therapeutic potential of small interfering rna for brain …therapeutic potential of small...

21
Therapeutic Potential of Small Interfering RNA for Brain Diseases Amy E. Lovett-Racke Contents 1 RNA Interference .......................................................................... 276 2 Spinocerebellar Ataxia and Other Polyglutamine-Associated Ataxias .................... 279 3 Alzheimer’s Disease ....................................................................... 281 4 Parkinson’s Disease ........................................................................ 284 5 Amyotrophic Lateral Sclerosis ............................................................. 284 6 Multiple Sclerosis .......................................................................... 286 7 Brain Tumors ............................................................................... 288 8 Conclusions ................................................................................ 291 References ...................................................................................... 291 Abstract RNA interference (RNAi) is an evolutionarily conserved mechanism found in all eukaryotes to degrade messenger RNA (mRNA) that has sequence homology to short double-stranded RNA (dsRNA). It is believed that the usual biological function of RNAi is to safeguard the host cells from RNA viruses and transposons, which can endanger the genome. Expression of dsRNA in the host cell signals a cascade of intracellular events that generate small interfering RNA (siRNA) segments that mediate the degradation of complementary mRNA and subsequent failure to express protein. Recently, engineered siRNA has been used to study the normal biological function and significance of a vast number of genes in a variety of cell types, as well as the role of specific genes in disease patho- genesis. Thus, siRNA has therapeutic potential to specifically suppress the expres- sion of proteins associated with disease processes. The exquisite specificity of siRNA-mediated gene suppression provides an opportunity to target genes with single point mutations found in hereditary disorders, genes unique to pathogenic tumor cells in malignancies, and genes that are critical for mediating the pathology of many diseases. The fact that RNAi functions in the cell at the transcription level A.E. LovettRacke Department of Molecular Virology, Immunology and Medical Genetics, Ohio State University, 43065, Columbus, OH, USA e-mail: [email protected] V.A. Erdmann et al. (eds.), Therapeutic Ribonucleic Acids in Brain Tumors, 275 DOI: 10.1007/9783642004759_13, # SpringerVerlag Berlin Heidelberg 2009

Upload: others

Post on 27-Jan-2021

3 views

Category:

Documents


0 download

TRANSCRIPT

  • Therapeutic Potential of Small InterferingRNA for Brain Diseases

    Amy E. Lovett-Racke

    Contents

    1 RNA Interference . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 276

    2 Spinocerebellar Ataxia and Other Polyglutamine-Associated Ataxias . . . . . . . . . . . . . . . . . . . . 279

    3 Alzheimer’s Disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 281

    4 Parkinson’s Disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 284

    5 Amyotrophic Lateral Sclerosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 284

    6 Multiple Sclerosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 286

    7 Brain Tumors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 288

    8 Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 291

    References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 291

    Abstract RNA interference (RNAi) is an evolutionarily conserved mechanismfound in all eukaryotes to degrade messenger RNA (mRNA) that has sequence

    homology to short double-stranded RNA (dsRNA). It is believed that the usual

    biological function of RNAi is to safeguard the host cells from RNA viruses and

    transposons, which can endanger the genome. Expression of dsRNA in the host cell

    signals a cascade of intracellular events that generate small interfering RNA

    (siRNA) segments that mediate the degradation of complementary mRNA and

    subsequent failure to express protein. Recently, engineered siRNA has been used

    to study the normal biological function and significance of a vast number of genes

    in a variety of cell types, as well as the role of specific genes in disease patho-

    genesis. Thus, siRNA has therapeutic potential to specifically suppress the expres-

    sion of proteins associated with disease processes. The exquisite specificity of

    siRNA-mediated gene suppression provides an opportunity to target genes with

    single point mutations found in hereditary disorders, genes unique to pathogenic

    tumor cells in malignancies, and genes that are critical for mediating the pathology

    of many diseases. The fact that RNAi functions in the cell at the transcription level

    A.E. Lovett‐RackeDepartment of Molecular Virology, Immunology and Medical Genetics, Ohio State University,

    43065, Columbus, OH, USA

    e-mail: [email protected]

    V.A. Erdmann et al. (eds.), Therapeutic Ribonucleic Acids in Brain Tumors, 275DOI: 10.1007/978‐3‐642‐00475‐9_13, # Springer‐Verlag Berlin Heidelberg 2009

  • means that virtually any gene can be targeted for suppression, providing a unique

    strategy to develop therapeutics for many neurological diseases, many of which

    have no effective therapies.

    1 RNA Interference

    RNA interference (RNAi) was initially discovered in plants, when RNA for

    pigment-producing genes was introduced into petunias to enhance color (Napoli

    et al. 1990; Van der Krol et al. 1990). Surprisingly, these petunias expressed very

    little color, and this paradoxical effect was termed cosuppression. A similar loss of

    gene function was observed in fungi transformed with albino transgene that

    silenced the endogenous albino gene and was referred to as quelling (Romano

    and Macino 1992). Like many other genetic discoveries, the recognition that these

    specific and potent suppressive effects were provoked by double-stranded RNA

    (dsRNA) was made in Caenorhabditis elegans when dsRNA homologous to unc-22mRNA was injected into C. elegans, producing a twitching phenotype associatedwith unc-22 loss of function (Fire et al. 1998). The clear association of dsDNA with

    suppression of a complementary gene was termed RNAi and led to Drs. Fire and

    Mello receiving the Nobel Prize in Physiology or Medicine in 2006. The observa-

    tion that 22–25 nucleotide RNA fragments in plants undergoing posttranscriptional

    gene silencing led to the realization that small interfering RNA (siRNA) were

    ultimately responsible for mediating the silencing effect of dsRNA observed in

    these diverse organisms (Hamilton and Baulcombe 1999).

    Since these original observations, RNAi, as a posttranscriptional gene-silencing

    mechanism, has been well defined. It is thought that RNAi evolved as a means to

    reduce the effect of potentially dangerous dsRNA, not usually expressed in eukar-

    yotes, but commonly by viruses. Dicer, a cytoplasmic ribonuclease III-like enzyme,

    recognizes dsRNA and cleaves it into 21–23 nucleotide segments that contain 2–3

    nucleotide overhangs (Fig. 1). These 21–23 nucleotide fragments, termed siRNA,

    bind to a multiprotein complex called the RNA-induced silencing complex (RISC).

    The RISC causes linearization of the siRNA and separation of the strands. The

    single-stranded RNA within the RISC, known as the guide strand, hybridizes to

    complementary messenger RNA (mRNA). An enzyme within the RISC, argo-

    naute 2, cleaves the mRNA into fragments so that translation of the transcript is

    no longer possible. The guide strand within the RISC can repeatedly bind comple-

    mentary mRNA, continuing the degradation of the target mRNA, allowing pro-

    longed suppression of the target gene.

    Scientists have since used the RNAi pathway to target self-genes as a mecha-

    nism to understand and define the normal biological function of gene products. This

    has been accomplished by delivering to cells plasmid or viral vectors containing

    specific RNA sequences that are processed into short hairpin DNA that contain a

    21-base dsRNA sequence that can be cleaved by Dicer into siRNA. Alternatively,

    synthetic siRNA have been shown to readily transfect cells and were shown to be

    276 A.E. Lovett-Racke

  • capable of directly activating the RISC, degrading the target mRNA. Similarly,

    RNAi has been used to suppress genes known to be associated with diseases, as well

    as discover genes critical to pathogenic processes. Because siRNA usually only

    recognize mRNA that are completely homologous, siRNA have been developed

    Long ds RNA

    shRNA expressed from plasmid or viral vectors

    Dicer

    Dicer

    siRNA

    Synthetic siRNA

    siRNA

    5’

    5’

    5’3’

    5’3’

    5’

    5’

    RNA induced silencing complex (RISC)

    Argonaute 2

    5’AAAmRNA

    5’

    AAA

    5’

    NUCLEUS

    Plasmid or viral vector containing

    siRNAsequence

    CYTOPLASM

    RNA degradation

    5’

    mRNAcomplementary

    to siRNA

    Virus with dsRNAgenome

    Plasmids can carry siRNAsequences

    Viruses can be engineered to contain siRNA

    sequences

    5’

    RISC with guide strand can continue to bind mRNA

    Guide strand of siRNA

    Synthetic siRNA

    5’

    5’

    5’

    5’AAA

    5’

    Fig. 1 Mechanism of RNA interference. Double-stranded RNA (dsRNA) is cleaved by theenzyme Dicer into 21–23 nucleotide fragments, termed small interfering RNA (siRNA). The

    dsRNA can come from viruses with dsRNA genomes, or from plasmids or viral vectors carrying

    sequences for shRNA. The siRNA enter a multiprotein complex called the RNA-induced silencing

    complex (RISC) that linearizes and separates the siRNA. The guide strand binds complementary

    mRNA and initiates degradation of the mRNA, preventing translation and protein production

    Therapeutic Potential of Small Interfering RNA for Brain Diseases 277

  • that efficiently distinguish between wild-type DNA sequences and point mutations

    in autosomal dominant genes. For neurological diseases, the question arose as to

    whether axons had the machinery for RNAi. Developing axons and growth cones

    have local mRNA that are translated in response to specific extracellular signaling

    molecules which are critical for axon development and neurotransmitter produc-

    tion. These same signaling pathways would be necessary in axonal repair following

    injury. The molecular mechanisms that mediate the translation of mRNA in axons

    and growth cones in not well defined and thus mechanisms that regulate mRNA in

    cell bodies may not be the same. However, developing axons express argonaute and

    Dicer, and can assemble functional RISC when distal axons are transfected with

    siRNA (Hengst et al. 2006).

