Download as pdf or txt
Download as pdf or txt
You are on page 1of 8

Review

Gene Therapy

Gene therapy for severe combined


immunodeficiencies
H Bobby Gaspar† & Adrian J Thrasher
1. Introduction †Institute of Child Health, Molecular Immunology Unit, 30 Guilford Street, London, WC1N 1EH, UK

2. Molecular basis of severe


Severe combined immune deficiencies (SCIDs) are a group of monogenic
combined immunodeficiency
diseases resulting in profound disturbances of lymphocyte development and
3. Haematopoietic stem cell
function. Affected individuals are prone to life-threatening infections and
transplantation for severe
without treatment do not survive beyond the first year of life. Haematopoi-
combined immunodeficiency
etic stem cell transplantation from a well-matched donor offers high rates
Expert Opin. Biol. Ther. Downloaded from informahealthcare.com by University of Guelph on 05/14/13

4. Enzyme replacement therapy of survival, but in the absence of a suitable matched donor, parental haplo-
for ADA-SCID
identical transplants are associated with greater complications, lower
5. Gene therapy for SCID-X1 success rates and in some instances poor long-term immune recovery.
6. Gene therapy for ADA-SCID Alternative therapeutic options based on correction of the defective gene
7. Gene therapy for other severe by retroviral gene delivery have been used to correct X-linked SCID
combined immunodeficiencies (SCID-X1) and adenosine deaminase-deficient SCID (ADA-SCID). A number
of clinical trials have established that ex vivo gene transfer into haemato-
8. Insertional mutagenesis and
poietic progenitor cells allows effective recovery of immune defects and
risks of gene therapy
that gene therapy can offer a successful alternative to transplantation. The
9. Expert opinion and conclusion:
development of leukaemia as a result of insertional mutagenesis in one trial
future prospects for severe
For personal use only.

of gene therapy for SCID-X1 has raised concerns regarding the toxicity of
combined immunodeficiency
retroviral vector-based gene delivery. These side effects are now being
gene therapy
studied in detail and measures to prevent such events through alternative
vectors delivery systems are in development at present.

Keywords: adenosine deaminase, gamma chain, gene therapy, insertional mutagenesis,


retroviral vector, severe combined immunodeficiency

Expert Opin. Biol. Ther. (2005) 5(9):1175-1182

1. Introduction

The most severe forms of primary immunodeficiency are known as severe combined
immunodeficiencies (SCIDs). These are a group of diseases in which T lymphocyte
development and in some cases function are severely disrupted and associated with
diverse disorders of development and functionality of B lymphocytes and natural
killer (NK) cells [1]. Affected infants often become seriously unwell in the first few
months of life with opportunistic infections, chronic diarrhoea and failure to thrive,
and without treatment most will die in the first year of life. To date, the mainstays of
treatment have been supportive care, antibiotic therapy and immunoglobulin replace-
ment, followed in almost all cases by allogeneic haematopoietic stem cell transplanta-
tion (HSCT) [2,3]. Over the past 20 years, improved understanding of the molecular
For reprint orders, please
contact: basis of these conditions and advances in gene transfer technology have resulted in the
reprints@ashley-pub.com development of successful gene therapy strategies for two forms of SCID.

2. Molecular basis of severe combined immunodeficiency

In recent years the genetic basis of almost three-quarters of all primary immune
deficiencies have been established [4]. X-linked SCID (SCID-X1) accounts for
Ashley Publications
www.ashley-pub.com ∼ 40 – 50% of all SCIDs and is caused by mutations in the gene encoding the
common cytokine receptor gamma chain (γc). This is a subunit of the cytokine

10.1517/14712598.5.9.1175 © 2005 Ashley Publications Ltd ISSN 1471-2598 1175


Gene therapy for severe combined immunodeficiencies

receptor complex for interleukins (ILs) 2, 4, 7, 9, 15 and myelosuppressive conditioning. Under these circumstances,
21 [5,6]. In the absence of γc signalling, many aspects of tissue damage caused by myeloablative chemotherapy is
immune cell development and function are compromised. avoided and mature T cells from the unmanipulated graft
The classical immunophenotype of SCID-X1 is the absence provides rapid protection from viral infection. Matched
of T and NK cells, and persistence of dysfunctional B cells unrelated donor transplants are increasingly used in major
(T-B+NK- SCID) [7], although individuals with partial T cell centres, and with the advent of reduced intensity conditioning,
development have been identified. A much rarer autosomal results are steadily improving and in some cases are equivalent
recessive form of T-B+NK- SCID arises from mutations in to those achieved using matched sibling donors [13,14].
Janus kinase (JAK)3, an intracellular tyrosine kinase that is For a significant proportion of patients, a well-matched
activated through γc binding and subsequently activates the related or unrelated donor is unavailable and in these cases
transcription factor signal transducer and activator of transplant can be undertaken from a mismatched (halploidenti-
transcription 5 [8]. cal) parental donor. Host/donor human leukocyte antigen
Deficiency of adenosine deaminase (ADA) accounts for up disparity necessitates rigorous depletion of donor T cells to
Expert Opin. Biol. Ther. Downloaded from informahealthcare.com by University of Guelph on 05/14/13

