional factor of in vivo clonal selection and expansion via proto-oncogene activation documented with γ-retroviral HSC gene transfer.57,59  A number of reviews discuss recent insights into proviral integration into the genome, made possible by high throughput retrieval of integration sites and next-generation sequencing.60-63  The most commonly used lentiviral vectors typically harbor ubiquitous internal promoters to drive transgene expression, such as that of human phosphoglycerate kinase or elongation factor-1α (EF-1α). These are not strong promoters, but they appear adequate for correction of enzymopathies, for which modest amounts of transgene product are therapeutic (on the order of femtograms protein per cell).64  Whether phosphoglycerate kinase, EF-1α, or WAS promoter-driven vectors will prove to be sufficient to redress defects in structural proteins or signaling molecules or receptors is yet to be determined. A concern is that the use of stronger, ubiquitous polII promoter/enhancers will increase the risk of trans-activating neighboring genes and malignant transformation back toward the level of genotoxicity encountered with intact LTR regulatory elements. However, there is evidence that enhancers located within a vector are less prone to activation of neighboring genes than the same enhancer contained within an LTR.65  Expression of miRNAs and shRNAs also require robust expression levels,66,67  but the polIII promoters they use may pose a lesser risk of deregulating endogenous gene expression than polII promoters. RNA-based anti-HIV moieties composing an shRNA to tat/rev, a TAR decoy, and an anti-CCR5 ribozyme were recently evaluated after lentiviral-mediated gene transfer to autologous CD34+ cells in subjects with AIDS-related lymphoma.68  Whereas the gene-marking levels were approximately 2 logs lower than in the ALD study, well below levels expected to provide a therapeutic benefit, sustained expression of the shRNA was detected for up to 24 months in one of the patients, without discernable toxicity. For the most common inherited blood disorders, including the thalassemias and sickle cell anemia, high-level globin chain expression (on the order of picograms per cell)64  requires the vectors to incorporate powerful erythroid-specific enhancers (Figure 1D),23  reviewed elsewhere.69  Here the safety concern posed by inclusion of powerful elements is in part mitigated by their tissue specificity, limiting the probability that an adjacent oncogene would be trans-activated in nonerythroid cells.43,64  A single patient treated with such a vector has been followed for more than 3 years.24  This subject, afflicted with HbE thalassemia, showed sustained expression of the vector-encoded globin starting 5 to 6 months after transplantation, most of which could be attributed to a single expanded clone. This patient is currently leukemia-free despite the prolonged clonal expansion and continues to produce an additional 2 to 3 grams per dL of hemoglobin comprising the vector-encoded β(T37Q)–globin chain. The proviral insertion in this clone resulted in aberrant splicing and dysregulated expression of the HMGA2 gene. A recent murine study links HMGA2 overexpression to clonal myeloid expansion, without leukemic transformation.70  This example serves as a cautionary note regarding the risk for clonal expansion with any integrating vector, even those without strong constitutive enhancers.

Powerful enhancers, especially nonspecific ones, would probably require to be flanked by genetic elements with enhancer-promoter blocking activity.71,72  The optimization use of such elements, however, still remains elusive,73,74  their utility is unproven in human or relevant large animal models, and inclusion in vectors may even precipitate mutagenic events, as demonstrated by the alternative splicing from HMGA2 to the cHS4 chicken insulator core element in the aforementioned expanded clone.24  Additional genetic switches and posttranscriptional regulatory mechanisms75  may add a further layer of control to these various vector designs.

Several other types of integrating retroviruses are being developed as potential gene therapy vectors for HSCs but are much less far along in development and have not yet been used in clinical trials. The human foamy virus has not been associated with disease in any species, has a broad target cell range, and efficiently transduces hematopoietic cells.76  Its integration profile is remarkably random, and it has been used to phenotypically correct CD18 integrin deficiency in a canine model of leukocyte adhesion deficiency.77  The avian sarcoma leucosis virus has also been developed and tested in a nonhuman primate HSC transplantation model. It also has a relatively random integration pattern and has the advantage that the LTR promoter/enhancer is completely inactive in mammalian cells.78 

The inclusion of suicide genes in vectors, which would allow ablation of vector-containing cells in the context of an adverse event, has been little investigated in HSCs compared with other cell types, but new studies are exploring their efficacy in preclinical models. To this end, a number of new highly effective suicide genes have been reported,79,80  including the human-derived, dimerizable caspase-9 gene.81  In sum, all of the vector designs shown in Figure 1B through D are expected to reduce the risk of insertional oncogenesis relative to that of LTR-driven γ-retroviral vectors. A number of preclinical models have been developed to try to assess genotoxic risk qualitatively or quantitatively before new vectors are used clinically. These include in vitro immortalization of murine myeloid progenitor cells,82-84  serial transplantation in a murine model,50  transplantation of transduced HSCs from genetically tumor-prone mouse strains,85  or long-term follow-up of nonhuman primates.56  It is reassuring that most of these models come to similar conclusions on the relative genotoxicity of different vector backbones. However, the proof can only come from clinical studies, which will take several more years to come to fruition. It is noteworthy that the vector configurations shown in Figure 1B through D are already undergoing clinical testing (Table 1).

The aforementioned strategies all continue to rely on semirandom integration of the vector provirus into the HSC genome and, as such, will never be free of genotoxic risk. An alternative goal is the targeting of transgene delivery to a predetermined chromosomal location, or the repair of a mutated locus, greatly decreasing the risk of insertional mutagenesis. Targeted gene delivery and gene repair would be optimal if clinically relevant targeting efficiencies can be achieved without off-target genotoxicity or immediate toxicity to transduced cells.

The gold standard for targeted gene delivery is homologous recombination (HR). HR is a DNA repair mechanism that has been successfully used to repair mutated genes and is therefore applicable in principle to cell-based therapies of monogenic diseases.86  Gene targeting by HR requires the use of homologous DNA surrounding the targeted site, usually delivered as plasmid DNA. Introducing large amounts of plasmid DNA into target cells is inefficient and toxic and has thus posed a major challenge in HSCs. The efficiency of DNA entry and of HR can be increased with the use of adenoviral87  and adeno-associated virus vectors,88,89  but these vector types are not well suited for use in HSCs. Another technique to promote specific HR uses triplex-forming oligonucleotides that bind the major groove of duplex DNA,90  which are coupled to a donor DNA sequence. Using nanoparticles for intracellular oligonucleotide delivery, this approach has been shown to target the endogenous β-globin locus in human CD34+ cells, resulting in levels of globin gene modification in the range of 0.5% to 1.0%.91 

The efficiency of HR versus nonhomologous recombination can be increased by the introduction of DNA double-strand breaks at the targeted site using an endonuclease.92  This requires the transient expression of an endonuclease, which can be directed to a specific sequence using modular zinc finger proteins,86  homing endonucleases,93  or transcription activator-like effectors derived from phytopathogenic bacteria.94  Zinc finger nucleases were recently shown to afford remarkable targeting frequencies at the CCR5 locus, disrupting an average 17% of all CCR5 loci in CD34+ cord blood cells, with retained ability to engraft immunodeficient mice and demonstration of engrafted human CCR5-disrupted cells resistant to HIV infection.95  Significant questions remain regarding the efficiency of targeting in bona fide HSCs, and the risk of inflicting off-target effects, which may result in translocations or occult genotoxicity.96  The ability to accurately predict and monitor the off-target effects of modified endonucleases is still an open question.97,98 

Gene targeting via HR in HSCs has lagged behind other target cell types because of the challenges inherent in efficiently and nontoxically introducing the endonuclease and corrective targeting constructs into these fragile and rare cells unable to be cloned or effectively expanded ex vivo. However, there has been recent progress using nonintegrating lentiviral vectors or optimized nucleofection.95,99  Off-target genotoxicity and efficiency of long-term HSC correction will be difficult to assess in xenograft models and will require both large-animals studies and pilot clinical trials in a patient population with sufficiently serious disease complications to justify introduction of these potentially risky approaches.