    One of the most significant obstacles to RNAi in both research and its advance-

    ment to therapeutics is delivery of the siRNA to the target cells, especially in the

    central nervous system (CNS) (Begley 2004). In particular, cells in the CNS are

    protected by the blood–brain barrier (BBB) which significantly limits access to

    these tenuous cells. Nonspecific delivery systems, such as liposomes and cholester-

    ol, have been beneficial in enhancing siRNA uptake and reducing degradation.

    Viral delivery of shRNA that can be cleaved by Dicer into siRNA has been used to

    deliver siRNA and can provide some cell specificity based on the type of virus and

    cell surface molecules that mediate viral entry. In addition, viruses add the benefit

    of prolonged expression of the siRNA which should result in extended suppression

    of the target gene. Synthetic siRNA have been shown to be effective at suppressing

    target genes when administered intravenously if the target is in highly vascularized

    tissue, such as the liver and spleen (Lovett-Racke et al. 2004; Thakker et al. 2004).

    This hydrodynamic approach is limited because siRNA delivered in large volumes

    of saline intravenously, which enhances tissue perfusion, can cause heart damage

    and may be inappropriate for humans. Chemical modifications of synthetic siRNA

    that reduce digestion of siRNA may improve bioavailability. Substitutions with

    locked nucleic acids, 20F, 20O-Me, and 20H residues can stabilize siRNA andenhanced benefits have been observed in vivo in mouse models using these chemi-

    cal modifications (Elman et al. 2005; Morrissey et al. 2005). Conjugation of siRNA

    to carriers may also enhance cell uptake and specificity of delivery. Peptides,

    aptamers or monoclonal antibodies that recognize specific cell surface receptors

    can be conjugated to siRNA, allowing entry into cells with the required receptor

    (Muratovska and Ecles 2004; Juliano 2005; Chu et al. 2006; Kumar et al. 2008).

    Although intracerebral and intrathecal administration of siRNA has been used in

    experimental models of CNS disease and is possible in humans, it is not the optimal

    strategy in humans given the potential risks. Systemic administration of siRNA

    does not efficiently cross the BBB, providing a serious obstacle to the development

    of siRNA-based therapies for human neurological disease. However, strategies to

    facilitate transport across the BBB are being developed. Trojan horse liposomes

    have been developed that may overcome many of the obstacles with siRNA

    delivery to the CNS (Boado 2007). Nucleic acids are encapsulated in a liposome

    that protects the nucleic acid from nuclease-mediated degradation (Fig. 2).

    The liposome is constructed of polyethyleneglycol that stabilizes the liposomes.

    278 A.E. Lovett-Racke

  • Monoclonal antibodies specific for receptors on the BBB are engineered into the

    polyethyleneglycol which allow transcytosis across the BBB by the liposomes. The

    Trojan horse liposomes can contain siRNA or plasmids that generate shRNA. In the

    case of plasmids, they can be engineered such that the promoter is specific for a

    particular cell type so that the shRNA is only expressed in the appropriate CNS cell

    type. Alternatively, the liposomes can contain two types of monoclonal antibodies,

    one that recognizes a BBB to allow transport across the BBB and a second

    monoclonal antibody that binds to a cell type-specific receptor to promote uptake

    of the nucleic acid by the appropriate cell. This strategy would be feasible with both

    plasmids and siRNA. Although delivery has been a major challenge in all forms of

    gene therapy, technology is advancing to the point that this will soon be overcome.

    2 Spinocerebellar Ataxia and Other Polyglutamine-AssociatedAtaxias

    Polyglutamine-associated ataxias are a group of autosomal dominant neurodegen-

    erative disorders. As such, one allele of the affected gene contains a mutation that

    results in expanded CAG repeats, while the other allele is normal. The mutant

    proteins cause intranuclear inclusions that are toxic to neurons. Since RNAi targets

    Blood Brain Barrier CNS Target CellsTrojan Horse Liposome

    MAb to BBB

    MAb to Cell

    siRNAplasmid

    Fig. 2 Delivering siRNA to cells in the CNS. Trojan horse liposomes are composed of poly-ethyleneglycol bound to monoclonal antibodies that recognize proteins unique to the blood-brain

    barrier to allow transport into the CNS and monoclonal antibodies to a cell type-specific receptor

    to allow transport of the siRNA into the target cell

    Therapeutic Potential of Small Interfering RNA for Brain Diseases 279

  • and degrades complementary mRNA sequences, it was theoretically possible that

    siRNA containing the mutation could be used to specifically target the mutant

    mRNA, allowing the nonmutant mRNA to translate into normal protein. The

    potential of siRNA as a therapeutic option for CNS diseases was first observed in

    a mouse model for spinocerebellar ataxia type 1 (SCA1), which is a dominantly

    inherited, progressive neurodegenerative disease caused by an expanded polyglu-

    tamine (CAG) sequence in the ataxin 1 protein. Initially, plasmids containing short

    hairpin RNA (shRNA) sequences within the human ataxin 1 complementary DNA

    sequence to target the ataxin 1 protein were tested in neuronal cell lines (Xia et al.

    2004). Two sequences that flanked the CAG repeat region coding for the poly-

    glutamine sequence suppressed ataxin 1 levels. These shRNA sequences were

    subsequently inserted into recombinant adeno-associated virus (AAV) vectors to

    determine if ataxin 1-specific siRNA had a biological effect in the transgenic mouse

    model for SCA1. Intracerebellar injection of the AAV-siRNA resulted in enhanced

    motor function, improved neuropathologic findings, and total loss of ataxin 1

    inclusions in transfected Purkinje cells, demonstrating a clear therapeutic benefit.

    Spinocerebellar ataxia type 3 (SCA3), also known as Machado-Joseph disease,

    is also an autosomal dominant neurodegenerative disease that results from an

    expanded polyglutamine repeat within the ataxin 3 gene. Initial attempts to sup-

    press ataxin 3 by RNAi targeting various sequences within the ataxin 3 gene

    resulted in efficient, but not allele-specific, suppression of the mutant and normal

    alleles (Miller et al. 2003). To enhance the probability that the mutant allele would

    be preferentially targeted, siRNA were developed that were specific for a single

    nucleotide polymorphism in the ataxin 3 gene which is in linkage disequilibrium

    with the expanded CAG repeat, and thus typically segregates with the disease allele.

    Development of synthetic siRNA that had the single mutation failed to differentiate

    between the mutant and normal alleles. However, siRNA that added a mutation

    within the siRNA sequence, so that it contained two mismatched bases with the

    normal sequence and a single mismatch to the mutant allele, demonstrated signifi-

    cant suppression of the mutant allele while maintaining expression of the normal

    allele. Subsequently, ataxin3-specific siRNA was developed that placed the mutant

    base in the center of the siRNA sequence, which demonstrated allele-specific

    suppression. Recent studies have developed strategies for designing siRNA that

    specifically distinguish between alleles that differ by a single nucleotide (Schwarz

    et al. 2006). The ataxin3 siRNA sequences with single base changes were subse-

    quently inserted in plasmid and AAV vectors as shRNA and demonstrated similar

    inhibition of the mutant ataxin3 in vitro, showing that efficacy is maintained when

    the siRNA is produced by different modes.

    Huntington disease is also a dominantly inherited neurodegenerative disease

    which causes dementia and motor deficits, ultimately resulting in death. Expansive

    CAG repeats, typically greater than 35, within the huntingtin gene create a protein

    that is prone to aggregation and toxic to neurons, predominately in the cerebral

    cortex and striatum. Although the huntingtin gene is expressed in all cells, its

    function remains unknown. Mice deficient in the huntingtin gene fail to produce

    viable offsprings, indicating that the huntingtin gene product is essential for normal

    280 A.E. Lovett-Racke

  • development (Duyao et al. 1995; Nasir et al. 1995; Zeitlin et al. 1995). Initially,

    a novel strategy was used to target the huntingtin gene by inserting shRNA into a

    transposon system, which inserts efficiently into almost any DNA sequence, pro-

    moting continuous suppression of the target gene (Chen et al. 2005). Using this

    system, expression of the normal huntingtin gene was suppressed in various cell

    lines. To repress the mutant huntingtin gene in vivo, AAV was used to deliver of

    mutant-specific huntingtin shRNA to the brain in mouse models of Huntington

    Disease. This resulted in improved motor function, and decreased mutant hunt-

    ingtin gene expression (Harper et al. 2005; Machida et al. 2006). Since shRNA can

    integrate into the genome randomly, off-target effects can be observed and were

    noted in a model of Huntington Disease (Denovan-Wright et al. 2008). Therefore,

    synthetic siRNA may provide a safer alternative for therapeutic use of RNAi to

    suppress specific genes. To enhance cell uptake in vivo of synthetic siRNA,

    cholesterol conjugated-synthetic siRNA specific for the mutant huntingtin gene

    were developed. Mice were injected with cholesterol conjugated-siRNA in the

    striata following administration of AAV vectors containing huntingtin cDNA

    encoding amino acids 1–400 with expanded (100 CAG) or typical (18 CAG)

    number of CAG repeats (DiFiglia et al. 2007). Mice receiving the mutant huntingtin

    cDNA with expanded CAG repeats developed motor deficits and neuropathology

    similar to Huntington Disease, while the mice that received the huntingtin cDNA

    with the wild-type number of CAG repeats remained healthy. Mice that received

    both the expanded CAG huntingtin cDNA and the mutant-specific siRNA had

    prolonged survival of striatal neurons, fewer neutrophil aggregates, smaller inclu-

    sion size, and improved motor function, indicating that siRNA had similar

    efficacy as the shRNA. Currently, therapeutic intervention in Huntington Disease

    is ineffective. Since Huntington Disease is a dominant familial disorder, families

    live with the burden that many will be stricken with this devastating disease.

