Developments in the discovery of drugs for spinal muscular atrophy: successful beginnings and future...

11
1. Introduction 2. Strategies for SMA drug discovery: increasing SMN 3. Compensating for functional consequences of SMN deficits 4. Other strategies besides increasing SMN 5. Is SMA a single disease? Unraveling disease mechanism and alternative targets 6. Neuroprotection, alone or in combination with SMN enhancers 7. Conclusion 8. Expert opinion Review Developments in the discovery of drugs for spinal muscular atrophy: successful beginnings and future prospects Rebecca M. Pruss Trophos, SA Parc Scientifique de Luminy Case, Marseille Cedex, France Introduction: Spinal muscular atrophy (SMA) is an autosomal recessive disease caused by mutations in a gene that produces a protein called survival motor neuron (SMN). SMN has an important role in snRNP assembly in all cells but that may not be its only role; the reasons for SMN deficiency resulting in neu- romuscular dysfunction and motor neuron degeneration remain active areas of research. Besides increasing SMN, compensating for SMN deficiencies or neuroprotection may be therapeutic options for SMA. Age of onset and the rate of disease progression are variable and therapeutic strategies should be appropriate to subtypes of SMA patients. Areas covered: The article discusses SMA, their targets and where these targets can be found. Additionally, the article reviews small molecules identified as disease modifiers and how these small molecules were discovered. The article also describes and discusses emerging concepts regarding the disease mecha- nisms. The author compiled this review using scientific literature, patent databases, company and patient association and government websites. Expert opinion: Small molecules targeting various processes implicated in SMA are reaching the clinic. These molecules and targets, although not yet validated, are providing insight into the complexity of a ‘simple’ genetic dis- ease such as SMA. SMA is not a single disease and so various therapeutic strat- egies are needed. Biomarkers and regulatory guidelines are required to select patients for clinical trials, decide when to initiate treatment and how to develop combinations of investigational drugs. Keywords: biomarkers, disease mechanisms, drug discovery, neurodegeneration, neuromuscular disease Expert Opin. Drug Discov. (2011) 6(8):827-837 1. Introduction Spinal muscular atrophy (SMA) is a rare neuromuscular disease that presents in infants and young children. SMA is an autosomal recessive disease affecting an esti- mated 1 in 6000 births and is caused by progressive degeneration of motor neurons innervating proximal skeletal muscles due to deletions or mutations in a gene cod- ing for a protein called survival motor neuron (SMN) [1]. Independently, and nearly simultaneously, the SMN protein, which is expressed in all cells, was discovered to be a central scaffolding element necessary for the assembly of small nuclear ribonu- clear protein (snRNP) complexes involved in mRNA splicing [2-4]. Discovering both the gene responsible for SMA and a function for the encoded protein was a remark- able coincidence, making SMA an exception with respect to other inherited neuro- degenerative diseases such as Huntington’s disease and familial forms of Parkinson’s disease, Alzheimer’s disease or amyotrophic lateral sclerosis (ALS). 10.1517/17460441.2011.586692 © 2011 Informa UK, Ltd. ISSN 1746-0441 827 All rights reserved: reproduction in whole or in part not permitted Expert Opin. Drug Discov. Downloaded from informahealthcare.com by RMIT University on 09/03/13 For personal use only.

Transcript of Developments in the discovery of drugs for spinal muscular atrophy: successful beginnings and future...

1. Introduction

2. Strategies for SMA drug

discovery: increasing SMN

3. Compensating for functional

consequences of SMN deficits

4. Other strategies besides

increasing SMN

5. Is SMA a single disease?

Unraveling disease mechanism

and alternative targets

6. Neuroprotection, alone or in

combination with SMN

enhancers

7. Conclusion

8. Expert opinion

Review

Developments in the discovery ofdrugs for spinal muscular atrophy:successful beginnings and futureprospectsRebecca M. PrussTrophos, SA Parc Scientifique de Luminy Case, Marseille Cedex, France

Introduction: Spinal muscular atrophy (SMA) is an autosomal recessive disease

caused by mutations in a gene that produces a protein called survival motor

neuron (SMN). SMN has an important role in snRNP assembly in all cells but

that may not be its only role; the reasons for SMN deficiency resulting in neu-

romuscular dysfunction and motor neuron degeneration remain active areas

of research. Besides increasing SMN, compensating for SMN deficiencies or

neuroprotection may be therapeutic options for SMA. Age of onset and the

rate of disease progression are variable and therapeutic strategies should be

appropriate to subtypes of SMA patients.

Areas covered: The article discusses SMA, their targets and where these targets

can be found. Additionally, the article reviews small molecules identified as

disease modifiers and how these small molecules were discovered. The article

also describes and discusses emerging concepts regarding the disease mecha-

nisms. The author compiled this review using scientific literature, patent

databases, company and patient association and government websites.

Expert opinion: Small molecules targeting various processes implicated in

SMA are reaching the clinic. These molecules and targets, although not yet

validated, are providing insight into the complexity of a ‘simple’ genetic dis-

ease such as SMA. SMA is not a single disease and so various therapeutic strat-

egies are needed. Biomarkers and regulatory guidelines are required to select

patients for clinical trials, decide when to initiate treatment and how to

develop combinations of investigational drugs.

Keywords: biomarkers, disease mechanisms, drug discovery, neurodegeneration,

neuromuscular disease

Expert Opin. Drug Discov. (2011) 6(8):827-837

1. Introduction

Spinal muscular atrophy (SMA) is a rare neuromuscular disease that presents ininfants and young children. SMA is an autosomal recessive disease affecting an esti-mated 1 in 6000 births and is caused by progressive degeneration of motor neuronsinnervating proximal skeletal muscles due to deletions or mutations in a gene cod-ing for a protein called survival motor neuron (SMN) [1]. Independently, and nearlysimultaneously, the SMN protein, which is expressed in all cells, was discovered tobe a central scaffolding element necessary for the assembly of small nuclear ribonu-clear protein (snRNP) complexes involved in mRNA splicing [2-4]. Discovering boththe gene responsible for SMA and a function for the encoded protein was a remark-able coincidence, making SMA an exception with respect to other inherited neuro-degenerative diseases such as Huntington’s disease and familial forms of Parkinson’sdisease, Alzheimer’s disease or amyotrophic lateral sclerosis (ALS).

10.1517/17460441.2011.586692 © 2011 Informa UK, Ltd. ISSN 1746-0441 827All rights reserved: reproduction in whole or in part not permitted

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y R

MIT

Uni

vers

ity o

n 09

/03/

13Fo

r pe

rson

al u

se o

nly.

