Skip to main content
Human Gene Therapy logoLink to Human Gene Therapy
. 2015 Apr 10;26(4):210–219. doi: 10.1089/hum.2015.047

Clinical Applications of Gene Therapy for Primary Immunodeficiencies

Maria Pia Cicalese 1,,2, Alessandro Aiuti 1,,2,,3,
PMCID: PMC4410188  PMID: 25860576

Abstract

Primary immunodeficiencies (PIDs) have represented a paradigmatic model for successes and pitfalls of hematopoietic stem cells gene therapy. First clinical trials performed with gamma retroviral vectors (γ-RV) for adenosine deaminase severe combined immunodeficiency (ADA-SCID), X-linked SCID (SCID-X1), and Wiskott–Aldrich syndrome (WAS) showed that gene therapy is a valid therapeutic option in patients lacking an HLA-identical donor. No insertional mutagenesis events have been observed in more than 40 ADA-SCID patients treated so far in the context of different clinical trials worldwide, suggesting a favorable risk–benefit ratio for this disease. On the other hand, the occurrence of insertional oncogenesis in SCID-X1, WAS, and chronic granulomatous disease (CGD) RV clinical trials prompted the development of safer vector construct based on self-inactivating (SIN) retroviral or lentiviral vectors (LVs). Here we present the recent results of LV-mediated gene therapy for WAS showing stable multilineage engraftment leading to hematological and immunological improvement, and discuss the differences with respect to the WAS RV trial. We also describe recent clinical results of SCID-X1 gene therapy with SIN γ-RV and the perspectives of targeted genome editing techniques, following early preclinical studies showing promising results in terms of specificity of gene correction. Finally, we provide an overview of the gene therapy approaches for other PIDs and discuss its prospects in relation to the evolving arena of allogeneic transplant.

Introduction

Primary immunodeficiencies (PIDs) represent a heterogeneous group of monogenic conditions determined by altered immune responses of innate and/or adaptive immunity.1 More than 260 disorders have been identified, resulting from mutations in over 300 genes.2,3 Their number is rapidly increasing thanks to next-generation sequencing technologies and increased clinical awareness.2

The incidence of PIDs ranges from 1 in 600 to 1 in 500,000 live newborns, depending upon the specific disorder.4,5 Patients with PIDs display phenotypes that can range from being asymptomatic to manifestation of life-threatening conditions (e.g., various forms of severe combined immunodeficiency, SCID). With new information on genes affecting the immune system and discovery of new pathogenic mutations and molecular mechanisms, different clinical presentations are attributed to gene defects that, in the past, appeared to have a traditional presentation only.2,6

Additionally, an increasing number of syndromes are also characterized by immune dysregulation with autoimmunity and susceptibility to lymphoreticular malignancy.5,7,8

While differing in clinical severity, early diagnosis and treatment remain a mainstay for all forms of PIDs to prevent organ damage and life-threatening infections and to improve prognosis and quality of life.6,9 Major efforts have recently been undertaken to develop methods for detection of PID in the neonatal period; in particular, a triplex RT-qPCR measuring the levels of TRECs and KRECs has been shown to provide a suitable screening for the vast majority of severe immunodeficiency diseases characterized by T- or B-lymphopenia in newborns.6 Universal newborn screening in the United States has helped to establish the true incidence of SCID in California (1 in 66,250 live births) and has led to the improvement of survival outcome.9 Recently, tandem mass spectrometry for analysis of metabolites from dried blood spots has been proposed as an easy and cheap method for adenosine deaminase (ADA) SCID screening.10,11

Bone marrow transplantation (BMT) still remains the definitive cure for most of the PIDs, and the outcomes of patients treated in European centers are improving over time.12,13 Survival is excellent in HLA genoidentical donor setting and is progressively increasing in other settings thanks to the improvement in conditioning regimens, prophylaxis and treatment of infectious complications, GvHD prevention, stem cells selection and manipulation, and choice of unrelated donors.13–17

In the last 15 years, gene therapy (GT) has been successfully implemented for the treatment of PID patients who lacked a suitable donor. In some cases, efficacy of gene therapy has been counterbalanced by the occurrence of insertional oncogenesis. The understanding of the molecular events that led to oncogenesis and improved vector technology allowed to progress with safer gene therapy approaches for PID.

Here we review the most recent results on clinical trials for X-linked SCID (SCID-X1), ADA-SCID, Wiskott–Aldrich syndrome (WAS), and chronic granulomatous disease (CGD) and discuss perspectives for new technologies and other diseases.

Gene Therapy for SCID-X1

X-linked SCID is actually the most common form of SCID, accounting for 40–50% of SCID cases reported worldwide.5 Mutations in the IL2RG gene are leading to defective expression of the common gammachain (γc), a subunit shared by a host of cytokine receptors, including interleukin (IL)-2, 4, 7, 9, 15, and 21 receptor complexes, which play a vital role in lymphocyte development and function.5 As a consequence, SCID-X1 patients present profound immunological defects caused by low numbers or complete absence of T and NK cells, and presence of nonfunctional B-cells.18 Death from community-acquired or opportunistic infections usually occurs before 1 year of age unless allogeneic hematopoietic stem cell transplantation is performed.19

While allogeneic transplantation from an HLA-identical donor has a high survival rate, persistent defects in humoral or cellular functions have been reported for some patients, resulting in partial immune recovery, autoimmunity, and/or retarded growth.19,20 On the other hand, transplantation from mismatched related, matched unrelated, or umbilical cord donors in patients with ongoing infection is associated with lower survival rates, often partial chimerism of hematopoietic lineages with persistent impairment of humoral immune function,21 and higher rates of complications as graft-versus-host disease.12–14,21

SCID-X1 was thought to be the most accurate model for assessing GT, because spontaneous reversion of the mutation in the γc-encoding IL2RG gene led to restoration of immunological competence, suggesting that transduced lymphocyte progenitors could carry a selective advantage over their nontransduced counterparts.5,22

Between 1999 and 2006, twenty subjects with SCID-X1 lacking HLA-identical bone marrow donors have been treated in two trials, conducted in Paris and London. The treatment consisted of an infusion of autologous CD34+ bone marrow cells transduced with a first-generation Moloney murine leukemia virus vector expressing the γc complementary DNA (MFG-γc) and containing duplicated viral enhancer sequences within the long terminal repeats (LTRs). Gene therapy resulted in correction of the immunodeficiency, with polyclonal and functional T-lymphocytes in 19/20 patients.3,23–26

Engraftment and correction of NK and B cells was lower, likely because patients did not receive conditioning. Immunoglobulin replacement treatment was stopped in 11/20 patients, allowing most patients to live a normal life.3,23 An 85% survival rate was observed with a median follow-up of 13 years, similar to the results obtained with matched-sibling donor hematopoietic stem cell transplantation (HSCT),19 demonstrating that gene therapy can be curative for X-SCID with long-lasting (10 years) beneficial effects.19 Four patients in the French trial and one patient in the British cohort have developed T-cell leukemia24 between 2 and 5 years after GT: four of them have been into remission after conventional chemotherapy, in one case followed by matched unrelated hematopoietic cell transplantation (HCT), and remain in long-term remission,25,26 while the remaining patient has died from chemotherapy-refractory leukemia. In all cases, the adverse event was the result of insertional oncogenesis caused by aberrant expression of the LMO2 (LIM domain only-2) or CCND2 (cyclin D2) oncogenes induced by the integration of the γc retroviral vector (RV) in the proximity of the gene regulatory regions.19 Second genome alterations were found in all cases and probably accounted for the advent of overt leukemia,3 favored by the selective advantage conferred to them by the concomitant expression of the γc gene.23 The occurrence of these serious complications prompted discontinuation of these trials.24

