throbber
10 May 2005 22:26
`
`AR
`
`AR261-BI74-24.tex
`
`XMLPublishSM(2004/02/24)
`P1: JRX
`10.1146/annurev.biochem.74.050304.091637
`
`Annu. Rev. Biochem. 2005. 74:711–38
`doi: 10.1146/annurev.biochem.74.050304.091637
`Copyright c(cid:1) 2005 by Annual Reviews. All rights reserved
`First published online as a Review in Advance on March 11, 2005
`
`GENE THERAPY: Twenty-First Century Medicine
`
`Inder M. Verma and Matthew D. Weitzman
`Laboratory of Genetics, The Salk Institute, La Jolla, California 92037;
`email: verma@salk.edu, weitzman@salk.edu
`
`Key Words
`viral vectors, retrovirus/lentivirus, adenovirus, AAV, clinical trials
`■ Abstract Broadly defined, the concept of gene therapy involves the transfer of
`genetic material into a cell, tissue, or whole organ, with the goal of curing a disease or at
`least improving the clinical status of a patient. A key factor in the success of gene ther-
`apy is the development of delivery systems that are capable of efficient gene transfer in
`a variety of tissues, without causing any associated pathogenic effects. Vectors based
`upon many different viral systems, including retroviruses, lentiviruses, adenoviruses,
`and adeno-associated viruses, currently offer the best choice for efficient gene delivery.
`Their performance and pathogenicity has been evaluated in animal models, and encour-
`aging results form the basis for clinical trials to treat genetic disorders and acquired
`diseases. Despite some initial success in these trials, vector development remains a
`seminal concern for improved gene therapy technologies.
`
`CONTENTS
`
`INTRODUCTION . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 711
`VIRAL VECTORS . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 713
`RNA VIRUS VECTORS . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 714
`Retroviruses . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 714
`DNA VIRUS VECTORS . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 719
`Adenovirus . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 719
`Adeno-Associated Virus . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 721
`Herpesvirus . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 723
`VECTOR TROPISM AND THE SPECIFICITY OF TRANSDUCTION . . . . . . . . . . 725
`VECTOR RECOGNITION, PROCESSING, AND INTEGRATION . . . . . . . . . . . . . . 727
`Gene Regulation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 728
`CLINICAL TRIALS: SUCCESSES AND SETBACKS . . . . . . . . . . . . . . . . . . . . . . . . 729
`PERSPECTIVES: WHAT IS NEXT? . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 732
`
`INTRODUCTION
`
`Gene therapy is a form of molecular medicine that has the potential to influence
`significantly human health in this century. It promises to provide new treatments
`for a large number of inherited and acquired diseases (1). The basic concept of
`
`0066-4154/05/0707-0711$20.00
`
`711
`
`Annu. Rev. Biochem. 2005.74:711-738. Downloaded from www.annualreviews.org
`
`by Pohang University of Science and Technology (POSTECH) on 10/03/13. For personal use only.
`
`SKI Exhibit 2036
`Page 1 of 30
`
`

