Topic 2.2. Potential targets for therapies

Return to Chapter Overview

Full Chapter PDF

Full TextAuthorsReferencesMethodology & Date

This chapter of the Clinical Management Guidelines for Friedreich Ataxia and the recommendations and best practice statements contained herein were endorsed by the authors and the Friedreich Ataxia Guidelines Panel in 2022.

Topic Contents

2.2 Potential targets for therapies
2.2.1 Therapies that decrease oxidative stress and enhance mitochondrial function
2.2.2 Anti-inflammatory therapy
2.2.3 Modulators of frataxin-controlled metabolic pathways
2.2.4 Therapies that increase FRDA gene expression
2.2.5 Frataxin replacement, stabilizers or enhancers

Disclaimer / Intended Use / Funding

Disclaimer
The Clinical Management Guidelines for Friedreich ataxia (‘Guidelines’) are protected by copyright owned by the authors who contributed to their development or said authors’ assignees.

These Guidelines are systematically developed evidence statements incorporating data from a comprehensive literature review of the most recent studies available (up to the Guidelines submission date) and reviewed according to the Grading of Recommendations, Assessment Development and Evaluations (GRADE) framework © The Grade Working Group.

Guidelines users must seek out the most recent information that might supersede the diagnostic and treatment recommendations contained within these Guidelines and consider local variations in clinical settings, funding and resources that may impact on the implementation of the recommendations set out in these Guidelines.

The authors of these Guidelines disclaim all liability for the accuracy or completeness of the Guidelines, and disclaim all warranties, express or implied to their incorrect use.

Intended Use
These Guidelines are made available as general information only and do not constitute medical advice. These Guidelines are intended to assist qualified healthcare professionals make informed treatment decisions about the care of individuals with Friedreich ataxia. They are not intended as a sole source of guidance in managing issues related to Friedreich ataxia. Rather, they are designed to assist clinicians by providing an evidence-based framework for decision-making.

These Guidelines are not intended to replace clinical judgment and other approaches to diagnosing and managing problems associated with Friedreich ataxia which may be appropriate in specific circumstances. Ultimately, healthcare professionals must make their own treatment decisions on a case-by-case basis, after consultation with their patients, using their clinical judgment, knowledge and expertise.
Guidelines users must not edit or modify the Guidelines in any way – including removing any branding, acknowledgement, authorship or copyright notice.

Funding
The authors of this document gratefully acknowledge the support of the Friedreich Ataxia Research Alliance (FARA). The views and opinions expressed in the Guidelines are solely those of the authors and do not necessarily reflect the official policy or position of FARA.


2.2 Potential targets for therapies

George Wilmot, Caterina Mariotti, David Lynch, Geneieve Tai and Massimo Pandolfo

The research of therapies that could have clinically meaningful results leading to a cure for the disease is continuing, with new compounds and new clinical trials being designed, commenced and currently ongoing. The research of new strategies is still based on the evaluation of potential therapeutic effects of drugs that are already commercially available and approved for other diseases, as well as new compounds specifically intended for the cure of FRDA and not available for other indications.

2.2.1 Therapies that decrease oxidative stress and enhance mitochondrial function

The pathology of FRDA is characterized by mitochondrial dysfunction and oxidative stress, demonstrated in both cell and animal models of FRDA (6, 14). Respiratory chain dysfunction, accumulation of iron in the mitochondria and impaired antioxidant responses lead to increased production of reactive oxygen species (15). The use of antioxidants has therefore been investigated as a potential therapy for FRDA.

2.2.2 Anti-inflammatory therapy

Inflammation contributes to the pathology of FRDA and has been detected in animal models as well as in tissues of people with FRDA (16, 17). The anti-inflammatory properties of steroids may play a role in altering oxidative damage caused by frataxin deficiency. This hypothesis arose after an improvement in neurological symptoms was reported in an individual with FRDA following corticosteroid treatment (18). Methylprednisolone has thus been explored as a treatment in FRDA (19).