A further trial was started at the NIH in 2003 as a treatment option for older X-SCID patients for whom HCT was not successful. Three patients (11, 10, and 14 years old) were treated with granulocyte colony stimulating factor (G-CSF)-mobilized peripheral blood CD34+cells transduced with γ-RV: T-cell numbers and function improved only in one subject, the youngest, and no immunological improvement was found in the other two.23

To improve safety while maintaining the efficacy profile for X-SCID gene therapy, a self-inactivating (SIN) γ-RV with deleted Moloney murine leukemia virus LTR U3 enhancer was exploited, expressing the IL2RG complementary DNA from the eukaryotic human elongation factor 1α (EF1α) short promoter, and having shown to be less mutagenic in vitro, although effective in the mouse model of X-SCID (enhancer-deleted SIN-γc).27,28 The interim results of the first nine patients treated in parallel phase 1/2 trials conducted in London, Paris, Boston, Cincinnati, and Los Angeles have been recently published,19 and the trial is still recruiting patients (Table 1). SCID-X1 children were enrolled if an HLA-identical sibling donor was not available or in the case of severe ongoing, therapy-resistant infections. Eight out of nine treated patients survived, while a preexisting disseminated adenovirus infection was fatal to one patient 4 months after GT, before the full reconstitution of the T-cell compartment. Up to 48 months of follow-up, immune reconstitution of T-cells occurred in the other 7 patients and was comparable to that observed in the previous trials conducted in Paris and London. Importantly, integration analysis showed a polyclonal integration profile with reduced numbers of clones near known lymphoid proto-oncogenes and genes implicated in serious adverse events in previous GT trials.19

Table 1.

Ongoing Gene Therapy Clinical Trials for Primary Immunodeficiency

Disease Country Sponsor Vector Outcome Reference Clinical Trials.gov ref no.
SCID-X1 France
UK
US
Assistance Publique—Hôpitaux de Paris, Great Ormond Street Hospital, Children's Hospital Boston SIN γ-RV 8/9 patients alive, immune recovery with clinical benefit Hacein-Bey-Abina et al.19 NCT01410019
NCT01175239
NCT01129544
SCID-X1 US St. Jude Children's Research Hospital, National Institute of Allergy and Infectious Diseases SIN-LV Two young adult patients, improved IgG production De Ravin et al.30 NCT01512888
NCT01306019
ADA-SCID Italy GlaxoSmithKline γ-RV 15/18 patients off ERT, clinical benefit Aiuti et al.,54 Cicalese et al.117 NCT00598481
ADA- SCID UK Great Ormond Street Hospital γ-RV 4/6 patients off ERT, clinical benefit Gaspar et al.55 NCT01279720
ADA- SCID US Donald B. Kohn, UCLA γ-RV 9/10 patients off ERT, clinical benefit Candotti et al.,56 Carbonaro Sarracino et al.118 NCT00794508
ADA-SCID UK
US
Great Ormond Street Hospital;
Donald B. Kohn, UCLA
SIN-LV 5 patients, immune and metabolic recovery Gaspar et al.119 NCT01380990
NCT01852071
WAS Italy IRCCS San Raffaele SIN-LV 6 patients, immune, hematological and clinical improvement Aiuti et al.,87 Scaramuzza et al.120 NCT01515462
WAS France
UK
Genethon SIN-LV France: 4/5 patients alive, clinical improvement Bosticardo et al.89 NCT01347346
NCT01347242
WAS US Children's Hospital Boston SIN-LV 2 patients, immune and hematological improvement Williams121 NCT01410825
CGD Germany Hubert Serve, Johann Wolfgang Goethe University Hospitals SIN γ-RV     NCT01906541
CGD Germany
Switzerland
UK
US
Genethon
UCLA
SIN-LV   Kaufmann et al.109 NCT01855685
NCT02234934

ADA, adenosine deaminase; CGD, chronic granulomatous disease; ERT, enzyme replacement therapy; LV, lentiviral; RV, retroviral vector; SCID, severe combined immunodeficiency; SIN, self-inactivating; WAS, Wiskott–Aldrich syndrome.

Studies are classified as active and ongoing based on information retrieved from ClinicalTrials.gov. Updated information on recruitment status can be found on ClinicalTrials.gov.

A new approach based on the use of SIN lentiviral vector (LV; CL20-i4-EF1α-hγc-OPT) expressing a γc gene has been developed by Sorrentino and colleagues29 and is used in a two-site clinical trial. Typical X-SCID patients will be enrolled at the St. Jude Children's Research Center in Memphis, while atypical children and adolescents between 2 and 20 years of age are treated at NIH (Table 1). The latter arm of the trial uses nonmyeloablative conditioning with a total busulfan dose of 6 mg/kg/body weight to improve the efficacy of engraftment of gene-corrected cells.30 Preliminary results in two young adults after 15 and 9 months from GT show restoration of Ig production and B-cell function, increasing gene-marked NK cells, and clinical improvement.30

Gene Therapy for ADA-SCID

ADA-SCID, caused by mutations in the ADA gene impairing ADA activity, stability, and survival and leading to accumulation of toxic metabolites in plasma, red blood cells, and tissues, represents the second most frequent form of SCIDs, accounting for 15–20% of all cases of severe combined immunodeficiencies.31,32 In its typical early severe onset form, it is usually fatal in the first year of life.33 Apart from the profound lymphopenia (T, B, and NK) and the absence of cellular and humoral immune function,34 nonimmunological alterations as manifestation of the metabolic organ damage have been described.35,36

HLA-matched sibling donor (MSD) or family donor (MFD) SCT is the gold standard in ADA-SCID therapy and is associated to excellent overall survival (86% for MSD and 83% for MFD). Data from a large cohort of ADA-SCID patients transplanted with alternative donors over 20 years clearly show the importance of donor matching in improving outcome, with 67% overall survival (OS) in HLA-matched unrelated donors (MUDs) (67%) and 43% and 29% OS in haplo and mismatched unrelated donor SCT, respectively.37,38 Moreover, the metabolic nature of the disease and the need for conditioning regimens make mismatched transplantation for this form of SCID more difficult to manage than other forms, even for the associated risks.21,32 Beyond this, once patients survive the procedure and engraft donor cells, relatively complete immune reconstitution is achieved.37

Patients who receive enzyme replacement therapy (ERT) usually improve immune functions and are well detoxified, but in the long-term, they present with T-cell numbers that are below normal levels and show gradual decline of functional assays, whereas B-cell function defects are not fully repaired, with only 50% of patients able to discontinue Ig replacement therapy.37 Moreover, a significant part of patients show immune dysregulations, development of antibodies against bovine ADA, and autoimmune manifestations over time.39–46