`

`10 May 2005 22:26
`
`AR
`
`AR261-BI74-24.tex
`
`XMLPublishSM(2004/02/24)
`
`P1: JRX
`
`712
`
`VERMA (cid:1) WEITZMAN
`
`gene therapy is simple: introduce into target cells a piece of genetic material that
`will result in either a cure for the disease or a slowdown in the progression of
`the disease. To achieve this goal, gene therapy requires technologies capable of
`gene transfer into a wide variety of cells, tissues, and organs. One of the biggest
`stumbling blocks to successful widespread application of such genetic treatments
`is the development of safe and effective vectors with which to ferry genetic material
`into a cell.
`The process of gene delivery and expression is known as transduction. Suc-
`cessful transduction requires overcoming a number of obstacles that are common
`to all vector systems (2). The first issue to be addressed is that of production. An
`ideal vector should be one that can be produced in a highly concentrated form,
`using a convenient and reproducible production scheme. This has been a chal-
`lenge for many of the currently used vector systems, but in many cases creative
`approaches have overcome this barrier. The vector must be capable of targeting
`the cell type most appropriate for the disease, whether it be dividing or nondi-
`viding cells. Understanding of the transduction process through studies of vector
`uptake, intracellular trafficking, and gene regulation has facilitated the develop-
`ment of efficient vehicles for gene delivery. In many cases it would be desirable
`to achieve stable, sustained gene expression, which requires either integration of
`the vector DNA into the host DNA or maintenance as an episome. When us-
`ing integrating vector systems, it is important to consider the potential hazards
`of insertional mutagenesis, and thus vectors capable of site-specific integration
`will be attractive. In many cases, expression of the therapeutic gene will require
`exquisite regulation, and thus the transcriptional unit must be capable of respond-
`ing to manipulations of its regulatory elements. Finally, no pathogenic or adverse
`effects should be elicited by vector transduction, including undesirable immune
`responses.
`Vectors that have been developed to overcome these obstacles fall into two
`broad categories: nonviral and viral vectors (1). The nonviral vectors consist of
`naked DNA delivered by injection, liposomes (cationic lipids mixed with nu-
`cleic acids), nanoparticles, and other means. Although nonviral vectors can be
`produced in relatively large amounts and are likely to present minimal toxic or
`immunological problems, presently they suffer from inefficient gene transfer. In
`addition, expression of the foreign gene tends to be transient, precluding their
`application to many diseased states in which sustained and high-level expression
`of the transgene is required. It is likely that future gene therapy protocols will use
`novel innovations to improve on the efficiency of nonviral vector systems, often
`building upon observations from viral vector transduction. Viral vectors are de-
`rived from viruses with either RNA or DNA genomes and are represented as both
`integrating and nonintegrating vectors. The former holds the promise of lifelong
`expression of the deficient gene product. Efficient gene transduction can also be
`achieved from vectors that are maintained as episomes, especially in nondividing
`cells.
`
`Annu. Rev. Biochem. 2005.74:711-738. Downloaded from www.annualreviews.org
`
`by Pohang University of Science and Technology (POSTECH) on 10/03/13. For personal use only.
`
`SKI Exhibit 2036
`Page 2 of 30
`
`

`

`10 May 2005 22:26
`
`AR
`
`AR261-BI74-24.tex
`
`XMLPublishSM(2004/02/24)
`
`P1: JRX
`
`GENE THERAPY
`
`713
`
`VIRAL VECTORS
`
`The basic concept of viral vectors is to harness the innate ability of viruses to de-
`liver genetic material into the infected cell. In general, the major preoccupation of
`viruses is to replicate and produce copious amounts of progeny. Most viruses gain
`little by killing the host, but unfortunately many viral infections lead to deleterious
`effects on the host, accompanied by destruction of infected host cells. Damag-
`ing effects can be caused by induction of genes whose products are hazardous
`to the host or by acquiring host genomic material that can lead to pathogenesis.
`The basic principle of turning these pathogens into delivery systems relies on the
`ability to separate the components needed for replication from those capable of
`causing disease (see Figure 1). The first step of viral vector design is, therefore,
`
`Figure 1 Principle of generating a viral vector. (a) Converting a virus into a recombinant
`vector. Schematic of a generic viral genome is shown with genes that are involved in replica-
`tion, production of the virion, and pathogenicity of the virus. The genome is flanked by cis-
`acting sequences that provide the viral origin of replication and the signal for encapsidation.
`The packaging construct contains only genes that encode functions required for replication
`and structural proteins. The vector construct contains the essential cis-acting sequences and
`the transgene cassette that contains the required transcriptional regulatory elements. (b) The
`packaging and vector constructs are introduced into the packaging cell by transfection, by
`infection with helper virus, or by generating stable cell lines. Proteins required for replica-
`tion and assembly of the virion are expressed from the packaging construct, and the repli-
`cated vector genomes are encapsidated into virus particles to generate the recombinant viral
`vector.
`
`Annu. Rev. Biochem. 2005.74:711-738. Downloaded from www.annualreviews.org
`
`by Pohang University of Science and Technology (POSTECH) on 10/03/13. For personal use only.
`
`SKI Exhibit 2036
`Page 3 of 30
`
`

