Beta-thalassemia is a genetic disorder with mutations in the β-globin gene that reduce or abolish β-globin protein production. Patients with β-thalassemia major (Cooley's anemia) become severely anemic by 6 to 18 months of age, and are transfusion dependent for life, while those with thalassemia intermedia, a less-severe form of thalassemia, are intermittently or rarely transfused. An allogeneically matched bone marrow transplant is curative, although it is restricted to those with matched donors. Gene therapy holds the promise of “fixing” one's own bone marrow cells by transferring the normal β-globin or γ-globin gene into hematopoietic stem cells (HSCs) to permanently produce normal red blood cells. Requirements for effective gene transfer for the treatment of β-thalassemia are regulated, erythroid-specific, consistent, and high-level β-globin or γ-globin expression. Gamma retroviral vectors have had great success with immune-deficiency disorders, but due to vector-associated limitations, they have limited utility in hemoglobinopathies. Lentivirus vectors, on the other hand, have now been shown in several studies to correct mouse and animal models of thalassemia. The immediate challenges of the field as it moves toward clinical trials are to optimize gene transfer and engraftment of a high proportion of genetically modified HSCs and to minimize the adverse consequences that can result from random integration of vectors into the genome by improving current vector design or developing novel vectors. This article discusses the current state of the art in gene therapy for β-thalassemia and some of the challenges it faces in human trials.

Beta thalassemias are among the most common single-gene defects worldwide, and pose a severe health and economic burden to patients and families at risk. They are caused by different mutations in the β-globin gene cluster, resulting in reduced or absent adult hemoglobin (HbA), which is composed of two alpha- and two beta-globin chains (α2β2), leading to severe anemia. This disorder is characterized by reduced HbA production (β+ thalassemia) or complete absence of β-globin synthesis (β0 thalassemia). In humans, the degree of imbalance in the α- and β-globin protein is directly linked to the clinical severity of β-thalassemia. This imbalance also hinders erythroid precursor maturation, resulting in ineffective erythropoiesis. Humans that are heterozygotic for β-thalassemia mutations are very mildly anemic and lead normal lives. Homozygosity results in disease: thalassemia intermedia, in which individuals are moderately anemic due to reduced β-globin production, and thalassemia major or Cooley's anemia, characterized by absent or severely reduced β-globin production and complete transfusion dependence for survival. Patients with severe β-thalassemia major show significant levels of iron accumulation in multiple organs and tissues from transfusional hemosiderosis, and inadequately transfused patients develop massive erythroid hyperplasia and extramedullary hematopoiesis in the body's attempt to compensate for the red blood cell loss. Iron overload, associated with chronic transfusions, is the major cause of death.1 

Hematopoietic stem-cell (HSC) transplantation using bone marrow, umbilical cord blood, or mobilized peripheral blood as a source of HSCs has been performed in numerous patients with thalassemia. The results of HSC transplant are remarkable if a matched sibling donor is available and the transplants are performed in well-transfused and chelated children with good organ status. The accompanying review by Dr. Angelucci summarizes the current status of transplantation for thalassemia (see p. 456 in this issue).

HSC transplant is limited by the availability of matched donors and is associated with immunological complications, including graft rejection and graft-versus-host disease.2–4  Gene transfer using autologous bone marrow can potentially permanently cure thalassemia major, and could resolve the limitations of finding a matched donor and eliminate the risks of graft-versus-host disease and graft rejection that are associated with allogeneic bone marrow transplantation.

For gene transfer of a normal β-globin gene to be an option, several conditions must be met, including: 1) efficient gene transfer and engraftment of a high number of HSCs, 2) regulated expression in the erythroid lineage, 3) consistent and therapeutic levels of β-globin gene expression, and 4) safe levels of expression, ideally devoid of position effects.