The first gene therapy attempts, aimed to provide treatment for patients lacking an HLA-identical sibling donor, started in the early 1990s, targeting T-lymphocytes from patients on ERT.47,48 Despite that this approach was not sufficient to discontinue stably ERT, it was shown that the transduced T-cells could safely persist for more than 10 years.49,50 Importantly, a recently identified population of T-cells with stem cell properties was shown to significantly contribute to the pool of long-term living T-cells in these patients by tracking of insertion sites.50 Early attempts at gene therapy targeting CD34 progenitor cells were unsuccessful because of a low transduction rate and the decision to keep patients on ERT.51

Ameliorations in transduction and protocol changes were introduced in the GT study designs in order to improve the engraftment of modified stem cells, as well as to provide a selective pressure for the corrected cells that would ultimately translate into clinical benefit for the patients.52 The engraftment of infused stem cells was optimized by the inclusion of a mild preconditioning regimen with busulfan (4 mg/kg i.v.), to make space for the corrected progenitors.53 Finally, the selective pressure for outgrowth of gene-modified progeny was provided by the withdrawal of ERT before GT. Results obtained at TIGET, Italy, showed in 8/10 treated patients ADA levels sufficient to gain decrease of toxic metabolites and allow functional immune recovery. Thymic activity was restored to normal with polyclonal T-cell receptor repertoires. Normal serum immunoglobulin levels were detected in 50% of patients, allowing for discontinuation of immunoglobulin therapy and production of antibodies after immunization. Importantly, no leukemic or adverse events related to the therapy were observed.54 A recent update presented at the ESID meeting showed the results of 18 patients treated with an F-U of >1 year, who are all alive; among them, 15 are off-ERT (Table 1).117

Subsequently, other patients were treated in London and the United States with a slightly different approach in terms of conditioning regimen and vector design52 (Table 1). As a result, in 31 out of 42 globally treated patients GT was efficacious, leading to the ERT discontinuation and persistent immune reconstitution, long-term multilineage engraftment, and sustained systemic detoxification.3,54–56,117,118,122 Furthermore, the study by Candotti et al. compared patients treated with or without chemotherapy confirming the importance of preconditioning on the engraftment of myeloid cells and immune reconstitution.56 Importantly, the presence of shared vector integrations among multiple hematopoietic lineages demonstrated stable engraftment of multipotent HSC.57

Differently from SCID-X1 trial, and despite the use of the same first-generation γ-RV vectors, there were no genotoxic events in GT-treated SCID patients. Integrations were also found in ADA-SCID patients within and/or near potentially oncogenic loci, but did not result in selection or expansion of malignant cell clones in vivo57,58 suggesting that ADA deficiency in itself may create an unfavorable milieu for leukemogenesis. It is important to continue to monitor these patients long-term.

Based on safety issues arisen in clinical trials of retroviral GT for the treatment of other PIDs, alternatively strategies based on ADA encoding LVs were developed. Mortellaro et al. developed an SIN LV in which the expression of the human ADA gene was driven by a PGK promoter. Mice treated with GT early in life were rescued from their lethal phenotype and displayed adequate immune reconstitution and metabolic correction, similar to bone marrow transplantation.59 To further improve ADA expression, the group of Dr. Kohn and Dr. Gaspar designed an LV that included a codon-optimized human cADA gene under the control of the short-form elongation factor-1α promoter (LV EFS ADA) that displayed high-efficiency gene transfer and adequate ADA expression to rescue ADA−/− mice from their lethal phenotype with good T- and B-cell reconstitution. An in vitro immortalization assay demonstrated that LV EFS ADA had significantly less transformation potential compared with gRV vectors, without clonal skewing.60

On this basis, two phase I/II clinical trials with the use of LV EFS ADA have started in the United Kingdom and the United States for the treatment of ADA-SCID children (Table 1). To date, 5 patients aged between 1.2 and 4.5 years have been treated, after conditioning with busulfan i.v. at a single dose of ∼5 mg/kg. At a mean follow-up of about 1 year, there has been significant immunological recovery, with a rise of total T-cell and CD4+ counts and normalization in mitogen responses.119

The promising results from gene therapy trials led to issue recommendations from the EBMT Inborn Error Working Party, according to which gene therapy is considered a valid option to all patients without an HLA-identical sibling donor, regardless of the age, availability of an MUD, and outcome of PEG-ADA therapy.61

Gene Therapy for WAS

WAS is a rare, complex, X-linked PID disorder caused by mutations in the WAS gene62 characterized by recurrent infections, microthrombocytopenia, eczema, and increased risk of autoimmune manifestations and tumors.63 The prevalence is estimated to be 1–10 out of a million male individuals, with an incidence of 4 out of a million male live births. The WAS protein (WASp) is a key regulator of actin polymerization in hematopoietic cells64; thus, absence or residual WASp expression causes functional defects in different leukocyte subsets, as defective function of T- and B-cells, alteration in NK cell immunological formation synapse, and impaired migration of all leukocyte subsets.65,66 The life expectancy of WAS patients is severely reduced, unless they are successfully cured by bone marrow transplantation (BMT).15

At present, HSCT from HLA-identical sibling donor (MSD) is the treatment of choice for WAS, with a reported 82–88% long-term survival in different European and American centers in the past decade,67–69 with a survival close to 100% for patients transplanted after year 2000.15 MUD transplant has reported recently survivals of 85–90%, but better results are obtained when patients are transplanted before the age of 5, and autoimmune complications are more frequent when complete chimerism is not achieved.67 HSCT from alternative donors (including mismatched family donors and umbilical cord blood) has led to more disappointing results.

In this scenario, therapy with WAS gene-corrected autologous HSCs could represent a valid alternative approach for patients lacking a suitable donor or older than 5 years.70 Extensive preclinical studies have been performed in the last 15 years to evaluate the feasibility and efficacy of gene transfer by means of both γ-RV71–74 and LV.75,76

Based on the encouraging results of preclinical studies, a first phase I/II study on humans was conducted since 2007 in Hannover, including 10 patients, treated with WASp-expressing LTR-driven γ-RV following reduced-intensity myeloablation.77–79 Stable engraftment of gene-corrected cells in multiple lineages (HSCs, lymphoid cells, and myeloid cells) lead to restoration of WASp expression. As previously observed in mixed chimerism preclinical models,80 a clear proliferative and selective advantage of corrected lymphoid cells over myeloid lineage was also evident in patients. These results were confirmed in a larger cohort of patients, who showed partial to complete resolution of immunodeficiency, autoimmunity, and bleeding.77,81 However, the analysis of vector common insertion sites revealed a marked clustering between patients, with hotspots found within the proto-oncogenes (LMO2 and MDS/Evi1), already known to be associated in other GT trials with the development of leukemia and myelodysplasia.3,77,82,83 Between 14 months and 5 years after GT, 7 out of 10 treated patients developed hematologic malignancies.77,81 These included four cases of T-cell acute lymphoblastic leukemia (T-ALL), two primary T-ALL with secondary acute myeloid leukemia (AML), and one AML, all LMO-2 related. Despite chemotherapy and secondary allogeneic HSCT, two patients died from leukemia.77 These data indicate that LMO2-driven leukemogenesis is not specific for γc-SCID GT, but it is also seen in WAS GT. The strong viral promoter in the context of an RV, the relatively high vector copy number per cell (1.7–5.2), and the disease background might have contributed to the increased risk of insertional mutagenesis.77

While retroviral WAS gene therapy was still at preclinical level, alternative approaches with LVs were developed to overcome the issues related to γ-RV.84 The own WAS promoter was chosen to drive WASp expression to reduce the risk of insertional oncogenesis and allow a more physiological expression of the transgene. Extensive preclinical studies showed the lack of toxicity in the mouse model of the disease.85 Moreover, human CD34+ cells were effectively transduced in vitro with the vector and engrafted in immunodeficient mice.86 Clinical trials were then started in Europe and the United States (Table 1), using different conditioning regimens and enrolling patients with severe clinical score and without a suitable BMT donor.5 Results of the first three patients treated at TIGET have been recently published.87 After a reduced intensity conditioning with busulfan and fludarabine, patients received autologous HSCs, transduced with the LV encoding the human WASp cDNA. All patients showed a multilineage engraftment of corrected cells, both in bone marrow and peripheral blood compartment, with stable levels of WASp expression.