to 20% of SCID cases [9]. The ADA gene maps to 20q13.11 avoid graft-versus-host disease, and the donor graft is rich in
and is an enzyme that is expressed in all tissues of the body, haematopoetic progenitor cells, but lacks mature T cell popula-
although at variable levels, with the highest activity in the tions. Thus, T cell immune recovery is delayed for 6 months or
thymus and lymphoid tissues. ADA plays an essential role in more during which time the recipient remains susceptible to
purine metabolite salvage pathways, and an absence of ADA viral infection. Controversy remains as to whether myeloblation
activity results in the accumulation of the substrates deoxy- with chemotherapy is required prior to the return of donor
adenosine (dAdo) and deoxyadenosinetriphosphate (dATP), cells. The lack of chemotherapy avoids short-term tissue
which through a variety of mechanisms lead to inhibition of toxicity, but there is an increased risk of rejection and most
DNA synthesis, impaired cell division and apoptosis. These probably poor engraftment of early donor haematopoietic
defects have a profound effect on lymphocyte development progenitors leading to continuing humoral defects and
and a T-B-NK- is seen in the most severely affected infants impaired long-term immune reconstitution. Nevertheless, the
For personal use only.

with preservation of NK cell numbers in some cases. Most overall survival for haploidentical transplants with or without
ADA-SCID patients (85 – 90%) present in the first year of chemotherapy is in the order of 70 – 80% for SCID-X1 [2,3],
life, although delayed presentation as a result of residual ADA but may be significantly less for other forms of SCID
activity is seen in a minority of cases. (ADA-SCID, 29% 3-year survival; T-B-NK+ SCID, 32% 3-year
Defects of antigen receptor gene rearrangement, causing a survival) [2,3,15]. Therefore, there is clearly a need for an alterna-
T-B-NK+ phenotype, account for most other cases of SCID. tive to haploidentical transplant to minimise toxicity, enhance
The failure to produce functional T or B cell receptors arises long-term immune recovery and improve overall survival.
either due to mutations in the recombinase activating genes
(RAG 1 and 2) or the Artemis gene [10,11]. These encode 4. Enzyme replacement therapy for ADA-SCID
lymphoid cell-specific factors required during the recombina-
tion of variable, diverse and junctional (VDJ) regions of An alternative modality of treatment for the ADA-deficient
antigen-specific receptor genes. Approximately 10% of form of SCID (ADA-SCID) is exogenous enzyme replace-
T-B-NK+ SCIDs remain undiagnosed at a molecular level, ment with polyethylene glycol-conjugated bovine ADA
suggesting that other genes involved in VDJ recombination (PEG-ADA). Regular intramuscular injections of PEG-ADA
may be involved. Table 1 details the molecular basis of SCID result in rapid systemic detoxification, with reduction of dAdo
disorders and highlights the lymphocyte subset pattern and dATP to near normal levels within a few weeks of starting
usually associated with each condition. therapy. Immune recovery follows and although no formal
data exists, reports suggest that ∼ 70% of children show an
3.Haematopoietic stem cell transplantation improvement in lymphocyte counts to near normal
for severe combined immunodeficiency levels [16,17]. Of these 50% remain on immunoglobulin
replacement due to continued humoral impairment. The
Until recently, HSCT has been the only curative therapy for long-term prognosis for children on PEG-ADA without any
SCID. Data on transplant outcome is now available from a corrective procedure being undertaken is unclear (M Hersh-
wide number of sources and centres, the largest data source field, ESID 2002). PEG-ADA has been used in conjunction
being the combined efforts of a European Stem Cell Trans- with gene therapy although this may have had a detrimental
plantation registry [2,12]. If a genotypically matched family effect on the success of initial clinical trials (see section below).
donor is available, bone marrow transplantation is a highly
successful procedure with recent results indicating a long-term 5. Gene therapy for SCID-X1
survival rate of ∼ 90% for all forms of SCID. The high
survival rates are partly due to the fact that the absence of Many incremental advances in gene transfer technology have
T and/or NK cells allows engraftment in the absence of now been translated into successful gene therapy for SCID-X1.

1176 Expert Opin. Biol. Ther. (2005) 5(9)


Gaspar & Thrasher

Table 1. Severe combined immunodeficiencies.

Disorder Mutated gene Molecular defect Phenotype


SCID-X1 Common γc Absence of functional receptors T-B+NK-
for IL-2, 4, 7, 9, 15 and 21
JAK-3 deficiency JAK3 Defect of signalling via IL-2, 4, 7, T-B+NK-
9, 15 and 21
IL-7 receptor deficiency IL-7 receptor α Absence of IL-7 receptor α T-B+NK+
RAG 1,2 deficiency RAG1 and 2 Defective VDJ recombination T-B-NK+
Artemis Artemis Defective VDJ recombination; T-B-NK+
radiation sensitivity
Adenosine deaminase deficiency ADA Block in purine salvage T-B-NK+/-
Expert Opin. Biol. Ther. Downloaded from informahealthcare.com by University of Guelph on 05/14/13

metabolism
T cell receptor deficiencies CD3 γεζδ Defective T cell signalling T-B+NK+
CD45 deficiency CD45 Abnormal T cell signalling T+B+NK+
γc: Cytokine receptor gamma chain; ADA: Adenosine deaminase; IL: Interleukin; JAK: Janus kinase; NK: Natural killer; RAG: Recombinase activating gene;
SCID: Severe combined immunodeficiency; VDJ: Variable, diverse and junctional.

