154
Views
21
CrossRef citations to date
0
Altmetric
Review

Current and emerging treatment options in the management of Friedreich ataxia

, , &
Pages 491-499 | Published online: 30 Jul 2010

Abstract

Friedreich ataxia (FRDA) is the most common autosomal recessive ataxia. Oxidative damage within the mitochondria seems to have a key role in the disease phenotype. Therefore, FRDA treatment options have been mostly directed at antioxidant protection against mitochondrial damage. Available evidence seems to suggest that patients with FRDA should be treated with idebenone, because it is well tolerated and may reduce cardiac hypertrophy and, at higher doses, also improve neurological function, but large controlled clinical trials are still needed. Alternatively, gene-based strategies for the treatment of FRDA may involve the development of small-molecules increasing frataxin gene transcription. Animal and human studies are strongly needed to assess whether any of the potential new treatment strategies, such as iron-chelating therapies or treatment with erythropoietin or histone deacetylase inhibitors and other gene-based strategies, may translate into an effective therapy for this devastating disorder. In this review, we try to provide an answer to some questions related to current and emerging treatment options in the management of FRDA.

Introduction

Friedreich ataxia (FRDA, OMIM #229300) is the most common autosomal recessive ataxia among Caucasian population, and it is caused by mutations in the FXN gene (OMIM *606829), mainly an expanded GAA triplet repeat in the intron 1.Citation1 Age at onset is typically 5–25 years. Sensory neurons in the dorsal root ganglia are lost initially, with secondary degeneration of the spinocerebellar tract, pyramidal tract, and dorsal columns.Citation2 FRDA is, therefore, characterized by progressive gait and limb ataxia, dysarthria, loss of vibration and proprioceptive sense, areflexia, abnormal eye movements, and pyramidal signs. Involvement of the auditory sensory neurons and pathways may also be found, as in optic atrophy.Citation3 Ataxia of mixed cerebellar and sensory type is the cardinal symptom. The first symptom is usually gait instability, though scoliosis may already be present when neurologic symptoms appear, and, in rare cases, hypertrophic cardiomyopathy is diagnosed before the onset of ataxia.

In patients with FRDA, voxel-based morphometry showed a symmetrical volume loss in dorsal medulla, infero-medial portions of the cerebellar hemispheres, the rostral vermis, and in the dentate region.Citation4 No volume loss in cerebral hemispheres was observed. The atrophy of the cerebellum and medulla correlated with the severity of the clinical deficit and disease duration.Citation4 Moreover, some magnetic resonance imaging-based studies found cerebral white matter atrophy or dysfunction.Citation5,Citation6

A possible manifestation of this disease is hypertrophic cardiomyopathy, described in up to two-third of patients with FRDA.Citation7 Ventricular arrhythmias can also occur. Later in the course of the disease, the hypertrophied heart can develop systolic dysfunction and heart failure and arrhythmias are possible causes of death in these patients.Citation8 Diabetes, scoliosis, and pes cavus are other possible manifestations of FRDA. Clinical course is variable, but on average 10–15 years after onset, patients lose the ability to walk, stand, and sit without support.Citation3 Age at diagnosis, which may incorporate other genetic and environmental factors, may be more important than GAA length in predicting cardiomyopathy, scoliosis, and disease progression.Citation9

In FRDA the genetic abnormality results in the deficiency of frataxin, a protein targeted to the mitochondrion.Citation10 In about 98% of patients, the disease is caused by a triplet GAA expansion within the first intron of the frataxin gene on chromosome 9q13, which impedes transcription of the gene and limits protein production.Citation1,Citation11 The repeat expansions can range from 70 to 90 repeats (normal less than 40) to over 1,000, with inverse correlation of age at onset, severity of the disease, and associated systemic symptoms.Citation2 Heterozygous carriers are clinically healthy. FRDA is the most common disease-causing triplet-repeat expansion identified so far, about 1 in 100 Europeans being a carrier. No other disease has been recognized to date to be caused by an expansion of GAA triplets.Citation3 Some patients are compound heterozygotes with the GAA expansion in one allele and one of a variety of point mutations in the other allele.Citation2 The FRDA-associated expansion shows instability when transmitted from parent to child. Expansions and contractions can both be observed and are equally likely after maternal transmission, whereas contractions are most common after paternal transmission.Citation3 In this regard, FRDA resembles the other diseases associated with very large expansions in noncoding regions, including fragile X syndrome and myotonic dystrophy, and differs from the diseases that are caused by CAG repeats in coding regions, such as dominant ataxias and Huntington disease, in which size increases typically occur after paternal transmission.Citation3

FRDA pathogenetic theories and their relevance for therapeutic approaches

Although the exact physiological function of frataxin is not known, its involvement in iron–sulfur cluster biogenesis has been suggested.Citation12 Frataxin iron-binding capacity is quite robust. Even when 5 of the most conserved residues from the putative iron-binding region are altered, at least 2 iron atoms per monomer can be bound.Citation13 Current evidence suggests that loss of frataxin impairs mitochondrial iron handlingCitation14 and respiratory chain function and contributes to increased oxidative stress and cellular damage.Citation2