2.2.3 Modulators of frataxin-controlled metabolic pathways

FRDA is caused by the reduced expression of frataxin, a protein found mostly in the mitochondria (20, 21), where it acts as an activator of iron-sulfur (Fe-S) cluster biosynthesis. The resulting Fe-S deficiency impairs the activity of many cellular proteins, including respiratory chain subunits and Krebs cycle enzymes in the mitochondria, and triggers a homeostatic response that increases cellular and mitochondrial iron uptake (22). However, as the Fe-S cluster biosynthetic pathway is impaired, iron eventually accumulates in mitochondria where it may engage in redox reactions generating toxic free radicals, activate signaling pathways leading to neurodegeneration (23), and trigger cell death, in particular by ferroptosis (24).

It has been proposed that frataxin is also involved in various other pathways, including iron metabolism, transport and storage (25), as well as regulation of apoptosis (26). Furthermore, several metabolic pathways are perturbed because of frataxin deficiency, in particular those that control antioxidant responses and mitochondrial biogenesis. Therapies that modulate such pathways include nuclear factor erythroid-derived 2-related factor 2 (Nrf2) activators and peroxisome proliferator activated receptor (PPAR)-γ agonists.

2.2.4 Therapies that increase FRDA gene expression

Approximately 96% of individuals with FRDA have a homozygous mutation consisting of the expansion of GAA trinucleotide repeats within the first intron of the FXN gene (20), leading to the formation of heterochromatin (27). As a result, transcription of FXN mRNA is reduced (28, 29). Agents that counter heterochromatin formation can upregulate FXN mRNA, including histone deacetylase (HDAC) inhibitors (30, 31). Other agents directly boost FXN expression regardless of the presence of expanded GAA repeats. Those clinically tested include erythropoietin and derivatives, Interferon gamma (12, 32).

2.2.5 Frataxin replacement, stabilizers or enhancers

Frataxin replacement therapy has been proposed by pairing synthetic frataxin protein with a delivery system using a protein fragment called a trans-activator of transcription (TAT) to enable frataxin delivery into the mitochondria (33, 34). Another method of frataxin supplementation is through delivery of a normal copy of the FXN gene via gene replacement therapy (35-37).

Gene replacement and editing

Gene replacement therapy is perhaps the most promising in terms of correcting frataxin loss in FRDA, with numerous strategies currently being explored (https://curefa.org/pipeline). FRDA presents as a favorable candidate for gene replacement therapy due to several factors. About 96% of individuals with FRDA have the same single gene mutation which leads to gene silencing and a reduction of the frataxin protein levels. Because individuals with FRDA already produce frataxin, it is less likely that an immune response will be produced. Furthermore, while carriers for FRDA possess one faulty copy of the gene and produce half the normal frataxin levels, these individuals do not exhibit any symptoms, indicating that even a small increase of frataxin has the likelihood to be beneficial.

There are several approaches to gene therapy (38). Adeno-associated viruses (AAV) are viral vectors that do not integrate into the host genome, avoiding genotoxicity. Their DNA persists for a long time in transfected cells as an episome, potentially lifelong. This makes AAV a vector of choice for perennial tissues such as the brain, spinal cord, and heart, which are most affected in FRDA. The potential efficacy of AAV-based gene therapy for FRDA was first demonstrated in a conditional cardiac and skeletal muscle FXN knockout mouse model (Mck-Cre-FxnL3/L mice) that was treated intravenously with adeno-associated virus rh10 vector expressing human FXN, leading to prevention of cardiac disease onset if given early, as well as a complete reversal of cardiomyopathy when given after the development of symptoms (36). In a separate study, a parvalbumin-conditional FXN knockout mouse model (Pvalb cKO) with FXN delivered through an AAV9 vector resulted in a complete reversal of sensory ataxia but not of manifestations of central nervous system (CNS) disease (37).