These have included activation of cells with high concentra- system was also restored, maybe not quite as effectively, but to a
tions of cytokines (thereby making them susceptible to sufficient degree that discontinuation of immunoglobulin
gammaretrovirus vector-mediated gene transfer), and trans- therapy was possible in a number of patients.
duction in containers coated with a recombinant fibronectin The authors have also initiated a similar study, using an
fragment (RetroNectin™) that is believed to facilitate MFG-based retroviral vector packaged in PG13 cells and
For personal use only.

co-localisation of virus particle and the target cell [18]. In the pseudotyped with a gibbon ape leukaemia virus (GALV) enve-
first landmark study, a conventional amphotropic retroviral lope [21]. Patients who lacked a matched sibling or unrelated
vector encoding the human γc cDNA (regulated by Moloney donor were enrolled and similar to the French study, autolo-
murine leukaemia virus long-terminal repeat sequences), was gous CD34+ cells were purified and transduced in a 96-h
used to transduce autologous CD34+ cells (separated by transduction protocol and returned to the patient without the
conventional magnetic bead technology from a bone marrow use of cytoreductive conditioning. To date, seven children (six
harvest). The cells were reinfused into the patients in the of whom were under the age of 1 year and one aged 3 years)
absence of preconditioning. have been enrolled. In all cases there has been evidence of
The results obtained from the first five patients have been immune recovery with kinetics of T and NK cell recovery
reported in the scientific literature [19,20]. To date, 11 infants in similar to the previous study. Recovery into the normal range
total have been treated, with good immunological reconstitution has been seen in 3 patients, all of whom have discontinued
in the majority of patients. One child showed no evidence in the prophylactic antibiotic and antibody replacement therapy. In
periphery of genetically modified cells and probably seques- two patients immune recovery is suboptimal and patients
trated the graft into an enlarged spleen. A further patient had remain on prophylactic treatment, and in the last two patients
poor immune recovery, in this case due to a low genetically with the shortest follow-up, there is continued improvement of
modified cell dose (< 1 × 106 γc transduced CD34+ cells). Both immune parameters. Clinically, all patients have cleared viral
these patients subsequently underwent a successful unrelated infections, demonstrate improved growth parameters and are
donor HSCT. In nearly all patients, NK cells appeared between living at home with no restrictions on socialisation or contact.
2 and 4 weeks after infusion of cells, followed by new thymic The levels of gene marking in all lineages are also similar in
T lymphocyte emigrants at 10 – 12 weeks. With some variation, both studies. All T cells show evidence of transgene integra-
the number and distribution of these T cells normalised rapidly tion with a copy number of 1 – 2 transgene copies per cell.
(more rapidly than observed following haploidentical transplan- Persistent long-term marking in myeloid cells (between 0.1
tation). Functional analysis in terms of proliferative responses to and 1%) suggests that long-lived stem or progenitor cells have
mitogenic, T cell receptor (TCR) and specific antigen stimula- been successfully transduced. It is difficult to know whether
tion was normal. By phenotypic analysis of TCRVβ usage and thymopoiesis arises from late prethymic T cell precursors or
TCR spectratyping, a complex and diverse T cell population from less committed progenitors that arise earlier in the hier-
was demonstrated. Analysis of naive T cell markers, CD45RA+ archy of haematopoietic stem cells. This will have important
and of T cell receptor excision circles (TRECs – by-products of implications for the longevity of T cell reconstitution and can
TCR rearrangement and, thus, surrogate markers of thymic really only be resolved by longitudinal study of naive T cell
activity) suggests increased thymic activity and the emergence of production, and by isolation of common integration sites
thymus educated T lymphocytes. Functionality of the humoral between myeloid and lymphoid populations. Ultimately, the

Expert Opin. Biol. Ther. (2005) 5(9) 1177


Gene therapy for severe combined immunodeficiencies

longevity of functional reconstitution can only be determined PEG-ADA [30]. In this individual, who 10 years after gene
by clinical monitoring. therapy had 1 – 3% gene transduced T cells, the cessation of
Two older patients (aged 20 years and 15 years), one in each PEG-ADA treatment led to a rapid increase in the proportion
of the above studies, were also treated and in both cases, despite of gene-modified T cells, eventually reaching nearly 100% of
effective gene transfer to bone marrow CD34+ cells, there was all T lymphocytes. Absolute CD3+ T cell counts also
no evidence of immune recovery in response to gene therapy increased and T cell proliferative responses were restored.
and only limited and transient presence of the transgene was These data suggest that significant gene marking can be
observed [22]. The reasons for failure are difficult to define, but achieved by retroviral gene transfer, but that the withdrawal
there may be intrinsic host-dependent restrictions to efficacy. of enzyme replacement is a prerequisite for the proliferation
In particular, in older individuals in whom there may have of gene marked T cells. Clinical observations also highlight
been thymic involution over many years, there is likely to be a the systemic nature of ADA deficiency [31,32] and suggest
limitation to initiation of normal thymopoiesis. In such cases, that systemic detoxification by high levels of engraftment of
early intervention is likely to achieve greater success. gene-modified cells in all haematopoietic lineages may be
Expert Opin. Biol. Ther. Downloaded from informahealthcare.com by University of Guelph on 05/14/13

beneficial. Two studies have now incorporated significant


6. Gene therapy for ADA-SCID changes into ADA-SCID gene therapy protocols to achieve
these goals. In both trials, patients received a mild
The first human gene therapy studies were conducted on non-myeloablative conditioning regimen prior to the infu-
patients with ADA deficiency in the early 1990s [23-26]. sion of gene-modified cells, thereby allowing engraftment of
Despite continued and in some cases significant presence of a greater number of modified cells (PEG-ADA was either
the transgene in patient cells over many years, it is generally not used or withdrawn prior to treatment). In the first of
agreed that these initial studies were unsuccessful in correcting these trials, patients who had not previously received
the immune defect in ADA-SCID. A major factor was the PEG-ADA were treated with Busulphan at a dose of 4 mg/kg
continued use of PEG-ADA enzyme replacement therapy, prior to return of gene transduced autologous CD34+
which in itself improved immune function, but may also have cells [33]. Five children have now been treated and in all cases
For personal use only.