Earlier studies on globin gene regulation and gamma-retroviral globin gene-encoding vectors were pivotal in providing critical insights into elements required for the high-level, regulated expression of globin genes. Gamma-retroviral vectors were limited by their size, stable transmission, and high level of expression of the β-globin gene due to: 1) the requirement for large elements of the locus control region (LCR) for high level expression; initial efforts to incorporate minimal core LCR elements resulted in vectors with low titers5  and low levels of expression that were prone to position effects6 ; 2) transcriptional interference of the retroviral long terminal repeats (LTR) with LCR elements resulted in unstable proviral transmission with vectors prone to sequence rearrangements7  and poor transgene expression.8–9 ; and 3) the β-globin gene is an intron-dependent gene and needs to be inserted in reverse orientation in viral vectors.10–11  Several modifications allowed efficacious expression: 1) deletion of deleterious sequences in intron 2 of the β-globin gene to allow stable transmission and expression of β-globin gene from retroviral vectors (Figure 1A)12–13 ; 2) the β-globin cDNA was used with post-transcriptional regulatory elements14 ; 3) the α-locus HS-40 regulatory region was used in place of the large β-globin LCR15–18 ; and 4) alternative erythroid-specific promoters such as ankyrin19–20  and mutant hereditary persistence of fetal hemoglobin (HPFH) γ-globin promoters were used,21–22  but had limited success. Studies by Emery et al.23  and Rivella et al.24  showed that incorporation of the chicken hypersensitive site-4 insulator (cHS4) element in gamma-retrovirus vectors improved the performance of the β-globin transgene expression to a significant extent, although its therapeutic relevance was not studied.

Figure 1.

(A) Prototypic design of a γ-retroviral vector β/γ globin gene and the small LCR core elements. The β-globin gene (exons depicted in blue shaded boxes) is placed in the reverse orientation in the retroviral vector to prevent splicing of the introns in the viral genomic RNA during production of the virus. Arrow indicates the direction of transcription. In the γ-retroviral vector construct, HS2, HS3, and HS4 denotes LCR core derivatives.12  (B) Successful β/γ-globin LV vectors used by different groups. TNS9 vector is based on the publication by May et al.24  and encodes the human beta globin gene. The βA87Q vector encodes a mutated β-globin gene at the 87th codon, conferring it with anti-sickling properties,55  and also carries two copies of the 5′ 250-bp cHS4 insulator core. This vector was used for the gene therapy trial for β-thalassemia in France led by Dr. Philippe Leboulch. The BG-I vector, described by Putheenvetil et al.,32  expresses β-globin from a 254-bp β-globin promoter, contains large LCR elements, and is flanked by the 1.2-kb full-length cHS4 insulator in the LTRs. The vector d432β- vector28  also contains γ-globin 3′ regulatory untranslated regions (shaded purple) and enhancer elements downstream of the γ-globin coding sequences (3′e*). mLARβΔγV5 vector30  consists of extended regions of β-globin LCR. The GLOBE vector is described in the article by Miccio et al.29  Triangles represent deletion of the 372-bp purine-rich sequences in the β-globin IVS 2 (a 562-bp deletion of IVS2 in the GLOBE vector) and the 400-bp enhancer deletion in the LTR to generate a self-inactivating vector design. RRE, rev response element; HS2, HS3, and HS4, DNase hypersensitive sites 2, 3, and 4 present in the β-globin LCR and the length of respective hypersensitive sites are indicated adjacent to them; cPPT, central polypurine tract; LTR, long terminal repeat; 3′e, β-globin 3′ enhancer element; ψ, packaging signal; Ψ+, extended packaging signal; βP, β-globin gene promoter. The β or γ globin genes are depicted in the reverse orientation, with β-globin exons in blue, γ-globin exons in green, and β-globin introns shown as unshaded boxes

Figure 1.