Current strategies to genetically engineer HSCs are confined by our inability to expand or subclone genetically modified HSCs, whether adult or cord blood-derived. Whether this is the result of properties inherent to HSCs or lack of knowledge regarding appropriate culture conditions, any strategy for mitigation of genotoxicity dependent on screening of vector insertion sites before cell administration is not feasible at this time. Recent advances in pluripotent stem cell technology may, however, transform the face of stem cell engineering, allowing much better characterization of corrected cells before clinical use. The ground-breaking discovery of Yamanaka, who successfully reprogrammed mouse fibroblasts to a pluripotent state similar to that of embryonic stem cells after γRV-mediated introduction of the transcription factors Oct4, Sox2, Klf4, and c-myc,100  opens up new prospects for therapeutic stem cell engineering. The feasibility of expanding pluripotent stem cells without compromising their stem cell properties makes it possible to subclone and select genetically modified cells, as well as to perform extensive efficacy and safety testing in the selected clonal derivatives (Figure 2). Thus, relatively inefficient techniques, such as classic HR, which are inapplicable to HSCs because of their inefficiency, now become relevant. These concepts were dramatically illustrated by Hanna et al in a mouse model of sickle cell anemia.101  In this study, the βS-globin gene was corrected by HR in a fibroblast obtained from a humanized sickle cell transgenic mouse, which was then reprogrammed to a pluripotent state by retroviral transduction100  and subjected to in vitro directed hematopoietic differentiation in the presence of HoxB4 protein. Transplantation of the specified hematopoietic cells did not achieve full hematopoietic reconstitution but effectively blunted the sickle cell syndrome.101 

Figure 2

Evolving paradigms in HSC engineering. (A) Current strategies are restricted by the use of nonclonable adult HSCs. (B) The advent of patient-specific pluripotent stem cells may open new strategies for genetic engineering and biosafety testing.

Figure 2

Evolving paradigms in HSC engineering. (A) Current strategies are restricted by the use of nonclonable adult HSCs. (B) The advent of patient-specific pluripotent stem cells may open new strategies for genetic engineering and biosafety testing.

Close modal

Patient-specific induced pluripotent stem (iPS) cells can be generated from various cell types obtained from patients with inherited or acquired disorders, using a range of techniques.102,103  Use of nonintegrating or excisable vectors for generation of iPS cells may avoid issues of insertional mutagenesis and incomplete silencing of reprogramming factors. However, new genetic material must be permanently introduced to correct the underlying disease mutation. Recognizing that iPS-like cells can arise by LV-mediated insertional mutagenesis alone,104  one approach is to pursue targeted correction, via engineered zinc finger nucleases,105  bacterial artificial chromosomes,106  and adeno-associated virus-mediated HR.88  An alternative to HR and its enhanced variations is to screen iPS clones for the integration of lentiviral or other vectors into putative genomic “safe harbors” (ie, sites that sustain transgene expression without interfering with endogenous gene expression).107,108  This capitalizes on the high efficiency of lentiviral transduction and their lack of nonspecific occult genotoxicity but is potentially constrained by imperfect knowledge of all possible mechanisms by which integrated foreign DNA can dysregulate gene expression.

The ability to generate patient-specific iPS cells and correct their abnormalities via genetic repair or transgene delivery at well-characterized “safe harbor” sites108  does not imply that the use of pluripotent stem cells as a source of HSCs is ready for implementation or that it may ever become suitable for human application. Several major questions need to be further investigated: how to best generate iPS cells efficiently with minimal genotoxicity imparted through the reprogramming process; how to identify and qualify iPS clones that are suitable for clinical investigation, which addresses the genetic, epigenetic, tumorigenic, and differentiation potential of individual iPS clones; how to genetically engineer iPS clones effectively but without adding any genotoxic insults through the repair process; how to generate engraftable HSC-like cells capable of full and durable hematopoietic reconstitution in transplanted recipients; and how to scale up the differentiation culture processes and ensure the depletion of cells with teratoma formation potential. Some of these concerns apply to the use of pluripotent stem cells or reprogrammed cells in general, whereas others specifically relate to their potential use for hematopoietic applications.

Recent studies have documented frequent genetic alterations in human ES and iPS cells, including point mutations (some affecting oncogenes), deletions, and gene duplications.109-111  These occur independently of the reprogramming vectors and are thus distinct from the problems related to insertional mutagenesis. These events may be in part linked to the reprogramming phase, which is known to be enhanced in the absence of p53.112-114  Successful reprogramming could even require accumulation of genetic and/or epigenetic alterations in cells undergoing extended cell culture, including pluripotent stem cells, and the low efficiency of reprogramming may result from a requirement for rare permissive mutations.115-117  In one recent report comparing the genome sequence of fibroblasts before and after reprogramming,118  several mutations found in the iPS cells could be traced back to the original fibroblast; however, others arose during the reprogramming period. No further genomic alterations were detected after further clonal expansion, suggesting the unique susceptibility or requirement for genomic changes during reprogramming. More studies are needed to better gauge the intrinsic genotoxicity of reprogramming, which may be unavoidable even if reprogramming were to be induced by chemical or other means that avoid the use of integrating vectors. The immunogenicity of iPS-derived cells may also cause concern, although currently available information is limited to the pluripotent stem cells themselves.119 

The generation of engraftable adult HSCs from human ES and iPS cells remains elusive to date and probably represents the single largest hurdle to use of pluripotent or reprogrammed cells for hematologic applications. The first hematopoietic cells arise in the primitive streak of the embryo, yielding a distinctive “primitive” hematopoiesis120  that cannot reconstitute adult hosts, and produces, for example, erythroid cells with embryonic hemoglobins that would be unable to function optimally for oxygen delivery in postnatal life. The immediate precursors of “definitive” HSCs are arterial endothelial cells, which generate HSCs capable of long-term multilineage repopulation of adult hosts beginning in the dorsal aorta of the aorta-gonad-mesonephros region and the chorioallantoic vessels of the placenta.121-126  It is these early CD34+, c-kit+, CD41+ HSCs that migrate to the yolk sac and the fetal liver, where they vastly expand before relocating to the bone marrow around birth.120  To date, candidate HSCs derived from human ES and iPS cells by and large fail to engraft and reconstitute irradiated adult recipients.127-130  Even production of engrafting HSCs from murine ES cells, studied intensively for more than 20 years, is extremely inefficient. Only the ectopic expression of the transcription factor HoxB4 in the hematopoietic progeny of murine ESCs has resulted in long-term efficient in vivo engraftment.131  Ectopic expression of HoxB4 has been shown to result in abnormal myeloid/lymphoid ratios in mice, and leukemogenesis in dogs and monkeys, suggesting that this approach to driving hematopoiesis from pluripotent ES or IPS cells is not clinically relevant.84,132,133 

Another recent approach, bypassing the need for iPS cells and the obstacles to generating HSCs from embryonic-type cells, consists of direct reprogramming of skin cells to a multipotent progenitor stage via introduction of a single transcription factor, Oct4.134  Unlike ES and iPS-derived hematopoietic cells, Oct4-reprogrammed progenitor cells possess desirable traits, such as the expression of adult globin genes on erythroid differentiation, and robust albeit short-term myeloid engraftment potential in immunodeficient mice. The exact nature and therapeutic potential of these cells are presently unknown, but these findings point to tantalizing discoveries that may come out of reprogramming and trans-differentiation research.135  Another game-changing advance would entail the ability to reprogram human adult HSCs to an expandable state without diminishing their long-term self-renewal properties and their safety, a goal that has remained elusive to date.

