This review addresses the current status of gene therapy for immunodeficiencies, chronic granulomatous disease, suicide gene therapy for graft-versus-host disease, viral infections, malignant hematologic disorders, hemophilia, and the hemoglobin disorders. New developments in vector design have fostered improved expression as well as enhanced safety, particularly of integrating retroviral vectors. Several immunodeficiencies have been treated successfully by stem cell–targeted, retroviral-mediated gene transfer with reconstitution of the immune system following infusion of the transduced cells. In a trial for hemophilia B, long-term expression of human FIX has been observed following adeno-associated viral vector–mediated gene transfer into the liver. This approach should be successful in treating any disorder in which liver production of a specific protein is therapeutic.

The field of gene therapy as applied to blood disorders has continued to advance rapidly since my review was completed in 2008.1  That review provided a historical prospective and described the initial successes in treating severe combined immunodeficiency and the first evidence in humans of the genotoxicity of integrating retroviruses. Further success in clinical trials,2,3  particularly in the immunodeficiencies,4-7  and also the development of a successful approach for hemophilia B8  are among the recent clinical advances.

Much of the effort to develop clinical gene therapy has focused on viral vector systems for gene transfer.9-11  Viruses have naturally evolved to insert genetic information into target cells, and the field of gene therapy attempts to take advantage of this proclivity. The basic strategy is to express the individual viral proteins on independent expression cassettes within producer cells and introduce the vector genome with the packaging signals and transgene either by transfection or transduction. There are a number of vector systems that are in wide use both experimentally and clinically as summarized in Table 1.

Table 1

Comparison of the most commonly used vectors for gene therapy

Characteristicsα-Retroviral vectorγ-Retroviral vectorFoamy virus vectorrAAVLentivirus vector
Tropism Dividing and nondividing cells Dividing cells Dividing and nondividing cells Dividing and nondividing cells Dividing and nondividing cells 
Host genome Neutral Integration near regulatory elements Integration nearly uniform No integration Integration in genes 
Transgene expression Stable Stable Stable Stable in nondividing cells Stable 
Packaging capacity Not defined ∼8 kb ∼9 kb ∼5 kb ∼8 kb 
Advantages Neutral, integration, pattern high-titer production Large packaging capacity; long-term expression Large packaging capacity; long-term expression High production yields; low immunogenicity; long-term expression Large packaging capacity; long-term expression 
Disadvantages None have emerged High risk of insertional mutagenesis Risk of insertional mutagenesis Small packaging capacity Risk of insertional mutagenesis 
Characteristicsα-Retroviral vectorγ-Retroviral vectorFoamy virus vectorrAAVLentivirus vector
Tropism Dividing and nondividing cells Dividing cells Dividing and nondividing cells Dividing and nondividing cells Dividing and nondividing cells 
Host genome Neutral Integration near regulatory elements Integration nearly uniform No integration Integration in genes 
Transgene expression Stable Stable Stable Stable in nondividing cells Stable 
Packaging capacity Not defined ∼8 kb ∼9 kb ∼5 kb ∼8 kb 
Advantages Neutral, integration, pattern high-titer production Large packaging capacity; long-term expression Large packaging capacity; long-term expression High production yields; low immunogenicity; long-term expression Large packaging capacity; long-term expression 
Disadvantages None have emerged High risk of insertional mutagenesis Risk of insertional mutagenesis Small packaging capacity Risk of insertional mutagenesis 

rAAV, adeno-associated viral vector.

Adenoviral vectors

Vectors based on adenovirus have the advantage of having a large transgene capacity allowing large or multiple gene expression cassettes to be included within the vector genome.10,12  Adenoviral vectors have had limited use in the treatment of blood disorders, although they have been quite effective in facilitating the generation of viral-specific T-cell populations as discussed in detail in a later section.

Adeno-associated viral vectors (rAAVs)

rAAVs are less immunogenic than adenoviral vectors and are capable of transducing quiescent cells and establishing a stable episome without vector integration in most cases.11,13  Only the inverted terminal repeats are required as part of the vector genome, and packaging can be achieved with various strategies to develop high-titer vector preparations. Further, the incidence of preexisting immunity is generally low, particularly for certain serotypes such as serotypes 8 and 9. However, rAAV can carry only a transgene up to 4700 nucleotides. The AAV genome is single stranded, and vector preparations are composed of a mixture of vector particles having 1 of the 2 strands of the virus. Upon transduction, the required annealing of the 2 strands delays gene expression. This limitation can be overcome by utilizing a self-complementary design in which the 2 strands of the transgene are on a single hairpin genome in an inverted orientation, which allows quick assembly into a transcription unit following transduction.14,15 

Retroviral vectors

The γ-retroviral vectors initially used in clinical trials had an intact long terminal repeat (LTR). The LTR enhance-promoter combination was associated with genotoxicity. However, subsequently self-inactivating vectors in which the enhancer-promoter are not present in the integrated vector genome have been shown to be clinically efficacious in the treatment of certain immunodeficiencies.16  Lentiviral vectors based on HIV-1 were developed largely in response to the challenge of transducing nonmitotic, hematopoietic stem cells.9  Although cytokine activation to trigger the cells into the G1 phase of the cell cycle is still required, mitosis is not required as the preintegration complex of HIV vectors can cross the nuclear membrane without mitosis.9  Another advantage of the HIV preintegration complex is that it persists in transduced cells for an extended period prior to integration, thereby allowing a longer window of opportunity for integration to occur. Foamy viruses, another type of retrovirus, do not cause disease and have not infected humans.17  The genome of foamy viruses integrates diffusely in cellular DNA rather than within genes or at regulatory elements (Table 1). A foamy viral vector has been used to achieve stem cell–targeted gene transfer and resolution of leukocyte adhesion deficiency in a canine model.18 

The α-retroviral vectors are being considered for therapeutic gene transfer as well.19-21  Such vectors integrate more uniformly within the genome, particularly within intergenic regions compared with γ-retroviral vectors, which are found near transcriptional start sites and cytosine guanine dinucleotide islands, and lentiviral vectors, which tend to integrate within genes.22  This favorable integration pattern has been shown to result in lower genotoxicity compared with lentiviral and γ-retroviral vectors.20  A split-packaging design for self-inactivating α-retroviral vectors has been developed and shown to be capable of generating high-titer vector particles.19  The therapeutic potential for this class of vectors has been shown by functional correction of chronic granulomatous disease (CGD) in a cell line as well as within transduced human cells that engraft in the mouse model.21 

Genotoxicity of integrating vectors

Integrating retroviruses are inherently mutagenic.16,22  Enhancer-mediated protooncogene activation has resulted in leukemia in a number of participants in the trials for immunodeficiencies.4-7,16,22,23  Myelodysplasia has also been observed in participants in a trial for CGD secondary to protooncogene activation.24  Various efforts have been made to improve the safety of integrating retroviral vectors. These include elimination of the enhancer element in the context of a self-inactivating vector design, the use of internal cellular promoters with lower activation potential,25,26  and the addition of insulator elements to the LTRs of the integrated vector.27  Historically, the chicken β-globin locus insulator28  has been used, but more recent studies have identified a number of additional elements that have insulating potential.29-31 

Various assays have been used to evaluate the steps taken to improve vector safety, although, as a noted in a recent review, none of the assays is fully predictive.22  One factor that may affect safety is the relative distribution of vector integrations (Table 1). For example, lentiviral vectors are less likely to integrate into regulatory elements than γ-retroviral vectors.32,33  However, recent studies suggest preferred regions of integration for both types of vectors, so the difference in integration pattern is presumably only relative with respect to safety.32  Also, the methodology for determining vector distribution is still imperfect.34  Alternative mechanisms of oncogenesis include alternative splicing, gene inactivation, truncation of cellular messenger RNA or protein, and micro RNA activation.22  Each of these mechanisms has been observed in animal models. Clonal dominance developed in 1 participant in a gene therapy trial for thalassemia and was associated with interruption of the HMGA2 gene by the vector genome resulting in the generation of a truncated messenger RNA with missing regulatory elements.35 

Generally, bone marrow transplantation is used to treat severe immunodeficiencies. The outcome for individuals with a matched related donor is outstanding (>90%) but much less satisfactory for individuals who have a matched unrelated donor or a partially mismatched, related donor.4-6,36,37  For those patients, gene transfer offers a potentially more satisfactory alternative form of treatment. In early trials, T lymphocytes were transduced, but much of the recent work has focused on transducing bone marrow hematopoietic stem cells expressing the CD34+ phenotype.

