Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2009 Jul 1.
Published in final edited form as: Curr Opin Hematol. 2008 Jul;15(4):375–380. doi: 10.1097/MOH.0b013e328302c807

Recent Advances in Gene Therapy for Severe Congenital Immunodeficiency Diseases

Robert Sokolic 1, Chimène Kesserwan 2, Fabio Candotti 3
PMCID: PMC2666972  NIHMSID: NIHMS90584  PMID: 18536577

Abstract

Purpose of Review

To discuss new data on safety and efficacy of the ongoing gene therapy trials for primary immune deficiencies, the first reports of new trials and the preclinical developments that are likely to be translated to the clinic in the near future.

Recent findings

Both clinical successes and severe adverse events continue to be reported in trials of gammaretroviral gene therapy for SCID-X1, ADA-SCID and CGD. Insertion site analyses of recently reported trials in all of these diseases have discovered preferential insertion in the 5’ ends of genes, including potentially dangerous genes such as proto-oncogenes, and signal transduction and proliferation genes. Preclinical work in rodent and canine models has tested novel vectors, including lentiviruses and foamy viruses.

Summary

Gene therapy for the most common forms of SCID can lead to immune reconstitution in most patients, although a minority of patients has derived minimal clinical benefit and some have suffered severe adverse events, including death. Ongoing pre-clinical work attempts to address the latter shortcomings. In the meantime, in the presence of a careful risk-benefit assessment, gene therapy remains an appropriate subject of clinical investigation.

Keywords: Primary Immunodeficiency, Gene Therapy, Clinical Trials, Insertional Oncogenesis, Insertion Site Analysis

Introduction

This review will cover recent clinical advances in gene therapy for primary immune deficiencies, including the X-linked and adenosine deaminase-deficient forms of severe combined immune deficiency (SCID-X1 and ADA-SCID), chronic granulomatous disease (CGD) and Wiskott-Aldrich Syndrome (WAS). We will briefly review the historical background to the recently published and currently ongoing trials of gene transfer for these illnesses. We will also cover the current problems facing clinical gene therapy and some of the pre-clinical work that may address these issues.

Recent advances in clinical Gene Therapy

In less than 20 years, the application of gene therapy for primary immune deficiency has gone from minimal success to a potentially completely curative procedure, albeit with substantial risks. Early gene therapy trials conducted in the 1990s for adenosine deaminase (ADA)-deficient severe combined immune deficiency (SCID)[1], chronic granulomatous disease (CGD) [2] and leukocyte adhesion deficiency type I (LAD I) [3] did not achieve long-lasting clinical benefits. However, following on technical advances achieved through extensive pre-clinical studies [4], subsequent trials culminated in the first irrefutably successful results described in the 21st century.

Successful Gene Therapy Trials for SCID-X1

The first successful clinical gene therapy trial was reported by Drs. Fischer and Cavazzana-Calvo in 2000[5]. This initial report described two patients with the X-linked form of SCID (SCID-X1) due to deficiency of the γc cytokine receptor. No myelosuppressive preparatory regimen was used. Additional data from this trial were reported in 2002 [6], 2003 [7] and 2007 [8••]. Overall, ten patients with classic SCID-X1 were treated, and achieved long-lasting immune reconstitution. Most patients derived clinical benefit. Signs and symptoms of infection resolved. T-cell counts, proliferative responses and diversity normalized. All patients had proviral integration in ~ 100% of their T-cells. Most of the successfully treated patients had normal levels of IgG, IgA and IgM. One of the two patients who did not achieve immune reconstitution was a 10-month-old boy with disseminated bacille Calmette-Guérin (BCG) infection and splenomegaly [6]. This patient ultimately required allogeneic hematopoietic cell transplantation (HCT).

The potentially curative outcome of gene therapy for SCID-X1 was confirmed by Drs. Gaspar and Thrasher in Great Britain. They reported successful reconstitution of immunity in ten patients with classic SCID-X1 [9,10••]. As of the time of the last full report, all patients showed increased lymphocyte counts, most to the normal range. T-cell function and diversity were mostly normalized as well. Six patients no longer need intravenous immunoglobulin (IVIG). Taken together, the French and British reports show, for the first time, clear efficacy of gene therapy for a human genetic disease and also point out some of the limitations of the approach, including the failure of gene therapy in two older patients (age 15–20 years) [11].

Older patients who failed transplant were the targeted population of a recent United States trial evaluating the efficacy of gene therapy for SCID-X1 as rescue therapy after a failed allogeneic HCT. In this report, a team of investigators from the National Institutes of Health, described the treatment of three patients aged 10, 11 and 14 years. After a follow-up of one to two years, only one patient was found to have improvement in T-cell number and proliferation, with increased numbers of CD45RA+ T-cells and T-cell receptor excision circles. No patient was able to discontinue IVIG, but all three have reported subjective improvements in well-being, and all have shown increased growth rates [12].