(A) Prototypic design of a γ-retroviral vector β/γ globin gene and the small LCR core elements. The β-globin gene (exons depicted in blue shaded boxes) is placed in the reverse orientation in the retroviral vector to prevent splicing of the introns in the viral genomic RNA during production of the virus. Arrow indicates the direction of transcription. In the γ-retroviral vector construct, HS2, HS3, and HS4 denotes LCR core derivatives.12  (B) Successful β/γ-globin LV vectors used by different groups. TNS9 vector is based on the publication by May et al.24  and encodes the human beta globin gene. The βA87Q vector encodes a mutated β-globin gene at the 87th codon, conferring it with anti-sickling properties,55  and also carries two copies of the 5′ 250-bp cHS4 insulator core. This vector was used for the gene therapy trial for β-thalassemia in France led by Dr. Philippe Leboulch. The BG-I vector, described by Putheenvetil et al.,32  expresses β-globin from a 254-bp β-globin promoter, contains large LCR elements, and is flanked by the 1.2-kb full-length cHS4 insulator in the LTRs. The vector d432β- vector28  also contains γ-globin 3′ regulatory untranslated regions (shaded purple) and enhancer elements downstream of the γ-globin coding sequences (3′e*). mLARβΔγV5 vector30  consists of extended regions of β-globin LCR. The GLOBE vector is described in the article by Miccio et al.29  Triangles represent deletion of the 372-bp purine-rich sequences in the β-globin IVS 2 (a 562-bp deletion of IVS2 in the GLOBE vector) and the 400-bp enhancer deletion in the LTR to generate a self-inactivating vector design. RRE, rev response element; HS2, HS3, and HS4, DNase hypersensitive sites 2, 3, and 4 present in the β-globin LCR and the length of respective hypersensitive sites are indicated adjacent to them; cPPT, central polypurine tract; LTR, long terminal repeat; 3′e, β-globin 3′ enhancer element; ψ, packaging signal; Ψ+, extended packaging signal; βP, β-globin gene promoter. The β or γ globin genes are depicted in the reverse orientation, with β-globin exons in blue, γ-globin exons in green, and β-globin introns shown as unshaded boxes

Close modal

In the mid 1990s, lentiviral (LV) vectors derived from HIV made possible the development of globin LV vectors. LV vectors can package full-length, unspliced RNA due to the presence of a strong RNA export element, the rev response element (RRE), and can carry large transgene cassettes with introns and regulatory LCR elements. Moreover, globin LV vectors can stably transmit globin regulatory elements and the coding sequences of the β-globin gene. The promise of “additive gene therapy” as a therapeutic option for β-thalassemia was therefore made possible with these vectors for the first time in the field of gene transfer for the treatment of hemoglobinopathies.

Correction of Murine Thalassemia

In a pioneering study, May et al. showed phenotypic correction of thalassemia mice (Hbbth3/+ mice that resemble human thalassemia intermedia phenotype), with an average increase of hemoglobin by 3 to 4 g/dL per vector copy with the TNS9 vector. Subsequently, others have shown correction of murine thalassemia using similar β- or γ-globin-encoding LV vectors (Figure 1B and Table 1).25–29  A major finding that emerged from these studies was that thalassemia mice with an average copy number of one did not show correction, and multiple copies were required for a therapeutic effect.27  Variable globin transgene expression was evident in thalassemia mice,26,28  which was often sub-therapeutic for β0-thalassemia.26  Therefore, subsequent studies focused on higher and more consistent expression for effective correction of thalassemia major.

Table 1.

Comparisons of different β- or γ-globin vectors studied successfully in mouse and human models of β-thalassemia

Comparisons of different β- or γ-globin vectors studied successfully in mouse and human models of β-thalassemia
Comparisons of different β- or γ-globin vectors studied successfully in mouse and human models of β-thalassemia

Status of clinical trials denoted by asterisks: *US trial planned using this vector or with minor modifications; **clinical trial ongoing in France; ***clinical trial planned in Europe

Improved Expression and Position Effects of LV Vectors

Higher and more consistent globin expression at a lower copy number was addressed by different groups using several strategies. Hanawa et al. included larger HS2–4 elements of the LCR.30  Lisowski et al.31  showed that inclusion of HS1, in addition to HS2–4, improved the overall expression. Puthenveetil et al.32  and Arumugam et al.33–34  from our group, and Emery et al.35–36  flanked the LV and γ-retrovirus vectors, respectively, with a chromatin insulator from cHS4. Chromatin insulators are DNA elements that can shield the genes from their surrounding chromatin environment. Insulators exert two functions: they prevent gene silencing from the effect of heterochromatin (barrier activity), and they prevent activation of a gene promoter by an adjacent enhancer (enhancer blocking activity).37  Incorporation of insulator elements in vectors can therefore function to diminish position effects and silencing and block surrounding gene enhancers.38  We showed correction of β0 thalassemia major (Cooley's anemia) in the human erythroid progeny of human CD34+ cells in vitro and in xenografts, and in the red blood cell progeny of thalassemia intermedia murine HSC using cHS4-insulated LV vector BG-I.33–34 

