<<

Review

1 Central & Peripheral Nervous Systems Huntington’s disease: progress

5 and potential in the fi eld

Edward C Stack & Robert J Ferrante† 1. Background † Boston University School of Medicine , Departments of Neurology , Pathology, and Psychiatry, 2. Existing clinical Edith Nourse Rogers Veterans Administration Medical Center , Experimental Neuropathology Unit 10 management and need and Translational Therapeutics Laboratory , GRECC Unit, Building 18 , 3. Mechanisms of cell death and Bedford, MA 01730 , USA potential therapeutic targets in Huntington’s disease While the first description of Huntington’s disease was reported over a century ago, no therapy exists that can halt or ameliorate the inexorable 4. Therapeutic candidates 15 disease progression. Tremendous progress, however, has been made in 5. Conclusion significantly broadening the understanding of pathogenic mechanisms in 6. Expert opinion this neurological disorder that may eventually lead to successful treatment strategies. Huntington’s disease is caused by the expansion of a CAG repeat in the huntingtin gene, which results in the expression of a mutant form 20 of the that is toxic to . Several mechanisms have been iden- tified in mediating this toxicity, such as protein aggregation, mitochondrial dysfunction, oxidative stress, transcriptional dysregulation, aberrant apop- tosis, altered proteosomal function and . With increasing understanding of each of these pathogenic mechanisms, therapeutic 25 strategies have attempted to target specific aspects of each. There have been many encouraging reports of preclinical efficacy in transgenic Huntington’s disease mice, from which a number have been extended to human clinical trials with some success. This review focuses on these studies and the compounds that hold promise for treating human 30 Huntington’s disease.

Keywords: , excitotoxicity , mitochondria , oxidative stress , transcription , ubiquitin-proteosome system

35 Expert Opin. Investig. Drugs (2007) 16(12):1-21

1. Background

Huntington’s disease (HD) is an inherited autosomal dominant disorder of 40 the CNS. Roughly 250,000 Americans are affected by or are at risk of inheriting this fatal disease. Clinically, HD is characterized by cognitive and memory impairments, heightened irritability, depression, weight loss and choreic motor abnormalities [1] . Interestingly, subtle behavioral alterations occur prior to clinical diagnosis [2,3] , consistent with evidence of altered neuropathology before manifest 45 disease [4] . Once manifest disease occurs, however, the duration of HD is ∼ 15 – 20 years, with clinical symptoms becoming increasingly disabling prior to death [1] . Despite great progress, a direct causative pathway from the HD gene mutation to neuronal death has not yet been established. As there is no proven therapy to delay or slow the progression of HD, there is a growing demand on 50 improved clinical management for affected individuals and their families. While other brain regions are altered in HD, the most prominent neuro- pathological feature is marked gross atrophy of the neostriatum with concomitant neuronal degeneration within the caudate and putamen. There is a topographic progression of neuronal loss and astrogliosis first observed in the dorso-medial 55 aspect of the striatum and progressing ventro-laterally, with relative sparing of the

10.1517/13543784.16.12.1 © 2007 Informa UK Ltd ISSN 1354-3784 1 Huntington’s disease: progress and potential in the fi eld

1 ventral striatum [5] . Not all neurons are affected equally inhibitors sertraline and and/or benzodiazepines, within the neostriatum [6] . While large cholinergic neurons such as clonazepam, diazepam, risperidone or sulpiride [26] . and medium-sized NADPH-diaphorase aspiny neurons There is no significant evidence that any given antipsychotic remain relatively spared in HD, medium-sized spiny drug is better than another in the treatment of symptoms. 5 GABAergic projection neurons of the striatum, which make up ∼ 95% of striatal content, are disproportionately 3. Mechanisms of cell death and potential affected early and most severely [6-9] . There is also a reduc- therapeutic targets in Huntington’s disease tion in striatal neurochemicals that parallels striatal neuro- degeneration in HD [10,11] , with substance P-expressing striatal 3.1 Mutant Huntingtin aggregation (mHtt) 10 projection neurons appearing more vulnerable [12] . In While the gene responsible for HD was discovered almost addition, striatal medium spiny neurons show both 15 years ago [15] , the relationship between mHtt and the proliferative and degenerative changes leading to neuronal multiple molecular pathways that appear to mediate neuronal dysfunction and cell death [13,14] . death in HD is still not well understood. HD is caused by an expanded trinucleotide CAG repeat activity has been proposed to mediate mHtt aggregation [27] . 15 in the gene coding for the protein, huntingtin (Htt) [15] . There is ample evidence regarding transglutaminase In HD patients, the CAG repeat is expanded beyond expression in HD [28-30] and a role for transglutaminase the normal repeat range, > 38 repeats, with an inverse in HD pathogenesis is now well accepted. relationship between the CAG repeat number and the age of Interestingly, containing polyglutamine expansions, onset, with higher repeats associated with younger age [16] . such as mHtt, are degraded in a limited context by the 20 Expression of Htt is observed throughout the brain within the ubiquitin–proteosome system (UPS) [31] . However, recent nucleus, cytoplasm, axons and dendrites of neurons [17-19] . data suggests that the proteosome may not cleave polygluta- While the function of normal Htt is not known, it is mine sequences within the mutant protein [31] . Ongoing thought to play a role in cellular transport mechanisms [17,20] . debate continues, as with other neurodegenerative disorders Cleaved during , mutant Htt (mHtt) releases a in which protein aggregates are a hallmark of disease, 25 persistent N-terminal fragment that contains the expanded questioning whether inclusions formed by the aggregated polyglutamine amino acid sequence. This fragment, which is N-terminal truncation of mHtt cause neuronal death believed to confer toxicity, forms aggregates with itself and through alterations of nuclear transport or DNA arrange- other proteins seen in both the cytoplasm and nucleus [18] . ments affecting transcription. Recent studies have suggested The aberrant protein interactions of mHtt and formation a protective role for aggregation [19,32,33] . Through the use of 30 of mHtt aggregates may lead to neuronal toxicity through an automated microscopy technique to assess the time frame a toxic gain of function mechanism. Observations where in which neurons expressing mHtt expire, improved survival nuclear aggregates have been seen in interneuron of neurons that contained mHtt aggregates has been populations known to be resistant to HD-induced shown [32] . Whether the formation of mHtt aggregates is [19] , suggest that soluble mHtt is the protective or toxic, it is clear that coincident with mHtt 35 harbinger of neurodegeneration. In contrast, additional aggregation, there are additional pathogenic cascades at evidence suggests that the aggregated form of mHtt is toxic work in HD. and can induce neurodegeneration [21] . 3.2 Oxidative stress and mitochondrial dysfunction 2. Existing clinical management and need Several lines of evidence lend support for mitochondrial 40 dysfunction and increased oxidative stress in HD [34] . There Existing care in HD is focused primarily on symptom is a general reduction in striatal glucose utilization in both management. As such, a number of drug agents have been human and transgenic mouse HD brain that precedes tissue used to treat the motor and behavioral changes found in loss [35,36] , as well as a reduction in the activity of several HD. Both [22] and [23] , mitochondrial complexes [37,38] . Lactate levels are elevated in 45 N -methyl-D-aspartate (NMDA) antagonists, have been used the striatum and cortex, with the increase correlated with to treat chorea. While has also been used in the the CAG repeat size [39] . Biomarkers of oxidative stress are management of dystonia and psychosis in HD, treatment is also elevated in human and transgenic mouse HD brain and complicated by extrapyramidal side effects [24] . More recent serum, such as DNA oxidative modifications and strand data regarding the type 2 vesicular monoamine transporter breaks [40-42] and deletions in mitochondrial DNA [43] . 50 inhibitor, tetrabenezine, which selectively blocks dopamine Recent evidence suggests that mHtt interacts directly release, demonstrates significant improvement in chorea with mitochondria in HD [44] . This interaction causes an in ambulatory HD patients [25] . Clinical practice in HD alteration in mitochondrial buffering, leading to also employs a number of antipsychotic agents for the mitochondrial dysfunction. In addition, it has been reported management of depression, anxiety and other psychiatric that mHtt represses peroxisome proliferator-activated 55 disturbances. These include the selective serotonin uptake receptor-γ co-activator 1α (PGC-1 α ) [45] . PGC-1α is a