The immunological function restoration involved T- and B-cell compartment, as well as cytotoxic activity of NK cells and suppressive activity of Treg. Furthermore, platelet counts increased with respect to the pre-GT phase, and platelets presented with normal volume. These biological improvements lead to a clinical benefit for all treated patients, with a reduction of severity and frequency of infections and bleeding and the absence of autoimmune manifestations. The levels of corrected cells in the bone marrow were significantly higher than the engraftment levels achieved in the previous RV trial, suggesting a higher gene transfer efficiency of LV. In terms of safety, analysis of LV insertion profile in vivo showed that, in contrast to RV-GT, LV integrations are less prone to cluster near genes involved in hematopoietic functions and potential proto-oncogenes. Moreover, highly represented genes targeted by the vector in these WAS patients were also hit in other LV-GT trials,88 where no clonal expansion or leukemia have been reported. An SIN-LV configuration and the presence of an autologous WASp human promoter in the vector construct were crucial in developing a safer GT approach for WAS.89 A longer evaluation of all patients treated, together with data of other LV-GT trials, will be important to confirm the safety and efficacy of this approach.

Another LV vector using a viral MND-derived promoter has also been used to further increase WASp expression in mice, and results indicate that the γ-RV-derived promoter leads to a stronger transgene expression as compared with the WAS-promoter vector. However, this occurs in association with myeloid clonal expansion and transcriptional dysregulation, highlighting the potential risk of the use of a strong viral promoter.3,90

Gene Therapy for CGD

Mutations impairing the expression of gp91phox, p22phox, p47pox, or p67phox molecules are affecting the superoxide production in phagocytic cells, leading to CGD disorder, in which life-threatening abscesses and/or skin, liver, lung, or bone granuloma, and inflammatory complications are characteristic.23,91 Available therapeutic strategies include antibiotic long-life prophylaxis, IFN-γ administration, and HCT.23,92 HSCT has recently shown a high success rate as an early intervention in patients with very low superoxide production and in patients with a history of severe invasive fungal infection, organ abscesses, and/or significant inflammatory or autoimmune signs.17,93 This constitutes an argument in favor of the GT approach for patients without a matched donor. Early clinical trials performed with γRV without conditioning showed only transitory functional correction of ≤0.5% of peripheral blood granulocytes.94,95 Since gene-transduced neutrophils have no survival advantage over defective neutrophils and have a lifespan of only a few days, engraftment of relatively high numbers of gene-transduced HSCs is required by preparatory conditioning.91

Most recent trials for X-CGD were conducted in five different centers worldwide (Frankfurt, Zurich, London, NIH, and Seoul) using γRV vector-transduced, mobilized CD34+ cells and nonmyeloablative conditioning with low-dose (8–10 mg/kg) busulfan93,96–98±fludarabine,92 or melphalan alone (140 mg/m2)98 in more than 10 patients. The treatments resulted in initial transient improvement of functional neutrophils up to 30%, with clearance of severe fungal infections and clinical benefit, followed by a yet-unexplained difficulty in achieving long-term engraftment of significant levels of transduced cells, with loss of the expression of the therapeutic gene gp91phox.99 The methylation of the viral promoter leading to silencing of transgene expression is an hypothesis suggested for loss of engraftment.83

Alternatively, ectopic gp91phox expression in HSPC could cause the production of reactive oxygen species that may damage DNA, alter cell growth, or induce apoptosis.100–102 Moreover, immune-mediated mechanisms against gp91phox-expressing cells could have contributed to the lack of long-term persistence.102 On the other hand, the first-generation γ-RV used in these protocols have also been associated with a high incidence of severe adverse events in the patients with persistent gene marking. A myelodysplastic syndrome (MDS) occurred in three patients (two in Frankfurt, with fatal outcome, and one in Zurich). The second child treated in Zurich displayed a clonal expansion without monosomy 7 or MDS, and this clone disappeared after a successful early HSCT.93 The frequency of these adverse events highlights the fact that only gp91phox-transduced cells with gain-of-function events could persist in patients treated with GT protocols employing LTR-driven RV.102

All these events were associated with the insertion near MDS-EVI-1 proto-oncogenes, suggesting the necessity to improve the safety and the efficacy of gene transfer technology.83,92,97,99,103 At the same time, different strategies to restrict transgene expression to the mature phagocyte compartment were developed using SIN LVs and have been tested in preclinical and clinical development (Table 1). These include gp91phox-encoding vector driven by synthetic chimeric promoter in combination with different myeloid transcription factor binding sites or the A2UCOE element linked to a myeloid promoter driving gp91phox expression in murine myeloid cells.102,104–106 However, as A2UCOE protects from promoter methylation, its chromatin remodeling properties could have considerable side effects in HSCs,105,107 and so further studies are needed to proceed to clinical applications.108

Another recent approach to improve and maximize transgene expression in myeloid cells while avoiding expression in HSCs is based on the use of an miR-126 target sequence fused to the transgene driven by a myeloid-specific promoter. Transgene expression is provided by the myeloid-specific promoter in myeloid cells and stringent control of gp91phox expression by a miR-126 target sequence in HSCs support further development of this microRNA approach as an alternative gene transfer technique for CGD.102,106

A multicentric trial in collaboration between the United Kingdom, Switzerland, and Germany using an LV with the chimeric promoter was approved and is currently recruiting (Table 1).109 Preclinical studies for the above dual-regulated LV gene therapy approach are currently ongoing.109

New Technologies and Future Plans

The success of gene therapy achieved in the last years has been the result of improved technology and enlarged knowledge on PID and their molecular mechanisms. As the safety of the patients remains a crucial point, the use of new-generation vectors, such as SIN vectors or LVs, showing high efficacy in terms of sustainable transgene expression and reduced risk of insertional mutagenesis tendency in vitro and in vivo, has been preferred for certain PIDs, characterized by an increased risk of oncogenesis for their genetic background.