Although insulated vectors provided consistent/uniform expression, and therefore lead to superior correction, the caveat is that LV vectors with the full-length (1.2 kb) cHS4 element in the LTR exhibit marked reductions in infectious titers.39  LV vectors carrying the large globin gene and LCR have modest titers even without insertion of large insulator elements in the LTR. The further sharp decline in titers limits the large-scale production of vector for human trials. The smaller core elements of cHS4 were therefore explored, but the core does not retain the insulator activity of the full-length insulator.33,36  The cHS4 insulator can provide consistent expression of globin genes33–35  and can impart enhancer-blocking effects40–41  if its effect on viral titers can be overcome.39  Miccio et al.29  generated LV vectors with high titers by shortening the globin insert in the vector. They removed HS4, retaining only HS2 and HS3 of the LCR, and removed a larger portion of intron 2 of the β-globin gene. This vector has high titers and is able to efficiently correct murine thalassemia (and more recently, human thalassemia intermedia and major42 ), although high vector copies are required to correct murine thalassemia major with this vector (Table 1)

Most murine transplant studies are performed with a myeloablative regimen that allows near-complete donor engraftment, and thus there is high chimerism of genetically modified HSCs. Clinically, unless myeloablative conditioning is used, this may not be necessary. Persons et al. showed that chimerism levels of only 10% to 30% normal HSCs are sufficient to result in nearly complete hematologic and pathologic correction of thalassemia mice,43  and our group has shown that 15% to 20% genetically corrected HSCs are sufficient to correct sickle-cell disease.44  This level of chimerism require a significant amount of pre-transplant chemotherapy conditioning for cytoreduction of endogenous HSCs, a large stem cell dose, and a high proportion of genetically modified HSCs. This is in contrast to gene-transfer approaches for immunodeficiency disorders, in which minimal to no cytoreduction is sufficient for phenotypic correction due to the tremendous selective advantage of gene-modified T-cell precursors and T cells.

Gene Therapy Clinical Trials in Thalassemia

The status of potential clinical trials in the upcoming future and an open clinical trial for β-thalassemia is included in Table 1. Leboulch et al. have opened a trial in France that has enrolled two patients. One patient with E-β0-thalassemia was transplanted with CD34+ cells transduced with the β-globin vector (Figure 1B) following myeloablative pre-transplant conditioning, and has shown sustained improvement over 2 years in hemoglobin and transfusion independence. At a molecular level, this has occurred due to selective proliferation of one myelo-erythroid clone with a viral integration that has apparently conferred this clone a proliferative advantage, as described in the subsequent section.

A safety concern is the risk of insertional mutagenesis with randomly integrating viral vectors. Insertional oncogenesis by the viral LTR enhancer has been shown in humans, in the context of gene therapy for X-linked severe combined immunodeficiency syndrome (X-SCID1) with γ-retroviral vectors in four patients in the French X-SCID1 trial45  and one patient in the London X-SCID1 trial,46  despite a very high degree of success with gene transfer. Four of five patients showed insertional activation of the LMO2 oncogene. Recently, in a similarly successful chronic granulomatous disease trial in Germany, insertional activation of MDS-EVI-1 by the viral LTR enhancer resulted in myelodysplastic syndrome in two patients that was accompanied by the silencing of the transgene expression due to methylation of CpG island in the viral LTR promoter, resulting in the abolishment of a therapeutic effect of the transgene.47  A study by Imren et al.48  with β-globin LV vector-marked human hematopoietic cells in immune-deficient mice showed that 86% of insertions occurred within genes, including genes involved in hematopoiesis and in human leukemia, although genetic dysregulation of these genes was not addressed in this study. Hargrove et al. showed direct evidence that globin LV vectors can cause insertional dysregulation of cellular genes in primary, clonal, murine, β-thalassemic erythroid cells in which the globin locus control region was active.49  Similar results were published subsequently by Arumugam et al. using the in vitro immortalization assay in primary murine hematopoietic progenitors.50  The French trial described below has now confirmed the LCR-mediated cellular gene activation in a patient with hemoglobin E-β0 thalassemia.49 