2 Expert Opin. Investig. Drugs (2007) 16(12) Stack & Ferrante

1 transcriptional co-activator regulating a number of genes stems from observations of similarities between kainic, and metabolic processes that protect against reactive oxygen glutamic and lesions and the striatal pathology species and is important in the mitochondrial regulation observed in rodent and primate models of HD [62-65] . of ATP [46] . Reduced levels of PGC-1α result in striatal Increases in striatal glutamate in the brains of HD 5 neurodegeneration and motor abnormalities in HD mice, patients [66] , as well as alterations in presynaptic glutamate along with increased sensitivity to oxidative stressors. receptors in the R6/2 murine model of HD [52] , lend Importantly, delivery of lentiviral-mediated PGC-1α additional support to the role of aberrant glutamate excitotoxi- expression into the striatum of R6/2 mice significantly city in HD pathogenesis. Given the fact that increased levels improved the pathological phenotype. of excitatory amino acids are not elevated in HD, the 10 concept of slow excitotoxicity in HD was suggested by Albin 3.3 Transcriptional dysregulation and Greenamyer and Beal as an alternative excitotoxic Another profound aspect of disease pathology in HD is hypothesis in which normal circulating levels of glutamate the alteration in gene transcription [47] . While the precise could result in neuronal dysfunction and death [67,68] . molecular basis for transcriptional dysregulation is unknown, Given the multiple mechanisms driving HD neuro- 15 abundant evidence suggests a direct interaction between the pathology, therapeutic intervention at any number of mHtt protein and transcription factors [48,49] . Through accessible points should prove beneficial for treating HD sequestration of transcription factors into mHtt aggregates, (F igure 1 and F igure 2 ). Importantly, targeting multiple it is thought mHtt brings about alterations in gene expres- pathogenic mechanisms may hold the greatest potential sion as observed in both human HD and murine models of to ameliorate or prevent disease progression in HD. 20 HD [50-52] . Importantly, transcriptional alterations associated Notwithstanding, recent therapeutic trials in murine models with mHtt appear presymptomatically, suggesting such of HD have provided preclinical proof-of-principle and dysregulation is not an epiphenomenon. As such, there is rationales for clinical trials in human HD, with some now strong evidence that transcriptional dysfunction is success in HD patients. related to histone hypoacetylation and hypermethylation 25 in HD [53,54] . Experimental studies in murine models 4. Therapeutic candidates have demonstrated significant hypoacetylation of histone H4 [55-57] , while hypermethylation of histone H3 is observed 4.1 Mitochondrial dysfunction and oxidative stress in human HD patients and HD mice [56-58] . Given the role of oxidative stress associated with mitochondrial dysfunction, several preclinical antioxidant strategies have 30 3.4 Apoptosis been employed with promising success. First among these is Pro-apoptotic signaling cascades initiated by mHtt likely the guanidine compound creatine, which, while produced play a role in mHtt-induced striatal neurodegeneration. In endogenously, is also obtained from the diet (see T able 1 for apoptotic-induced cell death, signaling cascades activate a compound list) [69] . In addition to its antioxidant capacity, multiple proteases that destroy proteins essential for neuronal creatine also buffers intracellular energy reserves, stabilizes 35 survival, along with a concurrent activation of genes intracellular calcium and inhibits activation of the mito- involved in cell suicide [59] . The primary constituents of the chondrial transition pore [70] . In neurons, creatine can exist apoptotic cascade are the proteases known as as free or phosphocreatine (PCr). According to the caspases. There are at least four initiator caspases and at least PCr shuttle hypothesis, sites of energy production are three effector caspases, including caspase-3, -6 and -7 [60] . connected with sites of energy consumption when creatine 40 Expanded polyglutamine stretches have been shown to kinase mediates the transfer of a phosphoryl group from sequentially activate the initiator caspases [60] . There is PCr to ADP, creating ATP [71] . In HD, there is a significant increasing evidence implicating apoptosis-mediated cell shift in the ratio of PCr to phosphate [39] . Thus, creatine death in the pathogenesis of neurodegenerative diseases. One administration may be able to restore normal metabolic important event in the apoptotic cascade is the release of activity. To this end, several preclinical studies have provided 45 cytochrome C by mitochondria into the cytoplasm, activating ample evidence of the neuroprotective benefit of creatine in caspase-9 and leading to the subsequent activation of down- chemical and animal models of neurodegenerative disease, stream executioner caspases [61] . Given apoptotic activity in including HD [72-78] . HD, pharmacologic inhibition of proteins involved in In the R6/2 mouse, creatine significantly improved various levels of the signaling cascade may represent a survival and motor performance, ameliorated brain and 50 potential beneficial therapeutic strategy to treat HD. striatal atrophy and reduced striatal mHtt aggregation in a dose-dependent manner. Oral creatine administration also 3.5 Excitotoxicity increased brain levels of creatine. This effect has been I n HD, excessive glutamatergic input to the striatum is confirmed in another animal model of HD [72] , suggesting hypothesized to contribute to the striatal neurodegeneration the significant promise of creatine administration in the 55 observed. Evidence supporting the excitotoxic hypothesis treatment of HD.

Expert Opin. Investig. Drugs (2007) 16(12) 3 Huntington’s disease: progress and potential in the fi eld

1 mHtt aggregation improvements in striatal neuropathology, with concomitant autophagy reductions in both mHtt aggregation and 8OH2′ dG levels. and the UPS In addition, there was a significant creatine-mediated improvement in striatal ATP levels. While drug trials in Mitochondrial 5 dysfuntion & mice confirm therapeutic direction, the challenge is in deter- oxidative stress mining what dose might be of value in patients, as the pharmacokinetics of mice and humans is dissimilar. As such, Pathogenic Therapeutic Transcriptional a much higher dose may be feasible for humans. In this mechanisms targets dysregulation regard, a dose escalation study up to 40 g/d to determine of HD for HD 10 whether there is a maximally tolerated dose in HD, as well as whether there are doses at which serum and brain levels Apoptosis of creatine are maximized, has been initiated. Preliminary results suggest that creatine (20 – 30 g/d) is effective in slowing disease progression. 15 Another antioxidant compound that has demonstrated Excitotoxicity preclinical efficacy in multiple murine models of HD is coenzyme Q 10 (CoQ10 ) [84-87] . CoQ10 , also known as ubiqui none, is a lipid-soluble benzoquinone that possesses Figure 1. Mechanisms of disease and potential targets. significant antioxidant properties when reduced to ubiquinol, 20 Ample evidence from in vitro and in vivo studies in mouse models or through a CoQ 10 -induced increase in α -tocopherol [88] . and human HD has implicated mHtt and its ability to form It is located in the inner mitochondrial membrane and is aggregates as being toxic to neurons. With the aggregation of mHtt, essential for Complex I and II electron transfer activities multiple pathogenic mechanisms associated with mitochondrial during oxidative phosphorylation [89] , playing a vital role dysfunction, oxidative stress, transcriptional dysregulation, apop- in ATP production. Importantly, CoQ10 administration 25 tosis and excitotoxicity are all activated. While each mechanism has been demonstrated to significantly increase brain is thought to play a role in mHtt-induced neurodegeneration, the relative pathogenicity of each is indicated from its horizontal mitochondrial CoQ10 concentrations [90] . position, with mHtt aggregation occupying the pivotal role. Initial preclinical therapeutic trials using CoQ10 in a HD: Huntington’s disease; mHtt: Mutant Huntington gene; striatal lesion model of HD demonstrated significant UPS: Ubiquitin–proteosome system. [84] . Malonate-induced lesions within the 30 striatum were significantly reduced by CoQ10 . Expanding 1 Several clinical trials show safe and tolerable doses of these results, the authors and others conducted preclinical creatine in HD patients in the range of 5 – 10 g/d [41,79-81] . therapeutic trials using CoQ10 in murine models of Creatine treatment in human HD resulted in a significant HD [85,86] . CoQ10 treatment significantly extends survival reduction in brain glutamate [79] and oxidative stress, as and delays the typical decline in weight loss and motor 355 measured by 8-hydroxy-2′ -deoxyguanosine (8OH2′ dG) [41] . performance as assessed on the rotarod. In addition, CoQ10 The 8OH2 ′ dG findings are the first instance of parallel administration significantly attenuates brain weight loss, efficacy using a common peripheral biomarker in the admin- gross brain atrophy and ventricular enlargement and striatal istration of a therapeutic agent in HD mice and HD neuron atrophy. These data have given way to several human patients. However, no studies have been sufficiently powered safety and tolerability trials using CoQ10 [39,91,92] . 4010 to detect a significant slowing of progression or improve- In all instances, CoQ10 has been found to be both safe ment in clinical measures. Although in a 1-year open-label and tolerable in HD patients. CoQ 10 treatment has resulted pilot study, creatine (10 g/d) administered for 12 months in a significant decrease in cortical lactate [39] , as well as a resulted in unchanged Unified Huntington’s disease Rating non-significant trend towards slowing in total functional Scale (UHDRS) scores, suggesting that creatine may be capacity decline over 30 months [92] . In addition, there were 4515 effective in stabilizing disease progression [82] . Although the significant beneficial effects on cognitive function, including optimal dose of creatine is not yet certain, it is possible that Stroop color naming and word reading tasks [92] . However, the dose of creatine supplementation in the above studies as the single target dose did not provide significance in the may have been underestimated. specified primary outcome of the trial, it remains unclear Recently, the authors examined the effects of a high whether a higher CoQ10 dose would provide greater efficacy 5020 dose creatine administration in multiple murine models in HD patients. A number of studies in other neurodegen- of HD [83]. High-dose creatine administration was well erative diseases suggest that a higher CoQ10 dose is possible. tolerated by both R6/2 and CAG140 mice and, at dosages A double-blind, randomized, controlled trial in Parkinson’s > 200% of previously successful preclinical dosing strategies [74] , disease (PD) patients, using CoQ10 at 1200 mg/d, slowed significant improvement in survival and motor performance the rate of deterioration in the Unified PD Rating Scale 5525 was demonstrated. Further analysis revealed significant score [93] . Follow-up studies in both PD and amyotropic

4 Expert Opin. Investig. Drugs (2007) 16(12) Stack & Ferrante

1 Creatine Mitochodrial dysfunction coenzyme Q10 mHtt Aggregation and oxidative stress ethyl-EPA dimebon Cystamine Cysteamine 5 Complex II and III mHtt Aggregates Rapamycin SMER 10, 18, and 28 Ca++/Permeability CCI-779 transition Lithium ATP prodution CytC 10 Phenylbutyrate Protease butyrate Caspase 3 activity M826 SAHA Mithramycin Apoptosis Chromomycin UPS and autophagy

15 RNA TF II Neuron death Pol II SP1130 H3

M/Ac

20 Transcriptional dysfunction

Figure 2. Targeting pathogenic mechanisms with existing and emerging therapeutic compounds. Aggregation of mHtt can be inhibited through cystamine or mercaptamine inhibition of transglutaminase. The antioxidant and bioenergetic compounds creatine and 25 coenzyme Q10 can improve mitochondrial function, while ethyl-EPA and dimebolin interact with and infl uence mitochondrial membrane function. Improvements in mitochondrial health can promote neuronal health and survival. The histone deacetylase inhibitors sodium butyrate, phenylbutyrate and SAHA, all promote maintenance of euchromatin and transcriptional activity. The antibiotics mithramycin and chromomycin also do this via interactions with GC-rich regions in the DNA. In addition, these compounds also reduce histone methylation, which is associated with heterochromatin and transcriptional repression. Proteosome activity is enhanced by rapamycin and its ester analog CCI-779, as well as lithium and multiple SMER molecules. Improved proteosomal activity can promote mHtt clearance. 30 Antiapoptotic activity is inhibited by the caspase inhibitor minocycline. The reversible inhibitor of caspase activity, M826, also reduces pro-apoptotic signaling, improving neuronal health. EPA: Eicosapentaenoic acid; mHtt: Mutant Huntington gene; SAHA: Suberoylanilide hydroxamic acid; SMER: Small molecule enhancers; UPS: Ubiquitin–proteosome system.