`

`10 May 2005 22:26
`
`AR
`
`AR261-BI74-24.tex
`
`XMLPublishSM(2004/02/24)
`
`P1: JRX
`
`714
`
`VERMA (cid:1) WEITZMAN
`
`to identify the viral sequences required for replication, assembly of viral parti-
`cles, packaging of the viral genome, and delivery of the transgene into the target
`cells. Next, dispensable genes are deleted from the viral genome to reduce repli-
`cation and pathogenicity, as well as expression of immunogenic viral antigens.
`The gene of interest together with transcriptional regulatory elements (referred to
`as the transgene) are inserted into the vector construct, and a recombinant virus
`is generated by supplying the missing gene products required for replication and
`virion production. The more genes that are removed from the virus, the more repli-
`cation defective the vector will be, and there is less chance of recombination to
`generate the infectious parental virus. The nature of the virus biology will usu-
`ally determine the means of production. For example, retroviruses are produced
`in packaging cell lines, and vector particles accumulate in the culture medium.
`In contrast, adenovirus and adeno-associated virus (AAV) vectors are generally
`produced from transfections, and cells must be lysed to liberate the viral particles.
`In this review, we describe the salient features and applications of some of the most
`commonly used viral vectors. There are a number of other emerging vector systems
`that are still in their infancy and have been extensively discussed in other excellent
`reviews (1, 3).
`
`RNA VIRUS VECTORS
`
`The most commonly used RNA virus vectors are derived from retroviruses, and
`these were among the first viral delivery systems to be developed for gene therapy
`applications. Retroviruses are a large family of enveloped RNA viruses found in
`all vertebrates, and they can be classified into oncoretroviruses, lentiviruses, and
`spumaviruses.
`
`Retroviruses
`The enveloped virus particle contains two copies of the viral RNA genome, sur-
`rounded by a cone-shaped core (for an in-depth review of retrovirus biology, see
`Reference 4). The viral RNA contains three essential genes, gag, pol, and env, and
`is flanked by long terminal repeats (LTR). The gag gene encodes for the core pro-
`teins capsid, matrix, and nucleocapsid, which are generated by proteolytic cleavage
`of the gag precursor protein. The pol gene encodes for the viral enzymes protease,
`reverse transcriptase, and integrase, which are usually derived from the gag-pol
`precursor. The env gene encodes for the envelope glycoproteins, which mediate
`virus entry. Oncoretroviruses are simple viruses encoding only the structural genes
`gag, pol, and env, whereas lentiviruses and spumaviruses have a more complex
`organization and encode for additional viral proteins (see Figure 2).
`After binding to its receptor, the viral capsid containing the RNA genome enters
`the cell through membrane fusion. The viral RNA genome is subsequently con-
`verted into a double-stranded proviral DNA by the viral enzyme reverse
`
`Annu. Rev. Biochem. 2005.74:711-738. Downloaded from www.annualreviews.org
`
`by Pohang University of Science and Technology (POSTECH) on 10/03/13. For personal use only.
`
`SKI Exhibit 2036
`Page 4 of 30
`
`

`

`10 May 2005 22:26
`
`AR
`
`AR261-BI74-24.tex
`
`XMLPublishSM(2004/02/24)
`
`P1: JRX
`
`GENE THERAPY
`
`715
`

`
`gag
`
`LTR
`
`Retrovirus
`
`pol
`
`Lentivirus
`
`env
`
`LTR
`

`
`gag
`
`LTR
`
`pol
`
`vpu
`
`vif
`vpr
`tat
`
`env
`
`LTR
`
`nef
`
`rev
`
`L5
`E3
`
`Adenovirus
`
`L2
`
`L3
`
`L4
`
`E2
`
`L1
`VA
`
`IVa2
`