Overall, the current experience indicates that, provided sufficient numbers of transduced hematopoietic stem cells (HSC’s) are engrafted, gene therapy is a highly successful therapeutic modality for SCID-X1 infants, while its efficacy decreases in older patients.

Adverse events in the SCID-X1 trials

Unfortunately, despite successful results, the gene therapy trials for SCID-X1 highlighted the substantial risks of the procedure. About two and a half years after treatment, the fourth patient treated on the French trial developed a monoclonal lymphoproliferation caused by the insertion of the therapeutic vector near LMO2, a known T-cell oncogene. Dysregulation of LMO2 induced by the murine leukemia virus (MLV) enhancer was concluded to be the basis of the leukemic event in this patient and in a second case that occurred shortly thereafter [7,13]. Over the last five years, additional reports have described other patients with T-cell lymphoproliferations [7,1316] (and www.asgt.org, posted 12-18-07, accessed 1-29-08). Two of the patients were very young (1 and 3 months of age at the time of gene therapy), and received cell doses in excess of 10 million cells/kg. Very young age, however, was not common to all of the cases of leukemia{ [10,14] and (www.asgt.org, posted 12-18-07, accessed 1-29-08)}. Overall, as of the last reports, twenty classic SCID-X1 patients have been treated in the French and British versions of this trial. Three of these patients had poor immune reconstitution and one of these died after allogeneic hematopoietic cell transplantation. Five patients developed leukemia and one of these patients died as well [17]. Thus, the overall mortality was 10% and the overall morbidity was 30%.

The question of why five out of 20 patients treated on the French and British SCID-X1 trials developed leukemia is one of the most pressing current problems in clinical gene therapy. Some controversy exists over the relative roles of IL2RG, LMO2 and/or their combination in leukemogenicity [15,18•,19•,20,21••]. A recent report noted that, in addition to frequently integrating near the 5’ ends of genes, the vector used in the French trial preferentially integrated into common fragile sites [22••]. Because LMO2 itself is a fragile site, this finding may help to explain why targeting of this particular oncogene was seen so frequently. In addition to these considerations, it is generally accepted that the preference of retroviral vectors for integration in gene-rich regions[23,24], and particularly near the transcriptional start sites of genes has likely contributed to severe adverse events. Recent surveys of clinical samples from the British and French SCID-X1 trials [8••,10••] have confirmed this tendency, a finding that was described earlier in vitro and in experimental animal systems[2326].

Success of gene therapy for ADA-SCID

Uniquely among the primary immunodeficiencies, ADA-SCID can be treated with enzyme replacement therapy (ERT) using pegylated bovine ADA (PEG-ADA) [27]. Early trials of gene therapy for ADA-SCID [1,28,29] treated patients who, for ethical reasons, were kept on PEG-ADA. The initial attempts at targeting HSC’s resulted in low transduction efficiency [28,30,31] and low engraftment of gene-corrected cells [29].

In a landmark study published in 2002, Aiuti and colleagues treated ADA-SCID patients who did not have access to PEG-ADA with low dose busulfan prior to infusion of autologous bone marrow CD34+ cells that had been genetically corrected using a retroviral vector, with the goal of facilitating the engraftment of corrected cells [32]. The initial report showed convincingly that immunity could be restored with this approach. To date, data on twelve patients have been reported in various forms [32,33••,34•] and indicated long-term multilineage engraftment of gene-corrected cells, associated with improvement in T-cell counts and proliferation to exogenous stimuli, leading to clear clinical benefits. No serious adverse events have been observed in the trial. An extensive analysis of the pattern of retroviral integration in five patients has recently been published and showed that integrations have occurred within and/or near the LMO2, BLM, CCND2 and BCL2 genes, among others, although these events did not lead to clonal selection [33••].

The efficacy of these trials surpassed those of a concurrent collaborative trial conducted by or own group and that of Dr. Kohn at Childrens Hospital Los Angeles (CHLA). The emphasis in this trial was on vector design. Patients were kept on PEG-ADA and myelosuppressive chemotherapy was not used [35].

A Japanese trial recently reported in abstract form [36•] also did not use marrow conditioning, but involved discontinuation of PEG-ADA before reinfusion of gene-corrected cells. Patients on this trial showed slow immunological improvement compared to the Milan study.

The initial results of a British study of gene therapy for ADA-SCID that also included withdrawal of PEG-ADA as well as myelosuppressive chemotherapy were also published in 2006 and showed immune recovery manifested by increasing T-cell numbers and T-cell proliferative responses in the first treated patient [37••].