In a conditional knockout mouse model where frataxin was removed from the heart, transferrin receptor-1 was upregulated, resulting in increased iron uptake from transferrin. Citation15 There is also marked downregulation of ferritin that is required for iron storage and decreased expression of the iron exporter, ferroportin 1, leading to decreased cellular iron efflux. The increased mitochondrial iron uptake is facilitated by upregulation of the mitochondrial iron transporter, mitoferrin 2.Citation15 This stimulation of iron uptake probably attempts to rescue the deficit in mitochondrial iron metabolism that is due to downregulation of mitochondrial iron utilization (heme and iron–sulfur cluster synthesis and iron storage in mitochondrial ferritin). Therefore, increased mitochondrial iron uptake coupled with decreased utilization and release leads to mitochondrial iron-loading.Citation16

Abnormalities of the neuronal cytoskeleton due to oxidative stress and increased protein glutathionylation have been also suggested to have a potential role in FRDA.Citation17 Oxidative damage within the mitochondria seems to have a key role in the disease phenotype.Citation18 A combined deficiency of a Krebs-cycle enzyme, aconitase, and 3 mitochondrial respiratory- chain complexes was reported in endomyocardial biopsy samples from patients with this disorder.Citation19 All 4 enzymes share iron – sulfur cluster-containing proteins that are damaged by iron overload through generation of oxygen free radicals.Citation19 Using phosphorus magnetic resonance spectroscopy, Lodi et alCitation20 demonstrated a maximum rate of muscle mitochondrial adenosine triphosphate (ATP) production below the normal range in all the 12 studied FRDA patients and a strong negative correlation between mitochondrial ATP production and the number of GAA repeats in the smaller allele, suggesting that FRDA is a nuclear-encoded mitochondrial disorder.Citation20 Moreover, Giacchetti et alCitation21 reported an influence of the mitochondrial DNA polymorphisms on the FRDA phenotype. These authors studied 99 patients with FRDA and 48 control individuals, all from southern Italy. They found that patients belonging to the haplogroup U class had a delay of 5 years in the disease onset and a lower rate of cardiomyopathy. Citation21 Mitochondrial DNA polymerase (POLG) CAG repeat instability has been also proposed as a predisposing factor that, in combination with environmental risk factors, may affect age of onset and FRDA progression.Citation22 For the described reasons, FRDA treatment options have been mostly directed at antioxidant protection against mitochondrial damage. Interestingly, other studies demonstrated that in FRDA mitochondrial iron accumulation did not induce oxidative stress and that FRDA is a neurodegenerative disease not associated with oxidative damage.Citation23

To date, no randomized controlled trial using antioxidants or any other pharmacological treatment has shown significant benefit on neurological symptoms associated FRDA.Citation24 Moreover, the design of clinical trials in FRDA presents some problems, such as the lack of reliable biomarkers that correlate with clinical dysfunction and the rarity of the condition. Therefore, therapeutic strategies are still unclear. In this review, we try to provide an answer to some questions related to current and emerging treatment options in the management of FRDA. References for this review were identified by searches of PubMed until May 2010 with the term “Friedreich*”; articles especially considering potential therapeutic approaches were considered. Emphasis was placed on comprehensive reviews and original articles published after 1998. Other articles were identified through references from relevant articles. Only papers published in English were reviewed.

Which pharmacological agents were effective in FRDA cellular and animal models?

PPARγ agonists

A recent microarray analysis of heart and skeletal muscle in a mouse model of frataxin deficiency showed molecular evidence of increased lipogenesis in skeletal muscle, and alteration of fiber-type composition in heart, consistent with insulin resistance and cardiomyopathy, respectively.Citation25 Since the peroxisome proliferator-activated receptor gamma (PPARγ) pathway is known to regulate both processes, dysregulation of this pathway may play a role in FRDA. PPARγ coactivator 1-alpha (PGC-1a) downregulation may contribute to the blunted antioxidant response observed in cells from FRDA patients.Citation26 PGC-1a is a transcriptional master regulator of mitochondrial biogenesis and antioxidant responses, and can be restored by agonist pioglitazione in FRDA cells,Citation26 suggesting a potential therapeutic approach for FRDA. Moreover, Marmolino et alCitation27 investigated the effect of another PPARγ agonist (Azelaoyl PAF) on the frataxin protein and mRNA expression profile in human neuroblastoma cells (SKNBE) and primary fibroblasts from skin biopsies from FRDA patients and healthy controls. Azelaoyl PAF increased both messenger RNA and protein levels of intracellular frataxin in SKNBE cells and fibroblasts from FRDA patients.Citation27