X-SCID

X-linked severe combined immunodeficiency (X-SCID) reflects the lack of the common γ-chain that is part of several interleukin (IL) receptors including the IL-7 receptor, which is required for T-cell development. Affected patients have deficient T cells and natural killer cells and poorly functional B cells. As recently reviewed,4  between 1999 and 2006, 20 individuals with X-SCID were treated in 2 gene therapy trials, 1 in Paris38  and 1 in London (Table 2).39  All subjects lacked an HLA-identical donor. Seventeen of the 20 treated participants are alive and display nearly full correction of the T-cell deficiency by genetically modified T cells when evaluated between 5 and 12 years after the gene transfer procedure. However, half of the trial participants remain on immunoglobulin replacement. The natural killer cell deficiency also persisted. Older participants with hypomorphic mutations responded less well to the gene therapy procedure, possibly because of loss of thymic function with advancing age. Unfortunately, 5 of the participants developed T-cell leukemia within 3 to 6 years after the gene transfer procedure. Four were successfully treated with standard antileukemic therapy, and 1 died of refractory leukemia.4,6  Vector integration analysis identified insertions near the LM02 protooncogene in 4 participants. The leukemogenic process was thought to be initiated by vector-mediated protooncogene activation,1  but other mutations must have occurred over time before evolution to full neoplasia. The initial clinical trials closed after the development of leukemia in the 5 participants. Subsequently, a self-inactivating γ-retroviral vector has been developed for treatment of X-SCID,26  and patients are again being enrolled. A self-inactivating lentiviral vector with an insulator element has also been designed and shown directly not to elevate LM02 expression in T cells.27  Two clinical trials utilizing this vector have also been opened and are enrolling participants.