    Genetic therapies, such as siRNA, provide the best hope of preventing the expres-

    sion of the mutant gene and suppressing the disease phenotype.

    3 Alzheimer’s Disease

    Alzheimer’s disease (AD) is a neurodegenerative disorder characterized by demen-

    tia that affects up to 30 million people worldwide and accounts for the majority of

    cognitive impairment in elderly patients inWestern Europe and North America. AD

    is characterized by two major hallmarks, extracellular senile plaques containing

    b-amyloid fragments cleaved from the amyloid precursor protein (APP) and intra-cellular neurofibrillary tangles composed of hyperphosphorylated tau protein. The

    most critical element for the use of siRNA as a therapeutic agent in the treatment of

    diseases is the identification of relevant and specific target genes. The molecular

    characterization of plaques and neurofibrillary tangles, and the discovery of genes

    involved in the pathogenesis of both familial and sporadic AD, have identified

    several potential targets for RNAi. APP was the first gene directly associated with

    Therapeutic Potential of Small Interfering RNA for Brain Diseases 281

  • AD (Mullan et al. 1992). Autosomal dominant mutations in APP result in an early-

    onset, aggressive form of the disease (Goate et al. 1991). Approximately 20 APP

    missense mutations near the cleavage site of secretases have been found, suggesting

    that altered cleavage of APP may negatively affect b-amyloid deposition (Fig. 3).Given the relative large number of APP mutations, identification of APP mutations

    associated with pathology is a critical first step before targeting a specific mutant

    allele with siRNA. Plasmids expressing shRNA, containing siRNA sequences,

    specific for the Swedish APP allele were developed, since this is the most common

    mutation in AD (Miller et al. 2004). The Swedish APP allele contains two adjacent

    point mutations which were targeted with siRNA that altered the position of the

    mutations within the siRNA sequence. The mutant APP allele could be suppressed

    to varying degrees in vitro based on the location of the mutations within the siRNA

    sequence, while maintaining the normal APP expression. Using a novel lentiviral-

    based mouse model for b-amyloid overexpression, herpes simplex viral vectorsexpressing a shRNA specific for the Swedish APP allele were found to significantly

    g-secretase

    a-secretase

    g-secretase

    b-secretase

    AmyloidPrecursor

    Protein

    membrane

    a-APP

    P3

    P7

    b-APP

    Ab39-43

    P7

    NormalAPP

    Processing

    PathogenicAPP

    Processing

    APPCleavageProducts

    APP PointMutations

    APHnic PEN2

    PS1 PS2

    APHnic PEN2

    PS1 PS2

    Insoluble Ab peptides

    Fig. 3 Multiple proteins in the processing amyloid precursor protein (APP) may be targets forsiRNA-based therapy in Alzheimer Disease. APP is normally processed into a-APP, P3, and P7 bya-secretase and g-secretase. However, mutations in APP and g-secretase are associated withabnomal APP cleavage, generating b-APP, Ab39–43 peptides and P7. Aggregation of Ab39–43peptides results in neurodegeneration

    282 A.E. Lovett-Racke

  • reduce b-amyloid accumulation, suggesting that in vivo suppression of mutant APPis achievable via RNAi (Hong et al. 2006).

    AD, as well as the inherited frontotemporal dementia with Parkinsonism linked to

    chromosome 17, progressive supranuclear palsy, and corticobasal ganglionic degen-

    eration (Houlden et al. 2001), are collectively called tauopathies due to mutations in

    the tau gene that cause an altered sequence or aberrant splicing (Lewis et al. 2001;

    Oddo et al. 2003). A mutant allele of the tau protein was successfully targeted in vitro

    using shRNA plasmids, demonstrating that a single nucleotide mutation could be

    used to suppress mutant tau by siRNA (Miller et al. 2004). As observed in the APP

    siRNA studies, the position of the mutation within the siRNA sequence was critical

    for selective inhibition of the mutant allele (Miller et al. 2003, 2004).

    Additional targets for AD include the apolipoprotein E e4 allele (ApoE4), thesecretases, and the presenilin genes. In addition to increasing age, ApoE4 is

    considered the most significant risk factor for familial and nonfamilial AD (Corder

    et al. 1993). Homozygosity for ApoE4 was shown to be sufficient to cause AD in

    most individuals over age 80. Suppression of ApoE4 with siRNA could potentially

    reduce the probability of developing AD or delay its onset, at least in heterozygotes.

    Because ApoE4 plays a critical role in cholesterol and triglyceride transport, it is

    necessary for normal metabolism, and, thus, the risk-benefit ratio of gene targeting

    in ApoE4 homozygotes is less clear.

    More than 150 mutations have been identified in the presenilin (PS) genes, which

    result in an early-onset aggressive form of AD. Presenilins (PS1/PS2) are the

    catalytic subunit of g-secretase, which also includes nicastrin, APH-1a, and PEN2.Mutations in PS1/PS2 enhance production of the highly self-aggregating Ab42peptide. AAP cleavage occurs via two pathways. Typically, APP is cleaved by

    a- and g-secretases into nontoxic, soluble a-APP protein and two smaller proteinsP3 and P7 (Nawrot 2004). In healthy individuals, these proteins protect neuronal

    cells from oxidative stress and play a role in wound repair. In AD patients, APP is

    cleaved at the N-terminus of the Ab region by b-secretase and at the C-terminus byg-secretase, generating b-APP protein, P7 and 39–43 amino acid long b-amyloidpeptides (Fig. 3). PS1 control and missense sequences were cloned into pSilencer

    plasmids that generate siRNA and then transfected into PS1/APP-expressing cell

    lines, resulting in reduced PS1 and Ab42 peptide (Luo et al. 2004). Given the largenumber of mutations found in the PS genes, therapeutic siRNA would need to be

    designed for individual patients or AD families based on the specific pathogenic PS

    mutation in that patient or family. Since b-secretase-deficient mice have no knowndeficits, this particular enzyme may also be a good therapeutic target (Luo et al.

    2001). Suppression of b-secretase, also known as beta-site APP-cleaving protein(BACE1), with siRNA inhibited Ab production, as well as the neurotoxicityassociated with oxidative stress, in neuronal cultures from normal and Swedish

    APP mutant mice (Kao et al. 2004). Using lentivirus vectors expressing siRNA that

    target BACE1, amyloid production, behavioral deficits and neurodegeneration were

    significantly reduced in an APP transgenic model of AD, indicating that reducing

    BACE1 reduced APP cleavage and the associated pathology (Singer et al. 2005).

    The relatively large number of targets for therapeutic intervention in AD and our

    Therapeutic Potential of Small Interfering RNA for Brain Diseases 283

  • enhanced understanding of designing mutant or allele-specific siRNA make AD a

    good candidate for developing therapeutic siRNA.

    4 Parkinson’s Disease

    Parkinson’s disease (PD) is a prevalent neurodegenerative disease, second only to

    AD. Both familial and idiopathic forms of PD exist with characteristic symptoms

    including resting tremor, rigidity, bradykinesia, and a unique freezing phenomenon

    that typically appears on one side of the body. The symptoms result from a loss of the

    dopaminergic neurons of the substantia nigra pars compacta and progressive exhaus-

    tion of striatal dopamine. Pathologically, intracytoplasmic inclusions, called Lewy

    Bodies, are present in the neurons. Since the pathology appears to be limited to

    dopaminergic neurons in the substantia nigra, finding proteins unique to these

    neurons is critical to identifying therapeutic targets for siRNA intervention. Current

    pharmacotherapy is replacement of levodopa, but side-effects such as dyskinesias can

    offset therapeutic benefit over time. Thus, the need for an effective therapy is great.