Because of its role in snRNP assembly in all cells, homozy-gous mutations or deletions in the SMN gene are lethal inother species besides humans, who benefit from a secondnearly identical gene resulting from a duplication in chromo-some region 5q13 where the SMN gene is located. A singlecopy of the centromeric SMN2 gene prevents embryoniclethality that would ordinarily result from homozygous lossof the telomeric SMN1 gene in SMA patients. Indeed, SMAdisease severity decreases with increasing SMN protein, whichin turn is related to SMN2 copy number [5-8]. Even so, up tofour copies of SMN2 cannot fully prevent SMA becauseSMN levels remain suboptimal. This is due to nucleotidechanges that produce differential splicing of exon 7, which isabsent in the majority of SMN2 transcripts [9]. The resultingprotein referred to as SMND7 has an altered C-terminal:16 amino acids coded by exon 7 are replaced by fouramino acids derived from exon 8. SMND7 levels remain lowbecause it is rapidly degraded by the ubiquitin-proteosomepathway, at least partially related to decreased capacity tooligomerize compared to full length SMN (FL-SMN) orbecause the new C-terminal creates a degradation signal [10,11].The combined understanding of the genetic basis ofSMA as well as the structure and function of SMN proteinhas led to various strategies to discover and developSMA therapeutics.Because SMA is due to SMN deficiency, increasing SMN

levels by various means is considered a promising approachto treat SMA. This includes gene and cell therapy as well assmall molecule pharmacotherapy. Besides increasing SMNprotein, compensating for the functional deficits due toSMN deficiency is another potential approach. Ultimately,like most complex diseases, combination therapeutics may

be required to fully restore SMN protein levels, provideneuroprotection and treat symptoms as well as disease mech-anisms. Here, we consider only small molecules including oli-gonucleotides that have been discovered for SMA, howthey were identified and discuss potential future trends fordiscovering and developing SMA therapeutics.

2. Strategies for SMA drug discovery:increasing SMN

Three major mechanisms to produce more FL-SMN fromthe SMN2 gene have been targeted: i) increasing exon 7inclusion in SMN2 transcripts, ii) increasing transcriptionfrom the SMN2 promoter and iii) stabilizing SMN or theSMND7 protein.

For all these strategies, small molecule screening campaignscan be said to have been successful. While their clinical valida-tion still remains to be demonstrated, these compounds serveas chemical genetic tools that are helping to understand theSMA disease process and identify further therapeutic strate-gies. Knowing the full potential of any one strategy will prob-ably guide development of combination therapies in order toachieve maximal clinical efficacy.

Table 1 lists a number of small molecules that affectone or more of these steps. One of the first approachesused to increase SMN from SMN2 was splicing modulation.Hydroxyurea and sodium butyrate were two of the first com-pounds to be identified using SMA patient cell lines to assayfor increased exon 7 inclusion in SMN2 transcripts; bothwere found to increase SMN protein as well as amelioratedisease phenotype in SMA mouse models [12-14]. Because oftheir structural similarity to butyrate (and because theywere already used clinically to treat children), phenylbutyrateand valproic acid were tested and found active in similarassays both increasing SMN2 expression and exon7 inclusion [15-17]. The ability of butyrate and valproic acidto inhibit HDAC was thought to lead to increased expres-sion of certain splicing and transcription factors that regulatethe expression of SMN. Potent and specific HDAC inhibi-tors are currently approved or under development to treatvarious types of cancer and a number of them have beentested and found to increase SMN levels in cells and mousemodels of SMA (Table 1). However, their broad epigeneticmodulatory activity makes them unlikely candidates to treata chronic, pediatric-onset disease such as SMA. Whethersufficiently selective and potent HDAC inhibitors can befound to eliminate these concerns remains to be determined.In addition, their mechanism of action needs further study.The splicing factor Sfrs10 (also known as Htra2-b1) hasbeen implicated in SMN2 exon 7 inclusion and is increasedby HDAC inhibitors [15,18]. However, a recent study foundthat Sfrs10 deletion, although lethal, had little effect onSMN2 splicing and that motor neuron-specific deletionhad no phenotypic consequences [19]. Besides calling intoquestion the validity of in vitro assay systems, these results

Article highlights.

. Screening strategies targeting mechanisms implicated inspinal muscular atrophy (SMA) have been successful;although not yet clinically validated, they are providinguseful tools to investigate SMA disease mechanisms andidentify new targets and strategies.

. Survival motor neuron (SMN) has cell-specific functionsbesides splicing; understanding these functions and theconsequence of SMN deficits will provide new targetsfor SMA drug discovery.

. SMA is not a single disease and so therapeutic strategiesand options will need to be designed for SMA subtypesbased on age of onset, disease severity and prognosis.

. Biomarkers related to pharmacology and diseaseprogression are needed; they should provide surrogateefficacy end points.

. Products targeting different mechanisms will benecessary and guidelines are required in order to decidewhen to start treatment and how to evaluatecombinations of investigational drugs for SMA.

This box summarizes key points contained in the article.

Developments in the discovery of drugs for spinal muscular atrophy: successful beginnings and future prospects

828 Expert Opin. Drug Discov. (2011) 6(8)

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y R

MIT

Uni

vers

ity o

n 09

/03/

13Fo

r pe

rson

al u

se o

nly.

have implications on the development of potential pharma-codynamic biomarkers based on splicing factor levels as sur-rogate markers for compounds that favor exon 7 inclusion.Further understanding of the complexity of the splicingmechanism involved in SMN2 exon 7 inclusion includingthe role of post-translational modification of splicing factorscould provide further insight into potential strategies andtargets [20,21].

Other screening assays specifically targeting each of thepossible routes for increasing SMN have used reporter genesor cell-free assays to detect SMN2 splicing modulators,SMN2 expression enhancers, SMN or SMND7 stabilizersand SMN-dependent snRNP assembly modulators (Table 2).Because SMN levels actually affect SMN2 splicing, com-pounds having even a modest direct effect on SMNlevels could have a larger impact on SMN expression overtime [22].

2.1 Methods targeting increased SMN2 exon

7 inclusionThe complexity of SMN2 splicing and various approaches todiscover SMA therapies targeting exon 7 inclusion (alongwith other SMA drug discovery strategies) has recently beenreviewed [23]. Besides HDAC inhibitors, assays designedspecifically to screen for enhancers of SMN2 exon 7 inclusion

have identified two additional classes of compounds,tetracycline derivatives and antisense oligonucleotides.

2.1.1 TetracyclinesAclarubicin, an anthracycline chemotherapeutic, was discov-ered as a splicing modulator in a cell-based screen usingSMA patient fibroblasts and confirmed in a neuronal cellline stably expressing a mini-gene reporter for exon 7 inclu-sion [24]. Although other similar compounds such as doxoru-bicin and or tetracyclines were not active in these whole cellassays, a cell free splicing assay found that structurally similartetracyclines promoted exon 7 inclusion (although aclarubicinwas not active in this assay) [25]. Paratek is now developing oneof the compounds active in this cell-free assay, PTK-SMA1,which is selective for SMN2 exon 7 inclusion at low concen-trations although it affects splicing of other transcripts athigher concentrations. This means dose finding studies willbe needed to optimize the therapeutic effects while avoidingpotential side effects when this class of compounds enters clin-ical development. Biomarkers related to the pharmacologicaleffect of the compounds could be developed (e.g.,SMN2 transcripts versus other gene transcripts) to serve thispurpose. Ideally, they would be able to be measured in bloodcells or serum taken from healthy subjects as well as SMApatients. How tetracyclines modulate SMN2 transcript

Table 1. Drugs and drug candidates that increase SMN production from the SMN2 gene.

Drug Mechanism of action Ref.