`
`E1A E1B
`
`ITR
`
`Adeno-associated virus
`
`Rep
`
`Cap
`
`ITR
`
`ITR
`
`E4
`
`ITR
`
`X Essential elements retained in vectors
`
`X Genes supplied by packaging construct / cell line
`
`X Nonessential genes often deleted
`
`Figure 2 Schematics of the commonly used viruses that are converted to recombinant
`viral vectors. The colored boxes indicate genes or cis-acting elements that are either
`essential [and therefore retained in vectors (red ) orsupplied by packaging constructs
`or cell lines (green)] or that are nonessential and often deleted (blue). Only the major
`genetic elements are shown, and viruses are not drawn to scale. In second- and third-
`generation derivatives of each vector system, more viral genes are deleted (see text for
`details).
`
`Annu. Rev. Biochem. 2005.74:711-738. Downloaded from www.annualreviews.org
`
`by Pohang University of Science and Technology (POSTECH) on 10/03/13. For personal use only.
`
`SKI Exhibit 2036
`Page 5 of 30
`
`

`

`10 May 2005 22:26
`
`AR
`
`AR261-BI74-24.tex
`
`XMLPublishSM(2004/02/24)
`
`P1: JRX
`
`716
`
`VERMA (cid:1) WEITZMAN
`
`transcriptase. The proviral DNA is associated with viral proteins, including nu-
`cleocapsid, reverse transcriptase, and integrase, in a preintegration complex and
`translocates to the nucleus, where the integrase mediates integration of the provirus
`into the host cell genome. Host cell transcription factors initiate transcription from
`the LTR, and new viral particles are formed at the plasma membrane. Gag-pol and
`gag precursors assemble together with two copies of viral RNA, and env glyco-
`proteins are incorporated into the viral membrane during the budding process. In
`the newly formed virion, gag and gag-pol precursors are subjected to processing
`by the viral protease, which results in maturation of the virion.
`
`ONCORETROVIRAL VECTORS These vectors have been derived from a number of
`different oncoretroviruses, including murine leukemia virus (MLV), spleen necro-
`sis virus, Rous sarcoma virus, and avian leukosis virus. Replication-defective MLV
`vectors are generated by replacing all viral protein encoding sequences with the
`exogenous promoter-driven transgene of interest. In addition to the packaging sig-
`nal, the vectors retain viral LTRs and adjacent sequences, which are essential for
`reverse transcription and integration. Vector RNA production is either driven by
`the U3 region of the LTR or by a CMV/LTR hybrid with higher transcriptional ac-
`(cid:1)
`tivity. In these vectors, the 3
`U3 region of the LTR is intact and is copied over to the
`(cid:1)
`5
`LTR during reverse transcription, allowing efficient integration and LTR-driven
`transgene expression in the transduced cell.
`Packaging of retroviral vectors is achieved by providing the structural proteins
`in trans in the packaging cells. The first packaging cell lines expressed gag, pol, and
`env from a complete proviral DNA, lacking only the packaging signal (5). How-
`ever, sequence homology between the vector and packaging constructs facilitated
`recombination, allowing generation of a replication competent virus. To prevent
`recombination, packaging cells have been developed that express gag/pol and env
`from separate constructs. Furthermore, expression from the packaging constructs
`is no longer driven by the viral LTR but by constitutive promoters that allow a
`high level of virus production (5, 6). To avoid the laborious and time-consuming
`practice of generating cell lines, high-titer vectors can be produced by transient
`transfection (7). Viruses are recovered from the supernatants of actively growing
`producer cells.
`Major concerns in the use of retroviral vectors are the possibility of vector
`mobilization and recombination with defective (endogenous) retroviruses in the
`target cell. This prompted the development of self-inactivating vectors (8). In these
`vectors LTR-driven transcription is prevented in transduced cells by deletion in the
`U3, and transgene expression is driven instead by an internal promoter, allowing
`the use of regulated and tissue specific promoters.
`
`In addition to gag, pol, and env, lentiviruses encode three to
`LENTIVIRAL VECTORS
`six additional viral proteins, which contribute to virus replication and persistence
`of infection (for in-depth reviews of lentiviruses, see References 9 and 10). Two
`of the accessory proteins, tat and rev, are present in all lentiviruses and mediate
`
`Annu. Rev. Biochem. 2005.74:711-738. Downloaded from www.annualreviews.org
`
`by Pohang University of Science and Technology (POSTECH) on 10/03/13. For personal use only.
`
`SKI Exhibit 2036
`Page 6 of 30
`
`