    The first gene linked to PD was PARK1, later identified as a-synuclein whichcontained single base mutations associated with familial disease (Polymeropoulos

    et al. 1997). Duplicate and triplicate gene copies of a-synuclein have also beenidentified in some families with PD, indicating that excess native a-synuclein isneurotoxic (Singleton et al. 2003). Idiopathic PD has been associated with genetic

    variability in the promoter of the a-synuclein gene (Pals et al. 2004). Lewy bodies arecomposed primarily of a-synuclein which appears to be misfolded and aggregateddue to the mutation or overexpression. RNAi of normal a-synuclein in dopaminergicneuroblastoma cells protected the cells from N-methyl-4-phenylpyridinium toxicity

    and reduced dopamine transport, illustrating a role for a-synuclein dopamine homeo-stasis (Fountaine and Wade-Martins 2007). An allele-specific siRNA was developed

    for the A53T mutation in human a-synuclein that specifically suppressed the targetgene in vitro (Sapru et al. 2006). Using a lentivirus expression system, shRNA

    specific for a-synuclein suppressed human a-synuclein in vivo in rat brains thatwere transgenic for human a-synuclein. Unfortunately, there are currently no goodanimal models of PD to evaluate the therapeutic potential of the a-synuclein siRNAthat specifically targets mutant alleles. Other proteins implicated in PD, such as

    PTEN, sirtuin 2, and LRRK-2, have been inhibited with siRNA in vitro, and

    preliminary data suggest that they may be potential targets for siRNA therapy in

    PD (Outeiro et al. 2007; Zhu et al. 2007; Habig et al. 2008).

    5 Amyotrophic Lateral Sclerosis

    Amyotrophic lateral sclerosis (ALS) is a progressive, usually fatal, neurodegenera-

    tive disease caused by motor neuron deterioration due to oxidative stress. Only

    5–10% of ALS is associated with a specific genetic or familial factor. The most

    284 A.E. Lovett-Racke

  • common genetic defect in ALS is a point mutation in copper/zinc superoxide

    dismutase (SOD1), an enzyme responsible for scavenging free radicals. Since

    SOD1 performs critical functions in cells, silencing of the mutant SOD1 must be

    specific, so that the normal allele is unaffected. This was accomplished in 2004

    when SOD1 mutant-specific siRNA were introduced into neuroblastoma cell lines

    that expressed mutant SOD1 (Maxwell et al. 2004; Yokota et al. 2004). Mutant-

    specific siRNA for SOD1 protected the neuroblastoma cells from cyclosporin

    A-induced cell death. In addition, siRNA was found to be more efficient at

    suppressing mutant SOD1 than ribozyme or DNA enzyme.

    Since more than 100 SOD1 mutations have been found, siRNA would need to be

    developed for each specific mutation found in an individual ALS patient or ALS

    family. However, this may not be possible because not all nucleic acid sequences

    are able to elicit a silencing effect, probably due to the ability of a sequence to bind

    the RISC or the ability of enzymes within the RISC to linearize and separate the

    siRNA strands. Therefore, a siRNA-based strategy was developed that would

    target all SOD1 sequence and replace the wild-type SOD1 (Xia et al. 2005). Two

    constructs were developed that contained shRNA sequences specific for both

    mutant and wild-type SOD1. However, these constructs also contained an RNAi-

    resistant SOD1 transgene that were generated by inserting silent point mutations in

    the wild-type SOD1 sequence that were not recognized by the shRNA. Transfection

    of these constructs into HEK293 cells demonstrated that endogenous and

    mutant SOD1 expression were suppressed, but that the transfected cells were not

    susceptible to hydrogen peroxide toxicity because the shRNA-resistant SOD1 gene

    was able to reverse the affects of the hydrogen peroxide. This study demonstrated

    the feasibility of using siRNA to target all mutations within the SOD1 gene by

    targeting all endogenous SOD1 and replacing the SOD1 gene with a wild-type

    functioning gene that is resistant to RNAi. Extending this strategy to animal models

    of ALS will determine if this will be feasible in patients.

    Allele-specific siRNA for SOD1 have been evaluated in mouse models of ALS.

    Intramuscular and intraspinal injection of lentivirus expressing mutant human

    SOD1 substantially retarded disease onset and progression in mice that overexpress

    a mutant form of SOD1 (Raoul et al. 2005; Ralph et al. 2005; Xia et al. 2006).

    Motor neuron survival in the brainstem and spinal cord was significantly improved

    and nearly 80% of mice had a normal life span, demonstrating that siRNA can be

    used in vivo to specifically suppress the mutant human allele. Intramuscular

    injection with AAV constructs carrying SOD1 siRNA and a green fluorescent

    protein gene demonstrated that retrograde transport of the AAV occurs, allowing

    expression of the siRNA in the spinal cord (Miller et al. 2005). To determine if

    siRNA can be used to actually prevent dominantly inherited diseases such as ALS,

    transgenic mice that express a shRNA specific for mutant SOD1 that was ubiqui-

    tously expressed throughout the brain were developed (Saito et al. 2005). These

    mice were crossed to the human mutant SOD1 transgenic mouse that spontaneously

    develops an ALS-like disease. However, the double transgenic mice failed to

    develop disease when monitored for 300 days, twice the time it takes until the

    mice transgenic for mutant SOD1 to die. In addition, these mice had no CNS

    Therapeutic Potential of Small Interfering RNA for Brain Diseases 285

  • pathology suggesting that the transgenic siRNA had completely prevented the

    expression of mutant SOD1.

    Another issue that must be addressed with diseases such as ALS is whether

    siRNA have therapeutic benefit after the onset of symptoms. To generate stable

    siRNA that could potentially be used in ALS patients, allele-specific mutant SOD1

    synthetic siRNA were developed with chemical modification to enhance stability

    and cellular uptake (Wang et al. 2008). Stability of the siRNA was enhanced

    by 20fluoro modifications at the 20-hydroxyl position which should maintain theA-helical structure of the siRNA required for initiating the RNAi cascade. Phos-

    porothioate modifications were also made to enhance uptake of the siRNA that was

    then infused into the spinal cord using an osmotic pump. Long-term infusion

    resulted in widespread distribution of the siRNA, decreased levels of mutant

    SOD1, slowed disease progression, and increased survival. This approach to patient

    treatment may initially be the safest option since infusion dose and rate of synthetic

    siRNA can be closely monitored and stopped if adverse effects are observed.

    Since mutations in the SOD1 gene only account for 2–3% of ALS cases, siRNA

    targets that are common to all forms of ALS may be more widely applicable. Motor

    neuron death in ALS is hypothesized to be mediated by Fas–Fas ligand apoptosis.

    To determine if motor neurons might be preserved if this apoptotic pathway is

    suppressed, Fas-specific siRNA was infused into the CNS of mutant SOD1 trans-

    genic mice (Locatelli et al. 2007). The number of Fas positive cells in the spinal

    cord decreased by nearly 50% and other molecules in the Fas pathway were

    similarly reduced. Motor deficits were delayed and survival prolonged in the treated

    mice, but not to the extent observed with mutant SOD1-specific siRNA. Suppres-

    sing genes such as Fas which are ubiquitously expressed also poses the problem that

    unforeseen side-effects are more likely than when targeting mutant alleles or genes

    unique to the target cell population. Expression of Fas in the CNS is believed to play

    a critical role in protecting neurons and resident CNS cells from potentially

    damaging affects of inflammatory cells that express FasL, resulting in apoptosis

    of the inflammatory cells. Thus, silencing Fas in the CNS may be associated with

    increased inflammation which could be problematic.

    6 Multiple Sclerosis

    Multiple sclerosis (MS) is an inflammatory, demyelinating disease of unknown

    etiology. The neurologic deficits appear to be mediated by demyelination and

    axonal transaction mediated by inflammatory cells. Thus, targeting components

    of the immune and nervous system may be necessary for optimal therapeutic

    intervention in MS. Diseases such as MS may be more easily modulated with

    siRNA than other CNS diseases because systemic administration of siRNA can

    readily target immune cells and potentially the site of active lesions due to BBB

    breakdown. Several inflammatory mediators appear to play a critical etiological

    role in MS. Specifically, it was demonstrated that systemic administration of

    286 A.E. Lovett-Racke

  • interferon gamma causes disease exacerbations (Panitch et al. 1987). Also, immu-

    nomodulation with agents that promote an anti-inflammatory cytokine profile are

    approved and currently being used for MS therapy (Hartung et al. 2004). In

    experimental autoimmune encephalomyelitis (EAE), a murine model of MS, the

    disease is mediated by myelin-specific CD4þ T cells that produce IFN-g, andsuppression of IFN-g in myelin-specific CD4þ T cells reduces the ability of thesecells to transfer disease to naı̈ve recipient mice (Olsson 1992; Racke et al. 1994,