abrogated the survival advantage of gene-modified cells. Over there has been substantial reconstitution of T, B and NK cell
a decade on, detailed analysis of the patients originally treated numbers; although similar to the SCID-X1 studies, patients
provides important information about the longevity and effi- receiving low levels (< 1 × 106/kg) of gene-modified CD34+
cacy of gene transfer [27]. In one patient treated > 10 years ago cells showed decreased benefit. Molecular analysis shows a
by repeated gammaretroviral vector-mediated ADA gene diverse TCR repertoire and an increase in TREC activity
transfer into stimulated peripheral blood lymphocytes, there levels, indicating the successful engraftment of prethymic
is > 10% gene marking in peripheral blood mononuclear cells progenitor populations. Lineage-specific transgene analysis
(PBMC), and ADA activity in PBMCs remains at ∼ 25% of by quantitative polymerase chain reaction shows high-level
normal. In a later study, umbilical cord blood CD34+ cells marking in T, NK and B cells, and persistence of
harvested from antenatally diagnosed ADA-SCID patients gene-modified granulocytes, monocytes and megakarocytes
were transduced and reinfused in the first week of life. Again at levels between 5 – 20%, again suggesting that multipotent
little clinical benefit was seen, and the level of gene marking progenitors have engrafted. In a second study using an opti-
was low (1 – 10% of T lymphocytes) in all three children [28]. mised GALV pseudotyped gammaretroviral vector, one
Recent clonal integration analysis using linear amplification patient has been treated following cessation of PEG-ADA
method-polymerase chain reaction (LAM-PCR) demonstrates one month before gene therapy and using a single dose of
that transgene-containing T lymphocytes are monoclonal or Melphalan (140 mg/m2) preconditioning before the return
oligoclonal (with 1 – 5 different integration sites) > 8 years of autologous gene-modified cells (Gaspar et al.,
after gene therapy, and that single prelymphoid clones ASGT 2005). T, B and NK cell recovery with metabolic
contribute between 25 and 100% of genetically corrected correction of dATP levels have now been sustained for
lymphocytes, whereas marking in other lineages is > 1 year following the procedure. The results from both
negligible [29]. These findings clearly demonstrate that human studies are extremely encouraging. The key to correction of
T cells have a lifespan in the peripheral circulation of the immunological and metabolic abnormalities in
> 10 years, and also that transgenes regulated by gammaretro- ADA-SCID appears to be the delivery of large amounts of
viral sequences continue to be expressed in peripheral T cells ADA enzyme whether exogenously in the form of
and resist in vivo silencing. PEG-ADA, or intracellularly as gene-modified cells. The use
The continuation of PEG-ADA throughout these early of conditioning may facilitate the initial engraftment of a
studies almost certainly compromised the efficient engraft- greater number of gene-modified cells. Certainly if immune
ment of transduced cells. This assumption is supported by an function in these patients is sustained, and further patients
important observation in one patient participating in another show a similar safety profile and immune response, this
gene therapy study who demonstrated that a survival strategy holds great promise for ADA-SCID and potentially
advantage for gene transduced cells did exist in the absence of other haematopoietic conditions.

1178 Expert Opin. Biol. Ther. (2005) 5(9)


Gaspar & Thrasher

Table 2. Clinical trials of gene therapy for SCID (previous and current).

Defect Cellular target Conditioning Vector/envelope No. of patients


*Common γc BM CD34 None MFG-ampho 12
*Common γc BM CD34 None MFG-GALV 8
*Common γc PBSC CD34 None MFG-GALV 2
ADA Peripheral T cells None LASN-ampho 2
ADA BM/peripheral lymphocytes None DCA-ampho 7
ADA UCB CD34+ None LASN-ampho 3
ADA BM CD34+ None LgAL-ampho 3
ADA Peripheral T cells None LASN-ampho 1
Expert Opin. Biol. Ther. Downloaded from informahealthcare.com by University of Guelph on 05/14/13

*ADA BM CD34+ Busulphan 4 mg/kg GIADA1-ampho 5


*ADA BM CD34+ Melphalan 140 mg/m2 SFFV-GALV 1
*ADA BM CD34+ None GCsap-M-GALV 4
MND-ADA-GALV
*ADA BM CD34+ None GCsap-M-GALV 2
JAK-3 BM CD34+ None MSCV 1
*Currently active study.
γc: Cytokine receptor gamma chain; ADA: Adenosine deaminase; Ampho: Amphotropic envelope; BM: Bone marrow; GALV: Gibbon ape leukaemia virus envelope;
JAK: Janus kinase; PBSC: Peripheral blood stem cells; UCB: Umbilical cord blood.
For personal use only.