`

`10 May 2005 22:26
`
`AR
`
`AR261-BI74-24.tex
`
`XMLPublishSM(2004/02/24)
`
`P1: JRX
`
`GENE THERAPY
`
`717
`
`transactivation of viral transcription and nuclear export of unspliced viral RNA,
`respectively. Although vectors based on Simian, Equine and Feline lentiviruses
`have been developed (11), we focus on human immunodeficiency virus type 1
`(HIV-1)-based vectors because they have been most extensively studied.
`In addition to gag, pol, and env, HIV-1 encodes six accessory proteins (tat, rev,
`vif, vpr, nef, and vpu). The retroviral vector design provided an excellent template
`for development of lentiviral vectors. The HIV-1-based lentiviral vectors are devoid
`of all viral sequences apart from essential cis-acting sequences, including the LTRs
`and the packaging signal (see Figure 2). The rev responsive element (RRE) is also
`included in the vector RNA. The viral rev protein is provided in trans to ensure
`efficient nuclear export of the full-length viral RNA genomes through binding to the
`RRE. Initially the endogenous LTR was used to drive vector RNA expression via
`transactivation by the tat protein (12), but further generations of the vector utilize a
`CMV/LTR hybrid promoter, which increases vector production and allows vector
`production to be independent of tat expression (13). The accessory genes, vif, vpr,
`nef, and vpu, are dispensable for lentiviral vector production and transduction, and
`they are deleted from the packaging construct (13). The central poly purine tract
`(cPPT), from the pol ORF, can be included in cis to improve nuclear import of
`the proviral DNA and hence accelerate transduction (14, 15). Furthermore, the
`biosafety of vectors is improved by the development of self-inactivating vectors,
`which are less likely to be mobilized following infection with HIV (16, 17).
`The HIV-1 env glycoprotein has a highly restricted host range in that it infects
`cells containing CD4 and coreceptors. To broaden the host range of lentiviral
`vectors, they can be pseudotyped with the vesicular stomatitis virus glycoprotein
`(VSV-G) env, which is provided in trans and imparts a wide tropism (12). Vectors
`are harvested from the supernatant, and those pseudotyped with VSV-G can be
`concentrated to produce high-titer preps. Titers can be determined using assays that
`measure the amount or activity of proteins incorporated in the vector particles, such
`as the p24gag ELISA assay. Stable packaging cell lines have now been developed,
`in which the producer cells express the structural proteins from minimal packaging
`constructs and expression is driven by an inducible promoter to minimize the
`toxicity of the VSV-G envelope protein (18, 19). Other viral glycoproteins have also
`been used to pseudotype lentiviral vectors and provide altered cell tropism (20, 21).
`
`FOAMY VIRAL VECTORS Foamy viral (FV) vectors have recently been developed
`and are quite similar to retroviral and lentiviral vectors (22). The FV genome
`contains three additional ORFs (tas/bel1, bel-2, and bel-3), with tas/bel1 being
`the coactivator of viral transcription (23). In addition to the packaging signal that
`(cid:1)
`(cid:1)
`consists of the 5
`-untranslated region and the 5
`portion of the gag ORF present in all
`(cid:1)
`retroviral vectors, FV vectors contain the 3
`region of the pol ORF, which is critical
`(cid:1)
`U3
`for efficient packaging of these vectors. Similar to other retroviral vectors, the 5
`region of the LTR in the vectors’ plasmid can been replaced by a CMV promoter,
`which increases vector expression and makes vector production independent of
`tas/bel1.
`
`Annu. Rev. Biochem. 2005.74:711-738. Downloaded from www.annualreviews.org
`
`by Pohang University of Science and Technology (POSTECH) on 10/03/13. For personal use only.
`
`SKI Exhibit 2036
`Page 7 of 30
`
`