There are, however, several issues that need to be resolved before AAV-based gene therapy becomes a reality in FRDA. Some naturally occurring AAVs, such as AAV9, can effectively cross the blood-brain barrier (BBB) after systemic administration, but only for a limited time after birth. For this reason, while AAV9-based gene therapy has been effective in treating diseases such as spinal muscular atrophy (SMA), that affects babies, there are ongoing efforts to generate new capsids that can penetrate the CNS in older children and adults after systemic administration (39). However, this may require very high intravenous doses of the vector, which, at least in the case of AAV9 and related capsids, may trigger a severe reaction with liver toxicity and cytokine release. While this reaction could be potentially lethal, it is at least partially preventable with immune suppressive treatment with steroids (40). Furthermore, an inflammatory reaction with neuron loss in the dorsal root ganglia (DRG) has been observed in some animal models (40). This is a particularly worrisome complication in FRDA, where DRG pathology is already present. Acquired immunity to AAVs, a common occurrence in the general population, is another problem, as neutralizing antibodies may inactivate the gene therapy vector and T cells may attack transfected cells presenting capsid fragments on their surface. This is also a major obstacle to re-administer AAV to patients who have previously received it. Overall, these difficulties impose the development of new capsids with improved biodistribution and ability to cross the BBB, as well as of strategies to control innate and acquired immune responses, allowing administration to individuals carrying anti-AAV antibodies and re-administration of a therapeutic vector if needed (41).

Proper control of transgene expression is also necessary. Frataxin expression must reach heterozygous carrier levels at least, but cannot be excessive, as it has demonstrated that very high levels are toxic, causing mitochondrial dysfunction and cardiac toxicity in mouse models (42). This requires a combination of appropriate vector biodistribution and promoter choice.

The possibility of dual routes of administration is an emerging option for what we may consider the first-generation gene therapy for FRDA, while “optimal” vectors are being developed. This approach aims at reaching peripheral organs (heart, pancreas, DRG, peripheral nerves, muscle) via a relatively low dose systemic administration, and the CNS via intrathecal or intraparenchymal administration, targeting key affected structures as the dentate nuclei in the cerebellum.

Other approaches may involve different viral vectors, such as Herpes virus-based, or non-viral vectors such as lipid nanoparticles. These are still in an early-preclinical phase.

Delivery of brain-derived neurotrophic factor (BDNF) is another approach to gene therapy in FRDA (43). BDNF has numerous neuroprotective properties including anti-apoptosis, antioxidation and autophagy suppression (44). The stargazer mouse model with severe cerebellar ataxia exhibited improved ataxia and motor impairment when crossed with mice overexpressing transgenic BDNF (45). In another study, a gene encoding BDNF was delivered via a herpesviral amplicon vector to a knockout mouse model which prevented the onset of cerebellar neuropathology and ataxia (46).  Overexpression is an issue with BDNF as well, having been shown to cause learning and short-term memory impairment (47).

A lack of animal models that accurately depict FRDA is another barrier in the development of gene therapy in FRDA. Conditional knockout mouse models are useful in providing proof-of-concept, but models with a pathologically low systemic frataxin expression, as is the case in the human disease, are still unsatisfactory. A YG8JR mouse model carrying a human FXN gene with 800 GAA repeats has recently been developed and is the most genetically alike to individuals with FRDA. However, the phenotype of this model, as of other GAA repeat expansion-carrying mouse models, appears to be late disease onset and mild disease presentation. There is also a lack of models in larger animals which may be more useful with respect to translation to humans.

David Lynch, MD, PhD
Professor of Neurology and Pediatrics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
Email: lynchd@mail.med.upenn.edu

Caterina Mariotti, MD
Neurologist, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
Email: caterina.mariotti@istituto-besta.it

Massimo Pandolfo, MD
Professor (Clinical), McGill University, Montreal, Quebec, Canada
Email: massimo.pandolfo@mcgill.ca

Geneieve Tai, BBiomedSc(Hons)
Research Assistant, Murdoch Children’s Research Institute, Parkville, Victoria, Australia.
Email: geneieve.tai@mcri.edu.au

George Wilmot, MD, PhD
Associate Professor, Department of Neurology, Emory University, Atlanta, Georgia, USA
Email: gwilmot@emory.edu

1. Parkinson MH, Schulz JB, Giunti P. Co-enzyme Q10 and idebenone use in Friedreich’s ataxia. J Neurochem. 2013;126 Suppl 1:125-41.

