throbber
Hematology, 2000, Vol. 4, pp. 437-455
`Reprints available direct! y from the publishcr
`Photocopying permitted by liccnsc on! y
`
`© 2000 OPA (Overseas Publishers Association) N.V.
`Published by liœnse under
`the Harwood Academie Publishers imprint,
`part of The Gordon and Breach Publishing Group.
`Printed in Malaysia.
`
`Erythropoiesis
`
`Review Article
`Slow and Steady Wins The Race? Progress in the
`Development of Vectors for Gene Therapy of
`{3-Thalassemia and Sickle Cell Disease
`
`JANE M. MCINERNEYa, MICHAEL J. NEMETHb and CHRISTOPHER H. LOWREYa,b,•
`
`"Department of Medicine, Dartmouth Medical Sclwol, Hanover, NH, USA; bDepartment of Pharmacologtj and Toxicologtj, Dartmouth
`Medical Sclwol, Hanover, NH, USA
`
`(Received 9 June 1999; In final form 23 June 1999)
`
`The cloning of the human .8-globin genes more than
`20 years ago led to predictions that ,8-thalassemia and
`sickle cell disease would be among the first mono(cid:173)
`genie diseases to be successfully treated by gene
`replacement therapy. However, despite the world(cid:173)
`wide enrollment of more than 3,000 patients in
`approved gene transfer protocols, none have involved
`therapy for these diseases. This has been due to sev(cid:173)
`era! technical hurdles that need to be overcome before
`gene replacement therapy for ,8-thalassemia and
`sickle cell disease can become practical. These prob(cid:173)
`lems include inefficient transduction of hematopoi(cid:173)
`etic stem cells and an inability to achieve consistent,
`long-term, high-level expression of transferred ,8-like
`globin genes with current gene transfer vectors. In
`this review we highlight the relationships between
`understanding the fondamental mechanisms of .8-
`globin gene locus function and basic vector biology
`and the development of strategies for .8-globin gene
`replacement therapy. Des pite slow initial progress in
`this field, recent advances in a variety of critical areas
`provide hope that clinical trials may not be far away.
`
`Keywords: Gene therapy, ,8-thalassemia, sickle cell disease,
`,8-globin, retrovirus, lentivirus, chroma tin structure
`
`INTRODUCTION
`
`Mutations of the a- and ,B-globin gene loci com(cid:173)
`prise the most prevalent group of inherited single
`gene diseases. It has been estimated that seven
`percent of the world's population are heterozy(cid:173)
`gous carriers of clinically significant mutations
`affecting the expression of the globin genes or
`the function of their gene products [1]. Mutations
`within the genes of the .B-globin locus give rise
`to ,B-thalassemia and the ,B-hemoglobinopathies,
`including sickle cell disease (SCD). While the
`high prevalence of these mutations appears to
`have arisen because they lessen the morbidity
`of malarial infection in the heterozygous state
`(reviewed in [2]), they continue to result in
`early death and chronic debilitating disease in
`the homozygous state [1]. The underlying patho(cid:173)
`physiologies of these diseases in volve destruction
`of erythrocytes due to abnormal polymerization
`
`*Corresponding au thor. Fax: (603) 650-1129. E-mail: c.lowrey@dartmouth.edu
`
`437
`
`SKI Exhibit 2057
`Page 1 of 19
`
`