More recently, our own group has used a similar protocol with PEG-ADA withdrawal and low dose myelosuppresive chemotherapy. Four patients with ADA-SCID have been treated with this approach to date. The first treated patient was discovered after gene therapy to have had bone marrow trisomy 8 that pre-dated chemotherapy. The patient suffered from persistent myelosuppresion post-chemotherapy and ultimately required hematopoietic stem cell transplantation[38]. Two other patients have been followed for at least six months and remain off PEG-ADA treatment without opportunistic infection, and one of these is also off IVIG. The patients have shown general clinical improvement as well as improvement in T-cell counts and in T-cell proliferative responses. Finally, a fourth child recently treated remains off PEG-ADA, but otherwise has not had enough time since treatment to evaluate the outcome[39].

Recent data on gene therapy for CGD and WAS

A recent trial of gene therapy for CGD was conducted by Drs. Grez and Seger in Germany and Switzerland. The retroviral vector used in this study carries gp91phox under the control of the spleen focus forming virus long terminal repeat (LTR), a promoter that is highly active in myeloid cells. High levels of functionally corrected phagocytes were observed, resulting in reduction of infections in three treated patients. Oligoclonal expansion of gene-corrected myeloid clones was also observed in the first two treated patients. This likely was due to insertional activation of the cancer-associated genes MDS1-EVI1, PRDM16 and SETBP1 [40••,41]. Recent findings form this trial indicate that both of these patients developed monosomy 7, presumed to be related to these insertional events [42]. Clearly, these results raise serious concerns regarding the vector LTR used in this trial.

More recently, Malech and colleagues have reported on the use of genetically modified cells to treat infections in CGD, rather than attempting immune reconstitution per se [43,44]. Conditioning consisted of busulfan 10 mg/kg given over two days. One of two treated patients had resolution of multiple staphylococcal liver abscesses and retained 1% correction of myeloid cells for more than one year. The second patient was felt to have an immune-mediated rejection of the transgene product that is currently being characterized.

Finally, at the 2007 meeting of the American Society of Hematology a preliminary report was given on a trial of retroviral gene therapy for WAS [45]. Based on prior optimization in vitro [46], Dr. Klein and colleagues treated two patients, both of whom have derived clinical benefit from the procedure. Both patients’ genetic marking is increasing in T and NK cells. No evidence of adverse events or clonal dominance have been noted on this trial to this time.

Implications of Recent Clinical Results for Pre-Clinical Development of Gene Therapy

The occurrence of severe adverse events in SCID-X1 gene therapy patients [47] has stimulated extensive preclinical research efforts aimed at reducing the risk of insertional oncogenesis in future applications of gene therapy to this and other primary immune deficiencies. The tendency of gammaretrovirus vectors to integrate near transcription start sites of transcriptionally active genes [8••,10••,23,24,26,33••] and the intrinsic promoter enhancing activity of retroviral LTR’s are seen as critical risk factors. To overcome the latter, Thornhill, Thrasher, et al have developed self inactivating (SIN) gammaretroviral vectors for gene therapy of SCID-X1[48]. SIN vectors lack the enhancer sequences of retroviral LTR’s and are therefore expected to be less prone to activating promoters near the integration sites [49,50]. However, these vectors necessarily require the use of an internal promoter, and this element must also need to be devoid of significant enhancer properties if ectopic expression of genes neighboring the integration site is to be avoided.

Vectors based on lentiviruses are an appealing alternative to gammaretrovirus vectors because their LTRs have no meaningful activity in the absence of the TAT transcriptional regulator, and their pattern of integration has been shown to be less biased towards transcription start sites [23]. At least one paper [51••] suggests that lentiviral vectors are less likely than are MLV-derived vectors to integrate into sites related to cancer, cell growth or oncogenesis. For these reasons, lentiviral vectors are being developed and tested for a variety of primary immunodeficiencies, including the X-linked [52••], ADA-deficient [53•,54•] and Artemis-deficient[55•] forms of SCID, as well as CGD [56] and WAS [57•,58•].

Vectors based on foamy virus [59] offer another alternative that is being evaluated for gene therapy of primary immunodeficiencies. Foamy vectors offer a number of advantages, including the observation that foamy virus is not known to be a human pathogen [59] and that the integration pattern of foamy viruses has been described as having a less strong preference for integrating near genes [25,60••]. Foamy vectors have been developed for gene therapy of LAD I [25], a disease that has also been the object of efforts to increase the efficiency of retrovirus vectors in the canine model of the human condition [61••,62].

Despite advances made in the above-mentioned viral vectors, they all suffer from the drawback that their use implies gene addition strategies that are less desirable than genetic repair approaches. Of the various approaches to genetic repair being studied [6366], the zinc finger nuclease approach has been applied for SCID-X1 [67], as an example of a disease where the strong selective advantage of gene-corrected cells could compensate for the low efficiency that at present characterizes all gene repair methods.