HSC engineering is now at a crossroads. HSC gene therapy has proven benefits in patients with severe immunodeficiencies, but the MLV-derived LTR-driven vectors used in initial clinical trials pose too great a risk of genotoxicity for further clinical development. As their clinical use comes to an end, with the possible exception of ADA deficiency, the first chapter of HSC gene therapy is now closed. New approaches are needed, and their development will greatly benefit from important lessons learned in the early age of gene therapy. Clinical experience with LTR enhancer-deleted MLV vector- or lentiviral vector-transduced HSCs is not mature enough for meaningful risk assessment, although results from animal models and in vitro transformation assays suggest that either of these vector classes will be significantly less genotoxic and thus safer. Any integrating vector can integrate near and activate oncogenes, but removal of strong ubiquitous enhancers from both self-inactivating MLV vectors and lentiviral vectors will greatly decrease the latter risk. However, other genetic effects, such as abnormal splicing events,24  or inactivation of tumor suppressor genes, may still occur. Lentiviral vectors, as well as several novel types of vectors in preclinical development, including those derived from human foamy virus or avian sarcoma leucosis virus, have integration profiles that differ subtly in the targeting of genes or their promoter region, which may result in significantly different safety profiles.36,78,136  Lentiviral vectors encoding ubiquitous promoters of moderate strength afford adequate titers and express transgenes at levels that seem to be appropriate for enzymatic deficiencies. The design of vectors that are better suited for conditions requiring higher protein expression is at the present less well defined.

Targeted gene delivery approaches, including the use of zinc finger nucleases, meganucleases, transcription activator-like effector nucleases, and triplex-forming oligonucleotides, have made great advances. Their targeting frequencies, albeit 1 to 2 logs lower than retroviral-mediated gene transfer efficiency, are on the way to reaching clinically relevant values.

Somatic cell reprogramming opens the door to many genetic engineering approaches, including screening for retroviral integrations in potential genomic safe harbors108  and targeted gene delivery, including nuclease-based approaches and adeno-associated virus-mediated homologous recombination. The advent of iPS cells is far from clinically relevant and poses a number of fascinating biologic questions, spanning a broad range of issues that concern the epigenetic and genetic status of iPS cells, their differentiation potential, and their propensity to transform, whether they have been genetically modified or not. In particular, the generation of human HSCs from ES or iPS cells remains an enigma, one that will hopefully soon be resolved.

Genotoxicities remain a fundamental concern in all the aforementioned approaches: insertional mutagenesis in the case of retroviral-mediated gene transfer, off-target effects when using nucleases to induce double-strand breaks to enhance targeted gene delivery, genetic alterations incurred during reprogramming to a pluripotent state, and genetic alterations arising throughout extended cell culture. Inclusion of suicide genes137  allowing in vivo ablation of dangerous clones may be worthwhile and is under preclinical development.

The first 2 decades of HSC gene therapy have been rich in lessons, providing both strong support for the therapeutic potential of this approach and sobering lessons on the shortcomings of the genetic engineering of stem cells. As the chapter on LTR-driven vectors comes to a close, several new chapters are already being written. The immediate future will evaluate SIN-γRVs, SIN-LVs, and lineage-restricted vectors, all of which should reduce the risk of trans-activating proto-oncogenes after semirandom integrations. Next, targeted gene delivery systems have the potential to further reduce the risk of integrating vectors at undesirable chromosomal locations. Later, if pluripotent stem cells fulfill their promise for the generation of HSCs and if the genotoxicity issues of their own prove not to be prohibitive, genetically corrected cells in which vector integration or gene repair can be fully ascertained before cell infusion will become available. Despite the significant challenges, at least one of these new directions will eventually lead to safe and effective HSC therapies for hereditary and acquired disorders. HSC engineering remains one of the most tantalizing medical research objectives for the 21st century.

The authors thank Jason Plotkin for help with figures.

I.R. and M.S. were supported by the National Institutes of Health (grants HL053750, CA59350, and CA08748), the Experimental Therapeutics Center at Memorial Sloan-Kettering Cancer Center, the Niarchos Foundation, the Leonardo Giambrone Foundation, the Cooley's Anemia Foundation, and NYSTEM.

National Institutes of Health

Contribution: I.R., C.E.D., and M.S. wrote the manuscript.

Conflict-of-interest disclosure: M.S. holds patents on globin gene transfer and chimeric antigen receptors for immune engineering. The remaining authors declare no competing financial interests.

Correspondence: Michel Sadelain, Center for Cell Engineering, Molecular Pharmacology and Chemistry Program, Memorial Sloan-Kettering Cancer Center, Box 182, 1275 York Ave, New York, NY 10065; e-mail: m-sadelain@ski.mskcc.org.

1
Dunbar
 
CE
Gene transfer to hematopoietic stem cells: implications for gene therapy of human disease.
Annu Rev Med
1996
, vol. 
47
 (pg. 
11
-
20
)
2
Kohn
 
DB
Gene therapy for genetic haematological disorders and immunodeficiencies.
J Intern Med
2001
, vol. 
249
 
4
(pg. 
379
-
390
)
3
Hacein-Bey-Abina
 
S
von Kalle
 
C
Schmidt
 
M
, et al. 
A serious adverse event after successful gene therapy for X-linked severe combined immunodeficiency.
N Engl J Med
2003
, vol. 
348
 
3
(pg. 
255
-
256
)
4
Nienhuis
 
AW
Dunbar
 
CE
Sorrentino
 
BP
Genotoxicity of retroviral integration in hematopoietic cells.
Mol Ther
2006
, vol. 
13
 
6
(pg. 
1031
-
1049
)
5
Kustikova
 
O
Brugman
 
M
Baum
 
C
The genomic risk of somatic gene therapy.
Semin Cancer Biol
2010
, vol. 
20
 
4
(pg. 
269
-
278
)
6
Barese
 
CN
Dunbar
 
CE
Contributions of gene marking to cell and gene therapies.
Hum Gene Ther
2011
, vol. 
22
 
6
(pg. 
659
-
668
)
7
Miller
 
AD
Rosman
 
GJ
Improved retroviral vectors for gene transfer and expression.
Biotechniques
1989
, vol. 
7
 