351 lateral sclerosis patients have demonstrated safe and tolerable been observed in rats treated with whole body doses up to 3000 mg/d [94,95] . γ -irradiation [98] by significantly reducing reactive oxygen Addressing this, the authors conducted a high dose trial species, cytochrome C translocation and caspase-3 using CoQ10 in R6/2 mice [96] , with dosages 10 times those activation. Importantly, mitochondrial dysfunction in HD 5 previously reported [84,85] . High dose CoQ10 treatment in mediated by mHtt has been shown to promote altered 40 R6/2 resulted in significant survival extension with signifi- calcium perme ability and associated cytochrome C cant improvements in motor performance. As with previous release [99] . The ability of EPA to interact with and promote preclinical trials using CoQ10 , high dose CoQ10 treatment mitochondrial fitness has stimulated interest in EPA as a in R6/2 resulted in improved neuropathology, with marked potential therapy for the treatment of HD. 10 reductions in striatal mHtt aggregation. Coupled with signifi- Using a purified derivative of EPA known as ethyl-EPA, 45 cant improvement in brain ATP levels and a reduction in an animal trial in the R6/1 murine model of HD showed brain 8OH2′ dG, these results demonstrate the pluripotent significant improvements in multiple motor and behavioral efficacy of high dose CoQ10 in the treatment of HD. A abnormalities [100] . A subsequent 6-month using multicenter Phase II – III clinical trial using high-dose ethyl-EPA in advanced HD patients demonstrated signifi- 15 CoQ10 has been initiated. cant improvement in several orofacial aspects of the 50 Therapies targeting alternative aspects of mitochondrial UHDRS [101] . The improvements in the UHDRS were con- function may also be effective. In this regard, the n-3 comitant with improved neuropathology, assessed through fatty acid eicosapentaenoic acid (EPA) possesses hypo- MRI. More recently, however, ethyl-EPA treatment in HD triglyceridemic activity, shown to occur through EPA inter- patients was reported to have no effect on the UHDRS [102] . 20 actions with mitochondria [97] , acting as a mitochondrial While secondary analysis revealed ethyl-EPA-induced 55 proliferator. EPA-induced hippocampal neuroprotection has improvements in motor function, further studies will

Expert Opin. Investig. Drugs (2007) 16(12) 5 Huntington’s disease: progress and potential in the fi eld

1

HD trials Phase II Human Human HD trials Phase III Human HD trials Phase III Human HD trials Phase III Present status

5

10 19% extension in N/A N/A 17.4% extension in survival; improved rotarod performance survival; improved rotarod performance Improved limb clasping, stide length, and locomotion 14.5% extension in survival; improved rotarod performance Outcome measures N/A R6/2 N171-82Q R6/1 R6/2 N171-82Q 15 HD mouse model

10 le 20 Suberoylanilide hydroxamic acid; SMER: Small molecule enhancer.

Increases brain levels of Coenzyme Q Increases brain levels of phosphocreatine Pharmacokineitc profi

25 channels

30 ++

An antihistamine that interacts with mitochondria and limits the mitochondrial pore transition Mitochondrial Supplement; inhibitor of mitochondrial pore transition; antioxidant Antioxidant; supports ATP production as an electron carrier; stabilizes slow Ca That targets mitochondrial and interacts with peroxisome proliferator activated receptors Mode of action

35

40 -methylhydantoin N 3 n CH COOH

n ) 45 6 H NH 2 4 (C

NH 4 3 O O O N 3 N O N 14 24 9 H H H O O 4 11 21 -Aminosarcosine; 2-imino-

C 2,3,4,5-Tetrahydro-2,8-dimethyl-5- (2-(6-methyl-3-pyridyl)ethyl)-1H-pyrido(4,3-b) indole C Ubiquinone; 2,3-dimethlbenzoquinone Ethyl-EPA 5,8,11,14,17-Eicosapentaenoic acid HO N Nomenclature/structure C

50 10 . Emerging therapeutic compounds for Huntington’s disease. 1 Table Coenzyme Q Dimebolin Compound EPA: Eicosapentaenoic acid; HD: Huntington’s disease; mHtt: Mutant Huntington gene; mTor: Mammalian target of rapomycin; SAHA: Mitochondrial dysfunction and oxidative stress Creatine 55

6 Expert Opin. Investig. Drugs (2007) 16(12) Stack & Ferrante

1

N/A N/A Present status 5

10 20.8% extension in survival; improved rotarod performance Improved rotarod performance Outcome measures R6/2 R6/2 15 HD mouse model le 20 availability Suberoylanilide hydroxamic acid; SMER: Small molecule enhancer.

Penetrates CNS. High bio in brain Penetrates CNS. Concentration in brain persists longer than in other tissues Pharmacokineitc profi

25

30

An inhibitor of histone deacetylase, transcriptional modifier An inhibitor of histone deacetylase, transcriptional modifier A histone deacetylase inhibitor that acts as a transcriptional modifier An inhibitor of neuronal apoptosis, can also bind DNA and alter epigenetic histone modifications to influence transcription Mode of action OH H O O 35 O O O O H O O HO

O 40 H O OH O O N O H O HO 6 ) 2 O

OH 45 (CH COOH COONa

OH O H 2

O N N O 3 24 Na 2 HO O O 2 O O 20 76 7 10 OH H H H H 4 14 9 52 C

C C Suberoylanilide hydroxamic acid HO Nomenclature/structure C 50 . Emerging therapeutic compounds for Huntington’s disease (continued). 1 anscriptional dysregulation r Table SAHA Phenylbutyrate Compound EPA: Eicosapentaenoic acid; HD: Huntington’s disease; mHtt: Mutant Huntington gene; mTor: Mammalian target of rapomycin; SAHA: Mithramycin T Sodium butyrate 55

Expert Opin. Investig. Drugs (2007) 16(12) 7 Huntington’s disease: progress and potential in the fi eld

1

Present status 5

10 Outcome measures

15 HD mouse model le 20 Suberoylanilide hydroxamic acid; SMER: Small molecule enhancer.

Penetrates CNS. Concentration in brain persists longer than in other tissues Pharmacokineitc profi

25

30

An inhibitor of neuronal apoptosis, can also bind DNA and alter epigenetic histone modifications to influence transcription Inhibitor of mTor, a protein kinase that regulates autophagy Mode of action OH OH O O

35 O O O O OH O OH HO OH

40 O O MeO OMe O O O OH HO O O O

45 O O HO

OMe O O

13 O O 26 N O O NO O 82 79 O H H 52 51 O C

C HO Nomenclature/structure HO 50 . Emerging therapeutic compounds for Huntington’s disease (continued). 1 Table Rapamycin Compound EPA: Eicosapentaenoic acid; HD: Huntington’s disease; mHtt: Mutant Huntington gene; mTor: Mammalian target of rapomycin; SAHA: The ubiquitin proteosomal system, autophagy and the aggregation of mHtt Chromomycin 55

8 Expert Opin. Investig. Drugs (2007) 16(12) Stack & Ferrante

1

Present status 5

10 Outcome measures

15 HD mouse model le 20 Suberoylanilide hydroxamic acid; SMER: Small molecule enhancer.

Penetrates CNS Penetrates CNS Pharmacokineitc profi

. e s a n i m a 25 t u l g s

n a r t

f o

n o i t i b

30 i h n

Inhibitor of mHtt aggregation, through inhibition of transglutaminase Inhibitor of mHtt aggregation, through i Also possesses antioxidant properties Rapamycin ester Inhibitor of mTor Small molecule enhancer of autophagy. Mechanism unknown Mode of action

35 O OH O

40 O O O O O 2 N O NH 45 N O O O

1

HO N O

16 2 O O 3 S 2 NO N N 87 17 NS 12 7 -Dithiobisthannamine H H ′ HO H H 56 16 4 2

C C 2-Aminoethanethiol 2-Mercaptethylamine 2-2 42-[3-Hydroxy-2-(hydroxymethyl)-2-methylpropanoate] rapamycin C HO Nomenclature/structure C 50 . Emerging therapeutic compounds for Huntington’s disease (continued). 1 Table Compound EPA: Eicosapentaenoic acid; HD: Huntington’s disease; mHtt: Mutant Huntington gene; mTor: Mammalian target of rapomycin; SAHA: Cysteamine SMER 10 Cystamine CCI-779, temsirolimus 55

Expert Opin. Investig. Drugs (2007) 16(12) 9 Huntington’s disease: progress and potential in the fi eld

1

Present status 5

10 Outcome measures

15 HD mouse model le 20 Suberoylanilide hydroxamic acid; SMER: Small molecule enhancer.

Penetrates CNS Pharmacokineitc profi

25

30

A tetracycline antibiotic that inhibits apoptosis through inhibition of caspase-3 An inhibitor of apoptosis via reversible inhibition An inhibitor of voltage- dependent sodium channels, resulting in attenuation of glutamate release Mode of action

35

N OH O 40 O 2 H N O NH OH O N N O N N OH 45 O OH N

3 7 6 H N F O O 2 3 7 O N N N 1 O 27 48 O 5 H H F H N OH N N 23 29 8 F

C C 2-Amino-6-trifluoromethoxy benzothiazole 7-dimethylamino-6-demethyl-6-deoxytetracycline O F Nomenclature/structure C 50 . Emerging therapeutic compounds for Huntington’s disease (continued). 1 Table Excitotoxicity M826 Compound EPA: Eicosapentaenoic acid; HD: Huntington’s disease; mHtt: Mutant Huntington gene; mTor: Mammalian target of rapomycin; SAHA: Apoptosis Minocycline 55