Conclusions

After extensive theoretical and preclinical work, human gene therapy for SCID was translated to the clinic in 1990. Within twelve years, this form of treatment was shown to be efficacious. This rapid and successful progress can be attributed to advances in vectorology, cell culture conditions and the use of preparative myelosuppressive therapy. While still incompletely characterized, the risks of gene therapy for the primary immunodeficiencies remain high, and warrant a careful analysis of the risk-benefit ratio. Pre-clinical and clinical advances continue to be made in the safety and efficacy of gene transfer vectors and gene correction approaches. While it is likely that future,gene therapy for primary immunodeficiencies will evolve beyond the use of integrating vectors for gene addition approaches. However, integrating vectos currently remain the state of the clinical science. Carefully monitored clinical trials of gene therapy are appropriate for patients with these severe illnesses who lack of a matched related donor. In some cases, gene therapy for immune deficiency has brought clinical benefit, and can be considered to have been life-saving despite the serious complications that have been reported in some patients. Based on the documented clinical successes, further progress in the development of gene therapy for primary immune deficiency can be expected.

Acknowledgements

This work was supported by the Intramural Research Program of the National Human Genome Research Institute.

Contributor Information

Robert Sokolic, Staff Clinician, Disorders of Immunity Section, Genetics and Molecular Biology Branch, National Human Genome Research Institute, National Institutes of Health, 10 Center Drive, MSC 1451, Building 10-CRC, Room 6-3330, Bethesda, MD 20892-1451, Voice: 301-451-1498 FAX: 301-451-5408, sokolicr@mail.nih.gov.

Chimène Kesserwan, Clinical Fellow, Disorders of Immunity Section, Genetics and Molecular Biology Branch, National Human Genome Research Institute, National Institutes of Health, 10 Center Drive, MSC 1611, Building 10-CRC, Room 6-3330, Bethesda, MD 20892-1611, Voice: 301-451-3949 FAX: 301-480-3015, kesserwanc@mail.nih.gov.

Fabio Candotti, Senior Investigator, Head, Disorders of Immunity Section, Genetics and Molecular Biology Branch, National Human Genome Research Institute, National Institutes of Health, 49 Convent Drive, MSC 4442, Building 49, Room 3A04, Bethesda, MD 20892-4442, Voice: 301-435-2944 FAX: 301-480-3678, fabio@nhgri.nih.gov.