`

`438
`
`J. M. MCINERNEY et al.
`
`of hemoglobin molecules containing the sickle (3-
`chain (sickle cell disease) and o:-chain imbalance
`((3-thalassemia) [3,4]. Because erythrocytes are
`derived from the hematopoietic stem cell (HSC),
`transfer of a functional human adult ((3) or fetal (-y)
`globin gene to a patient's stem cells could cure or
`lessen the severity of these diseases. -y-globin gene
`transfer is particularly attractive for seo since the
`-y-globin protein does not participate in the sick(cid:173)
`ling reaction and thus confers greater protection
`than the (3-globin protein [5]. A major rationale
`for using (3-thalassemia and seo as models for
`the development of gene therapy strategies is
`that less than full reconstitution of normal (3-
`or -y-globin gene expression is likely to pro(cid:173)
`duce significant clinical benefit as co-inheritance
`of mutations that confer hereditary persistence
`of fetal hemoglobin (HPFH) have an attenuated
`phenotype [6-8]. A second rationale for (3-globin
`replacement therapy is that significant clinical
`benefits have been achieved using the pharma(cid:173)
`cologie agents 5-azacytidine, hydroxyurea and
`sodium butyrate as inducers of -y-globin synthe(cid:173)
`sis (reviewed in [9,10]). Gene replacement therapy
`offers significant advantages over these phar(cid:173)
`macologie strategies in that a single treatment
`could permanently restore (3-globin (or -y-globin)
`protein production and both known and poten(cid:173)
`tial side-effects of long-term drug therapy could
`be avoided [9]. Successful gene transfer therapy
`would also offer a potentially safer,less expensive
`and more widely applicable curative therapy than
`allogeneic bone marrow transplantation [11,12].
`The (3-globin genes were among the first human
`genes to be cloned [13-15]. This led to specula(cid:173)
`tion that genetic diseases involving these genes
`would be among the first diseases to be treated
`using gene therapy. However, ten years after the
`approval of the first human gene transfer proto(cid:173)
`col [16] and after the treatment of more than three
`thousand patients worldwide, no (3-thalassemia
`or seo patient has been treated in an approved
`gene therapy trial [16,17]. This disappointing lack
`of progress has largely been due to several tech(cid:173)
`nical hurdles which must be overcome before
`
`gene therapy can be applied to these diseases.
`These challenges include developing more effi(cid:173)
`cient methods for the stable transfer of genetic
`material into the genomic ONA of hematopoietic
`stem cells and achieving long-term, high-level
`expression from integrated {3- or -y-globin genes.
`So far, most gene therapy approaches to the
`hemoglobinopathies have utilized retroviral vec(cid:173)
`tors. This gene transfer system has several advan(cid:173)
`tages including the ability to efficiently transduce
`a variety of cell types, the ability to stahly inte(cid:173)
`grate the therapeutic gene into host genomic
`ONA, a size capacity able to accommodate (3-
`globin regulatory and coding sequences and a
`proven clinical safety record [18,19]. Overall,
`retroviral vectors have been the most commonly
`used vector for gene transfer protocols in gen(cid:173)
`eral and for protocols targeting bone marrow
`stem cells in particular [16,17]. Unfortunately,
`retroviral vectors also have shortcomings includ(cid:173)
`ing relatively low level transduction efficiency
`of hematopoietic stem cells. Recent studies have
`shown that this is partly due to low level expres(cid:173)
`sion of appropriate viral receptors on the sur(cid:173)
`face of hematopoietic stem cells [20]. eommonly
`used retroviral vectors, which are based on the
`Moloney Murine Leukemia Virus (MML V) and
`related members of the same retroviral species,
`are also unable to efficiently integrate into the
`genomic ONA of non-cycling target cells. Because
`of these problems, expression of transferred genes
`in human peripheral blood cells remains too
`low to achieve therapeutic benefit [21,22]. New
`strategies designed to overcome these problems
`include up-regulation of retroviral receptors on
`hematopoietic stem cells [23], induction of stem
`cell cycling with hematopoietic growth factors
`and drugs [24,25], pseudo-typing of retroviral
`vector envelope proteins [26], and the recent use
`of lentiviral vectors, which are retroviruses which
`do not require cell cycling for stable integration
`into hematopoietic stem cells [27]. Another novel
`strategy involves the transfer, not of the (3- or -y(cid:173)
`globin genes themselves, but of genes coding for
`regula tory proteins which up-regulate expression
`
`SKI Exhibit 2057
`Page 2 of 19
`
`