10 Expert Opin. Investig. Drugs (2007) 16(12) Stack & Ferrante

1 be required to determine the therapeutic potential methylation of H3 within the striatum. Together, these data of ethyl-EPA. clearly link HDACi treatment with improved transcription. The antihistamine dimebolin (2,3,4,5-tetrahydro-2,8- With the prospect of HDACi compounds offering therapeu- dimethyl-5-(2-(6-methyl-3-pyridyl)ethyl)-1H -pyrido(4,3-b) tic benefit in the treatment of HD, a dose-finding study 5 indole), is an orally active small molecule that has multiple using sodium phenylbutyrate recently demonstrated doses in mechanisms of action. It may exert a neuroprotective effect the range of 12 – 15 g/d were safe and well-tolerated [108] . by interacting with the mitochondrial permeability transition The authors’ own preliminary data using 15 g/d in a safety pore and preventing the calcium-induced opening of the and tolerability trial in HD patients confirms the above pore [103]. Studies of dimebolin in animal models of study and, in addition, shows that sodium phenylbutyrate 10 Alzheimer’s disease have showed improved cognitive ability, was therapeutically salient in significantly improving while inhibiting β -amyloid [104] . Preliminary results in hypoacetylation levels in blood buffy-coat specimens. Alzheimer’s disease patients have also been promising; for In addition to compounds that directly interact with example, dimebolin may also regulate calcium homeostasis HDACs, another class of potential HD therapeutics exist and reduce the excitotoxicity, thus there may be potential that interact directly with DNA and that may be able to 15 benefit in administering dimebolin to HD patients. As such, influence transcriptional activity in HD. Two such com- a Phase II clinical trial examining safety and tolerability of pounds are mithramycin and chromomycin, anthracycline dimebolin is presently recruiting HD patients. antibiotics that act through modulating gene transcription. By binding to -cytosine-rich regions within gene 4.2 Transcriptional dysregulation promoters, anthracyclines displace transcriptional elements 20 The transcriptional repression observed in HD likely results that activate and repress transcription [109] . Importantly, from alterations in chromatin packaging associated with anthracyclines have been reported to interact directly with epigenetic modifications of histone proteins. In general, thera- histones H3 and H4 [110] . This has led to several preclinical peutic manipulation of transcription may offer significant studies investigating the potential utility of mithramycin or benefit in treating HD, as well as other neurodegenerative chromomycin in murine models of HD [56-58] . 25 disorders. In particular, pharmacological targeting of histone Mithramycin administration in R6/2 HD mice resulted methylation and acetylation status may be a unique method in the largest significant extension in survival (29.1%) by which to achieve transcriptional homeostasis and, by compared with any other preclinical therapeutic trial in HD extension, neuroprotection in HD. Several preclinical trials to date [56] . The mithramycin-mediated improvement in with compounds directed toward altered histone profiles in survival was concomitant with significant improvements 30 HD have been performed. One strategy has been to target in motor performance and striatal morphology. Notably, histone acetylation by administering histone deacetylase mithramycin induced a significant decrease in methylated inhibitors (HDACi) [55,105] . H3. In a follow-up study, the authors demonstrated The HDACi’s sodium butyrate or suberoylanilide mithramycin-mediated improvements in the methylation hydroxamic acid (SAHA) in a Dr osophila model of HD, and acetylation profile of H3 and H4 within the striatum of 35 provides significant neuroprotection [105] . These results N171-82Q mice [57] . Treatment with chromomycin or were supported by in vitro analyses that demonstrated mithramycin in R6/2 and N171-82Q HD mice significantly mHtt-induced inhibition of the histone increased acetylation of H4 and significantly reduced proteins CBP and p300, and improved acetylation profile trimethylation of H3 at 9. Additional analyses of H3 of histone H4 after sodium butyrate. Sodium butyrate revealed an anthracycline-mediated shift toward greater 40 and SAHA were also shown to offer neuroprotection in the acetylation and reduced methylation compared with R6/2 murine model of HD [55,106] . Sodium butyrate and untreated controls. This latter finding may be of particular SAHA improved motor performance and, while not reported importance given that methylation of H3 at lysine 9 is for SAHA, sodium butyrate significantly improved thought to be a dominant marker of transcriptional repression survival [55] . Both compounds also markedly improved and the balance between methylation and acetylation of H3 45 striatal morphology. Importantly, both sodium butyrate and at lysine 9 is believed to play an important role in the SAHA improved acetylation of histone H4. Improvements transcriptional disruption observed in HD [58] . in H4 acetylation mediated by sodium butyrate were While it may seem incongruous that cytotoxic antitumor concomitant with transcriptional improvements in R6/2 compounds may play a positive role in neurodegenerative striatum, assessed by microarray gene profiling, resulting in disorders, parallels between cancer and neurodegenerative 50 improved mRNA expression [55] . disorders have been suggested. However, if cellular stresses Confirming and expanding these data, administration of and transcriptional signals elicit different responses in the HDACi phenylbutyrate in the N171-82Q murine model dividing cells versus cells that are terminally differentiated of HD resulted in significant neuroprotection as well [107] . Of (leading to oncogenesis in the former and neurodegeneration interest, in addition to phenylbutyrate-mediated improvements in the latter), then different pathogenic mechanisms may 55 in H4 acetylation, there was also a significant reduction in underlie each. Importantly, previous clinical use of these

Expert Opin. Investig. Drugs (2007) 16(12) 11 Huntington’s disease: progress and potential in the fi eld

1 compounds has been associated with negative side effects, for use in human patients. Employed in several clinical including fever, nausea or vomiting, fatigue and depression. contexts, recent effectiveness has been demonstrated in Both agents cross the blood–brain barrier and mithramycin cancer chemotherapy [119] . More recent evidence demon- has been used chronically in a number of human strated that rapamycin significantly improved neuronal 5 conditions. The preclinical mithramycin and chromomycin survival, compared with wild type flies. In addition, the data provide a rationale for clinical trials of these clinically rapamycin ester CCI-779 significantly improved motor approved anthracyclines to test for efficacy in the treatment performance and striatal neuropathology in the N171-82Q of HD. murine model of HD [120] . A subsequent screen of small molecules capable of 10 4.3 The ubiquitin–proteosome system, autophagy modulating autophagy independent of mTOR was and the aggregation of mutant Htt performed [121] . Of ∼ 100 compounds screened, there were The degradation of mHtt is another potential therapeutic a dozen small molecule enhancers (SMERs)of autophagy, target in HD. In normal neurons, organelle and protein from which three positive hits (SMER 10, 18 and 28) were turnover is a critical feature that promotes health and function. shown to reduce mHtt aggregation in vitro . Further analysis 15 Altered proteolysis may result in aberrant protein changes in demonstrated significant protection against mHtt toxicity in denaturation or misfolding. Two distinct routes mediating an in vivo model of HD. While the precise therapeutic proteolysis in neurons are the ubiquitin–proteosomal mechanism of SMERs remains elusive, analysis of pathway and the lysosomal pathway. Proteins destined for phosphorylation status in targets of mTOR showed no degradation mediated by the UPS must first be tagged SMER-induced effects, suggesting the SMERs act 20 for degradation. In general, the UPS is responsible for downstream of mTOR to induce autophagy. the degradation of transiently expressed proteins [111] . An additional study of potential inducers of autophagy as Importantly, proteins destined for proteosomal degradation a treatment for HD, which operate independently of mTOR, must first be sufficiently unfolded to fit through the narrow has identified lithium as a potential therapeutic compound opening of the proteosome [112] . for HD. Lithium significantly reduces clearance of mHtt 25 In addition, degradation of proteins and organelles in and mHtt-induced cell death in vitro [122] . The similar mood bulk is accomplished through the lysosomal pathway, in a stabilizing drug also reduced mHtt-induced process termed autophagy [113] . Through this pathway, cell death and mHtt aggregation in vitro . The in vitro cellular components destined for degradation are enveloped lithium-induced clearance of mHtt mediated by autophagy in double membrane bound vesicles, called autophagosomes, is independent of mTOR activity and dependent on inositol 30 which fuse with lysosomes. Once fused, hydrolytic lysosomal monophosphatase 1 (IMPase) activity and the stimulatory degrade the contents. While the mechanisms effect was blocked with subsequent addition of inositol regulating autophagy are not completely characterized, it is a triphosphate. Interestingly, combined inhibition of mTOR process regulated by protein kinases, including the well and IMPase by rapamycin and lithium, respectively, resulted characterized mammalian target of rapamycin (mTOR) [114] . in additive clearance of mHtt in vitro [122] . 35 Phosphorylated mTOR is linked with protein synthesis, While strategies targeting enhanced clearance may whereas dephosphorylation of mTOR induces autophagy [115] . promote improved neuronal survival, therapeutic attenua- Furthermore, mTOR mediated autophagy has been linked tion of mutant protein aggregation may also prove thera- to glucose levels, with increased glucose stimulating auto- peutically valuable in treating HD. In this regard, phagy and enhanced mHtt clearance through reduced cystamine may hold significant promise. Cystamine is a 40 mTOR phosphorylation [116] . It is worth noting, however, -containing compound that possesses multiple that autophagy can be induced through activity of the modes of action, from antioxidant properties [123] to inhibi- insulin receptor substrate-2, independent of mTOR tion of transglutaminase [124] . Indeed, recent preclinical data activity [117] . This results in a significant reduction of from multiple laboratories has demonstrated the potential mHtt aggregation in vitro and is dependent on therapeutic benefit of cystamine in treating polyglutamine 45 normal autophagosome formation mediated by Beclin1 disorders, including HD [125-127] . More recently, the dimer and hVps34. of cystamine, mercaptamine, a product of cystamine reduc- Notwithstanding, with the importance of mTOR tion, has completed Phase I human trials determining in autophagy and the role of autophagy in HD, compounds maximum dose tolerability and safety [128] . These data, in that can interact with mTOR to promote autophagy concert with previous clinical use of mercaptamine for 50 may prove exceptionally beneficial in HD. Using the treatment of cystinosis [129] , demonstrate the unique potential specific inhibitors of autophagy 3-methyladenine or of cystamine and its analogs in the treatment of HD. N 6,N 6-dimethyladenosine, the number and size of mHtt aggregates increases [118] . In contrast, induction of autophagy 4.4 Apoptosis by rapamycin resulted in a significant reduction in mHtt Among antiapoptotic drug candidates, the tetracycline 55 aggregation. Rapamycin, a macrolide antibiotic, is approved antibiotic minocycline has emerged as a potentially beneficial