    1995). Surprisingly, mice genetically deficient in IFN-g develop severe EAE,indicating that systemic loss of IFN-g results in immune dysregulation that mayactually enhance EAE (Ferber et al. 1996; Willenborg et al. 1996). Since IFN-g isexpressed by different types of immune cells that may be critical for normal

    immune regulation, siRNA specific for a transcription factor, T-bet, was developed

    to specifically inhibit IFN-g in CD4þ T cells since it appears that T-bet specificallyregulates IFN-g gene transcription in CD4þ T cells. Transfection of myelin-specificT cells in vitro with T-bet-specific siRNA prevented these cells from inducing EAE

    following transfer of these siRNA-transfected cells into naive mice (Lovett-Racke

    et al. 2004). More important, systemic administration of T-bet-specific siRNA at

    the time of EAE induction prevented the onset of disease, indicating that inhibition

    of a transcription factor critical to the development of the pathogenic T cells was

    sufficient to prevent disease. Since the cause of MS is unknown, potential therapies

    must be able to treat established disease. To address this issue, the T-bet-specific

    siRNA was administered to mice with established EAE and significantly amelio-

    rated disease, suggesting that T-bet was a viable target in ongoing disease patho-

    genisis (Gocke et al. 2007). More recently, it has been postulated that another

    population of CD4þ T cells that express IL-17 may be pathogenic in MS and EAE(Langrish et al. 2005). Interestingly, these IL-17-producing T cells have been

    shown to be suppressed with administration of T-bet siRNA in EAE, suggesting

    that both known populations of potentially pathogenic T cells may be targeted by

    the same siRNA (Gocke et al. 2007). Another transcription factor, NR4A2 (an

    orphan nuclear receptor), was found by microarray to be upregulated in peripheral

    blood T cells fromMS patients (Doi et al. 2008). Further analysis demonstrated that

    IFNg and IL-17 expression was enhanced by NR4A2. Suppression of NR4A2 withsiRNA in vitro suppressed IFNg and IL-17 production. Similar to T-bet siRNA,NR4A2 siRNA reduced the ability of encephalitogenic T cells to transfer EAE into

    recipient mice, suggesting that NR4A2 is critical for the pathogenic features of

    myelin-specific T cells (Doi et al. 2008). Other immune cell markers that are readily

    targeted in the periphery, such as IL-23 (Cua et al. 2003) and osteopontin (Chabas

    et al. 2001), which appear to play an essential role in the function of pathogenic T

    cells in immune-mediated demyelinating disease and whose deficiency does not

    appear to compromise the immune system, may also be potential targets for siRNA

    therapy in MS.

    Although MS typically begins as a relapsing-remitting disease, most patients

    ultimately develop permanent functional deficits and advance to secondary pro-

    gressive MS. Permanent neurologic deficits in MS patients appear to result from

    axonal damage mediated by repeated inflammation, and currently no therapeutic

    Therapeutic Potential of Small Interfering RNA for Brain Diseases 287

  • agents promote axonal repair. Nogo-A is a potent inhibitor of neurite outgrowth and

    probably plays a critical role in negatively regulating regeneration and plasticity in

    the adult nervous system (Buffo et al. 2000; Prinjha et al. 2000). Studies inhibiting

    Nogo-A or its receptor with monoclonal antibodies or peptide inhibitors in experi-

    mental models of acute spinal cord injury have shown enhanced functional recov-

    ery, apparently mediated by axonal sprouting (GrandPre et al. 2000, 2002). The

    Nogo receptor associates with a transmembrane receptor, p75NTR, which initiate

    inhibitory signals that cause growth cone collapse and cessation of axonal growth.

    Suppression of p75NTR with siRNA in rat retinal ganglion cells decreased Rho-A

    activation and increase neurite sprouting, supporting the hypothesis that siRNA can

    be used to target inhibitory proteins to promote axonal repair (Ahmed et al. 2006).

    Suppression of axonal growth inhibitors, such as Nogo-A or the Nogo receptor,

    with siRNA may be beneficial in the promoting axonal repair in MS. Since there is

    episodic BBB breakdown in EAE and MS, systemic administration of siRNA may

    be sufficient to target the areas of the lesions, while areas of the CNS that are not

    affected by lesions, and potentially adversely affected by suppressing these pro-

    teins, would not be exposed to the siRNA. The glial reaction to injury results in the

    production of many other inhibitory and potentially toxic molecules that damage

    axons and prevent axonal repair which are also potential therapeutic targets for MS

    siRNA therapies (Fawcett and Asher 1999). Other CNS proteins that contribute to

    demyelination and wallerian degeneration may also be targets for RNAi in MS and

    other CNS diseases with similar mediators of pathology and repair.

    7 Brain Tumors

    Primary brain tumors remain one of the most difficult tumors to treat. Glioblastoma

    multiforme (GBM) is the most malignant form of human astrocytomas, accounting

    for almost 22% of all brain tumors. The worldwide incidence of brain tumors is

    approximately 3 per 100,000. Five-year survival for GBM is less than 3%, empha-

    sizing the critical need for better therapies for CNS malignancies. In addition,

    peripheral malignancies that metastasize to the CNS are often as difficult to treat

    as primary brain tumors. Although new therapies such as Herceptin, which inhibits

    growth of Her-2 positive cancer cells, has been shown to be very effective in breast

    cancer, this agent is not effective against breast cancer cells that have metastasized

    to the CNS (Bendell et al. 2003). Treatment of brain malignancies is hindered by

    the inability of most therapeutic agents to cross the BBB, as well as poor respon-

    siveness of brain tumors to current therapeutic agents. Molecular characterization

    of brain tumors has defined several molecules that appear to be essential for tumor

    survival, proliferation, metastasis and angiogenesis. Thus, siRNA technology tar-

    geted against specific molecules critical for tumors may be more effective therapies

    for brain tumors.

    One molecule that appears to be a potential target for the treatment of many

    cancers is epidermal growth factor receptor (EGFR). Proliferation of 90% of GBM

    288 A.E. Lovett-Racke

  • is driven by EFGR (Kaun et al. 2001; Kleihues and Ohgaki 1999), indicating that

    minimizing EFGR-mediated signaling may be an effective therapeutic option for

    GBM. In addition, EGFR plays an oncogenic role in nearly 70% of peripheral solid

    tumors that metastasize to the CNS (Nicholson et al. 2001). In tumors, the EGFR

    gene is frequently amplified which enhances the proliferative capacity of the tumor

    cells, or the EGFR gene contains mutations that alter signaling. The most common

    EGFRmutation is an in-frame deletion of exons 2–7 which generates the EGFR vIII

    protein that is not very responsive to its natural ligands, but signals continuously

    promoting the proliferation of cancer cells (Kaun et al. 2001). In an attempt to target

    the EGFR vIII protein which is observed in 40% of GBM, siRNA were developed

    that targeted the exon1–exon 8 junction sequence of mutant EGFR (Fan and Weiss

    2005). In human glioblastoma cell lines, this siRNA reduced phosphorylated Akt,

    enhanced apoptosis, and caused partial arrest of the cell cycle at the G2M phase. To

    determine if amplified wild-type EGFR could be targeted in glioma cells, plasmids

    that expressed shRNA targeting native EGFR were transfected into human glio-

    blastoma cell lines (Kang et al. 2005; Saydam et al. 2005). The transfected cells had

    a diminished capacity to proliferate and enhanced apoptosis. Transwell studies

    demonstrated that invasiveness was also inhibited. More importantly, suppression

    of EGFR with shRNA inhibited the growth of human glioblastoma cells in athymic

    mice, indicating that targeting EGFR reduced the capacity of malignant brain

    tumors to thrive in vivo.

    Since the ultimate goal in the treatment of brain tumors is to develop therapeu-

    tics that are specific and effective, combining targeted delivery with RNAi may

    provide optimal benefit in treating CNS tumors. Plasmids expressing shRNA

    specific for wild-type EGFR were encapsulated in monoclonal antibody-coated

    pegylated liposomes, often referred to as Trojan horse liposomes (Zhang et al.

    2004). One monoclonal antibody recognizes the mouse transferrin receptor that can

    mediate transport of the liposomes across the BBB, and another monoclonal

    antibody recognized the human insulin receptor that is expressed on human glio-

    blastoma cells (Fig. 2). Receptor-mediated transport would allow the liposomes to

    cross the BBB and specifically deliver the plasmids containing EGFR shRNA to the

    glioma cells. Human glioblastoma cells were injected into the brain of scid mice

    and the antibody-coated liposomes carrying the plasmids carrying shRNA specific

    for EGFR were infused intravenously once per week. EGFR expression on tumor

    cells was significantly decreased and survival increased almost 90% in this in vivo

    model of human glioblastoma. This study demonstrated that two of the greatest

    hurdles in brain tumor therapies, BBB and efficacy of the therapeutic agent, can be

    overcome with targeted delivery of siRNA.

    Growth factors, such as vascular endothelial growth factor (VEGF) and pleio-

    trophin (PTN), have also been shown to play a role in glioblastoma pathogenesis,

    and siRNA has been used to target these growth factors with some success

    (Grzelinski et al. 2006; Niola et al. 2006). Growth factor receptors, such as

    EGFR, VEGFR, platelet-derived growth factor receptor (PDGFR), and insulin

    growth factor receptor (IGFR), are all frequently overexpressed in malignant

    gliomas and share a common signaling pathway. Therefore, signaling molecules

    Therapeutic Potential of Small Interfering RNA for Brain Diseases 289

  • in this pathway, such as PI3 kinase, may be more effective targets for siRNA

    therapy since all of them can be suppressed simultaneously. The catalytic subunit

    of PI3 kinase, p110 beta, was target with plasmid-expressed siRNA in human

    glioma cell lines, resulting in decreased proliferation, arrest of cell cycle, decreased

    invasiveness, and increased apoptosis (Pu et al. 2006). Growth of human glioma

    cells in nude mice was also significantly impaired, illustrating the potential of PI3

    kinase as a therapeutic siRNA target.