7.Gene therapy for other severe combined disorders. Other severe immunodeficiencies are also targets
immunodeficiencies for treatment by gene therapy and are at various stages of
development. Preclinical studies in ZAP-70 deficiency show
The molecular basis of autosomal recessive T-B+NK- SCID is that reconstitution of ZAP-70-mediated signalling in T cells
mutation of the receptor tyrosine kinase gene JAK-3 [8]. The can be restored by retroviral gene transfer [36,37]. In
dependence of γc on signalling through JAK-3 is responsible Wiskott-Aldrich syndrome, where mutations in the WASP
for a clinical and immunological phenotype identical to that gene result in a heterogenous phenotype of combined
of SCID-X1, and the rationale for gene therapy is therefore immunodeficiency, thrombocytopaenia and eczema, a
similar. Correction of a murine model of JAK-3-deficient number of studies including correction of a murine model
SCID has been achieved using both myelosuppresssive and, show considerable promise and may allow development of
more relevant to clinical studies, conditioning-free protocols. clinical trials in the near future [38-41].
One patient has been treated by retroviral vector-mediated
transduction of haematopoietic stem cells, but no immune 8.Insertional mutagenesis and risks of
recovery was seen, although no published data exists gene therapy
(Sorrentino et al., unpublished data). Patients with muta-
tions of the RAG-1 and -2 genes characteristically present For retroviruses, which depend on chromosomal integration
with absence of both B and T cells. Moloney-based gamma- for stability of transduction, the most prominent safety
retroviral vectors have recently been shown to effectively concern has been for insertional mutagenicity. Prior to the
reconstitute RAG-2-deficient mice in the absence of detecta- inception of the clinical studies detailed above, data from
ble toxicity, even though gene expression was not tightly numerous animal studies and > 300 clinical trials in which
regulated [34]. One way to obviate toxicity arising from patients have received retroviral vectors, and from theoretical
dysregulated gene expression in any condition, and to achieve considerations, the risk of clinically manifesting insertional
physiological activity, is to correct genetic mutations by gene mutagenesis had been judged to be low. However, in one
repair or homologous recombination. It has recently been SCID-X1 gene therapy study, 3 of 11 patients who had shown
shown that RAG-2-/- mutant embryonic stem cells, repaired successful immune recovery following gene therapy developed
by standard homologous recombination technology, can be T cell leukaemia 3 years after treatment ([42,43] and
grown in vitro to provide sufficient haematopoietic progeni- Cavazzana-Calvo et al., ASGT Genotoxicity Retreat, St. Louis,
tors for engraftment and correction of RAG-2 mutant June 2005). Detailed study of the leukaemic clones shows that
mice [35]. This is the first example of gene therapy combined in two cases vector insertion into the LMO-2 proto-oncogene
with a therapeutic cloning strategy, and clearly has important locus led to upregulation of LMO-2 protein expression, most
implications for the future treatment of many genetic probably as a result of retroviral long terminal repeat (LTR)

Expert Opin. Biol. Ther. (2005) 5(9) 1179


Gene therapy for severe combined immunodeficiencies

enhancer activity. Analysis of the third patient most recently with significant short- and long-term morbidities. Carefully
described is ongoing. This is the first demonstration of inser- monitored clinical studies and analysis of larger numbers of
tional mutagenesis in human gene therapy, and follows the patients will be required to address these important issues.
recent description of a similar event leading to myeloid
leukaemia in mice, in which the oncogene was identified as 9.Expert opinion and conclusion:
Evi-1 [44]. From more recent studies it is clear that retroviruses future prospects for severe combined
show preference for integration into transcriptionally active immunodeficiency gene therapy
genes and gammaretroviruses (as used in the SCID-X1 and
ADA-SCID trials) in particular integrate preferentially into a The results from the SCID-X1 and ADA-SCID studies show
5-kb region around the transcription start site [45,46]. Retro- that haematopoietic stem cell gene therapy can offer excellent
viral vector dose escalation studies in murine models also immune system and clinical recovery. The use of low intensity
demonstrate that the potential for leukaemogenesis is related preconditioning coupled with gene therapy, as used for
to increased vector dose and thereby increased transgene copy ADA-SCID, offers much hope for many other inherited
Expert Opin. Biol. Ther. Downloaded from informahealthcare.com by University of Guelph on 05/14/13

number [47]. These studies also suggest that combinatorial haematological and immunological disorders where initial
insertional events may be important in leukaemogenesis. cytoreduction may be a prerequisite for successful outcome. It
Thus, the use of gammaretroviral vectors per se may represent is likely that these first few studies will allow further techno-
an inherent risk, but the lack of serious adverse events (SAEs) logical development and initiation of clinical trials in other
in other retroviral vector-based clinical studies would suggest conditions over the next decade.
that other factors may be equally as important. The high It is clear that clinical progress can only be achieved in
incidence in this SCID-X1 study, compared, for example, combination with improvements in basic vector design to
with ADA-SCID gene therapy where similar numbers of promote both safety and efficacy. For gammaretroviral
patients have been treated for equivalent and in some cases vectors, where insertional events may pose a consistent risk,
longer periods of time, may imply that the γc transgene itself it is necessary to limit LTR enhancer activity in order to
may play a synergistic role with oncogene activation. At prevent transcription of non-target genes. This may be
For personal use only.