2. Hawi A, Heald S, Sciascia T. Use of an adaptive study design in single ascending-dose pharmacokinetics of A0001 (alpha-tocopherylquinone) in healthy male subjects. J Clin Pharmacol. 2012;52(1):65-77.

3. Lynch DR, Willi SM, Wilson RB, Cotticelli MG, Brigatti KW, Deutsch EC, et al. A0001 in Friedreich ataxia: biochemical characterization and effects in a clinical trial. Mov Disord. 2012;27(8):1026-33.

4. Garcia-Gimenez JL, Sanchis-Gomar F, Pallardo FV. Could thiazolidinediones increase the risk of heart failure in Friedreich’s ataxia patients? Mov Disord. 2011;26(5):769-71.

5. Coppola G, Marmolino D, Lu D, Wang Q, Cnop M, Rai M, et al. Functional genomic analysis of frataxin deficiency reveals tissue-specific alterations and identifies the PPARgamma pathway as a therapeutic target in Friedreich’s ataxia. Hum Mol Genet. 2009;18(13):2452-61.

6. Delatycki MB, Camakaris J, Brooks H, Evans-Whipp T, Thorburn DR, Williamson R, et al. Direct evidence that mitochondrial iron accumulation occurs in Friedreich ataxia. Ann Neurol. 1999;45(5):673-5.

7. Pandolfo M, Arpa J, Delatycki MB, Le Quan Sang KH, Mariotti C, Munnich A, et al. Deferiprone in Friedreich ataxia: a 6-month randomized controlled trial. Ann Neurol. 2014;76(4):509-21.

8. Sturm B, Stupphann D, Kaun C, Boesch S, Schranzhofer M, Wojta J, et al. Recombinant human erythropoietin: effects on frataxin expression in vitro. Eur J Clin Invest. 2005;35(11):711-7.

9. Boesch S, Nachbauer W, Mariotti C, Sacca F, Filla A, Klockgether T, et al. Safety and tolerability of carbamylated erythropoietin in Friedreich’s ataxia. Mov Disord. 2014;29(7):935-9.

10. Boesch S, Strum B, Hering S, Goldenberg H, Poewe W, Scheiber-Mojdehkar B. Friedreich’s ataxia: clinical pilot trial with recombinant human erythropoietin. Ann Neurol. 2007;62(5):521-4.

11. Sacca F, Puorro G, Marsili A, Antenora A, Pane C, Casali C, et al. Long-term effect of epoetin alfa on clinical and biochemical markers in friedreich ataxia. Mov Disord. 2016;31(5):734-41.

12. Seyer L, Greeley N, Foerster D, Strawser C, Gelbard S, Dong Y, et al. Open-label pilot study of interferon gamma-1b in Friedreich ataxia. Acta Neurol Scand. 2015;132(1):7-15.

13. Lynch DR, Hauser L, McCormick A, Wells M, Dong YN, McCormack S, et al. Randomized, double-blind, placebo-controlled study of interferon-gamma 1b in Friedreich Ataxia. Ann Clin Transl Neurol. 2019;6(3):546-53.

14. Babcock M, de Silva D, Oaks R, Davis-Kaplan S, Jiralerspong S, Montermini L, et al. Regulation of mitochondrial iron accumulation by Yfh1p, a putative homolog of frataxin. Science. 1997;276(5319):1709-12.

15. Armstrong JS, Khdour O, Hecht S, M. Does oxidative stress contribute to the pathology of Friedreich’s ataxia? A radical question. FASEB J. 2010;24:2152–63.

16. Shen Y, McMackin MZ, Shan Y, Raetz A, David S, Cortopassi G. Frataxin deficiency promotes excess microglial DNA damage and inflammation that Is rescued by PJ34. PLoS One. 2016;11(3):e0151026.

17. Koeppen AH, Ramirez RL, Becker AB, Mazurkiewicz JE. Dorsal root ganglia in Friedreich ataxia: satellite cell proliferation and inflammation. Acta Neuropathol Commun. 2016;4(1):46.

18. Shinnick JE, Isaacs CJ, Vivaldi S, Schadt K, Lynch DR. Friedreich ataxia and nephrotic syndrome: a series of two patients. BMC Neurol. 2016;16:3.