In the first trial for gene transfer for the treatment of β-thalassemia, Leboulch et al. observed a therapeutic effect in one patient that was associated with the emergence of a dominant erythroid clone with insertion of the vector in the HMGA2 gene locus.51  The patient had approximately 10% gene marked hematopoiesis, of which a large percentage is contributed by this clone. The HMGA2 gene expression is increased in erythroid cells and not in myeloid cells, indicating dysregulation of the gene by the LCR in erythroid lineage. At the burst-forming unit-erythroid (BFUe) level, the amount of β87 globin produced by this clone is similar compared with other gene-marked BFUe. So far, this expansion of the dominant clone has resulted in a therapeutic benefit for 2 years following gene transfer, and there has been no perturbation of hematopoiesis. This LV had two copies if the cHS4 “core” that recombined and the integrated LV is flanked by one copy of the recombined cHS4 core (reported by Leboulch at the American Society of Gene Therapy Meeting, 2009; at the 5th Stem Cell Clonality and Genotoxicity Retreat, 2009; and at the Recombinant DNA Advisory Committee; for the latter, a webcast is available online at: http://oba.od.nih.gov/rdna/rac_past_meeting_2009_webcasts.html#dec09).51  At this point, the cause and effect are unknown and the field awaits further developments.

Using a sensitive in vitro immortalization assay, we have explored the genotoxicity of the β-globin LCR enhancer and promoter, with and without different sized cHS4 insulator elements in the LTR of the self-inactivating LV using primary hematopoietic progenitors to generate in vitro immortalization assay mutants. LCR carrying LV had approximately 200-fold lower transforming potential compared with the conventional γ-retroviral vectors. A further 3-fold reduction in transforming activity was observed with the LCR containing LV carrying either the full-length cHS4 or the 650-bp cHS4 (comprising of the 5′ 250 bp core and the distal3′ 400 bp).50  Our data indicate that toxicology studies of LCR containing LV in mice will likely not yield insertional oncogenesis with the number of mice that can be practically studied.

The clonal dominance seen in the patient with β-thalassemia treated in France suggests that the cHS4 core alone is unable to shield the HMGA2 gene from the LCR enhancer. Recently, a structure-function analysis of cHS4 showed unique insulator activity in the distal 400-bp cHS4 sequences, which, when combined with the 5′ core, restored full barrier31  and enhancer blocking46  activity. Overall, the identification of more potent insulators is necessary to impart safety to randomly integrating vectors.

Most traditional gene-therapy strategies add the function of a full-length gene to a population of target cells while the endogenous mutant gene is still present. Genetic correction using homologous recombination of the β-sickle globin has been done in mouse embryonic stem cells and induced pluripotent stem cells reprogrammed from fibroblasts in a sickle cell anemia mouse model. The corrected induced pluripotent stem cells were then differentiated into HSCs and transplanted, with complete correction of sickle-cell anemia.52–53  The reprogramming of somatic cells into induced pluripotent stem cells opens a new approach for treating β-thalassemia.54  Ye et al. reprogrammed the skin fibroblasts of a patient with homozygous β° thalassemia into induced pluripotent stem cells, and showed that these cells could be differentiated into hematopoietic cells that synthesized hemoglobin. These reports suggest that induced pluripotent stem cells could offer a new approach for the treatment of β-thalassemia. These approaches are at an experimental level at the present time.

The therapeutic efficacy of LV-mediated globin gene transfer in HSC has been adequately demonstrated over the past decade, resulting in the correction of mouse and human models of Cooley's anemia and thalassemia intermedia. The safety of gene transfer has been the primary goal of the researchers in designing globin vectors. The design of lineage and differentiation stage-restricted vectors represents one major step in reducing the risk of trans-activating oncogenes. The risk of insertional oncogenesis can likely be reduced significantly by rational vector design with tightly regulating expression of the vector-encoded transgene and by minimizing interactions between vector elements and flanking transgenes with optimal insulator elements. In this respect, HSC gene transfer to treat β-thalassemia is now becoming a therapeutic reality. However, caution regarding the insertional mutagenesis capability of any randomly integrating vector remains, until homologous recombination and induced pluripotent stem cell technology becomes translatable to the clinic.

This work was supported by NIH grant no. U54 HL06–008.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Off-label drug use: None disclosed.