Table 2

Ongoing gene therapy studies for blood disorders

DisorderVector, dose range, and number and ages of patientsTransgene and promoterRoute of administration and cell targetScientific and clinical outcomesReference
Hemophilia B AAV8; 2 × 1011, 6 × 1011, or 2 × 1012 vg/kg body weight; 6 patients (27-64 y old) FIX gene, regulated by the human apolipoprotein hepatic control region and human α-1-antitrypsin promoter IV delivery targeting hepatocytes Durable circulating FIX at 2% to 11% normal levels; decreased frequency (2 of 6 patients) or cessation (8 of 10) of spontaneous hemorrhage 
X-SCID γ-Retrovirus; 10 patients (4-36 mo old); CD34+ cells were infused (without conditioning) at doses of 60 × 106 to 207 × 106 cells per patient IL-2 receptor common γ-chain, retroviral LTR Ex vivo, CD34+ hematopoietic stem and progenitor cells Functional polyclonal T-cell response restored in all patients; 1 patient developed acute T-cell lymphoblastic leukemia 39 
γ-Retrovirus; 9 patients (1-11 mo old); CD34+ cells were infused (without conditioning) at doses of 1 × 106 to 22 × 106 cells per kg IL-2 receptor common γ-chain, retroviral LTR Ex vivo, CD34+ hematopoietic stem and progenitor cells Functional T-cell numbers reached normal ranges. Transduced T cells were detected for up to 10.7 y after gene therapy. Four patients developed acute T-cell lymphoblastic leukemia, and 1 died. 38 
Adenosine deaminase deficiency resulting in severe combined immunodeficiency (ADA-SCID) γ-Retrovirus; 6 patients (6-39 mo old); CD34+ cells were infused (after nonmyeloablative conditioning with melphalan [Alkeran], 140 mg/m2 body surface area, or busulfan [Myleran], 4 mg/kg) at doses of <0.5 × 106 to 5.8 × 106 cells per kg ADA gene, retroviral LTR Ex vivo, CD34+ hematopoietic stem and progenitor cells Restoration of immune function in 4 of 6 patients; 3 of 6 taken off enzyme-replacement therapy; 4 of 6 remain free of infection 40 
γ-Retrovirus; 10 patients (1-5 mo old); CD34+ cells were infused (after nonmyeloablative conditioning with busulfan, 4 mg/kg) at doses of 3.1 × 106 to 13.6 × 106 cells per kg ADA gene, retroviral LTR Ex vivo, CD34+ hematopoietic stem and progenitor cells Nine of 10 patients had immune reconstitution with increases in T-cell counts (median count at 3 y, 1.07 × 109/L) and normalization of T-cell function. Eight of 10 patients do not require enzyme-replacement therapy. 42 
CGD A range of studies, using γ-retrovirus vectors pseudotyped either with gibbon ape leukemia virus envelope or with an amphotrophic envelope; various nonmyeloablative conditioning strategies Gp91phox, retroviral LTR Ex vivo, CD34+ hematopoietic stem and progenitor cells Twelve of 12 patients showed short-term functional correction of neutrophils with resolution of life-threatening infections. Three patients developed myeloproliferative disease. 57 
Wiskott-Aldrich syndrome (WAS) γ-Retrovirus; 10 patients; CD34+ cells were infused (after nonmyeloablative conditioning with busulfan, 4 mg/kg) WAS gene, retroviral LTR Ex vivo, CD34+ hematopoietic stem and progenitor cells Nine of 10 patients showed improvement of immunologic function and platelet count. Four patients developed acute T-cell lymphoblastic leukemia. 7, 23 
β-Thalassemia Self-inactivating HIV-1–derived lentivirus; 1 patient (18 y old) received fully myeloablative conditioning with busulfan; 3.9 × 106 CD34+ cells per kg Mutated adult β-globin (βA(T87Q)) with antisickling properties, LCR control Ex vivo, CD34+ hematopoietic stem and progenitor cells Patient has been transfusion independent for 7 y. Blood hemoglobin is maintained between 9 and 10 g/dL, of which one-third contains vector-encoded β-globin. 35 
Adrenoleukodystrophy (ALD) Self-inactivating HIV-1–derived lentivirus; 2 patients (7 and 7.5 y old) received myeloablative conditioning with cyclophosphamide (Cytoxan) and busulfan; transduced CD34+ cells, 4.6 × 106 and 7.2 × 106 cells per kg, respectively. Two younger patients also treated with short-term follow-up. Wild-type ABCD1 complementary DNA under the control of the MND viral promoter Ex vivo, CD34+ hematopoietic stem and progenitor cells Nine percent to 14% of granulocytes, monocytes, and T and B lymphocytes expressing the ALD protein; beginning 14-16 mo after infusion of the genetically corrected cells, progressive cerebral demyelination in the 2 patients attenuated. 61-63 
Gene therapy for cancer      
 B-cell leukemia and lymphoma Self-inactivating lentivirus expressing a chimeric T-cell receptor; a single patient was conditioned with pentostatin (Nipent; 4 mg/m2) and cyclophosphamide (600 mg/m2) before receiving 1.5 × 105 transduced T cells per kg (total 3 × 108 T cells, of which 5% were transduced) Anti-CD19 scFv derived from FMC63 murine monoclonal antibody, human CD8α hinge and trans-membrane domain, and human 4-1BB and CD3ζ signaling domains Ex vivo, autologous T cells, IV infusion, split over 3 d Transduced T cells expanded more than 1000 times in vivo, with delayed development of the tumor lysis syndrome and complete remission, ongoing 10 mo after treatment. Engineered cells persisted at high levels for 6 mo in the blood and bone marrow. 80 
Murine stem cell virus–based splice-gag (retroviral) vector expressing CD19 CAR; 8 patients (47-63 y old) with progressive B-cell malignancies received cyclophosphamide and fludarabine (Fludara) before CAR-transduced autologous T cells and IL-2. Patients received 0.3 × 107 to 3.0 × 107 CAR+ T cells per kg, of which an average of 55% were transduced. Anti-CD19 scFv derived from the FMC63 mouse hybridoma, a portion of the human CD28 molecule and the intracellular component of the human TCR-ζ molecule Ex vivo, autologous T cells, single IV infusion, followed (3 h) by a course of IL-2 Varied levels of anti-CD19-CAR–transduced T cells could be detected in the blood of all patients. One patient died on trial, with influenza A pneumonia, nonbacterial thrombotic endocarditis, and cerebral infarction. Four patients had prominent elevations in serum levels of interferon γ and tumor necrosis factor, correlating with severity of acute toxicities. Six of the 8 patients treated obtained objective remissions. 81 
 Acute leukemia SFG retrovirus expressing an inducible suicide system for improved safety of stem cell transplantation to prevent GVHD; transduced haploidentical T cells (1 × 106 to 1 × 107 T cells per kg); 5 patients (3-17 y old) FK506-binding protein linked to modified human caspase 9 with truncated CD19 as a selectable marker; in the presence of the drug, the iCasp9 promolecule dimerizes and activates apoptosis; retroviral LTR Ex vivo, allodepleted haploidentical T cells, infused IV into recipients of allogeneic bone marrow transplants The genetically modified T cells were detected in peripheral blood from all 5 patients and increased in number over time. A single dose of dimerizing drug, given to 4 patients in whom GVHD developed, eliminated >90% of the modified T cells within 30 min after administration and ended the GVHD without recurrence. 69 
DisorderVector, dose range, and number and ages of patientsTransgene and promoterRoute of administration and cell targetScientific and clinical outcomesReference
Hemophilia B AAV8; 2 × 1011, 6 × 1011, or 2 × 1012 vg/kg body weight; 6 patients (27-64 y old) FIX gene, regulated by the human apolipoprotein hepatic control region and human α-1-antitrypsin promoter IV delivery targeting hepatocytes Durable circulating FIX at 2% to 11% normal levels; decreased frequency (2 of 6 patients) or cessation (8 of 10) of spontaneous hemorrhage 
X-SCID γ-Retrovirus; 10 patients (4-36 mo old); CD34+ cells were infused (without conditioning) at doses of 60 × 106 to 207 × 106 cells per patient IL-2 receptor common γ-chain, retroviral LTR Ex vivo, CD34+ hematopoietic stem and progenitor cells Functional polyclonal T-cell response restored in all patients; 1 patient developed acute T-cell lymphoblastic leukemia 39 
γ-Retrovirus; 9 patients (1-11 mo old); CD34+ cells were infused (without conditioning) at doses of 1 × 106 to 22 × 106 cells per kg IL-2 receptor common γ-chain, retroviral LTR Ex vivo, CD34+ hematopoietic stem and progenitor cells Functional T-cell numbers reached normal ranges. Transduced T cells were detected for up to 10.7 y after gene therapy. Four patients developed acute T-cell lymphoblastic leukemia, and 1 died. 38 
Adenosine deaminase deficiency resulting in severe combined immunodeficiency (ADA-SCID) γ-Retrovirus; 6 patients (6-39 mo old); CD34+ cells were infused (after nonmyeloablative conditioning with melphalan [Alkeran], 140 mg/m2 body surface area, or busulfan [Myleran], 4 mg/kg) at doses of <0.5 × 106 to 5.8 × 106 cells per kg ADA gene, retroviral LTR Ex vivo, CD34+ hematopoietic stem and progenitor cells Restoration of immune function in 4 of 6 patients; 3 of 6 taken off enzyme-replacement therapy; 4 of 6 remain free of infection 40 
γ-Retrovirus; 10 patients (1-5 mo old); CD34+ cells were infused (after nonmyeloablative conditioning with busulfan, 4 mg/kg) at doses of 3.1 × 106 to 13.6 × 106 cells per kg ADA gene, retroviral LTR Ex vivo, CD34+ hematopoietic stem and progenitor cells Nine of 10 patients had immune reconstitution with increases in T-cell counts (median count at 3 y, 1.07 × 109/L) and normalization of T-cell function. Eight of 10 patients do not require enzyme-replacement therapy. 42 
CGD A range of studies, using γ-retrovirus vectors pseudotyped either with gibbon ape leukemia virus envelope or with an amphotrophic envelope; various nonmyeloablative conditioning strategies Gp91phox, retroviral LTR Ex vivo, CD34+ hematopoietic stem and progenitor cells Twelve of 12 patients showed short-term functional correction of neutrophils with resolution of life-threatening infections. Three patients developed myeloproliferative disease. 57 
Wiskott-Aldrich syndrome (WAS) γ-Retrovirus; 10 patients; CD34+ cells were infused (after nonmyeloablative conditioning with busulfan, 4 mg/kg) WAS gene, retroviral LTR Ex vivo, CD34+ hematopoietic stem and progenitor cells Nine of 10 patients showed improvement of immunologic function and platelet count. Four patients developed acute T-cell lymphoblastic leukemia. 7, 23 
β-Thalassemia Self-inactivating HIV-1–derived lentivirus; 1 patient (18 y old) received fully myeloablative conditioning with busulfan; 3.9 × 106 CD34+ cells per kg Mutated adult β-globin (βA(T87Q)) with antisickling properties, LCR control Ex vivo, CD34+ hematopoietic stem and progenitor cells Patient has been transfusion independent for 7 y. Blood hemoglobin is maintained between 9 and 10 g/dL, of which one-third contains vector-encoded β-globin. 35 
Adrenoleukodystrophy (ALD) Self-inactivating HIV-1–derived lentivirus; 2 patients (7 and 7.5 y old) received myeloablative conditioning with cyclophosphamide (Cytoxan) and busulfan; transduced CD34+ cells, 4.6 × 106 and 7.2 × 106 cells per kg, respectively. Two younger patients also treated with short-term follow-up. Wild-type ABCD1 complementary DNA under the control of the MND viral promoter Ex vivo, CD34+ hematopoietic stem and progenitor cells Nine percent to 14% of granulocytes, monocytes, and T and B lymphocytes expressing the ALD protein; beginning 14-16 mo after infusion of the genetically corrected cells, progressive cerebral demyelination in the 2 patients attenuated. 61-63 
Gene therapy for cancer      
 B-cell leukemia and lymphoma Self-inactivating lentivirus expressing a chimeric T-cell receptor; a single patient was conditioned with pentostatin (Nipent; 4 mg/m2) and cyclophosphamide (600 mg/m2) before receiving 1.5 × 105 transduced T cells per kg (total 3 × 108 T cells, of which 5% were transduced) Anti-CD19 scFv derived from FMC63 murine monoclonal antibody, human CD8α hinge and trans-membrane domain, and human 4-1BB and CD3ζ signaling domains Ex vivo, autologous T cells, IV infusion, split over 3 d Transduced T cells expanded more than 1000 times in vivo, with delayed development of the tumor lysis syndrome and complete remission, ongoing 10 mo after treatment. Engineered cells persisted at high levels for 6 mo in the blood and bone marrow. 80 
Murine stem cell virus–based splice-gag (retroviral) vector expressing CD19 CAR; 8 patients (47-63 y old) with progressive B-cell malignancies received cyclophosphamide and fludarabine (Fludara) before CAR-transduced autologous T cells and IL-2. Patients received 0.3 × 107 to 3.0 × 107 CAR+ T cells per kg, of which an average of 55% were transduced. Anti-CD19 scFv derived from the FMC63 mouse hybridoma, a portion of the human CD28 molecule and the intracellular component of the human TCR-ζ molecule Ex vivo, autologous T cells, single IV infusion, followed (3 h) by a course of IL-2 Varied levels of anti-CD19-CAR–transduced T cells could be detected in the blood of all patients. One patient died on trial, with influenza A pneumonia, nonbacterial thrombotic endocarditis, and cerebral infarction. Four patients had prominent elevations in serum levels of interferon γ and tumor necrosis factor, correlating with severity of acute toxicities. Six of the 8 patients treated obtained objective remissions. 81 
 Acute leukemia SFG retrovirus expressing an inducible suicide system for improved safety of stem cell transplantation to prevent GVHD; transduced haploidentical T cells (1 × 106 to 1 × 107 T cells per kg); 5 patients (3-17 y old) FK506-binding protein linked to modified human caspase 9 with truncated CD19 as a selectable marker; in the presence of the drug, the iCasp9 promolecule dimerizes and activates apoptosis; retroviral LTR Ex vivo, allodepleted haploidentical T cells, infused IV into recipients of allogeneic bone marrow transplants The genetically modified T cells were detected in peripheral blood from all 5 patients and increased in number over time. A single dose of dimerizing drug, given to 4 patients in whom GVHD developed, eliminated >90% of the modified T cells within 30 min after administration and ended the GVHD without recurrence. 69 

Adapted from Seymour and Thrasher.2 

CAR, chimeric antigen receptor; GVHD, graft-vs-host disease; IV, intravenous/intravenously; LCR, locus control region.