Expression Splicing Stability

Sodium butyrate + [12]

Vanadate + [83]

Valproic acid + + [15]

Phenylbutyrate + + [16]

Hydroxyurea + + [13,14]

HDAC inhibitorsVorinostat (SAHA)Trichostatin AM344Romidepsin (FK-228)LBH589

+ + [84,85]

[86]

[87]

[88]

[89]

Tetracyclines +AclarubicinPTK-SMA1

[24]

[25]

Oligonucleotides + [26]

PRO105ISIS-SMNRx

[27]

[28]

Salbutamol + [90]

AminoglycosidesTobramycinTC007

+ [40]

[91]

[42,43]

QuinazolinesD156844, D157495 or RG3039

+ [31,33-38]

Indoprofen and analogsALB-X

+ [44,92,93]

[45,94,95]

SMN: Survival motor neuron.

Pruss

Expert Opin. Drug Discov. (2011) 6(8) 829

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y R

MIT

Uni

vers

ity o

n 09

/03/

13Fo

r pe

rson

al u

se o

nly.

splicing is not actually known, although their activity in a cell-free assay indicates that they interact with some component ofthe splicing reaction, the RNA or protein splicing factors.Further understanding of the mechanism could reveal targetsor chemical strategies to improve this approach.

2.1.2 OligonucleotidesOligonucleotides that were designed to hybridize to a spliceo-some recognition site in intron 7 were found to efficientlyblock SMN2 exon 7 excision in vitro [26]. This highly selec-tive approach has been further developed and found to beeffective in mouse models of SMA [27,28]. Other RNA anti-sense approaches using viral vector delivery have beenreviewed recently [23]. Small synthetic oligonucleotides arecurrently being developed as SMA therapeutics by Prosensa(PRO105) [29] and ISIS in collaboration with Genzyme(ISIS-SMARx) [30]. The current products are in preclinicaldevelopment stages. Issues that are being addressed includebetter understanding of where, when, how often and howmuch to deliver, what side effects are possible (e.g., pro-inflammatory effects) and what delivery systems will beneeded to get oligonucleotides across the BBB.

2.2 Increasing SMN2 expressionIncreased SMN2 copy number is correlated with reduceddisease severity because some SMN2 transcripts give rise toFL-SMN. Therefore, increasing expression from a patient’sSMN2 genes is another possible strategy to increase SMNproduction. Two groups have reported the development of areporter gene assay driven by the SMN2 promoter [31,32] using

neuronal cell lines in order to mimic as close as possible thetranscriptional regulation present in motor neurons. One ofthese screening strategies has led to the discovery of a familyof C-5 substituted quinazolines that increase SMN expres-sion [31,33,34]. Chemical optimization of the hits comingfrom this screening campaign lead to the identification of apotential drug candidate, D156844, that is orally active,crosses the BBB and is active in a mouse model ofSMA [35,36]. Proteomic methods found that the compoundtargets and inhibits DcpS, an mRNA decapping enzyme [37].It is thought that this stabilizes SMN2 transcripts and therebyincreases SMN protein. However, it is not clear whether orhow DcpS inhibition selectively increases SMN (activity inthe original reporter gene assay suggests not) and if not,what side effects could result. This novel target and mecha-nism that were uncovered using a cell-based assay is a goodexample of biological serendipity: you may not get what youwant, but if you try, sometimes you get what you need!Whether the effects in SMA animal models are due to thismechanism or to an increase in SMN remain to be deter-mined (however, this is the case for other compounds thatare active in SMA animal models). This drug discovery pro-gram was financed by a patient organization, Familiesof SMA, and was recently licensed to Repligen; the leadmolecule, RG3039 (or D157495), is in the preclinical stageof development [38].

2.3 Promoting SMN stabilityStabilizing either FL-SMN or SMND7 is expected to increaseprotein levels. SMN occurs in oligomers and oligomerization

Table 2. Screening approaches used to identify small molecule SMA therapeutics.

Assay type Compounds identified Ref.

Phenotypic screening on whole cells measuring SMN protein or FL-SMN transcriptsSMA patient lymphoblasts Hydroxyurea, sodium butyrate [12,13]

SMA patient fibroblasts Valproic acid [15]

Phenylbutyrate [16]

Aminoglycosides [40]

Forskolin [39]

Reporter gene assaysMinigene SMN2-exon 6 -- 8-luciferase Vanadate

Indoprofen analogsPolyphenols: curcumin, resveratrol

[83]

[44,92,93]

[96]

Minigene SMN2-exon 6 -- 8-b lactamase Aclarubicin [24]

SMN2 promoter-b lactamase Quinazolines [31]

SMN2 promoter-secreted alkaline phosphatase Taxol [32]

Cell-free assaysCell free splicing assay Tetracyclines [25]

SMN-dependent snRNP assembly Antioxidants [46-48]

Pluripotent stem cellsSMA patient-derived iPSCs Valproic acid

Tobramycin[57]

Phenotypic screen for neuroprotectionTrophic factor-deprived primary motor neurons Cholesterol oximes: olesoxime [75,76]

FL-SMN: Full length SMN; iPSC: Induced pluripotent stem cell; SMA: Spinal muscular atrophy; SMN: Survival motor neuron; snRNP: Small nuclear

ribonuclear protein.

Developments in the discovery of drugs for spinal muscular atrophy: successful beginnings and future prospects

830 Expert Opin. Drug Discov. (2011) 6(8)

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y R

MIT

Uni

vers

ity o

n 09

/03/

13Fo

r pe

rson

al u

se o

nly.

stabilizes both SMN and SMND7. SMND7 has a shorter half-life than SMN [10] and this was shown to be associated with areduced ability to oligomerize and form complexes comparedto FL-SMN [39]. Oligomerization appears to protect SMNand SMND7 from degradation by the ubiquitin-proteosomepathway since proteosome inhibitors slowed their degrada-tion [39]. Besides a general impact on conformation affectingoligomerization, the new C-terminal of SMND7 appearsto create a ‘degron’ or degradation signal that acceleratesproteosome-mediated turnover [11]. SMN oligomerizationand stability can be increased by cAMP and protein kinase Aactivation suggesting that pharmacological approaches couldbe used to increase SMN levels by this mechanism [39].

While SMND7 is unstable, replacing the C-terminal tail byother sequences or extending it by suppressing a terminationcodon in exon 8 with aminoglycoside antibiotics producesstable, correctly targeted and functional protein [40,41]. Subcu-taneous delivery of a novel derivative of this class of antibiot-ics, TC007 [42], improved the size of muscle fibers and motorfunction in an SMA mouse model although neither this newermolecule nor G418 improved survival of these mice [41,43].Poor BBB penetration may be the reason for poor efficacyof these compounds. PTC Therapeutics, which has beendeveloping stop codon suppressing compounds for musculardystrophies and other indications, has also reported workingon compounds of this type for SMA.

Indoprofen analogs also appear to increase SMN stability.Although originally discovered in a high-throughput screenusing the minigene reporter assay designed to identifySMN2 splicing modulators (Table 2), indoprofen did not affectSMN2 transcript profile in SMA patient fibroblasts but didincrease SMN levels [44]. The effects of indoprofen were notfound with other COX inhibitors. The Spinal Muscular Atro-phy Project, financed by the NIH, has been developing opti-mized indoprofen analogs to eliminate effects on COX,increase potency and allow passage across the BBB. The mech-anism of action of these compounds appears to be, likeaminoglycosides, due to stop codon readthrough [45].