Progresses in vector design and HSC biology are favoring the extension of clinical trials to several PID variants, particularly to some challenging ones, for which the current available technologies are not sufficient. Preclinical experiments are ongoing for PIDs, such as Artemis deficiency, CD3γ deficiency, JAK3-SCID, LAD-1, PNP deficiency, RAG1/2 deficiency, X-HIM, XLA, XLP, ZAP70 deficiency, and IPEX, that would benefit from gene therapy approaches.23

Furthermore, these results have now been translated from PIDs to other blood-borne disorders, such as lysosomal storage disorders, (β)-thalassemia, and sickle cell disease, which require a higher therapeutic threshold. Clinical trials with β globin lentivirus vectors are now open at multiple sites, and transfusion independence following GT has been reported in one patient with β-thalassemia.110 In the metabolic diseases field, gene therapy has led to successful ABCD1 gene transfer by LV in autologous engineered cells of patients with X-linked adrenoleukodystrophy111 and stable LV ARSA gene replacement in patients with metachromatic leukodystrophy.88

On the other hand, BMT has become much safer and more successful over time for PID patients, thanks to early diagnosis, also because of newborn screening programs, and to the improved outcome of transplants from MUDs determined by new conditioning regimens, accuracy of typing, and new cells manipulation processes. The long-term benefits, safety, and cost-effectiveness of gene therapy versus allogeneic BMT should be evaluated thoroughly in the next years, together with practical issues, such as the choice of vector, the patient's bone marrow stem cell reservoir, and the manufacturing ability to transduce a high number of HSCs.

Gene editing will represent a further step to provide a correction in the defective genes at their genomic locus, maintaining appropriate regulatory control of gene expression and reducing the risk of genotoxicity through ectopic vector insertion. Zinc finger nucleases (ZFNs), meganucleases (MN), transcription activator-like effector nucleases (TALENS), and, more recently, clustered, regularly interspaced, short palindromic repeat (CRISPR) nucleases are all being developed to create highly specific gene targeting.112–116 The efficiency of gene editing using these techniques has been shown in cell lines and certain primary cell lineages, although remains limited in primary HSCs.91 Proof of principle for the γ-chain gene has been recently obtained in vitro and in animal models.112

In conclusion, gene therapy for PID is quickly moving from being an experimental approach to a standard cellular therapy, as demonstrated by the adoption of vector manufacture by mainstream pharmaceutical companies, on the basis of the encouraging results. Further refinement and standardization of the technology will be important for the future clinical development and to enter into the arena of approved therapies.

Author Disclosure Statement

M.P.C. and A.A. declare that no competing financial interests exist.