ADA-SCID

Deficiency in ADA leads to SCID. Over the years, there have been several attempts to perform gene therapy in such individuals (Table 2). The early efforts were unsuccessful, presumably because of inadequate transduction of target T cells or primitive hematopoietic cells. However, since 2000, 40 patients have been treated in Italy, the United Kingdom, and the United States.40-43  CD34+ cells were transduced with a γ-retroviral vector encoding the ADA gene. Low-intensity conditioning with either busulfan or melphalan was required to allow engraftment of primitive stem cells with subsequent recovery from the immunodeficiency. Integration site analysis demonstrated vector insertions near protooncogenes, including LM02, but none of these patients developed leukemia. Moreover, only minor changes in transcriptional activity were observed in T-cell clones harboring 1 or 2 copies of the vector genome.44 

WAS

WAS is an X-linked disorder characterized by microthrombocytopenia, immunodeficiency, eczema, and a proclivity to develop an autoimmune disorder and/or lymphoma secondary to a mutation in the WAS protein (WASp) gene.5,45  The disease is variably severe depending on the exact nature of the mutation in the WASp gene, but individuals with early onset severe forms have been identified and are the appropriate candidates for attempted gene therapy.46  An initial clinical trial was conducted in Hanover, Germany, using a γ-retroviral vector in which the LTR enhancer/promoter combination drove expression of the WASp gene (Table 2).23  Gene transfer was targeted to bone marrow and/or peripheral blood CD34+ cells ex vivo, and participants received myelosuppression prior to reinfusion of the transduced cells. Although 9 of the 10 participants that were enrolled did well clinically for several years, ultimately, 4 developed leukemia secondary to insertional mutagenesis.4,7  Subsequently, self-inactivating lentiviral vectors were developed by a number of groups.25,47-50  Vectors have been tested in which the WASp coding sequences are under the control of various promoters.49  Full correction of the WAS phenotype was achieved in the murine model with the vector having a retroviral promoter, whereas only partial correction was achieved with the vector having the 1.6-kb WAS promoter fragment.49  We have also found that the γ-retroviral promoter is much stronger than either the EF1α or the WAS 1.6-kb promoter in various model systems.50  Several other studies suggested that a 1.6-kb promoter fragment from the WASp gene achieves adequate levels for correction in the preclinical model as well as normal levels of expression in human CD34+ cells.25,51  Methodology for large-scale manufacture of the lentiviral vector has permitted the initiation of a number of clinical trials.7,52  A recent report describes correction of the immune deficiency in 3 participants in a clinical trial who have now been followed up to 18 months after the gene therapy procedure.53 

CGD

CGD reflects a deficiency in neutrophil function due to mutations in nicotinamide adenine dinucleotide phosphate oxidase that result in chronic bacterial and fungal infections. The clinical characteristics of the disease have recently been reviewed in detail.54  Nicotinamide adenine dinucleotide phosphate is a complex of 5 separate proteins, each of which may be deficient and cause the CGD phenotype, although the most common defect is in gp91phox.44  Early trials done without myeloablation or limited myeloablation resulted in only transient production of genetically modified neutrophils, although a clinical benefit with clearance of a chronic infection was noted in several of the early patients.1,5,54-56  The initial clinical trials used γ-retroviral vectors with an intact LTR, and not surprisingly, protooncogene activation occurred resulting in myelodysplasia in all of the successfully treated participants (Table 2).24,57  In contrast to X-SCID, ADA-SCID, and WAS, the gene-corrected cells in CGD do not have an engraftment advantage, and thus, as treatment efforts evolve, regimens are likely to be more fully myeloablative.57  In an effort to achieve greater safety, lineage- and stage-restricted lentiviral vectors are being developed for treatment of CGD.58,59  Other vector systems are also being considered, although they are in much earlier stages of development.21,60 

Many leukodystrophies are potential candidates for treatment by gene therapy.61,62  To date, 2 have been studied in detail, X-linked ALD (X-ALD) and metachromatic leukodystrophy (MLD). X-ALD is caused by mutations in the ABCD1 gene that encodes a transporter protein. This deficiency leads to the accumulation of very long chain fatty acids in plasma and tissue and progressive demyelination in the central nervous system (CNS). X-ALD has been successfully treated with hematopoietic stem cell transplantation, although cessation of progression of the disorder occurs 12 to 18 months after treatment. To date, 4 patients have received autologous hematopoietic stem cells transduced with the lentiviral vector encoding the transporter (Table 2).63  Arrest of progression of the disorder occurred in the first 2 patients that were treated.61  Over time, their percentage of genetically modified myeloid and lymphoid cells stabilized at ∼10%. The favorable outcome following stem cell transplantation, whether allogeneic or autologous and gene corrected, is thought to rely on the migration of monocytes/macrophages into the CNS and subsequent conversion to microglia that are long-lived and provide the therapeutic benefit. Integration site analysis in these 2 participants of the trial demonstrated the typical lentiviral pattern with no evidence of protooncogene activation.64 

MLD is an inherited autosomal recessive disorder secondary to a deficiency of the lysosomal enzyme arylsulfatase A.61  Massive accumulation of nonmetabolite sulfatides damages both the central and peripheral nervous systems. A mouse knockout model of MLD has been developed and used for the exploration of gene therapy approaches.65  Stem cell–targeted gene transfer followed by autologous transplantation is one approach.66  An alternative that is also being explored in the mouse model is the direct injection of AAV vectors encoding ARSA into the CNS.67  Early correction is essential because the most common form of MLD develops in the second year of life with rapid, progressive CNS dysfunction. Thus, the introduction directly of AAV vectors into the brain seems more preferable than stem cell–targeted gene transfer in that correction of the phenotype with the latter approach requires several months, during which the patients continue to deteriorate.

The basic strategy with this approach is to genetically modify donor T cells ex vivo before infusion for enhancement of engraftment and prevention of viral infections. GVHD may develop in this context so that methodologies have been evolved to introduce a suicide gene into the T lymphocytes before infusion into patients. By the time the T cells are ablated, engraftment has been established and immunity is generally sufficient to protect from infections. A number of genetic systems have been used or explored experimentally.68  Two have been tested in clinical trials, 1 with a vector having the thymidine kinase gene, and posttreatment is with ganciclovir.68  The other system involves caspase-9.69  T cells containing this gene can be abrogated using a dimerizing drug (AP1903). Overall, this approach shows promise for control of GVHD.

Adoptive transfer of T-lymphocyte populations has been used as a strategy for preventing and treating viral infections in immunocompromised individuals.70  Adenoviral-transduced, antigen-presenting cell lines were used to derive the T cell–specific populations to treat or prevent viral infections in stem cell transplant recipients.70,71  More recent studies rely on the generation of cytotoxic T lymphocytes using immunostimulatory cells that have been transduced with peptide mixtures of the viral antigens.72,73  Current efforts to use gene transfer for prevention or treatment of viral infections are focused on AIDS.74-76  The goal in these efforts is to introduce 1 or more genetic elements into autologous hematopoietic stem cells that are then used to reconstitute the hematopoietic system of the AIDS patient to create a population of HIV-resistant T cells. Several studies have focused on the knockdown or knockout of the CCR5 gene as it encodes a potent coreceptor for HIV infection. Inhibition of critical processes such as viral entry or replication reflects an alternative strategy of stem cell modification for HIV therapy.74 

A number of creative approaches have been developed to use gene transfer in the context of treating cancer.1,77  These include generating T-cell populations in vitro with specificity for antigens expressed on tumor cells for reinfusion, the development of tumor vaccines by expressing genes that enhance the immune response to injected tumor cells, and the creation of CARs on T cells in which a single-chain antibody with specificity for an antigen expressed on human tumor cells linked to internal domains that participate in cell activation.77,78  Remission has been achieved in patients with B-lymphoid malignancies who received CAR-modified T cells (Table 2).79-81  Fortunately, no evidence of T-cell transformation has been observed in these patients, although the period of observation is still rather short. Oncolytic adenoviruses with relative tumor cell specificity are being developed for a variety of cancers.82  Species D adenovirus oncolytics have been shown to infect and kill B-cancer cells, but development of genetic variants to enhance activity has not yet been reported, nor have clinical trials begun to date.83  Another strategy to use gene transfer to enhance cancer treatment involves the transfer of a drug-resistance gene into autologous hematopoietic stem cells that are given to patients in the context of myelosuppression. Treatment is then given to amplify the genetically modified cell populations.84  This strategy has been tested in a small clinical trial that demonstrated that intensification of chemotherapy was feasible, and there was an apparent improvement in outcome.85  Overall, gene transfer technology continues to offer many opportunities to improve the treatment of individuals with malignant hematopoietic disorders.