Iron chelators

Iron chelators that target the mitochondrion have been proposed (ie, deferiprone).Citation28,Citation29 Adding the chelator deferiprone at clinical concentrations to inducibly frataxin-deficient HEK-293 cells resulted in chelation of mitochondrial labile iron, involved in oxidative stress.Citation30 This led to restoration of impaired mitochondrial membrane and redox potentials, increased ATP production and oxygen consumption, and attenuation of mitochondrial DNA damage and reversal of hypersensitivity to apoptosis.Citation30 On the other hand, a direct consequence of chelating mitochondrial free iron in various cell systems is a concentration and time-dependent loss of aconitase activity, which was shown to precede decreased cell proliferation.Citation31 Therefore, if chelating excessive mitochondrial iron may be beneficial at some stage of the disease, attention should be paid to not fully deplete mitochondrial iron store.Citation31 Li et alCitation32 investigated the regulation of frataxin expression by iron and reported that frataxin mRNA levels decreased significantly in multiple human cell lines treated with the iron chelator desferal. In addition, frataxin mRNA and protein levels decreased in fibroblast and lymphoblast cells derived from both normal controls and from patients with FRDA.Citation32 Lymphoblasts and fibroblasts of FRDA patients show evidence of cytosolic iron depletion, which may occur as frataxin-deficient cells overload their mitochondria with iron,Citation32 as already discussed. Therapeutic efforts should focus on an approach that combines iron removal from mitochondria with a treatment that increases cytosolic iron levels to maximize residual frataxin expression in FRDA patients.Citation32

Antioxidant agents

Coenzyme Q10 has been widely used for the treatment of neurodegenerative disorders, as well as its analog idebenone, which shares an identical modified parahydroxybenzoate ring with Coenzyme Q10, but has a short carbon tail. Idebenone was cytoprotective in fibroblasts from patients with FRDA.Citation33 Because of the role of mitochondrial oxidative damage in FRDA, Jauslin et alCitation34 compared the efficacy of mitochondria-targeted and untargeted antioxidants derived from coenzyme Q10 and from vitamin E at preventing oxidative stress-induced cell death in cultured fibroblasts from FRDA patients in which glutathione synthesis was blocked. Ubiquinones have been shown to protect mitochondria from oxidative damage, but only a small proportion of externally administered ubiquinone is taken up by mitochondria.Citation35 Conjugation of the lipophilic triphenylphosphonium cation to a ubiquinone moiety has produced a compound, MitoQ, which accumulates selectively into mitochondria. MitoQ passes easily through all biological membranes and, because of its positive charge, is accumulated several 100-fold within mitochondria driven by the mitochondrial membrane potential.Citation35 The mitochondria-targeted antioxidant MitoQ was 800-fold more potent than the untargeted analog idebenone. Citation34 The mitochondria-targeted antioxidant MitoVit E was 350-fold more potent than the water soluble analog.Citation34 Therefore, targeted antioxidants may have therapeutic potential in FRDA and in other disorders involving mitochondrial oxidative damage, but have not been investigated in FRDA patients to date.

A dose-escalation trial found that idebenone was efficacious in a frataxin-deficient mouse model of FRDA.Citation36 Low-dose idebenone (10 and 30 mg/kg per day) showed no benefit, whereas high-dose idebenone (90 mg/kg per day) delayed the cardiac disease onset, progression, and death of frataxin-deficient animals by 1 week.Citation36

Gene-based strategies

FRDA is a loss of function disorder, therefore gene-based strategies designed to increase frataxin levels could be an ideal therapy for this disease, although there are still technological limitations to their clinical applicability.Citation37 Viral vectors expressing frataxin partially corrected sensitivity to oxidative stress in FRDA fibroblasts.Citation38,Citation39 Furthermore, in mice with a localized frataxin reduction in the brainstem functional impairment could be corrected by exposure to HSV-1 vector expressing frataxin cDNA.Citation40 Other studies reported that compounds specifically targeting the GAA repeat, such as DNA sequence-specific polyamidesCitation41 or pentamidine,Citation42 were capable of increasing frataxin transcription. The exact mechanism by which these DNA-binding compounds increase transcription through GAA repeats still needs further characterization.Citation37

HDAC inhibitors

Alternative gene-based strategy for the treatment of FRDA would involve the development of small-molecules increasing frataxin gene transcription.Citation37 Expanded GAA repeats may silence frataxin expression inducing heterochromatin formation, and/or forming non B-DNA structures, such as triplex and sticky DNA, which block gene transcription. Therefore, possible treatments for FRDA may include drugs that facilitate chromatin opening, such as histone deacetylase (HDAC) inhibitors.Citation37 Gene silencing at expanded FXN alleles is accompanied by hypoacetylation of histones H3 and H4 and trimethylation of histone H3, observations that are consistent with a heterochromatin-mediated repression mechanism.Citation43 Herman et alCitation43 reported the synthesis and characterization of a class of HDAC inhibitors that reversed FXN silencing in primary lymphocytes from individuals with FRDA. These molecules directly affected the histones associated with FXN, increasing acetylation at particular lysine residues on histones H3 and H4. One compound, BML-210, showed a significant increase in frataxin message levels by approximately 2-fold.Citation43 Butyric acid is another HDAC inhibitor reported to increase frataxin expression.Citation44 Furthermore, compounds with pimelic diphenylamide basic structure were able to upregulate frataxin in cells from FRDA patients and in a mouse model.Citation45 HDAC3 could be the likely cellular target of the pimelic diphenylamides HDAC inhibitors and the target for therapeutic intervention in FRDA.Citation46 Although both the HDAC3 and HDAC1/2-specific compounds share a similar mechanism of inhibition of their target enzymes, only HDAC3-specific compounds increase frataxin gene expression and frataxin protein in cells.Citation46 Rai et alCitation47 treated KIKI mice (homozygous mice carrying a [GAA]230 repeat in the first intron of the mouse frataxin gene) with a novel HDAC inhibitor, compound 106, which substantially increases frataxin mRNA levels in cells from FRDA individuals. The treatment increased histone H3 and H4 acetylation in chromatin near the GAA repeat and restored wild-type frataxin levels in the nervous system and heart, as determined by quantitative reverse transcription polymerase chain reaction and semiquantitative western blot analysis.Citation47 Lack of acute toxicity, normalization of frataxin levels, and of the transcription profile changes resulting from frataxin deficiency may provide support to a possible efficacy of this or related compounds in FRDA patients.Citation47