19. Patel M, Schadt K, McCormick A, Isaacs C, Dong YN, Lynch DR. Open-label pilot study of oral methylprednisolone for the treatment of patients with Friedreich ataxia. Muscle Nerve. 2019;60(5):571-5.

20. Campuzano V, Montermini L, Molto MD, Pianese L, Cossee M, Cavalcanti F, et al. Friedreich’s ataxia: autosomal recessive disease caused by an intronic GAA triplet repeat expansion. Science. 1996;271(5254):1423-7.

21. Campuzano V, Montermini L, Lutz Y, Cova L, Hindelang C, Jiralerspong S, et al. Frataxin is reduced in Friedreich ataxia patients and is associated with mitochondrial membranes. Hum Mol Genet. 1997;6(11):1771-80.

22. Martelli A, Schmucker S, Reutenauer L, Mathieu JRR, Peyssonnaux C, Karim Z, et al. Iron regulatory protein 1 sustains mitochondrial iron loading and function in frataxin deficiency. Cell Metab. 2015;21(2):311-23.

23. Chen K, Lin G, Haelterman NA, Ho TS, Li T, Li Z, et al. Loss of Frataxin induces iron toxicity, sphingolipid synthesis, and Pdk1/Mef2 activation, leading to neurodegeneration. Elife. 2016;5.

24. Turchi R, Faraonio R, Lettieri-Barbato D, Aquilano K. An overview of the ferroptosis hallmarks in Friedreich’s ataxia. Biomolecules. 2020;10(11).

25. Llorens JV, Soriano S, Calap-Quintana P, Gonzalez-Cabo P, Molto MD. The role of iron in Friedreich’s ataxia: Insights from studies in human tissues and cellular and animal models. Front Neurosci. 2019;13:75.

26. Wong A, Yang J, Cavadini P, Gellera C, Lonnerdal B, Taroni F, et al. The Friedreich’s ataxia mutation confers cellular sensitivity to oxidant stress which is rescued by chelators of iron and calcium and inhibitors of apoptosis. Hum Mol Genet. 1999;8(3):425-30.

27. Saveliev A, Everett C, Sharpe T, Webster Z, Festenstein R. DNA triplet repeats mediate heterochromatin-protein-1-sensitive variegated gene silencing. Nature. 2003;422(6934):909-13.

28. Gottesfeld JM, Rusche JR, Pandolfo M. Increasing frataxin gene expression with histone deacetylase inhibitors as a therapeutic approach for Friedreich’s ataxia. J Neurochem. 2013;126 Suppl 1:147-54.

29. Silva AM, Brown JM, Buckle VJ, Wade-Martins R, Lufino MM. Expanded GAA repeats impair FXN gene expression and reposition the FXN locus to the nuclear lamina in single cells. Hum Mol Genet. 2015;24(12):3457-71.

30. Soragni E, Xu C, Cooper A, Plasterer HL, Rusche JR, Gottesfeld JM. Evaluation of histone deacetylase inhibitors as therapeutics for neurodegenerative diseases. Methods Mol Biol. 2011;793:495-508.

31. Rai M, Soragni E, Jenssen K, Burnett R, Herman D, Coppola G, et al. HDAC inhibitors correct frataxin deficiency in a Friedreich ataxia mouse model. PLoS One. 2008;3:e1958.

32. Marcotulli C, Fortuni S, Arcuri G, Tomassini B, Leonardi L, Pierelli F, et al. GIFT-1, a phase IIa clinical trial to test the safety and efficacy of IFNgamma administration in FRDA patients. Neurol Sci. 2016;37(3):361-4.

33. Vyas PM, Tomamichel WJ, Pride PM, Babbey CM, Wang Q, Mercier J, et al. A TAT-frataxin fusion protein increases lifespan and cardiac function in a conditional Friedreich’s ataxia mouse model. Hum Mol Genet. 2012;21(6):1230-47.

34. Britti E, Delaspre F, Feldman A, Osborne M, Greif H, Tamarit J, et al. Frataxin-deficient neurons and mice models of Friedreich ataxia are improved by TAT-MTScs-FXN treatment. J Cell Mol Med. 2018;22(2):834-48.