    Identification of molecules critical in tumor survival, proliferation, and invasive-

    ness remains a critical issue in developing siRNA-based therapeutics given the vast

    heterogeneity of brain tumors. Numerous genes have been shown to be critical in

    different types of human brain cancers and attempts to target them with siRNA have

    shown promise. Downregulation of Notch-1 and its ligands, which are overex-

    pressed in primary human gliomas, inhibit proliferation, and induce apoptosis

    (Purow et al. 2005). In addition, transfection of glioma cells with siRNA

    specific for Notch-1 prior to injection into mice enhances survival. However,

    Notch signaling is critical in developmental cell fate and adult nervous system

    plasticity, which may limit Notch as a viable therapeutic target. Proteases that

    enhance extracellular matrix degradation promote tumor invasion and progression.

    Cathepsin B, urokinase-type plasminogen activator receptor, and matrix metallo-

    proteinase have been inhibited with siRNA both in vitro and in vivo in several brain

    tumor models (Gondi et al. 2004, 2007; Lakka et al. 2005; Tummalapalli et al.

    2007; Kargiotis et al. 2008). Both tumor invasion and angiogenesis were reduced,

    and established intracranial tumors in mice demonstrated significant regression

    following intracranial injection of plasmids expressing these siRNAs. Similar to

    Notch-1, these proteases are abundantly expressed and play central roles in normal

    biological functions such as immune surveillance. Therefore, these targets should

    be studied cautiously and evaluated carefully to determine if the therapeutic benefit

    is greater than potential side-effects in fatal illness such as brain tumors.

    However, proteins like tenascin-C which is typically expressed in adults under

    pathological conditions, including human brain tumors, may be better therapeutic

    targets. Although the role of tenascin-C in brain tumors in unclear, its expression is

    associated with tumor grade. To determine if tenascin-C could be targeted in

    patients, ten high-grade and low-grade glioma patients with poor prognosis were

    chosen for siRNA therapy following surgical transection (Zukiel et al. 2006).

    Plasmids carrying shRNA specific for tenascin-C were injected directly into neo-

    plastic brain tissue that was inaccessible to surgical removal. Postsurgical imaging

    showed reduced tumor burden in the areas of injection of the shRNA, but some

    patients had new tumors in remote areas, suggesting that the efficacy of the RNAi

    therapy was limited to the injection site.

    RNAi technology has also been used to enhance the efficacy of other therapeutic

    modalities in models of human glioblastomas. Suppression of galectin 1, a hypoxia-

    regulated proangiogenic factor that promotes glioblastoma cell migration, enhances

    the therapeutic effect of the proautophagic agent temozolomide in vivo (Le Mercier

    et al. 2008). The anti-apoptotic molecule Bcl-2 has also been targeted with siRNA,

    enhancing apoptosis of glioma cell lines, but the apoptotic effect was significantly

    290 A.E. Lovett-Racke

    Joseph GeorgeRectangle

  • increased with the chemotherapeutic agent taxol (George et al. 2008). A VEGF

    siRNA, shown to significantly reduce vascularization of glioblastoma cells in nude

    mice, did not appear to alter tumor growth in vivo (Niola et al. 2006). However,

    using a retroviral vector that expressed VEGF siRNA and an IL-4 transgene, tumor

    growth was totally abolished, illustrating the synergistic effect between a siRNA

    and transgene. Since combination therapy has proved to be the most beneficial

    therapeutic option for most types of cancers, the use of siRNA with other thera-

    peutic modalities may prove to be the future for brain tumors.

    8 Conclusions

    Most pharmacotherapies that are currently being used for treatment of neurodegen-

    erative diseases, inflammatory CNS disorders, and brain tumors target molecules

    that are localized downstream in the pathogenic cascade. As a consequence, their

    effects are often not specific and their efficacy is moderate with regard to disease

    modulation. Furthermore, some of these therapeutics, such as chemotherapeutic

    agents for brain tumors and MS, are often associated with adverse effects that limit

    their use. RNAi technology has two major advantages over conventional therapeu-

    tic strategies. They can be used diagnostically to identify gene mutations associated

    with a clinical phenotype, and they can directly inhibit the gene product involved in

    the pathology of a disease. With our ever-accumulating knowledge of pathways that

    mediate pathogenic processes, it will soon be possible to design RNAi-based

    therapies that are specific for certain pathologic conditions and have relatively

    few systemic adverse effects.

    References

    Ahmed Z, Suggate EL, Brown ER et al (2006) Schwann cell-derived factor-induced modulation of

    the NgR/p75NTR/EGFR axis disinhibits axon growth through CNSmyelin in vivo and in vitro.

    Brain 129:1517–1533

    Begley DJ (2004) Delivery of therapeutic agents to the central nervous system: the problems and

    the possibilities. Pharmacol Ther 104:29–45

    Bendell JC, Domchek SM, Burstein HJ et al (2003) Central nervous system metastases in women

    who receive trastuzumab-based therapy for metastatic breast carcinoma. Cancer 97:2972–2977

    Boado RJ (2007) Blood-brain barrier transport of non-viral gene and RNAi therapeutics. Pharm

    Res 24:1772–1787

    Buffo A, Zagrebelsky M, Huber AB et al (2000) Application of neutralizing antibodies against

    NI-35/250 myelin-associated neurite growth inhibitory proteins to the adult rat cerebellum

    induces sprouting of uninjured Purkinje cell axons. J Neurosci 20:2275–2286

    Chabas D, Baranzini SE, Mitchell D et al (2001) The influence of the proinflammatory cytokine,

    osteopontin, on autoimmune demyelinating disease. Science 294:1731–1735

    Chen ZJ, Kren BT, Wong PY et al (2005) Sleeping Beauty-mediated down-regulation of hunting-

    tin expression by RNA interference. Biochem Biophys Res Commun 329:646–652

    Therapeutic Potential of Small Interfering RNA for Brain Diseases 291

    Joseph GeorgeRectangle

  • Chu TC, Twu KY, Ellington AD et al (2006) Aptamer mediated siRNA delivery. Nucleic Acids

    Res 34:e73

    Corder EH, Saunders AM, Strittmatter WJ et al (1993) Gene dose of apolipoprotein E type 4 allele

    and the risk of Alzheimer’s disease in late onset families. Science 261:921–923

    Cua DJ, Sherlock J, Chen Y et al (2003) Interleukin-23 rather than interleukin-12 is the critical

    cytokine for autoimmune inflammation of the brain. Nature 421:744–748

    Denovan-Wright EM, Rodriguez-Lebron E, Lewin AS et al (2008) Unexpected off-targeting

    effects of anti-huntingtin ribozymes and siRNA in vivo. Neurobiol Dis 29:446–455

    DiFiglia M, Sena-Esteves M, Chase K et al (2007) Therapeutic silencing of mutant huntingtin with

    siRNA attenuates striatal and cortical neuropathology and behavioral deficits. Proc Natl Acad

    Sci USA 104:17204–17209

    Ding H, Schwarz DS, Keene A et al (2003) Selective silencing by RNAi of a dominant allele that

    causes amyotrophic lateral sclerosis. Aging Cell 2:209–217

    Doi Y, Oki S, Ozawa T et al (2008) Orphan nuclear receptor NR4A2 expressed in T cells from

    multiple sclerosis mediates production of inflammatory cytokines. Proc Natl Acad Sci USA

    105:8381–8386

    Duyao MP, Auerbach AB, Ryan A et al (1995) Inactivation of the mouse Huntington’s disease

    gene homolog Hdh. Science 269:407–410

    Elman J, Thonberg H, Ljumberg K et al (2005) Locked nucleic acid (LNA) mediated improve-

    ments in siRNA stability and functionality. Nucleic Acids Res 33:439–447

    Fan QW, Weiss WA (2005) RNA interference against a glioma-derived allele of EGFR induces

    blockade at G2M. Oncogene 24:829–837

    Fawcett JW, Asher RA (1999) The glial scar and central nervous system repair. Brain Res Bull

    49:377–391

    Ferber IA, Brock S, Taylor-Edwards C et al (1996) Mice with a disrupted IFN-gamma gene are

    susceptible to the induction of experimental autoimmune encephalomyelitis (EAE). J Immunol

    156:5–7

    Fire A, Xu S, Montgomery MK, Kostas SA et al (1998) Potent and specific genetic interference by

    double-stranded RNA in Caenorhabditis elegans. Nature 391:806–811

    Fountaine TM, Wade-Martins R (2007) RNA interference-mediated knockdown of alpha-

    synuclein protects human dopaminergic neuroblastoma cells from MPP(+) toxicity and

    reduces dopamine transport. J Neurosci Res 85:351–363

    George J, Banik NL, Ray SK (2008) Bcl-2 siRNA augments taxol mediated apoptotic death in

    human glioblastoma U138MG and U251MG cells. Neurochem Res. Doi: 10.1007/s11064–

    008–9659-z

    Goate A, Chartier-Harlin M-C, Mullan M et al (1991) Segregation of a missense mutation in the

    amyloid precursor protein gene with familial Alzheimer’s disease. Nature 349:704–706