`

`10 May 2005 22:26
`
`AR
`
`AR261-BI74-24.tex
`
`XMLPublishSM(2004/02/24)
`
`P1: JRX
`
`718
`
`VERMA (cid:1) WEITZMAN
`
`FV vectors are produced by transient transfection of the vector construct as well
`as the packaging constructs encoding for the structural proteins gag, pol, and env
`in 293T cells (22). Because the FV envelope has a broad cellular host range, it is
`used by the vector, and therefore, the env sequences are included in the packaging
`construct or expressed from a separate construct. The FV env contains, in contrast
`to other retroviruses, an ER sorting signal, which allows FV particles to bud from
`intracellular membranes, and therefore, the majority of the infectious virions is cell
`associated (24). Consequently, the infectious particles have to be released from the
`packaging cells by a freeze-thawing process.
`Because retroviral vectors were some of the first to be utilized for gene delivery,
`they have been extensively used in many applications (2, 25). Their ability to
`integrate and provide long-term gene expression has made them particularly useful
`for generating stable cell lines that express a transgene of choice and for marking
`studies of cell lineage. Their dependence upon cell division has restricted in vivo
`applications to gene delivery in actively dividing tissues, such as stem cells and
`cancer cells. In contrast, lentivirus vectors have been used for gene delivery in vivo
`by direct administration in many organs, including brain, eye, liver, and muscle
`(26). They have also been used for ex vivo transduction of hematopoietic cells,
`followed by bone marrow transplantation (27).
`The limited cellular tropism of the natural envelope of wild-type viruses is one
`of the barriers for retroviral transduction. However, retroviruses have the ability to
`incorporate env glycoproteins from related as well as unrelated viruses, thus allow-
`ing pseudotyping with alternative glycoproteins. A number of different envelopes
`have been used to generate pseudotyped retroviral vectors with broad host ranges,
`including the VSV-G glycoprotein or the amphotropic MLV envelope. Pseudo-
`typing also allows transfer of specific tropisms to the vector. Neurotropism and
`retrograde axonal transport was accomplished by the vector via pseudotyping with
`the G protein of Mokola lyssaviruses (28), and the filovirus (Ebola Zaire) envelope
`supported transduction of airway epithelia (21). Interestingly, the entry pathway
`of the retroviral vector has minimal effect on the transduction efficiency.
`Reverse transcription and nuclear translocation of the preintegration complex
`are thought to be limiting steps in retroviral transduction, especially in terminally
`differentiated postmitotic cells. Proviral DNA synthesis of all retroviruses depends
`strongly on cellular conditions, and low nucleoside pools or absence of cellular
`cofactors might explain the incomplete reverse transcription in quiescent or sta-
`tionary cells. In contrast to other retroviral vectors, FV vector particles can contain
`fully reverse-transcribed viral DNA, owing to activation of the process of reverse
`transcription before virus assembly. This suggests that FV vector gene transfer
`might be more efficient in certain postmitotic cells in which reverse transcription
`is limited.
`Because HIV-1 is a human pathogen, there are biosafety concerns about the
`use of HIV-1-based lentiviral vectors. The current HIV-1 lentiviral vector sys-
`tem is depleted of all accessory proteins, and viral sequences in the vectors have
`been minimized. Thus, the replication of these vectors is highly disabled, and the
`
`Annu. Rev. Biochem. 2005.74:711-738. Downloaded from www.annualreviews.org
`
`by Pohang University of Science and Technology (POSTECH) on 10/03/13. For personal use only.
`
`SKI Exhibit 2036
`Page 8 of 30
`
`