Conclusive remarks

In conclusion, gene-based strategies (including HDAC inhibitors), the agonist pioglitazione, iron chelators that target the mitochondrion such as deferiprone, coenzyme Q10, vitamin E (especially their mitochondria-targeted forms), and idebenone were found to be effective in FRDA cellular and animal models. Clinical studies with some of these agents are reviewed in the next paragraphs. Moreover, there is some preliminary evidence that flavin adenine dinucleotideCitation48 and heminCitation44 may rescue the phenotype of frataxin deficiency in cellular and animal models, but further studies are still needed.

Is there a role for idebenone and other antioxidants in FRDA patients?

Idebenone

The antioxidant idebenone is a short-chain benzoquinone derivative with a structure similar to coenzyme Q10 but with a more favorable pharmacokinetic profile.Citation49 In vitro studies have shown that it acts both as an antioxidant, preventing damage to the mitochondrial membrane, and as an electron carrier, supporting mitochondrial function and ATP production. Citation50 High doses of idebenone are safe and well tolerated in patients with FRDA.Citation49 The idebenone half-life was relatively consistent across dose levels (2.6–21.7 hours).Citation49 It exhibited dose-dependent pharmacokinetics in daily doses up to 2,250 mg.Citation51 Idebenone plasma levels are thought to correlate with central nervous system concentrations because of its ability to penetrate central nervous system.Citation49

Schulz et alCitation52 measured concentrations of 8-hydroxy- 2′-deoxyguanosine (8OH2′dG), a marker of oxidative DNA damage, in urine and of dihydroxybenzoic acid (DHBA), a marker of hydroxyl radical attack, in plasma of 33 patients with FRDA. They found a 2.6-fold increase in normalized urinary 8OH2′dG but no change in plasma DHBA as compared with controls. Oral treatment with 5 mg/kg/day of idebenone for 8 weeks significantly decreased urinary 8OH2′dG concentrations.Citation52

Idebenone is a promising drug for treatment of FRDA.Citation53 Early trials have demonstrated that low-dose idebenone (5 mg/kg per day) reduced cardiac hypertrophy (as determined by echocardiography)Citation54,Citation55 in the majority of patients with FRDA, with no influence upon clinical progression of the neurological disease.Citation54,Citation56

Recently, a randomized, placebo-controlled trial has been conducted on 48 patients with genetically confirmed FRDA.Citation57 Treatment with higher doses of idebenone (up to 45 mg/kg) was generally well tolerated and associated with improvement also in neurological function and activities of daily living in patients with FRDA. The degree of improvement correlated with the dose of idebenone, suggesting that higher doses may be necessary to have a beneficial effect on neurological function.Citation57 It has been also suggested that the disease stage and patient age at which idebenone treatment is initiated may be important factors in the effectiveness of the therapy.Citation58,Citation59 Larger randomized trial focusing on the response to idebenone therapy of both neurological and heart symptoms are required to confirm whether an early diagnosis of FRDA can be exploited to initiate such antioxidant treatment in order to prevent the progression of this disorder.

Other antioxidants

To evaluate the long-term efficacy of a combined antioxidant and mitochondrial enhancement therapy on the bioenergetics and clinical course of 10 patients with FRDA, Hart et alCitation60 performed an open-labeled pilot trial over 47 months with a combined coenzyme Q10 (400 mg/d) and vitamin E (2,100 IU/d) therapy. They reported a significant improvement in cardiac and skeletal muscle mitochondrial bioenergetics as assessed using phosphorus magnetic resonance spectroscopy, and heart function assessed by echocardiographic fraction shortening significantly improved. Although improved fraction shortening was reported, there was no impact upon the degree of cardiac hypertrophy evident before therapy was started.Citation60 These results must be interpreted with caution because of limited patient numbers and the absence of a placebo group.