`

`GENE THERAPY IN HEMOGLOBINOP A THY
`
`439
`
`of the ')'-globin genes. The stage selector protein
`described by Jane and colleagues is an example
`of such a protein which could participate in the
`reactivation of the fetal globin genes [28,29).
`The second major obstacle to successful gene
`therapy for the ;3-hemoglobinopathies is the
`inability to obtain consistent long-term, high(cid:173)
`level expression of integrated therapeutic genes.
`The importance of this concept was emphasized
`in the report of the Panel to Assess the NIH
`Investment in Research on Gene Therapy which
`noted that " ... very little researclz effort is focused
`on
`the meclzanisms that govem maintenance or
`shutoff of gene expression following gene delivery
`in gene therapy experiments" [30]. The report
`urged the NIH ". . . to give high priority to
`basic researc/z to elucidate lww recipient cells and
`particularly stem cells, lwzdle and express foreign
`DNA sequences." [30]. This concem is especially
`relevant to gene therapy for 3-thalassemia and the
`hemoglobinopathies where !ife-long expression
`from the integrated therapeutic gene is a goal.
`Clearly, the normal J-globin genes are capa(cid:173)
`ble of long-term, high-level expression. This has
`provided a major rationale for studying normal 3-
`globin locus structure and function to define the
`mechanisms by which appropriate expression of
`the endogenous genes is achieved. By incorpo(cid:173)
`rating globin gene regulatory elements (such as
`promoters and enhancers) into gene therapy vec(cid:173)
`tors, it has been hoped that similar expression
`of transferred genes could be attained. How(cid:173)
`ever, after twenty years of investigation into the
`regulation of J-globin gene expression and the
`application of the results of this research to the
`development of ;J-globin gene transfer vectors,
`expression levels adequate to begin human trials
`have yet to be achieved.
`In this review we focus on how understanding
`the basic science of gene regulation in general,
`and of the 8-globin locus in particular, has driven
`the development of 8-globin gene replacement
`vectors. We begin by discussing the role chro(cid:173)
`matin structure plays in the regulation of genes
`integrated into genomic DNA, as understanding
`
`this process appears to be important to devel(cid:173)
`oping vectors capable of high-level expression.
`We then review current information on the reg(cid:173)
`ulation of the genes of the /3-globin locus. Next
`we discuss the development of /3-globin gene
`transfer vectors. Because retroviruses have been
`the principle mode! for the development of gene
`replacement strategies for the /3-globin related
`diseases, we have primarily focused on this sys(cid:173)
`tem. Finally, we discuss how novel insights into
`the chroma tin structure of the 8-globin locus are
`leading to the development of a new generation of
`retroviral vectors specifically designed to address
`the problems of inconsistent and low-level gene
`expression.
`
`CHROMA TIN STRUCTURE AND GENE
`EXPRESSION
`
`Within the nucleus of eukaryotic cells genomic
`DNA is packaged by specifie proteins in a highly
`structured fashion [31]. This combination of pro(cid:173)
`teins and nuclear DNA is termed chromatin.
`The structure of chromatin is highly dynamic
`and plays an important role in the regulation
`of normal gene expression (reviewed in [32])
`and expression resulting from gene therapy vec(cid:173)
`tors integrated into nuclear chromatin [33]. The
`first leve! of packaging involves the wrapping of
`DNA about disc-shaped, multi-protein structures
`ca lied nucleosomes. The nucleosomes themselves
`are comprised of four related proteins termed his(cid:173)
`tones. Each nucleosome packages approximately
`200 bp of DNA which is wrapped twice around
`the structure. Regions of DNA which contain
`actively expressed genes (such as the p-globin
`locus in erythroid cells) are loosely packaged and
`the nucleosomes of the region assume a beads(cid:173)
`on-a-string arrangement. This is associated with
`the formation of enzymatically modified histone
`proteins which become hyperacetylated. Regions
`of DNA which do not contain actively expressed
`genes (such as the p-globin gene locus in non(cid:173)
`erythroid cells) are much more compacted. The
`
`SKI Exhibit 2057
`Page 3 of 19
`
`