References

  • 1.Al-Herz W, Bousfiha A, Casanova JL, et al. . Primary immunodeficiency diseases: an update on the classification from the international union of immunological societies expert committee for primary immunodeficiency. Front Immunol 2011;8:2–54 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Locke BA, Dasu T, Verbsky JW. Laboratory diagnosis of primary immunodeficiencies. Clin Rev Allergy Immunol 2014;46:154–168 [DOI] [PubMed] [Google Scholar]
  • 3.Fischer A, Hacein-Bey-Abina S, Touzot F, et al. . Gene therapy for primary immunodeficiencies. Clin Genet 2015. [Epub ahead of print] [DOI] [PubMed] [Google Scholar]
  • 4.Zhang L, Thrasher AJ, Gaspar HB. Current progress on gene therapy for primary immunodeficiencies. Gene Ther 2013;20:963–969 [DOI] [PubMed] [Google Scholar]
  • 5.Mukherjee S, Thrasher AJ. Gene therapy for PIDs: progress, pitfalls and prospects. Gene 2013;525:174–181 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Borte S, von Dobeln U, Fasth A, et al. . Neonatal screening for severe primary immunodeficiency diseases using high-throughput triplex real-time PCR. Blood 2012;119:2552–2555 [DOI] [PubMed] [Google Scholar]
  • 7.Kildebeck E, Checketts J, Porteus M. Gene therapy for primary immunodeficiencies. Curr Opin Pediatr 2012;24:731–738 [DOI] [PubMed] [Google Scholar]
  • 8.Rivat C, Santilli G, Gaspar HB, et al. . Gene therapy for primary immunodeficiencies. Hum Gene Ther 2012;23:668–675 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Chinen J, Notarangelo LD, Shearer WT. Advances in basic and clinical immunology in 2013. J Allergy Clin Immunol 2014;133:967–976 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.La Marca G, Malvagia S, Casetta B, et al. . Progress in expanded newborn screening for metabolic conditions by LC–MS/MS in Tuscany: update on methods to reduce false tests. J Inherit Metab Dis 2008;2:395–404 [DOI] [PubMed] [Google Scholar]
  • 11.Azzari C, la Marca G, Resti M. Neonatal screening for severe combined immunodeficiency caused by an adenosine deaminase defect: a reliable and inexpensive method using tandem mass spectrometry. J Allergy Clin Immunol 2011;127:1394–1399 [DOI] [PubMed] [Google Scholar]
  • 12.Buckley RH. Transplantation of hematopoietic stem cells in human severe combined immunodeficiency: longterm outcomes. Immunol Res 2011;49:25–43 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Pai SY, Logan BR, Griffith LM, et al. . Transplantation outcomes for severe combined immunodeficiency, 2000–2009. N Engl J Med 2014;371:434–446 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Gennery AR, Slatter MA, Grandin L, et al. . Transplantation of hematopoietic stem cells and long-term survival for primary immunodeficiencies in Europe: entering a new century, do we do better? J Allergy Clin Immunol 2010;126:602–610 [DOI] [PubMed] [Google Scholar]
  • 15.Moratto D, Giliani S, Bonfim C, et al. . Long-term outcome and lineage-specific chimerism in 194 patients with Wiskott-Aldrich syndrome treated by hematopoietic cell transplantation in the period 1980–2009: an international collaborative study. Blood 2011;118:1675–1684 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Bertaina A, Merli P, Rutella S, et al. . HLA-haploidentical stem cell transplantation after removal of alphabeta+ T and B cells in children with nonmalignant disorders. Blood 2014;124:822–826 [DOI] [PubMed] [Google Scholar]
  • 17.Gungor T, Teira P, Slatter M, et al. . Reduced-intensity conditioning and HLA-matched haemopoietic stem-cell transplantation in patients with chronic granulomatous disease: a prospective multicentre study. Lancet 2014;383:436–448 [DOI] [PubMed] [Google Scholar]
  • 18.Fischer A, Cavazzana-Calvo M. Gene therapy of inherited diseases. Lancet 2008;371:2044–2047 [DOI] [PubMed] [Google Scholar]
  • 19.Hacein-Bey-Abina S, Pai SY, Gaspar HB, et al. . A modified γ-retrovirus vector for X-linked severe combined immunodeficiency. N Engl J Med 2014;371:1407–1417 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Neven B, Leroy S, Decaluwe H, et al. . Long-term outcome after haematopoietic stem cell transplantation of a single-centre cohort of 90 patients with severe combined immunodeficiency: long-term outcome of HSCT in SCID. Blood 2009;113:4114–4124 [DOI] [PubMed] [Google Scholar]
  • 21.Antoine C, Muller S, Cant A, et al. . Long-term survival and transplantation of haemopoietic stem cells for immunodeficiencies: report of the European experience 1968–99. Lancet 2003;361:553–560 [DOI] [PubMed] [Google Scholar]
  • 22.Bousso P, Wahn V, Douagi I, et al. . Diversity, functionality, and stability of the T cell repertoire derived in vivo from a single human T cell precursor. Proc Natl Acad Sci USA 2000;97:274–278 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Candotti F. Gene transfer into hematopoietic stem cells as treatment for primary immunodeficiency diseases. Int J Hematol 2014;99:383–392 [DOI] [PubMed] [Google Scholar]
  • 24.Cavazzana-Calvo M, Fischer A, Hacein-Bey-Abina S, et al. . Gene therapy for primary immunodeficiencies: part 1. Curr Opin Immunol 2012;24:580–584 [DOI] [PubMed] [Google Scholar]
  • 25.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;363:355–364 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.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;3:97ra79. [DOI] [PubMed] [Google Scholar]
  • 27.Thornhill SI, Schambach A, Howe SJ, et al. . Self-inactivating gammaretroviral vectors for gene therapy of X-linked severe combined immunodeficiency. Mol Ther 2008;16:590–598 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Zychlinski D, Schambach A, Modlich U, et al. . Physiological promoters reduce the genotoxic risk of integrating gene vectors. Mol Ther 2008;16:718–725 [DOI] [PubMed] [Google Scholar]
  • 29.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;116:900–908 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.De Ravin SS, Choi U, Theobald N, et al. . Lentiviral gene transfer for treatment of children 2 years old with X-linked severe combined immunodeficiency. Mol Ther 2013;21:S118 [Google Scholar]
  • 31.Aiuti A, Brigida I, Ferrua F, et al. . Hematopoietic stem cell gene therapy for adenosine deaminase deficient-SCID. Immunol Res 2009;44:150–159 [DOI] [PubMed] [Google Scholar]
  • 32.Gaspar HB, Aiuti A, Porta F, et al. . How I treat ADA deficiency. Blood 2009;114:3524–3532 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Grunebaum E, Cohen A, Roifman CM. Recent advances in understanding and managing adenosine deaminase and purine nucleoside phosphorylase deficiencies. Curr Opin Allergy Clin Immunol 2013;13:630–638 [DOI] [PubMed] [Google Scholar]
  • 34.Hirschhorn R, Candotti F. Immunodeficiency due to defects of purine metabolism. In: Primary Immunodeficiency Diseases: A Molecular and Genetic Approach, 2nd ed. Ochs H, Smith CIE, Puck JM, eds. (Oxford University Press, New York, NY: ). 2006; pp. 169–196 [Google Scholar]
  • 35.Honig M, Albert MH, Schulz A, et al. . Patients with adenosine deaminase deficiency surviving after hematopoietic stem cell transplantation are at high risk of CNS complications. Blood 2007;109:3595–3602 [DOI] [PubMed] [Google Scholar]
  • 36.Sauer AV, Mrak E, Hernandez RJ, et al. . ADA-deficient SCID is associated with a specific microenvironment and bone phenotype characterized by RANKL/OPG imbalance and osteoblast insufficiency. Blood 2009;114:3216–3226 [DOI] [PubMed] [Google Scholar]
  • 37.Hassan A, Booth C, Brightwell A, et al. . Outcome of hematopoietic stem cell transplantation for adenosine deaminase-deficient severe combined immunodeficiency Blood 2012;120:3615–3624; quiz 3626 [DOI] [PubMed] [Google Scholar]
  • 38.Pai SY, Logan BR, Griffith LM, et al. . Transplantation outcomes for severe combined immunodeficiency, 2000–2009. N Engl J Med 2014;371:434–446 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Weinberg K, Hershfield MS, Bastian J, et al. . T lymphocyte ontogeny in adenosine deaminase-deficient severe combined immune deficiency after treatment with polyethylene glycol-modified adenosine deaminase. J Clin Invest 1993;92:596–602 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Kohn DB. Gene therapy for childhood immunological diseases. Bone Marrow Transplant 2008;41:199–205 [DOI] [PubMed] [Google Scholar]