9
(pg. 
980
-
990
)
8
Riviere
 
I
Brose
 
K
Mulligan
 
RC
Effects of retroviral vector design on expression of human adenosine deaminase in murine bone marrow transplant recipients engrafted with genetically modified cells.
Proc Natl Acad Sci U S A
1995
, vol. 
92
 
15
(pg. 
6733
-
6737
)
9
Hildinger
 
M
Eckert
 
HG
Schilz
 
AJ
John
 
J
Ostertag
 
W
Baum
 
C
FMEV vectors: both retroviral long terminal repeat and leader are important for high expression in transduced hematopoietic cells.
Gene Ther
1998
, vol. 
5
 
11
(pg. 
1575
-
1579
)
10
Aiuti
 
A
Cassani
 
B
Andolfi
 
G
, et al. 
Multilineage hematopoietic reconstitution without clonal selection in ADA-SCID patients treated with stem cell gene therapy.
J Clin Invest
2007
, vol. 
117
 
8
(pg. 
2233
-
2240
)
11
Schwarzwaelder
 
K
Howe
 
SJ
Schmidt
 
M
, et al. 
Gammaretrovirus-mediated correction of SCID-X1 is associated with skewed vector integration site distribution in vivo.
J Clin Invest
2007
, vol. 
117
 
8
(pg. 
2241
-
2249
)
12
Boztug
 
K
Schmidt
 
M
Schwarzer
 
A
, et al. 
Stem-cell gene therapy for the Wiskott-Aldrich syndrome.
N Engl J Med
2010
, vol. 
363
 
20
(pg. 
1918
-
1927
)
13
Gaspar
 
HB
Cooray
 
S
Gilmour
 
KC
, et al. 
Hematopoietic stem cell gene therapy for adenosine deaminase-deficient severe combined immunodeficiency leads to long-term immunological recovery and metabolic correction.
Sci Transl Med
2011
, vol. 
3
 
97
pg. 
97ra80
 
14
Gaspar
 
HB
Cooray
 
S
Gilmour
 
KC
, et al. 
Long-term persistence of a polyclonal T cell repertoire after gene therapy for X-linked severe combined immunodeficiency.
Sci Transl Med
2011
, vol. 
3
 
97
pg. 
97ra79
 
15
Kohn
 
DB
Candotti
 
F
Gene therapy fulfilling its promise.
N Engl J Med
2009
, vol. 
360
 
5
(pg. 
518
-
521
)
16
Ott
 
MG
Schmidt
 
M
Schwarzwaelder
 
K
, 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
, vol. 
12
 
4
(pg. 
401
-
409
)
17
Cartier
 
N
Hacein-Bey-Abina
 
S
Bartholomae
 
CC
, et al. 
Hematopoietic stem cell gene therapy with a lentiviral vector in X-linked adrenoleukodystrophy.
Science
2009
, vol. 
326
 
5954
(pg. 
818
-
823
)
18
Aubourg
 
P
Blanche
 
S
Jambaque
 
I
, et al. 
Reversal of early neurologic and neuroradiologic manifestations of X-linked adrenoleukodystrophy by bone marrow transplantation.
N Engl J Med
1990
, vol. 
322
 
26
(pg. 
1860
-
1866
)
19
Muller
 
LU
Williams
 
DA
Finding the needle in the hay stack: hematopoietic stem cells in Fanconi anemia.
Mutat Res
2009
, vol. 
668
 
1
(pg. 
141
-
149
)
20
Rosenbaum
 
C
Peace
 
D
Rich
 
E
Van Besien
 
K
Granulocyte colony-stimulating factor-based stem cell mobilization in patients with sickle cell disease.
Biol Blood Marrow Transplant
2008
, vol. 
14
 
6
(pg. 
719
-
723
)
21
Gothot
 
A
van der Loo
 
JC
Clapp
 
DW
Srour
 
EF
Cell cycle-related changes in repopulating capacity of human mobilized peripheral blood CD34(+) cells in non-obese diabetic/severe combined immune-deficient mice.
Blood
1998
, vol. 
92
 
8
(pg. 
2641
-
2649
)
22
Takatoku
 
M
Sellers
 
S
Agricola
 
BA
, et al. 
Avoidance of stimulation improves engraftment of cultured and retrovirally transduced hematopoietic cells in primates.
J Clin Invest
2001
, vol. 
108
 
3
(pg. 
447
-
455
)
23
May
 
C
Rivella
 
S
Callegari
 
J
, et al. 
Therapeutic haemoglobin synthesis in beta-thalassaemic mice expressing lentivirus-encoded human beta-globin.
Nature
2000
, vol. 
406
 
6791
(pg. 
82
-
86
)
24
Cavazzana-Calvo
 
M
Payen
 
E
Negre
 
O
, et al. 
Transfusion independence and HMGA2 activation after gene therapy of human beta-thalassaemia.
Nature
2010
, vol. 
467
 
7313
(pg. 
318
-
322
)
25
Persons
 
DA
The challenge of obtaining therapeutic levels of genetically modified hematopoietic stem cells in beta-thalassemia patients.
Ann N Y Acad Sci
2010
, vol. 
1202
 (pg. 
69
-
74
)
26
Hanawa
 
H
Yamamoto
 
M
Zhao
 
H
Shimada
 
T
Persons
 
DA
Optimized lentiviral vector design improves titer and transgene expression of vectors containing the chicken beta-globin locus HS4 insulator element.
Mol Ther
2009
, vol. 
17
 
4
(pg. 
667
-
674
)
27
Urbinati
 
F
Arumugam
 
P
Higashimoto
 
T
, et al. 
Mechanism of reduction in titers from lentivirus vectors carrying large inserts in the 3′LTR.
Mol Ther
2009
, vol. 
17
 
9
(pg. 
1527
-
1536
)
28
Rivella
 
S
Sadelain
 
M
Genetic treatment of severe hemoglobinopathies: the combat against transgene variegation and transgene silencing.
Semin Hematol
1998
, vol. 
35
 
2
(pg. 
112
-
125
)
29
Bestor
 
TH
Gene silencing as a threat to the success of gene therapy.
J Clin Invest
2000
, vol. 
105
 
4
(pg. 
409
-
411
)
30
Ellis
 
J
Silencing and variegation of gammaretrovirus and lentivirus vectors.
Hum Gene Ther
2005
, vol. 
16
 
11
(pg. 
1241
-
1246
)
31
Heim
 
DA
Dunbar
 
CE
Hematopoietic stem cell gene therapy: towards clinically significant gene transfer efficiency.
Immunol Rev
2000
, vol. 
178
 (pg. 
29
-
38
)
32
Stein
 
S
Ott
 
MG
Schultze-Strasser
 
S
, et al. 
Genomic instability and myelodysplasia with monosomy 7 consequent to EVI1 activation after gene therapy for chronic granulomatous disease.
Nat Med
2010
, vol. 
16
 
2
(pg. 
198
-
204
)
33
Kim
 
YJ
Kim
 
YS
Larochelle
 
A
, et al. 
Sustained high-level polyclonal hematopoietic marking and transgene expression 4 years after autologous transplantation of rhesus macaques with SIV lentiviral vector-transduced CD34+ cells.
Blood
2009
, vol. 
113
 
22
(pg. 
5434
-
5443
)
34
Beard
 
BC
Mezquita
 
P
Morris
 
JC
Kiem
 
HP
Efficient transduction and engraftment of G-CSF-mobilized peripheral blood CD34+ cells in nonhuman primates using GALV-pseudotyped gammaretroviral vectors.
Mol Ther
2006
, vol. 
14
 