Several different approaches have been considered for using viral vectors to achieve therapy for hemophilia.1,86  Over the years, Amit Nathwani at University College London and Andrew Davidoff at St. Jude Children’s Research Hospital have shown that intravenous injection of rAAV8 particles resulted in long-term hepatic production of human factor IX in the rhesus macaque model. These observations have now been translated into a clinical trial (Table 2).8  To date, 10 participants have been enrolled, and all show evidence of vector-derived FIX production. The length of follow-up ranged from 6.7 months to 3.3 years. The FIX levels varied from 1% to as much as 8% or 9% in the individual patients at different times, with participants that received the highest dose having the higher levels. Seven have stopped routine prophylaxis with recombinant FIX, and the remaining 3 individuals have reduced the frequency of recombinant protein infusion. The only complication in the trial was the development of transient elevation in transaminases in 1 and perhaps in 2 other participants, which was abrogated with a short course of prednisolone.8  Overall, these results are highly encouraging with respect to application of gene therapy for FIX deficiency. Efforts are underway to modify the AAV capsid to reduce its immunogenicity.87  Despite the larger size of the FVIII gene, an AAV vector has been constructed with a modified coding sequence capable of being packaged and transmitting an expressible FXIII gene.88  A clinical trial for hemophilia A is in the early stages of planning.

The hemoglobin disorders, severe β-thalassemia and sickle cell anemia, were identified early as potential targets for therapeutic gene transfer.1  However, despite considerable effort over the years to develop vector systems and to test them in animal models, progress into the clinic has been limited and not fully successful.1,89  We have estimated that it would require 20% of the primitive hematopoietic cells to be genetically modified to achieve a definitive therapeutic benefit.89  Although levels of gene transfer of 7% to 14% were observed in 2 participants in a clinical trial for X-linked ALD,62  comparable levels have not been achieved in individuals with thalassemia. In 1 small trial, 2 patients failed to engraft with the transduced stem cells and had to receive backup bone marrow for hematopoietic rescue (Phillip Leboulch, personal communication). The other participant developed significant amounts of the transgene globin product, but this apparently primarily reflected dominance by a large clone having a retroviral integration into the HMG2A gene.35  This participant remains transfusion independent for a period now up to 7 years.

There has been a long-standing interest in reactivating the fetal γ-globin gene in an effort to compensate for deficient β-globin synthesis in thalassemia or to inhibit sickling in patients with sickle cell anemia.90,91  Efforts in this area were advanced with the discovery that BCL11A is a major regulator of developmentally stage-specific repression of the γ-globin gene.92  Genetic deletion of BCL11A has been used to correct sickle cell disease in adult mice by interference with fetal hemoglobin silencing.93  In addition, therapeutic levels of fetal hemoglobin have been achieved in erythroid progeny of β-thalassemic cells after lentiviral vector–mediated gene transfer of an inhibitory BCL11A short hairpin RNA.94  Efforts to advance these preclinical observations to small-scale clinical trials are of great interest.

Fanconi anemia is an inherited bone marrow failure disorder characterized by aplastic anemia and an enhanced risk for the development of leukemia. The syndrome may occur as a consequence of a defect in 1 of at least 15 genes. The development of gene therapy approaches for this disorder have focused on the protein encoded by the FANCA gene,95,96  which is most commonly mutated. An international working group has been established to chart the path and facilitate the development of gene therapy for Fanconi anemia.95,96  Gene therapy for patients with Fanconi anemia is particularly challenging because of low numbers of hematopoietic stem cells and sensitivity to myelosuppressive regimens.

Over the past decade, the use of chimeric nucleases to create specific double-stranded breaks to alter specific sites in the genome has progressed significantly.97  Several classes of enzymes have been developed that enable a broad range of genetic modification by inducing DNA double-strand breaks that stimulate error-prone nonhomologous end joining or homology-directed repair at specific genomic locations.98  Various viral vectors including AAV99  lentiviral100  and adenoviral vectors101  have been adapted for various purposes to achieve gene editing in target cells. The methodology has been used to establish HIV-1 resistance in T cells by disrupting the HIV coreceptor CC chemokine receptor 5.102  The mutation that results in synthesis of a sickle β-globin has been corrected in human induced pluripotent stem (iPS) cells providing proof of principal that this is a potentially viable approach.103-105  The nuclease methodology has also been used to achieve correction of α-thalassemia in iPS cells106  as well as genetic correction of β-thalassemia mutations in such cells.107  Another potential application is the creation of genomic safe harbors that facilitate high β-globin transgene expression. Genomic safe harbors are identified by screening of iPS cells from thalassemic individuals to identify clones with favorable integration sites.108  Basic research continues to increase the repertoire of target sites amenable to genetic modification.109  This technology could also be applicable to other blood disorders such as immunodeficiencies or CGD.

The author thanks Derek A. Persons and Brian P. Sorrentino for their review of the manuscript and for providing useful suggestions for improvement, and Pat Streich for her assistance in the preparation of this manuscript.

This work was supported by grants from the National Institutes of Health National Heart, Lung, and Blood Institute (P01HL 53749 and R01DK095169), the Assisi Foundation (94-000 R12), and the American Lebanese Syrian Associated Charities.

Contribution: A.W.N. wrote and approved the manuscript.

Conflict-of-interest disclosure: The author declares no competing financial interests.

Correspondence: Arthur W. Nienhuis, St. Jude Children’s Research Hospital, 262 Danny Thomas Pl – MS#341, Memphis, TN 38105; e-mail: arthur.nienhuis@stjude.org.

1
Nienhuis
 
AW
Development of gene therapy for blood disorders.
Blood
2008
, vol. 
111
 
9
(pg. 
4431
-
4444
)
2
Seymour
 
LW
Thrasher
 
AJ
Gene therapy matures in the clinic.
Nat Biotechnol
2012
, vol. 
30
 
7
(pg. 
588
-
593
)
3
Limberis
 
MP
Phoenix rising: gene therapy makes a comeback.
Acta Biochim Biophys Sin (Shanghai)
2012
, vol. 
44
 
8
(pg. 
632
-
640
)
4
Cavazzana-Calvo
 
M
Fischer
 
A
Hacein-Bey-Abina
 
S
Aiuti
 
A
Gene therapy for primary immunodeficiencies: part 1.
Curr Opin Immunol
2012
, vol. 
24
 
5
(pg. 
580
-
584
)
5
Aiuti
 
A
Bacchetta
 
R
Seger
 
R
Villa
 
A
Cavazzana-Calvo
 
M
Gene therapy for primary immunodeficiencies: part 2.
Curr Opin Immunol
2012
, vol. 
24
 
5
(pg. 
585
-
591
)
6
Rivat
 
C
Santilli
 
G
Gaspar
 
HB
Thrasher
 
AJ
Gene therapy for primary immunodeficiencies.
Hum Gene Ther
2012
, vol. 
23
 
7
(pg. 
668
-
675
)
7
Galy
 
A
Thrasher
 
AJ
Gene therapy for the Wiskott-Aldrich syndrome.
Curr Opin Allergy Clin Immunol
2011
, vol. 
11
 
6
(pg. 
545
-
550
)
8
Nathwani
 
AC
Tuddenham
 
EG
Rangarajan
 
S
, et al. 
Adenovirus-associated virus vector-mediated gene transfer in hemophilia B.
N Engl J Med
2011
, vol. 
365
 
25
(pg. 
2357
-
2365
)
9
Cooray
 
S
Howe
 
SJ
Thrasher
 
AJ
Retrovirus and lentivirus vector design and methods of cell conditioning.
Methods Enzymol
2012
, vol. 
507
 (pg. 
29
-
57
)
10
Beatty
 
MS
Curiel
 
DT
Chapter two—adenovirus strategies for tissue-specific targeting.
Adv Cancer Res
2012
, vol. 
115
 (pg. 
39
-
67
)
11
Xiao
 
PJ
Lentz
 
TB
Samulski
 
RJ
Recombinant adeno-associated virus: clinical application and development as a gene-therapy vector.
Ther Deliv
2012
, vol. 
3
 
7
(pg. 
835
-
856
)
12
Puntel
 
M
AKM
 
GM
Farrokhi
 
C
, et al. 
Safety profile, efficacy, and biodistribution of a bicistronic high-capacity adenovirus vector encoding a combined immunostimulation and cytotoxic gene therapy as a prelude to a phase I clinical trial for glioblastoma.
Toxicol Appl Pharmacol
2013
, vol. 
268
 
3
(pg. 
318
-
330
)
13
Nathwani
 
AC
Rosales
 
C
McIntosh
 
J
, et al. 
Long-term safety and efficacy following systemic administration of a self-complementary AAV vector encoding human FIX pseudotyped with serotype 5 and 8 capsid proteins.
Mol Ther
2011
, vol. 
19
 