12 Expert Opin. Investig. Drugs (2007) 16(12) Stack & Ferrante

1 therapeutic intervention for treatment in HD. Minocycline additional study to improve and assess both M826 possesses potent antiapoptotic capacity through inhibitory and brain penetration in vivo [141,142] . effects on caspase-1 and -3. In addition, minocycline also attenuates disruptions in mitochondrial function, including 4.5 Excitotoxicity 5 the release of cytochrome C [61,130] . Importantly, minocy- G lutamate excitotoxicity is also thought to play a cline also readily crosses the blood–brain barrier. Clinically, role in HD pathogenesis. Given extensive evidence in chronic administration of minocycline has yielded a good support of an excitotoxic hypothesis for HD, compounds safety record [131] . From a therapeutic standpoint, minocy- that counter excessive glutamate release may, therefore, cline has shown significant improvement in multiple models be candidates for therapeutic intervention in HD. 10 of neurodegeneration, including brain trauma, spinal cord One such FDA-approved compound is riluzole injury, PD and HD [61,87,132-135] . (2-amino-6-trifluoromethoxy benzothiazole), a potent In preclinical trials using minocycline in murine models antiglutamatergic agent, which attenuates glutamate release of HD, several studies have demonstrated a significant through its ability to inhibit voltage-dependent sodium neuroprotective effect. Minocycline significantly inhibited channels [143] . In HD, the potential benefit of riluzole was 15 caspase-1 and caspase-3 activation in R6/2 mice [61] . first suggested by preclinical studies in rats and non-human Minocycline also significantly reduced mHtt cleavage. primates, using the 3-nitroproprionic (3-NP) chemical In addition to their role in apoptotic signaling cascades, model of HD [144,145] . In the 3-NP model of HD, riluzole caspases also play a role in cleaving mHtt, yielding the offers significant improvements in motor performance, toxic fragment [136] . Inhibition of caspase activity was with significant neuroprotection observed. Expanding 20 associated with improved survival and motor behavior in these findings, riluzole was found to significantly increase R6/2 mice. Extending these findings, minocycline has survival in R6/2 mice concomitant with significant improve- been shown to significantly inhibit both initiator and ments in motor behavior [146] . There was also a marked effector caspases, including caspase-1, -3, -8 and -9, as riluzole-mediated reduction in ubiquitin-positive mHtt well as the pro-apoptotic Bid cleavage [134] . In addition, aggregates within the striatum. Riluzole was also found to 25 minocycline also inhibited both the release of cytochrome C protect medium spiny neurons against glutamate-induced and Smac/Diablo from mitochondria in R6/2 mice, apoptosis in vitro [147] . The aberration in corticostriatal indicating that mitochondria are a direct target of function resulting in excessive glutamate release is widely minocycline-mediated neuroprotection [130,133,134] . thought to contribute to the selective striatal pathology These preclinical minocycline studies have given way to observed in HD [68,148] . Interestingly, riluzole administration 30 pilot clinical trials assessing safety and tolerability in human significantly reduces aberrant excitatory postsynaptic HD. At doses of 100 and 200 mg/d, minocycline was found currents in R6/2 mice, lending further support for riluzole to be well tolerated by patients [137] . In terms of cognitive therapy in HD [149] . outcomes, there were no clinically relevant differences in To that end, several clinical trials in human HD have cognition assessed by UHDRS. Similar results in pilot trials been conducted. In a 6-week safety and tolerability trial 35 using minocycline at 100 mg/d over 6 months have been with riluzole that assessed motor performance and brain reported [138,139] . Excellent safety and tolerability data for lactate levels [150] , riluzole was found safe and well tolerated, minocycline treatment in HD patients has led the Huntington with a non-significant trend toward lower basal ganglia Study Group to conduct a Phase II trial that is presently lactate levels. Analysis of motor function demonstrated a underway. Of interest is a recent clinical trial in amyotropic significant decrease in chorea, as measured via the UHDRS. 40 lateral sclerosis using 400 mg/d that showed no efficacy [140] . Further clinical evaluations in HD patients confirmed It has been suggested that the target dose was too great, riluzole’s efficacy [151,152] . Interestingly, the efficacy of other resulting in the negative findings. glutamatergic NMDA antagonists, amantadine [22] and In addition to minocycline-mediated caspase inhibition, memantine [23] , has had mixed results [153-156] . Each of these a recent report demonstrated a novel reversible inhibitor drug agents is associated with significant side effects. 45 of caspase-3 that was shown to provide significant neuro- protection in a chemical rat model of HD [141] . In a 4.6 Supplementary therapeutic strategies preclinical proof-of-principle trial, M826, a pyrazinone In addition to potential therapies as described above, there mono-, demonstrated significant protection against are several other therapeutic approaches that may prove malonate lesions, with a pharmacokinetic profile indicating the beneficial in treating HD and, thus, deserve mention here. 50 ability of M826 to inhibit capsase-3 6 h postadministration. RNA interference (RNAi) is one such therapy that takes Striatal lesion volumes were significantly reduced following advantage of a functionally conserved pathway present in all M826 administration and the number of neurons expressing eukaryotes [157] . The molecular machinery mediating RNAi active caspase-3 was also significantly reduced. While these activity includes both micro RNA (miRNA) and short results demonstrate significant neuroprotective potential, the interfering RNA (siRNA). Through associations between 55 route of administration (intracerebral injection) will require various proteins, including Argonaute-2 and individual

Expert Opin. Investig. Drugs (2007) 16(12) 13 Huntington’s disease: progress and potential in the fi eld

1 RNAi molecules, a functional complex is formed that can motor and behavioral alterations, demonstrating improved then target homologous mRNA. Once bound to the functional capacity [168] . homologous mRNA species, Argonaute-2 cleaves, and thus Using recommendations for trial criteria from the Core inactivates, the homologous mRNA [158] . Both miRNA Assessment Program for Intracerebral Transplantation in 5 and siRNA can prevent translation of homologous mRNA HD [169] , an initial pilot grafting paradigm was employed when each possesses a limited number of mismatches [159] . where three HD patients received bilateral transplantation of Through this mechanism, RNAi could be manipulated to fetal striatal tissue into the caudate and putamen [170] . Graft reduce expression of protein products known to cause survival was determined through comparison of pre- and disease. In the case of HD, the ability to effectively 1 year postsurgical MRI, with marked improvement in 10 target and downregulate mHtt expression may hold signal, consistent with graft survival. In all three patients, significant promise. striatal tissue transplantation resulted in improved motor In this regard, several preclinical studies have shown behavior, as assessed by the UHDRS. the potential promise of RNAi therapy in HD. Using an In a subsequent safety and tolerability trial employing adeno-associated viral vector (AAV) expressing a short hair- fetal striatal tissue transplants into the caudate and 15 pin RNA precursor targeting the Htt gene, mHtt expression putamen [171] , several HD patients showed marked improve- is reduced in the striatum of N171-82Q mice [160] . RNAi ment in UHDRS scores. However, complications in the use targeting mHtt also improved motor behavior, with improved of immunosuppressant therapies postoperatively were gait and rotarod performance. Similar results were also observed, making analyses impossible. Additional trials with obtained in the R6/1 murine model of HD [161] . cross-species striatal transplantation were performed using 20 Through elimination of the toxic mHtt via specific porcine fetal tissues in human HD patients [172] . Even with targeting of the mHtt protein, these preclinical studies therapy to suppress immunological xenograft rejection, demonstrate the promise of RNAi-based therapies for the no surviving striatal transplants were observed and no treatment of HD. However, it is important to note that functional improvements noted. Together, these latter trials such therapeutic intervention in HD may yet be many years represent the difficulties of treating human HD with clinical 25 off. While intracerebral infusion of AAV containing striatal transplants. RNAi may be suitable in animal studies, safe and While completed clinical trials demonstrate safety effective delivery of RNAi molecules to humans has yet to and tolerability, with adequate surgical procedures to be firmly established. perform the tissue transplants, certain aspects of tissue or Striatal neuron transplant, via striatal tissue graft or cell preparation and delivery for surgical implantation remain 30 dissociated striatal suspension, has also been suggested to unresolved. Furthermore, given ethical concerns regarding hold promise as a therapeutic intervention in HD. The the use of fetal tissue or the use of alternative cells such as rationale for neural transplantation arises from the fact that porcine fetal grafts, issues arise regarding immunological neurodegeneration eliminates specific neuronal populations, function and management of tissue rejections [173] . Finally, which can theoretically be replaced with the addition of new while striatal cell transplant may hold promise for the 35 neurons, akin to organ transplant. In this system, it is pro- treatment of HD, one caveat that remains is the significant posed that transplanted neuronal tissue would re-establish gross neuropathology observed as disease progresses, with the anatomical and functional aspects of the damaged and extra-striatal neuropathology present. Indeed, cortical neuro- lost neurons [162] . Several preclinical and clinical studies in pathology in HD likely contributes to many of the PD have provided proof-of-principle data suggesting behavioral and cognitive disruptions associated with advanced 40 the potential benefit of transplantation in the treatment disease. As previous studies have not examined behavioral of PD [163,164] . and motor performance beyond 1 year postoperatively, it Initial studies using rodent chemical lesion models of HD remains to be seen whether striatal transplantation will have have demonstrated successful striatal transplant survival, a long-term, broad therapeutic benefit. including dopamine terminal innervation of the transplant 45 and a recovery of striatal choline acetyltransferase and 5. Conclusion decarboxylase [165] . Subsequent studies have demonstrated functional recovery of motor behaviors The search for effective strategies aimed at halting or after striatal transplantation [166] . Similar studies in a reversing the insidious march of HD will require extensive non-human primate chemical lesion model of HD have preclinical and clinical validation to provide the necessary 50 demonstrated successful stereotaxic implantation of safety and tolerability data for effective clinical use. All of cross-species striatal neuronal grafts (rat to baboon) into the the compounds described in this review are at some stage of caudate-putamen [167] . Post-transplant analyses revealed this process. Many have demonstrated significant potential graft survival, with expression of striatal markers evident. in preclinical trials involving mice. The development of Additional studies in non-human primates confirmed that genetic models has greatly expanded our understanding of 55 grafting of striatal tissue into lesioned caudate ameliorated HD pathogenesis. These models also provide complex,

14 Expert Opin. Investig. Drugs (2007) 16(12) Stack & Ferrante

1 yet accessible, biological systems with which candidate Alternatively, optimal therapeutic dosing may be therapeutic compounds can be tested for efficacy and mode underestimated. The reliance on human equivalent dose of action. While such models greatly enhance the discovery extrapolation measurements derived from body surface area potential, it is important to understand the difficulty inher- criteria in animals may be deficient [174] , as it likely does not 5 ent in predicting the transference of success from mouse to accurately predict the maximum recommended safe dose man. Interestingly, recent evidence shows a parallel in effi- in humans. This is evident in human trials where human cacy in both HD patients and murine models of HD using equivalency dosing of bioenergetic agents based on pre- antioxidant therapies in reducing peripheral oxidative stress clinical murine trials in mice has not demonstrated similar levels of 8OH2′ dG [41,96] . Difficulties notwithstanding, efficacy in patients. However, while dose extrapolation may 10 preclinical therapeutic trials with murine models provide not be straightforward, it is well accepted that the pheno- perhaps the best foundation on which to base human types from mouse models of neurological diseases closely clinical trials. Importantly, data from preclinical trials using correlate with human diseases, thus providing a system with multiple models is likely to be most informative when which to validate known CNS drug targets. As drug trials in assessing potential benefit in human HD. mice can confirm therapeutic potential, the challenge is to 15 identify an efficacious dose in humans. 6. Expert opinion An additional factor to consider when assessing potential clinical benefit is that most compounds described above A major goal of existing clinical research in HD is to are already available for use in humans. This is likely to improve early detection of disease and premanifest detection accelerate clinical validation. However, an important consid- 20 of neuronal dysfunction with translation to therapeutic eration that must be addressed is the myriad of mechanisms trials. Biomarkers are urgently needed for diagnosis, disease underlying HD pathogenesis. It is likely that mHtt induces progression and for potential disease-modifying therapies disruptions in transcriptional activity that ultimately results that are being developed and evaluated in clinical trials, in neurodegeneration. Due to the regional specificity of especially at the preclinical stage. The development of early neurodegeneration, there are likely other important mecha- 25 premanifest biomarkers is of great importance, as these may nisms, such as mitochondrial dysfunction and associated improve the power and cost-effectiveness of drug trials. oxidative stress, which play an important role in HD While many different approaches have been undertaken to pathogenesis. Given the multiple pathogenic mechanisms identify biomarkers, profiling objective biomarker measure- active in HD, combinatorial treatment paradigms will be ments of HD has proven difficult at the present time. essential for treatment of HD. In this regard, it is important 30 The optimal biomarker should be easily quantified and to acknowledge the multiple therapeutic mechanisms measured, reproducible and not subject to wide variation. inherent in certain compounds, such as cystamine. Given In therapeutic evaluation, linear change with disease pro- that cystamine acts both as a tissue transglutaminase inhibi- gression that closely correlates with clinico-pathological tor and a potent antioxidant, its potential benefit, when assessments of the disease is critical. As there may be a paired with the transcriptional modulator chromomycin, 35 prolonged period of time in which neurons become may yield synergistic results enhancing the therapeutic benefit dysfunctional before clinical expression of disease, preclinical of each. While the precise combination that may yield the detection of biomarkers offers the promise of administering most significant benefit is not yet known, it is likely that a disease-modifying during the premanifest combinatorial paradigm will prove most suitable for effec- period, further delaying or ameliorating disease symptoms. tively treating HD. Given the relative safety of creatine and 40 The early identification of premanifest biomarkers in HD CoQ10 , it is likely that either high-dose CoQ10 or creatine, that correspond to disease activity, disease progression and or both agents, will represent a cornerstone defense in disease response to therapy would greatly facilitate the ameliorating the progression of HD. accurate evaluation of the effectiveness of new therapies and improve the safety and efficiency of clinical trials. Declaration of interest 45 While successful preclinical trials demonstrating improved phenotype in HD transgenic mice have yet to be fully This work was supported by NIH grants NS045242 validated in HD patients, this may be the consequence of (Robert J Ferrante), NS045806 (Robert J Ferrante), and underpowered clinical trials in humans, preventing a the Veterans Administration (Robert J Ferrante and comparison of therapeutic efficacy between mouse and human. Edward C Stack). 50