Another pilot study investigated the potential for high-dose CoQ10/vitamin E therapy to modify clinical progression in FRDA.Citation61 Fifty patients were randomly divided into high-dose or low-dose CoQ10/vitamin E groups. At baseline, serum CoQ10 and vitamin E levels were significantly decreased in patients.Citation61 During the trial, CoQ10 and vitamin E levels significantly increased in both groups. Serum CoQ10 level resulted to be the best predictor of a positive clinical response to CoQ10/vitamin E therapy.Citation61

Conclusive remarks

Overall, because of the lack of controlled studies, the variable doses used and the association with other antioxidant medications, vitamin E has not been appropriately tested in FRDA, and no conclusions can yet be drawn about its safety and efficacy in this disorder.Citation62 More research is needed to identify the role of vitamin E, and of other antioxidant agents, if any, in the management of FRDA and other neurodegenerative disorders.Citation63 A meta-analysis (which included 68 randomized trials with 232,606 healthy participants and patients with various diseases) reported that treatment with β-carotene, vitamin A, and vitamin E may increase all-cause mortality.Citation64 Further study of causes of mortality is needed. These findings contradicts observational studies, claiming that synthetic antioxidant supplements improve health.Citation64 Therefore, more research is needed to establish the real safety of such compounds, including vitamin E.Citation63

In conclusion, available evidence () seems to suggest that patients with FRDA should be treated with idebenone, because it is well tolerated and may reduce cardiac hypertrophy and, at higher doses (up to 45 mg/kg), also improve neurological function, but large controlled clinical trials are still needed.

Table 1 Clinical trials in Friedreich ataxia

Other pharmacological agents could be useful in FRDA patients?

Carnitine and creatine

L-carnitine and creatine are natural compounds that may enhance cellular energy transduction.Citation63 A placebo-controlled triple-phase crossover trial of L-carnitine (3 g/d) and creatine (6.75 g/d) has been performed in 16 patients with genetically confirmed FRDA.Citation65 Ataxia rating scale and echocardio-graphic parameters remained unchanged.Citation65

Erythropoietin

Peripherally administered erythropoietin (EPO), which could increase the amount of frataxin protein at posttranslational level in primary fibroblast cell culturesCitation66 or isolated lymphocytesCitation67 derived from FRDA patients, crosses the blood – brain barrier. Citation68 It may stimulate neurogenesis, neuronal differentiation, and activate brain neurotrophic, antiapoptotic, antioxidant, and anti-inflammatory signaling.Citation68 Boesch et alCitation69 performed an open-label clinical pilot study with recombinant human EPO. Twelve FRDA patients received 5,000 units recombinant human EPO 3 times weekly subcutaneously. Treatment with recombinant human EPO showed a persistent and significant increase in frataxin levels after 8 weeks and a reduction of oxidative stress markers (urinary 8-hydroxydeoxyguanosine and serum peroxides).Citation69 The same research groupCitation70 subsequently reported a 6-month open-label clinical pilot study of safety and efficacy of EPO treatment in 8 FRDA patients (2,000 units thrice a week). Scores in Ataxia Rating Scales improved significantly, with a persistent increase of frataxin levels and a reduction of oxidative stress parameters, but increases in hematocrit requiring phlebotomies occurred in 4 of 8 patients. Safety monitoring with regular blood cell counts and parameters of iron metabolism is a potential limitation of this approach, which to date has not been evaluated with a double-blind controlled study.Citation70

Conclusive remarks

To date, pharmacological approaches other than idebenone and symptomatic therapies are not routinely indicated in patients with FRDA.

Which symptomatic countermeasures are available for FRDA patients?

Symptomatic therapy, which comprises antidiabetic therapy in case of diabetes and cardiologic therapy in case of rhythm abnormalities or heart failure,Citation71 as well as baclofen for spasticity,Citation72 may markedly improve quality of life and prognosis of affected individuals. The levorotatory form of 5-hydroxytryptophan could be able to modify the cerebellar symptoms in patients with FRDA, but the effect is only partial and not clinically major.Citation73 Recent preliminary evidence suggests that riluzole may be potentially effective as symptomatic therapy in diverse forms of cerebellar ataxia.Citation74

Furthermore, in patients with cerebellar ataxia, coordinative training improved motor performance and reduced ataxia symptoms, enabling them to achieve personally meaningful goals in everyday life.Citation75 Rehabilitation therapies usually focus on strategies and compensatory techniques for maintaining or improving abilities to continue to participate in all environmental contexts for as long as possible.Citation76 The benefits of physical exercise programs have been demonstrated for patient with other degenerative disabilities that include ataxia, but at present there is little evidence supporting specific physical therapy interventions that would address impairments or functional concerns in patients with FRDA.Citation76 Patients with FRDA may improve aerobic fitness by participating in stationary cycling for 20–25 minutes at 70%–85% of their maximum heart rate.Citation77 The plan of care should include daily attention to range of motion, including muscle length and soft-tissue extensibility, as well as maintaining independence in mobility.Citation77

Maintaining biomechanical alignment is another important therapeutic consideration.Citation76 Orthopedic problems such as foot deformities and scoliosis are often treated with orthosesCitation78 or surgeryCitation79 and may result in a temporary improvement in function.Citation76 Early intervention for biomechanical changes in the foot significantly improves alignment and thus weight bearing ability and mobility outcomes.Citation79

Conclusion and perspectives

In conclusion, to date the best care for patients with FRDA has not been defined according to evidence-based criteria, and all efforts should be made to obtain solid standards of care, although this goal is difficult to accomplish for a rare disease.Citation80 The first phase 2 trial of idebenone has shown dose-dependent effects on neurological scale scores in children and adolescents with FRDA.Citation57 Large trials are needed to investigate whether all patients with FRDA can benefit from idebenone treatment, regardless of age and disease stage.Citation80

Additional studies with animal models will be essential for an enhanced understanding of the disease pathophysiology and for the development of better therapies.Citation81 Animal and human studies are strongly needed to assess if any of the described new treatment strategies, such as iron-chelating therapies or treatment with EPO or HDAC inhibitors and other gene-based strategies, may translate into an effective therapy for this devastating disorder.