    Gocke AR, Cravens PD, Ben L et al (2007) T-bet regulates the fate of Th1 and Th17 lymphocytes

    in autoimmunity. J Immunol 178:1341–1348

    Gondi CS, Lakka SS, Dinh DH et al (2004) RNAi-mediated inhibition of cathepsin B and uPAR

    leads to decreased cell invasion, angiogenesis and tumor growth in gliomas. Oncogene

    23:8486–8496

    Gondi CS, Lakka SS, Dinh DH et al (2007) Intraperitoneal injection of a hairpin RNA-expressing

    plasmid targeting urokinase-type plasminogen activator (uPA) receptor and uPA retards

    angiogenesis and inhibits intracranial tumor growth in nude mice. Clin Cancer Res 13:4051–

    4060

    GrandPre T, Nakamura F, Vartanian T et al (2000) Identification of the Nogo inhibitor of axon

    regeneration as a Reticulon protein. Nature 403:439–444

    GrandPre T, Li S, Strittmatter SM (2002) Nogo-66 receptor antagonist peptide promotes axonal

    regeneration. Nature 417:547–551

    Grzelinski M, Urban-Klein B, Martens T et al (2006) RNA interference-mediated gene silencing

    of pleiotrophin through polyethlenimine-complexed small interfering RNAs in vivo exerts

    antitumoral effects in glioblastoma xenografts. Hum Gene Ther 17:751–766

    292 A.E. Lovett-Racke

    Joseph GeorgeRectangle

  • Habig K, Walter M, Poths S et al (2008) RNA interference of LRRK2-microarray expression

    analysis of a Parkinson’s disease key player. Neurogenetics 9:83–94

    Hamilton AJ, Baulcombe DC (1999) A species of small antisense RNA in posttranscriptional gene

    silencing in plants. Science 286:950–952

    Harper SQ, Staber PD, He X et al (2005) RNA interference improves motor and neuropathological

    abnormalities in a Huntington’s disease mouse model. Proc Natl Acad Sci USA 102:5820–5825

    Hartung HP, Bar-Or A, Zoukos Y (2004) What do we know about the mechanism of action of

    disease-modifying treatments in MS? J Neurol 251(suppl 5):v12–v29

    Hengst U, Cox LJ, Macosko EZ et al (2006) Functional and selective RNA interference in

    developing axons and growth cones. J Neurosci 26:5727–5732

    Hong CS, Goins WF, Goss JR et al (2006) Herpes simplex virus RNAi and neprilysin gene transfer

    vectors reduce accumulation of Alzheimer’s disease-related amyloid-beta peptide in vivo.

    Gene Ther 13:1068–1079

    Houlden H, Baker M, Morris HR et al (2001) Corticobasal degeneration and progressive supra-

    nuclear palsy share a common tau haplotype. Neurology 56:1702–1706

    Juliano RL (2005) Peptide-oligonucleotide conjugates for the delivery of antisense and siRNA.

    Curr Opin Mol Ther 7:132–136

    Kang CS, Pu PY, Li YH et al (2005) An in vitro study on the suppressive effect of glioma cell

    growth induced by plasmid-based small interference RNA (siRNA) targeting human epidermal

    growth factor receptor. J Neurooncol 74:267–273

    Kao SC, Krichevsky AM, Kosik KS et al (2004) BACE1 suppression by RNA interference in

    primary cortical neurons. J Biol Chem 279:1942–1949

    Kargiotis O, Chetty C, Gondi CS et al (2008) Adenovirus-mediated transfer of siRNA against

    MMP-2 mRNA results in impaired invasion and tumor-induced angiogenesis, induces apop-

    tosis in vitro and inhibits tumor growth in vivo in glioblastoma. Oncogene 27:4830–4840

    Kaun CT, Wikstrand CJ, Bigner DD (2001) EGF mutant receptor vIII as a molecular target in

    cancer therapy. Endocr Relat Cancer 8:83–96

    Kleihues P, Ohgaki H (1999) Primary and secondary glioblastomas from concept to clinical

    diagnosis. Neuro-oncol 1:44–51

    Kumar P, Ban HS, Kim SS et al (2008) T cell-specific siRNA delivery suppresses HIV-1 infection

    in humanized mice. Cell 134:577–586

    Lakka SS, Gondi CS, Dinh DH et al (2005) Specific interference of urokinase-type plasminogen

    activator receptor and matrix metalloproteinase-9 gene expression induced by double-stranded

    RNA results in decreased invasion, tumor growth and angiogenesis in gliomas. J Biol Chem

    280:21882–21892

    Langrish CL, Chen Y, Blumenschein M et al (2005) IL-23 drives a pathogenic T cell population

    that induces autoimmune inflammation. J Exp Med 201:233–240

    Le Mercier M, Mathieu V, Haibe-Kains B et al (2008) Knocking down galectin 1 in human hs683

    glioblastoma cells impairs both angiogenesis and endoplasmic reticulum stress responses.

    J Neuropathol Exp Neurol 67:456–469

    Lewis J, Dickson DW, Lin WI et al (2001) Enhanced neurofibrillary degeneration in transgenic

    mice expressing mutant tau and APP. Science 293:1487–1491

    Locatelli F, Corti S, Papadimitriou D et al (2007) Fas small interfering RNA reduces motoneuron

    death in amyotrophic lateral sclerosis mice. Ann Neurol 62:81–92

    Lovett-Racke AE, Rocchini AE, Choy J et al (2004) Silencing T-bet defines a critical role in the

    differentiation of autoreactive T lymphocytes. Immunity 21:719–731

    Luo Y, Bolon B, Kahn S et al (2001) Mice deficient in BACE1, the Alzheimer’s beta-secretase,

    have normal phenotype and abolished beta-amyloid generation. Nat Neurosci 4:231–232

    Luo HM, Deng H, Xiao F et al (2004) Down-regulation amyloid beta-protein 42 production

    by interfering with transcript of presenilin 1 gene with siRNA. Acta Pharmacol Sin

    25:1613–1618

    Machida Y, Okada T, Kurosawa M et al (2006) rAAV-mediated shRNA ameliorated neuro-

    pathology in Huntingtin disease model mouse. Biochem Biophys Res Commun 343:190–197

    Therapeutic Potential of Small Interfering RNA for Brain Diseases 293

  • Maxwell MM, Pasinelli P, Kazantsev AG et al (2004) RNA interference-mediated silencing of

    mutant superoxide dismutase rescues cyclosporin A-induced death in cultured neuroblastoma

    cells. Proc Natl Acad Sci USA 101:3178–3183

    Miller VM, Xia H, Marrs GL et al (2003) Allele-specific silencing of dominant disease genes. Proc

    Natl Acad Sci USA 100:7195–7200

    Miller VM, Gouvion CM, Davidson BL et al (2004) Targeting Alzheimer’s disease genes

    with RNA interference: an efficient strategy for silencing mutant alleles. Nucleic Acids Res

    32:661–668

    Miller TM, Kaspar BK, Kops GJ et al (2005) Virus-delivered small RNA silencing sustains

    strength in amyotrophic lateral sclerosis. Ann Neurol 57:773–776

    Morrissey DV, Lockridge JA, Shaw L et al (2005) Potent and persistent in vivo anti-HBV activity

    of chemically modified siRNA. Nat Biotechnol 23:1002–1007

    Mullan M, Crawford F, Axelman K et al (1992) A pathogenic mutation for probable Alzheimer’s

    disease in the APP gene at the N-terminus of beta amyloid. Nat Genet 1:345–347

    Muratovska A Ecles MR (2004) Conjugate for efficient delivery of short interfering RNA (siRNA)

    into mammalian cells. FEBS Lett 558:63–68

    Napoli C, Lemieux C, Jorgensen R (1990) Introduction of a chimeric chalcone synthase gene

    into petunia results in reversible co-suppression of homologous genes in trans. Plant Cell

    2:279–289

    Nasir J, Floresco SB, O’Kusky JR et al (1995) Targeted disruption of the Huntington’s disease

    gene results in embryonic lethality and behavioral and morphological changes in heterozy-

    gotes. Cell 81:811–823

    Nawrot B (2004) Targeting BACE with small inhibitory nucleic acids – a future for Alzheimer’s

    disease therapy. Acta Biochem Pol 51:431–444

    Nicholson RI, Gee JM, Harper ME (2001) EGFR and cancer prognosis. Eur J Cancer 37:S9–S15

    Niola F, Evangelisti C, Campangnolo L et al (2006) A plasmid-encoded VEGF siRNA reduces

    glioblastoma angiogenesis and its combination with interleukin-4 blocks tumor growth in a

    xenograft mouse model. Cancer Biol Ther 5:174–179

    Oddo S, Caccamo A, Shepherd JD et al (2003) Triple-transgenic model of Alzheimer’s disease

    with plaques and tangles: intracellular Abeta and synaptic dysfunction. Neuron 39:409–421