`

`440
`
`J. M. MCINERNEY et al.
`
`structure regardless of their site of integration
`within the genome.
`
`REGULATION OF HUMAN ,6-GLOBIN
`GENE EXPRESSION
`
`Perhaps because the human ,B-globin genes were
`among the first mammalian genes to be cloned,
`the human ,B-globin gene locus has served as a
`premiere model for studying the regulation of
`complex mammalian loci. As shown in Figure 1,
`the genes of the locus are arranged in their order
`of developmental expression, from the embryonic
`e-globin gene to the fetal -y-globin genes and the
`adult 8-and ,B-globin genes [2]. Early studies of
`the locus identified promoters and both intronic
`and downstream enhancers of ,B-globin locus
`gene expression (Figure lB). In earl y gene transfer
`experiments, these elements were able to direct
`erythroid specifie expression of the genes but
`expression levels in both transgenic mice and
`tissue culture celllines were very low and highly
`variable [38,39]. Clearly, important elements for
`gene regulation were missing.
`
`nucleosomes in these regions are thought to form
`tightly condensed helical arrays called solenoids
`or 30 nanometer fibers [31]. Inactive areas of
`chromatin are also characterized by DNA methy-
`lation and hypoacetylation of histone proteins
`(reviewed in [34]). Higher levels of packag(cid:173)
`ing also occur in inactive areas but are much
`less well defined. Experimentall~r, differences in
`chromatin structure are often assessed by their
`sensitivity to digestion by nucleases, which are
`enzymes capable of digesting DNA. DNase I is
`a nuclease commonly used in chromatin studies.
`Tightly compacted, inactive areas of chromatin
`are highly resistant to DNase I digestion, while
`transcriptionally active areas of chromatin are
`more sensitive. Transcriptional regulatory ele(cid:173)
`ments such as promoters and enhancers are
`often hypersensitive to nuclease digestion due
`to the displacement or disruption of nucleosomes
`within these areas exposing DNA to the nucle(cid:173)
`ase. These regions of locally altered chromatin
`structure are termed nuclease or DNase I hyper(cid:173)
`sensitive sites or "HSs".
`Chromatin structure is relevant to the devel(cid:173)
`opment of gene transfer vectors because the
`expression of a stably integrated therapeutic gene
`is likely to depend on the surrounding chroma tin
`structure. If the gene is integrated into a com(cid:173)
`pacted, transcriptionally-inactive region of the
`genome it is unlikely to be expressed. This phe(cid:173)
`nomenon is termed "position-dependent expres(cid:173)
`sion" as expression is dependent on the site of
`integration. Position-independent expression or,
`stated in a different way, consistent expression
`is an important goal in the development of gene
`transfer vectors. Furthermore, it has been shown
`that integrated genes which initially express at
`high levels may be subject to transcriptional
`silencing over time [35-37]. This is thought to
`result from a closing down of the chroma tin struc(cid:173)
`ture around the integrated gene. As is discussed
`below, it would be advantageous to be able to
`include elements within gene therapy vectors
`which are able to independently open and main(cid:173)
`tain surrounding domains of active chromatin
`
`100~[3
`~·GLOBIN
`
`:
`
`CHAIN
`SYNTHESIS
`('l'o)
`
`E
`
`EMBAVO
`
`FETlJS
`
`1
`
`1
`BIRTH
`
`~
`ADUL T
`
`6 MONTHS
`
`A
`
`8
`
`• LCR Ill
`5 4 3 2 1
`..
`
`Erythrold spaclllc
`DNasal HSs
`
`E
`
`-10kb
`Chromosome 11 p15.5
`
`FIGURE 1 The_ human .6-&lobin gene locus. A) Expression
`IS developmentally
`regulated.
`the _.6-g!obm genes
`of
`B) Orgamzahon of the locus. Globin genes are depicted
`as black boxes, gene promoters as white boxes and local
`enhanc~rs as di~monds. The .6-globin locus control region
`(LCR) 1s compnsed of five domains of altered chromatin
`strucrure termed DNase 1 HSs. Four of these HSs are erythtoid
`specifie.
`
`SKI Exhibit 2057
`Page 4 of 19
`
`