2
(pg. 
212
-
217
)
35
Brenner
 
MK
Rill
 
DR
Holladay
 
MS
, et al. 
Gene marking to determine whether autologous marrow infusion restores long-term haemopoiesis in cancer patients.
Lancet
1993
, vol. 
342
 
8880
(pg. 
1134
-
1137
)
36
Mitchell
 
RS
Beitzel
 
BF
Schroder
 
AR
, et al. 
Retroviral DNA integration: ASLV, HIV, and MLV show distinct target site preferences.
PLoS Biol
2004
, vol. 
2
 
8
pg. 
E234
 
37
Wu
 
X
Li
 
Y
Crise
 
B
Burgess
 
SM
Transcription start regions in the human genome are favored targets for MLV integration.
Science
2003
, vol. 
300
 
5626
(pg. 
1749
-
1751
)
38
Schroder
 
AR
Shinn
 
P
Chen
 
H
Berry
 
C
Ecker
 
JR
Bushman
 
F
HIV-1 integration in the human genome favors active genes and local hotspots.
Cell
2002
, vol. 
110
 
4
(pg. 
521
-
529
)
39
Kohn
 
DB
Update on gene therapy for immunodeficiencies.
Clin Immunol
2010
, vol. 
135
 
2
(pg. 
247
-
254
)
40
Qasim
 
W
Gaspar
 
HB
Thrasher
 
AJ
Progress and prospects: gene therapy for inherited immunodeficiencies.
Gene Ther
2009
, vol. 
16
 
11
(pg. 
1285
-
1291
)
41
Howe
 
SJ
Mansour
 
MR
Schwarzwaelder
 
K
, et al. 
Insertional mutagenesis combined with acquired somatic mutations causes leukemogenesis following gene therapy of SCID-X1 patients.
J Clin Invest
2008
, vol. 
118
 
9
(pg. 
3143
-
3150
)
42
Hacein-Bey-Abina
 
S
Garrigue
 
A
Wang
 
GP
, et al. 
Insertional oncogenesis in 4 patients after retrovirus-mediated gene therapy of SCID-X1.
J Clin Invest
2008
, vol. 
118
 
9
(pg. 
3132
-
3142
)
43
Ryu
 
BY
Evans-Galea
 
MV
Gray
 
JT
Bodine
 
DM
Persons
 
DA
Nienhuis
 
AW
An experimental system for the evaluation of retroviral vector design to diminish the risk for proto-oncogene activation.
Blood
2008
, vol. 
111
 
4
(pg. 
1866
-
1875
)
44
McCormack
 
MP
Young
 
LF
Vasudevan
 
S
, et al. 
The Lmo2 oncogene initiates leukemia in mice by inducing thymocyte self-renewal.
Science
2010
, vol. 
327
 
5967
(pg. 
879
-
883
)
45
Schmidt
 
M
Carbonaro
 
DA
Speckmann
 
C
, et al. 
Clonality analysis after retroviral-mediated gene transfer to CD34+ cells from the cord blood of ADA-deficient SCID neonates.
Nat Med
2003
, vol. 
9
 
4
(pg. 
463
-
468
)
46
Kohn
 
DB
Sadelain
 
M
Glorioso
 
JC
Occurrence of leukaemia following gene therapy of X-linked SCID.
Nat Rev Cancer
2003
, vol. 
3
 
7
(pg. 
477
-
488
)
47
Seggewiss
 
R
Pittaluga
 
S
Adler
 
RL
, et al. 
Acute myeloid leukemia is associated with retroviral gene transfer to hematopoietic progenitor cells in a rhesus macaque.
Blood
2006
, vol. 
107
 
10
(pg. 
3865
-
3867
)
48
Calmels
 
B
Ferguson
 
C
Laukkanen
 
MO
, et al. 
Recurrent retroviral vector integration at the Mds1/Evi1 locus in nonhuman primate hematopoietic cells.
Blood
2005
, vol. 
106
 
7
(pg. 
2530
-
2533
)
49
Modlich
 
U
Kustikova
 
OS
Schmidt
 
M
, et al. 
Leukemias following retroviral transfer of multidrug resistance 1 (MDR1) are driven by combinatorial insertional mutagenesis.
Blood
2005
, vol. 
105
 
11
(pg. 
4235
-
4246
)
50
Kustikova
 
O
Fehse
 
B
Modlich
 
U
, et al. 
Clonal dominance of hematopoietic stem cells triggered by retroviral gene marking.
Science
2005
, vol. 
308
 
5725
(pg. 
1171
-
1174
)
51
Yu
 
SF
von Ruden
 
T
Kantoff
 
PW
, et al. 
Self-inactivating retroviral vectors designed for transfer of whole genes into mammalian cells.
Proc Natl Acad Sci U S A
1986
, vol. 
83
 
10
(pg. 
3194
-
3198
)
52
Naldini
 
L
Blomer
 
U
Gallay
 
P
, et al. 
In vivo gene delivery and stable transduction of nondividing cells by a lentiviral vector.
Science
1996
, vol. 
272
 
5259
(pg. 
263
-
267
)
53
Hirata
 
RK
Miller
 
AD
Andrews
 
RG
Russell
 
DW
Transduction of hematopoietic cells by foamy virus vectors.
Blood
1996
, vol. 
88
 
9
(pg. 
3654
-
3661
)
54
Beard
 
BC
Dickerson
 
D
Beebe
 
K
, et al. 
Comparison of HIV-derived lentiviral and MLV-based gammaretroviral vector integration sites in primate repopulating cells.
Mol Ther
2007
, vol. 
15
 
7
(pg. 
1356
-
1365
)
55
De Palma
 
M
Montini
 
E
Santoni de Sio
 
FR
, et al. 
Promoter trapping reveals significant differences in integration site selection between MLV and HIV vectors in primary hematopoietic cells.
Blood
2005
, vol. 
105
 
6
(pg. 
2307
-
2315
)
56
Hematti
 
P
Hong
 
BK
Ferguson
 
C
, et al. 
Distinct genomic integration of MLV and SIV vectors in primate hematopoietic stem and progenitor cells.
PLoS Biol
2004
, vol. 
2
 
12
pg. 
e423
 
57
Biffi
 
A
Bartolomae
 
CC
Cesana
 
D
, et al. 
Lentiviral vector common integration sites in preclinical models and a clinical trial reflect a benign integration bias and not oncogenic selection.
Blood
2011
, vol. 
117
 
20
(pg. 
5332
-
5339
)
58
Deichmann
 
A
Hacein-Bey-Abina
 
S
Schmidt
 
M
, et al. 
Vector integration is nonrandom and clustered and influences the fate of lymphopoiesis in SCID-X1 gene therapy.
J Clin Invest
2007
, vol. 
117
 
8
(pg. 
2225
-
2232
)
59
Deichmann
 
A
Brugman
 
MH
Bartholomae
 
CC
, et al. 
Insertion sites in engrafted cells cluster within a limited repertoire of genomic areas after gammaretroviral vector gene therapy.
Mol Ther
2011
, vol. 
19
 
11
(pg. 
2031
-
2039
)
60
Ciuffi
 
A
Mechanisms governing lentivirus integration site selection.
Curr Gene Ther
2008
, vol. 
8
 