`

`10 May 2005 22:26
`
`AR
`
`AR261-BI74-24.tex
`
`XMLPublishSM(2004/02/24)
`
`P1: JRX
`
`GENE THERAPY
`
`719
`
`possibility of homologous recombination is minimized. In addition, codon op-
`timization of the packaging construct further decreases the risk of homologous
`recombination. The use of vectors based on other lentiviruses might eliminate
`some of the concerns (11); however, the risk associated with the introduction of
`nonhuman lentiviral vectors in human tissues is unknown.
`
`DNA VIRUS VECTORS
`
`Of the vectors derived from viruses with DNA genomes, the most prominent are
`those based on adenovirus (Ad) and the adeno-associated virus (AAV). Aden-
`oviruses contain a double-stranded DNA (dsDNA) genome of ∼36 kb, whereas
`AAVs consist of a single-stranded DNA molecule that is relatively small (∼4.7 kb).
`The basic principals of vector design (Figure 1) also apply to vectors derived from
`DNA viruses. However, there are important practical differences in terms of con-
`struction, production, and purification of these vectors.
`
`Adenovirus
`Ads have been isolated from a large number of species and tissue types (for an
`in-depth review of Ad biology see Reference 29). The human Ad family consists of
`more than 50 serotypes that can infect and replicate in a wide range of organs, such
`as the respiratory tract, the eye, urinary bladder, gastrointestinal tract, and liver.
`The Ad genome consists of a double-stranded linear DNA molecule (size: ∼36
`kb) with overlapping transcription units on both strands. Extensive splicing results
`in the production of over 50 proteins; 11 of which are structural virion proteins.
`The viral life cycle occurs in an early and a late phase, divided by the onset of viral
`DNA replication. Adenoviral genes fall into three major groups, depending on
`the time course of their expression during the viral replicative cycle: early (E1A,
`E1B, E2, E3, and E4), delayed (IX and IVa2), and the major late transcription unit
`(see Figure 2). The latter is processed into five mRNAs (L1–L5) that share the
`same carboxy terminus. These transcription units are transcribed by the cellular
`RNA polymerase II, whereas the viral-associated (VA) RNA is transcribed by
`RNA polymerase III. The viral genome contains two identical origins for DNA
`replication within each terminal repeat. The E2 region encodes proteins required
`for replication, including the viral polymerase, and proteins from the E1 and E4
`regions also contribute to efficient DNA replication. The gene products of the E3
`region are involved in immune surveillance and suppression but are nonessential
`for infection in vitro.
`The Ad genome is packaged in a nonenveloped icosahedral protein capsid. The
`fiber protein projects from the virion, and the carboxy-terminal knob domain forms
`a high-affinity complex with a host cell surface receptor protein. For the majority
`of Ad serotypes this receptor is the Coxsackie-adenovirus receptor (CAR) (30).
`In addition to attachment, efficient virus internalization requires an interaction
`
`Annu. Rev. Biochem. 2005.74:711-738. Downloaded from www.annualreviews.org
`
`by Pohang University of Science and Technology (POSTECH) on 10/03/13. For personal use only.
`
`SKI Exhibit 2036
`Page 9 of 30
`
`