`

`GENE THERAPY IN HEMOGLOBINOP ATHY
`
`441
`
`The description of the ,8-globin locus control
`in 1987 apparently identified
`region (LCR)
`these critical elements (Figure lB). The LCR is
`comprised of five DNase 1 HSs, four of which
`(5' HS 1-4) are erythroid specifie and are
`able to direct consistent, high-level, position(cid:173)
`independent expression of linked globin genes in
`transgenic mice [40,41]. The functional activities
`of the LCR are primarily mediated by specifie
`core sequences found near the centers of each
`HS domain [42-52]. Each of the 200-400 bp HS
`core elements contain evolutionarily conserved
`binding sites for factors primarily expressed in
`hematopoietic cells, such as NF-E2, GATA-1,
`SSP, and EKLF, as well as more ubiquitous
`factors such as Sp1, YY1, and CDP (reviewed
`in [53]). On a chromatin structural level these
`nuclease sensitive regions are characterized by
`highly specifie nucleosome positioning including
`regions where nucleosomes are disrupted or
`displaced [54,55]. The active elements of the
`LCR HSs closely correspond to the regions
`of nucleosome displacement [47,48,52,54,55]. In
`transgenic mice containing multiple copies of
`linked transgenes and individual LCR elements,
`HS2, HS3, and HS4 are each able to confer
`position independent expression [56]. However,
`only when all of these LCR elements are
`present as full-length, or smaller "mini" or
`"micro" LCRs is near-normal expression of the
`transgene consistently obtained [41,51,57]. In
`single copy transgenic mice HS3 is uniquely able
`to confer position-independent expression [58]. A
`well characterized, classical enhancer element is
`present within the core region of HS2 [59,60]. This
`enhancer activity requires binding sites for the
`hematopoietic transcription factor NF-E2 [52,61].
`The other LCR HSs do not contain significant
`enhancer activity. While relatively little research
`has been performed on HSs 1 and 5, HS1 may
`be important for consistent expression and for
`mediating interactions between the LCR and
`the ,8-globin promoter [62] and, as discussed
`below, HS5 may function as a chroma tin structure
`insulator [63].
`
`Despite intense investigation the mechanisms
`by which the elements of the LCR are able to
`influence expression of the genes of the human ,8-
`globin locus remain unclear. Most current models
`propose the formation of a holocomplex in which
`the core elements of the LCR functionally inter act.
`This holocomplex is then envisioned to form a
`three dimensionallooping structure that is able
`to interact with the promoters of the globin genes
`(reviewed in [53,64]).
`
`DEVELOPMENT OF ,8-GLOBIN GENE
`TRANSFER VECTORS
`
`Retroviral Transfer Vectors
`
`Since the development of the first gene transfer
`trial more than ten years aga, retroviral vec(cid:173)
`tors have been the most frequently used method
`for stable integration of genes into target cell
`genomes [17]. Retroviruses are enveloped RNA
`viruses which enter cells through interactions
`with specifie receptors on the cell surface [65].
`The presence of these receptors on target cells
`is an important determinant of the virus's abil(cid:173)
`ity to transduce a specifie cell type. The infecting
`virus particle carries a single-strand RNA genome
`which is converted into a double-strand DNA
`provirus by the virally-encoded reverse transcrip(cid:173)
`tase within the infected cell. The viral integrase,
`in cooperation with cellular proteins, then medi(cid:173)
`ates incorporation of the DNA provirus into the
`genomic DNA of the target cell [66].
`The retroviral system has been adapted to
`serve as a safe and effective method for stahly
`transferring potentially therapeutic genes into
`the genomic DNA of target cells (Figure 2).
`The retroviral gene transfer vector is commonly
`constructed within the context of a bacterial
`plasmid. The vector contains the viral long
`terminal repeats (LTRs) and packaging signal ('lj;)
`as well as the gene or genes to be transferred.
`Because the retroviral structural genes gag, pol
`and env are not contained within the vector, it
`is unable to direct the formation of infectious
`
`SKI Exhibit 2057
`Page 5 of 19
`
`