  • 41.Malacarne F, Benicchi T, Notarangelo LD, et al. . Reduced thymic output, increased spontaneous apoptosis and oligoclonal B cells in polyethylene glycol-adenosine deaminase-treated patients. Eur J Immunol 2005;35:3376–3386 [DOI] [PubMed] [Google Scholar]
  • 42.Notarangelo LD, Stoppoloni G, Toraldo R, et al. . Insulin-dependent diabetes mellitus and severe atopic dermatitis in a child with adenosine deaminase deficiency. Eur J Pediatr 1992;151:811–814 [DOI] [PubMed] [Google Scholar]
  • 43.Ozsahin H, Arredondo-Vega FX, Santisteban I, et al. . Adenosine deaminase deficiency in adults. Blood 1997;89:2849–2855 [PubMed] [Google Scholar]
  • 44.Sauer AV, Brigida I, Carriglio N, et al. . Autoimmune dysregulation and purine metabolism in adenosine deaminase deficiency. Front Immunol 2012;3:265. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Sauer AV, Brigida I, Carriglio N, et al. . Alterations in the adenosine metabolism and CD39/CD73 adenosinergic machinery cause loss of Treg cell function and autoimmunity in ADA-deficient SCID. Blood 2012;119:1428–1439 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Brigida I, Sauer AV, Ferrua F, et al. . B-cell development and functions and herapeutic options in adenosine deaminase-deficient patients. J Allergy Clin Immunol 2014;133:799–806 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Bordignon C, Mavilio F, Ferrari G, et al. . Transfer of the ADA gene into bone marrow cells and peripheral blood lymphocytes for the treatment of patients affected by ADA-deficient SCID. Hum Gene Ther 1993;4:513–520 [DOI] [PubMed] [Google Scholar]
  • 48.Blaese RM. Optimism regarding the use of RNA/DNA hybrids to repair genes at high efficiency. J Gene Med 1999;1:144–147 [DOI] [PubMed] [Google Scholar]
  • 49.Muul LM, Tuschong LM, Soenen SL, et al. . Persistence and expression of the adenosine deaminase gene for 12 years and immune reaction to gene transfer components: long-term results of the first clinical gene therapy trial. Blood 2003;101:2563–2569 [DOI] [PubMed] [Google Scholar]
  • 50.Biasco L, Scala S, Basso Ricci L, et al. . In vivo tracking of T cells in humans unveils decade-long survival and activity of genetically modified T memory stem cells. Sci Transl Med 2015;7:273ra13. [DOI] [PubMed] [Google Scholar]
  • 51.Gaspar HB, Qasim W, Davies EG, et al. . How I treat severe combined immunodeficiency. Blood 2013;122:3749–3758 [DOI] [PubMed] [Google Scholar]
  • 52.Montiel-Equihua CA, Thrasher AJ, Gaspar HB. Gene therapy for severe combined immunodeficiency due to adenosine deaminase deficiency. Curr Gene Ther 2012;12:57–65 [DOI] [PubMed] [Google Scholar]
  • 53.Aiuti A, Slavin S, Aker M, et al. . Correction of ADA-SCID by stem cell gene therapy combined with nonmyeloablative conditioning. Science 2002;296:2410–2413 [DOI] [PubMed] [Google Scholar]
  • 54.Aiuti A, Cattaneo F, Galimberti S, et al. . Gene therapy for immunodeficiency due to adenosine deaminase deficiency. N Engl J Med 2009;360:447–458 [DOI] [PubMed] [Google Scholar]
  • 55.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;3:97ra80. [DOI] [PubMed] [Google Scholar]
  • 56.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;120:3635–3646 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Biasco L, Ambrosi A, Pellin D, et al. . Integration profile of retroviral vector in gene therapy treated patients is cell-specific according to gene expression and chromatin onformation of target cell. EMBO Mol Med 2011;3:89–101 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.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;117:2233–2240 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Mortellaro A, Hernandez RJ, Guerrini MM, et al. . Ex vivo gene therapy with lentiviral vectors rescues adenosine deaminase (ADA)-deficient mice and corrects their immune and metabolic defects. Blood 2006;108:2979–2988 [DOI] [PubMed] [Google Scholar]
  • 60.Carbonaro DA, Zhang L, Jin X, et al. . Preclinical demonstration of lentiviral vector-mediated correction of immunological and metabolic abnormalities in models of adenosine deaminase deficiency. Mol Ther 2014;22:607–622 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.EBMT/ESID Guidelines for Haematopoietic Stem Cell Transplantation for Primary Immunodeciencies, 2011. www.ebmt.org/Contents/About-EBMT/Who-We-Are/ScientificCouncil/Documents/EBMT_ESID%20GUIDELINES%20FOR%20INBORN%20ERRORS%20FINAL%202011.pdf (accessed April16, 2015)
  • 62.Derry JM, Kerns JA, Weinberg KI, et al. . WASP gene mutations in Wiskott-Aldrich syndrome and X-linked thrombocytopenia. Hum Mol Genet 1995;4:1127–1135 [DOI] [PubMed] [Google Scholar]
  • 63.Catucci M, Castiello MC, Pala F, et al. . Autoimmunity in wiskott-Aldrich syndrome: an unsolved enigma. Front Immunol 2012;3:209. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Thrasher AJ. WASp in immune-system organization and function. Nat Rev Immunol 2002;2:635–646 [DOI] [PubMed] [Google Scholar]
  • 65.Huang H, Manton KG. Newborn screening for severe combined immunodeficiency (SCID): a review. Front Biosci 2005;10:1024–1039 [DOI] [PubMed] [Google Scholar]
  • 66.Silvin C, Belisle B, Abo A. A role for Wiskott-Aldrich syndrome protein in T-cell receptor-mediated transcriptional activation independent of actin polymerization. J Biol Chem 2001;276:21450–21457 [DOI] [PubMed] [Google Scholar]
  • 67.Ozsahin H, Cavazzana-Calvo M, Notarangelo LD, et al. . Long-term outcome following hematopoietic stem-cell transplantation in Wiskott-Aldrich syndrome: collaborative study of the European Society for Immunodeficiencies and European Group for Bloodand Marrow Transplantation. Blood 2008;111:439–445 [DOI] [PubMed] [Google Scholar]
  • 68.Filipovich AH, Stone JV, Tomany SC, et al. . Impact of donor type on outcome of bone marrow transplantation for Wiskott-Aldrich syndrome: collaborative study of the International Bone Marrow Transplant Registry and the National Marrow Donor Program. Blood 2001;97:1598–1603 [DOI] [PubMed] [Google Scholar]
  • 69.Kobayashi R, Ariga T, Nonoyama S, et al. . Outcome in patients with Wiskott-Aldrich syndrome following stem cell transplantation: an analysis of 57 patients in Japan. Br J Haematol 2006;135:362–366 [DOI] [PubMed] [Google Scholar]
  • 70.Fischer A, Hacein-Bey-Abina S, Cavazzana-Calvo M. 20 years of gene therapy for SCID. Nat Immunol 2010;11:457–460 [DOI] [PubMed] [Google Scholar]
  • 71.Candotti F, Facchetti F, Blanzuoli L, et al. . Retrovirus-mediated WASP gene transfer corrects defective actin polymerization in B cell lines from Wiskott-Aldrich syndrome patients carrying “null” mutations. Gene Ther 1999;6:1170–1174 [DOI] [PubMed] [Google Scholar]
  • 72.Wada T, Jagadeesh GJ, Nelson DL, et al. . Retrovirus-mediated WASP gene transfer corrects Wiskott-Aldrich syndrome T-cell dysfunction. Hum Gene Ther 2002;13:1039–1046 [DOI] [PubMed] [Google Scholar]
  • 73.Klein C, Nguyen D, Liu CH, et al. . Gene therapy for Wiskott-Aldrich syndrome: rescue of T-cell signaling and amelioration of colitis upon transplantation of retrovirally transduced hematopoietic stem cells in mice. Blood 2003;101:2159–2166 [DOI] [PubMed] [Google Scholar]
  • 74.Strom TS, Gabbard W, Kelly PF, et al. . Functional correction of T cells derived from patients with the Wiskott-Aldrich syndrome (WAS) by transduction with an oncoretroviral vector encoding the WAS protein. Gene Ther 2003;10:803–809 [DOI] [PubMed] [Google Scholar]
  • 75.Galy A, Roncarolo MG, Thrasher AJ. Development of lentiviral gene therapy for Wiskott Aldrich syndrome. Expert Opin Biol Ther 2008;8:181–190 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Dupré L, Trifari S, Follenzi A, et al. . Lentiviral vector-mediated gene transfer in T cells from Wiskott-Aldrich syndrome patients leads to functional correction. Mol Ther 2004;10:903–915 [DOI] [PubMed] [Google Scholar]
  • 77.Braun CJ, Boztug K, Paruzynski A, et al. . Gene therapy for Wiskott-Aldrich Syndrome—long-term efficacy and genotoxicity. Sci Transl Med 2014;6:227ra33. [DOI] [PubMed] [Google Scholar]
  • 78.Thrasher AJ, Burns SO. WASP: a key immunological multitasker. Nat Rev Immunol 2010;10:182–192 [DOI] [PubMed] [Google Scholar]