6
(pg. 
419
-
429
)
61
Bushman
 
F
Lewinski
 
M
Ciuffi
 
A
, et al. 
Genome-wide analysis of retroviral DNA integration.
Nat Rev Microbiol
2005
, vol. 
3
 
11
(pg. 
848
-
858
)
62
Lu
 
R
Neff
 
NF
Quake
 
SR
Weissman
 
IL
Tracking single hematopoietic stem cells in vivo using high-throughput sequencing in conjunction with viral genetic barcoding.
Nat Biotechnol
2011
, vol. 
29
 
10
(pg. 
928
-
933
)
63
Brady
 
T
Roth
 
SL
Malani
 
N
, et al. 
A method to sequence and quantify DNA integration for monitoring outcome in gene therapy.
Nucleic Acids Res
2011
, vol. 
39
 
11
pg. 
e72
 
64
Chang
 
AH
Sadelain
 
M
The genetic engineering of hematopoietic stem cells: the rise of lentiviral vectors, the conundrum of the ltr, and the promise of lineage-restricted vectors.
Mol Ther
2007
, vol. 
15
 
3
(pg. 
445
-
456
)
65
Modlich
 
U
Navarro
 
S
Zychlinski
 
D
, et al. 
Insertional transformation of hematopoietic cells by self-inactivating lentiviral and gammaretroviral vectors.
Mol Ther
2009
, vol. 
17
 
11
(pg. 
1919
-
1928
)
66
Anderson
 
J
Li
 
MJ
Palmer
 
B
, et al. 
Safety and efficacy of a lentiviral vector containing three anti-HIV genes–CCR5 ribozyme, tat-rev siRNA, and TAR decoy–in SCID-hu mouse-derived T cells.
Mol Ther
2007
, vol. 
15
 
6
(pg. 
1182
-
1188
)
67
Li
 
MJ
Kim
 
J
Li
 
S
, et al. 
Long-term inhibition of HIV-1 infection in primary hematopoietic cells by lentiviral vector delivery of a triple combination of anti-HIV shRNA, anti-CCR5 ribozyme, and a nucleolar-localizing TAR decoy.
Mol Ther
2005
, vol. 
12
 
5
(pg. 
900
-
909
)
68
DiGiusto
 
DL
Krishnan
 
A
Li
 
L
, et al. 
RNA-based gene therapy for HIV with lentiviral vector-modified CD34(+) cells in patients undergoing transplantation for AIDS-related lymphoma.
Sci Transl Med
2010
, vol. 
2
 
36
pg. 
36ra43
 
69
Sadelain
 
M
Boulad
 
F
Lisowki
 
L
Moi
 
P
Riviere
 
I
Stem cell engineering for the treatment of severe hemoglobinopathies.
Curr Mol Med
2008
, vol. 
8
 
7
(pg. 
690
-
697
)
70
Ikeda
 
K
Mason
 
PJ
Bessler
 
M
3′UTR-truncated Hmga2 cDNA causes MPN-like hematopoiesis by conferring a clonal growth advantage at the level of HSC in mice.
Blood
2011
, vol. 
117
 
22
(pg. 
5860
-
5869
)
71
Giles
 
KE
Gowher
 
H
Ghirlando
 
R
Jin
 
C
Felsenfeld
 
G
Chromatin boundaries, insulators, and long-range interactions in the nucleus.
Cold Spring Harb Symp Quant Biol
2010
, vol. 
75
 (pg. 
79
-
85
)
72
Emery
 
DW
The use of chromatin insulators to improve the expression and safety of integrating gene transfer vectors.
Hum Gene Ther
2011
, vol. 
22
 
6
(pg. 
761
-
774
)
73
Li
 
CL
Xiong
 
D
Stamatoyannopoulos
 
G
Emery
 
DW
Genomic and functional assays demonstrate reduced gammaretroviral vector genotoxicity associated with use of the cHS4 chromatin insulator.
Mol Ther
2009
, vol. 
17
 
4
(pg. 
716
-
724
)
74
Desprat
 
R
Bouhassira
 
EE
Gene specificity of suppression of transgene-mediated insertional transcriptional activation by the chicken HS4 insulator.
PLoS One
2009
, vol. 
4
 
6
pg. 
e5956
 
75
Brown
 
BD
Naldini
 
L
Exploiting and antagonizing micro-RNA regulation for therapeutic and experimental applications.
Nat Rev Genet
2009
, vol. 
10
 
8
(pg. 
578
-
585
)
76
Erlwein
 
O
McClure
 
MO
Progress and prospects: foamy virus vectors enter a new age.
Gene Ther
2010
, vol. 
17
 
12
(pg. 
1423
-
1429
)
77
Bauer
 
TR
Allen
 
JM
Hai
 
M
, et al. 
Successful treatment of canine leukocyte adhesion deficiency by foamy virus vectors.
Nat Med
2008
, vol. 
14
 
1
(pg. 
93
-
97
)
78
Hu
 
J
Renaud
 
G
Gomes
 
TJ
, et al. 
Reduced genotoxicity of avian sarcoma leukosis virus vectors in rhesus long-term repopulating cells compared to standard murine retrovirus vectors.
Mol Ther
2008
, vol. 
16
 
9
(pg. 
1617
-
1623
)
79
Preuss
 
E
Treschow
 
A
Newrzela
 
S
, et al. 
TK. 007: A novel, codon-optimized HSVtk(A168H) mutant for suicide gene therapy.
Hum Gene Ther
2010
, vol. 
21
 
8
(pg. 
929
-
941
)
80
Straathof
 
KC
Pule
 
MA
Yotnda
 
P
, et al. 
An inducible caspase 9 safety switch for T-cell therapy.
Blood
2005
, vol. 
105
 
11
(pg. 
4247
-
4254
)
81
Di Stasi
 
A
Tey
 
S-K
Dotti
 
G
, et al. 
Inducible apoptosis as a safety switch for adoptive cell therapy.
N Engl J Med
2011
, vol. 
365
 
18
(pg. 
1673
-
1683
)
82
Modlich
 
U
Bohne
 
J
Schmidt
 
M
, et al. 
Cell-culture assays reveal the importance of retroviral vector design for insertional genotoxicity.
Blood
2006
, vol. 
108
 
8
(pg. 
2545
-
2553
)
83
Du
 
Y
Jenkins
 
NA
Copeland
 
NG
Insertional mutagenesis identifies genes that promote the immortalization of primary bone marrow progenitor cells.
Blood
2005
, vol. 
106
 
12
(pg. 
3932
-
3939
)
84
Zhang
 
XB
Beard
 
BC
Trobridge
 
GD
, et al. 
High incidence of leukemia in large animals after stem cell gene therapy with a HOXB4-expressing retroviral vector.
J Clin Invest
2008
, vol. 
118
 
4
(pg. 
1502
-
1510
)
85
Montini
 
E
Cesana
 
D
Schmidt
 
M
, 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
, vol. 
119
 
4
(pg. 
964
-
975
)
86
Porteus
 
MH
Carroll
 
D
Gene targeting using zinc finger nucleases.
Nat Biotechnol
2005
, vol. 
23
 
8
(pg. 
967
-
973
)
87
Suzuki
 
K
Mitsui
 
K
Aizawa
 
E
, et al. 
Highly efficient transient gene expression and gene targeting in primate embryonic stem cells with helper-dependent adenoviral vectors.
Proc Natl Acad Sci U S A
2008
, vol. 
105
 