`

`10 May 2005 22:26
`
`AR
`
`AR261-BI74-24.tex
`
`XMLPublishSM(2004/02/24)
`
`P1: JRX
`
`720
`
`VERMA (cid:1) WEITZMAN
`
`between the viral penton base and the cellular integrin αv receptor (31). After
`entry, the virus rapidly escapes from the endosome, and transport to the nucleus
`is accompanied by gradual disruption of the virus particle (32). The viral genome
`is imported through the nuclear pore and associates with the nuclear matrix to
`facilitate initiation of the primary transcription events (33). The Ad genome is
`transcribed and replicated at discrete replication centers in the nucleus of the
`infected cell (34), and the viral DNA does not normally integrate into the host
`genome.
`Adenoviral infection causes an initial nonspecific host response with synthesis
`of cytokines (tumor necrosis factor as well as interleukin 1 and 6), followed by a
`specific response of cytotoxic T lymphocytes directed against virus-infected cells
`that display viral peptide antigens (29). In addition, there is activation of B cells
`and the necessary CD4-positive T cells, leading to a humoral response. Serologic
`surveys found antibodies against Ad serotypes 1, 2, and 5 in 40% to 60% of
`children. The immune response of the host against adenoviral proteins is the major
`hurdle to the efficient and safe use of adenoviral vectors.
`Most adenoviral vectors are derived from Ad serotype 5; however, Ad vectors
`have also been generated from other serotypes, including human Ad2, Ad7, and
`Ad4 as well as nonhuman viruses. Replication-defective Ad vectors are designed
`by replacing crucial adenoviral coding regions (35). In the first generation of
`Ad vectors, the E1 gene was replaced with the transgene (36). Because E1A is
`the principal protein that activates the expression of other Ad transcription units’
`genes (37), and other E1 proteins play crucial roles in viral replication, these
`vectors are replication defective on most cell lines. E1-deleted vectors can be
`propagated in cell lines that provide the E1 gene products in trans, such as the
`human 293 cell line (38). Transgenes of up to 4.7–4.9 kb can be incorporated
`into E1-deleted adenoviral vectors (39). The cloning capacity of Ad vectors can
`be further increased by deletion of additional dispensable sequences from the
`Ad genome, such as the nonessential E3 region (40). Combining the E1 and E3
`deletions provides a total cloning capacity of 8.3 kb in one mutant virus. Newly
`developed E1-complementing cell lines with a minimal amount of viral sequences
`help reduce the chances of homologous recombination between the vector and
`the host genome, and thus, less replication-competent Ad is generated (41, 42).
`Vectors can be produced at titers of up to 1013 particles/ml and are purified by
`cesium chloride gradient ultracentrifugation or column chromatography.
`Although E1-deleted viruses are defective for replication, in some cell types
`they can produce virus proteins that serve as foreign antigens to induce a cellu-
`lar immune response. Many attempts have been made to reduce immunogenicity
`by engineering the second generation of Ad vectors that are additionally deleted
`in other viral transcription units, such as E2 and E4. Because the E2 region en-
`codes proteins that are essential for replication of the viral chromosome, it has
`to be provided in trans in the packaging cells. A number of deletions have been
`introduced into the E4 region, which encodes proteins required for efficient viral
`DNA replication and late protein synthesis. Partial deletions of the E4 region can
`
`Annu. Rev. Biochem. 2005.74:711-738. Downloaded from www.annualreviews.org
`
`by Pohang University of Science and Technology (POSTECH) on 10/03/13. For personal use only.
`
`SKI Exhibit 2036
`Page 10 of 30
`
`

`

`10 May 2005 22:26
`
`AR
`
`AR261-BI74-24.tex
`
`XMLPublishSM(2004/02/24)
`
`P1: JRX
`
`GENE THERAPY
`
`721
`
`produce viable vectors, and E4-complementing cell lines have been developed for
`more extensive deletions (43–46). Although deletions in the E4 region increase
`the cloning capacity of the Ad vector, some reports indicate that the E4 region may
`exert a positive effect on long-term expression (47–49).
`Theoretically it should be possible to reduce viral genes to a minimum and create
`“gutted” vectors that carry no viral sequences, apart from the inverted terminal
`repeats (ITRs) and the cis-acting packaging signal (50). These vectors require
`helper viruses for propagation, generating a problem in the purification of a helper-
`free virus. An important step toward a third generation of Ad vectors was the
`development of high-capacity, helper-dependent vectors based on the Cre/loxP-
`system of site-specific DNA excision (51–53). Using the CreloxP-system, 25 kb
`of adenoviral genome can be deleted from an Ad vect

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