Acknowledgment

This study was supported by Telethon Grant GUP09004.

Disclosure

The authors report no conflicts of interest in this work.

References

  • CampuzanoVMonterminiLMoltoMDFriedreich’s ataxia: autosomal recessive disease caused by an intronic GAA triplet repeat expansionScience19962715254142314278596916
  • FogelBLPerlmanSClinical features and molecular genetics of autosomal recessive cerebellar ataxiasLancet Neurol20076324525717303531
  • PandolfoMFriedreich ataxiaArch Neurol200865101296130318852343
  • Della NaveRGinestroniAGiannelliMBrain structural damage in Friedreich’s ataxiaJ Neurol Neurosurg Psychiatr2008791828517634216
  • FrancaMCJrD’AbreuAYasudaCLA combined voxel-based morphometry and 1H-MRS study in patients with Friedreich’s ataxiaJ Neurol200925671114112019280106
  • PaganiEGinestroniADella NaveRAssessment of brain white matter fiber bundle atrophy in patients with Friedreich ataxiaRadiology2010255388288920501725
  • HsuDTCardiac manifestations of neuromuscular disorders in childrenPaediatr Respir Rev2010111353820113990
  • KippsAAlexanderMColanSDThe longitudinal course of cardiomyopathy in Friedreich’s ataxia during childhoodPediatr Cardiol200930330631018716706
  • La PeanAJeffriesNGrowCRavinaBDi ProsperoNAPredictors of progression in patients with Friedreich ataxiaMov Disord200823142026203218759347
  • CampuzanoVMonterminiLLutzYFrataxin is reduced in Friedreich ataxia patients and is associated with mitochondrial membranesHum Mol Genet1997611177117809302253
  • ShishkinAAVoineaguIMateraRLarge-scale expansions of Friedreich’s ataxia GAA repeats in yeastMol Cell2009351829219595718
  • LeviSRovidaEThe role of iron in mitochondrial functionBiochim Biophys Acta20091790762963618948172
  • CorreiaARWangTCraigEAGomesCMIron-binding activity in yeast frataxin entails a trade off with stability in the alpha1/beta1 acidic ridge regionBiochem J2010426219720320001966
  • PopescuBFPickeringIJGeorgeGNNicholHThe chemical form of mitochondrial iron in Friedreich’s ataxiaJ Inorg Biochem2007101695796617475338
  • HuangMLBeckerEMWhitnallMRahmantoYSPonkaPRichardsonDRElucidation of the mechanism of mitochondrial iron loading in Friedreich’s ataxia by analysis of a mouse mutantProc Natl Acad Sci U S A200910638163811638619805308
  • RichardsonDRHuangMLWhitnallMBeckerEMPonkaPRahmantoYSThe ins and outs of mitochondrial iron-loading: the metabolic defect in Friedreich’s ataxiaJ Mol Med201088432332919997898
  • SparacoMGaetaLMSantorelliFMFriedreich’s ataxia: oxidative stress and cytoskeletal abnormalitiesJ Neurol Sci20092871211111819800081
  • GakhOParkSLiuGMitochondrial iron detoxification is a primary function of frataxin that limits oxidative damage and preserves cell longevityHum Mol Genet200615346747916371422
  • RustinPvon Kleist-RetzowJCChantrel-GroussardKSidiDMunnichARotigAEffect of idebenone on cardiomyopathy in Friedreich’s ataxia: a preliminary studyLancet1999354917747747910465173
  • LodiRCooperJMBradleyJLDeficit of in vivo mitochondrial ATP production in patients with Friedreich ataxiaProc Natl Acad Sci U S A19999620114921149510500204
  • GiacchettiMMonticelliADe BiaseIMitochondrial DNA haplogroups influence the Friedreich’s ataxia phenotypeJ Med Genet200441429329515060107
  • HeidariMMHoushmandMHosseinkhaniSNafissiSScheiber-MojdehkarBKhatamiMAssociation between trinucleotide CAG repeats of the DNA polymerase gene (POLG) with age of onset of Iranian Friedreich’s ataxia patientsNeurol Sci200829648949319043662
  • SeznecHSimonDBoutonCFriedreich ataxia: the oxidative stress paradoxHum Mol Genet200514446347415615771
  • KearneyMOrrellRWFaheyMPandolfoMAntioxidants and other pharmacological treatments for Friedreich ataxiaCochrane Database Syst Rev20094CD00779119821439
  • CoppolaGMarmolinoDLuDFunctional genomic analysis of frataxin deficiency reveals tissue-specific alterations and identifies the PPARgamma pathway as a therapeutic target in Friedreich’s ataxiaHum Mol Genet200918132452246119376812
  • MarmolinoDMantoMAcquavivaFPGC-1alpha down-regulation affects the antioxidant response in Friedreich’s ataxiaPLoS One201054e1002520383327
  • MarmolinoDAcquavivaFPinelliMPPAR-gamma agonist Azelaoyl PAF increases frataxin protein and mRNA expression: new implications for the Friedreich’s ataxia therapyCerebellum2009829810319104905