References

  • 1.Blaese RM, Culver KW, Miller AD, et al. T lymphocyte-directed gene therapy for ADA- SCID: initial trial results after 4 years. Science. 1995;270:475–480. doi: 10.1126/science.270.5235.475. [DOI] [PubMed] [Google Scholar]
  • 2.Malech HL, Maples PB, Whiting-Theobald N, et al. Prolonged production of NADPH oxidase-corrected granulocytes after gene therapy of chronic granulomatous disease. Proceedings of the National Academy of Sciences. 1997;94:12133–12138. doi: 10.1073/pnas.94.22.12133. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Bauer TR, Jr, Hickstein DD. Gene therapy for leukocyte adhesion deficiency. Current Opinion in Molecular Therapeutics. 2000;2:383–388. [PubMed] [Google Scholar]
  • 4.Kohn DB. Gene therapy using hematopoietic stem cells. Current Opinion in Molecular Therapeutics. 1999;1:437–442. [PubMed] [Google Scholar]
  • 5.Cavazzana-Calvo M, Hacein-Bey S, de Saint Basile Geneviève , et al. Gene Therapy of Human Severe Combined Immunodeficiency (SCID)-X1 Disease. Science. 2000;288:669–672. doi: 10.1126/science.288.5466.669. [DOI] [PubMed] [Google Scholar]
  • 6.Hacein-Bey-Abina S, Le Deist F, Carlier F, et al. Sustained correction of X-linked severe combined immunodeficiency by ex vivo gene therapy. New England Journal of Medicine. 2002;346:1185–1193. doi: 10.1056/NEJMoa012616. [DOI] [PubMed] [Google Scholar]
  • 7.Hacein-Bey-Abina S, Von Kalle C, Schmidt M, et al. LMO2-associated clonal T cell proliferation in two patients after gene therapy for SCID-X1. Science. 2003;302:415–419. doi: 10.1126/science.1088547. [DOI] [PubMed] [Google Scholar]
  • 8. Deichmann A, Hacein-Bey-Abina S, Schmidt M, et al. Vector integration is nonrandom and clustered and influences the fate of lymphopoiesis in SCID-X1 gene therapy. Journal of Clinical Investigation. 2007;117:2225–2232. doi: 10.1172/JCI31659. Report confirming preferential integration of the gammaretrovirus used for gene therapy of SCID-X1 at the 5' ends of genes. Differences in integration sites between the pre-infusion CD34+ cells and their T-cell progeny were also found, suggesting in vivo selection of cells containing retroviral vector integrations that provide a proliferative advantage.
  • 9.Gaspar HB, Parsley KL, Howe S, et al. Gene therapy of X-linked severe combined immunodeficiency by use of a pseudotyped gammaretroviral vector. Lancet. 2004;364:2181–2187. doi: 10.1016/S0140-6736(04)17590-9. [DOI] [PubMed] [Google Scholar]
  • 10. Schwarzwaelder K, Howe SJ, Schmidt M, et al. Gammaretrovirus-mediated correction of SCID-X1 is associated with skewed vector integration site distribution in vivo. Journal of Clinical Investigation. 2007;117:2241–2249. doi: 10.1172/JCI31661. Report from the British gene therapy trial for SCID-X1 indicating several similarities with the findings reported in reference #8. A revised frequency of common integration sites identified in this trial also suggests the existence of differences among the French and British transduction procedures.
  • 11.Thrasher AJ, Hacein-Bey-Abina S, Gaspar HB, et al. Failure of SCID-X1 gene therapy in older patients. Blood. 2005;105:4255–4257. doi: 10.1182/blood-2004-12-4837. [DOI] [PubMed] [Google Scholar]
  • 12.Chinen J, Davis J, De Ravin SS, et al. Gene therapy improves immune function in preadolescents with X-linked severe combined immunodeficiency. Blood. 2007;110:67–73. doi: 10.1182/blood-2006-11-058933. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Hacein-Bey-Abina S, von Kalle C, Schmidt M, et al. A serious adverse event after successful gene therapy for X-linked severe combined immunodeficiency. New England Journal of Medicine. 2003;348:255–256. doi: 10.1056/NEJM200301163480314. [DOI] [PubMed] [Google Scholar]
  • 14.Check E. Gene therapy put on hold as third child develops cancer. Nature. 2005;433:561–561. doi: 10.1038/433561a. [DOI] [PubMed] [Google Scholar]
  • 15.Pike-Overzet K, van der Burg M, Wagemaker G, et al. New insights and unresolved issues regarding insertional mutagenesis in X-linked SCID gene therapy. Molecular Therapy: the Journal of the American Society of Gene Therapy. 2007;15:1910–1916. doi: 10.1038/sj.mt.6300297. [DOI] [PubMed] [Google Scholar]
  • 16.Gray JT, Hacein-Bey-Abina S, Cavazzana-Calvo M, Fischer A. The Effect of the B2 Mutation on Hematopoietic Reconstitution in the French X-SCID Gene Therapy Trial. Mol Ther. 2006;13:S254–S255. [Google Scholar]
  • 17.Qasim W, Gaspar HB, Thrasher AJ. Update on clinical gene therapy in childhood. Archives of Disease in Childhood. 2007;92:1028–1031. doi: 10.1136/adc.2006.108787. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Pike-Overzet K, de Ridder D, Weerkamp F, Baert MR, et al. Gene therapy: is IL2RG oncogenic in T-cell development? Nature. 2006;443:E5–E7. doi: 10.1038/nature05218. This brief communication presents data from mouse-human hybrid fetal thymus culture to refute the contention that IL2RG transgenesis in and of itself is leukemogenic.