present there is no direct evidence for a contribution of achieved by the use of specialised DNA sequences, termed
dysregulated γc expression in lymphoid cells, although the enhancer blockers (or insulators), that can interfere with an
very nature of γc as a lymphoid growth factor makes this a enhancers ability to communicate with a target promoter
possibility and is being studied carefully. Interestingly, analysis when positioned between the two [49]. Targeting of retroviral
of one leukaemic tumour arising from infection with replica- vectors to ‘safe’ regions in the genome is another possibility,
tion-competent retrovirus shows clonal integrations in both but it is likely that the next generation of gammaretroviral
LMO-2 and γc [48]. The chances of finding coincident inte- vectors to be developed for clinical use will be self-inactivating
grations into both genes within the same tumour are exceed- (SIN) vectors, where transgene expression is controlled by a
ingly small and provide genetic evidence for a synergistic non-viral promoter with limited or no enhancer activity.
mechanism. Cells with high proliferative potential such as Alternative SIN vectors based on lentiviruses or foamy
thymocytes are also likely to be more susceptible to transfor- viruses that obviate prolonged ex vivo culture may allow the
mation following an insertional event than quiescent cells if preservation of larger numbers of multipotential progenitor
they acquire additional adverse mutations unrelated to the cells, but at the same time may produce higher numbers of
gene therapy itself. This increased risk cannot yet be quanti- insertion events in each cell. Methods to minimise the
fied. The detailed molecular analysis of insertion events in number of integration events per cell and to limit the
patients undergoing gene therapy will greatly assist in the number of engrafting clones, for example, by more stringent
delineation of integration points within the genome, but is purification of stem cell (or defined target cell) populations,
unlikely to be able to predict potential for leukaemogenesis may therefore also be beneficial. In particular, lentiviral
unless recurrent hotspots associated with clinical disease vectors provide greater capacity for incorporation of more
become evident. The accurate characterisation of adverse complex and physiological regulatory sequences. The rela-
events, the utilisation of protocols to test toxicity in a rigorous tive risk for each type of vector modification needs to be
way and the development of methods to minimise risks are determined in clinically relevant animal model systems, and
therefore essential. the effectiveness of these models to predict side effects in
The applicability of any novel therapy, including gene humans has to be validated.
therapy, ultimately depends on the balance of risks against The development of homologous recombination or gene
those of alternative treatments. Of eighteen SCID-X1 patients repair to correct mutations, or the construction of mitotically
now treated in two clinical trials, three have developed SAEs, stable extrachromosomal vectors would obviate many of these
two of which have been treated and are in remission, and one problems, but existing technologies have until recently been
of whom has died due to leukaemic relapse. The alternative inefficient. Recent innovations, such as zinc-finger technology
for this group of patients is mismatched allogeneic HSCT, whereby double-strand breaks induced by zinc-finger nucle-
which carries a 20% 1-year mortality and is also associated ases can create specific sequence alterations by stimulating

1180 Expert Opin. Biol. Ther. (2005) 5(9)


Gaspar & Thrasher

homologous recombination between the chromosome and an efficiency will be sufficient to provide clinical benefit. The
extrachromosomal DNA donor, show efficient correction of future for gene therapy of SCID is exciting, but has been
γc-deficient cells, at least in in vitro studies, and may hold clouded by the occurrence of toxicity. As for all novel thera-
considerable promise if gene delivery to haematopoietic peutic modalities, an increased understanding of mechanisms
progenitor cells can be demonstrated [50]. Once again, SCID and increased sophistication of technology will translate into
may be a perfect initial target for this strategy, as even limited even more effective and safe application.

Bibliography 10. SCHWARZ K, GAUSS GH, LUDWIG L 19. CAVAZZANA-CALVO M,


Papers of special note have been highlighted as et al.: RAG mutations in human HACEIN-BEY S, DE SAINT BG et al.:
either of interest (•) or of considerable interest (••) B cell-negative SCID. Science (1996) Gene therapy of human severe combined
to readers. 274(5284):97-99. immunodeficiency (SCID)-X1 disease.
11. MOSHOUS D, CALLEBAUT I, Science (2000) 288(5466):669-672.
1. FISCHER A, CAVAZZANA-CALVO M,
Expert Opin. Biol. Ther. Downloaded from informahealthcare.com by University of Guelph on 05/14/13

DE CHASSEVAL R et al.: Artemis, a novel •• A landmark study. The first


DE SAINT BG et al.: Naturally occurring
DNA double-strand break repair/V(D)J demonstration of successful
primary deficiencies of the immune system.
recombination protein, is mutated in human gene therapy for a genetic disorder.
Annu. Rev. Immunol. (1997) 15:93-124.
severe combined immune deficiency. 20. HACEIN-BEY-ABINA S, LE DEIST F,
2. ANTOINE C, MULLER S, CANT A et al.:
Cell (2001) 105(2):177-186. CARLIER F et al.: Sustained correction of
Long-term survival and transplantation of
12. FISCHER A, LANDAIS P, FRIEDRICH W X-linked severe combined
haemopoietic stem cells for
et al.: European experience of bone-marrow immunodeficiency by ex vivo gene therapy.
immunodeficiencies: report of the European
transplantation for severe combined N. Engl. J. Med. (2002) 346(16):1185-1193.
experience 1968-99. Lancet (2003)
361(9357):553-560. immunodeficiency. Lancet (1990) 21. GASPAR HB, PARSLEY KL, HOWE S
336(8719):850-854. et al.: Gene therapy of X-linked severe
3. BUCKLEY RH, SCHIFF SE, SCHIFF RI
13. AMROLIA P, GASPAR HB, HASSAN A combined immunodeficiency by use of a
et al.: Hematopoietic stem-cell
pseudotyped gammaretroviral vector.
For personal use only.