55

Expert Opin. Investig. Drugs (2007) 16(12) 15 Huntington’s disease: progress and potential in the fi eld

1 Bibliography glutamic acid decarboxylase, choline polyglutamine neurodegenerative disorders. 1. KOROSHETZ WJ, MYERS RH, acetyltransferase and dopamine in basal N ature ( 2003 ) 421 : 373 -379. MARTIN JB: The neurology of ganglia. Brain ( 1974 ) 97 : 457-472. 22. VERHAGEN METMAN L, Huntington’s disease. In: M ovement 12. REINER A, ALBIN RL, ANDERSON KD, MORRIS MJ, FARMER C et al. : 5 Disorders in Neurology and Neuropsychiatry . D’AMATO CJ, PENNEY JB, YOUNG AB: Huntington’s disease: a randomized, Joseph A, Young R (Eds), Blackwell Differential loss of striatal projection controlled trial using the Scientifi c Publications, Boston neurons in Huntington’s disease. P roc. Natl. NMDA-antagonist amantadine. ( 1992): 173 -190. Acad. Sci. USA ( 1988 ) 85 : 5733-5737. N eurology (2002 ) 59 :694 -699. 2. HAHN-BARMA V, DEWEER B, 13. FERRANTE RJ, KOWALL NW, 23. ONDO WG, MEJIA NI, HUNTER CB: DURR A et al. : Are cognitive changes 10 RICHARDSON EP Jr: Proliferative A pilot study of the clinical effi cacy and the fi rst symptoms of Huntington’s and degenerative changes in striatal safety of memantine for Huntington’s disease? A study of gene carriers. spiny neurons in Huntington’s disease: disease. P arkinsonism Relat. Disor d. J. Neurol. Neurosurg. Psychiatry ( 1998 ) a combined study using the section-Golgi ( 2006 ):[Epub ahead of print]. 64 : 172-177. method and calbindin D28k 24. BONELLI RM, WENNING GK: 3. LAWRENCE AD, HODGES JR, immunocytochemistry. J. Neurosci. (1991 ) Pharmacological management of 15 ROSSER AE et al. : Evidence for specifi c 11 : 3877 -3887. Huntington’s disease: an evidence-based cognitive defi cits in preclinical 14. GRAVELAND GA, WILLIAMS RS, review. C urr. Pharm. Des. ( 2006 ) Huntington’s disease. B rain ( 1998 ) DIFIGLIA M: Evidence for degenerative 12 : 2701 -2720. 121 : 1329 -1341. and regenerative changes in neostriatal 25. HUNTINGTON STUDY GROUP: 4. GOMEZ-TORTOSA E, spiny neurons in Huntington’s disease. Tetrabenazine as antichorea therapy in 20 MACDONALD ME, FRIEND JC et al. : Science (1985 ) 227 : 770 -773. Huntington disease: a randomized Quantitative neuropathological changes 15. HUNTINGTON’S DISEASE controlled trial. N eurology ( 2006) in presymptomatic Huntington’s disease. COLLABORATIVE RESEARCH 66 : 366 -372. A nn. Neurol. (2001 ) 49 : 29 -34. GROUP: A novel gene containing a 26. RAMASWAMY S, SHANNON KM, 5. VONSATTEL JP, MYERS RH, trinucleotide repeat that is expanded KORDOWER JH: Huntington’s disease: 25 STEVENS TJ, FERRANTE RJ, and unstable on Huntington’s disease pathological mechanisms and therapeutic BIRD ED, RICHARDSON EP Jr: . Cell ( 1993 ) strategies. C ell Transplant. ( 2007 ) Neuropathological classifi cation of 72 : 971 -983. 16 : 301 -312. Huntington’s disease. J. Neuropathol. 16. DJOUSSE L, KNOWLTON B, 27. ZAINELLI GM, DUDEK NL, ROSS CA, Exp. Neurol. (1985 ) 44 : 559-577. HAYDEN M et al. : Interaction of KIM SY, MUMA NA: Mutant huntingtin 30 6. KOWALL NW, FERRANTE RJ, normal and expanded CAG repeat sizes protein: a substrate for transglutaminase 1, MARTIN JB: Patterns of cell loss in infl uences age at onset of Huntington 2, and 3. J. Neuropathol. Exp. Neurol. Huntington’s disease. T rends Neurosci. disease. A m. J. Med. Genet. A ( 2003 ) ( 2005 ) 64 : 58 -65. ( 1987) 10 : 24 -29. 119 : 279 -282. 28. HOFFNER G, KAHLEM P, DJIAN P: 7. FERRANTE RJ, BEAL MF, 17. DIFIGLIA M, SAPP E, CHASE K Perinuclear localization of huntingtin et al. : Huntingtin is a cytoplasmic 35 KOWALL NW, RICHARDSON EP Jr, as a consequence of its binding to MARTIN JB: Sparing of protein associated with vesicles in human microtubules through an interaction acetylcholinesterase-containing and rat brain neurons. N euron ( 1995 ) with β -tubulin: relevance to Huntington’s striatal neurons in Huntington’s disease. 14 : 1075 -1081. disease. J. Cell Sci. ( 2002 ) 115 : 941-948. B rain Res. (1987 ) 411 : 162 -166. 18. DIFIGLIA M, SAPP E, CHASE KO 29. KARPUJ MV, GARREN H, SLUNT H 8. FERRANTE RJ, KOWALL NW, et al. : Aggregation of huntingtin in 40 et al. : Transglutaminase aggregates BEAL MF, RICHARDSON EP Jr, neuronal intranuclear inclusions and huntingtin into nonamyloidogenic BIRD ED, MARTIN JB: Selective dystrophic neurites in brain. Science (1997 ) polymers, and its enzymatic activity sparing of a class of striatal neurons in 277 : 1990 -1993. increases in Huntingtonx’s disease Huntington’s disease. Science ( 1985 ) 19. KUEMMERLE S, GUTEKUNST CA, brain nuclei. P roc. Natl. Acad. Sci. USA 230 : 561 -563. KLEIN AM et al. : Huntington aggregates ( 1999) 96 : 7388-7393. 45 9. FERRANTE RJ, KOWALL NW, may not predict neuronal death in 30. LESORT M, CHUN W, JOHNSON GV, RICHARDSON EP Jr, BIRD ED, Huntington’s disease. Ann. Neurol. (1999 ) FERRANTE RJ: Tissue transglutaminase MARTIN JB: Topography of enkephalin, 46 : 842 -849. is increased in Huntington’s disease brain. substance P and acetylcholinesterase 20. VELIER J, KIM M, SCHWARZ C J. Neurochem. ( 1999 ) 73 :2018 -2027. staining in Huntington’s disease striatum. et al. : Wild-type and mutant huntingtins 31. HOLMBERG CI, STANISZEWSKI KE, N eurosci. Lett. ( 1986 ) 71 : 283 -288. 50 function in vesicle traffi cking in the MENSAH KN, MATOUSCHEK A, 10. BIRD ED: Chemical pathology of secretory and endocytic pathways. MORIMOTO RI: Ineffi cient degradation Huntington’s disease. A nnu. Rev. E xp. Neurol. (1998 ) 152 : 34 -40. of truncated polyglutamine proteins by P harmacol. Toxicol. (1980 ) 20 : 533 -551. 21. SANCHEZ I, MAHLKE C, YUAN J: the proteasome. EMBO J. ( 2004 ) 11. BIRD ED, IVERSEN LL: Huntington’s Pivotal role of oligomerization in expanded 23 :4307 -4318. 55 chorea. Post-mortem measurement of