  • 19.Thrasher AJ, Gaspar HB, Baum C, et al. Gene therapy: X-SCID transgene leukaemogenicity. Nature. 2006;443:E5–E6. doi: 10.1038/nature05219. discussion E6–E7. [DOI] [PubMed] [Google Scholar]
  • 20. Dave UP, Jenkins NA, Copeland NG. Gene therapy insertional mutagenesis insights. Science. 2004;303:333. doi: 10.1126/science.1091667. This brief communication discusses the results of preclinical murine studies performed by French, British and united States groups, and the relevance of these studies to the adverse events seen in the SCID-X1 clinical gene therapy trial.
  • 21. Pike-Overzet K, de Ridder D, Weerkamp F, et al. Ectopic retroviral expression of LMO2, but not IL2Rgamma, blocks human T-cell development from CD34+ cells: implications for leukemogenesis in gene therapy. Leukemia. 2007;21:754–763. doi: 10.1038/sj.leu.2404563. In this paper the authors seek to differentiate between the ability of LMO2 and IL2RG to cause derangements of T-cell development and provide some insight into the relative contributions of each of these genes to the severe adverse events noted in the SCID-X1 clinical gene therapy trial.
  • 22. Bester AC, Schwartz M, Schmidt M, et al. Fragile sites are preferential targets for integrations of MLV vectors in gene therapy. Gene Therapy. 2006;13:1057–1059. doi: 10.1038/sj.gt.3302752. The authors demonstrate that LMO2 is located within the FRA11E fragile site, and furthermore that integrations in the SCID-X1 trials were represented in fragile sites in a higher percentage than would be expected by chance. This provides another mechanism to explain why LMO2 integrations were repeatedly seen in the leukemias that occurred on the French and British trials.
  • 23.Mitchell RS, Beitzel BF, Schroder ARW, et al. Retroviral DNA Integration: ASLV, HIV, and MLV Show Distinct Target Site Preferences. PLoS Biology. 2004;2:e234. doi: 10.1371/journal.pbio.0020234. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Wu X, Li Y, Crise B, Burgess SM. Transcription Start Regions in the Human Genome Are Favored Targets for MLV Integration. Science. 2003;300:1749–1751. doi: 10.1126/science.1083413. [DOI] [PubMed] [Google Scholar]
  • 25.Bauer TR, Jr, Hai M, Adler RL, et al. Reduced Genotoxic Risk Using Foamy Viral Vectors for the Treatment of Canine Leukocyte Adhesion Deficiency. Blood. 2007;110:3744. [Google Scholar]
  • 26.Hematti P, Hong B-K, Ferguson C, et al. Distinct Genomic Integration of MLV and SIV Vectors in Primate Hematopoietic Stem and Progenitor Cells. PLoS Biology. 2004;2:e423. doi: 10.1371/journal.pbio.0020423. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Hershfield MS, Buckley RH, Greenberg ML, et al. Treatment of adenosine deaminase deficiency with polyethylene glycol-modified adenosine deaminase. N Engl J Med. 1987;316:589–596. doi: 10.1056/NEJM198703053161005. [DOI] [PubMed] [Google Scholar]
  • 28.Bordignon C, Notarangelo LD, Nobili N, et al. Gene therapy in peripheral blood lymphocytes and bone marrow for ADA- immunodeficient patients. Science. 1995;270:470–475. doi: 10.1126/science.270.5235.470. [DOI] [PubMed] [Google Scholar]
  • 29.Hoogerbrugge PM, van Beusechem VW, Fischer A, et al. Bone marrow gene transfer in three patients with adenosine deaminase deficiency. Gene Therapy. 1996;3:179–183. [PubMed] [Google Scholar]
  • 30.Kohn DB, Weinberg KI, Nolta JA, et al. Engraftment of gene-modified umbilical cord blood cells in neonates with adenosine deaminase deficiency. Nature Medicine. 1995;1:1017–1023. doi: 10.1038/nm1095-1017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Kohn DB, Hershfield MS, Carbonaro D, et al. T lymphocytes with a normal ADA gene accumulate after transplantation of transduced autologous umbilical cord blood CD34+ cells in ADA-deficient SCID neonates. Nat Med. 1998;4:775–780. doi: 10.1038/nm0798-775. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.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: 10.1126/science.1070104. [DOI] [PubMed] [Google Scholar]
  • 33. Aiuti A, Cassani B, Andolfi G, et al. Multilineage hematopoietic reconstitution without clonal selection in ADA-SCID patients treated with stem cell gene therapy. Journal of Clinical Investigation. 2007;117:2233–2240. doi: 10.1172/JCI31666. This is the first reported insertion site analysis for the Italian ADA-SCID trial. Again, the clinically used MLV-based vector is shown to preferentially integrate into the 5' ends of genes. Although the LMO2 locus was a common integration site, no clonal dominance was demonstrated. Although the precise implications for this trial and/or other MLV-based gene therapy trials is not clear, there are both worrisome and reassuring aspects of the data.
  • 34. Aiuti A, Cassani B, Benninghoff U, et al. Long-Term Safety and Correction of Immune and Metabolic Defects in ADA-SCID Children Treated with Gene Therapy. Human Gene Therapy. 2007;18:941–942. This is the latest report on the original Italian ADA-SCID gene therapy trial. The authors note that 12 patients have been treated and none have developed a dominant clonal proliferation.