Punam Malik, MD, TCHRF 6564, Division of Experimental Hematology, Cincinnati Children's Hospital Medical Center, ML 7013, 3333 Burnet Avenue, Cincinnati, OH 45229; Phone: 513-636-0364; Fax: 513-636-3768; e-mail: Punam.Malik@cchmc.org

1
Weatherall
DJ
Stamatoyannopoulos
G
Neinhuis
AW
Majerus
PW
Varmus
H
The thalassemias
The molecular basis of blood diseases
1994
2nd ed
Philadelphia
W.B. Saunders Company
157
205
2
Lucarelli
G
Galimberti
M
Giardini
C
et al
Bone marrow transplantation in thalassemia. The experience of Pesaro
Ann N Y Acad Sci
1998
850
270
5
3
Giardini
C
Lucarelli
G
Bone marrow transplantation for beta-thalassemia
Hematol Oncol Clin North Am
1999
13
1059
1064
4
Gaziev
D
Galimberti
M
Lucarelli
G
et al
Bone marrow transplantation from alternative donors for thalassemia: HLA-phenotypically identical relative and HLA-nonidentical sibling or parent transplants
Bone Marrow Transplant
2000
25
815
821
5
Plavec
I
Papayannopoulou
T
Maury
C
Meyer
F
A human beta-globin gene fused to the human beta-globin locus control region is expressed at high levels in erythroid cells of mice engrafted with retrovirus-transduced hematopoietic stem cells
Blood
1993
81
1384
1392
6
Chang
JC
Liu
D
Kan
YW
A 36-base-pair core sequence of locus control region enhances retrovirally transferred human beta-globin gene expression
Proc Natl Acad Sci U S A
1992
89
3107
3110
7
Novak
U
Harris
EA
Forrester
W
Groudine
M
Gelinas
R
High-level beta-globin expression after retroviral transfer of locus activation region-containing human beta-globin gene derivatives into murine erythroleukemia cells
Proc Natl Acad Sci U S A
1990
87
3386
3390
8
Sadelain
M
Wang
CH
Antoniou
M
Grosveld
F
Mulligan
RC
Generation of a high-titer retroviral vector capable of expressing high levels of the human beta-globin gene
Proc Natl Acad Sci U S A
1995
92
6728
6732
9
Walsh
CE
Liu
JM
Miller
JL
Nienhuis
AW
Samulski
RJ
Gene therapy for human hemoglobinopathies
Proc Soc Exp Biol Med
1993
204
289
300
10
Antoniou
M
Geraghty
F
Hurst
J
Grosveld
F
Efficient 3′-end formation of human beta-globin mRNA in vivo requires sequences within the last intron but occurs independently of the splicing reaction
Nucleic Acids Res
1998
26
721
729
11
Custódio
N
Carmo-Fonseca
M
Geraghty
F
Pereira
HS
Grosveld
F
Antoniou
M
Inefficient processing impairs release of RNA from the site of transcription
EMBO J
1999
18
2855
2866
12
Leboulch
P
Huang
GM
Humphries
RK
et al
Mutagenesis of retroviral vectors transducing human beta-globin gene and beta-globin locus control region derivatives results in stable transmission of an active transcriptional structure
EMBO J
1994
13
3065
3076
13
Miller
AD
Bender
MA
Harris
EA
Kaleko
M
Gelinas
RE
Design of retrovirus vectors for transfer and expression of the human beta-globin gene
J Virol
1988
11
62
11
4337
4345
14
Higashimoto
T
Urbinati
F
Perumbeti
A
et al
The woodchuck hepatitis virus post-transcriptional regulatory element reduces readthrough transcription from retroviral vectors
Gene Ther
2007
9
14
17
1298
304
15
Emery
DW
Morrish
F
Li
Q
Stamatoyannopoulos
G
Analysis of gamma-globin expression cassettes in retrovirus vectors
Hum Gene Ther
1999
10
877
888
16
Lung
H
Meeus
IS
Weinberg
RS
Atweh
GF
In vivo silencing of the human gamma-globin gene in murine erythroid cells following retroviral transduction
Blood Cells Mol Dis
2000
26
613
619
17
Ren
S
Wong
BY
Li
J
Luo
XN
Wong
PM
Atweh
GF
Production of genetically stable high-titer retroviral vectors that carry a human gamma-globin gene under the control of the alpha-globin locus control region
Blood
1996
87
2518
2524
18