transplantation for the treatment of severe et al.: Nonmyeloablative stem cell


transplantation for congenital Lancet (2004) 364(9452):2181-2187.
combined immunodeficiency. N. Engl.
J. Med. (1999) 340(7):508-516. immunodeficiencies. Blood (2000) 22. THRASHER AJ, HACEIN-BEY-ABINA S,
96(4):1239-1246. GASPAR HB et al.: Failure of SCID-X1
4. FISCHER A, LE DEIST F,
14. RAO K, AMROLIA PJ, JONES A et al.: gene therapy in older patients. Blood (2005)
HACEIN-BEY-ABINA S et al.: Severe
Improved survival after unrelated donor 105(11):4255-4257.
combined immunodeficiency. A model
bone marrow transplantation in children • The age of patients undergoing gene
disease for molecular immunology and
with primary immunodeficiency using a therapy may be important, as there may be
therapy. Immunol. Rev. (2005) 203:98-109.
reduced-intensity conditioning regimen. age-related restrictions to the reinitiation of
5. NOGUCHI M, YI H, ROSENBLATT HM thymic function.
Blood (2005) 105(2):879-885.
et al.: Interleukin-2 receptor gamma chain
15. BUCKLEY RH, SCHIFF SE, SCHIFF RI 23. BLAESE RM, CULVER KW, MILLER AD
mutation results in X-linked severe
et al.: Haploidentical bone marrow stem cell et al.: T lymphocyte-directed gene therapy
combined immunodeficiency in humans.
transplantation in human severe combined for ADA-SCID: initial trial results after 4
Cell (1993) 73(1):147-157.
immunodeficiency. Semin. Hematol. (1993) years. Science (1995) 270:475-480.
6. PARRISH-NOVAK J, DILLON SR,
30(4 Suppl. 4):92-101. 24. BORDIGNON C, NOTARANGELO LD,
NELSON A et al.: Interleukin 21 and its
16. HERSHFIELD MS: PEG-ADA NOBILI N et al.: Gene therapy in peripheral
receptor are involved in NK cell expansion
replacement therapy for adenosine blood lymphocytes and bone marrow for
and regulation of lymphocyte function.
deaminase deficiency: an update after ADA- immunodeficient patients. Science
Nature (2000) 408(6808):57-63.
8.5 years. Clin. Immunol. Immunopathol. (1995) 270(5235):470-475.
7. WHITE H, THRASHER A, VEYS P,
(1995) 76(3 Pt 2):S228-S232. 25. HOOGERBRUGGE PM,
KINNON C, GASPAR HB: Intrinsic
17. HERSHFIELD MS: PEG-ADA: an VAN BEUSECHEM VW, FISCHER A
defects of B cell function in X-linked severe
alternative to haploidentical bone marrow et al.: Bone marrow gene transfer in three
combined immunodeficiency. Eur. J.
transplantation and an adjunct to gene patients with adenosine deaminase
Immunol. (2000) 30(3):732-737.
therapy for adenosine deaminase deficiency. deficiency. Gene Ther. (1996) 3(2):179-183.
8. MACCHI P, VILLA A, GILIANI S et al.:
Hum. Mutat. (1995) 5(2):107-112. 26. KOHN DB, WEINBERG KI, NOLTA JA
Mutations of Jak-3 gene in patients with
18. DEMAISON C, BROUNS G, et al.: Engraftment of gene-modified
autosomal severe combined immune
BLUNDELL MP et al.: A defined window umbilical cord blood cells in neonates with
deficiency (SCID). Nature (1995)
for efficient gene marking of severe adenosine deaminase deficiency. Nat. Med.
377(6544):65-68.
combined imunodeficient-repopulating cells (1995) 1(10):1017-1023.
9. HIRSCHHORN R: Overview of
using a gibbon ape leukaemia 27. MUUL LM, TUSCHONG LM,
biochemical abnormalities and molecular
virus-pseudotyped retroviral vector. SOENEN SL et al.: Persistence and
genetics of adenosine deaminase deficiency.
Hum. Gene Ther. (2000) 11:91-100. expression of the adenosine deaminase gene
Pediatr. Res. (1993) 33(1 Suppl.):S35-S41.
for 12 years and immune reaction to gene

Expert Opin. Biol. Ther. (2005) 5(9) 1181


Gene therapy for severe combined immunodeficiencies

transfer components: long-term results of 36. OTSU M, STEINBERG M, FERRAND C 44. LI Z, DULLMANN J,
the first clinical gene therapy trial. Blood et al.: Reconstitution of lymphoid SCHIEDLMEIER B et al.: Murine
(2003) 101(7):2563-2569. development and function in ZAP-70- leukemia induced by retroviral gene
• Analysis of patients treated after 10 years deficient mice following gene transfer into marking. Science (2002) 296(5567):497.
shows that gene-modified cells persist in bone marrow cells. Blood (2002) 45. WU X, LI Y, CRISE B, BURGESS SM:
significant numbers. 100(4):1248-1256. Transcription start regions in the human
28. KOHN DB, HERSHFIELD MS, 37. TAYLOR N, BACON KB, SMITH S et al.: genome are favored targets for MLV
CARBONARO D et al.: T lymphocytes Reconstitution of T cell receptor signaling integration. Science (2003)
with a normal ADA gene accumulate after in ZAP-70-deficient cells by retroviral 300(5626):1749-1751.
transplantation of transduced autologous transduction of the ZAP-70 gene. 46. LAUFS S, NAGY KZ, GIORDANO FA,
umbilical cord blood CD34+ cells in J. Exp. Med. (1996) 184(5):2031-2036. HOTZ-WAGENBLATT A, ZELLER WJ,
ADA-deficient SCID neonates. Nat. Med. 38. CHARRIER S, STOCKHOLM D, FRUEHAUF S: Insertion of retroviral
(1998) 4(7):775-780. SEYE K et al.: A lentiviral vector encoding vectors in NOD/SCID repopulating human
29. SCHMIDT M, CARBONARO DA, the human Wiskott-Aldrich syndrome peripheral blood progenitor cells occurs
Expert Opin. Biol. Ther. Downloaded from informahealthcare.com by University of Guelph on 05/14/13