  • 35.Candotti F, Podsakoff G, Schurman S, et al. Corrective gene transfer into bone marro CD34+ cells for Adenosine Deaminase (ADA) deficiency: Results in four patients after one year of follow-up. Mol Ther. 2003;7:S448–S449. [Google Scholar]
  • 36. Ariga T. Hematopoietic stem sell (HSC) gene therapy for two patients with adenosine deaminase (ADA) deficiency without myelopreparative conditioning: a suggestion for the optimal protocol of HSC gene therapy for ADA. Jounal of Gene Medicine. 2006;8:1439. This abstract is a report of the Japanese trial of HSC gene therapy of ADA-SCID after withdrawal of PEG-ADA and without myelosuppression. Partial reconstitution of immunity is demonstrated in two patients.
  • 37. Gaspar HB, Bjorkegren E, Parsley K, et al. Successful reconstitution of immunity in ADA-SCID by stem cell gene therapy following cessation of PEG-ADA and use of mild preconditioning. Mol Ther. 2006;14:505–513. doi: 10.1016/j.ymthe.2006.06.007. This is the first report of the British trial of ADA-SCID gene therapy after withdrawal of PEG-ADA and myelosuppression with melphalan. This paper is the first confirmation of the efficacy of gene therapy in this disease first reported by the Italian group, and provides an alternative conditioning regimen.
  • 38.Engel BC, Podsakoff GM, Ireland JL, et al. Prolonged pancytopenia in a gene therapy patient with ADA-deficient SCID and trisomy 8 mosaicism: a case report. Blood. 2007;109:503–506. doi: 10.1182/blood-2006-06-031476. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Sokolic R, Podsakoff G, Muul L, et al. Comparative Results of Gene Therapy for Adenosine Deaminase Deficiency with or without PEG-ADA Withdrawal and Myelosuppressive Chemotherapy. Blood. 2007;110:501. [Google Scholar]
  • 40. 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. Nature Medicine. 2006;12:401–409. doi: 10.1038/nm1393. [see comment]. This is the first report of successful gene therapy for a myeloid disease and contains the initial characterization of the expansion of myeloid clones mediated by the retroviral activation of known oncogenes.
  • 41.Ott MG, Seger R, Stein S, et al. Advances in the treatment of Chronic Granulomatous Disease by gene therapy. Current Gene Therapy. 2007;7:155–161. doi: 10.2174/156652307780859044. [DOI] [PubMed] [Google Scholar]
  • 42.Williams DA. An International Conversation on Stem Cell Gene Therapy. Mol Ther. 2007;15:2058–2059. doi: 10.1038/sj.mt.6300358. [DOI] [PubMed] [Google Scholar]
  • 43.Kang EM, Linton G, Theobald N, et al. Transplantation of Genetically Modified Cells for Treatment of Infection in Patients with X-Linked Chronic Granulomatous Disease (X-CGD) Molecular Therapy: the Journal of the American Society of Gene Therapy. 2007;15:S295. [Google Scholar]
  • 44.Kang EM, Linton G, Theobald N, et al. Treatment of Infections in Patients with X-Linked Chronic Granulomatous Disease (XCGD) with Gene Therapy; ASH Annual Meeting Abstracts; 2007. p. 504. [Google Scholar]
  • 45.Boztug K, Dewey RA, Diez IA, et al. Hematopoietic Stem Cell Gene Therapy for Wiskott-Aldrich Syndrome. Blood. 2007;110:502. [Google Scholar]
  • 46.Boztug K, Baumann U, Ballmaier M, et al. Large granular lymphocyte proliferation and revertant mosaicism: two rare events in a Wiskott-Aldrich syndrome patient. Haematologica. 2007;92:e43–e45. doi: 10.3324/haematol.11222. [DOI] [PubMed] [Google Scholar]
  • 47.Hacein-Bey-Abina S, von Kalle C, Schmidt M, et al. A serious adverse event after successful gene therapy for X-linked severe combined immunodeficiency. New England Journal of Medicine. 2003;348:255–256. doi: 10.1056/NEJM200301163480314. [see comment]. [DOI] [PubMed] [Google Scholar]
  • 48.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: 10.1038/sj.mt.6300393. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Modlich U, Bohne J, Schmidt M, et al. Cell-culture assays reveal the importance of retroviral vector design for insertional genotoxicity. Blood. 2006;108:2545–2553. doi: 10.1182/blood-2005-08-024976. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Schambach A, Galla M, Maetzig T, et al. Improving Transcriptional Termination of Self-inactivating Gamma-retroviral and Lentiviral Vectors. Mol Ther. 2007;15:1167–1173. doi: 10.1038/sj.mt.6300152. [DOI] [PubMed] [Google Scholar]
  • 51. Cattoglio C, Facchini G, Sartori D, et al. Hot spots of retroviral integration in human CD34+ hematopoietic cells. Blood. 2007;110:1770–1778. doi: 10.1182/blood-2007-01-068759. [see comment]. This study compares common insertion sites of both gammaretrovirus and lentivirus vectors in human CD34+ cells. The authors show that gammaretroviruses are more likely than lentiviruses to integrate into proto-oncogenes, cancer associated genes and growth-regulating genes.