3. Compensating for functional consequencesof SMN deficits

3.1 SMN, snRNPs and spliceosome functionBecause snRNP assembly and spliceosome activity areimpaired by SMN deficits, assays focused on the discoveryof small molecules that enhance snRNP assembly could be avalid and interesting drug discovery strategy. Indeed, novelhigh-throughput screens have been developed that specificallymeasure snRNP competent SMN in cell extracts [46-48].Although these assays have so far only identified inhibitors,they served to discover that oxidative stress severely impairsSMN-mediated snRNP assembly. Following up on this obser-vation, a study in SMA patients treated for up to 24 weekswith various antioxidant formulations found a progressiveincrease in snRNP assembly competence in their cells [48].

3.2 Other functions of SMNAlthough SMN deficits impair snRNP assembly andspliceosome activity, this affects all cells so it remains to bedetermined why motor neuron degeneration is the major con-sequence in SMA. Intellect and other neurological functionsare spared as are most other tissues and organs (with the pos-sible exception of cardiac tissue; see Section 5.1 below). Otherpossible roles of SMN, particularly in neurons, are nowemerging. There is mounting evidence that SMN participatesin other ribonuclear protein complexes and in particular ispresent in axon granules that transport RNA cargo to nerveterminals [49-52]. Such neuron-specific functions may explainwhy motor neurons with axons extending from the spinalcord to proximal muscles are particularly vulnerable toSMN deficiency compared to other cells.

4. Other strategies besides increasing SMN

Other genes besides SMN2 are able to modify SMA diseaseseverity. Both clinical genetics and studies in various SMAmodelsystems have identified candidate genes and pathways. Geneticanalysis of families with a child affected by SMA found thatsome of their unaffected siblings had the same SMN1 deletions.Further investigation led to the discovery that Plastin 3 overex-pression can prevent SMA disease onset [53,54]. Plastin 3 is anactin-regulating protein implicated in axonogenesis and canincrease neurite outgrowth in cultured motor neurons fromSMAmice or in a zebrafishmodel of SMA [54]. Cytoskeletal per-turbations have been found in SMA model systems. Knockingdown SMN expression in PC12 cells activates RhoA, a regulatorof actin dynamics that can cause neuronal growth cone collapse.Treating SMA mice with Y-27632 which inhibits ROCK, adownstream effecter of RhoA, improved maturation ofneuromuscular junctions (NMJs), increased muscle fiber sizeand prolonged survival [55]. Similar knockdown studies inNSC34 neuronal cells discovered that stathmin, a microtubuledestabilizing protein, is increased. Stathmin increases were thenfound to be present in SMA mice as well as perturbations inmicrotubule structure and dynamics in their motor neurons [56].Perturbations in microtubule-mediated organelle transport andneurofilament organization are also noted in SMA models (seebelow). These results suggest that treatments affecting cytoskele-tal dynamics may be therapeutically useful in SMA even if theydo not change SMN protein levels. Recent use of induced plu-ripotent stem cells derived from SMA patient show that theymay be useful for developing newmodels to explore the functionof SMN protein and disease mechanisms [57].

5. Is SMA a single disease? Unravelingdisease mechanism and alternative targets

5.1 What leads to neuromuscular pathology and

motor neuron cell death?Motor neuron cell death occurs naturally during developmentand is programmed in relation to muscle innervation. Motor

Pruss

Expert Opin. Drug Discov. (2011) 6(8) 831

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y R

MIT

Uni

vers

ity o

n 09

/03/

13Fo

r pe

rson

al u

se o

nly.

neuron survival depends on the extent of branching and syn-apse formation that facilitates interaction with the basal lam-ina and uptake of trophic factors supplied by muscle andSchwann cells. Connections between motor nerve presynapticterminals and the basal lamina facilitate access to trophic fac-tors and appear to be mediated by the interaction betweenlaminin subunits and synaptic vesicle proteoglycans exposedduring neurotransmitter release [58]. This implies that theamount of synaptic vesicle fusion occurring with motor neu-ron firing plays an important role in neuromuscular junction(NMJ) formation and stabilization. NMJ abnormalities arefound in most if not all SMA mouse models [59-65]. Theseinclude disorganized neurofilaments and synaptic vesicles inpresynaptic nerve terminals and immature postsynaptic struc-tures. These abnormalities are associated with decreased neu-romuscular transmission that is attributed to decreasedprobability of synaptic vesicle fusion and reduced quantalrelease [62,63,65]. Two recent descriptions of rescue or recoveryof SMA disease phenotype in mice suggest that early deficitsin synaptic vesicle release are reversible. A single intravenousadministration of a viral vector delivering the SMN gene atthe time of birth rescues severely affected SMA mice and theirrecovery is associated with normalized motor endplate currentindicative of increased quantal release [63]. Similarly, a recentlydescribed SMA mouse model in which SMN deficiency is tar-geted to motor neurons displays neuromuscular weaknessduring the first week of life but recovers during the secondweek so that ~ 70% live > 1 year [65]. Recovery was associatedwith normalization of quantal release at motor endplates.Even though these mice recover and survive, there is evidenceof simultaneous atrophy and hypertrophy in their skeletalmuscle fibers showing SMN depletion in motor neuronsalone is sufficient to generate muscle abnormalities. Neverthe-less, SMN may be important in other tissues as well: cardiacabnormalities have been noted both in severely affectedSMA patients [66] and SMA mice [67].

5.2 SMA: a spectrum of disease severity requires a

variety of therapeutic optionsDisease severity and progression in SMA patients areextremely variable and classified by type according to func-tional milestones the individual can achieve. Type 1 patientscan never sit independently, type 2 patients can sit but neverwalk independently and type 3 patients develop the abilityto walk but eventually lose this capability. These classifica-tions, as cited previously, are correlated with SMN proteinlevel and SMN2 copy number, which in turn correlates withthe rate of disease progression and the rate and extent ofdenervation as shown in a natural history study [8]. Remark-ably, in a few type 1 and type 2 SMA patients identified byprenatal testing who could be studied from birth, denervationas measured by the decrease in compound muscle actionpotential (CMAP) amplitude occurred very rapidly startingfrom nearly normal levels at birth [8]. Denervation intype 3 patients on the other hand was less marked and

CMAP was rather stable with time. Muscle fibers in biopsiestaken from SMA patients or normal controls have also foundmarked differences between type 1 and type 3 SMA patients.While muscle transcripts from type 3 patients were closerto those in normal muscle, transcripts from type 1 patientswere drastically different. There was evidence for simulta-neous atrophy and hypertrophy in type 3 SMA muscle fibersassociated with various signaling pathways that were notpresent in type 1 SMA fibers [68].

Because of the difference in disease severity and progres-sion, SMA may not be a single disease and therapies need tobe designed and developed for various subpopulations. Exceptfor the mouse model in which SMN depletion is targeted tomotor neurons and the type III SMA mice generated byback-crossing founders onto a C57Bl/6 background [69],

most SMA mouse models have a severe phenotype andare considered models of type 1 SMA. These models as wellas clinical evidence that denervation occurs soon after birthin severe forms of SMA suggest that the window of oppor-tunity for treatment may be very narrow in type 1 SMApatients [8,63,70,71]. Introduction of newborn screening forSMA will be essential to initiate treatment during thiscritical period.