SPECKMANN C et al.: Clonality analysis protein corrects immune and cytoskeletal preferentially in the vicinity of transcription
after retroviral-mediated gene transfer to defects in WASP knockout mice. Gene Ther. start regions and in introns. Mol. Ther.
CD34+ cells from the cord blood of (2005) 12(7):597-606. (2004) 10(5):874-881.
ADA-deficient SCID neonates. Nat. Med. 39. KLEIN C, NGUYEN D, LIU CH et al.: 47. MODLICH U, KUSTIKOVA OS,
(2003) 9(4):463-468. Gene therapy for Wiskott-Aldrich SCHMIDT M et al.: Leukemias following
30. AIUTI A, VAI S, MORTELLARO A et al.: syndrome: rescue of T-cell signaling and retroviral transfer of multidrug resistance 1
Immune reconstitution in ADA-SCID after amelioration of colitis upon transplantation (MDR1) are driven by combinatorial
PBL gene therapy and discontinuation of of retrovirally transduced hematopoietic insertional mutagenesis. Blood (2005)
enzyme replacement. Nat. Med. (2002) stem cells in mice. Blood (2003) 105(11):4235-4246.
8(5):423-425. 101(6):2159-2166. 48. DAVE UP, JENKINS NA,
31. ROGERS MH, LWIN R, FAIRBANKS L, 40. DUPRE L, TRIFARI S, FOLLENZI A COPELAND NG: Gene therapy
For personal use only.

GERRITSEN B, GASPAR HB: Cognitive et al.: Lentiviral vector-mediated gene insertional mutagenesis insights. Science
and behavioral abnormalities in adenosine transfer in T cells from Wiskott-Aldrich (2004) 303(5656):333.
deaminase deficient severe combined syndrome patients leads to functional • Evidence for possible synergism between
immunodeficiency. J. Pediatr. (2001) correction. Mol. Ther. (2004) LMO2 and γc in the molecular
139(1):44-50. 10(5):903-915. pathogenesis of T cell leukaemias in
32. ALBUQUERQUE W, GASPAR HB: 41. STROM TS, GABBARD W, KELLY PF, SCID-X1 trials.
Bilateral sensorineural deafness in adenosine CUNNINGHAM JM, NIENHUIS AW: 49. WEST AG, FRASER P: Remote control of
deaminase-deficient severe combined Functional correction of T cells derived gene transcription. Hum. Mol. Genet.
immunodeficiency. J. Pediatr. (2004) from patients with the Wiskott-Aldrich (2005) 14(Spec. No 1):R101-R111.
144(2):278-280. syndrome (WAS) by transduction with an 50. URNOV FD, MILLER JC, LEE YL et al.:
33. AIUTI A, SLAVIN S, AKER M et al.: oncoretroviral vector encoding the WAS Highly efficient endogenous human gene
Correction of ADA-SCID by stem cell gene protein. Gene Ther. (2003) 10(9):803-809. correction using designed zinc-finger
therapy combined with nonmyeloablative 42. HACEIN-BEY-ABINA S, VON KALLE C, nucleases. Nature (2005)
conditioning. Science (2002) SCHMIDT M et al.: A serious adverse 435(7042):646-651.
296(5577):2410-2413. event after successful gene therapy for
•• Another landmark study. The first X-linked severe combined Affiliation
successful treatment of ADA-SCID by immunodeficiency. N. Engl. J. Med. (2003) H Bobby Gaspar†1 & Adrian J Thrasher
gene therapy, and the first successful 348(3):255-256. †Author for correspondence

combination of chemotherapy with 1Institute of Child Health,


43. HACEIN-BEY-ABINA S, VON KALLE C,
gene therapy. SCHMIDT M et al.: LMO2-associated Molecular Immunology Unit, 30 Guilford Street,
34. YATES F, MALASSIS-SERIS M, clonal T cell proliferation in two patients London, WC1N 1EH, UK
STOCKHOLM D et al.: Gene therapy of after gene therapy for SCID-X1. Science Tel: +44 (0)207 905 2319;
RAG-2-/- mice: sustained correction of the (2003) 302(5644):415-419. Fax: +44 (0)207 905 2810;
immunodeficiency. Blood (2002) •• Retroviral vector integration into the E-mail: h.gaspar@ich.ucl.ac.uk
100(12):3942-3949. LMO2 oncogene results in LMO2
35. RIDEOUT WM 3RD, activation and clonal T cell proliferation.
HOCHEDLINGER K et al.: Correction of The first serious adverse events in SCID
a genetic defect by nuclear transplantation gene therapy trials.
and combined cell and gene therapy.
Cell (2002) 109(1):17-27.

1182 Expert Opin. Biol. Ther. (2005) 5(9)

You might also like