16 Expert Opin. Investig. Drugs (2007) 16(12) Stack & Ferrante

1 32. ARRASATE M, MITRA S, 42. MECOCCI P, MACGARVEY U, “H3 barcode hypothesis”. P roc. Natl. Acad. SCHWEITZER ES, SEGAL MR, KAUFMAN AE et al. : Oxidative Sci. USA (2006 ) 103 :6428 -6435. FINKBEINER S: Inclusion body damage to mitochondrial DNA shows 54. STRAHL BD, ALLIS CD: The language formation reduces levels of mutant marked age-dependent increases in of covalent histone modifi cations. N ature 5 huntingtin and the risk of neuronal human brain. Ann. Neurol. ( 1993 ) ( 2000 ) 403 :41 -45. death. N ature (2004 ) 431 : 805 -810. 34 :609 -616. 55. FERRANTE RJ, KUBILUS JK, LEE J 33. TAYLOR JP, TANAKA F, 43. POLIDORI MC, MECOCCI P, et al. : Histone deacetylase inhibition by ROBITSCHEK J et al. : Aggresomes BROWNE SE, SENIN U, BEAL MF: sodium butyrate chemotherapy protect cells by enhancing the degradation Oxidative damage to mitochondrial ameliorates the neurodegenerative of toxic polyglutamine-containing protein. DNA in Huntington’s disease parietal 10 phenotype in Huntington’s disease mice. H um. Mol. Genet. (2003 ) 12 : 749 -757. cortex. Neurosci. Lett. ( 1999 ) 272 : 53 -56. J. Neurosci. (2003 ) 23 : 9418 -9427. 34. GRUNEWALD T, BEAL MF: 44. PANOV AV, GUTEKUNST CA, 56. FERRANTE RJ, RYU H, KUBILUS JK Bioenergetics in Huntington’s disease. LEAVITT BR et al. : Early mitochondrial et al. : Chemotherapy for the brain: the A nn. NY Acad. Sci. ( 1999 ) 893 : 203 -213. calcium defects in Huntington’s disease antitumor antibiotic mithramycin 35. JENKINS BG, KLIVENYI P, are a direct effect of polyglutamines. prolongs survival in a mouse model of 15 KUSTERMANN E et al. : Nonlinear N at. Neurosci. (2002 ) 5 :731 -736. Huntington’s disease. J. Neurosci. ( 2004 ) decrease over time in N -acetyl aspartate 45. CUI L, JEONG H, BOROVECKI F, 24 :10335 -10342. levels in the absence of neuronal loss and PARKHURST CN, TANESE N, 57. STACK EC, DEL SIGNORE SJ, increases in and glucose in KRAINC D: Transcriptional repression LUTHI-CARTER R et al. : Modulation transgenic Huntington’s disease mice. of PGC-1 α by mutant huntingtin of nucleosome dynamics in Huntington’s 20 J. Neurochem. ( 2000 ) 74 : 2108-2119. leads to mitochondrial dysfunction and disease. H um. Mol. Genet. ( 2007) 36. KUHL DE, PHELPS ME, neurodegeneration. C ell ( 2006) 127 :59 -69. 16 :1164 -1175. MARKHAM CH, METTER EJ, 46. ROHAS LM, ST-PIERRE J, ULDRY M, 58. RYU H, LEE J, HAGERTY SW et al. : RIEGE WH, WINTER J: Cerebral JAGER S, HANDSCHIN C, ESET/SETDB1 and metabolism and atrophy in Huntington’s SPIEGELMAN BM: A fundamental histone H3 (K9) trimethylation in 25 disease determined by 18FDG and system of cellular energy homeostasis Huntington’s disease. P roc. Natl. Acad. computed tomographic scan. regulated by PGC-1 α . P roc. Natl. Sci. USA ( 2006) 103 :19176 -19181. A nn. Neurol. (1982 ) 12 : 425 -434. Acad. Sci. USA (2007 ) 104 :7933 -7938. 59. HENGARTNER MO: The biochemistry 37. BROWNE SE, BOWLING AC, 47. SUGARS KL, RUBINSZTEIN DC: of apoptosis. N ature ( 2000 ) 407 : 770 -776. MACGARVEY U et al. : Oxidative Transcriptional abnormalities in 60. FRIEDLANDER RM: Apoptosis and damage and metabolic dysfunction in Huntington disease. T rends Genet. 30 caspases in neurodegenerative diseases. Huntington’s disease: selective vulnerability ( 2003 ) 19 :233 -238. N. Engl. J. Med. ( 2003) 348 : 1365 -1375. of the basal ganglia. A nn. Neurol. ( 1997 ) 48. DUNAH AW, JEONG H, GRIFFIN A 61. CHEN M, ONA VO, LI M et al. : 41 : 646 -653. et al. : Sp1 and TAFII130 transcriptional Minocycline inhibits caspase-1 and 38. TABRIZI SJ, CLEETER MW, activity disrupted in early Huntington’s caspase-3 expression and delays mortality XUEREB J, TAANMAN JW, disease. Science ( 2002 ) 296 : 2238 -2243. 35 in a transgenic mouse model of COOPER JM, SCHAPIRA AH: 49. RUBINSZTEIN DC: How does the Huntington disease. N at. Med. (2000 ) Biochemical abnormalities and Huntington’s disease mutation damage 6 :797 -801. excitotoxicity in Huntington’s disease cells? Sci. Aging Knowledge Environ. (2003 ) 62. BEAL MF, FERRANTE RJ, SWARTZ KJ, brain. A nn. Neurol. ( 1999 ) 45 : 25 -32. 37 : PE26. KOWALL NW: Chronic quinolinic acid 39. KOROSHETZ WJ, JENKINS BG, 50. AUGOOD SJ, FAULL RL, EMSON PC: lesions in rats closely resemble Huntington’s 40 ROSEN BR, BEAL MF: Energy Dopamine D1 and D2 receptor gene disease. J. Neurosci. ( 1991 ) 11 :1649 -1659. metabolism defects in Huntington’s expression in the striatum in Huntington’s 63. COYLE JT, SCHWARCZ R: disease and effects of coenzyme Q10. disease. Ann. Neurol. ( 1997 ) 42 :215 -221. A nn. N eurol. ( 1997 ) 41 : 160 -165. Lesion of striatal neurones with kainic 51. BOROVECKI F, LOVRECIC L, ZHOU J acid provides a model for Huntington’s 40. BOGDANOV MB, ANDREASSEN OA, et al. : Genome-wide expression profi ling chorea. N ature ( 1976) 263 : 244-246. 45 DEDEOGLU A, FERRANTE RJ, of human blood reveals biomarkers 64. FERRANTE RJ, KOWALL NW, BEAL MF: Increased oxidative damage for Huntington’s disease. P roc. Natl. CIPOLLONI PB, STOREY E, BEAL MF: to DNA in a transgenic mouse model Acad. Sci. USA ( 2005 ) 102 : 11023 -11028. of Huntington’s disease. J. Neurochem. Excitotoxin lesions in primates as a model 52. CHA JH, FREY AS, ALSDORF SA et al. : (2001 ) 79 : 1246 -1249. for Huntington’s disease: histopathologic Altered neurotransmitter receptor and neurochemical characterization. 50 41. HERSCH SM, GEVORKIAN S, expression in transgenic mouse models E xp. Neurol. ( 1993 ) 119 :46 -71. MARDER K et al. : Creatine in Huntington of Huntington’s disease. P hilos. Trans. R 65. MCGEER EG, MCGEER PL: disease is safe, tolerable, bioavailable in Soc. Lond. Biol. Sci. ( 1999 ) 354 : 981 -989. brain and reduces serum 8OH2′dG. Duplication of biochemical changes of 53. HAKE SB, ALLIS CD: Histone H3 N eurology (2006 ) 66 : 250 -252. Huntington’s chorea by intrastriatal variants and their potential role in 55 indexing mammalian genomes: the

Expert Opin. Investig. Drugs (2007) 16(12) 17 Huntington’s disease: progress and potential in the fi eld

1 injections of glutamic and kainic acids. effects of creatine and cyclocreatine in 88. BEAL MF, FERRANTE RJ: N ature (1976 ) 263 : 517 -519. animal models of Huntington’s disease. Experimental therapeutics in transgenic 66. TAYLOR-ROBINSON SD, WEEKS RA, J. N eurosci. (1998 ) 18 : 156 -163. mouse models of Huntington’s disease. BRYANT DJ et al. : Proton magnetic 78. ZHU S, LI M, FIGUEROA BE et al. : N at. Rev. Neurosci. ( 2004 ) 5 : 373-384. 5 resonance spectroscopy in Huntington’s Prophylactic creatine administration 89. CHAN TS, WILSON JX, O’BRIEN PJ: disease: evidence in favour of the glutamate mediates neuroprotection in cerebral Coenzyme Q cytoprotective mechanisms. excitotoxic theory. M ov. Disord. ( 1996) ischemia in mice. J. Neurosci. (2004 ) M ethods Enzymol. ( 2004 ) 382 :89 -104. 11 :167 -173. 24 : 5909 -5912. 90. MATTHEWS RT, YANG L, BROWNE S, 67. ALBIN RL, GREENAMYRE JT: 79. BENDER A, AUER DP, MERL T et al. : BAIK M, BEAL MF: Coenzyme Q10 10 Alternative excitotoxic hypotheses. Creatine supplementation lowers brain administration increases brain N eurology (1992 ) 42 : 733 -738. glutamate levels in Huntington’s disease. mitochondrial concentrations and exerts 68. BEAL MF: Does impairment of energy J. Neurol. (2005 ) 252 : 36 -41. neuroprotective effects. P roc. Natl. metabolism result in excitotoxic neuronal 80. TABRIZI SJ, BLAMIRE AM, Acad. Sci. USA ( 1998) 95 : 8892 -8897. death in neurodegenerative illnesses? MANNERS DN et al. : High-dose creatine 91. FEIGIN A, KIEBURTZ K, COMO P A nn. Neurol. (1992 ) 31 : 119 -130. therapy for Huntington disease: a 2-year et al. : Assessment of coenzyme Q10 15 69. RYU H, ROSAS HD, HERSCH SM, clinical and MRS study. N eurology ( 2005 ) tolerability in Huntington’s disease. FERRANTE RJ: The therapeutic role of 64 : 1655 -1656. M ov. Disord. (1996 ) 11 : 321-323. creatine in Huntington’s disease. 81. VERBESSEM P, LEMIERE J, 92. HUNTINGTON STUDY GROUP: P harmacol. Ther. ( 2005 ) 108 : 193 -207. EIJNDE BO et al. : Creatine A randomized, placebo-controlled trial supplementation in Huntington’s of coenzyme Q10 and remacemide in 20 70. O’GORMAN E, BEUTNER G, DOLDER M, KORETSKY AP, disease: a placebo-controlled pilot trial. Huntington’s disease. N eurology ( 2001 ) BRDICZKA D, WALLIMANN T: N eurology ( 2003 ) 61 : 925-930. 57 :397 -404. The role of creatine kinase in inhibition 82. TABRIZI SJ, BLAMIRE AM, 93. SHULTS CW, OAKES D, KIEBURTZ K of mitochondrial permeability transition. MANNERS DN et al. : Creatine therapy et al. : Effects of coenzyme Q10 in early FEBS Lett. (1997 ) 414 : 253 -257. for Huntington’s disease: clinical and Parkinson’s disease: evidence of slowing 25 71. BESSMAN SP, GEIGER PJ: Transport MRS fi ndings in a 1-year pilot study. of the functional decline. A rch. Neurol. of energy in muscle: the phosphocreatine N eurology ( 2003 ) 61 :141 -142. ( 2002 ) 59 : 1541-1550. shuttle. Science ( 1981 ) 211 : 448 -452. 83. FORAN EF, DEL SIGNORE SJ, 94. FERRANTE KL, SHEFNER J, 72. ANDREASSEN OA, DEDEOGLU A, MARKEY A et al. : Dose ranging and ZHANG H et al. : Tolerance of high-dose FERRANTE RJ et al. : Creatine increases effi cacy study of high-dose creatine in (3,000 mg/day) coenzyme Q10 in ALS. 30 survival and delays motor symptoms in a Huntington disease mouse models. N eurology ( 2005 ) 65 :1834 -1836. transgenic animal model of Huntington’s 36th Annual Meeting of the Society for 95. SHULTS CW, BEAL MF, SONG D, disease. N eurobiol. Dis. ( 2001 ) 8 : 479 -491. Neuroscience . Atlanta, GA (2006 ). FONTAINE D: Pilot trial of high 73. DEDEOGLU A, KUBILUS JK, YANG L 84. BEAL MF, HENSHAW DR, dosages of coenzyme Q10 in patients et al. : Creatine therapy provides JENKINS BG, ROSEN BR, SCHULZ JB: with Parkinson’s disease. E xp. Neurol. 35 neuroprotection after onset of clinical Coenzyme Q10 and nicotinamide block (2004 ) 188 :491 -494. symptoms in Huntington’s disease striatal lesions produced by the 96. SMITH KM, MATSON S, transgenic mice. J. Neurochem. (2003 ) mitochondrial toxin malonate. A nn. Neurol. MATSON WR et al. : Dose ranging and 85 : 1359-1367. ( 1994 ) 36 :882 -888. effi cacy study of high-dose coenzyme Q10 74. FERRANTE RJ, ANDREASSEN OA, 85. FERRANTE RJ, ANDREASSEN OA, formulations in Huntington’s disease 40 JENKINS BG et al. : Neuroprotective DEDEOGLU A et al. : Therapeutic effects mice. B iochim. Biophys. Acta ( 2006 ) effects of creatine in a transgenic mouse of coenzyme Q10 and remacemide in 1762 : 616 -626. model of Huntington’s disease. J. Neurosci. transgenic mouse models of Huntington’s 97. FROYLAND L, MADSEN L, ( 2000) 20 : 4389 -4397. disease. J. Neurosci. ( 2002 ) 22 : 1592-1599. VAAGENES H et al. : 75. KLIVENYI P, FERRANTE RJ, 86. SCHILLING G, COONFIELD ML, is the principal target for nutritional ROSS CA, BORCHELT DR: and pharmacological control of 45 MATTHEWS RT et al. : Neuroprotective effects of creatine in a transgenic animal Coenzyme Q10 and remacemide triglyceride metabolism. J. Lipid Res. model of amyotrophic lateral sclerosis. hydrochloride ameliorate motor defi cits in ( 1997 ) 38 : 1851-1858. N at. Med. ( 1999) 5 : 347 -350. a Huntington’s disease transgenic mouse 98. LONERGAN PE, MARTIN DS, model. N eurosci. Lett. ( 2001) 315 : 149-153. 76. MATTHEWS RT, FERRANTE RJ, HORROBIN DF, LYNCH MA: KLIVENYI P et al. : Creatine and 87. STACK EC, SMITH KM, RYU H Neuroprotective effect of 50 cyclocreatine attenuate MPTP et al. : Combination therapy using eicosapentaenoic acid in hippocampus neurotoxicity. E xp. Neurol. (1999 ) minocycline and coenzyme Q10 in of rats exposed to γ -irradiation. 157 : 142 -149. R6/2 transgenic Huntington’s disease J. Biol. Chem. (2002 ) 277 :20804 -20811. mice. B iochim. Biophys. Acta ( 2006 ) 77. MATTHEWS RT, YANG L, 99. CHOO YS, JOHNSON GV, 1762 : 373 -380. JENKINS BG et al. : Neuroprotective MACDONALD M, DETLOFF PJ, 55 LESORT M: Mutant huntingtin directly