35. Gerard C, Xiao X, Filali M, Coulombe Z, Arsenault M, Couet J, et al. An AAV9 coding for frataxin clearly improved the symptoms and prolonged the life of Friedreich ataxia mouse models. Mol Ther Methods Clin Dev. 2014;1:14044.

36. Perdomini M, Belbellaa B, Monassier L, Reutenauer L, Messaddeq N, Cartier N, et al. Prevention and reversal of severe mitochondrial cardiomyopathy by gene therapy in a mouse model of Friedreich’s ataxia. Nat Med. 2014;20(5):542-7.

37. Piguet F, de Montigny C, Vaucamps N, Reutenauer L, Eisenmann A, Puccio H. Rapid and complete reversal of sensory ataxia by gene therapy in a novel model of Friedreich ataxia. Mol Ther. 2018;26(8):1940-52.

38. Ocana-Santero G, Diaz-Nido J, Herranz-Martin S. Future prospects of gene therapy for Friedreich’s ataxia. Int J Mol Sci. 2021;22(4).

39. Liu D, Zhu M, Zhang Y, Diao Y. Crossing the blood-brain barrier with AAV vectors. Metab Brain Dis. 2021;36(1):45-52.

40. Hinderer C, Katz N, Buza EL, Dyer C, Goode T, Bell P, et al. Severe toxicity in nonhuman primates and piglets following high-dose intravenous administration of an adeno-associated virus vector expressing human SMN. Hum Gene Ther. 2018;29(3):285-98.

41. Muhuri M, Maeda Y, Ma H, Ram S, Fitzgerald KA, Tai PW, et al. Overcoming innate immune barriers that impede AAV gene therapy vectors. J Clin Invest. 2021;131(1).

42. Belbellaa B, Reutenauer L, Messaddeq N, Monassier L, Puccio H. High levels of frataxin overexpression lead to mitochondrial and cardiac toxicity in mouse models. Mol Ther Methods Clin Dev. 2020;19:120-38.

43. Misiorek JO, Schreiber AM, Urbanek-Trzeciak MO, Jazurek-Ciesiolka M, Hauser LA, Lynch DR, et al. A comprehensive transcriptome analysis identifies FXN and BDNF as novel targets of miRNAs in Friedreich’s ataxia patients. Mol Neurobiol. 2020;57(6):2639-53.

44. Chen SD, Wu CL, Hwang WC, Yang DI. More insight into BDNF against neurodegeneration: anti-apoptosis, anti-oxidation, and suppression of autophagy. Int J Mol Sci. 2017;18(3).

45. Meng H, Larson SK, Gao R, Qiao X. BDNF transgene improves ataxic and motor behaviors in stargazer mice. Brain Res. 2007;1160:47-57.

46. Katsu-Jimenez Y, Loria F, Corona JC, Diaz-Nido J. Gene transfer of brain-derived neurotrophic factor (BDNF) prevents neurodegeneration triggered by FXN deficiency. Mol Ther. 2016;24(5):877-89.

47. Cunha C, Angelucci A, D’Antoni A, Dobrossy MD, Dunnett SB, Berardi N, et al. Brain-derived neurotrophic factor (BDNF) overexpression in the forebrain results in learning and memory impairments. Neurobiol Dis. 2009;33(3):358-68.

48. Li L, Voullaire L, Sandi C, Pook MA, Ioannou PA, Delatycki MB, et al. Pharmacological screening using an FXN-EGFP cellular genomic reporter assay for the therapy of Friedreich ataxia. PLoS One. 2013;8(2):e55940.

49. Yiu EM, Tai G, Peverill RE, Lee KJ, Croft KD, Mori TA, et al. An open-label trial in Friedreich ataxia suggests clinical benefit with high-dose resveratrol, without effect on frataxin levels. J Neurol. 2015;262(5):1344-53.