5
(pg. 
876
-
885
)
14
McCarty
 
DM
Fu
 
H
Monahan
 
PE
Toulson
 
CE
Naik
 
P
Samulski
 
RJ
Adeno-associated virus terminal repeat (TR) mutant generates self-complementary vectors to overcome the rate-limiting step to transduction in vivo.
Gene Ther
2003
, vol. 
10
 
26
(pg. 
2112
-
2118
)
15
Raj
 
D
Davidoff
 
AM
Nathwani
 
AC
Self-complementary adeno-associated viral vectors for gene therapy of hemophilia B: progress and challenges.
Expert Rev Hematol
2011
, vol. 
4
 
5
(pg. 
539
-
549
)
16
O’Reilly
 
M
Kohn
 
DB
Bartlett
 
J
, et al. 
Gene therapy for rare diseases: summary of a National Institutes of Health workshop, September 13, 2012.
Hum Gene Ther
2013
, vol. 
24
 
4
(pg. 
355
-
362
)
17
Erlwein
 
O
McClure
 
MO
Progress and prospects: foamy virus vectors enter a new age.
Gene Ther
2010
, vol. 
17
 
12
(pg. 
1423
-
1429
)
18
Bauer
 
TR
Tuschong
 
LM
Calvo
 
KR
, et al. 
Long-term follow-up of foamy viral vectormediated gene therapy for canine leukocyte adhesion deficiency.
Mol Ther
2013
, vol. 
21
 
5
(pg. 
964
-
972
)
19
Suerth
 
JD
Maetzig
 
T
Galla
 
M
Baum
 
C
Schambach
 
A
Self-inactivating alpharetroviral vectors with a split-packaging design.
J Virol
2010
, vol. 
84
 
13
(pg. 
6626
-
6635
)
20
Suerth
 
JD
Maetzig
 
T
Brugman
 
MH
, et al. 
Alpharetroviral self-inactivating vectors: long-term transgene expression in murine hematopoietic cells and low genotoxicity.
Mol Ther
2012
, vol. 
20
 
5
(pg. 
1022
-
1032
)
21
Kaufmann
 
KB
Brendel
 
C
Suerth
 
JD
, et al. 
Alpharetroviral vector-mediated gene therapy for X-CGD: functional correction and lack of aberrant splicing.
Mol Ther
2013
, vol. 
21
 
3
(pg. 
648
-
661
)
22
Corrigan-Curay
 
J
Cohen-Haguenauer
 
O
O’Reilly
 
M
, et al. 
Challenges in vector and trial design using retroviral vectors for long-term gene correction in hematopoietic stem cell gene therapy.
Mol Ther
2012
, vol. 
20
 
6
(pg. 
1084
-
1094
)
23
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
)
24
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
)
25
Avedillo Díez
 
I
Zychlinski
 
D
Coci
 
EG
, et al. 
Development of novel efficient SIN vectors with improved safety features for Wiskott-Aldrich syndrome stem cell based gene therapy.
Mol Pharm
2011
, vol. 
8
 
5
(pg. 
1525
-
1537
)
26
van der Loo
 
JCM
Swaney
 
WP
Grassman
 
E
, et al. 
Critical variables affecting clinical-grade production of the self-inactivating gamma-retroviral vector for the treatment of X-linked severe combined immunodeficiency.
Gene Ther
2012
, vol. 
19
 
8
(pg. 
872
-
876
)
27
Zhou
 
S
Mody
 
D
DeRavin
 
SS
, et al. 
A self-inactivating lentiviral vector for SCID-X1 gene therapy that does not activate LMO2 expression in human T cells.
Blood
2010
, vol. 
116
 
6
(pg. 
900
-
908
)
28
Gaszner
 
M
Felsenfeld
 
G
Insulators: exploiting transcriptional and epigenetic mechanisms.
Nat Rev Genet
2006
, vol. 
7
 
9
(pg. 
703
-
713
)
29
Gaussin
 
A
Modlich
 
U
Bauche
 
C
, et al. 
CTF/NF1 transcription factors act as potent genetic insulators for integrating gene transfer vectors.
Gene Ther
2012
, vol. 
19
 
1
(pg. 
15
-
24
)
30
Xi
 
H
Shulha
 
HP
Lin
 
JM
, et al. 
Identification and characterization of cell type-specific and ubiquitous chromatin regulatory structures in the human genome.
PLoS Genet
2007
, vol. 
3
 
8
pg. 
e136
 
31
Knight
 
S
Zhang
 
F
Mueller-Kuller
 
U
, et al. 
Safer, silencing-resistant lentiviral vectors: optimization of the ubiquitous chromatin-opening element through elimination of aberrant splicing.
J Virol
2012
, vol. 
86
 
17
(pg. 
9088
-
9095
)
32
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
)
33
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
)
34
Wu
 
C
Jares
 
A
Winkler
 
T
Xie
 
J
Metais
 
JY
Dunbar
 
CE
High efficiency restriction enzyme-free linear amplification-mediated polymerase chain reaction approach for tracking lentiviral integration sites does not abrogate retrieval bias.
Hum Gene Ther
2013
, vol. 
24
 
1
(pg. 
38
-
47
)
35
Cavazzana-Calvo
 
M
Payen
 
E
Negre
 
O
, et al. 
Transfusion independence and HMGA2 activation after gene therapy of human β-thalassaemia.
Nature
2010
, vol. 
467
 
7313
(pg. 
318
-
322
)
36
Fischer
 
A
Hacein-Bey-Abina
 
S
Cavazzana-Calvo
 
M
Strategies for retrovirus-based correction of severe, combined immunodeficiency (SCID).
Methods Enzymol
2012
, vol. 
507
 (pg. 
15
-
27
)
37
Kohn
 
DB
Pai
 
S-Y
Sadelain
 
M
Gene therapy through autologous transplantation of gene-modified hematopoietic stem cells.
Biol Blood Marrow Transplant
2013
, vol. 
19
 
1, suppl
(pg. 
S64
-
S69
)
38
Hacein-Bey-Abina
 
S
Hauer
 
J
Lim
 
A
, et al. 
Efficacy of gene therapy for X-linked severe combined immunodeficiency.
N Engl J Med
2010
, vol. 
363
 
4
(pg. 
355
-
364
)
39
Zhang
 
L
Thrasher
 
AJ
Gaspar
 
HB
Current progress on gene therapy for primary immunodeficiencies [published online ahead of print May 30, 2013].
Gen Ther
40
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 [published correction appears in Sci Transl Med. 2013;5(168):168er1].
Sci Transl Med
2011
, vol. 
3
 
97
pg. 
97ra80
 
41
Montiel-Equihua
 
CA
Thrasher
 
AJ
Gaspar
 
HB
Gene therapy for severe combined immunodeficiency due to adenosine deaminase deficiency.
Curr Gene Ther
2012
, vol. 
12
 
1
(pg. 
57
-
65
)
42
Aiuti
 
A
Cattaneo
 
F
Galimberti
 
S
, et al. 
Gene therapy for immunodeficiency due to adenosine deaminase deficiency.
N Engl J Med
2009
, vol. 
360
 
5
(pg. 
447
-
458
)
43
Candotti
 
F
Shaw
 
KL
Muul
 
L
, et al. 
Gene therapy for adenosine deaminase-deficient severe combined immune deficiency: clinical comparison of retroviral vectors and treatment plans.
Blood
2012
, vol. 
120
 
18
(pg. 
3635
-
3646
)
44
Cassani
 
B
Montini
 
E
Maruggi
 
G
, et al. 
Integration of retroviral vectors induces minor changes in the transcriptional activity of T cells from ADA-SCID patients treated with gene therapy.
Blood
2009
, vol. 
114
 
17
(pg. 
3546
-
3556
)
45
Thrasher
 
AJ
Burns
 
SO
WASP: a key immunological multitasker.
Nat Rev Immunol
2010
, vol. 
10
 
3
(pg. 
182
-
192
)
46
Mahlaoui
 
N
Pellier
 
I
Mignot
 
C
, et al. 
Characteristics and outcome of early-onset, severe forms of Wiskott-Aldrich syndrome.
Blood
2013
, vol. 
121
 
9
(pg. 
1510
-
1516
)
47
Dupré
 
L
Marangoni
 
F
Scaramuzza
 
S
, et al. 
Efficacy of gene therapy for Wiskott-Aldrich syndrome using a WAS promoter/cDNA-containing lentiviral vector and nonlethal irradiation.
Hum Gene Ther
2006
, vol. 
17
 
3
(pg. 
303
-
313
)
48
Marangoni
 
F
Bosticardo
 
M
Charrier
 
S
, et al. 
Evidence for long-term efficacy and safety of gene therapy for Wiskott-Aldrich syndrome in preclinical models.
Mol Ther
2009
, vol. 
17
 