In less severe SMA models, mice have a nearly normal lifespan and so far do not appear to offer similar end points toassess disease progression as those in type 2 and type 3 SMApatients. This makes identification and validation of therapiesfor less severely affected SMA patients, for whom preservingexisting neuromuscular function rather than reversing diseasewill be the objective, more challenging both conceptually andpractically. A porcine model of SMA is currently in develop-ment with the hope that it may be closer (both in size and dis-ease phenotype) to human SMA patients [72,73] and provide ameans to test therapies for mild as well as severe forms ofSMA. Because no suitable animal model exists to test thera-pies for type 2 or type 3 SMA patients, there are severalopen questions about whether treatments that increase SMNwill be effective in these patients, when the treatment(s)should be administered and how their effects will be mea-sured. New ethical issues regarding how and when to treatchildren who may have milder forms of SMA will also needto be addressed. In the future, should newborn screening beavailable, will it be possible to accurately predict the time ofdisease onset? SMN2 copy number is not sufficiently preciseeither to detect all type 1 patients who should be treatedimmediately or to predict the time of disease onset or severityif three or more copies of SMN2 are present. Will it be ethicalto initiate treatment before symptom onset? And if not, will itthen be too late?

6. Neuroprotection, alone or in combinationwith SMN enhancers

Besides increasing SMN, neuroprotection might havetherapeutic potential. Indeed the two approaches may be

Developments in the discovery of drugs for spinal muscular atrophy: successful beginnings and future prospects

832 Expert Opin. Drug Discov. (2011) 6(8)

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y R

MIT

Uni

vers

ity o

n 09

/03/

13Fo

r pe

rson

al u

se o

nly.

complementary. This approach was recently tested bycombining a nucleic acid SMN2 splicing modulator withIGF1 in a single viral vector and comparing the effects ofthis combination vector with those of individual vectors deliv-ering either a splicing enhancer or IGF1 in a severe SMAmouse model [74]. Interestingly, a significant effect on survivalwas produced by delivering IGF1 alone while the combinedvector was only slightly but not significantly more effective.Delivering the splicing enhancer alone had a small but insig-nificant effect although SMN protein expression wasenhanced. These results suggest that neuroprotection alone,which may have had little or no effect on SMN levels or splic-ing deficits resulting from SMN deficiency, could be a highlyeffective therapeutic option in SMA.

Neurotrophic factors, besides increasing neuronal sur-vival, enhance neurite outgrowth, which may improve neu-ronal cytoskeleton abnormalities resulting from SMNdeficits as mentioned previously (Plastin 3, RhoA, stath-min). Olesoxime (also called TRO19622) is a compoundcurrently being studied in clinical trials in type 2 andtype 3 SMA patients as well as in patients with ALS. Itsdiscovery and development for SMA was financed by theAssociation Francaise contres les Myopathies, a Frenchpatient organization. Olesoxime was discovered in a cell-based screen for compounds that rescue trophic factordeprived motor neurons; it favors both motor neuronsurvival and neurite outgrowth, has shown beneficial effectsin mouse models of ALS and SMA [75,76] and rescues othertypes of target deprived neurons [77]. The neuroprotectiveeffects of olesoxime are attributed to modulation of oxida-tive stress-induced mitochondrial permeability transitionthrough interaction with two outer mitochondrial mem-brane proteins, TSPO and VDAC. In addition, olesoximepromotes microtubule dynamics and preserves neuritesfrom toxic effects of microtubule-targeting chemotherapeu-tic drugs [78], which may explain its ability to promote neu-rite outgrowth. Despite having no effect on SMN levels,olesoxime appears to have a combination of actions thatcould be beneficial in SMA either alone or in the futurein combination with SMN enhancing therapies.

7. Conclusion

Since the discovery that SMA was due to mutations inSMN1 and that it could be possible to increase SMN produc-tion from a patient’s SMN2 gene, enormous effort and prog-ress have been made in identifying mechanisms, smallmolecules and gene therapies that could fulfill this objective.Screening models have been established that have identifiedsmall molecules with potential to be developed into drug can-didates focused on SMN production as well as neuroprotec-tion. New screening strategies are likely to evolve withfurther understanding of cell type specific functions ofSMN in neurons, skeletal muscle and cardiac tissue, all ofwhich may be affected in SMA. Assays based on SMA

patient-derived pluripotent stem cells may provide bettermodels to discover and evaluate new disease modifyingapproaches. iPierian has created a drug discovery screeningplatform based on patient-derived pluripotent stem cells andis tackling SMA therapeutics.

8. Expert opinion

The first clinical trials of SMN2 modulating therapies withhydroxyurea, valproic acid or sodium phenylbutyrate wereconducted because these drugs had already been approvedfor use in children; ongoing trials of additional compoundsor ones expected to start in the next few years were coveredin a recent review [32]. The trial results that have been reportedso far have not validated the SMN2 modulation strategy per-haps for methodological reasons (dose tested, trial duration,number of patients studied) although responder subpopula-tions could benefit from these treatments [79,80]. These andother trials have paved the way by establishing clinical trialmethodology showing that SMA disease progression is slowand variable and so trial duration will need to be long (twoor more years) to detect a disease modifying effect using anumber of motor function scales. Identifying and validatingbiomarkers both related to the product’s expected pharmacol-ogy (e.g., for the SMN approach, SMN protein, SMN2transcripts, snRNP assembly) should be used for dose selec-tion and to evaluate acute and long-term effects; they couldalso be used to screen and select possible responders for along-term clinical trial for efficacy. A clear understanding ofdisease progression, particularly in different patient subpopu-lations and disease-associated biomarkers, is also criticallyneeded. These could be more sensitive and less subjectivemeasures of disease progression and provide surrogate endpoints for efficacy earlier than an effect on motor functioncan be detected. Electromyography to assess CMAP andmotor unit number estimation has been shown to be feasiblein clinical trials even in young children and appears to becorrelated with disease severity and functional outcomemeasures; it appears to be promising as an early biomarkerindicative of efficacy and should be performed in SMA clini-cal trials to validate this hypothesis [8,81]. It will also be neces-sary to consider co-development of complementary therapies:combinations of products increasing SMN by differentmechanisms, combinations of neuroprotective therapies andproducts increasing SMN, or other strategies that mayemerge. Regulatory agencies are beginning to address how toevaluate combinations of investigational drugs in certain indi-cations [82]. The more we know about SMN and SMA diseasepathology, the more we understand that SMA is not such asimple monogenetic disease with only one therapeutic targetor strategy. The molecular tools developed so far are wideningour understanding and the scope of possibilities. Theseemerging novel ideas need to be explored. Because SMA is arare disease, preclinical and clinical research funding haslargely been provided by patient organizations and

Pruss

Expert Opin. Drug Discov. (2011) 6(8) 833

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y R

MIT

Uni

vers

ity o

n 09

/03/

13Fo

r pe

rson

al u

se o

nly.

government agencies. Priorities should include identificationof alternative SMN functions, neuroprotection, improvingneuronal cytoskeletal dynamics, reducing oxidative stress andother targets and mechanisms as they emerge from furtherunderstanding of disease mechanism. Identification and vali-dation of pharmacodynamic biomarkers for each therapeutic

strategy as well as SMA disease progression will alsobe indispensable.