50. Kulkarni SS, Canto C. The molecular targets of resveratrol. Biochim Biophys Acta. 2015;1852(6):1114-23.

51. Alfedi G, Luffarelli R, Condo I, Pedini G, Mannucci L, Massaro DS, et al. Drug repositioning screening identifies etravirine as a potential therapeutic for Friedreich’s ataxia. Mov Disord. 2019;34(3):323-34.

52. Jasoliya M, Sacca F, Sahdeo S, Chedin F, Pane C, Brescia Morra V, et al. Dimethyl fumarate dosing in humans increases frataxin expression: A potential therapy for Friedreich’s Ataxia. PLoS One. 2019;14(6):e0217776.

53. McMackin MZ, Durbin-Johnson B, Napierala M, Napierala JS, Ruiz L, Napoli E, et al. Potential biomarker identification for Friedreich’s ataxia using overlapping gene expression patterns in patient cells and mouse dorsal root ganglion. PLoS One. 2019;14(10):e0223209.

54. Hui CK, Dedkova EN, Montgomery C, Cortopassi G. Dimethyl fumarate dose-dependently increases mitochondrial gene expression and function in muscle and brain of Friedreich’s ataxia model mice. Hum Mol Genet. 2021;29(24):3954-65.

55. Shrader WD, Amagata A, Barnes A, Enns GM, Hinman A, Jankowski O, et al. alpha-Tocotrienol quinone modulates oxidative stress response and the biochemistry of aging. Bioorg Med Chem Lett. 2011;21(12):3693-8.

56. Zesiewicz T, Salemi JL, Perlman S, Sullivan KL, Shaw JD, Huang Y, et al. Double-blind, randomized and controlled trial of EPI-743 in Friedreich’s ataxia. Neurodegener Dis Manag. 2018;8(4):233-42.

57. Zesiewicz T, Sullivan K, Huang Y, Salemi J, Klein M, et al. EPI-743 (alpha-tocotrienol quinone) demonstrates long-term improvement in neurological function and disease progression in Friedreich’s ataxia. Neurology. 2017;88.

58. Sullivan K, Shaw J, Gooch C, Huang Y, Klein M, et al. EPI-743 for Friedreich’s ataxia patients with point mutations (P5.388). Neurology. 2016;86.

59. Lynch DR, Chin MP, Delatycki MB, Subramony SH, Corti M, Hoyle JC, et al. Safety and efficacy of omaveloxolone in Friedreich aAtaxia (MOXIe Study). Ann Neurol. 2021;89(2):212-25.

60. Reata announces that the FDA has asked the company to request a pre-NDA meeting for omaveloxolone for the treatment of Friedreich’s ataxia. 2021 [Available from: https://www.reatapharma.com/investors/news/news-details/2021/Reata-Announces-that-The-FDA-Has-Asked-The-Company-to-Request-a-Pre-NDA-Meeting-for-Omaveloxolone-for-the-Treatment-of-Friedreichs-Ataxia/default.aspx.

61. Cotticelli MG, Crabbe AM, Wilson RB, Shchepinov MS. Insights into the role of oxidative stress in the pathology of Friedreich ataxia using peroxidation resistant polyunsaturated fatty acids. Redox Biol. 2013;1:398-404.

62. Zesiewicz T, Heerinckx F, De Jager R, Omidvar O, Kilpatrick M, Shaw J, et al. Randomized, clinical trial of RT001: Early signals of efficacy in Friedreich’s ataxia. Mov Disord. 2018;33(6):1000-5.

63. Rodriguez-Pascau L, Britti E, Calap-Quintana P, Dong YN, Vergara C, Delaspre F, et al. PPAR gamma agonist leriglitazone improves frataxin-loss impairments in cellular and animal models of Friedreich ataxia. Neurobiol Dis. 2021;148:105162.

64. Meya U, Pina G, Pascual S, Cerrada-Gimenez M, Pizcueta P, Martinell M, et al. A phase 1 study to assess the safety, tolerability, pharmacokinetics, and effects on biomarkers of MIN-102 (Leriglitazone) (4149) Neurology. 2020;94(15 Supplement):4149.