37
(pg. 
13781
-
13786
)
88
Khan
 
IF
Hirata
 
RK
Wang
 
PR
, et al. 
Engineering of human pluripotent stem cells by AAV-mediated gene targeting.
Mol Ther
2010
, vol. 
18
 
6
(pg. 
1192
-
1199
)
89
Li
 
H
Haurigot
 
V
Doyon
 
Y
, et al. 
In vivo genome editing restores haemostasis in a mouse model of haemophilia.
Nature
2011
, vol. 
475
 
7355
(pg. 
217
-
221
)
90
Knauert
 
MP
Glazer
 
PM
Triplex forming oligonucleotides: sequence-specific tools for gene targeting.
Hum Mol Genet
2001
, vol. 
10
 
20
(pg. 
2243
-
2251
)
91
McNeer
 
NA
Chin
 
JY
Schleifman
 
EB
Fields
 
RJ
Glazer
 
PM
Saltzman
 
WM
Nanoparticles deliver triplex-forming PNAs for site-specific genomic recombination in CD34+ human hematopoietic progenitors.
Mol Ther
2011
, vol. 
19
 
1
(pg. 
172
-
180
)
92
Jasin
 
M
Genetic manipulation of genomes with rare-cutting endonucleases.
Trends Genet
1996
, vol. 
12
 
6
(pg. 
224
-
228
)
93
Paques
 
F
Duchateau
 
P
Meganucleases and DNA double-strand break-induced recombination: perspectives for gene therapy.
Curr Gene Ther
2007
, vol. 
7
 
1
(pg. 
49
-
66
)
94
Boch
 
J
TALEs of genome targeting.
Nat Biotechnol
2011
, vol. 
29
 
2
(pg. 
135
-
136
)
95
Holt
 
N
Wang
 
J
Kim
 
K
, et al. 
Human hematopoietic stem/progenitor cells modified by zinc finger nucleases targeted to CCR5 control HIV-1 in vivo.
Nat Biotechnol
2010
, vol. 
28
 
8
(pg. 
839
-
847
)
96
Radecke
 
S
Radecke
 
F
Cathomen
 
T
Schwarz
 
K
Zinc finger nuclease-induced gene repair with oligodeoxynucleotides: wanted and unwanted target locus modifications.
Mol Ther
2010
, vol. 
18
 
4
(pg. 
743
-
753
)
97
Gabriel
 
R
Lombardo
 
A
Arens
 
A
, et al. 
An unbiased genome-wide analysis of zinc finger nuclease specificity.
Nat Biotechnol
2011
, vol. 
29
 
9
(pg. 
816
-
823
)
98
Pattanayak
 
V
Ramirez
 
CL
Joung
 
JK
Liu
 
DR
Revealing off-target cleavage specificities of zinc finger nucleases by in vitro selection.
Nat Methods
2011
, vol. 
8
 
9
(pg. 
765
-
770
)
99
Lombardo
 
A
Genovese
 
P
Beausejour
 
CM
, et al. 
Gene editing in human stem cells using zinc finger nucleases and integrase-defective lentiviral vector delivery.
Nat Biotechnol
2007
, vol. 
25
 
11
(pg. 
1298
-
1306
)
100
Takahashi
 
K
Yamanaka
 
S
Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors.
Cell
2006
, vol. 
126
 
4
(pg. 
663
-
676
)
101
Hanna
 
J
Wernig
 
M
Markoulaki
 
S
, et al. 
Treatment of sickle cell anemia mouse model with iPS cells generated from autologous skin.
Science
2007
, vol. 
318
 
5858
(pg. 
1920
-
1923
)
102
Yamanaka
 
S
Strategies and new developments in the generation of patient-specific pluripotent stem cells.
Cell Stem Cell
2007
, vol. 
1
 
1
(pg. 
39
-
49
)
103
Warren
 
L
Manos
 
PD
Ahfeldt
 
T
, et al. 
Highly efficient reprogramming to pluripotency and directed differentiation of human cells with synthetic modified mRNA.
Cell Stem Cell
2010
, vol. 
7
 
5
(pg. 
618
-
630
)
104
Kane
 
NM
Nowrouzi
 
A
Mukherjee
 
S
, et al. 
Lentivirus-mediated reprogramming of somatic cells in the absence of transgenic transcription factors.
Mol Ther
2010
, vol. 
18
 
12
(pg. 
2139
-
2145
)
105
Zou
 
J
Maeder
 
ML
Mali
 
P
, et al. 
Gene targeting of a disease-related gene in human induced pluripotent stem and embryonic stem cells.
Cell Stem Cell
2009
, vol. 
5
 
1
(pg. 
97
-
110
)
106
Song
 
H
Chung
 
SK
Xu
 
Y
Modeling disease in human ESCs using an efficient BAC-based homologous recombination system.
Cell Stem Cell
2010
, vol. 
6
 
1
(pg. 
80
-
89
)
107
Papapetrou
 
EP
Lee
 
G
Malani
 
N
, et al. 
Genomic safe harbors permit high beta-globin transgene expression in thalassemia induced pluripotent stem cells.
Nat Biotechnol
2011
, vol. 
29
 
1
(pg. 
73
-
78
)
108
Sadelain
 
M
Papapetrov
 
EP
Bushman
 
FD
Safe harbours for the integration of new DNA in the human genome [published online ahead of print December 1, 2011].
Nat Rev Cancer
 
109
Gore
 
A
Li
 
Z
Fung
 
HL
, et al. 
Somatic coding mutations in human induced pluripotent stem cells.
Nature
2011
, vol. 
471
 
7336
(pg. 
63
-
67
)
110
Hussein
 
SM
Batada
 
NN
Vuoristo
 
S
, et al. 
Copy number variation and selection during reprogramming to pluripotency.
Nature
2011
, vol. 
471
 
7336
(pg. 
58
-
62
)
111
Laurent
 
LC
Ulitsky
 
I
Slavin
 
I
, et al. 
Dynamic changes in the copy number of pluripotency and cell proliferation genes in human ESCs and iPSCs during reprogramming and time in culture.
Cell Stem Cell
2011
, vol. 
8
 
1
(pg. 
106
-
118
)
112
Hong
 
H
Takahashi
 
K
Ichisaka
 
T
, et al. 
Suppression of induced pluripotent stem cell generation by the p53-p21 pathway.
Nature
2009
, vol. 
460
 
7259
(pg. 
1132
-
1135
)
113
Marion
 
RM
Strati
 
K
Li
 
H
, et al. 
A p53-mediated DNA damage response limits reprogramming to ensure iPS cell genomic integrity.
Nature
2009
, vol. 
460
 
7259
(pg. 
1149
-
1153
)
114
Li
 
H
Collado
 
M
Villasante
 
A
, et al. 
The Ink4/Arf locus is a barrier for iPS cell reprogramming.
Nature
2009
, vol. 
460
 
7259
(pg. 
1136
-
1139
)
115
Draper
 
JS
Moore
 
HD
Ruban
 
LN
Gokhale
 
PJ
Andrews
 
PW
Culture and characterization of human embryonic stem cells.
Stem Cells Dev
2004
, vol. 
13
 