Declaration of interest

RM Pruss is an employee of Trophos.

BibliographyPapers of special note have been highlighted as

either of interest (�) or of considerable interest(��) to readers.

1. Lefebvre S, Burglen L, Reboullet S, et al.

Identification and characterization of a

spinal muscular atrophy-determining

gene. Cell 1995;80:155-65

2. Liu Q, Dreyfuss G. A novel nuclear

structure containing the survival of motor

neurons protein. EMBO J

1996;15:3555-65

3. Liu Q, Fischer U, Wang F, Dreyfuss G.

The spinal muscular atrophy disease

gene product, SMN, and its associated

protein SIP1 are in a complex with

spliceosomal snRNP proteins. Cell

1997;90:1013-21

4. Fischer U, Liu Q, Dreyfuss G. The

SMN-SIP1 complex has an essential role

in spliceosomal snRNP biogenesis. Cell

1997;90:1023-9

5. Lefebvre S, Burlet P, Liu Q, et al.

Correlation between severity and SMN

protein level in spinal muscular atrophy.

Nat Genet 1997;16:265-9

6. Burghess AHM. When is a deletion not

a deletion? When it is converted. Am J

Hum Genet 2007;61:9-15

7. Mailman MD, Heinz JW, Papp AC,

et al. Molecular analysis of spinal

muscular atrophy and modification of

the phenotype by SMN2. Genet Med

2002;4:20-6

8. Swoboda KJ, Prior TW, Scott CB, et al.

Natural history of denervation in SMA:

relation to age, SMN2 copy number, and

function. Ann Neurol 2005;57:704-12

9. Lorson CL, Hahnen E, Androphy EJ,

Wirth B. A single nucleotide in the

SMN gene regulates splicing and is

responsible for spinal muscular atrophy.

Proc Natl Acad Sci USA

1999;96:6307-11

10. Lorson CL, Androphy EJ. An exonic

enhancer is required for inclusion of an

essential exon in the SMA-determining

gene SMN. Hum Mol Genet

2000;9:259-65

11. Cho S, Dreyfuss G. A degron created by

SMN2 exon 7 skipping is a principal

contributor to spinal muscular atrophy

severity. Genes Dev 2010;24:438-42

12. Chang JG, Hsieh-Li HM, Jong YJ, et al.

Treatment of spinal muscular atrophy by

sodium butyrate. Proc Natl Acad

Sci USA 2001;98:9808-13.. Proof of concept paper showing the

therapeutic potential of HDAC

inhibitors for the treatment of spinal

muscular atrophy.

13. China Medical College Hospital.

US6573300; 2003

14. Grzeschik SM, Ganta M, Prior TW,

et al. Hydroxyurea enhances SMN2 gene

expression in spinal muscular atrophy

cells. Ann Neurol 2005;58:194-202

15. Brichta L, Hofmann Y, Hahnen E, et al.

Valproic acid increases the SMN2

protein level: a well-known drug as a

potential therapy for spinal muscular

atrophy. Hum Mol Genet

2003;12:2481-9

16. Andreassi C, Angelozzi C, Tiziano FD,

et al. Phenylbutyrate increases SMN

expression in vitro: relevance for

treatment of spinal muscular atrophy.

Eur J Hum Genet 2004;12:59-65

17. Brahe C, Vitali T, Tiziano FD, et al.

Phenylbutyrate increases SMN gene

expression in spinal muscular atrophy

patients. Eur J Hum Genet

2005;13:256-9

18. Hofmann Y, Lorson CL, Stamm S, et al.

Htra2-beta 1 stimulates an exonic

splicing enhancer and can restore

full-length SMN expression to survival

motor neuron 2 (SMN2). Proc Natl

Acad Sci USA 2000;97:9618-23

19. Mende Y, Jakubik M, Reissland M, et al.

Deficiency of the splicing factor

Sfsr10 results in early embryonic in mice

and has no impact on full-length SMN/

Smn splicing. Hum Mol Gen

2010;19:2154-67

20. Novoyateleva T, Heinrich B, Tang Y,

Benderska N. Protein phosphatase

1 binds to the RNA recognition motif of

several splicing factors and regulates

alternative pre-mRNA processing.

Hum Mol Genet 2008;17:52-70

21. Martins de Araujo M, Bonnal S,

Hastings ML, et al. Differential 3’ splice

site recognition of SMN1 and

SMN2 transcripts by U2AF and

U2 snRNP. RNA 2009;15:515-23

22. Jodelka FM, Ebert AD, Duelli DM,

Hastings ML. A feedback loop

regulates splicing of the spinal

muscular atrophy-modifying gene,

SMN2. Hum Mol Genet

2010;19:4906-17

23. Lorson CL, Rindt H, Shababi M. Spinal

muscular atrophy: mechanisms and

therapeutic strategies. Hum Mol Genet

2010;19:R111-18

24. Andreassi C, Jarecki J, Zhou J, et al.

Aclarubicin treatment restores SMN

levels to cells derived from type I spinal

muscular atrophy patients.

Hum Mol Genet 2001;10:2841-9

25. Hastings ML, Berniac J, Liu YH, et al.

Tetracyclines that promote SMN2 exon

7 splicing as therapeutics for spinal

muscular atrophy. Sci Transl Med

2009;1:5ra12

26. Singh NK, Singh NN, Androphy EJ,

Singh RN. Splicing of a critical exon of

human Survival Motor Neuron is

regulated by a unique silencer element

located in the last intron. Mol Cell Biol

2006;26:1333-46

27. Williams JH, Schray RC, Patterson CA,

et al. Oligonucleotide-mediated survival

of motor neuron protein expression in

CNS improves phenotype in a mouse

model of spinal muscular atrophy.

J Neurosci 2009;29:7633-8

28. Hua Y, Sahashi K, Hung G, et al.

Antisense correction of SMN2 splicing in

the CNS rescues necrosis in a type III

SMA mouse model. Genes Dev

2010;24:1634-44

29. Available from: http://www.prosensa.eu/

technology-and-products/pre-clinical-

portfolio

Developments in the discovery of drugs for spinal muscular atrophy: successful beginnings and future prospects

834 Expert Opin. Drug Discov. (2011) 6(8)

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y R

MIT

Uni

vers

ity o

n 09

/03/

13Fo

r pe

rson

al u

se o

nly.

30. Available from: http://www.isispharm.

com/Pipeline/Therapeutic-Areas/

Neurodegenerative-Disease.htm#

ISIS-SMNRx

31. Jarecki J, Chen X, Bernardino A, et al.

Diverse small-molecule modulators of

SMN expression found by

high-throughput compound screening:

early leads towards a therapeutic for

spinal muscular atrophy.

Hum Mol Genet 2005;14:2003-18. The first report of an HTS to identify

new modulators of

SMN2 gene expression.