  • KontoghiorghesGJEfstathiouAKleanthousMMichaelidesYKolnagouARisk/benefit assessment, advantages over other drugs and targeting methods in the use of deferiprone as a pharmaceutical anti-oxidant in iron loading and non iron loading conditionsHemoglobin200933538639719814684
  • RichardsonDRFriedreich’s ataxia: iron chelators that target the mitochondrion as a therapeutic strategy?Expert Opin Investig Drugs2003122235245
  • KakhlonOManningHBreuerWCell functions impaired by frataxin deficiency are restored by drug-mediated iron relocationBlood2008112135219522718796625
  • GoncalvesSPaupeVDassaEPRustinPDeferiprone targets aconitase: implication for Friedreich’s ataxia treatmentBMC Neurol 200882018558000
  • LiKBesseEKHaDKovtunovychGRouaultTAIron-dependent regulation of frataxin expression: implications for treatment of Friedreich ataxiaHum Mol Genet200817152265227318424449
  • JauslinMLWirthTMeierTSchoumacherFA cellular model for Friedreich ataxia reveals small-molecule glutathione peroxidase mimetics as novel treatment strategyHum Mol Genet200211243055306312417527
  • JauslinMLMeierTSmithRAMurphyMPMitochondria-targeted antioxidants protect Friedreich ataxia fibroblasts from endogenous oxidative stress more effectively than untargeted antioxidantsFASEB J200317131972197412923074
  • CochemeHMKelsoGFJamesAMMitochondrial targeting of quinones: therapeutic implicationsMitochondrion20077SupplS94S10217449335
  • SeznecHSimonDMonassierLIdebenone delays the onset of cardiac functional alteration without correction of Fe-S enzymes deficit in a mouse model for Friedreich ataxiaHum Mol Genet200413101017102415028670
  • HebertMDTargeting the gene in Friedreich ataxiaBiochimie20089081131113918206656
  • FlemingJSpinoulasAZhengMPartial correction of sensitivity to oxidant stress in Friedreich ataxia patient fibroblasts by frataxinencoding adeno-associated virus and lentivirus vectorsHum Gene Ther200516894795616076253
  • Gomez-SebastianSGimenez-CassinaADiaz-NidoJLimFWade-MartinsRInfectious delivery and expression of a 135 kb human FRDA genomic DNA locus complements Friedreich’s ataxia deficiency in human cellsMol Ther200715224825417235301
  • LimFPalomoGMMauritzCFunctional recovery in a Friedreich’s ataxia mouse model by frataxin gene transfer using an HSV-1 amplicon vectorMol Ther20071561072107817375064
  • BurnettRMelanderCPuckettJWDNA sequence-specific polyamides alleviate transcription inhibition associated with long GAA. TTC repeats in Friedreich’s ataxiaProc Natl Acad Sci U S A200610331114971150216857735
  • GrantLSunJXuHSubramonySHChairesJBHebertMDRational selection of small molecules that increase transcription through the GAA repeats found in Friedreich’s ataxiaFEBS Lett2006580225399540516989817
  • HermanDJenssenKBurnettRSoragniEPerlmanSLGottesfeldJMHistone deacetylase inhibitors reverse gene silencing in Friedreich‘s ataxiaNat Chem Biol200621055155816921367
  • SarseroJPLiLWardanHSitteKWilliamsonRIoannouPAUpregulation of expression from the FRDA genomic locus for the therapy of Friedreich ataxiaJ Gene Med200351728112516053
  • RaiMSoragniEChouCJTwo new pimelic diphenylamide HDAC inhibitors induce sustained frataxin upregulation in cells from Friedreich’s ataxia patients and in a mouse modelPLoS One201051e882520098685
  • XuCSoragniEChouCJChemical probes identify a role for histone deacetylase 3 in Friedreich’s ataxia gene silencingChem Biol200916998098919778726
  • RaiMSoragniEJenssenKHDAC inhibitors correct frataxin deficiency in a Friedreich ataxia mouse modelPLoS One200834e195818463734
  • Gonzalez-CaboPRosSPalauFFlavin adenine dinucleotide rescues the phenotype of frataxin deficiencyPLoS One201051e887220111601
  • Di ProsperoNASumnerCJPenzakSRRavinaBFischbeckKHTaylorJPSafety, tolerability, and pharmacokinetics of high-dose idebenone in patients with Friedreich ataxiaArch Neurol200764680380817562928
  • MeierTBuyseGIdebenone: an emerging therapy for Friedreich ataxiaJ Neurol2009256Suppl 1253019283347
  • BodmerMVankanPDreierMKutzKWDreweJPharmacokinetics and metabolism of idebenone in healthy male subjectsEur J Clin Pharmacol200965549350119125241
  • SchulzJBDehmerTScholsLOxidative stress in patients with Friedreich ataxiaNeurology200055111719172111113228
  • CooperJMSchapiraAHFriedreich’s ataxia: coenzyme Q10 and vitamin E therapyMitochondrion20077SupplS127S13517485244