Nishino
T
Tubb
J
Emery
DW
Partial correction of murine beta-thalassemia with a gammaretrovirus vector for human gamma-globin
Blood Cells Mol Dis
2006
37
1
7
19
Sabatino
DE
Wong
C
Cline
AP
et al
A minimal ankyrin promoter linked to a human gamma-globin gene demonstrates erythroid specific copy number dependent expression with minimal position or enhancer dependence in transgenic mice
J Biol Chem
2000
15
28549
28554
20
Sabatino
DE
Seidel
NE
Aviles-Mendoza
GJ
et al
Long-term expression of gamma-globin mRNA in mouse erythrocytes from retrovirus vectors containing the human gamma-globin gene fused to the ankyrin-1 promoter
Proc Natl Acad Sci U S A
2000
97
13294
13299
21
Fragkos
M
Anagnou
NP
Tubb
J
Emery
DW
Use of the hereditary persistence of fetal hemoglobin 2 enhancer to increase the expression of oncoretrovirus vectors for human gamma-globin
Gene Ther
2005
12
1591
1600
22
Katsantoni
EZ
Langeveld
A
Wai
AW
et al
Persistent gamma-globin expression in adult transgenic mice is mediated by HPFH-2, HPFH-3, and HPFH-6 breakpoint sequences
Blood
2003
102
3412
3419
23
Emery
DW
Yannaki
E
Tubb
J
Stamatoyannopoulos
G
A chromatin insulator protects retrovirus vectors from chromosomal position effects
Proc Natl Acad Sci U S A
2000
97
9150
9155
24
Rivella
S
Callegari
JA
May
C
Tan
CW
Sadelain
M
The cHS4 insulator increases the probability of retroviral expression at random chromosomal integration sites
J Virol
2000
74
4679
4687
25
May
C
Rivella
S
Callegari
J
et al
Therapeutic haemoglobin synthesis in beta-thalassaemic mice expressing lentivirus-encoded human beta-globin
Nature
2000
406
82
86
26
Rivella
S
May
C
Chadburn
A
Riviere
I
Sadelain
M
A novel murine model of Cooley anemia and its rescue by lentiviral-mediated human beta-globin gene transfer
Blood
2003
101
2932
2939
27
Imren
S
Payen
E
Westerman
KA
et al
Permanent and panerythroid correction of murine beta thalassemia by multiple lentiviral integration in hematopoietic stem cells
Proc Natl Acad Sci U S A
2002
99
14380
14385
28
Persons
DA
Hargrove
PW
Allay
ER
Hanawa
H
Nienhuis
AW
The degree of phenotypic correction of murine beta -thalassemia intermedia following lentiviral-mediated transfer of a human gamma-globin gene is influenced by chromosomal position effects and vector copy number
Blood
2003
101
2175
2183
29
Miccio
A
Cesari
R
Lotti
F
et al
In vivo selection of genetically modified erythroblastic progenitors leads to long-term correction of beta-thalassemia
Proc Natl Acad Sci U S A
2008
105
10547
10552
30
Hanawa
H
Hargrove
PW
Kepes
S
Srivastava
DK
Nienhuis
AW
Persons
DA
Extended beta-globin locus control region elements promote consistent therapeutic expression of a gamma-globin lentiviral vector in murine beta-thalassemia
Blood
2004
104
2281
2290
31
Lisowski
L
Sadelain
M
Locus control region elements HS1 and HS4 enhance the therapeutic efficacy of globin gene transfer in beta-thalassemic mice
Blood
2007
110
4175
4178
32
Puthenveetil
G
Scholes
J
Carbonell
D
et al
Successful correction of the human beta-thalassemia major phenotype using a lentiviral vector
Blood
2004
104
3445
3453
33
Arumugam
PI
Urbinati
F
Velu
CS
Higashimoto
T
Grimes
HL
Malik
P
The 3′ region of the chicken hypersensitive site-4 insulator has properties similar to its core and is required for full insulator activity
PLoS One
2009
4
e6995
34
Arumugam
PI
Scholes
J
Perelman
N
Xia
P
Yee
JK
Malik
P
Improved human beta-globin expression from self-inactivating lentiviral vectors carrying the chicken hypersensitive site-4 (cHS4) insulator element
Mol Ther
2007
15
1863
1871
35
Emery
DW
Yannaki
E
Tubb
J
Nishino
T
Li
Q