  • 79.Boztug K, Schmidt M, Schwarzer A, et al. . Stem-cell gene therapy for the Wiskott-Aldrich syndrome. N Engl J Med 2010;363:1918–1927 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Westerberg LS, de la Fuente MA, Wermeling F, et al. . WASP confers selective advantage for specific hematopoietic cell populations and serves a unique role in marginal zone B-cell homeostasis and function. Blood 2008;112:4139–4147 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Paruzynski A, Glimm H, Schmidt M, et al. . Analysis of the clonal repertoire of gene-corrected cells in gene therapy. Methods Enzymol 2012;507:59–87 [DOI] [PubMed] [Google Scholar]
  • 82.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;118:3143–3150 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.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;16:198–204 [DOI] [PubMed] [Google Scholar]
  • 84.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;17:303–313 [DOI] [PubMed] [Google Scholar]
  • 85.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;17:1073–1082 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.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;21:175–184 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Aiuti A, Biasco L, Scaramuzza S, et al. . Lentiviral hematopoietic stem cell gene therapy in patients with Wiskott-Aldrich syndrome. Science 2013;341:1233151. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Biffi A, Montini E, Lorioli L, et al. . Lentiviral hematopoietic stem cell gene therapy benefits metachromatic leukodystrophy. Science 2013;341:1233158. [DOI] [PubMed] [Google Scholar]
  • 89.Bosticardo M, Ferrua F, Cavazzana M, et al. . Gene therapy for Wiskott-Aldrich Syndrome. Curr Gene Ther 2014;14:413–421 [DOI] [PubMed] [Google Scholar]
  • 90.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;119:4395–4407 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Qasim W, Gennery AR. Gene therapy for primary immunodeficiencies: current status and future prospects. Drugs 2014;74:963–969 [DOI] [PubMed] [Google Scholar]
  • 92.Kang EM, Marciano BE, DeRavin S, et al. . Chronic granulomatous disease: overview and hematopoietic stem cell transplantation. J Allergy Clin Immunol 2011;127:1319–1326; quiz 1327–1328 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Aiuti A, Bacchetta R, Seger R, et al. . Gene therapy for primary immunodeficiencies: Part 2. Curr Opin Immunol 2012;24:585–591 [DOI] [PubMed] [Google Scholar]
  • 94.Sekhsaria S, Fleisher TA, Vowells S, et al. . Granulocyte colony-stimulating factor recruitment of CD34+ progenitors to peripheral blood: impaired mobilization in chronic granulomatous disease and adenosine deaminase—deficient severe combined immunodeficiency disease patients. Blood 1996;88:1104–1112 [PubMed] [Google Scholar]
  • 95.Goebel WS, Dinauer MC. Gene therapy for chronic granulomatous disease. Acta Haematol 2003;110:86–92 [DOI] [PubMed] [Google Scholar]
  • 96.Kang EM, Choi U, Theobald N, et al. . Retrovirus gene therapy for X-linked chronic granulomatous disease can achieve stable long-term correction of oxidase activity in peripheral blood neutrophils. Blood 2010;115:783–791 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.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;12:401–409 [DOI] [PubMed] [Google Scholar]
  • 98.Grez M, Reichenbach J, Schwäble J, et al. . Gene therapy of chronic granulomatous disease: the engraftment dilemma. Mol Ther 2011;19:28–35 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Farinelli G, Capo V, Scaramuzza S, et al. . Lentiviral vectors for the treatment of primary immunodeficiencies. J Inherit Metab Dis 2014;37:525–533 [DOI] [PubMed] [Google Scholar]
  • 100.Bedard K, Krause KH. The NOX family of ROS-generating NADPH oxidases: physiology and pathophysiology. Physiol Rev 2007;87:245–313 [DOI] [PubMed] [Google Scholar]
  • 101.Yahata T, Takanashi T, Muguruma Y, et al. . Accumulation of oxidative DNA damage restricts the self-renewal capacity of human hematopoietic stem cells. Blood 2011;118:2941–2950 [DOI] [PubMed] [Google Scholar]
  • 102.Chiriaco M, Farinelli G, Capo V, et al. . Dual-regulated lentiviral vector for gene therapy of X-linked chronic granulomatosis. Mol Ther 2014;22:1472–1483 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Bianchi M, Hakkim A, Brinkmann V, et al. . Restoration of NET formation by gene therapy in CGD controls aspergillosis. Blood 2009;114:2619–2622 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.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;18:1087–1097 [DOI] [PubMed] [Google Scholar]
  • 105.Brendel C, Müller-Kuller U, Schultze-Strasser S, et al. . Physiological regulation of transgene expression by a lentiviral vector containing the A2UCOE linked to a myeloid promoter. Gene Ther 2012;19:1018–1029 [DOI] [PubMed] [Google Scholar]
  • 106.Sauer AV, Di Lorenzo B, Carriglio N, et al. . Progress in gene therapy for primary immunodeficiencies using lentiviral vectors. Curr Opin Allergy Clin Immunol 2014;14:527–534 [DOI] [PubMed] [Google Scholar]
  • 107.Williams S, Mustoe T, Mulcahy T, et al. . CpG-island fragments from the HNRPA2B1/CBX3 genomic locus reduce silencing and enhance transgene expression from the hCMV promoter/enhancer in mammalian cells. BMC Biotechnol 2005;5:17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Zhang F, Frost AR, Blundell MP, et al. . A ubiquitous chromatin opening element (UCOE) confers resistance to DNA methylation-mediated silencing of lentiviral vectors. Mol Ther 2010;18:1640–1649 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Kaufmann KB, Büning H, Galy A, et al. . Gene therapy on the move. EMBO Mol Med 2013;5:1642–1661 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Cavazzana-Calvo M, Payen E, Negre O, et al. . Transfusion independence and HMGA2 activation after gene therapy of human β-thalassaemia. Nature 2010;467:318–322 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Cartier N, Hacein-Bey-Abina S, Von Kalle C, et al. . Gene therapy of x-linked adrenoleukodystrophy using hematopoietic stem cells and a lentiviral vector. Bull Acad Natl Med 2010;194:255–264; discussion 264–268 [PubMed] [Google Scholar]
  • 112.Genovese P, Schiroli G, Escobar G, et al. . Targeted genome editing in human repopulating haematopoietic stem cells. Nature 2014;510:235–240 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Urnov FD, Miller JC, Lee YL, et al. . Highly efficient endogenous human gene correction using designed zinc-finger nucleases. Nature 2005;435:646–651 [DOI] [PubMed] [Google Scholar]
  • 114.Paques F, Duchateau P. Meganucleases and DNA double-strand break-induced recombination: perspectives for gene therapy. Curr Gene Ther 2007;7:49–66 [DOI] [PubMed] [Google Scholar]
  • 115.Wood AJ, Lo TW, Zeitler B, et al. . Targeted genome editing across species using ZFNs and TALENs. Science 2011;333:307. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Ran FA, Hsu PD, Wright J, et al. . Genome engineering using the CRISPR-Cas9 system. Nat Protoc 2013;8:2281–2308 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Cicalese MP, Ferrua F, Pajno R, et al. . Long-term safety and efficacy of retroviral-mediated gene therapy for ADA SCID. J Clin Immunol 2014;34Suppl 2:S311 [Google Scholar]
  • 118.Carbonaro Sarracino D, Shaw K, Sokolic R, et al. . Clinical Gene Therapy Trials for Adenosine Deaminase-Deficient Severe Combined Immune Deficiency (ADA-SCID). J Clin Immunol 2014;34Suppl 2:S313 [Google Scholar]
  • 119.Gaspar B, Buckland K, Rivat C, et al. . Immunological and Metabolic Correction After Lentiviral Vector Mediated Haematopoietic Stem Cell Gene Therapy for ADA Deficiency. J Clin Immunol 2014;34Suppl 2:S167 [Google Scholar]
  • 120.Scaramuzza S, Giannelli S, Ferrua F, et al. . Persistent multilineage engraftment and WASP restored expression after lentiviral mediated CD34+ cells gene therapy for the treatment of Wiskott-Aldrich Syndrome. Mol Ther 2014;22Suppl 1:S88 [Google Scholar]
  • 121.Williams DA, Update on gene therapy trials for severe combined immunodeficiency and Wiskott-Aldrich Syndrome. Hum Gene Ther 2014;25:A16 [Google Scholar]
  • 122.Shaw KL, Sokolic R, Davila A, et al. . Phase II clinical trial of gene therapy for adenosine deaminase-deficient Severe Combined Immune Deficiency (ADA SCID). Mol Ther 2014;22Suppl 1:S 107 [Google Scholar]

Articles from Human Gene Therapy are provided here courtesy of Mary Ann Liebert, Inc.

RESOURCES