    Olsson T (1992) Cytokines in neuroinflammatory disease: role of myelin autoreactive T cell

    production of interferon-gamma. J Neuroimmunol 40:211–218

    Outeiro TF, Kontopoulos E, Altmann SM et al (2007) Sirtuin 2 inhibitors rescue alpha-synuclein-

    mediated toxicity in models of Parkinson’s disease. Science 317:516–519

    Pals P, Lincoln S, Manning J et al (2004) Alpha-synuclein promoter confers susceptibility to

    Parkinson’s disease. Ann Neurol 56:591–595

    Panitch HS, Hirsch RL, Haley AS et al (1987) Exacerbation of multiple sclerosis in patients with

    gamma interferon. Lancet 1:893–895

    Polymeropoulos MH, Lavedan C, Leroy E et al (1997) Mutation in the alpha-synuclein gene

    identified in families with Parkinson’s disease. Science 276:2045–2047

    Prinjha R, Moore SE, Vinson M et al (2000) Inhibitor of neurite outgrowth in humans. Nature

    403:383–384

    Pu P, Kang C, Zhang Z et al (2006) Downregulation of PIK3CB by siRNA suppresses malignant

    glioma cell growth in vitro and in vivo. Technol Cancer Res Treat 5:271–280

    Purow BW, Haque RM, Noel MW et al (2005) Expression of Notch-1 and its ligands, Delta-like-1

    and Jagged-1, is critical for glioma cell survival and proliferation. Cancer Res 65:2353–2363

    Racke MK, Bonomo A, Scott DE et al (1994) Cytokine-induced immune deviation as a therapy for

    inflammatory autoimmune disease. J Exp Med 180:1961–1966

    Racke MK, Burnett D, Pak SH et al (1995) Retinoid treatment of experimental allergic

    encephalomyelitis: IL-4 production correlates with improved disease course. J Immunol

    154:450–458

    Ralph GS, Radcliffe PA, Day DM et al (2005) Silencing mutant SOD1 using RNAi protects

    against neurodegeneration and extends survival in an ALS model. Nat Med 11:429–433

    294 A.E. Lovett-Racke

  • Raoul C, Abbas-Terki T, Bensadoun JC et al (2005) Lentiviral-mediated silencing of SOD1

    through RNA interference retards disease onset and progression in a mouse model of ALS.

    Nat Med 11:423–428

    Romano N, Macino G (1992) Quelling: transient inactivation of gene expression in Neurospora

    crassa by transformation with homologous sequences. Mol Microbiol 6:3343–3353

    Saito Y, Yokota T, Mitani T et al (2005) Transgenic small interfering RNA cells halts amyotrophic

    lateral sclerosis in a mouse model. J Biol Chem 280:42826–42830

    Sapru MK, Yates JW, Hogan S et al (2006) Silencing of human alpha-synuclein in vitro and in rat

    brain using lentiviral-mediated RNAi. Exp Neurol 198:382–390

    Saydam O, Glauser DL, Heid I et al (2005) Herpes simplex virus 1 amplicon vector-mediated

    siRNA targeting epidermal growth factor receptor inhibits growth of human glioma cells in

    vivo. Mol Ther 12:803–812

    Schwarz DS, Ding H, Kennington L et al (2006) Designing siRNA that distinguish between genes

    that differ by a single nucleotide. PLoS Genet 2:e140

    Singer O, Marr RA, Rockenstein E et al (2005) Targeting BACE1 with siRNAs ameliorates

    Alzheimer disease neuropathology in a transgenic model. Nat Neurosci 8:1343–1349

    Singleton AB, Farrer M, Johnson J et al (2003) Alpha-synuclein locus triplication causes Parkin-

    son’s disease. Science 302:841

    Thakker DR, Natt F, Husken D et al (2004) Neurochemical and behavioral consequences of

    widespread gene knockdown in the adult mouse brain using nonviral RNA interference. Proc

    Natl Acad Sci USA 101:17270–17275

    Tummalapalli P, Gondi CS, Dinh DH et al (2007) RNA interference-mediated targeting of

    urokinase plasminogen activator receptor and matrix metalloproteinase-9 gene expression in

    the IOMM-lee malignant meningioma cell line inhibits tumor growth, tumor cell invasion and

    angiogenesis. Int J Oncol 31:5–17

    Van der Krol AR, Mur AL, Beld M et al (1990) Flavanoid genes in petunia: addition of a limited

    number of gene copies may lead to a suppression of gene expression. Plant Cell 2:291–299

    Wang H, Ghosh A, Baigude H et al (2008) Therapeutic gene silencing delivered by a chemically

    modified small interfering RNA against mutant SOD1 slows amyotrophic lateral sclerosis

    progression. J Biol Chem 283:15845–15852

    Willenborg DO, Fordham S, Bernard CC et al (1996) IFN-gamma plays a critical down-regulatory

    role in the induction and effector phase of myelin oligodendrocyte-induced autoimmune

    encephalomyelitis. J Immunol 157:3223–3227

    Xia H, Qinwen M, Eliason SL et al (2004) RNAi suppresses polyglutamine-induced neurodegen-

    eration in a model of spinocerebellar ataxia. Nat Med 10:816–820

    Xia XG, Zhou H, Zhou S et al (2005) An RNAi strategy for treatment of amyotrophic lateral

    sclerosis caused by mutant Cu, Zn superoxide dismutase. J Neurochem 92:362–367

    Xia X, Zhou H, Huang Y et al (2006) Allele-specific RNAi selectively silences mutant SOD1 and

    achieves significant therapeutic benefit in vivo. Neurobiol Dis 23:578–586

    Yokota T, Miyagishi M, Hino T et al (2004) siRNA-based inhibition specific for mutant SOD1

    with single nucleotide alternation in familial ALS, compared with ribozyme and DNA enzyme.

    Biochem Biophys Res Commun 314:283–291

    Zeitlin S, Liu JP, Chapman DL et al (1995) Increased apoptosis and early embryonic lethality in

    mice nullizygous for the Huntington’s disease gene homologue. Nat Genet 11:155–163

    Zhang Y, Zhang Y, Bryant J et al (2004) Intravenous RNA interference gene therapy targeting the

    human epidermal growth factor receptor prolongs survival in intracranial brain cancer. Clin

    Cancer Res 10:3667–3677

    Zhu Y, Hoell P, Ahlemeyer B et al (2007) Implications of PTEN in production of reactive oxygen

    species and neuronal death in in vitro models of stroke and Parkinson’s disease. Neurochem Int

    50:507–516

    Zukiel R, Nowak S, Wyszko E et al (2006) Suppression of human brain tumor with interference

    RNA specific for tenascin-C. Cancer Biol Ther 5:1002–1007

    Therapeutic Potential of Small Interfering RNA for Brain Diseases 295

    References1 RNA Interference2 Spinocerebellar Ataxia and Other Polyglutamine-Associated Ataxias3 Alzheimer's Disease4 Parkinson's Disease5 Amyotrophic Lateral Sclerosis6 Multiple Sclerosis7 Brain Tumors8 ConclusionsReferences

    /ColorImageDict > /JPEG2000ColorACSImageDict > /JPEG2000ColorImageDict > /AntiAliasGrayImages false /CropGrayImages true /GrayImageMinResolution 150 /GrayImageMinResolutionPolicy /Warning /DownsampleGrayImages true /GrayImageDownsampleType /Bicubic /GrayImageResolution 150 /GrayImageDepth -1 /GrayImageMinDownsampleDepth 2 /GrayImageDownsampleThreshold 1.50000 /EncodeGrayImages true /GrayImageFilter /DCTEncode /AutoFilterGrayImages true /GrayImageAutoFilterStrategy /JPEG /GrayACSImageDict > /GrayImageDict > /JPEG2000GrayACSImageDict > /JPEG2000GrayImageDict > /AntiAliasMonoImages false /CropMonoImages true /MonoImageMinResolution 600 /MonoImageMinResolutionPolicy /Warning /DownsampleMonoImages true /MonoImageDownsampleType /Bicubic /MonoImageResolution 600 /MonoImageDepth -1 /MonoImageDownsampleThreshold 1.50000 /EncodeMonoImages true /MonoImageFilter /CCITTFaxEncode /MonoImageDict > /AllowPSXObjects false /CheckCompliance [ /None ] /PDFX1aCheck false /PDFX3Check false /PDFXCompliantPDFOnly false /PDFXNoTrimBoxError true /PDFXTrimBoxToMediaBoxOffset [ 0.00000 0.00000 0.00000 0.00000 ] /PDFXSetBleedBoxToMediaBox true /PDFXBleedBoxToTrimBoxOffset [ 0.00000 0.00000 0.00000 0.00000 ] /PDFXOutputIntentProfile (None) /PDFXOutputConditionIdentifier () /PDFXOutputCondition () /PDFXRegistryName () /PDFXTrapped /False

    /CreateJDFFile false /Description >>> setdistillerparams> setpagedevice