`

`442
`
`J. M. MCINERNEY et al.
`
`(..._5_' -L-T-R'lf_T_a_bz_~_~_Sj_,~_fl_J--3-' _L_T_,R) ~
`
`Retroviral Vector
`(in bacterial plasmid)
`
`Packaging Cell
`
`Target Cell
`
`FIGURE 2 Retroviral gene transfer. A retroviral vector
`containing
`the gene
`to be
`transferred
`(transgene)
`is
`constructed in the context of a bacterial plasmid. The plasmid
`is stably introduced into a packaging cellline (step 1) where
`transcription of the retroviral sequences produces the single
`stranded RNA viral genome (step 2). The packaging cellline
`contains the retroviral structural genes (gag, pol and env)
`necessary for producing infectious virions (step 3) which
`then bind to a specifie cell surface receptor on the target
`cel!. Following uptake into the target cel!, the viral genome
`undergoes reverse transcription by the viral polymerase
`resulting in a double stranded DNA copy of the viral genome
`(step 4) which is then stably integrated into the genomic DNA
`of the target cell (step 5). The transgene is then transcribed,
`ultimately producing the protein product of the transferred
`gene (step 6).
`
`vmons in cells other than specially designed
`"packaging" cell lines which have been stably
`transduced with these genes [67]. This allows the
`structural genes of the virus to be replaced by
`the gene or genes to be transferred as well as
`their associated regulatory elements. Following
`transfection of the retroviral vector into the
`packaging cell an RNA copy of the vector is
`
`produced which is then packaged into functional
`viral particles. These viruses are then used to
`transfer the gene or genes of interest to the
`target cells where they are incorporated into the
`target cell genome. Infection of the target cell is
`mediated by binding to specifie receptors on the
`cell surface. Since the genes for the viral structural
`proteins are not packaged, the virus particles can
`infect a single cell, but subsequent production of
`virions by the target cell can not occur. Once
`the vector RNA is inside the target cell it is
`converted to double stranded DNA in the process
`of reverse transcription. This is mediated by the
`viral polymerase gene (pol) product which is
`packaged within the viral particle. The DNA form
`of the vector is then incorporated into the host
`genomic DNA. Transcription of the integrated
`viral sequences then leads to the production of
`the protein coded for by the transferred gene.
`While there are many positive aspects to using
`retroviruses for transferring ,8-globin genes into
`bone marrow stem cells, there are also disad(cid:173)
`vantages. One is that marrow stem cells have
`relatively low levels of viral receptors on their
`surfaces [20]. Strategies to overcome this problem
`include induction of receptor expression on the
`stem cells. A second disadvantage is an inabil(cid:173)
`ity of retroviruses of the Murine Sarcoma and
`Leukemia Virus species (i.e. MMLV) to integrate
`proviral DNA into the genomic DNA of non(cid:173)
`cycling cells [68]. Hematopoietic growth factors
`and the drug 5-fluorouracil have been used to
`induce stem cell cycling and increase transduc(cid:173)
`tion efficiency [24,25]. An alternative approach
`has been to use other viral-based gene transfer
`vectors. The most promising of these include vec(cid:173)
`tors based on adeno-associated viruses (AA V)
`and lentiviruses. Recombinant AA V vectors car(cid:173)
`rying the human 1'-globin gene have been used
`to mediate stable gene transfer with high-level
`expression in K562 human erythroid tissue cul(cid:173)
`ture cells [69-71]. Lentiviruses are also retro(cid:173)
`viruses but comprise a separate genus from the
`MMLV -related retroviruses. They are structurally
`distinct in that their genome and structure are
`
`SKI Exhibit 2057
`Page 6 of 19
`
`