Stamatoyannopoulos
G
Development of virus vectors for gene therapy of beta chain hemoglobinopathies: flanking with a chromatin insulator reduces gamma-globin gene silencing in vivo
Blood
2002
100
2012
2019
36
Aker
M
Tubb
J
Groth
AC
et al
Extended core sequences from the cHS4 insulator are necessary for protecting retroviral vectors from silencing position effects
Hum Gene Ther
2007
18
333
343
37
Recillas-Targa
F
Pikaart
MJ
Burgess-Beusse
B
et al
Position-effect protection and enhancer blocking by the chicken beta-globin insulator are separable activities
Proc Natl Acad Sci U S A
2002
99
6883
6888
38
Gaszner
M
Felsenfeld
G
Insulators: exploiting transcriptional and epigenetic mechanisms
Nat Rev Genet
2006
7
703
713
39
Urbinati
F
Arumugam
P
Higashimoto
T
et al
Mechanism of reduction in titers from lentivirus vectors carrying large inserts in the 3′LTR
Mol Ther
2009
17
1527
36
40
Ryu
BY
Persons
DA
Evans-Galea
MV
Gray
JT
Nienhuis
AW
A chromatin insulator blocks interactions between globin regulatory elements and cellular promoters in erythroid cells
Blood Cells Mol Dis
2007
39
221
228
41
Li
CL
Xiong
D
Stamatoyannopoulos
G
Emery
DW
Genomic and functional assays demonstrate reduced gammaretroviral vector genotoxicity associated with use of the cHS4 chromatin insulator
Mol Ther
2009
17
716
724
42
Roselli
EA
Mezzadra
R
Frittoli
MC
et al
Correction of beta-thalassemia major by gene transfer in haematopoietic progenitors of pediatric patients
EMBO Mol Med
2010
8
2
8
315
328
43
Persons
DA
Allay
ER
Sabatino
DE
Kelly
P
Bodine
DM
Nienhuis
AW
Functional requirements for phenotypic correction of murine beta-thalassemia: implications for human gene therapy
Blood
2001
97
3275
3282
44
Perumbeti
A
Higashimoto
T
Urbinati
F
et al
A novel human gamma-globin gene vector for genetic correction of sickle cell anemia in a humanized sickle mouse model: critical determinants for successful correction
Blood
2009
114
1174
1185
45
Hacein-Bey-Abina
S
Garrigue
A
Wang
GP
et al
Insertional oncogenesis in 4 patients after retrovirus-mediated gene therapy of SCID-X1
J Clin Invest
2008
118
3132
3142
46
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
47
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
48
Imren
S
Fabry
ME
Westerman
KA
et al
High-level beta-globin expression and preferred intragenic integration after lentiviral transduction of human cord blood stem cells
J Clin Invest
2004
114
953
962
49
Hargrove
PW
Kepes
S
Hanawa
H
et al
Globin lentiviral vector insertions can perturb the expression of endogenous genes in beta-thalassemic hematopoietic cells
Mol Ther
2008
16
525
533
50
Arumugam
PI
Higashimoto
T
Urbinati
F
et al
Genotoxic potential of lineage-specific lentivirus vectors carrying the beta-globin locus control region
Mol Ther
2009
17
1929
1937
51
Kaiser
J
Gene therapy. Beta-thalassemia treatment succeeds, with a caveat
Science
2009
326
1468
1469
52
Hanna
J
Wernig
M
Markoulaki
S
et al
Treatment of sickle cell anemia mouse model with iPS cells generated from autologous skin
Science
2007
318
1920
1923
53
Wu
LC
Sun
CW
Ryan
TM
Pawlik
KM
Ren
J
Townes
TM
Correction of sickle cell disease by homologous recombination in embryonic stem cells
Blood
2006
108
1183
1188
54
Ye
L
Chang
JC
Lin
C
Sun
X
Yu
J
Kan
YW
Induced pluripotent stem cells offer new approach to therapy in thalassemia and sickle cell anemia and option in prenatal diagnosis in genetic diseases
Proc Natl Acad Sci U S A
2009
106
9826
9830
55
Pawliuk
R
Westerman
KA
Fabry
ME
et al
Correction of sickle cell disease in transgenic mouse models by gene therapy
Science
2001
294
2368
2371