65. Minoryx’s clinical candidate leriglitazone shows clinical benefit in a proof of concept Phase 2 study in Friedreich’s ataxia. 2020 [Available from: https://www.minoryx.com/media/minoryx’s_clinical_candidate_leriglitazone_shows_clinical_benefit_in_a_proof_of_concept_phase_2_study_in_friedreichs_ataxia/.

66. Larimar Therapeutics reports positive topline phase 1 clinical trial data showing dose-dependent increases in frataxin levels in patients with Friedreich’s ataxia. 2021 [Available from: https://www.globenewswire.com/fr/news-release/2021/05/11/2227007/0/en/Larimar-Therapeutics-Reports-Positive-Topline-Phase-1-Clinical-Trial-Data-Showing-Dose-Dependent-Increases-in-Frataxin-Levels-in-Patients-with-Friedreich-s-Ataxia.html.

67. Shen X, Wong J, Prakash TP, Rigo F, Li Y, Napierala M, et al. Progress towards drug discovery for Friedreich’s Ataxia: Identifying synthetic oligonucleotides that more potently activate expression of human frataxin protein. Bioorg Med Chem. 2020;28(11):115472.

68. Erwin GS, Grieshop MP, Ali A, Qi J, Lawlor M, Kumar D, et al. Synthetic transcription elongation factors license transcription across repressive chromatin. Science. 2017;358(6370):1617-22.

69. Kemp KC, Cerminara N, Hares K, Redondo J, Cook AJ, Haynes HR, et al. Cytokine therapy-mediated neuroprotection in a Friedreich’s ataxia mouse model. Ann Neurol. 2017;81(2):212-26.

70. Herman D, Jenssen K, Burnett R, Soragni E, Perlman SL, Gottesfeld JG. Histone deacetylase inhibitors reverse gene silencing in Friedreich’s ataxia. Nat Chem Biol. 2006;2(10):551-8.

71. Soragni E, Miao W, Iudicello M, Jacoby D, De Mercanti S, Clerico M, et al. Epigenetic therapy for Friedreich ataxia. Ann Neurol. 2014;76(4):489-508.

72. Soragni E, Gottesfeld JM. Translating HDAC inhibitors in Friedreich’s ataxia. Expert Opin Orphan Drugs. 2016;4(9):961-70.

73. BioMarin highlights breadth of innovative development pipeline at R&D Day on October 18th in New York. 2017 [Available from: https://investors.biomarin.com/2017-10-18-BioMarin-Highlights-Breadth-of-Innovative-Development-Pipeline-at-R-D-Day-on-October-18th-in-New-York.

74. Vaquero A, Scher M, Erdjument-Bromage H, Tempst P, Serrano L, Reinberg D. SIRT1 regulates the histone methyl-transferase SUV39H1 during heterochromatin formation. Nature. 2007;450(7168):440-4.

75. Libri V, Yandim C, Athanasopoulos S, Loyse N, Natisvili T, Law PP, et al. Epigenetic and neurological effects and safety of high-dose nicotinamide in patients with Friedreich’s ataxia: an exploratory, open-label, dose-escalation study. Lancet. 2014;384(9942):504-13.

76. Reetz K, Hilgers RD, Isfort S, Dohmen M, Didszun C, Fedosov K, et al. Protocol of a randomized, double-blind, placebo-controlled, parallel-group, multicentre study of the efficacy and safety of nicotinamide in patients with Friedreich ataxia (NICOFA). Neurol Res Pract. 2019;1:33.

77. Knip M, Douek IF, Moore WP, Gillmor HA, McLean AE, Bingley PJ, et al. Safety of high-dose nicotinamide: a review. Diabetologia. 2000;43(11):1337-45

These Guidelines are systematically developed evidence statements incorporating data from a comprehensive literature review of the most recent studies available (up to the Guidelines submission date) and reviewed according to the Grading of Recommendations, Assessment Development and Evaluations (GRADE) framework © The Grade Working Group.

This chapter of the Clinical Management Guidelines for Friedreich Ataxia and the recommendations and best practice statements contained herein were endorsed by the authors and the Friedreich Ataxia Guidelines Panel in 2022.

It is our expectation that going forward individual topics can be updated in real-time in response to new evidence versus a re-evaluation and update of all topics simultaneously.