18 Expert Opin. Investig. Drugs (2007) 16(12) Stack & Ferrante

1 increases susceptibility of mitochondria width and fl exibility. B iopolymers models of Huntington disease. to the calcium-induced permeability ( 2000 ) 56 : 85 -95. N at. Genet. ( 2004 ) 36 : 585 -595. transition and cytochrome C release. 110. RABBANI A, FINN RM, 121. SARKAR S, PERLSTEIN EO, H um. Mol. Genet. ( 2004 ) 13 : 1407 -1420. THAMBIRAJAH AA, AUSIO J: IMARISIO S et al. : Small molecules 5 100. CLIFFORD JJ, DRAGO J, NATOLI AL Binding of antitumor antibiotic enhance autophagy and reduce toxicity et al. : Essential fatty acids given from daunomycin to histones in chromatin in Huntington’s disease models. conception prevent topographies of motor and in solution. B iochemistry (2004 ) N at . Chem. Biol. ( 2007 ) 3 :331 -338. defi cit in a transgenic model of 43 : 16497 -16504. 122. SARKAR S, FLOTO RA, BERGER Z Huntington’s disease. N euroscience ( 2002 ) 111. CIECHANOVER A: The ubiquitin et al. : Lithium induces autophagy by 109 : 81 -88. 10 proteolytic system: from a vague idea, inhibiting inositol monophosphatase. 101. PURI BK, BYDDER GM, through basic mechanisms, and onto J. Cell Biol. ( 2005) 170 : 1101 -1111. COUNSELL SJ et al. : MRI and human diseases and drug targeting. 123. REVESZ L, MODIG H: neuropsychological improvement in N eurology ( 2006 ) 66 : S7-S19. Cysteamine-induced increase of Huntington disease following ethyl-EPA 112. PICKART CM, VANDEMARK AP: cellular -level: a new treatment. N euroreport (2002 ) 13 : 123 -126. Opening doors into the proteasome. hypothesis of the radioprotective 15 102. PURI BK, LEAVITT BR, HAYDEN MR N at. Struct. Biol. ( 2000 ) 7 : 999-1001. mechanism. N ature ( 1965 ) 207 : 430 -431. et al. : Ethyl-EPA in Huntington disease: 113. RUBINSZTEIN DC: The roles of 124. LORAND L, SIEFRING GE Jr, a double-blind, randomized, intracellular protein-degradation LOWE-KRENTZ L: Formation of placebo-controlled trial. N eurology ( 2005 ) pathways in neurodegeneration. N ature γ -glutamyl-ε-lysine bridges between 65 : 286-292. (2006 ) 443 : 780 -786. membrane proteins by a Ca2+ -regulated 20 103. BACHURIN SO, SHEVTSOVA EP, 114. SCHMELZLE T, HALL MN: TOR, in intact erythrocytes. KIREEVA EG, OXENKRUG GF, a central controller of cell growth. J. Supramol. Struct. (1978 ) 9 : 427 -440. SABLIN SO: Mitochondria as a target C ell (2000 ) 103 :253 -262. 125. DEDEOGLU A, KUBILUS JK, for neurotoxins and neuroprotective 115. SOMWAR R, SUMITANI S, TAHA C, JEITNER TM et al. : Therapeutic agents. Ann. NY Acad. Sci. (2003 ) SWEENEY G, KLIP A: Temporal effects of cystamine in a murine model 993 : 334-344; discussion 345-339. 25 activation of p70 S6 kinase and Akt1 of Huntington’s disease. J. Neurosci. 104. BACHURIN S, BUKATINA E, by insulin: PI3-kinase-dependent (2002 ) 22 : 8942-8950. LERMONTOVA N et al. : Antihistamine and -independent mechanisms. 126. IGARASHI S, KOIDE R, agent dimebon as a novel neuroprotector A m. J. Physiol. ( 1998) 275 :E618 -E625. SHIMOHATA T et al. : Suppression of and a cognition enhancer. A nn. NY 116. RAVIKUMAR B, STEWART A, aggregate formation and apoptosis by Acad. Sci. (2001 ) 939 : 425 -435. 30 KITA H, KATO K, DUDEN R, transglutaminase inhibitors in cells 105. STEFFAN JS, BODAI L, PALLOS J RUBINSZTEIN DC: Raised intracellular expressing truncated DRPLA protein et al. : Histone deacetylase inhibitors glucose concentrations reduce aggregation with an expanded polyglutamine stretch. arrest polyglutamine-dependent and cell death caused by mutant huntingtin N at. Genet. ( 1998 ) 18 :111 -117. neurodegeneration in Dr osophila . N ature exon 1 by decreasing mTOR 127. KARPUJ MV, BECHER MW, (2001 ) 413 : 739 -743. 35 phosphorylation and inducing autophagy. SPRINGER JE et al. : Prolonged survival 106. HOCKLY E, RICHON VM, H um. Mol. Genet. (2003 ) 12 :985 -994. and decreased abnormal movements in WOODMAN B et al. : Suberoylanilide 117. YAMAMOTO A, CREMONA ML, transgenic model of Huntington disease, hydroxamic acid, a histone deacetylase ROTHMAN JE: Autophagy-mediated with administration of the transglutaminase inhibitor, ameliorates motor defi cits in a clearance of huntingtin aggregates triggered inhibitor cystamine. N at. Med. (2002 ) mouse model of Huntington’s disease. 8 :143 -149. 40 by the insulin-signaling pathway. P roc. Natl. Acad. Sci. USA ( 2003 ) J. Cell Biol. ( 2006 ) 172 : 719 -731. 128. DUBINSKY R, GRAY C: CYTE-I-HD: 100 : 2041 -2046. 118. RAVIKUMAR B, DUDEN R, Phase I dose fi nding and tolerability 107. GARDIAN G, BROWNE SE, CHOI DK RUBINSZTEIN DC: Aggregate-prone study of cysteamine (Cystagon) in et al. : Neuroprotective effects of proteins with polyglutamine and Huntington’s disease. M ov. Disord. (2006 ) phenylbutyrate in the N171-82Q polyalanine expansions are degraded by 21 : 530 -533. 45 transgenic mouse model of Huntington’s autophagy. H um. Mol. Genet. (2002 ) 129. MCDOWELL GA, TOWN MM, disease. J. Biol. Chem. ( 2005 ) 280 : 556 -563. 11 : 1107 -1117. VAN’T HOFF W, GAHL WA: 108. HOGARTH P, LOVRECIC L, 119. WENDEL HG, DE STANCHINA E, Clinical and molecular aspects of KRAINC D: Sodium phenylbutyrate in FRIDMAN JS et al. : Survival signaling nephropathic cystinosis. J. Mol. Med. Huntington’s disease: a dose-fi nding study. by Akt and eIF4E in oncogenesis (1998 ) 76 : 295 -302. 50 M ov. Disord. ( 2007 ):[Epub ahead of print]. and cancer therapy. N ature ( 2004) 130. ZHU S, STAVROVSKAYA IG, 109. CHAKRABARTI S, 428 :332 -337. DROZDA M et al. : Minocycline BHATTACHARYYA D, DASGUPTA D: 120. RAVIKUMAR B, VACHER C, inhibits cytochrome C release and Structural basis of DNA recognition by BERGER Z et al. : Inhibition of mTOR delays progression of amyotrophic anticancer antibiotics, chromomycin A(3), induces autophagy and reduces toxicity of lateral sclerosis in mice. N ature ( 2002) 55 and mithramycin: roles of minor groove polyglutamine expansions in fl y and mouse 417 : 74 -78.

Expert Opin. Investig. Drugs (2007) 16(12) 19