32. Pruss RM, Giraudon-Paoli M,

Morozova S, et al. Drug discovery and

development for spinal muscular atrophy:

lessons from screening approaches and

future challenes for clinical development

Future Med Chem 2010;2:1429-40

33. Vertex Pharmaceuticals, Inc. US7465738;

2008

34. Vertex Pharmaceuticals, Inc. WO2004/

112205A2

35. Thurmond J, Butchbach MER,

Palomo M, et al. Synthesis and

biological evaluation of novel

2,4-diaminoquinazoline derivatives as

SMN2 promoter activators for the

potential treatment of spinal muscular

atrophy. J Med Chem 2008;51:449-69

36. Butchbach ME, Singh J,

Thorsteinsdottir M, et al. Effects of

2,4-diaminoquinazoline derivatives on

SMN expression and phenotype in a

mouse model for spinal muscular

atrophy. Hum Mol Genet

2010;19:454-67

37. Singh J, Salcius M, Liu SW, et al.

DcpS as a therapeutic target for spinal

muscular atrophy. ACS Chem Biol

2008;3:711-22

38. http://www.repligen.com/products/

pipeline/rgsma

39. Burnett BG, Munoz E, Tandou A, et al.

Regulation of SMN protein stability.

Mol Cell Biol 2009;29:1107-15

40. Wolstencroft EC, Mattis V, Bajer AA,

et al. A non-sequence-specific

requirement for SMN protein activity:

the role of aminoglycosides in inducing

elevated SMN protein levels.

Hum Mol Genet 2005;14:1199-210.. This work showed that the SMN exon

7 sequence functions as a nonspecific

‘tail’ that facilitates proper localization

providing the rationale for

read-through strategies to restore

SMND7 protein activity.

41. Heier CR, DiDonato CJ. Translational

readthrough by the aminoglycoside

geneticin (G418) modulates SMN

stability in vitro and improves motor

function in SMA mice in vivo.

Hum Mol Genet 2009;18:1310-22

42. Chang CW, Hui Y, Elchert B, et al.

Pyranmycins, a novel class of

aminoglycosides with improved acid

stability: the SAR of D-pyranoses on

ring III of pyranmycin. Org Lett

2002;4:4603-6

43. Mattis VB, Ebert AD, Fosso MY, et al.

Delivery of a read-through inducing

compound, TC007, lessens the severity

of a spinal muscular atrophy animal

model. Hum Mol Genet

2009;18:3906-13

44. Lunn MR, Root DE, Martino AM, et al.

Indoprofen upregulated the survival

motor neuron protein through a

cyclooxygenase-independent mechanism.

Chem Biol 2004;11:1489-93

45. The Spinal Muscular Atrophy Project

http://asent.org/22308B47-E7FF-0F41-

24C00CE1DE6EC4AC

46. The trustees of the University of

Pennsylvania US2006/0223092A1

47. Wan L, Ottinger E, Cho S, Dreyfuss G.

Inactivation of the SMN complex by

oxidative stress. Mol Cell

2008;31:244-54

48. Dreyfuss G, Wan L US2010/0234402A1

49. Zhang HL, Pan F, Hong D, et al. Active

transport of the survival motor neuron

protein and the role of exon-7 in

cytoplasmic localization. J Neurosci

2003;23:6627-37

50. Rossoll W, Jablonka S, Andreassi C,

et al. Smn, the spinal muscular

atrophy-determining gene product,

modulates axon growth and localization

of beta-actin mRNA in growth cones of

motoneurons. J Cell Biol

2003;163:801-12

51. Rossoll W, Bassell GJ. Spinal muscular

atrophy and a model for survival of

motor neuron protein function in axonal

ribonucleoprotein complexes.

Results Probl Cell Differ

2009;48:289-326

52. Fallini C, Zhang H, Su Y, et al. The

survival of motor neuron (SMN) protein

interacts with the mRNA-binding protein

HuD and regulates localization of poly

(A) mRNA in primary motor neuron

axons. J Neurosci 2011;31:3914-25

53. Hahnen E, Forkert R, Marke C, et al.

Molecular analysis of candidate genes on

chromosome 5q13 in autosomal recessive

spinal muscular atrophy: evidence of

homozygous deletions of the SMN gene

in unaffected individuals.

Hum Mol Genet 1995;4:1927-33

54. Oprea GE, Krober S, McWhorter ML,

et al. Plastin 3 is a protective modifier of

autosomal recessive spinal muscular

atrophy. Science 2008;320:524-7.. Identification of Plastin 3 as a disease

modifier suggesting other approaches

than increasing SMN may be useful to

treat SMA.

55. Bowerman M, Beauvais A, Anderson CL,

Kothary R. Rho-kinase inactivation

prolongs survival of an intermediate

SMA mouse model. Hum Mol Genet

2010;19:1468-78

56. Wen HL, Lin YT, Ting CH, et al.

Stathmin, a microtubule-destabilizing

protein, is dysregulated in spinal

muscular atrophy. Hum Mol Genet

2010;19:1766-78

57. Ebert AD, Yu J, Rose FF, et al. Induced

pluripotent stem cells from a spinal

muscular atrophy patient. Nature

2009;457:277-80

58. Banks G, Noakes PG. Elucidating the

molecular mechanisms that underlie the

target control of motoneuron death. Int J

Dev Biol 2002;46:551-8

59. Le TT, Pham LT, Butchbach MER,

et al. SMNDelta7, the major product of

the centromeric survival motor neuron

(SMN2) gene, extends survival in mice

with spinal muscular atrophy and

associates with full-length SMN.

Hum Mol Genet 2005;14:845-57

60. Kariya S, Park GH, Maeno-Hikichi Y,

et al. Reduced SMN protein impairs

maturation of the neuromuscular

junctions in mouse models of spinal

muscular atrophy. Hum Mol Genet

2008;17:2552-69

61. Murray LM, Comley LH, Thomson D,

et al. Selective vulnerability of motor

neurons and dissociation of pre- and

post-synaptic pathology at the

neuromuscular junction in mouse models

of spinal muscular atrophy.

Hum Mol Genet 2008;17:949-62

62. Kong L, Wang X, Choe DW, et al.

Impaired synaptic vesicle release and

Pruss

Expert Opin. Drug Discov. (2011) 6(8) 835

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y R

MIT

Uni

vers

ity o

n 09

/03/

13Fo

r pe

rson

al u

se o

nly.

immaturity of neuromuscular junctions

in spinal muscular atrophy mice.

J Neurosci 2009;29:842-51

63. Foust KD, Wang X, McGovern VL,

et al. Rescue of the spinal muscular

atrophy phenotype in a mouse model by

early postnatal delivery of SMN.

Nat Biotechnol 2010;28:271-4.. A study showing a single intravenous

administration of a viral vector to

deliver SMN can rescue severely

affected SMA mice but that the

therapeutic window of oportunity

is narrow.