  • RustinPRotigAMunnichASidiDHeart hypertrophy and function are improved by idebenone in Friedreich’s ataxiaFree Radic Res200236446746912069112
  • HausseAOAggounYBonnetDIdebenone and reduced cardiac hypertrophy in Friedreich’s ataxiaHeart200287434634911907009
  • MariottiCSolariATortaDMaranoLFiorentiniCDi DonatoSIdebenone treatment in Friedreich patients: one-year-long randomized placebo-controlled trialNeurology200360101676167912771264
  • Di ProsperoNABakerAJeffriesNFischbeckKHNeurological effects of high-dose idebenone in patients with Friedreich’s ataxia: a randomised, placebo-controlled trialLancet Neurol200761087888617826341
  • ArtuchRAracilAMasAFriedreich’s ataxia: idebenone treatment in early stage patientsNeuropediatrics200233419019312368988
  • PinedaMArpaJMonteroRIdebenone treatment in paediatric and adult patients with Friedreich ataxia: long-term follow-upEur J Paediatr Neurol200812647047518234531
  • HartPELodiRRajagopalanBAntioxidant treatment of patients with Friedreich ataxia: four-year follow-upArch Neurol200562462162615824263
  • CooperJMKorliparaLVHartPEBradleyJLSchapiraAHCoenzyme Q10 and vitamin E deficiency in Friedreich’s ataxia: predictor of efficacy of vitamin E and coenzyme Q10 therapyEur J Neurol200815121371137919049556
  • PandolfoMDrug insight: antioxidant therapy in inherited ataxiasNat Clin Pract Neurol200842869618256680
  • OrsucciDFilostoMSicilianoGMancusoMElectron transfer mediators and other metabolites and cofactors in the treatment of mitochondrial dysfunctionNutr Rev200967842743819674340
  • BjelakovicGNikolovaDGluudLLSimonettiRGGluudCMortality in randomized trials of antioxidant supplements for primary and secondary prevention: systematic review and meta-analysisJAMA2007297884285717327526
  • ScholsLZangeJAbeleML-carnitine and creatine in Friedreich’s ataxia. A randomized, placebo-controlled crossover trialJ Neural Transm2005112678979615480852
  • AcquavivaFCastaldoIFillaARecombinant human erythropoietin increases frataxin protein expression without increasing mRNA expressionCerebellum20087336036518581197
  • SturmBStupphannDKaunCRecombinant human erythropoietin: effects on frataxin expression in vitroEur J Clin Invest2005351171171716269021
  • SirenALFasshauerTBartelsCEhrenreichHTherapeutic potential of erythropoietin and its structural or functional variants in the nervous systemNeurotherapeutics20096110812719110203
  • BoeschSSturmBHeringSGoldenbergHPoeweWScheiber-MojdehkarBFriedreich’s ataxia: clinical pilot trial with recombinant human erythropoietinAnn Neurol200762552152417702040
  • BoeschSSturmBHeringSNeurological effects of recombinant human erythropoietin in Friedreich’s ataxia: a clinical pilot trialMov Disord200823131940194418759345
  • FinstererJOverview on visceral manifestations of mitochondrial disordersNeth J Med2006643617116547358
  • BensmailDQuera SalvaMARocheNEffect of intrathecal baclofen on sleep and respiratory function in patients with spasticityNeurology20066781432143617060570
  • TrouillasPSerratriceGLaplaneDLevorotatory form of 5-hydroxytryptophan in Friedreich’s ataxia. Results of a double-blind drug-placebo cooperative studyArch Neurol19955254564607733839
  • RistoriGRomanoSViscontiARiluzole in cerebellar ataxia: a randomized, double-blind, placebo-controlled pilot trialNeurology2010741083984520211908
  • IlgWSynofzikMBrotzDBurkardSGieseMAScholsLIntensive coordinative training improves motor performance in degenerative cerebellar diseaseNeurology200973221823183019864636
  • MaringJRCroarkinEPresentation and progression of Friedreich ataxia and implications for physical therapist examinationPhys Ther200787121687169617911272
  • FillyawMJAdesPAEndurance exercise training in Friedreich ataxiaArch Phys Med Rehabil198970107867882802961
  • GoulipianCBensoussanLVitonJMMilhe-De BovisVRamonJDelarqueAOrthopedic shoes improve gait in Friedreich’s ataxia: a clinical and quantified case studyEur J Phys Rehabil Med2008441939818385634
  • DelatyckiMBHolianACorbenLSurgery for equinovarus deformity in Friedreich’s ataxia improves mobility and independenceClin Orthop Relat Res200543013814115662315
  • SchulzJBBoeschSBurkKDiagnosis and treatment of Friedreich ataxia: a European perspectiveNat Rev Neurol20095422223419347027
  • PuccioHMulticellular models of Friedreich ataxiaJ Neurol2009256Suppl 1182419283346