  • 52. Zhang F, Thornhill SI, Howe SJ, et al. Lentiviral vectors containing an enhancer-less ubiquitously acting chromatin opening element (UCOE) provide highly reproducible and stable transgene expression in hematopoietic cells. Blood. 2007;110:1448–1457. doi: 10.1182/blood-2006-12-060814. This paper demonstrates the use of a UCOE to mediate transcription of IL2RG in the murine model of SCID-X1.
  • 53. Carbonaro DA, Jin X, Petersen D, et al. In vivo transduction by intravenous injection of a lentiviral vector expressing human ADA into neonatal ADA gene knockout mice: a novel form of enzyme replacement therapy for ADA deficiency. Molecular Therapy: the Journal of the American Society of Gene Therapy. 2006;13:1110–1120. doi: 10.1016/j.ymthe.2006.02.013. This paper uses in vivo injection of virus as an alternative to ex vivo transduction of hematopoietic cells. Cells in the liver and lungs were the most commonly transduced and mediated restoration of immune function and extended survival.
  • 54. 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: 10.1182/blood-2006-05-023507. This is the first demonstration of the utility of lentivectors in preclinical experiments in a mouse model of ADA-SCID.
  • 55. Mostoslavsky G, Fabian AJ, Rooney S, et al. Complete correction of murine Artemis immunodeficiency by lentiviral vector-mediated gene transfer. PNAS. 2006;103:16406–16411. doi: 10.1073/pnas.0608130103. This is the first demonstration that lentivectors can be used in the treatment of artemis-deficient SCID.
  • 56.Naumann N, De Ravin SS, Choi U, et al. Simian immunodeficiency virus lentivector corrects human X-linked chronic granulomatous disease in the NOD/SCID mouse xenograft. Gene Therapy. 2007;14:1513–1524. doi: 10.1038/sj.gt.3303010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Dupre 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. Human Gene Therapy. 2006;17:303–313. doi: 10.1089/hum.2006.17.303. This paper demonstrates in a murine model that a lentiviral vector wherein WASp is under the control of its native promoter can correct WASp deficiency in all lineages in vivo.
  • 58. Charrier S, Dupre L, Scaramuzza S, et al. Lentiviral vectors targeting WASp expression to hematopoietic cells, efficiently transduce and correct cells from WAS patients. Gene Therapy. 2007;14:415–428. doi: 10.1038/sj.gt.3302863. This paper examined a different aspect of the correction of WAS with a SIN lenitviral vector using a tissue specific promoter.
  • 59.Rethwilm A. Foamy virus vectors: an awaited alternative to gammaretro-and lentiviral vectors. Current Gene Therapy. 2007;7:261–271. doi: 10.2174/156652307781369092. [DOI] [PubMed] [Google Scholar]
  • 60. Beard BC, Keyser KA, Trobridge GD, et al. Unique integration profiles in a canine model of long-term repopulating cells transduced with gammaretrovirus, lentivirus, or foamy virus. Human Gene Therapy. 2007;18:423–434. doi: 10.1089/hum.2007.011. This paper compared integration sites of three different types of vectors in canine repopulating cells. Notably, all three virus types integrated within proto-oncogenes, although the genes differed from vector to vector.
  • 61. Bauer TR, Jr, Hai M, Tuschong LM, et al. Correction of the disease phenotype in canine leukocyte adhesion deficiency using ex vivo hematopoietic stem cell gene therapy. Blood. 2006;108:3313–3320. doi: 10.1182/blood-2006-03-006908. This is the first paper to use foamy virus vector to treat a clinically relevant large animal that models a human disease and represents a significant milestone in the development of this system for human gene therapy.
  • 62.Sokolic RA, Bauer TR, Tuschong LM, et al. A selectable bicistronic retroviral vector corrects the molecular defect in a cell line derived from a patient with leukocyte adhesion deficiency. Biology of Blood and Marrow Transplantation. 2006;14:27–28. [Google Scholar]
  • 63.Moehle EA, Rock JM, Lee Y-LJ, et al. Targeted gene addition into a specified location in the human genome using designed zinc finger nucleases. Proceedings of the National Academy of Sciences. 2007;104:3055–3060. doi: 10.1073/pnas.0611478104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Calos MP. The phiC31 integrase system for gene therapy. Current Gene Therapy. 2006;6:633–645. doi: 10.2174/156652306779010642. [DOI] [PubMed] [Google Scholar]
  • 65.Radecke S, Radecke F, Peter I, Schwarz K. Physical incorporation of a single-stranded oligodeoxynucleotide during targeted repair of a human chromosomal locus. Journal of Gene Medicine. 2006;8:217–228. doi: 10.1002/jgm.828. [DOI] [PubMed] [Google Scholar]
  • 66.Ren X, Tahimic CG, Katoh M, et al. Human artificial chromosome vectors meet stem cells: new prospects for gene delivery. Stem Cell Reviews. 2006;2:43–50. doi: 10.1007/s12015-006-0008-9. [DOI] [PubMed] [Google Scholar]
  • 67.Urnov FD, Miller JC, Lee Y-L, et al. Highly efficient endogenous human gene correction using designed zinc-finger nucleases. Nature. 2005;435:646–651. doi: 10.1038/nature03556. [DOI] [PubMed] [Google Scholar]

RESOURCES