64. Michaud M, Arnoux T, Bielli S, et al.

Neuromuscular defects and breathing

disorders in a new mouse model of

spinal muscular atrophy. Neurobiol Dis

2010;38:125-35

65. Park G-H, Maeno-Hikichi Y, Awano T,

et al. Reduced suvival of motor neuron

(SMN) protein in motor neuronal

progenitors functions cell autonomously

to cause spinal muscular atrophy in

model mice expressing the human

centromeric (SMN2) gene. J Neurosci

2010;30:12005-19

66. Rudnik-Schoneborn S, Heller R, Berg C,

et al. Congenital heart disease is a

feature of severe infantile spinal

muscular atrophy. J Med Genet

2008;45:635-8

67. Shababi M, Habibi J, Yang HT, et al.

Cardiac defects contribute to the

pathology of spinal muscular atrophy

models. Hum Mol Genet

2010;19:4059-71

68. Millino C, Fanin M, Vettori A, et al.

Different atrophy-hypertrophy

transcription pathways in muscles

affected by severe and mild spinal

muscular atrophy BMC Medicine

2009;7:14-22

69. Tsai LK, Tsai MS, Ting CH, et al.

Restoring Bcl-x(L) levels benefits a mouse

model of spinal muscular atrophy.

Neurobiol Dis 2008;31:361-7

70. Passini MA, Bu J, Roskelley EM, et al.

CNS-targeted gene therapy improves

survival and motor function in

a mouse model of spinal muscular

atrophy. J Clin Invest

2010;120:1253-64

71. Valori CF, Ning K, Wyles M, et al.

Systemic delivery of scAAV9 expressing

SMN prolongs survival in a model of

spinal muscular atrophy. Sci Transl Med

2010;2:35ra42

72. Lorson MA, Spate LD, Prather RS,

Lorson CL. Identification and

characterization of the porcine (Sus

scrofa) survival motor neuron (SMN1)

gene: an animal model for therapeutic

studies. Dev Dyn 2008;237:2268-78

73. Lorson MA, Spate LD, Samuel MS,

et al. Disruption of the Survival Motor

Neuron (SMN) gene in pigs using

ssDNA. Transgenic Res 2011.

[Epub ahead of print]

74. Shababi M, Glascock J, Lorson CL.

Combination of SMN trans-splicing and

a neurotrophic factor increases the life

span and body mass in a severe

model of spinal muscular atrophy.

Hum Gene Ther 2011;22:135-44

75. Bordet T, Buisson B, Michaud M, et al.

Identification and characterization of

cholest-4-en-3-one, oxime (TRO19622),

a novel drug candidate for amyotrophic

lateral sclerosis. J Pharmacol Exp Ther

2007;322:709-20

76. Bordet T, Berna P, Abitbol JL,

Pruss RM Olesoxime (TRO19622):

A novel mitochondrial-targeted

neuroprotective compound.

Pharmaceuticals 2010;3:345-68

77. Martin LJ, Adams NA, Pan Y, et al.

The mitochondrial permeability

transition pore regulates nitric

oxide-mediated apoptosis of neurons

induced by target deprivation. J Neurosci

2011;31:359-70

78. Rovini A, Carre M, Bordet T, et al.

Olesoxime prevents microtubule-targeting

drug neurotoxicity: selective preservation

of EB comets in differentiated neuronal

cells. Biochem Pharmacol

2010;80:884-94

79. Mercuri E, Bertini E, Messina S, et al.

Randomized, double-blind,

placebo-controlled trial of phenylbutyrate

in spinal muscular atrophy. Neurology

2007;68:51-5. The first well-powered (107 patients

included) Phase II clinical evaluation

of the efficacy of an HDAC inhibitor

in SMA type 2 patients aged

30 -- 154 months.

80. Swoboda KJ, Scott CB, Reyna SP, et al.

Phase II open label study of valproic acid

in spinal muscular atrophy. PLoS One

2009;4:e5268

81. Lewelt A, Krosschell KJ, Scott C, et al.

Compound muscle action potential and

motor function in children with spinal

muscular atrophy. Muscle Nerve

2010;42:703-8

82. Guidance for Industry Codevelopment of

Two or More Unmarketed

Investigational Drugs for Use in

Combination. Available from: http://

www.fda.gov/downloads/Drugs/

GuidanceComplianceRegulatory

Information/Guidances/UCM236669.pdf

83. Zhang ML, Lorson CL, Androphy EJ,

Zhou J. An in vivo reporter system for

measuring increased inclusion of exon

7 in SMN2 mRNA: potential therapy of

SMA. Gene Ther 2001;8:1532-8

84. Hahnen E, Eyupoglu IY, Brichta L, et al.

In vitro and ex vivo evaluation of

second-generation histone deacetylase

inhibitors for the treatment of spinal

muscular atrophy. J Neurochem

2006;98:193-202

85. Riessland M, Ackermann B, Forster A,

et al. SAHA ameliorates the

SMA phenotype in two mouse models

for spinal muscular atrophy. Hum. Mol.

Genet 2010;19:1492-1506

86. Avila AM, Burnett BG, Taye AA, et al.

Trichostatin A increases SMN expression

and survival in a mouse model of spinal

muscular atrophy. J Clin Invest

2007;117:659-71

87. Riessland M, Brichta L, Hahnen E,

Wirth B. The benzamide M344, a novel

histone deacetylase inhibitor, significantly

increases SMN2 RNA/protein levels in

spinal muscular atrophy cells.

Hum Genet 2006;120:101-10

88. Hauke J, Riessland M, Lunde S, et al.

Survival motor neuron gene 2 silencing

by DNA methylation correlates with

spinal muscular atrophy disease severity

and can be bypassed by histone

deacetylase inhibition. Hum Mol Genet

2009;18:304-17

89. Garbes L, Riessland M, Holker I, et al.

LBH589 induces up to 10-fold SMN

protein levels by several independent

mechanisms and is effective even in cells

from SMA patients non-responsive to

valproate. Hum Mol Genet

2009;18:3645-58

90. Tiziano FC, Lomastro R, Pinto AM,

et al. Salbutamol increases survival motor

neuron (SMN) transcript levels in

leucocytes of spinal muscular atrophy

(SMA) patients: relevance for

clinical trial design. J Med Genet

2010;47:856-8

Developments in the discovery of drugs for spinal muscular atrophy: successful beginnings and future prospects

836 Expert Opin. Drug Discov. (2011) 6(8)

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y R

MIT

Uni

vers

ity o

n 09

/03/

13Fo

r pe

rson

al u

se o

nly.

91. Mattis VB, Rai R, Wang J, et al. Novel

aminoglycosides increase SMN levels in

spinal muscular atrophy fibroblasts.

Hum Genet 2006;120:589-601

92. Whitehead Institute for Biomedical

Research, Trustees of Columbia

University in the City of New York

WO2006/050451

93. Stockwell BR, Root DE WO2009/

0031435

94. US Department of Health and Human

Services, Albany Molecular Research, Saic

US2009/0312323A1

95. US Department of Health and Human

Services, Saic, Albany Molecular Research

US2010/0267712A1

96. Sakla MS, Lorson CL. Induction of

full-length survival motor neuron by

polyphenol botanical compounds.

Hum Genet 2008;122:635-43

AffiliationRebecca M. Pruss PhD

Chief Scientific Officer,

Trophos, SA Parc Scientifique de

Luminy Case 931,

13288 Marseille Cedex 9, France

Tel: +1 33 0 491 828281;

Fax: +1 33 0 491 828289;

E-mail: [email protected]

Pruss

Expert Opin. Drug Discov. (2011) 6(8) 837

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y R

MIT

Uni

vers

ity o

n 09

/03/

13Fo

r pe

rson

al u

se o

nly.