4
(pg. 
325
-
336
)
116
Baker
 
DE
Harrison
 
NJ
Maltby
 
E
, et al. 
Adaptation to culture of human embryonic stem cells and oncogenesis in vivo.
Nat Biotechnol
2007
, vol. 
25
 
2
(pg. 
207
-
215
)
117
Mayshar
 
Y
Ben-David
 
U
Lavon
 
N
, et al. 
Identification and classification of chromosomal aberrations in human induced pluripotent stem cells.
Cell Stem Cell
2010
, vol. 
7
 
4
(pg. 
521
-
531
)
118
Howden
 
SE
Gore
 
A
Li
 
Z
, et al. 
Genetic correction and analysis of induced pluripotent stem cells from a patient with gyrate atrophy.
Proc Natl Acad Sci U S A
2011
, vol. 
108
 
16
(pg. 
6537
-
6542
)
119
Zhao
 
T
Zhang
 
ZN
Rong
 
Z
Xu
 
Y
Immunogenicity of induced pluripotent stem cells.
Nature
2011
, vol. 
474
 
7350
(pg. 
212
-
215
)
120
Orkin
 
SH
Zon
 
LI
Hematopoiesis: an evolving paradigm for stem cell biology.
Cell
2008
, vol. 
132
 
4
(pg. 
631
-
644
)
121
Robert-Moreno
 
A
Guiu
 
J
Ruiz-Herguido
 
C
, et al. 
Impaired embryonic haematopoiesis yet normal arterial development in the absence of the Notch ligand Jagged1.
EMBO J
2008
, vol. 
27
 
13
(pg. 
1886
-
1895
)
122
Eilken
 
HM
Nishikawa
 
S
Schroeder
 
T
Continuous single-cell imaging of blood generation from haemogenic endothelium.
Nature
2009
, vol. 
457
 
7231
(pg. 
896
-
900
)
123
Zovein
 
AC
Hofmann
 
JJ
Lynch
 
M
, et al. 
Fate tracing reveals the endothelial origin of hematopoietic stem cells.
Cell Stem Cell
2008
, vol. 
3
 
6
(pg. 
625
-
636
)
124
Rhodes
 
KE
Gekas
 
C
Wang
 
Y
, et al. 
The emergence of hematopoietic stem cells is initiated in the placental vasculature in the absence of circulation.
Cell Stem Cell
2008
, vol. 
2
 
3
(pg. 
252
-
263
)
125
Kissa
 
K
Herbomel
 
P
Blood stem cells emerge from aortic endothelium by a novel type of cell transition.
Nature
2010
, vol. 
464
 
7285
(pg. 
112
-
115
)
126
Boisset
 
JC
van Cappellen
 
W
Andrieu-Soler
 
C
Galjart
 
N
Dzierzak
 
E
Robin
 
C
In vivo imaging of haematopoietic cells emerging from the mouse aortic endothelium.
Nature
2010
, vol. 
464
 
7285
(pg. 
116
-
120
)
127
Wang
 
L
Menendez
 
P
Shojaei
 
F
, et al. 
Generation of hematopoietic repopulating cells from human embryonic stem cells independent of ectopic HOXB4 expression.
J Exp Med
2005
, vol. 
201
 
10
(pg. 
1603
-
1614
)
128
Raya
 
A
Rodriguez-Piza
 
I
Guenechea
 
G
, et al. 
Disease-corrected haematopoietic progenitors from Fanconi anaemia induced pluripotent stem cells.
Nature
2009
, vol. 
460
 
7251
(pg. 
53
-
59
)
129
Tian
 
X
Hexum
 
MK
Penchev
 
VR
Taylor
 
RJ
Shultz
 
LD
Kaufman
 
DS
Bioluminescent imaging demonstrates that transplanted human embryonic stem cell-derived CD34(+) cells preferentially develop into endothelial cells.
Stem Cells
2009
, vol. 
27
 
11
(pg. 
2675
-
2685
)
130
Ledran
 
MH
Krassowska
 
A
Armstrong
 
L
, et al. 
Efficient hematopoietic differentiation of human embryonic stem cells on stromal cells derived from hematopoietic niches.
Cell Stem Cell
2008
, vol. 
3
 
1
(pg. 
85
-
98
)
131
Rideout
 
WM
Hochedlinger
 
K
Kyba
 
M
Daley
 
GQ
Jaenisch
 
R
Correction of a genetic defect by nuclear transplantation and combined cell and gene therapy.
Cell
2002
, vol. 
109
 
1
(pg. 
17
-
27
)
132
Schiedlmeier
 
B
Klump
 
H
Will
 
E
, et al. 
High-level ectopic HOXB4 expression confers a profound in vivo competitive growth advantage on human cord blood CD34+ cells but impairs lymphomyeloid differentiation.
Blood
2003
, vol. 
101
 
5
(pg. 
1759
-
1768
)
133
Zhang
 
XB
Schwartz
 
JL
Humphries
 
RK
Kiem
 
HP
Effects of HOXB4 overexpression on ex vivo expansion and immortalization of hematopoietic cells from different species.
Stem Cells
2007
, vol. 
25
 
8
(pg. 
2074
-
2081
)
134
Szabo
 
E
Rampalli
 
S
Risueno
 
RM
, et al. 
Direct conversion of human fibroblasts to multilineage blood progenitors.
Nature
2010
, vol. 
468
 
7323
(pg. 
521
-
526
)
135
Graf
 
T
Enver
 
T
Forcing cells to change lineages.
Nature
2009
, vol. 
462
 
7273
(pg. 
587
-
594
)
136
Trobridge
 
GD
Miller
 
DG
Jacobs
 
MA
, et al. 
Foamy virus vector integration sites in normal human cells.
Proc Natl Acad Sci U S A
2006
, vol. 
103
 
5
(pg. 
1498
-
1503
)
137
Sadelain
 
M
Eliminating cells gone astray.
N Engl J Med
2011
, vol. 
365
 
18
(pg. 
1735
-
1737
)
138
Hacein-Bey
 
H
Cavazzana-Calvo
 
M
Le Deist
 
F
, et al. 
gamma-c gene transfer into SCID X1 patients' B-cell lines restores normal high-affinity interleukin-2 receptor expression and function.
Blood
1996
, vol. 
87
 
8
(pg. 
3108
-
3116
)
139
Hacein-Bey-Abina
 
S
Von Kalle
 
C
Schmidt
 
M
, et al. 
LMO2-associated clonal T cell proliferation in two patients after gene therapy for SCID-X1.
Science
2003
, vol. 
302
 
5644
(pg. 
415
-
419
)
140
Gaspar
 
HB
Parsley
 
KL
Howe
 
S
, et al. 
Gene therapy of X-linked severe combined immunodeficiency by use of a pseudotyped gammaretroviral vector.
Lancet
2004
, vol. 
364
 
9452
(pg. 
2181
-
2187
)
141
Imren
 
S
Payen
 
E
Westerman
 
KA
, et al. 
Permanent and panerythroid correction of murine beta thalassemia by multiple lentiviral integration in hematopoietic stem cells.
Proc Natl Acad Sci U S A
2002
, vol. 
99
 
22
(pg. 
14380
-
14385
)
Sign in via your Institution