6
(pg. 
1073
-
1082
)
49
Astrakhan
 
A
Sather
 
BD
Ryu
 
BY
, et al. 
Ubiquitous high-level gene expression in hematopoietic lineages provides effective lentiviral gene therapy of murine Wiskott-Aldrich syndrome.
Blood
2012
, vol. 
119
 
19
(pg. 
4395
-
4407
)
50
Koldej
 
RM
Carney
 
G
Wielgosz
 
MM
, et al. 
Comparison of insulators and promoters for expression of the Wiskott Aldrich syndrome protein using lentiviral vectors [published online ahead of print June 20, 2013].
Hum Gene Ther Clin Dev
51
Scaramuzza
 
S
Biasco
 
L
Ripamonti
 
A
, et al. 
Preclinical safety and efficacy of human CD34(+) cells transduced with lentiviral vector for the treatment of Wiskott-Aldrich syndrome.
Mol Ther
2013
, vol. 
21
 
1
(pg. 
175
-
184
)
52
Merten
 
OW
Charrier
 
S
Laroudie
 
N
, et al. 
Large-scale manufacture and characterization of a lentiviral vector produced for clinical ex vivo gene therapy application.
Hum Gene Ther
2011
, vol. 
22
 
3
(pg. 
343
-
356
)
53
Scaramuzza
 
S
Ferrua
 
F
Castiello
 
MC
, et al. 
Lentiviral vector transduced CD34+ cells for the treatment of Wiskott-Aldrich syndrome [abstract].
Mol Ther
2012
, vol. 
20
 
suppl 1
(pg. 
S211
-
S212
Abstract 547
54
Holland
 
SM
Chronic granulomatous disease.
Hematol Oncol Clin North Am
2013
, vol. 
27
 
1
(pg. 
89
-
99, viii
)
55
Kang
 
EM
Malech
 
HL
Gene therapy for chronic granulomatous disease.
Methods Enzymol
2012
, vol. 
507
 (pg. 
125
-
154
)
56
Kang
 
HJ
Bartholomae
 
CC
Paruzynski
 
A
, et al. 
Retroviral gene therapy for X-linked chronic granulomatous disease: results from phase I/II trial.
Mol Ther
2011
, vol. 
19
 
11
(pg. 
2092
-
2101
)
57
Grez
 
M
Reichenbach
 
J
Schwäble
 
J
Seger
 
R
Dinauer
 
MC
Thrasher
 
AJ
Gene therapy of chronic granulomatous disease: the engraftment dilemma.
Mol Ther
2011
, vol. 
19
 
1
(pg. 
28
-
35
)
58
Barde
 
I
Laurenti
 
E
Verp
 
S
, et al. 
Lineage- and stage-restricted lentiviral vectors for the gene therapy of chronic granulomatous disease.
Gene Ther
2011
, vol. 
18
 
11
(pg. 
1087
-
1097
)
59
Santilli
 
G
Almarza
 
E
Brendel
 
C
, et al. 
Biochemical correction of X-CGD by a novel chimeric promoter regulating high levels of transgene expression in myeloid cells.
Mol Ther
2011
, vol. 
19
 
1
(pg. 
122
-
132
)
60
Chatziandreou
 
I
Siapati
 
EK
Vassilopoulos
 
G
Genetic correction of X-linked chronic granulomatous disease with novel foamy virus vectors.
Exp Hematol
2011
, vol. 
39
 
6
(pg. 
643
-
652
)
61
Biffi
 
A
Aubourg
 
P
Cartier
 
N
Gene therapy for leukodystrophies.
Hum Mol Genet
2011
, vol. 
20
 
R1
(pg. 
R42
-
R53
)
62
Cartier
 
N
Hacein-Bey-Abina
 
S
Bartholomae
 
CC
, et al. 
Lentiviral hematopoietic cell gene therapy for X-linked adrenoleukodystrophy.
Methods Enzymol
2012
, vol. 
507
 (pg. 
187
-
198
)
63
Orchard
 
P
Cartier
 
N
Hacein-Bey-Abina
 
S
, et al. 
Hematopoietic stem cell gene therapy with lentiviral vector in 4 patients with cerebral X-linked adrenoleukodystrophy: long-term outcome and comparison of efficacy with allogeneic hematopoietic stem cell transplantation [abstract].
Mol Ther
2013
, vol. 
21
 
suppl 1
(pg. 
S25
-
S26
Abstract 62
64
Bartholomae
 
CC
Cartier
 
N
Hacein-Bey-Abina
 
S
, et al. 
Integration profile of lentiviral vectors in gene therapy for X-adrenoleukodystrophy [abstract].
Mol Ther
2013
, vol. 
21
 
suppl 1
(pg. 
S106
-
S107
Abstract 279
65
Hess
 
B
Saftig
 
P
Hartmann
 
D
, et al. 
Phenotype of arylsulfatase A-deficient mice: relationship to human metachromatic leukodystrophy.
Proc Natl Acad Sci USA
1996
, vol. 
93
 
25
(pg. 
14821
-
14826
)
66
Matzner
 
U
Hartmann
 
D
Lüllmann-Rauch
 
R
, et al. 
Bone marrow stem cell-based gene transfer in a mouse model for metachromatic leukodystrophy: effects on visceral and nervous system disease manifestations.
Gene Ther
2002
, vol. 
9
 
1
(pg. 
53
-
63
)
67
Sevin
 
C
Benraiss
 
A
Van Dam
 
D
, et al. 
Intracerebral adeno-associated virus-mediated gene transfer in rapidly progressive forms of metachromatic leukodystrophy.
Hum Mol Genet
2006
, vol. 
15
 
1
(pg. 
53
-
64
)
68
Lupo-Stanghellini
 
MT
Provasi
 
E
Bondanza
 
A
Ciceri
 
F
Bordignon
 
C
Bonini
 
C
Clinical impact of suicide gene therapy in allogeneic hematopoietic stem cell transplantation.
Hum Gene Ther
2010
, vol. 
21
 
3
(pg. 
241
-
250
)
69
Di Stasi
 
A
Tey
 
SK
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
)
70
Leen
 
AM
Myers
 
GD
Sili
 
U
, et al. 
Monoculture-derived T lymphocytes specific for multiple viruses expand and produce clinically relevant effects in immunocompromised individuals.
Nat Med
2006
, vol. 
12
 
10
(pg. 
1160
-
1166
)
71
Leen
 
AM
Bollard
 
CM
Mendizabal
 
AM
, et al. 
Multicenter study of banked third-party virus-specific T cells to treat severe viral infections after hematopoietic stem cell transplantation.
Blood
2013
, vol. 
121
 
26
(pg. 
5113
-
5123
)
72
Gerdemann
 
U
Keirnan
 
JM
Katari
 
UL
, et al. 
Rapidly generated multivirus-specific cytotoxic T lymphocytes for the prophylaxis and treatment of viral infections.
Mol Ther
2012
, vol. 
20
 
8
(pg. 
1622
-
1632
)
73
Gerdemann
 
U
Keukens
 
L
Keirnan
 
JM
, et al. 
Immunotherapeutic strategies to prevent and treat human herpesvirus 6 reactivation after allogeneic stem cell transplantation.
Blood
2013
, vol. 
121
 
1
(pg. 
207
-
218
)
74
Kiem
 
HP
Jerome
 
KR
Deeks
 
SG
McCune
 
JM
Hematopoietic-stem-cell-based gene therapy for HIV disease.
Cell Stem Cell
2012
, vol. 
10
 
2
(pg. 
137
-
147
)
75
Chung
 
J
DiGiusto
 
DL
Rossi
 
JJ
Combinatorial RNA-based gene therapy for the treatment of HIV/AIDS.
Expert Opin Biol Ther
2013
, vol. 
13
 
3
(pg. 
437
-
445
)
76
De Keersmaecker
 
B
Allard
 
SD
Lacor
 
P
Schots
 
R
Thielemans
 
K
Aerts
 
JL
Expansion of polyfunctional HIV-specific T cells upon stimulation with mRNA electroporated dendritic cells in the presence of immunomodulatory drugs.
J Virol
2012
, vol. 
86
 
17
(pg. 
9351
-
9360
)
77
Stroncek
 
DF
Berger
 
C
Cheever
 
MA
, et al. 
New directions in cellular therapy of cancer: a summary of the summit on cellular therapy for cancer.
J Transl Med
2012
, vol. 
10
 pg. 
48
 