`

`5' LTR
`
`P-GlobinGene
`
`3' LTR
`
`ljl
`
`ljl
`
`'::;;:J
`
`·v~ HS Core
`
`Intron 2 Deletion
`Mutagcnesis of poly A/splicc sites
`'\1
`
`ljl
`
`Intron 2 Deletion
`'V
`'=:fi%>' 1
`
`ljl
`
`11
`
`HS2 • HS3- HS4 ---
`
`A
`
`B
`
`c
`
`D
`
`E
`
`much more complex. Functionally, they exhibit
`the ability to infect non-cycling cells such as qui(cid:173)
`escent bane marrow stem cells [27]. While it has
`been difficult to develop effective packaging cell
`lines for these vectors, significant progress has
`recently been made in this area [72]. To date no
`published reports have demonstrated the appli(cid:173)
`cation of these vectors to globin gene replacement
`therapy.
`
`First Generation Retroviral Gene Transfer
`Vectors
`
`Early attempts to develop retroviral gene ther(cid:173)
`apy vectors for the hemoglobinopathies utilized
`the human ,8-globin genes in combination with
`their proximal regulatory elements [73-78]. An
`example of these vectors is shawn in Figure 3B.
`These initial vectors were hindered by law titer
`and generally very law ,8-globin gene expres(cid:173)
`sion. Subsequent studies demonstrated that the
`law titer was due, in part, to sequences within
`the second intron and within other untrans(cid:173)
`lated sequences S' and 3' of the ,8-globin gene
`[78-80]. These sequences appeared to cause
`proviral instability and law titer by acting as
`polyadenylation signais or cryptic splice sites
`[81]. Higher titers were achieved with vectors
`containing the ,8-globin eDNA than with those
`containing the genomic sequence [78-80]. How(cid:173)
`ever, it was also shawn that sequences within
`the second intron contribute to high-level gene
`expression [43,78,79]. Therefore, to reduce the
`problem of genetic instability while retaining the
`intronic elements, ,8-globin retroviral vectors are
`now commonly designed so that the gene is in
`reverse orientation relative to viral transcription
`(Figure 3A vs. 3B) [73,75,82]. This reduces vec(cid:173)
`tor rearrangement caused by splicing of the viral
`RNA genome because potential splice sites do
`not have their characteristic sequence in this ori(cid:173)
`entation and consequently are not recognized by
`the RNA splicing machinery. Following integra(cid:173)
`tion of the double stranded provirus, the reversed
`
`GENE THERAPY IN HEMOGLOBINOPA THY
`
`443
`
`Retroviral Construct
`
`References
`
`[78, 79]
`
`[73 -79, 83]
`
`[60, 87, 90]
`
`[81, 89, 91, 92]
`
`[33,80]
`
`FIGURE 3 Evolution of retroviral ,B-globin gene transfer
`vectors. A) Retroviral vector containing the ,B-globin gene
`in the "sense" orientation relative to the viral genome. B) The
`orientation of the ,6-globin gene is reversed to decrease the
`incidence of splicing of the viral sequences during the RNA
`stage of viral production. C) lndividual LCR HS cores are
`added to the vector in an effort to increase ,6-globin expression.
`D) Multiple LCR HS cores are added to the vector, potential
`splice sites are mutated and an inhibitory portion of intron
`2 is deleted. E) Different orientations of LCR HS cores are
`evaluated to determine their ability to improve ,8-globin
`expression.
`
`gene is capable of normal expression on the oppo(cid:173)
`site strand.
`With these earl y vectors, ,8- globin expression in
`mouse erythroleukemia (MEL) cells was highly
`varied, averaging between 2 and 28% percent
`of endogenous mouse ,8-major gene expression
`[73-75,78,79]. These results strongly suggested
`that the site of integration of the retroviral vector
`has a profound effect on expression of the ,8-
`globin gene. In experiments involving gene trans(cid:173)
`fer to murine bane marrow stem cells expression
`was tissue-specifie and less varied, but reached
`at most one percent of mouse ,8-major expres(cid:173)
`sion [76,77,83]. ,8- globin vectors thatincorporated
`the 3' ,8-globin enhancer did not display consis(cid:173)
`tent improvement in overall expression levels or
`position-independent expression [77,79,84-86].
`
`SKI Exhibit 2057
`Page 7 of 19
`
`