78
Chang
 
Y-H
Connolly
 
J
Shimasaki
 
N
Mimura
 
K
Kono
 
K
Campana
 
D
A chimeric receptor with NKG2D specificity enhances natural killer cell activation and killing of tumor cells.
Cancer Res
2013
, vol. 
73
 
6
(pg. 
1777
-
1786
)
79
Kochenderfer
 
JN
Dudley
 
ME
Feldman
 
SA
, et al. 
B-cell depletion and remissions of malignancy along with cytokine-associated toxicity in a clinical trial of anti-CD19 chimeric-antigen-receptor-transduced T cells.
Blood
2012
, vol. 
119
 
12
(pg. 
2709
-
2720
)
80
Porter
 
DL
Levine
 
BL
Kalos
 
M
Bagg
 
A
June
 
CH
Chimeric antigen receptor-modified T cells in chronic lymphoid leukemia.
N Engl J Med
2011
, vol. 
365
 
8
(pg. 
725
-
733
)
81
Kochenderfer
 
JN
Rosenberg
 
SA
Treating B-cell cancer with T cells expressing anti-CD19 chimeric antigen receptors.
Nat Rev Clin Oncol
2013
, vol. 
10
 
5
(pg. 
267
-
276
)
82
Cerullo
 
V
Koski
 
A
Vähä-Koskela
 
M
Hemminki
 
A
Chapter eight—oncolytic adenoviruses for cancer immunotherapy: data from mice, hamsters, and humans.
Adv Cancer Res
2012
, vol. 
115
 (pg. 
265
-
318
)
83
Chen
 
CY
Senac
 
JS
Weaver
 
EA
, et al. 
Species D adenoviruses as oncolytics against B-cell cancers.
Clin Cancer Res
2011
, vol. 
17
 
21
(pg. 
6712
-
6722
)
84
Larochelle
 
A
Choi
 
U
Shou
 
Y
, et al. 
In vivo selection of hematopoietic progenitor cells and temozolomide dose intensification in rhesus macaques through lentiviral transduction with a drug resistance gene.
J Clin Invest
2009
, vol. 
119
 
7
(pg. 
1952
-
1963
)
85
Adair
 
JE
Beard
 
BC
Trobridge
 
GD
, et al. 
Extended survival of glioblastoma patients after chemoprotective HSC gene therapy.
Sci Transl Med
2012
, vol. 
4
 
133
pg. 
133ra57
 
86
High
 
KA
The gene therapy journey for hemophilia: are we there yet?
Blood
2012
, vol. 
120
 
23
(pg. 
4482
-
4487
)
87
Martino
 
AT
Basner-Tschakarjan
 
E
Markusic
 
DM
, et al. 
Engineered AAV vector minimizes in vivo targeting of transduced hepatocytes by capsid-specific CD8+ T cells.
Blood
2013
, vol. 
121
 
12
(pg. 
2224
-
2233
)
88
McIntosh
 
J
Lenting
 
PJ
Rosales
 
C
, et al. 
Therapeutic levels of FVIII following a single peripheral vein administration of rAAV vector encoding a novel human factor VIII variant.
Blood
2013
, vol. 
121
 
17
(pg. 
3335
-
3344
)
89
Nienhuis
 
AW
Persons
 
DA
Development of gene therapy for thalassemia.
Cold Springs Harb Perspect Med
2012
, vol. 
2
 
11
 
pii:a011833
90
Wilber
 
A
Nienhuis
 
AW
Persons
 
DA
Transcriptional regulation of fetal to adult hemoglobin switching: new therapeutic opportunities.
Blood
2011
, vol. 
117
 
15
(pg. 
3945
-
3953
)
91
Sankaran
 
VG
Orkin
 
SH
The switch from fetal to adult hemoglobin.
Cold Spring Harb Perspect Med
2013
, vol. 
3
 
1
pg. 
a011643
 
92
Sankaran
 
VG
Menne
 
TF
Xu
 
J
, et al. 
Human fetal hemoglobin expression is regulated by the developmental stage-specific repressor BCL11A.
Science
2008
, vol. 
322
 
5909
(pg. 
1839
-
1842
)
93
Xu
 
J
Peng
 
C
Sankaran
 
VG
, et al. 
Correction of sickle cell disease in adult mice by interference with fetal hemoglobin silencing.
Science
2011
, vol. 
334
 
6058
(pg. 
993
-
996
)
94
Wilber
 
A
Hargrove
 
PW
Kim
 
YS
, et al. 
Therapeutic levels of fetal hemoglobin in erythroid progeny of β-thalassemic CD34+ cells after lentiviral vector-mediated gene transfer.
Blood
2011
, vol. 
117
 
10
(pg. 
2817
-
2826
)
95
Tolar
 
J
Adair
 
JE
Antoniou
 
M
, et al. 
Stem cell gene therapy for Fanconi anemia: report for the 1st International Fanconi Anemia Gene Therapy Working Group Meeting.
Mol Ther
2011
, vol. 
19
 
7
(pg. 
1193
-
1198
)
96
Tolar
 
J
Becker
 
PS
Clapp
 
DW
, et al. 
Gene therapy for Fanconi anemia: one step closer to the clinic.
Hum Gene Ther
2012
, vol. 
23
 
2
(pg. 
141
-
144
)
97
Porteus
 
MH
Baltimore
 
D
Chimeric nucleases stimulate gene targeting in human cells.
Science
2003
, vol. 
300
 
5620
pg. 
763
 
98
Gaj
 
T
Gersbach
 
CA
Barbas
 
CF
ZFN, TALEN, and CRISPR/Cas-based methods for genome engineering.
Trends Biotechnol
2013
, vol. 
31
 
7
(pg. 
397
-
405
)
99
Porteus
 
MH
Cathomen
 
T
Weitzman
 
MD
Baltimore
 
D
Efficient gene targeting mediated by adeno-associated virus and DNA double-strand breaks.
Mol Cell Biol
2003
, vol. 
23
 
10
(pg. 
3558
-
3565
)
100
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
)
101
Li
 
L
Krymskaya
 
L
Wang
 
J
, et al. 
Genomic editing of the HIV-1 coreceptor CCR5 in adult hematopoietic stem and progenitor cells using zinc finger nucleases.
Mol Ther
2013
, vol. 
21
 
6
(pg. 
1259
-
1269
)
102
Perez
 
EE
Wang
 
J
Miller
 
JC
, et al. 
Establishment of HIV-1 resistance in CD4+ T cells by genome editing using zinc-finger nucleases.
Nat Biotechnol
2008
, vol. 
26
 
7
(pg. 
808
-
816
)
103
Zou
 
J
Mali
 
P
Huang
 
X
Dowey
 
SN
Cheng
 
L
Site-specific gene correction of a point mutation in human iPS cells derived from an adult patient with sickle cell disease.
Blood
2011
, vol. 
118
 
17
(pg. 
4599
-
4608
)
104
Wu
 
LC
Sun
 
CW
Ryan
 
TM
Pawlik
 
KM
Ren
 
J
Townes
 
TM
Correction of sickle cell disease by homologous recombination in embryonic stem cells.
Blood
2006
, vol. 
108
 
4
(pg. 
1183
-
1188
)
105
Sun
 
N
Liang
 
J
Abil
 
Z
Zhao
 
H
Optimized TAL effector nucleases (TALENs) for use in treatment of sickle cell disease.
Mol Biosyst
2012
, vol. 
8
 
4
(pg. 
1255
-
1263
)
106
Chang
 
C-J
Bouhassira
 
EE
Zinc-finger nuclease-mediated correction of α-thalassemia in iPS cells.
Blood
2012
, vol. 
120
 
19
(pg. 
3906
-
3914
)
107
Wang
 
Y
Zheng
 
CG
Jiang
 
Y
, et al. 
Genetic correction of β-thalassemia patient-specific iPS cells and its use in improving hemoglobin production in irradiated SCID mice.
Cell Res
2012
, vol. 
22
 
4
(pg. 
637
-
648
)
108
Papapetrou
 
EP
Lee
 
G
Malani
 
N
, et al. 
Genomic safe harbors permit high β-globin transgene expression in thalassemia induced pluripotent stem cells.
Nat Biotechnol
2011
, vol. 
29
 
1
(pg. 
73
-
78
)
109
Wilson
 
KA
McEwen
 
AE
Pruett-Miller
 
SM
Zhang
 
J
Kildebeck
 
EJ
Porteus
 
MH
Expanding the repertoire of target sites for zinc finger nuclease-mediated genome modification.
Mol Ther Nucleic Acids
2013
, vol. 
2
 pg. 
e88
 
Sign in via your Institution