`

`444
`
`J. M. MCINERNEY et al.
`
`From these early experiments it was evident
`that retroviral vectors incorporating the pro(cid:173)
`moters and enhancers of the ,B-globin genes
`were insufficient to direct high-level position(cid:173)
`independent expression of ,B-globin genes and
`that additional or alternative gene regula tory ele(cid:173)
`ments would be required to achieve high-level
`position-independent expression using retroviral
`gene therapy vectors.
`
`Second Generation Retroviral Vectors:
`Incorporation of LCR Elements
`
`The ,B-globin LCR is generally thought to be
`required for normal expression of the globin
`genes in human hematopoietic cells and in
`transgenic models of ,B-globin gene expression.
`The discovery of the LCR and the characterization
`of its functional properties led to the expectation
`that the inclusion of these elements in ,B-globin
`gene transfer vectors would finally provide
`high-level position-independent expression in
`erythroid cells. Because the packaging capacity
`of retroviral vectors is limited to approximately
`8-9 kb, the entire LCR region was too large for
`inclusion in ,B-globin vectors [67]. However, as
`noted above, experiments in transgenic mice had
`delineated functional HS cores of only a few
`hundred base pairs in size that contained most of
`the relevant LCR activity and conferred high-level
`position-independent expression. In numerous
`studies these elements were incorporated into a
`second generation of ,B-globin retroviral vectors
`(Figure 3C-3E).
`One of the first studies to utilize this strategy
`was reported by Novak et al. ,B-globin retroviral
`vectors were constructed so that each contained
`one of the HS core elements (Figure 3C) [87].
`These retroviral vectors were then used to trans(cid:173)
`duce MEL cells. An encouraging result from
`these experiments was that HS2, HS3, and HS4
`increased average ,B-globin expression by approx(cid:173)
`imately 20-fold, 4-fold, and 6-fold respectively.
`The combination of HS3 and HS4 increased
`
`expression 3-fold. However, the retroviruses in
`this study continued to show low producer cell
`titers in the range of 104 to 105 cfu/ml, typical
`of similarly designed vectors. Vectors containing
`HS2 and HS3-HS4 also demonstrated frequent
`genetic rearrangement. All vectors tested contin(cid:173)
`ued to show highly variable expression indicating
`that the inclusion of HS cores did not produce
`position independent expression.
`Previous studies in transgenic mice and tissue
`culture cell lines, as well as the retroviral study
`of Novak et al. above, indicated strong enhancer
`activity within HS2 [42,44,59,88]. In an attempt
`to circumvent the problem of genetic instability
`associated with the use of HS2 in retroviral
`vectors, Chang et al. reduced the size of HS2 to
`a 36 bp element containing tandem NF-E2/ AP-l
`binding si tes [ 60,61]. Incorpora ti on ofthis element
`into a ,B-globin vector increased expression two(cid:173)
`fold. It also resulted in reduced vector instability
`compared to vectors containing either a 732
`or 412 bp HS2 core element. However, vector
`rearrangement was still observed approximately
`fifty percent of the time and viral titer remained
`low- in the range of 104 cfu/ml. The use of
`multiple copies of the 36 bp HS2 enhancer
`element led to increased vector rearrangement.
`Attempts to overcome the problems of low ti ter
`and genetic rearrangement included studies by
`the Gelinas and Leboulch laboratories. In the first
`of these studies a 374 bp deletion within intron
`2 of the ,B-globin gene improved the titer of a ,B(cid:173)
`globin vector 10-fold without adversely affecting
`gene expression [89]. Leboulch et al. attempted
`to correct these problems in ,B-globin vectors
`which incorporated LCR elements by perform(cid:173)
`ing sequence analysis of the human ,B-globin
`gene and both S' and 3' untranslated regions
`[81]. This led to the identification of poten(cid:173)
`tial polyadenylation signais and splice sites. In
`this study, all ,B-globin LCR vectors containing
`the wild-type ,B-globin sequences were unsta(cid:173)
`ble and characterized by low titer regardless
`of the LCR derivatives used. Targeted muta(cid:173)
`genesis of putative splice sites within intron 2
`
`SKI Exhibit 2057
`Page 8 of 19
`
`

`

`GENE THERAPY IN HEMOGLOBINOP A THY
`
`445
`
`and the 3' flanking region of the ,13-globin gene
`and deletion of a 372 bp region within intron 2
`(Figure 3D) promoted the stability and increased
`titer 10-fold of an HS2-,13-globin vector. This vec(cid:173)
`tor was also stable in murine bone marrow cells.
`These mutations also allowed stable transmission
`of vectors with multiple LCR elements. Incor(cid:173)
`poration of HS2 increased expression 14-f

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket