Next Article in Journal
Characterisation of Lipoma-Preferred Partner as a Novel Mechanotransducer in Vascular Smooth Muscle Cells
Next Article in Special Issue
Development of a Polymeric Pharmacological Nanocarrier System as a Potential Therapy for Spinocerebellar Ataxia Type 7
Previous Article in Journal
The Chemical Inhibitors of Endocytosis: From Mechanisms to Potential Clinical Applications
Previous Article in Special Issue
Increased Actin Binding Is a Shared Molecular Consequence of Numerous SCA5 Mutations in β-III-Spectrin
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Inborn Errors of Metabolism with Ataxia: Current and Future Treatment Options

1
Department of Neurology, University Hospital Bern (Inselspital) and University of Bern, 3010 Bern, Switzerland
2
Center for Rare Diseases, University Hospital Bern (Inselspital) and University of Bern, 3010 Bern, Switzerland
3
Division of Pediatric Endocrinology, Diabetes and Metabolism, Department of Paediatrics, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland
4
Institute of Clinical Chemistry, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland
*
Author to whom correspondence should be addressed.
Cells 2023, 12(18), 2314; https://doi.org/10.3390/cells12182314
Submission received: 15 August 2023 / Revised: 9 September 2023 / Accepted: 15 September 2023 / Published: 19 September 2023
(This article belongs to the Special Issue Emerging Therapies for Hereditary Ataxia)

Abstract

:
A number of hereditary ataxias are caused by inborn errors of metabolism (IEM), most of which are highly heterogeneous in their clinical presentation. Prompt diagnosis is important because disease-specific therapies may be available. In this review, we offer a comprehensive overview of metabolic ataxias summarized by disease, highlighting novel clinical trials and emerging therapies with a particular emphasis on first-in-human gene therapies. We present disease-specific treatments if they exist and review the current evidence for symptomatic treatments of these highly heterogeneous diseases (where cerebellar ataxia is part of their phenotype) that aim to improve the disease burden and enhance quality of life. In general, a multimodal and holistic approach to the treatment of cerebellar ataxia, irrespective of etiology, is necessary to offer the best medical care. Physical therapy and speech and occupational therapy are obligatory. Genetic counseling is essential for making informed decisions about family planning.

1. Introduction

Hereditary (or inherited) cerebellar ataxias are a heterogeneous group of individually rare, progressive, and neurological disorders that affect the cerebellum and the cerebellar network, the parts of the brain responsible for coordinating movement and balance. Ataxias are characterized by an unsteady gait leading to recurrent falls, clumsiness, difficulty with coordination, slurred speech, and impaired fine motor skills. Neuro-ophthalmological and -otological manifestations can also be a component of and/or a factor leading to ataxia given the common infratentorial pathways’ and networks’ dysfunction. Ataxias can be caused by inborn errors of metabolism (IEM) which have mostly autosomal recessive, but sometimes X-linked or mitochondrial, inheritance.
Considering that the impaired metabolic pathways in IEM have an impact on numerous functions, cerebellar ataxia is only one component of the clinical picture, even though it may be the leading symptom. Vulnerabilities of specific neuronal populations in the central nervous system (CNS) can lead to disruptions in functional networks causing specific but often multifaceted patterns of neurological deficits along with psychiatric and systemic manifestations. Ataxia is the most common movement disorder in IEM. It occurs in various combinations with other movement disorders. For example, defects of intermediary metabolism are often accompanied by tremors, whereas IEM of complex molecules are more often present with dystonia. Based on the temporal pattern of the occurrence of symptoms, we can distinguish between intermittent metabolic ataxias, occurring due to the worsening of the underlying metabolic abnormality (i.e., metabolic decompensation, commonly occurring in cases of IEM of the intermediary metabolism), and the chronic static or progressive metabolic ataxias, which are usually caused by deficiencies of enzymes or transporters involved in the synthesis or breakdown of complex molecules, leading to chronic brain cell dysfunction and neurodegeneration, e.g., by intracellular storage. Ataxia, as a leading symptom, often occurs in milder defects, including adult-onset forms of IEM [1].
Currently, there is no cure for hereditary cerebellar ataxias, representing a significant unmet medical need.
Thus, it is of paramount importance to promptly diagnose an underlying IEM in order to prevent potentially irreversible damage to the CNS caused by metabolic decompensations, which often occur in disorders of intermediary metabolism. Currently, the general therapeutic approach aims to manage symptoms, prevent or slow down disease progression (by introducing disease-specific treatments, if available), and improve quality of life. Recently, promising gene therapy trials have been initiated for some of the IEM, e.g., first-in-human [2] trials for GM2-Gangliosidoses or metachromatic leukodystrophy [3,4], in the latter showing a remarkable long-term improvement in neurological state compared to natural history data. Preliminary experimental gene therapies for some IEM have been tested in animal studies [5]. Some IEM have a very specific pathomechanism. Thus, targeted treatments may lead to either stabilization/slowing down of the disease progression or to diminished signs and symptoms of the disease. A few general principles with regard to IEM can be formulated:
  • The earlier the disease starts, the more severe it generally is, and the severity of the disease often correlates with the residual activity of the affected gene product (e.g., enzyme);
  • Emergency treatment for acute decompensations of IEM of the intermediary metabolism generally includes a non-specific anticatabolic treatment with intravenous administration of high glucose with insulin, if needed, as well as temporary reduction or stopping of nutrition combined with more specific detoxification measures as indicated, such as ammonia scavengers in case of hyperammonemia (see disease-group-specific guidelines);
  • Regarding the general symptomatic therapeutic principles, anti-seizure medication, together with antipsychotics, antidepressants, and muscle-relaxing agents, such as baclofen or tizanidine or in case of focal/segmental spasticity/dystonia treatment with botulotoxin should be used to relieve the burden of multiple disease symptoms.
Some metabolic ataxias can be diagnosed by biochemical screenings, either as part of a standardized newborn screening or once the neurological abnormalities are first noticed. For IEM without biochemical markers, first-line diagnosis is usually performed by genetic testing, which may be impeded by the absence of variants, the presence of variants of unknown significance, or other inconclusive findings. Functional investigations remain important in the work-up of undiagnosed patients. For example, pyruvate dehydrogenase deficiency can be corroborated by relatively simple biochemical assays on cultured fibroblasts or clinically with a glucose loading test.
Here, we review the most important grouped or individual IEM that may present with ataxia as the leading or accompanying disease manifestation. We describe the clinical picture and the current and potential future treatment options with a focus on lysosomal storage disorders. Table 1 provides a summary of the main points.

2. Disorders of Amino Acid Metabolism

2.1. Organic Acidurias

The organic acidurias, the propionic (PA), methylmalonic (MMA) and isovaleric (IVA) acidemias are caused by defects in the catabolism of branched-chain amino acids (BCAA). They belong to the most common causes of intermittent ataxias. Organic acidurias belong to the intoxication type of IEM. Untreated, they can cause brain damage in many ways, often through impairing brain energy metabolism [22]. Along with episodes of ataxia, symptoms include developmental delay and cognitive impairment, seizures, episodic vomiting, and lethargy. Other organic acidurias can cause chronic ataxia, e.g., mevalonic aciduria can present as chronic progressive ataxia, whereas non-progressive metabolic ataxia may be due to SSADH (succinic semialdehyde dehydrogenase) deficiency or L-2-hydroxy-glutaric aciduria.

Isovaleric (ORPHA:33 and OMIM: 243500), Propionic (ORPHA:35 and OMIM: 606054), and Methylmalonic Acidemias (ORPHA:289916 and OMIM: 606054)

Isovaleric Acidemia (IVA) is an autosomal recessive disorder of leucine catabolism. It is caused by mutations in the isovaleryl-CoA dehydrogenase (IVD) gene, leading to an accumulation of derivatives of isovaleryl-CoA, including isovaleryl (C5)-carnitine, which is assessed as part of the newborn screening in some countries. IVA can cause delayed psychomotor and cognitive development in some cases [23]. Only a subset of patients seem to suffer from neurological and/or cognitive symptoms [23]. Infants with IVA can develop episodes of metabolic acidosis, which are rather rare after nine years of age [24]. There are three phenotypes of IVA: (1) the symptomatic “acute neonatal” form typically presents with the first acute catabolic episode at less than a week of age, with potentially life-threatening metabolic acidosis, a characteristic odour of “sweaty feet”, lethargy, dehydration, vomiting and sometimes impaired consciousness and seizures; (2) the symptomatic “chronic intermittent” form presenting later in childhood, and (3) the mostly asymptomatic “mild” form, caused by a common missense mutation.
PA and MMA are due to defects in the breakdown of isoleucine and valine. Very similar to IVA, they present as either acute neonatal, chronic progressive or late-onset intermittent forms. Diagnosis by urinary organic acid analysis and genetic confirmation, as well as treatments are analogous to IVA as well.
Therapy: Protein-restricted diet, specific amino-acid supplementation, vitamin and oligoelement supplementation. Medication includes L-carnitine, +/− glycine in the case of IVA, or +/− sodium benzoate, and intermittent antibiotics in the case of PA and MMA. Avoidance of catabolic states, including emergency treatment in case of intercurrent illness and other situations with risk of decompensation is highly necessary. Consensus guidelines for the diagnosis, treatment and follow-up have been published [25].

2.2. Maple Syrup Urine Disease (ORPHA:268145 and OMIM: 248600)

Maple syrup urine disease is an autosomal recessive disorder of the branched-chain α-ketoacid dehydrogenase (BCKAD) complex, caused by mutations in the BCKDHA, BCKDHB, DBT, or DLD genes, which code for its catalytic subunits. These mutations result in elevated branched-chain amino acids (BCAA) in plasma, α-ketoacids in urine, and increased production of the pathognomonic disease marker, alloisoleucine [26]. Urine and other body fluids have a characteristic, maple syrup-like odour. Three distinct clinical phenotypes are described. Classic MSUD present soon after birth and is severe and potentially fatal. The intermediate and intermittent forms can be overlapping, presenting at any time of life, with stress- or infection-related decompensations, however adult-onset is rather rare. The clinical presentation of MSUD depends on the BCKAD residual activity, the leucine tolerance and the metabolic response to illness. Movement disorders, such as characteristic “bicycling” movements in neonates, dystonia, spasticity, choreoathethosis, however also cerebellar ataxia may be present, along with cognitive impairment, hyperactivity, sleep disturbance and hallucinations [26]. Ocular motor impairment can occur, varying from up-gaze supranuclear palsy and adduction deficit to absence of voluntary eye movements with absent vestibulo-ocular reflexes [27,28].
Therapy: Low-protein diet with restriction of leucine, isoleucine, and valine. Thiamine supplementation in thiamine-responsive MSUD (pathogenic variants in DBT). Phenylbutyrate acts on the kinase of the BCKAD and could become an additional treatment option in MSUD [29] (see also NCT01529060). During periods of protein catabolism, such as fever, infections, exercise, trauma, or surgery, individuals with MSUD can develop neurological deterioration, which may manifest as intermittent ataxia, due to acute leucine intoxication. Thus, emergency management is essential. Patients with severe disease may need liver transplantation.

2.3. Hartnup Disease (ORPHA:2116 and OMIM: 234500)

Hartnup disease is caused by a deficiency of the sodium-dependent neutral amino acid transporter in the proximal kidney tubules and jejunum, resulting from a mutation in the solute carrier family 6 member 19 (SLC6A19) gene [30,31]. Biochemically, Hartnup disorder is diagnosed via a corresponding amino acid excretion pattern [32]. The ensuing tryptophan deficiency leads to secondary niacin and serotonin deficiencies, thought to be responsible for the pellagra-like skin and the neurological manifestations, including ataxia and neuropsychiatric symptoms. However, most patients are asymptomatic (i.e., low penetrance).
Therapy: Supplementation with niacin (nicotinamide) or tryptophan-rich diet in order to correct the secondary deficiency. Symptomatic patients should strictly protect themselves from sun exposure and avoid photosensitizing drugs.

3. Lysosomal Storage Diseases

Disorders of lysosomes (LSD) and lysosome-related organelles (LROs) can be subclassified according to the biochemical type of stored material (e.g., into sphingolipidoses, mucopolysaccharidoses, glycoproteinoses, and neutral lipid storage disorders), the integral membrane proteins, post-translational modification, or lipofuscin accumulation.

3.1. Gaucher Disease (ORPHA:355)

Gaucher disease (GD) is a sphingolipidosis caused by mutations in the GBA gene, which encodes the enzyme glucocerebrosidase, leading to accumulation of glucosylceramide and its deacylated form, glucosylsphingosine. Even though there is a continuum of signs and symptoms in reality, three phenotypes have been distinguished historically: non-neuronopathic (GD1), acute neuronopathic with infantile onset (GD2), and chronic neuronopathic (GD3). In GD2, early signs in affected children are abnormal eye movements, including horizontal gaze palsy and slow saccades, saccadic initiation problems, and strabismus. Furthermore, early slowing of development followed by regression and severe developmental retardation, as well as visceral signs with hepatosplenomegaly, are prominent. GD3 has a later-onset and commonly presents with cerebellar ataxia and dystonia. Patients may develop (myoclonus) epilepsy that might be severe and lead to neurological deterioration. Patients also show a horizontal supranuclear saccade or complete gaze palsy that is compensated for by head movements, so-called head thrusts, indicating the brainstem involvement [33]. Vestibulo-ocular responses are also impaired [33,34].
Therapy: Intravenous enzyme replacement therapy (ERT) with recombinant glucocerebrosidase is available for visceral manifestations in all forms. However, its use in the severe GD2 form is considered futile. Eliglustat is the first-line substrate reduction therapy (SRT) in adult GD1 patients, showing comparable efficacy to ERT [35]. Prior to the use of eliglustat [36], the pharmacogenomics of CYP2D6, the primary metabolizing enzyme of eliglustat needs to be clarified in all patients in order to determine the adequate dosing. Miglustat is a second-line alternative SRT for adult patients. The potential advantage of these small molecules is their penetrance into the CNS, in contrast to ERT. Various investigational compounds directed at the CNS (including other SRT, arimoclomol, ambroxol, and others) are currently evaluated in phase 1–3 clinical trials. Arimoclomol [37] is thought to act by enhancing the endogenous chaperone system via the HSP70 pathway. Ambroxol [38], an over-the-counter sold mucolytic drug, acts as a pharmacological chaperone. Both these drugs penetrate easily into the central nervous system and could therefore potentially improve cerebral complications of the disease. In addition, an AAV-based gene therapy, so far only for the visceral form (GD1), is currently recruiting (NCT05324943).

3.2. GM2-Gangliosidoses (Tay–Sachs and Sandhoff Disease) (ORPHA:845 and OMIM: 272800)

The GM2-gangliosidoses (vGM2), namely Tay–Sachs disease (TSD), Sandhoff disease (SD), and AB variant, are caused by mutations in the HEXA, HEXB, or GM2A genes, respectively, which code for the α- or β-subunits of β-hexosaminidase or the GM2 activator protein. They are clinically indistinguishable from one another (but certain subtle visceral and skeletal features only occur in SD) [39]. The GM2-gangliosidoses can be divided into infantile-, juvenile- and adult-onset forms. Phenotypically, juvenile- and adult-onset forms feature cerebellar ataxia combined with the involvement of a second motoneuron, along with slowly developing intellectual disability and psychiatric manifestations including bipolar disorder or psychosis. The diagnostic standard is an enzymatic assay of β-hexosaminidase activities (usually carried out in serum and/or peripheral white blood cells) followed by genetic confirmation. In a mouse model of SD disease, a recombinant AAV-α or AAV-β was injected, which led to improved survival of these mice only if the treatment was initiated pre-symptomatically or very early in the disease manifestations [40].
Therapy: Currently, there is no approved treatment for GM2. N-acetyl-L-leucine (ALL) has shown symptomatic benefits in motor domains, quality of life in a positive rater-blinded clinical trial, and in a case report [21,41]. Additionally, a disease-modifying, neuroprotective effect was demonstrated in animal models [42,43]. A first-in-human study of TSHA-101 gene therapy for the treatment of infantile-onset GM2 is ongoing in Kingston, Ontario, Kanada (NCT04798235) [2], where three infants receive the AAV9 viral vector containing HEXA and HEXB genes intrathecally.

3.3. Metachromatic Leukodystrophy (ORPHA:512)

Metachromatic leukodystrophy (MLD) is caused by mutations in the ARSA gene, leading to a deficiency of the enzyme arylsulfatase A (ARSA). Rarely, it can be related to mutations in the prosaposin gene PSAP, leading to a deficiency of ARSA activator protein saposin B (SAP-B) [44] that normally stimulates the degradation of sulfatides by ARSA. ARSA deficiency leads to sulfatide storage in microglia, oligodendrocytes, and Schwann cells [45], leading to demyelination because sulfatides are the major component of the myelin membrane. Neuronal sulfatide storage also leads to neurodegeneration and neuroinflammation. The early-onset MLD form is characterized by rapid progression leading to death within the first decade of life. The late-infantile form is the most severe as all patients are tetraplegic and in a vegetative state by the end of the second year [46]. The juvenile-onset disease is heterogeneous, manifesting with cerebellar ataxia, gait disturbance, upper motor neuron signs, cognitive disability, behavioral difficulties, and peripheral neuropathy [47]. If seizures occur, the disease is fatal within six years of onset. A milder form or MLD, usually adult-onset, presents with cognitive decline as the first symptom [48]. It is characterized by dementia, behavioral changes, and psychiatric manifestations [49]. The typical brain MRI shows a tigroid (striped) pattern of white matter T2-weighted hyperintensities in ~70% of cases, over time accompanied by cortical atrophy [24].
Therapy: Allogenic hematopoietic stem cell transplantation (HSCT) is the first-line treatment option in patients with no or early MLD disease, treating CNS manifestations but not peripheral neuropathy [24,25,26]. Ex vivo gene therapy with HSCT (atidarsagene autotemcel or arsa-cel) [50] for early-onset MLD has been facilitated and approved by the European Medicines Agency (EMA) in December 2020 [51]. A lentiviral vector is used to transfer a functional ARSA gene into autologous HSCs [3]. The gene-corrected HSCs (arsa-cel) are then intravenously transferred back into patients, enabling engraftment of the transduced cells with elevated levels of ARSA enzyme activity. The two-years long-term trial [4], including 26 surviving children with pre- or early symptomatic late-infantile or early-juvenile MLD, showed improved ambulation and either normal development or stabilization of motor skills overall. Their cognitive development was normal with absent or postponed brain white matter and cortical involvement. Patients with late-infantile onset showed improved peripheral nerve conduction velocity. The benefits of arsa-cel treatment were most apparent in patients who were pre-symptomatic when treatment started. A phase I/II, open-label, and monocentric study of direct intracranial administration of a replication-deficient adeno-associated virus gene transfer vector serotype rh.10 expressing the human ARSA cDNA to children with MLD is ongoing (NCT01801709) [52].

3.4. Neuronal Ceroid Lipofuscinoses (NCLs) (ORPHA:216)

The neuronal ceroid lipofuscinoses (NCLs) are LSDs characterized by progressive degeneration of the brain and frequently of the retina with intracellular accumulation of autofluorescent lipopigment [53]. NCL genes are expressed in various tissues but the disease manifests mainly as CNS dysfunction. Early affected brain structures are the thalamus and cerebellum [54]. Five of the NCL variants are caused by defects in soluble lysosomal proteins: CLN1 disease due to palmitoyl protein thioesterase-1 (PPT1) deficiency, CLN2 disease due to tripeptidyl peptidase-1 (TPP1) deficiency, CLN5 disease due to CLN5 protein deficiency, CLN10 disease due to cathepsin D deficiency, and CLN13 due to cathepsin F deficiency. All other NCL variants are due to defects in lysosomal endoplasmatic reticulum or cytoplasmic vesicular transmembrane proteins [55]. Genetically, at least 14 different forms of NCL have been found and assigned a ”CLNx” number. All NCLs, except CLN4 disease, are recessive disorders. Individually, NCLs are rare conditions, yet collectively, they are the most frequent childhood neurodegenerative condition [53]. Clinical manifestations include vision loss, dementia, epilepsy, movement disorders, and storage of ceroid lipofuscin in neurons. NCLs differ in age of onset, clinical features, and rate of progression [56]. In CLN1 disease, there is a rapid, prematurely fatal regression between 6 and 24 months old, characterized by seizures, myoclonus, ataxia, and visual failure, followed by severe spasticity and a decreased level of consciousness [57]. The adult-onset form begins with cognitive decline and depression, with cerebellar ataxia, parkinsonism, and vision loss setting in later [58]. CLN2 disease is associated with the classic late infantile-onset form of NCL [59], with onset between two and four years of age. Typical symptoms include a speech delay, developmental plateau, epilepsy, polymorphic seizures, and over time ataxia, refractory nonepileptic myoclonus, and spastic quadriparesis. Similar to CLN1 disease, children with CLN2 onset past four years of age have a milder course with more prominent ataxia and less epilepsy [60]. CLN3 is a juvenile-onset form, encompassing vision loss, cognitive decline, behavioral problems, epilepsy, and parkinsonism [61]. Stuttering dysarthria is characteristic and develops usually after ten years of age. Ataxia is also a symptom of CLN4, CLN5, CLN6, CLN10, and CLN11. Myoclonus can be observed in CLN4 and CLN7 as well. CLN12, also known as Kufor-Rakeb syndrome (PARK9), is caused by loss-of-function variants in the predominantly neuronal P-type ATPase gene, ATP13A2. CLN12 presents with parkinsonian features of a mask-like face, rigidity, and bradykinesia but without tremor. Spasticity, supranuclear upgaze palsy, and dementia are symptoms of most affected patients [62]. The preferred diagnostic method is genetic testing with the possible addition of brain imaging, which can show cerebellar atrophy.
Therapy: In general, supportive care paired with pharmacological strategies. For epilepsy, antiseizure medications (such as valproate, lamotrigine, topiramate, and levetiracetam) are administered although epilepsy due to NCL is often resistant to treatment [63]. Baclofen can help against spasticity but movement disorders in NCLs can be difficult to treat because of the complexity of movement disorder combinations and the treatment-refractory symptoms. Disease-modifying therapy for CLN2 for patients aged three and older is recombinant human cerliponase alfa, the proenzyme form of human TPP1, and the enzyme deficient in CLN2, administered into the cerebrospinal fluid via the lateral ventricles [64].

3.5. Niemann-Pick Type A and B (ORPHA:618899)

Niemann-Pick types A and B are caused by deficient activity of the enzyme acid sphingomyelinase (ASM) [65]. All patients with types A and B Niemann-Pick disease have mutations in the gene encoding ASM (SMPD1) and thus the disease is more accurately referred to as ASM deficiency (ASMD). Patients suffering from type A ASMD present with failure to thrive in the neonatal period, hypotonia, and failure to reach developmental milestones. They rarely live past two or three years of age. Patients with type B have hepatosplenomegaly and pathologic alterations of their lungs but usually little to no neurological involvement. In patients with type B, the age of onset and rate of disease progression varies and they frequently live into adulthood. Serum triglycerides and LDL-cholesterol are commonly elevated, while HDL-cholesterol is low. Elevated oxysterols may provide an important clue for diagnosis (especially C-triol), even though somewhat less consistent than in type C ([66] and see below). Variant phenotypes feature cerebellar ataxia and neurodevelopmental delay leading to learning difficulties with more severe systemic manifestations [67,68]. Patients carrying the Q294K variant have an increased risk of false negative diagnosis. Pseudo-normal results are obtained from ASM assays employing fluorometric substrates, leading to substantial under-diagnosis in patients with the Q294K mutation [69]. Therefore, best practice guidelines recommend the use of the tandem mass spectrometry ASM assay, which provides a reliable diagnosis in Q294K-dependent cases.
Therapy: Recombinant human ASM (olipudase alfa) [70,71], based on the results of phase 1 and within-patient dose escalation phase 1b clinical trials, is approved by both the US Food and Drug Administration (FDA) and the European Medicines Agency (EMA). Its approval in Switzerland is underway (Xenpozyme®). In children with chronic ASMD, olipudase alfa was well-tolerated and showed significant, comprehensive improvements in disease pathology across a range of clinically relevant endpoints [72]. This finding was confirmed in the phase 2/3, randomized, double-blinded, placebo-controlled, and repeat-dose study (NCT02004691) [73].

3.6. Niemann-Pick Type C (ORPHA:646)

Niemann-Pick disease type C (NPC) is caused by mutations in the NPC1 or NPC2 genes, leading to an impaired intracellular lipid transport with its storage in lysosomes and late endosomes [74,75]. The accumulated cholesterol undergoes non-enzymatic oxidation, leading to the formation of oxysterols [76]. Oxysterols are used as blood-based biomarkers of NPC, together with certain plasma bile acids [77] (e.g., 3β,5α,6β-trihydroxycholanic acid) and certain lysosphingolipids [78] (e.g., lyso-SM-509), replacing filipin staining as first-line diagnostics.
NPC disease is highly heterogeneous, comprising systemic, neurological, and psychiatric signs [79]. Early- and late-infantile forms of NPC are present with severe hepatosplenomegaly, cataplexy, epilepsy, and a regress of already-gained neurological skills. These forms are rapidly progressive and prematurely fatal. In comparison, the juvenile and adolescent forms of NPC progress more slowly and present with cerebellar ataxia, dysarthrophonia, dystonia, cognitive decline, psychiatric involvement, vertical supranuclear saccade (VSSP), gaze palsy (VSGP), and epilepsy [17]. VSSP and VSGP are commonly present in infantile cases and serve as a diagnostic clue [80,81,82]. Children with mutations in the NPC2 gene often develop respiratory insufficiency due to pulmonary alveolar lipoproteins.
Therapy: A holistic and multidisciplinary approach, combining disease-modifying, symptomatic treatments, and non-pharmacological approaches needs to be offered, with the aim to counteract the ongoing neurodegeneration/neuroinflammation and to improve the patients’ quality of life. Miglustat (OGT 918, N-butyl-deoxynojirimycin) is the only disease-modifying treatment for NPC [16], approved by the EMA, Health Canada, and regulatory authorities of other countries (Japan) but not the FDA. It is an iminosugar that crosses the blood brain barrier (BBB) and leads primarily to a substrate reduction. However, miglustat may not be used for patients who lack neurological symptoms or for those with an advanced disease state. Miglustat inhibits glucosylceramide synthase, which is needed in the initial stages of glycosphingolipid synthesis, also acting as a non-lysosomal glucosylcerebrosidase (Gba2) inhibitor. The initial clinical trial for NPC was positive [16], followed by several positive longitudinal cohort studies over 2–8 years assessing the neurological state of patients with NPC [83,84,85,86]. The less severely affected juvenile- and adult-onset patients tend to benefit more than the infantile-onset population. The VSGP/VSSP [80,81] also improves or at least stabilizes under treatment [83], as well as dysphagia [87,88].
One of the newly developed, but not yet approved drugs for NPC is N-acetyl-L-leucine (ALL) [89]. It crosses the BBB. It has beneficial effects not only on motor but also on disease-specific signs and symptoms and on the quality of life of patients with NPC without serious adverse events [18,19,90]. Studies in the Npc1−/−/ mouse model have shown both symptomatic and disease-modifying effects of ALL [42]. A multinational, Phase III, double-blind, randomized, placebo-controlled, crossover trial with ALL for NPC has been completed but the results have not been made public, yet (NCT05163288) [91].
Other substances are in development for NPC, such as chaperone-treatment with arimoclomol [92], AZ-3102 [79], efavirenz [93] or cyclodextrin [94], and are at different development stages. A phase 2, 12-week study evaluating the safety, tolerability, pharmacokinetics, and pharmacodynamics of oral AZ-3102 in patients with NPC or GM2 gangliosidosis (see above) has just been initiated (NCT05758922). The aim of this short study is to determine the clearance of AZ-3102 from the body and to identify the target dose for planned phase 3 studies.

3.7. Sialidosis (ORPHA:309294)

Sialidosis or neuraminidase deficiency (mucolipidosis I) is an autosomal recessive disorder caused by a defect in the enzyme sialidase (neuraminidase). Two main clinical variants have been described that may be accompanied by cerebellar ataxia. Sialidosis type I begins in adulthood, the primary features being myoclonic epilepsy and bilateral macular cherry-red spots. Sialidosis type II appears in childhood, comprising skeletal dysplasia, intellectual disability, and hepatosplenomegaly.
Therapy: There is no ERT for this condition; the treatment is limited to supportive care and symptomatic relief. Myoclonic epilepsy is often pharmacoresistant, partially responding/not responding to a ketogenic diet, further complicating the management of this disease with worsening cognition [95]. Recently, perampanel has been suggested as efficacious for treating myoclonic epilepsy in this condition [96], as it is in other progressive myoclonic epilepsies, such as Lafora disease [97], Unverricht-Lundborg disease [98], and dentatorubral-pallidoluysian atrophy [99]. Perampanel is a non-competitive, selective α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) glutamate receptor antagonist. AMPA receptors may play a pivotal role in the pathophysiology of epilepsy. Symptomatic treatment of cerebellar ataxia, if present, is indicated.

4. Disorders of Carbohydrate Metabolism

4.1. Galactosemia (ORPHA:79239 and OMIM: 230400)

Classic galactosemia (CG) is caused by mutations in the GALT gene, leading to a profound deficiency of galactose-1-phosphate uridylyltransferase (GALT), the central enzyme of galactose metabolism. GALT deficiency leads to an inability to metabolize galactose. Due to the high galactose content in mammalian milk, newborns with CG present with acute intoxication syndrome, a life-threatening condition. Thus, CG is included in the newborn screening programs of several countries. For the diagnosis, GALT enzyme activity is measured in red blood cells (absent or significantly decreased), and/or GALT gene analysis is performed [100]. Individuals with CG develop cognitive impairments and motor difficulties in 18–45% of cases [100,101,102,103]. They range from mild positional tremors to pronounced cerebellar ataxia, myoclonus, dystonia, and ocular motor abnormalities [104,105,106]. Neurological symptoms are more frequently reported in adult age; however, the incidence of subtle neurological signs in pediatric patients may have been underestimated in the past.
Therapy: A life-long galactose-restricted diet eliminating sources of lactose and galactose from dairy products but permitting galactose from non-milk sources that contribute minimal dietary galactose is required [100]. All types of fruits and vegetables, unfermented soy-based products, mature cheeses (with galactose content < 25 mg/100 g), and the food additives sodium or calcium caseinate are permitted in the CG-specific diet. Although higher in galactose, all fermented soy-based products can be allowed in small quantities as part of the diet [100]. Speech therapy, cognitive training, and treatment of the hypogonadotropic hypogonadism and primary ovarian insufficiency should be initiated if needed. Motor functions should be assessed and physical therapy should be considered if needed. However, CG-specific guidelines for the treatment of motor abnormalities are still lacking. Trihexylphenidyl has been used to treat tremors [104].

4.2. GLUT1 Deficiency (ORPHA:71277 and OMIM: 606777)

Impaired facilitated diffusion of glucose across the BBB by the glucose transporter family member 1 (GLUT1) leads to the autosomal dominant Glut1 deficiency syndrome with acute and/or chronic brain energy failure. Depending on the severity of the defect, clinical manifestations range from early-onset epileptic encephalopathy and psychomotor retardation with microcephaly to milder forms with fluctuating movement disorders (including ataxia, tremor, dystonia, chorea and/or spasticity, in variable combinations, or late-onset absence epilepsy). Abnormal eye–head movements in the context of primarily unspecific epilepsy and psychomotor retardation should prompt measurement of the glucose liquor-to-plasma ratio to ascertain hypoglychorrhachia. The diagnosis is confirmed by genetic analysis of the SLC2A1 gene.
Therapy: The treatment of choice is a ketogenic diet. Its rational is to circumvent the cerebral glucopenia by providing ketone bodies as an alternative source of energy to the brain. It should be started as early as possible as it can prevent irreversible damage [107].

4.3. Cerebral Creatine Deficiency (ORPHA:79172)

Cerebral creatine deficiency (CCD) is a group of rare diseases caused by mutations in the AGAT (CCD syndrome 3, CCDS3), GAMT (CCDS2) or X-linked SLC6A8 (CCDS1) genes, leading to a disturbed synthesis or transport of creatine within the cell. The creatine metabolism plays an essential role in energy turnover, thus CCD leads to early alterations of the mitochondrial proteomic landscape [108]. During periods of high energetic demand, creatine kinases catalyze the transfer of the high-energy phosphate group in phosphocreatine to ADP, allowing for the rapid generation/regeneration of ATP, thereby maintaining the energetic supply required for cellular function [109]. The half of the creatine not taken in through diet is endogenously synthesized by the conversion of glycine and arginine into guanidinoacetate (and ornithine) through the enzyme arginine:glycine amidinotransferase (AGAT) and the subsequent transformation of guanidinoacetate into creatine through the enzyme guanidinoacetate methyltransferase (GAMT). Creatine can then be transferred into brain and muscle tissue using the creatine transporter SLC6A8 [110], where it is converted to phosphocreatine by the creatine kinase acting as a cytoplasmic ATP buffer. The brain is the most affected organ in CDS because it has its own creatine production with high levels of AGAT and GAMT expression, particularly in the cortex [111]. The common clinical presentation in CCDS includes intellectual disability, expressive speech and language delay, autistic-like behavior, and epilepsy. However, each CCD has a unique phenotype. Ataxia is mainly found in GAMT deficiency.
Therapy: AGAT and GAMT deficiencies are treatable but not curable conditions. Long-term creatine supplementation usually restores brain creatine levels and improves clinical features but the efficacy of this treatment is limited by the low permeability of the BBB for creatine. Successful treatment strategies (e.g., chaperones or cyclocreatine [112,113]) for creatine transporter deficiency are much needed. The current treatment strategy is simply the supplementation of creatine, arginine, and/or glycine, with limited success. In GAMT deficiency, the treatment consists of a low-protein and arginine-restricted diet as well as ornithine supplementation aimed at reducing levels of neurotoxic guanidinoacetate, along with creatine supply [114]. Safety monitoring and the evaluation of treatment effects is required in all patients. CCDSs may be responsible for a considerable fraction of the diagnoses’ ‘mental retardation of unknown etiology’ in children and adults. Guanidinoacetic acid (GAA), a direct metabolic precursor of creatine, has recently been suggested as a possible alternative to creatine to increase brain creatine levels in experimental medicine. AGAT patients might benefit from oral GAA due to upgraded bioavailability and convenient utilization of the compound. However, possible drawbacks (e.g., brain methylation issues, neurotoxicity, and hyperhomocysteinemia) should be carefully considered as well [115].

5. Congenital Disorders of Glycosylation

A number of genetic glycosylation defects (CDGs) can lead to cerebellar dysfunction and ataxia. Phosphomannomutase 2 deficiency is the most common example.

Phosphomannomutase 2 Deficiency (ORPHA:79318 and OMIM: 212065)

Phosphomannomutase 2 deficiency (PMM2-CDG or CDG1a) is caused by a mutation in the PMM2 gene, leading to impaired protein N-glycosylation. It is the most common congenital disorder of glycosylation. PMM2 catalyzes the conversion of mannose-6-phosphate to mannose-1-phosphate, which is a precursor of guanosine diphosphate mannose (GDP-Man) and dolichol-P-mannose (Dol-Man), which are the donors of mannose in the endoplasmic reticulum. PMM2 deficiency leads to hypoglycosylation of various glycoproteins. The disease is multisystemic and heterogeneous [116]. It is usually diagnosed in childhood as it causes developmental delay. In patients up to 10 years of age, stroke-like episodes can occur. In adulthood, the leading symptoms include cerebellar ataxia and learning difficulties [117]. Neuro-ophthalmological and -otological manifestations include impaired smooth pursuit, nystagmus, ocular flutter, ocular motor apraxia, impaired optokinetic nystagmus, and impaired vestibulo-ocular reflexes [118,119,120,121]. These symptoms point toward the cerebellopontomesencephalic regions being affected. Strabismus and nystagmus might be secondary to visual impairment, even though they can be found in patients with PMM2-CDG with normal vision as well [117,121].
Therapy: Currently, there is no approved treatment for PMM2 [116]. A long-term mannose supplementation (≥2 years) led to improved protein glycosylation in the majority of patients, was well-tolerated, and suggested possible clinical improvements, which would need to be investigated with a prospective clinical trial [122]. However, short-term dietary supplementations with mannose at 100 mg/kg body weight (b.w.) every 3 h over lll9 days or 0.17 g/kg b.w. every 3.5 h over a period of 6 months [123,124], as well as a continuous i.v. mannose infusion of 5.7 g/kg b.w. over a period of 3 weeks [125], had failed to show improvements in glycosylation patterns or clinical benefits in previous studies. Acetazolamide, a carbonic anhydrase inhibitor, has been used to treat cerebellar manifestations of PMM2-CDG based on the hypothesis of a gain-of-function effect on the calcium channel CaV2.1 secondary to dysglycosylation, which might be attenuated by this treatment [126]. This approach has had some success, independently of the conjectured mechanism, as acetazolamide appears to improve ataxia due to unrelated diseases, such as CTX (see below) or mucopolysaccharidosis type 3. Currently, investigated agents include the repurposing of epalrestat, an aldose reductase inhibitor, which has been approved for the treatment of diabetes-related neuropathy in some countries [127] (NCT04925960) as well as a mannose-1-phosphate replacement therapy GLM101 (NCT05549219).

6. Disorders of Lipid Metabolism

6.1. Abetalipoproteinemia (ORPHA:14 and OMIM: 200100)

Abetalipoproteinemia (Bassen-Kornzweig disease) is a neurodegenerative disease caused by a defect in the microsomal triglyceride transfer protein that is critical for the transfer and metabolism of fats in the body. It presents with progressive cerebellar ataxia, seizures, and abnormal eye movements, including ophthalmoplegia with dissociated nystagmus, saccadic slowing, and hypometria [1,128]. Systemic symptoms, including symptoms of fat malabsorption steatosis and abnormal liver transaminases, also occur. Abetalipoproteinemia often presents in the first or second decade of life but may also start in adulthood. The biochemical hallmarks of this disease are very low cholesterol and triglycerides as well as absent ApoB. Low vitamin E leads to progressive neurological symptoms. The diagnosis is confirmed by MTTP gene analysis.
Therapy: Early intervention with a low-fat diet combined with reduced long-chain fatty acids and fat-soluble vitamin supplements (including 100–150 mg/kg/d of α-tocopherol and vitamin A) can prevent the onset of symptoms or ameliorate the disease [6].

6.2. Hypobetalipoproteinemia (ORPHA:31154)

Hypobetalipoproteinemias (HBL) describes a group of diseases characterized by reduced plasma concentrations of low-density lipoprotein-cholesterol (LDL-C) and by intestinal lipid malabsorption and fat-soluble vitamin deficiency. The most common primary monogenic HBL is familial HBL (FHBL), which is due to a defect of the APOB gene and has an autosomal co-dominant mode of inheritance [129]. Heterozygous FHBL typically has a rather mild clinical presentation, sometimes including a fatty liver [130]. In contrast, homozygous HBL can have very severe neurological consequences if left untreated, including severe ataxia, myopathy, dysarthria, absent reflexes, retinal degeneration, neuropathy, coagulopathy, hepatic steatosis, progressive demyelination of the central nervous system, lack of responsiveness to local anesthesia, psychomotor retardation, and more, leading to various impairments and a shorter lifespan. An individual genetic diagnosis can facilitate prognosis and treatment planning [131].
Therapy: A low-fat diet and vitamin E supplementation in moderate form. However, these measures cannot control or cure HBL, which is why regular follow-ups evaluating the symptoms and compliance with the diet are necessary [132].

6.3. Cerebrotendinous Xanthomatosis (ORPHA:909 and OMIM: 213700)

Cerebrotendinous xanthomatosis (CTX) is caused by a deficiency of mitochondrial sterol 27-hydroxylase (CYP27) leading to the accumulation of cholestanol (toxic for the CNS) and cholesterol in tendons, the CNS, and the lenses [133]. Symptoms may develop at any stage of life. The disorder is characterized by solid deposits known as xanthomas near large tendons, cerebellar ataxia, pyramidal, and extrapyramidal signs, which manifest in the second or third decade of life [134,135]. Based on a large-exome human analysis, this disease appears to be underdiagnosed [136]. The typical symptoms are progressive ataxia with spasticity. Other symptoms may include neuropathy, cognitive decline, seizures, and cataracts (which often appear early and may even be the first sign). In addition, ocular motor deficits can be present, including abnormal pursuit, increased saccadic intrusions, multistep saccades, and antisaccades [137].
Therapy: A life-long replacement therapy with the bile acid chenodeoxycholic acid should be started as soon as possible, ideally immediately following a positive newborn screening test, in order to prevent or reverse neurological symptoms [133]. A combination of this first-line therapy with an HMG-CoA reductase inhibitor (statin) is effective [138,139]. Chenodeoxycholic acid works by binding to and reducing the accumulation of cholestanol and related metabolites that are damaging to the CNS [134]. The starting dosage in adults is 250 mg tid, which can subsequently be increased to 1000 mg/day if the serum cholestanol and/or urine bile alcohols remain elevated. In the pediatric population, the starting dose is 5 mg/kg/day tid. In neonates, the safety and efficacy of the drug have not yet been established [140].

7. Disorders of Mineral, Metal, and Vitamin Metabolism

7.1. Ataxia with Vitamin E Deficiency (ORPHA:96 and OMIM: 277460)

Ataxia, due to vitamin E deficiency, can have various causes, such as mutations in the TTPA gene, which is responsible for the alpha-tocopherol transfer protein. Several hereditary metabolic ataxias are associated with vitamin E deficiency. However, secondary vitamin E deficiency from a variety of other, non-hereditary causes (e.g., malabsorption and malnutrition) can lead to ataxia as well [141,142]. This ataxia is slowly progressive, accompanied by neuropathy, and may resemble Friedreich ataxia. Some patients present with retinitis pigmentosa [143] or dystonia [144]. Horizontal microsaccadic oscillations, interfering with smooth pursuit, saccades, and optokinetic nystagmus as well as hyperventilation-induced ocular flutter and hippus have been described as well [145].
Therapy: Lifelong and early-initiated oral supplementation of high-dose vitamin E in a dosage of 800–1500 mg a day aims at normal plasma levels of vitamin E, which leads to neurological improvements [146]. Nonetheless, recovery may be slow and incomplete [147]. Heterozygotes are clinically healthy but have serum vitamin E concentrations 25% lower than normal.

7.2. Multiple Carboxylase Deficiency (ORPHA:148)

Multiple carboxylase deficiency (MCD) can either be due to biotinidase or holocarboxylase synthetase deficiencies. Biotinidase deficiency is caused by mutations in the BTD gene. Biotinidase catalyzes the removal of protein-bound biotin to generate free biotin. Its deficiency leads to decreased recycling and eventually to deficiency of biotin, an important co-factor of a number enzymes, especially carboxylases which are involved in gluconeogenesis, the catabolism of several branch-chain amino acids and fatty acid synthesis [148,149]. Manifestations can occur insidiously or acutely depending on the residual biotinidase activity (<10% is considered complete deficiency), mostly during childhood (juvenile-onset form) [14,150]. Symptoms include cerebellar ataxia, ketoacidosis, dermatitis, hearing loss, optic atrophy, seizures, myoclonus, nystagmus, and intellectual disability [151]. In some countries, biotinidase deficiency is part of the newborn screening program. Mutations in the holocarboxylase synthetase gene generally cause the severe infantile-onset form of the disease. This enzyme catalyzes the fixation of biotin to inactive apocarboxylases, producing four active carboxylases (including pyruvate carboxylase).
Therapy: Biotin 5–20 (−40) mg/day for prevention or improvement in signs and symptoms. Individuals with biotinidase deficiency, who are screened and treated early and continuously, usually remain completely asymptomatic.

7.3. Wilson Disease (ORPHA:905, OMIM: 277900)

Wilson’s disease is caused by mutations in the ATP7B gene, leading to a disturbance of a copper transporter and thus to pathological storage of copper in the liver and brain [152]. The first symptoms appear in childhood or young adulthood, including dystonia, parkinsonism, and/or tremor, but predominant ataxia or spasticity cases have been described.
Therapy: Chelation treatment with penicillamine or trientine needs to be initiated as soon as possible to prevent (further) neuronal damage. A reduction in intestinal copper uptake can be achieved with zinc administration. In addition to chelation treatment, pyridoxine supplementation is necessary. In some cases, a liver transplantation might become necessary. Dystonia/parkinsonism due to a manganese transporter defect is a more recently described disorder which is treated using a similar strategy that involves chelation of heavy metals with ethylene diamine tetra-acetic acid (EDTA) [153].

8. Disorders of Mitochondrial Energy Metabolism

8.1. Pyruvate Dehydrogenase (PDH) Deficiency (ORPHA:79243 and OMIM: 312170)

The pyruvate dehydrogenase complex (PDH) is composed of several subunits. Its deficiency is caused by a number of pathogenic variants in one of its genes, most commonly the PDHA1 gene. It is characterized by lactic acidosis, seizures, impaired cognition, intermittent cerebellar ataxia, and spasticity [154] due to reduced energy production.
Therapy: A ketogenic diet, defined by high fat but very low carbohydrate consumption, leads to increased fatty acid oxidation and increased production of ketone bodies [155,156], providing an alternative energy-generating pathway that bypasses the dependency on glycolysis. The disease signs and symptoms can partially be alleviated with a ketogenic regimen, though not completely resolved [157]. A trial with high dose thiamine should be performed in all patients, but generally, only mild and late-onset PDH deficiencies will be responsive. An alternative treatment option is dichloroacetate, which reduces signal transduction by the hypoxia-inducible factors and is an inhibitor of E1 kinase, thereby stimulating (residual) PDH complex activity. It has been used in cancer cells [158]. It might have an effect on most PDHA1 pathogenic variants and can act as a temporary treatment to reduce lactic acidosis, a common symptom of PDH deficiency [159]. Phenylbutyrate has been used in MSUD, where it acts on the kinase of the BCKAD, a member of the same family of PDH kinase [29,160]. It prevents the PDH kinase from phosphorylating the PDH complex, allowing the complex to remain active. However, it can only be used in patients with certain missense variants (p.P221L, p.R234G, p.G249R, p.R349C, and p.R349H) of the PDHA1 protein. In addition, carnitine or lipoic acid supplementation can be advantageous.

8.2. Coenzyme Q 10 (Ubiquinone) Deficiency (ORPHA:139485 and OMIM: 612016)

Coenzyme Q10 (CoQ10) plays an important role in electron transport, especially in oxidative phosphorylation (OXPHOS) in that it distributes electrons between the various dehydrogenases and the cytochrome segments of the respiratory chain, central to ATP production. Its deficiency is an important cause of treatable chronic progressive ataxia. Primary CoQ10 deficiency, due to a genetic defect in coenzyme Q biosynthesis, is a clinically heterogeneous condition. Several disease genes have been identified. The CABC1 gene, also called ADCK3, is one of the numerous genes involved in the ubiquinone biosynthesis pathway. Patients with a mutation in the CABC1 gene feature a progressive neurological disorder with cerebellar atrophy and seizures [161]. A muscle biopsy typically shows decreased levels of ubiquinone. A similar condition is COQ8A-ataxia but it is a mitochondrial disease in which defect coenzyme Q10 synthesis leads to dysfunction of the respiratory chain. The disease usually presents in childhood with progressive ataxia accompanied by developmental regression and cerebellar atrophy. Given the heterogeneous phenotype, it may be hard to distinguish from other mitochondrial diseases or a wide spectrum of childhood-onset cerebellar ataxias. Diagnostic confirmation is performed by genetic analysis, usually whole exome sequencing (WES) owing to the many differential diagnoses and the difficult biochemical testing of these disorders.
Therapy: The rapid diagnosis of this potentially treatable group of conditions is paramount. Treatment entails high dose CoQ10 substitution for life, albeit with variable success. Reported doses range from 60 to 700 mg/day (Class IV) [161,162,163]. COQ8A-ataxia is a potentially treatable condition with supplementation of the coenzyme Q10 as the main therapy. However, 50% of patients may not respond to the treatment.

8.3. Mitochondrial and Nuclear Defects of the Respiratory Chain Machinery

Many syndromal mitochondrial diseases may present with ataxia as an important symptom. Examples include myoclonic epilepsy with ragged red fibers (MERRF), mitochondrial encephalopathy, lactic acidosis, stroke-like episodes (MELAS), neuropathy, ataxia, retinitis pigmentosa (NARP), maternally inherited Leigh syndrome (MILS), and Kearns–Sayre syndrome. In addition, ataxia may also be associated with defects in one of the genes coding for a factor of the mitochondrial maintenance machinery, including tRNAs and their synthetases and polymerases and proteins directly involved in the mitochondrial energy metabolism. The diagnosis can be suspected from the clinical combination of symptoms with indicative biochemical markers, such as increased lactate (though present in less than 50% of cases), pyruvate, and Krebs cycle metabolites present in urine. The diagnosis can further be substantiated by a functional and genetic (mtDNA) workup in fibroblasts or preferably in cells from affected and generally energy intensive tissues such as muscle or kidney (alternatively urothelial cells). Genetic analysis of the more than 1500 nuclear genes with mitochondrial functions and the mitochondrial genome establishes the diagnosis [164].
Therapy: Despite many years of research investigating numerous different compounds, no specific treatment for “classical” mitochondrial diseases is available today or on the horizon [165]. In clinical practice, most often a “cocktail” of vitamins is prescribed, aiming at increased residual respiratory chain function or decreased accumulation of reactive oxygen species. However, this approach has uncertain effects, apart from some specific defects of co-factors, such as coenzyme Q10 or riboflavin-related disorders, with a primary or secondary deficiency of the substituted factor [12,13].

9. Peroxisomal Disorders

9.1. Zellweger Spectrum Disorders

The peroxisome biogenesis disorders in the Zellweger spectrum disorders (PBD-ZSD) range from severe (Zellweger Syndrome, ZS) and intermediate (Neonatal Adrenoleukodystrophy, NALD) to mild (Infantile Refsum disease, IRD) phenotypes. They are characterized by a defect in peroxisome formation and caused by mutations in one of 13 PEX genes [166,167]. As a result of the defect in peroxisome formation, both catabolic and anabolic pathways are involved. Despite the heterogeneity, the metabolic profile involves very long chain fatty acids (VLCFAs), phytanic- and pristanic acid, C27-bile acid intermediates and pipecolic acid in plasma, and a deficiency of plasmalogens in erythrocytes [168]. Based on when the disease starts, early-infantile, late-infantile/childhood-onset, and juvenile/adult onset versions are distinguished. The later the disease manifests, the milder the phenotype is.
Therapy: No effective causal treatment is known to date. A number of small molecules, including betaine (an organic chemical compound with both a positive and a negative charge in its molecule but uncharged on the surface), seemed promising [169,170,171,172]. However, the details of the conducted clinical trial (NCT01838941) have not been reported elsewhere suggesting negative results. The long-term administration of cholic acid (ISRCTN96480891) in a large cohort of ZSD patients did not significantly improve clinically relevant parameters despite reducing the toxic C27-bile acid intermediates [173] and beneficial effects in smaller cohorts [174,175]. Considering the gene therapy, this must be investigated in the PEX1 mouse model [176] before a human trial can be initiated.

9.2. Refsum Disease (ORPHA:773, OMIM: 266500)

Refsum disease is caused by a deficiency of phytanoyl-CoA hydroxylase (PhyH) due to mutations in the PHYH gene, i.e., an inability to degrade the branched-chain fatty acid, phytanic acid. The main neurological feature is progressive cerebellar ataxia, presenting in late childhood or early adulthood with the additional triad of ichthyosis, retinitis pigmentosa, and neuropathy.
Therapy: A diet of reduced phytanic acids can prevent or reverse symptoms of Refsum disease [62,63].

9.3. X-Linked Adrenoleukodystrophie (ALD) (ORPHA:43 and OMIM: 300100)

ALD is a peroxisomal disorder of beta-oxidation. It includes a spectrum of phenotypes, such as cerebral ALD (in males of 3 years and older), isolated or combined adrenal insufficiency (in males aged 6 months or older), and also the less severe adult form adrenomyeloneuropathy (AMN; in males usually after 18 years and females after 40 years of age) [177]. The age of onset and severity of clinical presentation vary; the phenotype does not correlate with the type of mutation and within the same family, different phenotypes can occur [178]. Once affected boys have reached 10 years of age, about one-third of them will have developed brain disease and half of them adrenal disease. The lifetime risk for adrenal insufficiency is 80% [179,180]. Most affected males will develop some neurological symptoms. ALD/AMN is an X-linked disorder caused by mutations in the adenosine triphosphate (ATP)-binding cassette (ABC), subfamily D, member 1 gene (ABCD1), which encodes an ABC transporter [181]. ALD leads to an accumulation of very long-chain fatty acids (VLCFAs) in all tissues, given the ABC transporter helps form the channel through which VLCFAs normally move into the peroxisome. ALD protein distribution correlates with regions of high metabolic activity (heart, skeletal muscle, and liver) and with critical neural regions [182]. The cerebral forms of ALD are characterized by diverse immune responses. The profound mononuclear response is characterized by microglial activation followed by apoptosis and differs from that in multiple sclerosis [183]. The inflammatory demyelination usually first affects the splenium of the corpus callosum and the occipitoparietal regions with asymmetric progressions toward the frontal or temporal lobes. Sometimes, lesions may be seen in the brainstem as well, especially with the involvement of the pons. Usually, the spinal cord remains unaffected except for bilateral corticospinal tract degeneration [184]. The primary manifestation of AMN Is spinal cord dysfunction with progressive stiffness and weakness of the legs (spastic paraparesis), abnormal sphincter control, neurogenic bladder, and sexual dysfunction. Motor abnormalities are often preceded by gonadal dysfunction. AMN is also a progressive cerebellar disorder. In males with AMN, numbness and pain from polyneuropathy may occur. The majority of childhood ALD patients and adult AMN patients have adrenal insufficiency. Cerebral symptoms such as cognitive decline, behavioral abnormalities, visual loss, impaired auditory discrimination, feeding difficulties, poor coordination, ataxia and seizures often develop over the course of the disease. Primary adrenal insufficiency may be the initial manifestation of ALD and can remain its only sign. It may present as fatigue, nonspecific gastrointestinal symptoms, vomiting, weakness, morning headaches, or, in some cases, as an acute Addisonian crisis [185]. Hyperpigmented skin due to increased ACTH secretion is possible. Fasting hypoglycemia may occur [186]. The first step in diagnosing ALD/AMN is the VLCFA panel, which is more sensitive in male patients than in female carriers [187]. Typically, these three VLCFA parameters are included: C22:0, C24:0, and C26:0 levels, as well as the ratios of C26:0 to docosanoic acid (C26:0/C22:0) and C24:0/C22:0 [188]. Genetic testing confirms the diagnosis. In addition, adrenal function is evaluated by morning cortisol and ACTH or stimulation testing. Confirmed ALD patients without neurological symptoms are advised to undergo regular surveillance neuroimaging. Prenatal DNA-based testing is available [189].
Therapy: Treatments differ as they are targeted to specific phenotypes. First line treatment for boys with early stages of cerebral ALD is hematopoietic stem cell transplant (HSCT) [190]. However, HSCT should not be undertaken in advanced neurologic disease or presymptomatic patients. Gluco- and mineralocorticoid replacement can treat adrenal insufficiency, which remains untreated by HSCT [191]. Treatment for adults with AMN is primarily supportive and similar to other myelopathies. HSCT does not seem beneficial to AMN patients without cerebral involvement as it may even exacerbate myelopathy symptoms [192]. The aim is the prevention and treatment of complications of spinal cord disease. Advanced ALD and AMN are treated supportively [193].

10. Disorders of Purine or Pyrimidine Metabolism

Lesch-Nyhan Disease and Its Variants (ORPHA:510 and OMIM:300322)

Lesch-Nyhan disease is an X-linked inherited disease predominantly affecting men and very rarely women [194]. It is caused by mutations in the HGPRT1 gene [195]. The risk to an affected child’s siblings depends on the carrier status of the mother, not the father. Carrier women have a 50% chance of transmitting the HPRT1 variant in each pregnancy. Sons will inherit the disease and daughters will be carriers [196].
The disorder arises from a deficiency of the purine salvage enzyme hypoxanthine–guanine phosphoribosyltransferase (HGprt). The disease is characterized by elevated uric acid, increasing the risk for nephrolithiasis, renal insufficiency up to renal failure, crystalluria, or gout with tophi [195]. It can be efficiently managed with allopurinol treatment but this treatment has no impact on the neurocognitive and behavioral consequences of the disease. The phenotype is variable and depends on the level of residual HGprt activity. The classical phenotype encompasses low or undetectable levels of the HGprt enzyme, rarely with enzyme activity above 1%. These patients feature the classic phenotype with hyperuricemia, dystonia, dysarthria, ocular motor involvement (including inability to initiate voluntary saccades (ocular motor apraxia) and impaired smooth pursuit [197]), intellectual disability, and self-injurious behavior leading to self-mutilation. Patients with HGprt enzyme activity that ranges from 1.6 to 8% of the normal level present with hyperuricemia without self-injury. Enzyme activity levels that exceed 8–10% of the norm typically present with hyperuricemia without self-injury. They may display ataxia and mild cognitive impairment depending on the enzyme levels [195].
Therapy: Multimodal treatment is necessary, including pharmacotherapy, physical therapy, behavioral therapy, and psychiatric interventions. Protective equipment and dental management counteract self-mutilation. Pharmacological management focuses on mood and anxiety stabilization, including atypical antipsychotics (e.g., risperidone [198], antidepressants, carbamazepine [199], or gabapentin [200]). Given the involvement of the basal ganglia and the associated L-Dopa deficiency, L-Dopa was administered. However, instead of the anticipated amelioration, this lead to an exacerbation of the motor symptoms [201]. S-adenosylmethionine (SAMe) has shown beneficial effects in patients who can tolerate the drug [202]. Deep brain stimulation was shown (in several case reports and case series) to reduce or eliminate self-injury and aggression and to modify dystonia in some cases [203,204,205,206].

11. Urea Cycle Enzyme Deficiencies

The metabolic pathway that allows ammonia detoxification (by conversion from peripheral (muscle) and enteral sources (protein ingestion) into urea) for excretion from the body is called the urea cycle. Urea cycle disorders (UCDs) are deficiencies of an enzyme (or transporter) in that pathway [207], namely the following:
  • Carbamyl phosphate synthetase I (CPSI) deficiency;
  • Ornithine transcarbamylase (OTC) deficiency;
  • Argininosuccinate synthetase (ASS) deficiency (also known as classic citrullinemia or type I citrullinemia (CTLN1);
  • Argininosuccinate lyase (ASL) deficiency (also known as argininosuccinic aciduria);
  • N-acetyl glutamate synthetase (NAGS) deficiency;
  • Arginase deficiency.
The severe neonatal forms of all UCDs, except for arginase deficiency, lead to hyperammonemia and potentially lethal metabolic decompensation in the days following birth. Survivors frequently suffer from severe neurological injuries. UCDs are autosomal recessively inherited, except for the x-linked OTC deficiency. In affected females, the severity of clinical symptoms depends on the pattern of X-inactivation in the liver (lyonization) and can be almost as severe as in males. The majority of patients become symptomatic in early childhood. However, a disease onset in later childhood or in adulthood is possible. Common symptoms of UCD are frequent vomiting, poor appetite, protein aversion, and food refusal [208]. Other symptoms are neurological (decreased level of consciousness, altered mental status, abnormal motor function, and seizures) and gastrointestinal (vomiting, poor feeding, diarrhea, nausea, and constipation). Severe deficiencies manifest 24–48 h postnatally in newborns, who seemed initially well. Due to the protein in human milk, the newborn becomes symptomatic soon after birth, with a clinical presentation similar to sepsis [209]. The accumulation of ammonia, glutamine, and other metabolites leads to cerebral edema [210]. Patients have a lifelong risk of increased catabolism leading to metabolic decompensation. Intellectual disability and developmental delays are typical [211]. Atypical symptoms are most commonly seen with partial enzyme deficiencies (frequently with OTC deficiency), including sleep disorders, psychiatric illness, headaches, anorexia, vomiting, lethargy, ataxia, and behavioral abnormalities after increased protein intake or elevated stress levels [20,212]. These patients often prefer vegetarian diets to reduce dietary protein intake. Elevated plasma ammonia concentration (>100 to 150 µmol/L) is the laboratory hallmark of UCDs. The plasma ammonia concentration needs to be measured in an arterial or venous blood sample; capillary blood is not reliable. Mild elevations of the ammonia concentration threshold are to be interpreted based on the clinical presentation and need to be double-checked with a follow-up measurement. Elevated plasma ammonia concentration with normal blood glucose and anion gap suggests a high probability of a UCD. In patients with partial defects, the ammonia needs to be measured at the time of decompensation as it may be within the normal range during the well-compensated periods [213]. Equally, plasma amino acids as well as urinary organic and orotic acids need to be analyzed during symptomatic episodes. Neuroimaging findings tend to vary with the severity and duration of the hyperammonemia [214]. Enzyme analysis and molecular genetic testing can confirm a specific diagnosis (Table 1).
Therapy: Therapy can entail a low-protein diet, essential amino acids supplementation, nitrogen scavengers (sodium benzoate or/and phenylbutyrate), carglumic acid in NAGS deficiency, and substitution with L-arginine or L-citrulline in proximal UCD. Emergency management entails high intravenous glucose and nitrogen scavengers. Clinical trials of AVV-mediated gene transfer are currently ongoing (NCT02991144, NCT05345171, and NCT03636438). Another approach is to administer mRNA coding for OTC packaged into lipid-based nanoparticles to adolescent and adult patients with OTCD (phase 1/2 studies: NCT05526066, NCT03767270). Special management of acute and life-threatening hyperammonemia is necessary (Table 1) [7]. Patients with severe disease burden may need a liver transplantation.

12. Common Therapeutic Strategies

12.1. Physical Therapy

Physical therapy plays a crucial role in managing cerebellar ataxias. A physical therapist can design an individualized exercise program to improve balance, coordination, and muscle strength. There is a robust body of evidence demonstrating the importance of physical therapy and neuro-rehabilitative strategies in cerebellar ataxia [215,216]. The currently ongoing randomized controlled trial “Rehabilitation for ataxia study” [217] (ACTRN12618000908235) investigates the efficacy of a 30-week-long outpatient and home-based rehabilitation program for individuals with hereditary cerebellar ataxia. Physical exercises may include gait training, balance exercises, and stretching to maintain a range of motion. Hippotherapy may also be helpful but reimbursement issues may limit its use in practice. Nevertheless, not all strategies lead to improvements [218]. Patients may benefit from assistive devices depending on the severity of ataxia and its impact on mobility to improve stability and support individuals in maintaining mobility and independence. In case of frequent falls, orthoses and helmets should be considered to prevent fall-related injuries that are a major cause of morbidity and mortality in these patients.

12.2. Occupational Therapy

Occupational therapists can help individuals with hereditary cerebellar ataxias develop strategies to adapt to daily living activities. They may provide assistive devices such as braces or splints to support weakened limbs, recommend modifications in the home environment, and suggest adaptive techniques to maintain function and improve independence.

12.3. Speech Therapy

Speech and swallowing difficulties are common in hereditary cerebellar ataxias. Speech therapy can help individuals improve their speech clarity, coordination, and swallowing function. Therapists may teach exercises to strengthen the muscles involved in speech and swallowing and provide strategies to compensate for difficulties.

12.4. Symptomatic Treatment

As for cerebellar ataxias due to cerebrovascular insult, tumor, or trauma and cerebellar ataxias of other etiologies (including degenerative, autoimmune, etc.), the aim of symptomatic treatment is to improve the load and burden of symptoms (e.g., ambulation, fine motor skills, and speech). The experience of symptoms can differ a lot between patients due to the large variety in symptom etiology and due to individual differences in personality, mental wellbeing, resilience, support systems, education, and means between patients. While medications cannot cure cerebellar ataxias, they may be prescribed to manage specific symptoms, not only for ataxias but also for other signs and symptoms of the aforementioned disease (e.g., dystonia, spasticity, or dyskinesia). Here, we focus on symptomatic treatments of cerebellar ataxia that have already been introduced so far, with varying levels of evidence.

12.4.1. Acetyl-L-Leucine

As mentioned in chapter 2.2 and 2.6, ALL is the L-enantiomer of N-Acetyl-DL-Leucine (ADLL), an acetylated derivative of the amino acid leucine, which is ubiquitously present in human food [89]. ALL is the active component of the racemate and is currently not being authorized anywhere in the world for the treatment of any condition. ALL and ADLL can cross the blood–brain barrier (BBB). The drug has been shown to be safe and well tolerated [90,219]. ALL’s mode of action is multimodal, leading to increased ATP production, decreasing the superoxide dismutase 1 and mitochondrial reactive oxygen species (ROS), and reducing lipid storage in neuronal and non-neuronal tissues [42]. ALL functions as a pro-drug of leucine, leading to leucine being metabolized to alpha-ketoisocaproate and oxidized in the citric acid cycle. In contrast to leucine, ALL completely lacks toxicity because it is primarily transported by the monocarboxylate transporter (MCT1), not the L-type amino acid transporter (LAT1) [220]. Thus, ALL enables the delivery of more leucine to tissues because the transport via MCT1 is not limited by the competition with the uptake of other essential amino acids, as is the case with the transport via LAT1 [220].
A case series of patients with cerebellar ataxia of various origins treated with ADLL suggested a clinically significant benefit of the treatment [221,222] that has also been supported by neuro-metabolic studies [223,224,225]. However, the evidence for the efficacy of ADLL is somewhat contradictory with a negative placebo-controlled clinical trial that failed to show the improvement in cerebellar ataxia signs and symptoms [226]. This might be due to the heterogeneity of selected cerebellar ataxia subtypes that lead to a higher response variability together with studying a racemate instead of the ALL-specific active compound. Further clinical trials in more homogeneous populations with cerebellar ataxia are needed to evaluate the efficacy of ALL.

12.4.2. Amantadine

Amantadine, a non-competitive N-methyl-D-aspartate antagonist, which has positive effects on some Parkinsonian features, seems to benefit patients with degenerative ataxias. It has shown improvements in the functional ataxia scoring scale after daily administrations of max. 200 mg/day (Class III and IV) for a few months [227,228] but no significant improvement was found in patients with Friedreich ataxia (Class II) [229].

12.4.3. Aminopyridines and Acetazolamide

Aminopyridines, such as 4-aminopyridine (4-AP), 3,4-diaminopyridine, and prolonged-release 4-aminopyridine (fampridine) are blockers of the so-called Kv1 voltage-activated potassium channels [230], increasing the excitability of neurons, especially of cerebellar Purkinje cells and other cerebellar cells [231,232]. In a calcium channel, “tottering” mouse model of episodic ataxia type 2 (EA2), aminopyridines seem to increase the threshold for attack initiation without mitigating the character of the attack per se [233]. Two randomized, double-blind, placebo-controlled trials have shown significant benefits of treatment, i.e., reduction in attack frequency, and an improved quality of life with aminopyridines in EA2 [234,235]. Acetazolamide, a carbonic anhydrase inhibitor, has been shown to reduce the number of attacks as well (compared to placebo), but it also showed more adverse events compared to fampridine [235]. Unfortunately, fampridine is not yet approved for the indication of EA2 but for gait disturbance in patients with multiple sclerosis [236,237,238]. Importantly, the positive effects of aminopyridines are not restricted to EA2; in a mouse model of spinocerebellar ataxia type 1, they seem to correct early dysfunction and delay the neurodegeneration, thus acting neuroprotectively [239]. There is evidence for improvements in cerebellar ataxic gait under aminopyridines [240] as well as improvements in tremor [241]. Aminopyridines also ameliorate cerebellar downbeat-nystagmus and improve vision in affected individuals [222,242]. The positive effects are seemingly not restricted to the cerebellum: aminopyridines are a promising treatment option for patients with gain-of-function KCNA2- and KCNA1-encephalopathy [243,244] as well as severe intractable epileptic syndromes with intellectual disability and other neuropsychiatric symptoms.

12.4.4. Omaveloxolone

Omaveloxolone is a potent activator of Nrf2 that has been shown to be effective at improving symptoms and slowing progression of Friedreich ataxia (FA) [245]. The treatment has been approved by the FDA (02/2023) for individuals from 16 to 40 years of age with genetically confirmed FA. FA is not a metabolic ataxia, even though the mutations in the frataxin gene lead to dysfunction in the mitochondria, consequently decreasing the production of adenosine triphosphate (ATP) which leads to a lack of cell energy. An impaired signaling of Nrf2, a transcription factor that regulates the cellular defense against oxidative stress, is one of the key pathophysiological mechanisms. Thus, applying Nrf2 leads to restored function.
The approval of omaveloxolone is supported by the efficacy and safety data from the MOXIe Part 2 trial, a randomized, double-blind, and placebo-controlled study together with the data from the open-label MOXIe extension trial [245,246]. The daily treatment with 150 mg omaveloxolone for 48 weeks was generally well tolerated and resulted in statistically significant improvements in neurological functions. The most common adverse reactions in MOXIe Part 2 (≥20% and greater than placebo) were elevated liver enzymes (AST/ALT), headache, nausea, abdominal pain, fatigue, diarrhea, and musculoskeletal pain.

12.4.5. Riluzole

Riluzole has multimodal effects which include inhibiting excitatory amino acid release and stabilizing the inactivated state of voltage-dependent sodium channels but also opening small-conductance calcium-activated potassium channels (that regulate the firing rate of neurons in deep cerebellar nuclei), thus reducing neuronal hyperexcitability [247,248]. In a double-blind placebo-controlled pilot trial including 40 patients with ataxia of different etiologies, riluzole (100 mg/day) demonstrated a safe risk–benefit profile and a mean decrease in 7 of 100 points on the International Cooperative Ataxia Rating Scale (ICARS) after 8 weeks (Class II) [249]. For reference, the cut-off for a clinically relevant difference is set at five points.

12.4.6. Varenicline

Varenicline is a partial agonist at α4β2 nicotinic acetylcholine receptors. In a randomized controlled trial with SCA3 patients, a dosage of 1 mg twice per day has mainly led to gait improvements (Class II) [250]. However, due to significant drawbacks in the study design, these results need to be interpreted with caution [251].

12.5. Genetic Counseling

Hereditary metabolic cerebellar ataxias are caused by genetic, usually autosomally recessive, and rarely X-linked patterns of inheritance, such as in X-linked adrenoleucodystrophy. Genetic counseling may help individuals and families understand the inheritance patterns, the concept of carrying a pathogenic variant, the risk of passing on the condition, and the availability of genetic testing options. This information can be valuable when it comes to family planning for persons of childbearing potential and their families trying to reach a well-informed decision. In addition, non-medical factors, including but not limited to psychosocial factors, personal circumstances, social support, accessibility, financial means, and more need to be considered in the decision-making process as well.
It is highly relevant that individuals affected by a hereditary metabolic ataxia are provided with the opportunity to work closely together with a team of healthcare professionals (including neurologists, metabolic experts, endocrinologists, physical therapists, occupational therapists, psychologists, and other specialists) to tailor the therapy to their individual needs. Additionally, support groups and organizations dedicated to ataxia can provide valuable resources, education, and emotional support for individuals and their families affected by these conditions.

Author Contributions

T.B.-E.: writing of the manuscript. J.H., J.S., A.V. and M.G.: revising with regard to specific aspects of the manuscript and relevant intellectual issues. All authors have read and agreed to the published version of the manuscript.

Funding

There was no funding for this review. The APC was funded by the Open Access Publication Fonds of the University of Bern.

Data Availability Statement

This is a review article.

Conflicts of Interest

Bremova-Ertl received honoraria for lecturing from Actelion and Sanofi-Genzyme. She acts as a consultant for Intrabio, Azafaros, and Zevra. She has received a grant from the Swiss Innovation Agency, University of Bern and Baasch-Medicus. Hofmann, Stucki, and Vossenkaul have nothing to disclose. Gautschi has received consulting fees from Sanofi Genzyme and is an investigator for industry-sponsored trials from Takeda, Horizon, Idorsia, Kaleido, Mallinckrodt, and Zevra Therapeutics (formerly Orphazyme A/S).

References

  1. Hollak, C.E.M.; Lachmann, R. (Eds.) Inherited Metabolic Disease in Adults; University Press: Oxford, UK, 2016; Available online: https://global.oup.com/academic/product/inherited-metabolic-disease-in-adults-9780199972135?cc=is&lang=en& (accessed on 15 July 2023).
  2. Sehgal, D.A. Phase 1/2, Open-Label Clinical Study to Evaluate the Safety and Efficacy of Intrathecal TSHA-101 Gene Therapy for Treatment of Infantile Onset GM2 Gangliosidosis. Available online: https://clinicaltrials.gov/study/NCT04798235 (accessed on 15 July 2023).
  3. Biffi, A.; Montini, E.; Lorioli, L.; Cesani, M.; Fumagalli, F.; Plati, T.; Baldoli, C.; Martino, S.; Calabria, A.; Canale, S.; et al. Lentiviral Hematopoietic Stem Cell Gene Therapy Benefits Metachromatic Leukodystrophy. Science 2013, 341, 1233158. [Google Scholar] [CrossRef] [PubMed]
  4. Fumagalli, F.; Calbi, V.; Sora, M.G.N.; Sessa, M.; Baldoli, C.; Rancoita, P.M.V.; Ciotti, F.; Sarzana, M.; Fraschini, M.; Zambon, A.A.; et al. Lentiviral haematopoietic stem-cell gene therapy for early-onset metachromatic leukodystrophy: Long-term results from a non-randomised, open-label, phase 1/2 trial and expanded access. Lancet 2022, 399, 372–383. [Google Scholar] [CrossRef] [PubMed]
  5. Markmann, S.; Christie-Reid, J.J.; Rosenberg, J.B.; De, B.P.; Kaminsky, S.M.; Crystal, R.G.; Sondhi, D. Attenuation of the Niemann-Pick type C2 disease phenotype by intracisternal administration of an AAVrh.10 vector expressing Npc2. Exp. Neurol. 2018, 306, 22–33. [Google Scholar] [CrossRef] [PubMed]
  6. Lee, J.; Hegele, R.A. Abetalipoproteinemia and homozygous hypobetalipoproteinemia: A framework for diagnosis and man-agement. J. Inherit. Metab. Dis. 2014, 37, 333–339. [Google Scholar] [CrossRef] [PubMed]
  7. Häberle, J.; Burlina, A.; Chakrapani, A.; Dixon, M.; Karall, D.; Lindner, M.; Mandel, H.; Martinelli, D.; Pintos-Morell, G.; Santer, R.; et al. Suggested guidelines for the diagnosis and management of urea cycle disorders: First revision. J. Inherit. Metab. Dis. 2019, 42, 1192–1230. [Google Scholar] [CrossRef] [PubMed]
  8. Baumgartner, M.R. Vitamin-responsive disorders: Cobalamin, folate, biotin, vitamins B1 and E. Handb. Clin. Neurol. 2013, 113, 1799–1810. [Google Scholar]
  9. Schuelke, M. Ataxia with Vitamin E Deficiency. In GeneReviews®; University of Washington: Seattle, WA, USA, 1993. [Google Scholar]
  10. Stockler-Ipsiroglu, S.; van Karnebeek, C.D.M. Cerebral creatine deficiencies: A group of treatable intellectual developmental disorders. Semin. Neurol. 2014, 34, 350–356. [Google Scholar]
  11. Jinnah, H.A.; Albanese, A.; Bhatia, K.P.; Cardoso, F.; Da Prat, G.; de Koning, T.J.; Espay, A.J.; Fung, V.; Garcia-Ruiz, P.J.; Gershanik, O.; et al. Treatable inherited rare movement disorders. Mov. Disord. 2017, 33, 21–35. [Google Scholar] [CrossRef]
  12. Pitceathly, R.D.; Keshavan, N.; Rahman, J.; Rahman, S. Moving towards clinical trials for mitochondrial diseases. J. Inherit. Metab. Dis. 2020, 44, 22–41. [Google Scholar] [CrossRef]
  13. Diodato, D.; Schiff, M.; Cohen, B.H.; Bertini, E.; Rahman, S.; Ardissone, A.; Christodoulou, J.; Falk, M.; Goldstein, A.; Haas, R.; et al. 258th ENMC international workshop Leigh syndrome spectrum: Genetic causes, natural history and preparing for clinical trials 25-27 March 2022, Hoofddorp, Amsterdam, The Netherlands. Neuromuscul. Disord. 2023, 33, 700–709. [Google Scholar] [CrossRef]
  14. Wolf, B.; Grier, R.E.; Allen, R.J.; Goodman, S.I.; Kien, C.L. Biotinidase deficiency: The enzymatic defect in late-onset multiple carboxylase deficiency. Clin. Chim. Acta 1983, 131, 273–281. [Google Scholar] [CrossRef] [PubMed]
  15. Canda, E.; Uçar, S.K.; Çoker, M. Biotinidase Deficiency: Prevalence, Impact And Management Strategies. Pediatr. Health Med. Ther. 2020, 11, 127–133. [Google Scholar]
  16. Patterson, M.C.; Vecchio, D.; Prady, H.; Abel, L.; Wraith, J. Miglustat for treatment of Niemann-Pick C disease: A randomised controlled study. Lancet Neurol. 2007, 6, 765–772. [Google Scholar] [CrossRef] [PubMed]
  17. Geberhiwot, T.; on behalf of the International Niemann-Pick Disease Registry (INPDR); Moro, A.; Dardis, A.; Ramaswami, U.; Sirrs, S.; Marfa, M.P.; Vanier, M.T.; Walterfang, M.; Bolton, S.; et al. Consensus clinical management guidelines for Niemann-Pick disease type C. Orphanet J. Rare Dis. 2018, 13, 1–19. [Google Scholar] [CrossRef] [PubMed]
  18. Cortina-Borja, M.; Vruchte, D.T.; Mengel, E.; Amraoui, Y.; Imrie, J.; Jones, S.A.; i Dali, C.; Fineran, P.; Kirkegaard, T.; Runz, H.; et al. Annual severity increment score as a tool for stratifying patients with Niemann-Pick disease type C and for recruitment to clinical trials. Orphanet J. Rare Dis. 2018, 13, 143. [Google Scholar] [CrossRef]
  19. Bremova-Ertl, T.; Claassen, J.; Foltan, T.; Gascon-Bayarri, J.; Gissen, P.; Hahn, A.; Hassan, A.; Hennig, A.; Jones, S.A.; Kolnikova, M.; et al. Efficacy and safety of N-acetyl-l-leucine in Niemann–Pick disease type C. J. Neurol. 2021, 269, 1651–1662. [Google Scholar] [CrossRef]
  20. Rüegger, C.M.; Lindner, M.; Ballhausen, D.; Baumgartner, M.R.; Beblo, S.; Das, A.; Gautschi, M.; Glahn, E.M.; Grünert, S.C.; Hennermann, J.; et al. Cross-sectional observational study of 208 patients with non-classical urea cycle disorders. J. Inherit. Metab. Dis. 2013, 37, 21–30. [Google Scholar] [CrossRef]
  21. Martakis, K.; Claassen, J.; Gascon-Bayari, J.; Goldschagg, N.; Hahn, A.; Hassan, A.; Hennig, A.; Jones, S.; Kay, R.; Lau, H.; et al. Efficacy and Safety of N-Acetyl-l-Leucine in Children and Adults With GM2 Gangliosidoses. Neurology 2023, 100, e1072–e1083. [Google Scholar] [CrossRef]
  22. Aminoaciduria–An Overview|ScienceDirect Topics. Available online: https://www.sciencedirect.com/topics/medicine-and-dentistry/aminoaciduria (accessed on 16 July 2023).
  23. Grünert, S.C.; Wendel, U.; Lindner, M.; Leichsenring, M.; Schwab, K.O.; Vockley, J.; Lehnert, W.; Ensenauer, R. Clinical and neurocognitive outcome in symptomatic isovaleric acidemia. Orphanet J. Rare Dis. 2012, 7, 9. [Google Scholar] [CrossRef]
  24. Schlune, A.; Riederer, A.; Mayatepek, E.; Ensenauer, R. Aspects of Newborn Screening in Isovaleric Acidemia. Int. J. Neonatal Screen. 2018, 4, 7. [Google Scholar] [CrossRef]
  25. Forny, P.; Hörster, F.; Ballhausen, D.; Chakrapani, A.; Chapman, K.A.; Dionisi-Vici, C.; Dixon, M.; Grünert, S.C.; Grunewald, S.; Haliloglu, G.; et al. Guidelines for the diagnosis and management of methylmalonic acidaemia and propionic acidaemia: First revision. J. Inherit. Metab. Dis. 2021, 44, 566–592. [Google Scholar] [CrossRef] [PubMed]
  26. Blackburn, P.R.; Gass, J.M.; e Vairo, F.P.; Farnham, K.M.; Atwal, H.K.; Macklin, S.; Klee, E.W.; Atwal, P.S. Maple syrup urine disease: Mechanisms and management. Appl. Clin. Genet. 2017, 10, 57–66. [Google Scholar] [CrossRef] [PubMed]
  27. Chhabria, S.; Tomasi, L.G.; Wong, P.W.K. Ophthalmoplegia and bulbar palsy in variant form of maple syrup urine disease. Ann. Neurol. 1979, 6, 71–72. [Google Scholar] [CrossRef] [PubMed]
  28. Zee, D.S.; Freeman, J.M.; Holtzman, N.A. Ophthalmoplegia in maple syrup urine disease. J. Pediatr. 1974, 84, 113–115. [Google Scholar] [CrossRef]
  29. Brunetti-Pierri, N.; Lanpher, B.; Erez, A.; Ananieva, E.A.; Islam, M.; Marini, J.C.; Sun, Q.; Yu, C.; Hegde, M.; Li, J.; et al. Phenylbutyrate therapy for maple syrup urine disease. Hum. Mol. Genet. 2010, 20, 631–640. [Google Scholar] [CrossRef]
  30. Kleta, R.; Romeo, E.; Ristic, Z.; Ohura, T.; Stuart, C.; Arcos-Burgos, M.; Dave, M.H.; Wagner, C.; Camargo, S.R.M.; Inoue, S.; et al. Mutations in SLC6A19, encoding B0AT1, cause Hartnup disorder. Nat. Genet. 2004, 36, 999–1002. [Google Scholar] [CrossRef]
  31. Seow, H.F.; Bröer, S.; Bröer, A.; Bailey, C.G.; Potter, S.J.; Cavanaugh, J.; Rasko, J.E.J. Hartnup disorder is caused by mutations in the gene encoding the neutral amino acid transporter SLC6A19. Nat. Genet. 2004, 36, 1003–1007. [Google Scholar] [CrossRef]
  32. Haijes, H.; Prinsen, H.C.; de Sain-van der Velden, M.G.M.; Verhoeven-Duif, N.M.; van Hasselt, P.M.; Jans, J.J. Accurate discrimination of Hartnup disorder from other aminoacidurias using a diagnostic ratio. Mol. Genet. Metab. Rep. 2019, 22, 100551. [Google Scholar] [CrossRef]
  33. Bremova-Ertl, T.; Schiffmann, R.; Patterson, M.C.; Belmatoug, N.; de Villemeur, T.B.; Bardins, S.; Frenzel, C.; Malinová, V.; Naumann, S.; Arndt, J.; et al. Oculomotor and Vestibular Findings in Gaucher Disease Type 3 and Their Correlation with Neurological Findings. Front. Neurol. 2018, 8, 711. [Google Scholar] [CrossRef]
  34. Chen, L.; Halmagyi, G.M.; Todd, M.J.; Aw, S.T. Vestibular and Saccadic Abnormalities in Gaucher’s Disease. JIMD Rep. 2013, 13, 111–118. [Google Scholar] [CrossRef]
  35. Shemesh, E.; Deroma, L.; Bembi, B.; Deegan, P.; Hollak, C.; Weinreb, N.J.; Cox, T.M. Enzyme replacement and substrate reduction therapy for Gaucher disease. Cochrane Database Syst. Rev. 2015, 2015, CD010324. [Google Scholar] [CrossRef]
  36. Belmatoug, N.; Di Rocco, M.; Fraga, C.; Giraldo, P.; Hughes, D.; Lukina, E.; Maison-Blanche, P.; Merkel, M.; Niederau, C.; Plöckinger, U.; et al. Management and monitoring recommendations for the use of eliglustat in adults with type 1 Gaucher disease in Europe. Eur. J. Intern. Med. 2016, 37, 25–32. [Google Scholar] [CrossRef] [PubMed]
  37. Kirkegaard, T.; Gray, J.; Priestman, D.A.; Wallom, K.-L.; Atkins, J.; Olsen, O.D.; Klein, A.; Drndarski, S.; Petersen, N.H.T.; Ingemann, L.; et al. Heat shock protein–based therapy as a potential candidate for treating the sphingolipidoses. Sci. Transl. Med. 2016, 8, 355ra118. [Google Scholar] [CrossRef] [PubMed]
  38. Zimran, A.; Altarescu, G.; Elstein, D. Pilot study using ambroxol as a pharmacological chaperone in type 1 Gaucher disease. Blood Cells Mol. Dis. 2013, 50, 134–137. [Google Scholar] [CrossRef]
  39. Cachon-Gonzalez, M.B.; Zaccariotto, E.; Cox, T.M. Genetics and Therapies for GM2 Gangliosidosis. Curr. Gene Ther. 2018, 18, 68–89. [Google Scholar] [CrossRef]
  40. Cachón-González, M.-B.; Wang, S.Z.; Ziegler, R.; Cheng, S.H.; Cox, T.M. Reversibility of neuropathology in Tay–Sachs-related diseases. Hum. Mol. Genet. 2013, 23, 730–748. [Google Scholar] [CrossRef]
  41. Bremova-Ertl, T.; Platt, F.; Strupp, M. Sandhoff Disease: Improvement of Gait by Acetyl-DL-Leucine: A Case Report. Neuropediatrics 2020, 51, 450–452. [Google Scholar] [CrossRef] [PubMed]
  42. Kaya, E.; Smith, D.; Smith, C.; Morris, L.; Bremova-Ertl, T.; Cortina-Borja, M.; Fineran, P.; Morten, K.J.; Poulton, J.; Boland, B.; et al. Acetyl-leucine slows disease progression in lysosomal storage disorders. Brain Commun. 2020, 3, fcaa148. [Google Scholar] [CrossRef]
  43. Kaya, E.; Smith, D.A.; Smith, C.; Boland, B.; Strupp, M.; Platt, F.M. Beneficial Effects of Acetyl-DL-Leucine (ADLL) in a Mouse Model of Sandhoff Disease. J. Clin. Med. 2020, 9, 1050. [Google Scholar] [CrossRef]
  44. Holtschmidt, H.; Sandhoff, K.; Kwon, H.Y.; Harzer, K.; Nakano, T.; Suzuki, K. Sulfatide activator protein. Alternative splicing that generates three mRNAs and a newly found mutation responsible for a clinical disease. J. Biol. Chem. 1991, 266, 7556–7560. [Google Scholar] [CrossRef]
  45. van Rappard, D.F.; Boelens, J.J.; Wolf, N.I. Metachromatic leukodystrophy: Disease spectrum and approaches for treatment. Best Pr. Res. Clin. Endocrinol. Metab. 2015, 29, 261–273. [Google Scholar] [CrossRef]
  46. Gieselmann, V.; Krägeloh-Mann, I. Metachromatic leukodystrophy—An update. Neuropediatrics 2010, 41, 1–6. [Google Scholar] [CrossRef]
  47. MacFaul, R.; Cavanagh, N.; Lake, B.D.; Stephens, R.; Whitfield, A. Metachromatic leucodystrophy: Review of 38 cases. Arch. Dis. Child. 1982, 57, 168–175. [Google Scholar] [CrossRef]
  48. Kehrer, C.; Elgün, S.; Raabe, C.; Böhringer, J.; Beck-Wödl, S.; Bevot, A.; Kaiser, N.; Schöls, L.; Krägeloh-Mann, I.; Groeschel, S. Association of Age at Onset and First Symptoms With Disease Progression in Patients With Metachromatic Leukodystrophy. Neurology 2020, 96, e255–e266. [Google Scholar] [CrossRef] [PubMed]
  49. Mahmood, A.; Berry, J.; Wenger, D.A.; Escolar, M.; Sobeih, M.; Raymond, G.; Eichler, F. Metachromatic Leukodystrophy: A Case of Triplets With the Late Infantile Variant and a Systematic Review of the Literature. J. Child Neurol. 2010, 25, 572–580. [Google Scholar] [CrossRef]
  50. Sessa, M.; Lorioli, L.; Fumagalli, F.; Acquati, S.; Redaelli, D.; Baldoli, C.; Canale, S.; Lopez, I.D.; Morena, F.; Calabria, A.; et al. Lentiviral haemopoietic stem-cell gene therapy in early-onset metachromatic leukodystrophy: An ad-hoc analysis of a non-randomised, open-label, phase 1/2 trial. Lancet 2016, 388, 476–487. [Google Scholar] [CrossRef]
  51. EMA. Libmeldy. European Medicines Agency. Available online: https://www.ema.europa.eu/en/medicines/human/EPAR/libmeldy (accessed on 17 July 2023).
  52. Institut National de la Santé Et de la Recherche Médicale, France. A Phase I/II, Open Labeled, Monocentric Study of Direct Intracranial Administration of a Replication Deficient Adeno-Associated Virus Gene Transfer Vector Serotype rh.10 Expressing the Human ARSA cDNA to Children with Metachromatic Leukodystrophy. Available online: https://clinicaltrials.gov/study/NCT01801709 (accessed on 17 July 2023).
  53. Williams, R.E.; Mole, S.E. New nomenclature and classification scheme for the neuronal ceroid lipofuscinoses. Neurology 2012, 79, 183–191. [Google Scholar] [CrossRef] [PubMed]
  54. Nelvagal, H.R.; Lange, J.; Takahashi, K.; Tarczyluk-Wells, M.A.; Cooper, J.D. Pathomechanisms in the neuronal ceroid lipofuscinoses. Biochim. Biophys. Acta (BBA) Mol. Basis Dis. 2019, 1866, 165570. [Google Scholar] [CrossRef] [PubMed]
  55. Cárcel-Trullols, J.; Kovács, A.D.; Pearce, D.A. Cell biology of the NCL proteins: What they do and don’t do. Biochim. Biophys. Acta (BBA) Mol. Basis Dis. 2015, 1852, 2242–2255. [Google Scholar] [CrossRef] [PubMed]
  56. Mink, J.W.; Augustine, E.F.; Adams, H.R.; Marshall, F.J.; Kwon, J.M. Classification and Natural History of the Neuronal Ceroid Lipofuscinoses. J. Child Neurol. 2013, 28, 1101–1105. [Google Scholar] [CrossRef]
  57. Santavuori, P.; Haltia, M.; Rapola, J. Infantile Type of So-called Neuronal Ceroid-lipofuscinosis. Dev. Med. Child Neurol. 1974, 16, 644–653. [Google Scholar] [CrossRef] [PubMed]
  58. Ramadan, H.; Al-Din, A.S.; Ismail, A.; Balen, F.; Varma, A.; Twomey, A.; Watts, R.; Jackson, M.; Anderson, G.; Green, E.; et al. Adult neuronal ceroid lipofuscinosis caused by deficiency in palmitoyl protein thioesterase 1. Neurology 2007, 68, 387–388. [Google Scholar] [CrossRef] [PubMed]
  59. Nickel, M.; Simonati, A.; Jacoby, D.; Lezius, S.; Kilian, D.; Van de Graaf, B.; Pagovich, O.; Kosofsky, B.; Yohay, K.; Downs, M.; et al. Disease characteristics and progression in patients with late-infantile neuronal ceroid lipofuscinosis type 2 (CLN2) disease: An observational cohort study. Lancet Child Adolesc. Health 2018, 2, 582–590. [Google Scholar] [CrossRef]
  60. Kohan, R.; Carabelos, M.N.; Xin, W.; Sims, K.; Guelbert, N.; Cismondi, I.A.; Pons, P.; Alonso, G.I.; Troncoso, M.; Witting, S.; et al. Neuronal ceroid lipofuscinosis type CLN2: A new rationale for the construction of phenotypic subgroups based on a survey of 25 cases in South America. Gene 2013, 516, 114–121. [Google Scholar] [CrossRef] [PubMed]
  61. Cialone, J.; Adams, H.; Augustine, E.F.; Marshall, F.J.; Kwon, J.M.; Newhouse, N.; Vierhile, A.; Levy, E.; Dure, L.S.; Rose, K.R.; et al. Females experience a more severe disease course in batten disease. J. Inherit. Metab. Dis. 2011, 35, 549–555. [Google Scholar] [CrossRef] [PubMed]
  62. Williams, D.R.; Hadeed, A.; al-Din, A.S.N.; Wreikat, A.-L.; Lees, A.J. Kufor Rakeb disease: Autosomal recessive, levodo-pa-responsive parkinsonism with pyramidal degeneration, supranuclear gaze palsy, and dementia. Mov. Disord. 2005, 20, 1264–1271. [Google Scholar] [CrossRef]
  63. Ashrafi, M.R.; Amanat, M.; Garshasbi, M.; Kameli, R.; Nilipour, Y.; Heidari, M.; Rezaei, Z.; Tavasoli, A.R. An update on clinical, pathological, diagnostic, and therapeutic perspectives of childhood leukodystrophies. Expert Rev. Neurother. 2019, 20, 65–84. [Google Scholar] [CrossRef]
  64. Lewis, G.; Morrill, A.M.; Conway-Allen, S.L.; Kim, B. Review of Cerliponase Alfa: Recombinant Human Enzyme Replacement Therapy for Late-Infantile Neuronal Ceroid Lipofuscinosis Type 2. J. Child Neurol. 2019, 35, 348–353. [Google Scholar] [CrossRef]
  65. Schuchman, E.H.; Desnick, R.J. Types A and B Niemann-Pick disease. Mol. Genet. Metab. 2017, 120, 27–33. [Google Scholar] [CrossRef]
  66. Klinke, G.; Rohrbach, M.; Giugliani, R.; Burda, P.; Baumgartner, M.R.; Tran, C.; Gautschi, M.; Mathis, D.; Hersberger, M. LC-MS/MS based assay and reference intervals in children and adolescents for oxysterols elevated in Nie-mann-Pick diseases. Clin. Biochem. 2015, 48, 596–602. [Google Scholar] [CrossRef]
  67. Pavlů-Pereira, H.; Asfaw, B.; Poupčtová, H.; Ledvinová, J.; Sikora, J.; Vanier, M.T.; Sandhoff, K.; Zeman, J.; Novotná, Z.; Chudoba, D.; et al. Acid sphingomyelinase deficiency. Phenotype variability with prevalence of intermediate phenotype in a series of twenty-five Czech and Slovak patients. A multi-approach study. J. Inherit. Metab. Dis. 2005, 28, 203–227. [Google Scholar] [CrossRef] [PubMed]
  68. Wasserstein, M.P.; Aron, A.; Brodie, S.E.; Simonaro, C.; Desnick, R.J.; McGovern, M.M. Acid sphingomyelinase deficiency: Prevalence and characterization of an intermediate phenotype of Niemann-Pick disease. J. Pediatr. 2006, 149, 554–559. [Google Scholar] [CrossRef] [PubMed]
  69. Blümlein, U.; Mengel, E.; Amraoui, Y. Acid sphingomyelinase deficiency: The clinical spectrum of 2 patients who carry the Q294K mutation and diagnostic challenges. Mol. Genet. Metab. Rep. 2022, 32, 100900. [Google Scholar] [CrossRef]
  70. McGovern, M.M.; Wasserstein, M.P.; Kirmse, B.; Duvall, W.L.; Schiano, T.; Thurberg, B.L.; Richards, S.; Cox, G.F. Novel first-dose adverse drug reactions during a phase I trial of olipudase alfa (recombinant human acid sphingomyelinase) in adults with Niemann–Pick disease type B (acid sphingomyelinase deficiency). Anesthesia Analg. 2016, 18, 34–40. [Google Scholar] [CrossRef] [PubMed]
  71. Wasserstein, M.P.; Jones, S.A.; Soran, H.; Diaz, G.A.; Lippa, N.; Thurberg, B.L.; Culm-Merdek, K.; Shamiyeh, E.; Inguilizian, H.; Cox, G.F.; et al. Successful within-patient dose escalation of olipudase alfa in acid sphingomyelinase deficiency. Mol. Genet. Metab. 2015, 116, 88–97. [Google Scholar] [CrossRef] [PubMed]
  72. Diaz, G.A.; Jones, S.A.; Scarpa, M.; Mengel, K.E.; Giugliani, R.; Guffon, N.; Batsu, I.; Fraser, P.A.; Li, J.; Zhang, Q.; et al. One-year results of a clinical trial of olipudase alfa enzyme replacement therapy in pediatric patients with acid sphingomyelinase deficiency. Anesthesia Analg. 2021, 23, 1543–1550. [Google Scholar] [CrossRef] [PubMed]
  73. Genzyme, a Sanofi Company. A Phase 2/3, Multicenter, Randomized, Double-Blinded, Placebo-Controlled, Repeat-Dose Study to Evaluate the Efficacy, Safety, Pharmacodynamics, and Pharmacokinetics of Olipudase Alfa in Patients with Acid Sphin-Gomyelinase Deficiency. Available online: https://clinicaltrials.gov/study/NCT02004691 (accessed on 19 July 2023).
  74. Vruchte, D.T.; Lloyd-Evans, E.; Veldman, R.J.; Neville, D.C.A.; Dwek, R.A.; Platt, F.M.; van Blitterswijk, W.J.; Sillence, D.J. Accumulation of Glycosphingolipids in Niemann-Pick C Disease Disrupts Endosomal Transport. J. Biol. Chem. 2004, 279, 26167–26175. [Google Scholar] [CrossRef]
  75. Lloyd-Evans, E. Acidic Ca2+ stores in neurodegeneration. Messenger 2016, 5, 37–55. [Google Scholar] [CrossRef]
  76. Reunert, J.; Lotz-Havla, A.S.; Polo, G.; Kannenberg, F.; Fobker, M.; Griese, M.; Mengel, E.; Muntau, A.C.; Schnabel, P.; Sommerburg, O.; et al. Niemann-Pick Type C-2 Disease: Identification by Analysis of Plasma Cholestane-3β,5α,6β-Triol and Further Insight into the Clinical Phenotype. JIMD Rep. 2015, 23, 17–26. [Google Scholar]
  77. Jiang, X.; Sidhu, R.; Porter, F.D.; Yanjanin, N.M.; Speak, A.O.; te Vruchte, D.T.T.; Platt, F.M.; Fujiwara, H.; Scherrer, D.E.; Zhang, J.; et al. A sensitive and specific LC-MS/MS method for rapid diagnosis of Niemann-Pick C1 disease from human plasma. J. Lipid Res. 2011, 52, 1435–1445. [Google Scholar] [CrossRef]
  78. Giese, A.-K.; Mascher, H.; Grittner, U.; Eichler, S.; Kramp, G.; Lukas, J.; Vruchte, D.T.; Al Eisa, N.; Cortina-Borja, M.; Porter, F.D.; et al. A novel, highly sensitive and specific biomarker for Niemann-Pick type C1 disease. Orphanet J. Rare Dis. 2015, 10, 1–8. [Google Scholar] [CrossRef]
  79. Bremova-Ertl, T.; Schneider, S. Current advancements in therapy for Niemann-Pick disease: Progress and pitfalls. Expert Opin. Pharmacother. 2023, 24, 1229–1247. [Google Scholar] [CrossRef]
  80. Bremova, T.; Strupp, M. Vertical Supranuclear Gaze Palsy in a Toddler With Niemann-Pick Type C. Pediatr. Neurol. 2017, 72, 94. [Google Scholar] [CrossRef]
  81. Bremova-Ertl, T.; Abel, L.; Walterfang, M.; Salsano, E.; Ardissone, A.; Malinová, V.; Kolníková, M.; Gascón Bayarri, J.; Reza Tavasoli, A.; Reza Ashrafi, M. Ocular motor biomarkers in Niemann-Pick disease type C: A prospective cross-sectional multicontinental study in 72 patients. medRxiv 2020, 28, 3040–3050. [Google Scholar] [CrossRef]
  82. Sousa, M.; Maamari, B.; Bremova, T.; Nuoffer, J.; Wiest, R.; Amstutz, D.; Krack, P.; Bartholdi, D.; Tinkhauser, G. Late adult-onset Niemann Pick type C (NPC): An “atypical” typical presentation at the age of 62. Park. Relat. Disord. 2023, 105460. [Google Scholar] [CrossRef] [PubMed]
  83. Abel, L.A.; Walterfang, M.; Stainer, M.J.; Bowman, E.A.; Velakoulis, D. Longitudinal assessment of reflexive and volitional saccades in Niemann-Pick Type C disease during treatment with miglustat. Orphanet J. Rare Dis. 2015, 10, 1–8. [Google Scholar] [CrossRef] [PubMed]
  84. Bowman, E.A.; Walterfang, M.; Abel, L.; Desmond, P.; Fahey, M.; Velakoulis, D. Longitudinal changes in cerebellar and subcortical volumes in adult-onset Niemann–Pick disease type C patients treated with miglustat. J. Neurol. 2015, 262, 2106–2114. [Google Scholar] [CrossRef] [PubMed]
  85. Fecarotta, S.; Romano, A.; Della Casa, R.; Del Giudice, E.; Bruschini, D.; Mansi, G.; Bembi, B.; Dardis, A.; Fiumara, A.; Di Rocco, M.; et al. Long term follow-up to evaluate the efficacy of miglustat treatment in Italian patients with Niemann-Pick disease type C. Orphanet J. Rare Dis. 2015, 10, 1–11. [Google Scholar] [CrossRef]
  86. Pineda, M.; Juríčková, K.; Karimzadeh, P.; Kolnikova, M.; Malinova, V.; Insua, J.L.; Velten, C.; Kolb, S.A. Disease characteristics, prognosis and miglustat treatment effects on disease progression in patients with Niemann-Pick disease Type C: An international, multicenter, retrospective chart review. Orphanet J. Rare Dis. 2019, 14, 1–10. [Google Scholar] [CrossRef] [PubMed]
  87. Walterfang, M.; Chien, Y.-H.; Imrie, J.; Rushton, D.; Schubiger, D.; Patterson, M.C. Dysphagia as a risk factor for mortality in Niemann-Pick disease type C: Systematic literature review and evidence from studies with miglustat. Orphanet J. Rare Dis. 2012, 7, 76. [Google Scholar] [CrossRef] [PubMed]
  88. Fecarotta, S.; Amitrano, M.; Romano, A.; Della Casa, R.; Bruschini, D.; Astarita, L.; Parenti, G.; Andria, G. The videofluoroscopic swallowing study shows a sustained improvement of dysphagia in children with Niemann-Pick disease type C after therapy with miglustat. Am. J. Med Genet. Part A 2011, 155, 540–547. [Google Scholar] [CrossRef] [PubMed]
  89. Churchill, G.C.; Strupp, M.; Galione, A.; Platt, F.M. Unexpected differences in the pharmacokinetics of N-acetyl-DL-leucine enantiomers after oral dosing and their clinical relevance. PLoS ONE 2020, 15, e0229585. [Google Scholar] [CrossRef]
  90. Bremova, T.; Malinová, V.; Amraoui, Y.; Mengel, E.; Reinke, J.; Kolníková, M.; Strupp, M. Acetyl-dl-leucine in Niemann-Pick type C: A case series. Neurology 2015, 85, 1368–1375. [Google Scholar] [CrossRef] [PubMed]
  91. IntraBio Inc. Effects of N-Acetyl-L-Leucine on Niemann-Pick Disease Type C (NPC): A Phase III, Randomized, Place-bo-Controlled, Double-Blind, Crossover Study. Available online: https://clinicaltrials.gov/study/NCT05163288 (accessed on 20 July 2023).
  92. Mengel, E.; Patterson, M.C.; Da Riol, R.M.; Del Toro, M.; Deodato, F.; Gautschi, M.; Grunewald, S.; Grønborg, S.; Harmatz, P.; Héron, B.; et al. Efficacy and safety of arimoclomol in Niemann-Pick disease type C: Results from a double-blind, randomised, placebo-controlled, multinational phase 2/3 trial of a novel treatment. J. Inherit. Metab. Dis. 2021, 44, 1463–1480. [Google Scholar] [CrossRef]
  93. Efficacy and Safety Clinical Trial with Efavirenz in Patients Diagnosed with Adult Niemann-Pick Type C with Cognitive Impairment. Available online: https://pubmed.ncbi.nlm.nih.gov/36482560/ (accessed on 20 July 2023).
  94. Hastings, C.; Liu, B.; Hurst, B.; Cox, G.F.; Hrynkow, S. Intravenous 2-hydroxypropyl-β-cyclodextrin (Trappsol® CycloTM) demonstrates biological activity and impacts cholesterol metabolism in the central nervous system and peripheral tissues in adult subjects with Niemann-Pick Disease Type C1: Results of a phase 1 trial. Mol. Genet. Metab. 2022, 137, 309–319. [Google Scholar]
  95. D’azzo, A.; Machado, E.; Annunziata, I. Pathogenesis, emerging therapeutic targets and treatment in sialidosis. Expert Opin. Orphan Drugs 2015, 3, 491–504. [Google Scholar] [CrossRef] [PubMed]
  96. Hu, S.-C.; Hung, K.-L.; Chen, H.-J.; Lee, W.-T. Seizure remission and improvement of neurological function in sialidosis with perampanel therapy. Epilepsy Behav. Case Rep. 2018, 10, 32–34. [Google Scholar] [CrossRef]
  97. Schorlemmer, K.; Bauer, S.; Belke, M.; Hermsen, A.; Klein, K.M.; Reif, P.S.; Oertel, W.H.; Kunz, W.S.; Knake, S.; Rosenow, F.; et al. Sustained seizure remission on perampanel in progressive myoclonic epilepsy (Lafora disease). Epilepsy Behav. Case Rep. 2013, 1, 118–121. [Google Scholar] [CrossRef]
  98. Crespel, A.; Gelisse, P.; Tang, N.P.L.; Genton, P. Perampanel in 12 patients with Unverricht-Lundborg disease. Epilepsia 2017, 58, 543–547. [Google Scholar] [CrossRef] [PubMed]
  99. Shiraishi, H.; Egawa, K.; Ito, T.; Kawano, O.; Asahina, N.; Kohsaka, S. Efficacy of perampanel for controlling seizures and improving neurological dysfunction in a patient with dentatorubral-pallidoluysian atrophy (DRPLA). Epilepsy Behav. Case Rep. 2017, 8, 44–46. [Google Scholar] [CrossRef]
  100. Welling, L.; Bernstein, L.E.; Berry, G.T.; Burlina, A.B.; Eyskens, F.; Gautschi, M.; Grünewald, S.; Gubbels, C.S.; Knerr, I.; Labrune, P.; et al. International clinical guideline for the management of classical galactosemia: Diagnosis, treatment, and follow-up. J. Inherit. Metab. Dis. 2016, 40, 171–176. [Google Scholar] [CrossRef]
  101. Waggoner, D.D.; Buist, N.R.M.; Donnell, G.N. Long-term prognosis in galactosaemia: Results of a survey of 350 cases. J. Inherit. Metab. Dis. 1989, 13, 802–818. [Google Scholar] [CrossRef] [PubMed]
  102. Schadewaldt, P.; Hoffmann, B.; Hammen, H.-W.; Kamp, G.; Schweitzer-Krantz, S.; Wendel, U. Longitudinal Assessment of Intellectual Achievement in Patients With Classical Galactosemia. Pediatrics 2010, 125, e374–e381. [Google Scholar] [CrossRef]
  103. Welsink-Karssies, M.M.; Ferdinandusse, S.; Geurtsen, G.J.; Hollak, C.E.M.; Huidekoper, H.H.; Janssen, M.C.H.; Langendonk, J.G.; van der Lee, J.H.; O’flaherty, R.; Oostrom, K.J.; et al. Deep phenotyping classical galactosemia: Clinical outcomes and biochemical markers. Brain Commun. 2020, 2. [Google Scholar] [CrossRef] [PubMed]
  104. Rubio-Agusti, I.; Carecchio, M.; Bhatia, K.P.; Kojovic, M.; Parees, I.; Chandrashekar, H.S.; Footitt, E.J.; Burke, D.; Edwards, M.J.; Lachmann, R.H.; et al. Movement Disorders in Adult Patients With Classical Galactosemia. Mov. Disord. 2013, 28, 804–810. [Google Scholar] [CrossRef]
  105. Kuiper, A. Movement disorders and nonmotor neuropsychological symptoms in children and adults with classical ga-lactosemia. J. Inherit. Metab. Dis. 2019, 42, 451–458. [Google Scholar] [CrossRef]
  106. Hermans, M.E.; van Weeghel, M.; Vaz, F.M.; Ferdinandusse, S.; Hollak, C.E.M.; Huidekoper, H.H.; Janssen, M.C.H.; van Kuilenburg, A.B.P.; Pras-Raves, M.L.; Wamelink, M.M.C.; et al. Multi-omics in classical galactosemia: Evidence for the involvement of multiple metabolic pathways. J. Inherit. Metab. Dis. 2022, 45, 1094–1105. [Google Scholar] [CrossRef] [PubMed]
  107. Klepper, J.; Akman, C.; Armeno, M.; Auvin, S.; Cervenka, M.; Cross, H.J.; De Giorgis, V.; Della Marina, A.; Engelstad, K.; Heussinger, N.; et al. Glut1 Deficiency Syndrome (Glut1DS): State of the art in 2020 and recommendations of the international Glut1DS study group. Epilepsia Open 2020, 5, 354–365. [Google Scholar] [CrossRef]
  108. Giusti, L.; Molinaro, A.; Alessandrì, M.G.; Boldrini, C.; Ciregia, F.; Lacerenza, S.; Ronci, M.; Urbani, A.; Cioni, G.; Mazzoni, M.R.; et al. Brain mitochondrial proteome alteration driven by creatine deficiency suggests novel therapeutic venues for creatine deficiency syndromes. Neuroscience 2019, 409, 276–289. [Google Scholar] [CrossRef]
  109. Wyss, M.; Braissant, O.; Pischel, I.; Salomons, G.S.; Schulze, A.; Stockler, S.; Wallimann, T. Creatine and creatine kinase in health and disease--a bright future ahead? Subcell Biochem. 2007, 46, 309–334. [Google Scholar]
  110. Braissant, O.; Henry, H.; Béard, E.; Uldry, J. Creatine deficiency syndromes and the importance of creatine synthesis in the brain. Amino Acids 2011, 40, 1315–1324. [Google Scholar] [CrossRef] [PubMed]
  111. Braissant, O. GAMT deficiency: 20years of a treatable inborn error of metabolism. Mol. Genet. Metab. 2014, 111, 1–3. [Google Scholar] [CrossRef] [PubMed]
  112. Dunbar, M.; Jaggumantri, S.; Sargent, M.; Stockler-Ipsiroglu, S.; van Karnebeek, C.D. Treatment of X-linked creatine transporter (SLC6A8) deficiency: Systematic review of the literature and three new cases. Mol. Genet. Metab. 2014, 112, 259–274. [Google Scholar] [CrossRef] [PubMed]
  113. Farr, C.V.; El-Kasaby, A.; Freissmuth, M.; Sucic, S. The Creatine Transporter Unfolded: A Knotty Premise in the Cerebral Cre-atine Deficiency Syndrome. Front. Synaptic Neurosci. 2020, 12, 588954. [Google Scholar] [CrossRef]
  114. Gordon, N. Guanidinoacetate methyltransferase deficiency (GAMT). Brain Dev. 2010, 32, 79–81. [Google Scholar] [CrossRef]
  115. Ostojic, S.M. Benefits and drawbacks of guanidinoacetic acid as a possible treatment to replenish cerebral creatine in AGAT deficiency. Nutr. Neurosci. 2017, 22, 302–305. [Google Scholar] [CrossRef]
  116. Altassan, R.; Péanne, R.; Jaeken, J.; Barone, R.; Bidet, M.; Borgel, D.; Brasil, S.; Cassiman, D.; Cechova, A.; Coman, D.; et al. International clinical guidelines for the management of phosphomannomutase 2-congenital disorders of glycosylation: Diagnosis, treatment and follow up. J. Inherit. Metab. Dis. 2019, 42, 5–28. [Google Scholar] [CrossRef]
  117. Vermeer, S.; Kremer, H.P.H.; Leijten, Q.H.; Scheffer, H.; Matthijs, G.; Wevers, R.A.; Knoers, N.A.V.M.; Morava, E.; Lefeber, D.J. Cerebellar ataxia and congenital disorder of glycosylation Ia (CDG-Ia) with normal routine CDG screening. J. Neurol. 2007, 254, 1356–1358. [Google Scholar] [CrossRef]
  118. Messenger, W.B.; Yang, P.; Pennesi, M.E. Ophthalmic findings in an infant with phosphomannomutase deficiency. Doc. Ophthalmol. 2014, 128, 149–153. [Google Scholar] [CrossRef]
  119. Coorg, R.; Lotze, T.E. Child Neurology: A case of PMM2-CDG (CDG 1a) presenting with unusual eye movements. Neurology 2012, 79, e131–e133. [Google Scholar] [CrossRef]
  120. Stark, K.L.; Gibson, J.B.; Hertle, R.W.; Brodsky, M.C. Ocular motor signs in an infant with carbohydrate-deficient glycoprotein syndrome type Ia. Am. J. Ophthalmol. 2000, 130, 533–535. [Google Scholar] [CrossRef] [PubMed]
  121. Morava, E.; Wosik, H.N.; Sykut-Cegielska, J.; Adamowicz, M.; Guillard, M.; Wevers, R.; Lefeber, D.J.; Cruysberg, J.R.M. Ophthalmological abnormalities in children with congenital disorders of glycosylation type I. Br. J. Ophthalmol. 2008, 93, 350–354. [Google Scholar] [CrossRef] [PubMed]
  122. Taday, R.; Grüneberg, M.; DuChesne, I.; Reunert, J.; Marquardt, T. Dietary mannose supplementation in phosphomannomutase 2 deficiency (PMM2-CDG). Orphanet J. Rare Dis. 2020, 15, 1–8. [Google Scholar] [CrossRef] [PubMed]
  123. Kjaergaard, S.; Kristiansson, B.; Stibler, H.; Freeze, H.H.; Schwartz, M.; Martinsson, T.; Skovby, F. Failure of short-term mannose therapy of patients with carbohydrate-deficient glycoprotein syndrome type 1A. Acta Paediatr. 1998, 87, 884–888. [Google Scholar] [CrossRef] [PubMed]
  124. Mayatepek, E.; Kohlmüller, D. Mannose supplementation in carbohydrate-deficient glycoprotein syndrome type I and phos-phomannomutase deficiency. Eur. J. Pediatr. 1998, 157, 605–606. [Google Scholar] [CrossRef] [PubMed]
  125. Mayatepek, E.; Schröder, M.; Kohlmüller, D.; Bieger, W.P.; Nützenadel, W. Continuous mannose infusion in carbohy-drate-deficient glycoprotein syndrome type I. Acta Paediatr. 1997, 86, 1138–1140. [Google Scholar] [CrossRef]
  126. Martínez-Monseny, A.F.; Bolasell, M.; Callejón-Póo, L.; Cuadras, D.; Freniche, V.; Itzep, D.C. AZATAX: Acetazolamide safety and efficacy in cerebellar syndrome in PMM2 congenital dis-order of glycosylation (PMM2-CDG). Ann. Neurol. 2019, 85, 740–751. [Google Scholar] [CrossRef]
  127. Iyer, S.; Sam, F.S.; DiPrimio, N.; Preston, G.; Verheijen, J.; Murthy, K.; Parton, Z.; Tsang, H.; Lao, J.; Morava, E.; et al. Repurposing the aldose reductase inhibitor and diabetic neuropathy drug epalrestat for the congenital disorder of glycosylation PMM2-CDG. Dis. Model. Mech. 2019, 12. [Google Scholar] [CrossRef]
  128. Yee, R.D.; Cogan, D.G.; Zee, D.S. Ophthalmoplegia and dissociated nystagmus in adetalipoproteinemia. Arch. Ophthalmol. 1976, 94, 571–575. [Google Scholar] [CrossRef]
  129. Tarugi, P.; Averna, M. Hypobetalipoproteinemia: Genetics, biochemistry, and clinical spectrum. Adv. Clin. Chem. 2011, 54, 81–107. [Google Scholar]
  130. Musialik, J.; Boguszewska-Chachulska, A.; Pojda-Wilczek, D.; Gorzkowska, A.; Szymańczak, R.; Kania, M. A Rare Mutation in The APOB Gene Associated with Neurological Manifestations in Familial Hypobetali-poproteinemia. Int. J. Mol. Sci. 2020, 21, 1439. [Google Scholar] [CrossRef] [PubMed]
  131. Rimbert, A.; Vanhoye, X.; Coulibaly, D.; Marrec, M.; Pichelin, M.; Charrière, S.; Peretti, N.; Valéro, R.; Wargny, M.; Carrié, A.; et al. Phenotypic Differences Between Polygenic and Monogenic Hypobetalipoproteinemia. Arter. Thromb. Vasc. Biol. 2020. [Google Scholar] [CrossRef]
  132. Hooper, A.J.; Burnett, J.R. Update on Primary Hypobetalipoproteinemia. Curr. Atheroscler. Rep. 2014, 16, 1–7. [Google Scholar] [CrossRef] [PubMed]
  133. Nie, S.; Chen, G.; Cao, X.; Zhang, Y. Cerebrotendinous xanthomatosis: A comprehensive review of pathogenesis, clinical manifestations, diagnosis, and management. Orphanet J. Rare Dis. 2014, 9, 1–11. [Google Scholar] [CrossRef] [PubMed]
  134. Stelten, B.M.L.; Dotti, M.T.; Verrips, A.; Elibol, B.; Falik-Zaccai, T.C.; Hanman, K.; Mignarri, A.; Sithole, B.; Steiner, R.D.; Verma, S.; et al. Expert opinion on diagnosing, treating and managing patients with cerebrotendinous xanthomatosis (CTX): A modified Delphi study. Orphanet J. Rare Dis. 2021, 16, 1–18. [Google Scholar] [CrossRef]
  135. de Silva, R.; Greenfield, J.; Cook, A.; Bonney, H.; Vallortigara, J.; Hunt, B.; Giunti, P. Guidelines on the diagnosis and management of the progressive ataxias. Orphanet J. Rare Dis. 2019, 14, 51. [Google Scholar] [CrossRef]
  136. Appadurai, V.; DeBarber, A.; Chiang, P.W.; Patel, S.B.; Steiner, R.D.; Tyler, C.; Bonnen, P.E. Apparent underdiagnosis of Cerebrotendinous Xanthomatosis revealed by analysis of ~60,000 human exomes. Mol. Genet. Metab. 2015, 116, 298–304. [Google Scholar] [CrossRef]
  137. Rosini, F.; Pretegiani, E.; Mignarri, A.; Optican, L.M.; Serchi, V.; De Stefano, N.; Battaglini, M.; Monti, L.; Dotti, M.T.; Federico, A.; et al. The role of dentate nuclei in human oculomotor control: Insights from cerebrotendinous xanthomatosis. J. Physiol. 2017, 595, 3607–3620. [Google Scholar] [CrossRef]
  138. Peynet, J.; Laurent, A.; De Liege, P.; Lecoz, P.; Gambert, P.; Legrand, A.; Mikol, J.; Warnet, A. Cerebrotendinous xanthomatosis: Treatments with simvastatin, lovastatin, and chenodeoxycholic acid in 3 siblings. Neurology 1991, 41, 434–436. [Google Scholar] [CrossRef]
  139. Verrips, A.; Wevers, R.A.; Van Engelen, B.G.; Keyser, A.; Wolthers, B.G.; Barkhof, F.; Stalenhoef, A.; De Graaf, R.; Janssen-Zijlstra, F.; Van Spreeken, A.; et al. Effect of simvastatin in addition to chenodeoxycholic acid in patients with cerebrotendinous xanthomatosis. Metabolism 1999, 48, 233–238. [Google Scholar] [CrossRef]
  140. Chenodeoxycholic Acid Leadiant 250 mg Hard Capsules–Summary of Medicine Characteristics|Patient info. Patient-Info.co.uk. Available online: https://patient-info.co.uk/chenodeoxycholic-acid-leadiant-250-mg-hard-capsules-87734/summary-of-medicine-characteristics (accessed on 15 July 2023).
  141. Satya-Murti, S.; Howard, L.; Krohel, G.; Wolf, B. The spectrum of neurologic disorder from vitamin E deficiency. Neurology 1986, 36, 917. [Google Scholar] [CrossRef]
  142. Traber, M.G.; Sokol, R.J.; Ringel, S.P.; Neville, H.E.; Thellman, C.A.; Kayden, H.J. Lack of Tocopherol in Peripheral Nerves of Vitamin E-Deficient Patients with Peripheral Neuropathy. N. Engl. J. Med. 1987, 317, 262–265. [Google Scholar] [CrossRef] [PubMed]
  143. Yokota, T.; Shiojiri, T.; Gotoda, T.; Arita, M.; Arai, H.; Ohga, T.; Kanda, T.; Suzuki, J.; Imai, T.; Matsumoto, H.; et al. Friedreich-like ataxia with retinitis pigmentosa caused by the His101Gln mutation of the alfa-Tocopherol transfer protein gene. Ann. Neurol. 1997, 41, 826–832. [Google Scholar] [CrossRef] [PubMed]
  144. Zhang, L.-W.; Liu, B.; Peng, D.-T. Clinical and genetic study of ataxia with vitamin E deficiency: A case report. World J. Clin. Cases 2022, 10, 8271–8276. [Google Scholar] [CrossRef]
  145. Koens, L.H.; Tuitert, I.; Blokzijl, H.; Engelen, M.; Klouwer, F.C.C.; Lange, F.; Leen, W.G.; Lunsing, R.J.; Koelman, J.H.T.M.; Verrips, A.; et al. Eye movement disorders in inborn errors of metabolism: A quantitative analysis of 37 patients. J. Inherit. Metab. Dis. 2022, 45, 981–995. [Google Scholar] [CrossRef] [PubMed]
  146. van de Warrenburg, B.P.C.; van Gaalen, J.; Boesch, S.; Burgunder, J.-M.; Dürr, A.; Giunti, P.; Klockgether, T.; Mariotti, C.; Pandolfo, M.; Riess, O. EFNS/ENS Consensus on the diagnosis and management of chronic ataxias in adulthood. Eur. J. Neurol. 2014, 21, 552–562. [Google Scholar] [CrossRef]
  147. Schuelke, M.; Mayatepek, E.; Inter, M.; Becker, M.; Pfeiffer, E.; Speer, A.; Hübner, C.; Finckh, B. Treatment of ataxia in isolated vitamin E deficiency caused by α-tocopherol transfer protein deficiency. J. Pediatr. 1999, 134, 240–244. [Google Scholar] [CrossRef]
  148. Moss, J.; Lane, M.D. The biotin-dependent enzymes. Adv. Enzym. Relat. Areas Mol. Biol. 1971, 35, 321–442. [Google Scholar]
  149. Wolf, B.; Hsia, Y.E.; Sweetman, L.; Feldman, G.; Boychuk, R.B.; Bart, R.D.; Crowell, D.H.; Di Mauro, R.M.; Nyhan, W.L. Multiple Carboxylase Deficiency: Clinical and Biochemical Improvement Following Neonatal Biotin Treatment. Pediatrics 1981, 68, 113–118. [Google Scholar] [CrossRef]
  150. Wolf, B.; Heard, G.S.; Weissbecker, K.A.; McVoy, J.R.S.; Grier, R.E.; Leshner, R.T. Biotinidase deficiency: Initial clinical features and rapid diagnosis. Ann. Neurol. 1985, 18, 614–617. [Google Scholar] [CrossRef]
  151. Aoki, Y.; Li, X.; Sakamoto, O.; Hiratsuka, M.; Akaishi, H.; Xu, L.; Briones, P.; Suormala, T.; Baumgartner, E.R.; Suzuki, Y.; et al. Identification and characterization of mutations in patients with holocarboxylase synthetase deficiency. Hum. Genet. 1999, 104, 143–148. [Google Scholar] [CrossRef]
  152. Bandmann, O.; Weiss, K.H.; Kaler, S.G. Wilson’s disease and other neurological copper disorders. Lancet Neurol. 2015, 14, 103–113. [Google Scholar] [CrossRef]
  153. Tuschl, K.; Meyer, E.; Valdivia, L.E.; Zhao, N.; Dadswell, C.; Abdul-Sada, A. Mutations in SLC39A14 disrupt manganese homeostasis and cause childhood-onset parkinsonism-dystonia. Nat. Commun. 2016, 7, 11601. [Google Scholar] [CrossRef]
  154. Robinson, B.H.; MacMillan, H.; Petrova-Benedict, R.; Sherwood, W.G. Variable clinical presentation in patients with defective E1 component of pyruvate dehydrogenase complex. J. Pediatr. 1987, 111, 525–533. [Google Scholar] [CrossRef]
  155. Cederbaum, S.D.; Blass, J.P.; Minkoff, N.; Brown, J.W.; Cotton, M.E.; Harris, S.H. Sensitivity to carbohydrate in a patient with familial intermittent lactic acidosis and pyruvate dehy-drogenase deficiency. Pediatr. Res. 1976, 10, 713–720. [Google Scholar] [CrossRef]
  156. Vasam, G.; Reid, K.; Burelle, Y.; Menzies, K.J. Nutritional Regulation of Mitochondrial Function. In Mitochondria in Obesity and Type 2 Diabetes; Academic Press: Cambridge, MA, USA, 2019; pp. 93–126. [Google Scholar] [CrossRef]
  157. Ebertowska, A.; Ludkiewicz, B.; Klejbor, I.; Melka, N.; Moryś, J. Pyruvate dehydrogenase deficiency: Morphological and met-abolic effects, creation of animal model to search for curative treatment. Folia Morphol. 2020, 79, 191–197. [Google Scholar] [CrossRef] [PubMed]
  158. Suh, Y.; Amelio, I.; Urbano, T.G.; Tavassoli, M. Clinical update on cancer: Molecular oncology of head and neck cancer. Cell Death Dis. 2014, 5, e1018. [Google Scholar] [CrossRef] [PubMed]
  159. Karissa, P.; Simpson, T.; Dawson, S.P.; Low, T.Y.; Tay, S.H.; Nordin, F.D.A.; Zain, S.M.; Lee, P.Y.; Pung, Y.-F. Comparison Between Dichloroacetate and Phenylbutyrate Treatment for Pyruvate Dehydrogenase Deficiency. Br. J. Biomed. Sci. 2022, 79, 10382. [Google Scholar] [CrossRef] [PubMed]
  160. Ferriero, R.; Boutron, A.; Brivet, M.; Kerr, D.; Morava, E.; Rodenburg, R.J.; Bonafé, L.; Baumgartner, M.R.; Anikster, Y.; Braverman, N.E.; et al. Phenylbutyrate increases pyruvate dehydrogenase complex activity in cells harboring a variety of defects. Ann. Clin. Transl. Neurol. 2014, 1, 462–470. [Google Scholar] [CrossRef]
  161. Mollet, J.; Delahodde, A.; Serre, V.; Chretien, D.; Schlemmer, D.; Lombes, A.; Boddaert, N.; Desguerre, I.; de Lonlay, P.; de Baulny, H.O.; et al. CABC1 Gene Mutations Cause Ubiquinone Deficiency with Cerebellar Ataxia and Seizures. Am. J. Hum. Genet. 2008, 82, 623–630. [Google Scholar] [CrossRef]
  162. Horvath, R.; Czermin, B.; Gulati, S.; Demuth, S.; Houge, G.; Pyle, A.; Dineiger, C.; Blakely, E.L.; Hassani, A.; Foley, C.; et al. Adult-onset cerebellar ataxia due to mutations in CABC1/ADCK3. J. Neurol. Neurosurg. Psychiatry 2012, 83, 174–178. [Google Scholar] [CrossRef] [PubMed]
  163. Lagier-Tourenne, C.; Tazir, M.; López, L.C.; Quinzii, C.M.; Assoum, M.; Drouot, N.; Busso, C.; Makri, S.; Ali-Pacha, L.; Benhassine, T.; et al. ADCK3, an Ancestral Kinase, Is Mutated in a Form of Recessive Ataxia Associated with Coenzyme Q10 Deficiency. Am. J. Hum. Genet. 2008, 82, 661–672. [Google Scholar] [CrossRef] [PubMed]
  164. Wortmann, S.B.; Mayr, J.; Nuoffer, J.M.; Prokisch, H.; Sperl, W. A Guideline for the Diagnosis of Pediatric Mitochondrial Disease: The Value of Muscle and Skin Biopsies in the Genetics Era. Neuropediatrics 2017, 48, 309–314. [Google Scholar] [CrossRef]
  165. Parikh, S.; Goldstein, A.; Karaa, A.; Koenig, M.K.; Anselm, I.; Brunel-Guitton, C.; Christodoulou, J.; Cohen, B.H.; Dimmock, D.; Enns, G.M.; et al. Patient care standards for primary mitochondrial disease: A consensus statement from the Mitochondrial Medicine Society. Anesthesia Analg. 2017, 19, 1380–1397. [Google Scholar] [CrossRef] [PubMed]
  166. Waterham, H.R.; Ebberink, M.S. Genetics and molecular basis of human peroxisome biogenesis disorders. Biochim. Biophys. Acta (BBA) Mol. Basis Dis. 2012, 1822, 1430–1441. [Google Scholar] [CrossRef]
  167. Braverman, N.E.; D’Agostino, M.D.; MacLean, G.E. Peroxisome biogenesis disorders: Biological, clinical and pathophysiological perspectives. Dev. Disabil. Res. Rev. 2013, 17, 187–196. [Google Scholar] [CrossRef]
  168. Wanders, R.J.; Waterham, H.R. Biochemistry of Mammalian Peroxisomes Revisited. Annu. Rev. Biochem. 2006, 75, 295–332. [Google Scholar] [CrossRef]
  169. Zhang, R.; Chen, L.; Jiralerspong, S.; Snowden, A.; Steinberg, S.; Braverman, N. Recovery of PEX1-Gly843Asp peroxisome dysfunction by small-molecule compounds. Proc. Natl. Acad. Sci. USA 2010, 107, 5569–5574. [Google Scholar] [CrossRef]
  170. Wei, H.; Kemp, S.; McGuinness, M.C.; Moser, A.B.; Smith, K.D. Pharmacological induction of peroxisomes in peroxisome bio-genesis disorders. Ann. Neurol. 2000, 47, 286–296. [Google Scholar] [CrossRef]
  171. Braverman, N. A Pilot, Open Label Trial Assessing the Safety and Efficacy of Betaine in Children with Peroxisome Biogenesis Disorders. Available online: https://clinicaltrials.gov/study/NCT01838941 (accessed on 15 July 2023).
  172. Berendse, K.; Ebberink, M.S.; Ijlst, L.; Poll-The, B.T.; Wanders, R.J.; Waterham, H.R. Arginine improves peroxisome functioning in cells from patients with a mild peroxisome biogenesis disorder. Orphanet J. Rare Dis. 2013, 8, 138. [Google Scholar] [CrossRef]
  173. Klouwer, F.C.C.; Koot, B.G.P.; Berendse, K.; Kemper, E.M.; Ferdinandusse, S.; Koelfat, K.V.K.; Lenicek, M.; Vaz, F.M.; Engelen, M.; Jansen, P.L.M.; et al. The cholic acid extension study in Zellweger spectrum disorders: Results and implications for therapy. J. Inherit. Metab. Dis. 2019, 42, 303–312. [Google Scholar] [CrossRef] [PubMed]
  174. Berendse, K.; Klouwer, F.C.C.; Koot, B.G.P.; Kemper, E.M.; Ferdinandusse, S.; Koelfat, K.V.K.; Lenicek, M.; Schaap, F.G.; Waterham, H.R.; Vaz, F.M.; et al. Cholic acid therapy in Zellweger spectrum disorders. J. Inherit. Metab. Dis. 2016, 39, 859–868. [Google Scholar] [CrossRef]
  175. Heubi, J.E.; Setchell, K.D.; Bove, K.E. Long-Term Cholic Acid Therapy in Zellweger Spectrum Disorders. Case Rep. Gastroenterol. 2018, 12, 360–372. [Google Scholar] [CrossRef] [PubMed]
  176. Hiebler, S.; Masuda, T.; Hacia, J.G.; Moser, A.B.; Faust, P.L.; Liu, A.; Chowdhury, N.; Huang, N.; Lauer, A.; Bennett, J.; et al. The Pex1-G844D mouse: A model for mild human Zellweger spectrum disorder. Mol. Genet. Metab. 2014, 111, 522–532. [Google Scholar] [CrossRef]
  177. Kemp, S.; Huffnagel, I.C.; Linthorst, G.E.; Wanders, R.J.; Engelen, M. Adrenoleukodystrophy—neuroendocrine pathogenesis and redefinition of natural history. Nat. Rev. Endocrinol. 2016, 12, 606–615. [Google Scholar] [CrossRef] [PubMed]
  178. Moser, H.W.; Raymond, G.V.; Dubey, P. Adrenoleukodystrophy: New approaches to a neurodegenerative disease. JAMA 2005, 294, 3131–3134. [Google Scholar] [CrossRef]
  179. Huffnagel, I.C.; Laheji, F.K.; Aziz-Bose, R.; Tritos, N.A.; Marino, R.; Linthorst, G.E. The Natural History of Adrenal Insufficiency in X-Linked Adrenoleukodystrophy: An International Col-laboration. J. Clin. Endocrinol. Metab. 2019, 104, 118–126. [Google Scholar] [CrossRef]
  180. Huffnagel, I.C.; Dijkgraaf, M.G.W.; Janssens, G.E.; van Weeghel, M.; van Geel, B.M.; Poll-The, B.T.; Kemp, S.; Engelen, M. Disease progression in women with X-linked adrenoleukodystrophy is slow. Orphanet J. Rare Dis. 2019, 14, 1–15. [Google Scholar] [CrossRef]
  181. Mosser, J.; Douar, A.-M.; Sarde, C.-O.; Kioschis, P.; Feil, R.; Moser, H.; Poustka, A.-M.; Mandel, J.-L.; Aubourg, P. Putative X-linked adrenoleukodystrophy gene shares unexpected homology with ABC transporters. Nature 1993, 361, 726–730. [Google Scholar] [CrossRef]
  182. Höftberger, R.; Kunze, M.; Weinhofer, I.; Aboul-Enein, F.; Voigtländer, T.; Oezen, I.; Amann, G.; Bernheimer, H.; Budka, H.; Berger, J. Distribution and cellular localization of adrenoleukodystrophy protein in human tissues: Implications for X-linked adrenoleukodystrophy. Neurobiol. Dis. 2007, 28, 165–174. [Google Scholar] [CrossRef]
  183. Eichler, F.S.; Ren, J.Q.; Cossoy, M.; Rietsch, A.M.; Nagpal, S.; Moser, A.B. Is microglial apoptosis an early pathogenic change in cerebral X-linked adrenoleukodystrophy? Ann. Neurol. 2008, 63, 729–742. [Google Scholar] [CrossRef] [PubMed]
  184. Powers, J.M. Adreno-leukodystrophy (adreno-testiculo-leukomyelo-neuropathic-complex). Clin. Neuropathol. 1985, 4. [Google Scholar]
  185. Acharya, S.V.; Gopal, R.A.; Bandgar, T.R.; Joshi, S.R.; Menon, P.S.; Shah, N.S. Clinical profile of adrenoleukodysrophy. Indian J. Pediatr. 2009, 76, 1045–1047. [Google Scholar] [CrossRef] [PubMed]
  186. Dubey, P.; Raymond, G.V.; Moser, A.B.; Kharkar, S.; Bezman, L.; Moser, H.W. Adrenal insufficiency in asymptomatic adrenoleukodystrophy patients identified by very long-chain fatty acid screening. J. Pediatr. 2005, 146, 528–532. [Google Scholar] [CrossRef]
  187. Jaspers, Y.R.J.; Ferdinandusse, S.; Dijkstra, I.M.E.; Barendsen, R.W.; van Lenthe, H.; Kulik, W.; Engelen, M.; Goorden, S.M.I.; Vaz, F.M.; Kemp, S. Comparison of the Diagnostic Performance of C26:0-Lysophosphatidylcholine and Very Long-Chain Fatty Acids Analysis for Peroxisomal Disorders. Front. Cell Dev. Biol. 2020, 8, 690. [Google Scholar] [CrossRef]
  188. Moser, A.B.; Kreiter, N.; Bezman, L.; Lu, S.-E.; Raymond, G.V.; Naidu, S.; Moser, H.W. Plasma very long chain fatty acids in 3,000 peroxisome disease patients and 29,000 controls. Ann. Neurol. 1999, 45, 100–110. [Google Scholar] [CrossRef]
  189. Lledó, B.; Bernabeu, R.; Ten, J.; Galán, F.M.; Cioffi, L. Preimplantation genetic diagnosis of X-linked adrenoleukodystrophy with gender determination using multiple displacement amplification. Fertil. Steril. 2007, 88, 1327–1333. [Google Scholar] [CrossRef]
  190. Mahmood, A.; Dubey, P.; Moser, H.W.; Moser, A. X-linked adrenoleukodystrophy: Therapeutic approaches to distinct phe-notypes. Pediatr. Transpl. 2005, 9, 55–62. [Google Scholar] [CrossRef]
  191. Petryk, A.; Polgreen, L.; Chahla, S.; Miller, W.; Orchard, P.J. No evidence for the reversal of adrenal failure after hematopoietic cell transplantation in X-linked adrenoleukodystrophy. Bone Marrow Transplant. 2012, 47, 1377–1378. [Google Scholar] [CrossRef]
  192. Kühl, J.-S.; Suarez, F.; Gillett, G.T.; Hemmati, P.G.; Snowden, J.A.; Stadler, M.; Vuong, G.L.; Aubourg, P.; Köhler, W.; Arnold, R. Long-term outcomes of allogeneic haematopoietic stem cell transplantation for adult cerebral X-linked adre-noleukodystrophy. Brain 2017, 140, 953–966. [Google Scholar] [CrossRef]
  193. Engelen, M.; Kemp, S.; Poll-The, B.-T. X-Linked Adrenoleukodystrophy: Pathogenesis and Treatment. Curr. Neurol. Neurosci. Rep. 2014, 14, 1–8. [Google Scholar] [CrossRef] [PubMed]
  194. De Gregorio, L.; Jinnah, H.; Harris, J.C.; Nyhan, W.L.; Schretlen, D.J.; Trombley, L.M.; O’neill, J.P. Lesch–Nyhan disease in a female with a clinically normal monozygotic twin. Mol. Genet. Metab. 2005, 85, 70–77. [Google Scholar] [CrossRef]
  195. Harris, J.C. Lesch-Nyhan syndrome and its variants: Examining the behavioral and neurocognitive phenotype. Curr. Opin. Psychiatry 2018, 31, 96–102. [Google Scholar] [CrossRef] [PubMed]
  196. Jinnah, H.A. HPRT1 Disorders. In GeneReviews®; Adam, M.P., Ed.; University of Washington: Seattle, WA, USA, 1993. [Google Scholar]
  197. A Jinnah, H.; Lewis, R.F.; E Visser, J.; E Eddey, G.; Barabas, G.; Harris, J.C. Ocular motor dysfunction in lesch-Nyhan disease. Pediatr. Neurol. 2001, 24, 200–204. [Google Scholar] [CrossRef]
  198. Allen, S.M.; Rice, S.N. Risperidone antagonism of self-mutilation in a Lesch-Nyhan patient. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 1996, 20, 793–800. [Google Scholar] [CrossRef] [PubMed]
  199. Roach, E.; Delgado, M.; Anderson, L.; Iannaccone, S.T.; Bums, D.K. Carbamazepine Trial for Lesch-Nyhan Self-Mutilation. J. Child Neurol. 1996, 11, 476–478. [Google Scholar] [CrossRef] [PubMed]
  200. McManaman, J.; Tam, D. Gabapentin for self-injurious behavior in Lesch-Nyhan syndrome. Pediatr. Neurol. 1999, 20, 381–382. [Google Scholar] [CrossRef] [PubMed]
  201. Visser, J.E.; Schretlen, D.J.; Bloem, B.R.; Jinnah, H.A. Levodopa is not a useful treatment for Lesch-Nyhan disease. Mov. Disord. 2011, 26, 746–749. [Google Scholar] [CrossRef]
  202. Glick, N. Dramatic reduction in self-injury in Lesch-Nyhan disease following S-adenosylmethionine administration. J. Inherit. Metab. Dis. 2006, 29, 687. [Google Scholar] [CrossRef]
  203. Pralong, E.; Pollo, C.; Coubes, P.; Bloch, J.; Roulet, E.; Tétreault, M.-H.; Debatisse, D.; Villemure, J.-G. Electrophysiological characteristics of limbic and motor globus pallidus internus (GPI) neurons in two cases of Lesch–Nyhan syndrome. Neurophysiol. Clin. 2005, 35, 168–173. [Google Scholar] [CrossRef]
  204. Pralong, E.; Pollo, C.; Villemure, J.-G.; Debatisse, D. Opposite effects of internal globus pallidus stimulation on pallidal neurones activity. Mov. Disord. 2007, 22, 1879–1884. [Google Scholar] [CrossRef] [PubMed]
  205. Taira, T.; Kobayashi, T.; Hori, T. Disappearance of self-mutilating behavior in a patient with lesch-nyhan syndrome after bi-lateral chronic stimulation of the globus pallidus internus. Case report. J. Neurosurg. 2003, 98, 414–416. [Google Scholar] [CrossRef] [PubMed]
  206. Cif, L.; Biolsi, B.; Gavarini, S.; Saux, A.; Gil Robles, S.; Tancu, C.; Vasques, X.; Coubes, P. Antero-ventral internal pallidum stimulation improves behavioral disorders in Lesch-Nyhan disease. Mov. Disord. 2007, 22, 2126–2129. [Google Scholar] [CrossRef] [PubMed]
  207. Matsumoto, S.; Häberle, J.; Kido, J.; Mitsubuchi, H.; Endo, F.; Nakamura, K. Urea cycle disorders-update. J. Hum. Genet. 2019, 64, 833–847. [Google Scholar] [CrossRef]
  208. Gardeitchik, T.; Humphrey, M.; Nation, J.; Boneh, A. Early Clinical Manifestations and Eating Patterns in Patients with Urea Cycle Disorders. J. Pediatr. 2012, 161, 328–332. [Google Scholar] [CrossRef]
  209. Maestri, N.E.; Clissold, D.; Brusilow, S.W. Neonatal onset ornithine transcarbamylase deficiency: A retrospective analysis. J. Pediatr. 1999, 134, 268–272. [Google Scholar] [CrossRef]
  210. Butterworth, R.F. Effects of hyperammonaemia on brain function. J. Inherit. Metab. Dis. 1998, 21, 6–20. [Google Scholar] [CrossRef]
  211. Waisbren, S.E.; Stefanatos, A.K.; Kok, T.M.Y.; Ozturk-Hismi, B. Neuropsychological attributes of urea cycle disorders: A sys-tematic review of the literature. J. Inherit. Metab. Dis. 2019, 42, 1176–1191. [Google Scholar] [CrossRef]
  212. Maestri, N.E.; Lord, C.; Glynn, M.; Bale, A.; Brusilow, S.W. The Phenotype of Ostensibly Healthy Women Who Are Carriers for Ornithine Transcarbamylase Deficiency. Medicine 1998, 77, 389–397. [Google Scholar] [CrossRef]
  213. Arn, P.H.; Hauser, E.R.; Thomas, G.H.; Herman, G.; Hess, D.; Brusilow, S.W. Hyperammonemia in Women with a Mutation at the Ornithine Carbamoyltransferase Locus. N. Engl. J. Med. 1990, 322, 1652–1655. [Google Scholar] [CrossRef]
  214. Gropman, A. Brain imaging in urea cycle disorders. Mol. Genet. Metab. 2010, 100, S20–S30. [Google Scholar] [CrossRef] [PubMed]
  215. Miyai, I.; Ito, M.; Hattori, N.; Mihara, M.; Hatakenaka, M.; Yagura, H.; Sobue, G.; Nishizawa, M. Cerebellar Ataxia Rehabilitation Trial in Degenerative Cerebellar Diseases. Neurorehabilit. Neural Repair 2011, 26, 515–522. [Google Scholar] [CrossRef] [PubMed]
  216. He, M.; Zhang, H.-N.; Tang, Z.-C.; Gao, S.-G. Balance and coordination training for patients with genetic degenerative ataxia: A systematic review. J. Neurol. 2020, 268, 3690–3705. [Google Scholar] [CrossRef] [PubMed]
  217. Milne, S.C.; A Corben, L.; Roberts, M.; Szmulewicz, D.; Burns, J.; Grobler, A.C.; Williams, S.; Chua, J.; Liang, C.; Lamont, P.J.; et al. Rehabilitation for ataxia study: Protocol for a randomised controlled trial of an outpatient and supported home-based physiotherapy programme for people with hereditary cerebellar ataxia. BMJ Open 2020, 10, e040230. [Google Scholar] [CrossRef]
  218. Bultmann, U.; Pierscianek, D.; Gizewski, E.R.; Schoch, B.; Fritsche, N.; Timmann, D.; Maschke, M.; Frings, M. Functional recovery and rehabilitation of postural impairment and gait ataxia in patients with acute cer-ebellar stroke. Gait Posture 2014, 39, 563–569. [Google Scholar] [CrossRef]
  219. Bicvic, A. Improvement of ataxia and downbeat-nystagmus in ataxia telangiectasia treated with acetyl-DL-leucine: A case series. Devekionebtak Neducube Child Neurol. 2020. [Google Scholar]
  220. Churchill, G.C.; Strupp, M.; Factor, C.; Bremova-Ertl, T.; Factor, M.; Patterson, M.C.; Platt, F.M.; Galione, A. Acetylation turns leucine into a drug by membrane transporter switching. Sci. Rep. 2021, 11, 1–10. [Google Scholar] [CrossRef]
  221. Strupp, M.; Teufel, J.; Habs, M.; Feuerecker, R.; Muth, C.; van de Warrenburg, B.P.; Klopstock, T.; Feil, K. Effects of acetyl-dl-leucine in patients with cerebellar ataxia: A case series. J. Neurol. 2013, 260, 2556–2561. [Google Scholar] [CrossRef]
  222. Kalla, R.; Strupp, M. Aminopyridines and Acetyl-DL-leucine: New Therapies in Cerebellar Disorders. Curr. Neuropharmacol. 2019, 17, 7–13. [Google Scholar] [CrossRef]
  223. Günther, L.; Beck, R.; Xiong, G.; Potschka, H.; Jahn, K.; Bartenstein, P.; Brandt, T.; Dutia, M.; Dieterich, M.; Strupp, M.; et al. N-Acetyl-L-Leucine Accelerates Vestibular Compensation after Unilateral Labyrinthectomy by Action in the Cerebellum and Thalamus. PLoS ONE 2015, 10, e0120891. [Google Scholar] [CrossRef]
  224. Becker-Bense, S.; Feuerecker, R.; Xiong, G.; Feil, K.; Bartenstein, P.; Strupp, M.; Dieterich, M. P37. Effects of acetyl-dl-leucine on the cerebral activation pattern in cerebellar ataxia (FDG-PET study). Clin. Neurophysiol. 2015, 126, e115. [Google Scholar] [CrossRef]
  225. Becker-Bense, S.; Kaiser, L.; Becker, R.; Feil, K.; Muth, C.; Albert, N.L.; Unterrainer, M.; Bartenstein, P.; Strupp, M.; Dieterich, M. Acetyl-DL-leucine in cerebellar ataxia ([18F]-FDG-PET study): How does a cerebellar disorder influence cortical sensorimotor networks? J. Neurol. 2023, 270, 44–56. [Google Scholar] [CrossRef]
  226. Feil, K.; Adrion, C.; Boesch, S.; Doss, S.; Giordano, I.; Hengel, H.; Jacobi, H.; Klockgether, T.; Klopstock, T.; Nachbauer, W.; et al. Safety and Efficacy of Acetyl-DL-Leucine in Certain Types of Cerebellar Ataxia: The ALCAT Randomized Clinical Crossover Trial. JAMA Netw. Open 2021, 4, e2135841. [Google Scholar] [CrossRef]
  227. Botez, M.; Young, S.N.; Botez, T.; Pedraza, O.L. Treatment of Heredo-Degenerative Ataxias with Amantadine Hydrochloride. Can. J. Neurol. Sci. J. Can. des Sci. Neurol. 1991, 18, 307–311. [Google Scholar] [CrossRef]
  228. Botez, M.; Botez-Marquard, T.; Elie, R.; Pedraza, O.L.; Goyette, K.; Lalonde, R. Amantadine hydrochloride treatment in heredodegenerative ataxias: A double blind study. J. Neurol. Neurosurg. Psychiatry 1996, 61, 259–264. [Google Scholar] [CrossRef] [PubMed]
  229. Filla, A.; De Michele, G.; Orefice, G.; Santarelli, F.; Trombetta, L.; Banfi, S.; Squitieri, F.; Napolitano, G.; Puma, D.; Campanella, G. A Double-Blind Cross-over Trial of Amantadine Hydrochloride in Friedreich’s Ataxia. Can. J. Neurol. Sci. J. Can. des Sci. Neurol. 1993, 20, 52–55. [Google Scholar] [CrossRef] [PubMed]
  230. Alviña, K.; Khodakhah, K. KCa Channels as Therapeutic Targets in Episodic Ataxia Type-2. J. Neurosci. 2010, 30, 7249–7257. [Google Scholar] [CrossRef] [PubMed]
  231. Etzion, Y.; Grossman, Y. Highly 4-aminopyridine sensitive delayed rectifier current modulates the excitability of guinea pig cerebellar Purkinje cells. Exp. Brain Res. 2001, 139, 419–425. [Google Scholar] [CrossRef]
  232. Alviña, K.; Khodakhah, K. The Therapeutic Mode of Action of 4-Aminopyridine in Cerebellar Ataxia. J. Neurosci. 2010, 30, 7258–7268. [Google Scholar] [CrossRef]
  233. Weisz, C.J.; Raike, R.S.; Soria-Jasso, L.E.; Hess, E.J. Potassium channel blockers inhibit the triggers of attacks in the calcium channel mouse mutant tottering. J. Neurosci. 2005, 25, 4141–4145. [Google Scholar] [CrossRef]
  234. Strupp, M.; Kalla, R.; Claassen, J.; Adrion, C.; Mansmann, U.; Klopstock, T.; Freilinger, T.; Neugebauer, H.; Spiegel, R.; Dichgans, M.; et al. A randomized trial of 4-aminopyridine in EA2 and related familial episodic ataxias. Neurology 2011, 77, 269–275. [Google Scholar] [CrossRef] [PubMed]
  235. Muth, C.; Teufel, J.; Schöls, L.; Synofzik, M.; Franke, C.; Timmann, D.; Mansmann, U.; Strupp, M. Fampridine and Acetazolamide in EA2 and Related Familial EA: A Prospective Randomized Placebo-Controlled Trial. Neurol. Clin. Pract. 2021, 11, e438–e446. [Google Scholar] [CrossRef] [PubMed]
  236. Research, C. Ampyra (Dalfampridine) Information; FDA: Silver Spring, MD, USA, 2018. [Google Scholar]
  237. Swissmedic 2019, © Copyright. Fampyra®, Retardtabletten (Fampridinum). Available online: https://www.swissmedic.ch/swissmedic/de/home/humanarzneimittel/authorisations/new-medicines/fampyra-retardtabletten_fampridinum.html (accessed on 21 July 2023).
  238. EMA (European Medicines Agency). Fampridine Accord. Available online: https://www.ema.europa.eu/en/medicines/human/EPAR/fampridine-accord (accessed on 21 July 2023).
  239. Hourez, R.; Servais, L.; Orduz, D.; Gall, D.; Millard, I.; D’Exaerde, A.d.K.; Cheron, G.; Orr, H.T.; Pandolfo, M.; Schiffmann, S.N. Aminopyridines Correct Early Dysfunction and Delay Neurodegeneration in a Mouse Model of Spinocerebellar Ataxia Type 1. J. Neurosci. 2011, 31, 11795–11807. [Google Scholar] [CrossRef]
  240. Schniepp, R.; Wuehr, M.; Neuhaeusser, M.; Benecke, A.K.; Adrion, C.; Brandt, T.; Strupp, M.; Jahn, K. 4-Aminopyridine and cerebellar gait: A retrospective case series. J. Neurol. 2012, 259, 2491–2493. [Google Scholar] [CrossRef] [PubMed]
  241. Schniepp, R.; Jakl, V.; Wuehr, M.; Havla, J.; Kümpfel, T.; Dieterich, M.; Strupp, M.; Jahn, K. Treatment with 4-aminopyridine improves upper limb tremor of a patient with multiple sclerosis: A video case report. Mult. Scler. J. 2012, 19, 506–508. [Google Scholar] [CrossRef]
  242. Claassen, J.; Spiegel, R.; Kalla, R.; Faldon, M.; Kennard, C.; Danchaivijitr, C.; Bardins, S.; Rettinger, N.; Schneider, E.; Brandt, T.; et al. A randomised double-blind, cross-over trial of 4-aminopyridine for downbeat nystagmus—effects on slowphase eye velocity, postural stability, locomotion and symptoms. J. Neurol. Neurosurg. Psychiatry 2013, 84, 1392–1399. [Google Scholar] [CrossRef]
  243. Hedrich, U.B.S.; Lauxmann, S.; Wolff, M.; Synofzik, M.; Bast, T.; Binelli, A.; Serratosa, J.M.; Martínez-Ulloa, P.; Allen, N.M.; King, M.D.; et al. 4-Aminopyridine is a promising treatment option for patients with gain-of-function KCNA2 -encephalopathy. Sci. Transl. Med. 2021, 13, eaaz4957. [Google Scholar] [CrossRef]
  244. Müller, P.; Takacs, D.S.; Hedrich, U.B.; Coorg, R.; Masters, L.; Glinton, K.E.; Dai, H.; Cokley, J.A.; Riviello, J.J.; Lerche, H.; et al. KCNA1 gain-of-function epileptic encephalopathy treated with 4-aminopyridine. Ann. Clin. Transl. Neurol. 2023, 10, 656–663. [Google Scholar] [CrossRef]
  245. Lynch, D.R.; Chin, M.P.; Delatycki, M.B.; Subramony, S.H.; Corti, M.; Hoyle, J.C.; Boesch, S.; Nachbauer, W.; Mariotti, C.; Mathews, K.D.; et al. Safety and Efficacy of Omaveloxolone in Friedreich Ataxia (MOXIe Study). Ann. Neurol. 2020, 89, 212–225. [Google Scholar] [CrossRef]
  246. Lynch, D.R.; Chin, M.P.; Boesch, S.; Delatycki, M.B.; Giunti, P.; Goldsberry, A.; Hoyle, J.C.; Mariotti, C.; Mathews, K.D.; Nachbauer, W.; et al. Efficacy of Omaveloxolone in Friedreich’s Ataxia: Delayed-Start Analysis of the MOXIe Extension. Mov. Disord. 2022, 38, 313–320. [Google Scholar] [CrossRef]
  247. Bryson, H.M.; Fulton, B.; Benfield, P. Riluzole. A review of its pharmacodynamic and pharmacokinetic properties and thera-peutic potential in amyotrophic lateral sclerosis. Drugs 1996, 52, 549–563. [Google Scholar] [CrossRef] [PubMed]
  248. Dimitriadi, M.; Kye, M.J.; Kalloo, G.; Yersak, J.M.; Sahin, M.; Hart, A.C. The Neuroprotective Drug Riluzole Acts via Small Conductance Ca2+-Activated K+ Channels to Ameliorate Defects in Spinal Muscular Atrophy Models. J. Neurosci. 2013, 33, 6557–6562. [Google Scholar] [CrossRef] [PubMed]
  249. Ristori, G.; Romano, S.; Visconti, A.; Cannoni, S.; Spadaro, M.; Frontali, M.; Pontieri, F.E.; Vanacore, N.; Salvetti, M. Riluzole in cerebellar ataxia: A randomized, double-blind, placebo-controlled pilot trial. Neurology 2010, 74, 839–845. [Google Scholar] [CrossRef] [PubMed]
  250. Zesiewicz, T.A.; Greenstein, P.E.; Sullivan, K.L.; Wecker, L.; Miller, A.; Jahan, I.; Chen, R.; Perlman, S.L. A randomized trial of varenicline (Chantix) for the treatment of spinocerebellar ataxia type 3. Neurology 2012, 78, 545–550. [Google Scholar] [CrossRef]
  251. Filla, A.; Sacca, F.; De Michele, G. A Randomized Trial of Varenicline (Chantix) for the Treatment of Spinocerebellar Ataxia Type 3. Neurology 2012, 78, 1538. [Google Scholar] [CrossRef]
Table 1. An overview of the diseases discussed in this review. In general, cerebellar ataxia is mainly observable in milder forms of IEM and manifests between late-infantile and adult-onsets. Late-adult onsets in persons above age 60 are rare but possible. Multidisciplinary approaches, combining physical therapy, psychological/behavioral/psychiatric intervention, protective equipment, and pharmacological treatment, incl. disease-modifying (where possible) and symptomatic approaches, should be applied, where necessary. Standardized monitoring for safety and evaluation of treatment effects is required in all patients. Mutation analysis is standard for confirmatory diagnostics.
Table 1. An overview of the diseases discussed in this review. In general, cerebellar ataxia is mainly observable in milder forms of IEM and manifests between late-infantile and adult-onsets. Late-adult onsets in persons above age 60 are rare but possible. Multidisciplinary approaches, combining physical therapy, psychological/behavioral/psychiatric intervention, protective equipment, and pharmacological treatment, incl. disease-modifying (where possible) and symptomatic approaches, should be applied, where necessary. Standardized monitoring for safety and evaluation of treatment effects is required in all patients. Mutation analysis is standard for confirmatory diagnostics.
Disease (in Alphabetical Order), ORPHAcode, OMIM, Prevalence (P)Causative Gene, Gene Locus, Protein and Mode of InheritanceMode of OccurenceDiagnosticsMovement DisordersOther Clinical SignsTreatment and ConsiderationsOutcome
Abetalipoproteinemia (Bassen-Kornzweig),
(ORPHA:14, OMIM: 200100)
P: 1/1,000,000
MTTP, 4q23, Microsomal triglyceride transfer protein (MTTP).
Autosomal recessive
Chronic progressivePlasma Vitamin EAtaxia, chorea, dystonia, parkinsonismRetinitis pigmentosa, dementia, seizures, ophthalmoplegia with dissociated nystagmus, saccadic slowing, hypometria, acanthocytosis, fat malabsorption steatosis, abnormal liver transaminasesDietary fat restriction, reduced long-chain fatty acids, vitamins E (α-tocopherol) 150 mg/kg/day & vitamin A supplementsEarly treatment prevents symptoms or ameliorates the disease [6]
Adrenomyeloneuropathy (AMN = late onset form, also in females),
(ORPHA:1393, OMIM: 300100)
P: unknown
ABCD1, Xq28, Adrenoleukodystrophy protein-adenosine triphosphate (ATP)-binding cassette (ABC), subfamily D, member 1 gene.
X-linked recessive
Chronic progressive
  • Very long-chain fatty acid (VLCFA):
  • Neuroimaging
  • Adrenocorticotropic hormone dosing or stimulation for adrenal insufficiency
  • Electrophysiology testing for AMN
Spinal cord dysfunction with spastic paraparesis, abnormal sphincter control, progressive incoordination and ataxiaInflammatory and noninflammatory demyelination, peripheral neuropathy, learning disabilities, behaviour problems, neurologic deterioration, blindness, seizures, adrenal insufficiency, hyperpigmented skin, neurogenic bladder, sexual dysfunction, impaired auditory discrimination, fasting hypoglycemiaRegular follow-up by neuroimaging; at early signs of progression: Hematopoietic stem cell transplantation (HSCT) for (early) cerebral ALD.
Primarily supportive for adults with AMN.
Gluco- and mineralocorticoid replacement for adrenal insufficiency
Treatment of early cerebral ALD, Considerably higher five-year survival in ALD
Arginase deficiency
(Argininemia),
(ORPHA:90, OMIM: 207800)
P: <1/1,000,000
ARG1, 6q23, Arginase.
Autosomal recessive
Intermittent or chronic progressive
  • variably n-↑ plasma ammonia
  • Neuroimaging
  • Plasma amino acids (↑↑ Arg, ↑ Cit)/↑ urinary orotic acid analyses
  • Enzyme analyses in erythrocytes
Progressive spastic quatriplegia, rarely ataxiaChronic vomiting, anorexia, growth failure, developmental delay, somnolence, seizures, sleep disorder, psychiatric illness, headache, lethargy, behaviour abnormalities, central hyperventilation, cerebral edemaArginine-restricted diet, nitrogen scavengers if needed, essential nutrients, essential amino acids, emergency management. In case of acute hyperammonemia:anticatabolic treatment and nitrogen scavenger.Depending on the subtype
Argininosuccinic aciduria
(ASL deficiency),
(ORPHA:23, OMIM: 207900)
P: 1–9/100,000
ASL, 7q11.21, Arginosuccinate lyase,
Autosomal recessive
Intermittent or chronic progressive
  • ↑ plasma ammonia concentration
  • ↑↑ Argininosuccinic acide in plasma and urine
  • High plasma citrulline (100–300 µmol/L)
  • ↓ plasma arginine
  • High plasma glutamine
  • Elevated liver enzymes in serum
  • Orotic aciduria
  • Neuroimaging
  • Enzyme analysis in red blood cells or fibroblasts
AtaxiaChronic vomiting, failure to thrive, developmental delay, somnolence, seizures, sleep disorder, psychiatric illness, headache, lethargy, behaviour abnormalities, central hyperventilation, cerebral edema, Trichorrhexis nodosa, Dry brittle hairProtein-restricted diet, nitrogen scavengers as needed, arginine-supplementation, essential amino acids and vitamins, emergency management. In case of acute hyperammonemia: anticatabolic treatment and nitrogen scavenger [7].Outcome depends on the severity of the enzyme defect, the extent and duration of initial and recurrent hyperammonemia, as well as diagnostic delay
Ataxia with CoQ10 (ubiquinone) deficiency
(ORPHA:139485, OMIM: 612016)
See also genetic forms of secondary CoQ10 deficiencies! P: unknown
ADCK3/SCAR9, 1q42.13,
(potentially other causes of primary CoQ10 deficiency: PDSS1, PDSS2, COQ2, COQ4, COQ5, COQ6, COQ7, COQ9).
All autosomal recessive
Chronic progressive
  • Decreased coenzyme Q
  • Increased serum and CSF lactate
  • Decreased activity of respiratory complex II + III
  • Decreased activity of respiratory complex I + III
  • Muscle biopsy shows mitochondrial aggregates, lipid droplets, and decreased levels of ubiquinone
Ataxia with exercise intolerance, hypotonia, proximal muscle weakness, Tremor, myoclonic jerksDevelopmental regression and cerebellar atrophy, variably seizures, mild mental impairment
Lactic acidosis
CoQ10 supplementation, 60 to 700 mg/day reported in the literature, variable response rateVariable response rate and symptom amelioration
Ataxia with vitamin E deficiency,
(ORPHA:96, OMIM: 277460)
P: 1–9/1,000,000
TTPA, 8q13, Alpha-tocopherol transfer protein.
Autosomal recessive
Chronic progressive
  • Plasma vitamin E undetectable
  • High serum cholesterol, triglyceride and beta-lipoprotein
  • Defective liver ‘tocopherol binding protein’
  • Brain MRI
  • TTPA sequencing
Ataxia, rarely dystonia, spasticitySomatosensory disturbance with loss of proprioception and areflexiaVitamin E supplements [8,9]
Monitor vitamin E levels at regular intervals,
Improvement in symptoms
Cerebral creatine deficiency,
(ORPHA:79172)
P: <1/1,000,000
GAMT, 19p13.3 guanidinoacetate methyltransferase
AGAT
SLC6A8. X-linked recessive but symptoms in females may occur.
Chronic progressive or static/non-progressive
  • low creatine excretion (but: ↑ creatine/creatinine-ratio in urine of transporter deficient males)
  • ↓ creatine in plasma (not transporter deficiency)
  • Low CSF creatine
  • Low CSF creatinine
  • Deficiency of creatine phosphate in the brain
  • Accumulation of guanidinoacetate in urine, plasma and brain (only GAMT deficiency)
  • Guanidinoacetate methyltransferase (GAMT) deficiency
  • MR-spectroscopy: ↓↓ creatine-peak
Chorea, dystonia, ataxia, hypotonia in infancyDevelopmental delay, epilepsy, behavioral abnormalities, myopathy
50% of female carriers with transporter defect have learning and behavioural problems
  • GAMT: creatine-supplementation, ornithine, +/− Na benzoate. Low-protein diet with restricted arginine
  • AGAT: creatine supplementation, in experimental medicine Guanidinoacetic acid (GAA) intake?; drawbacks e.g., brain methylation issues, neurotoxicity, and hyperhomocysteinemia
  • Creatine transporter deficiency: supplementation of creatine, arginine and glycine (unproven effect). chaperones or cyclocreatine?
Partial improvement in symptoms [10] (the earlier the better)
Cerebrotendinous xanthomatosis,
(ORPHA:909, OMIM: 213700)
P: unknown
CYP27A1, 2q33, Mitochondrial sterol 27-hydroxylase.
Autosomal recessive
Chronic progressive
  • Normal to slightly elevated plasma cholesterol
  • Elevated plasma cholestanol
  • Elevated urinary 7 alpha-hydroxylated bile alcohols
  • Sterol 27-hydroxylase deficiency
Ataxia, spasticity, pyramidal and extrapyramidal signsXanthomas near large tendons, neuropathy, cognitive decline, seizures, cataracts, ocular motor deficits: abnormal pursuit, increased saccadic intrusions, multistep saccades, antisaccade deficitsPrompt installation of treatment with chenodeoxycholic acid, 250 mg tid, or in patients < 18 years, 5 mg/kg/body weight tid possibly in combination with HMG-CoA reductase inhibitor (statin)Prevent or reverse neurological symptoms
Ceroid lipofuscinosis,
(ORPHA:216)
P: 1-9/100,000
Several variants, multiple gene products: PPT1, 1p34.2, CLN1: Palmitoyl protein thioesterease-1 (PPT1) deficiency; TPP1, 11p15.4, CLN2: tripeptidyl peptidase-1 (TPP1) deficiency, CLN5, 13q22.3, CLN5: CLN5 protein deficiency, CTSD, 11p15, CLN10: cathepsin D deficiency, CTSF, 11q13.2, CLN13: cathepsin F deficiency.
All other NCL forms are caused by defects in lysosomal, endoplasmic reticulum or cytoplasmic vesicular transmembrane proteins. Autosomal recessive but CLN4 (autosomal dominant).
Chronic progressive
  • Clinical clusters of signs and symptoms
  • Histology
  • MR Imaging may be suggestive
  • Genetic testing
Ataxia, myoclonus, spasticity, spastic quadriparesis, parkinsonismVision loss, dementia, epilepsy, decreased level of consciousness, depression, speech delay, stuttering dysarthria, developmental plateau, polymorphic seizuresSupportive care paired with pharmacological strategies:
  • Epilepsy: valproate lamotrigine, topiramate, levetiracetam
  • Spasticity: Baclofen
  • CLN2: intrathecal ERT (Recombinant human cerliponase alfa administered into the cerebrospinal fluid via the lateral ventricles)
Supportive, only CLN2 disease is presently treatable
Citrullinemia Type 1
(ORPHA:2475, OMIM: 215700)
P: 1-9/100,000
ASS1, 9q34, Arginosuccinate synthetase.
Autosomal recessive
Intermittent
  • Elevated plasma ammonia concentration >100 to 150 micromol/L measured in arterial or venous blood sample.
  • Plasma amino acid: ↑↑↑ Citrulline, ↑ glutamine
  • ↑ urine orotic acid
  • Neuroimaging during acute presentation may show cerebral edema, during prolonged hyperammonemia cortical atrophy, white matter cystic changes, hypomyelination
  • Enzyme analysis: Fibroblasts from skin biopsy for ASS and ASL deficiencies
  • DNA mutation analysis
Abnormal motor function, abnormal posturing, ataxia, spasticityFrequent vomiting, nausea, poor appetite, food refusal, protein aversion, diarrhea, constipation, decreased level of consciousness, altered mental status, seizures, central hyperventilation, cerebral edema, progressive encephalopathy, intellectual disability, developmental delayProtein-restricted diet, amino acid mixtures as needed,
Nitrogen scavengers as needed
Arginine-supplementation
Emergency procedure: avoidance of catabolic states, anti-catabolic treatment (i.v. glucose) [7]
Outcome depends on the severity of the enzyme defect, the extent and duration of initial and recurrent hyperammonemia, as well as diagnostic delay
Galactosemia, classical
(ORPHA:79239, OMIM: 230400)
and Clinical variant galactosemia,
combinded P: approximated 1/40,000–1/60,000
GALT, 9p13, Galactose-1-phosphate uridylyltransferase.
Autosomal recessive
Chronic progressive
  • NBS in some countries
  • ↑↑ galactose-1-phosphate in erythrocytes, ↑ free galactose in plasma and urine
  • In untreated patients—elevated blood galactose urine reducing substances (galactosuria),
  • Galactose-1-phosphate uridyltransferase deficiency in erythrocytes
  • In affected neonates: liver failure, renal disease (hyperchloremic metabolic acidosis, aminoaciduria, elevated liver enzymes, albuminuria), (E. coli) sepsis
Ataxia, dystonia, tremorOn exposure to lactose: neonatal intoxication syndrome with hepatocerebral syndrome, vomiting, hypotonia, liver and renal failure, fulminant sepsis.
Cognitive deficits, psychosocial challenges
Galactose-restricted diet for lifeImprovement in clinical state, stabilisation of the disease
Long-term complications occur despite diet
GLUT1 deficiency syndrome,
(ORPHA:71277, OMIM: 606777)
P: 1-9/1,000,000
SLC2A1, 1p34.2.
Autosomal dominant or autosomal recessive
Intermittent↓ glucose-ratio CRF/serum Fluctuating (exacerbation during fasting) movement disorders, with ataxia, tremor, dystonia, chorea and/or spasticitySevere form: Early-onset epileptic enephalopathy, secondary microcephaly, psychomotor retardation, intellectual disability; milder form: late-onset absence epilepsyKetogenic dietImproved if early diagnosis
Hartnup disease,
(ORPHA:2116, OMIM: 234500)
P: 1-9/100,000
SLC6A19, 5p15.33, Neutral amino acid transporter.
Autosomal recessive
Intermittent Amino acid excretion patterns (Neutral hyperaminoaciduria)Cerebellar ataxia, hyperreflexia, hypertonusSeizures, developmental delay, emotional instability and psychosis, Light-sensitive dermatitis, Supplementation with nicotinamide or tryptophan-rich diet; Patients on a high protein diet: sunlight protection and avoidance of photosensitizing drugs [11].Reduced severity of symptoms
Hypobetalipoproteinemia
(ORPHA:31154)
P: unknown
APOB, 2p24, Apolipoprotein B. Autosomal recessive Chronic progressive↓ Serum cholesterol and beta lipoproteinAtaxia, Ataxic movements of the hands, gait disturbanceRetinal degeneration, neuropathy, coagulopathy, hepatic steatosis, progressive demyelination of the central nervous system, lack of responsiveness to local anesthesia, dislike for animal fats and milk, psychomotor retardation, acantocytosisReduction in the proportion of fat in the patient’s diet and vitamin E supplementation in moderate form [11]Prevention of adverse events
Isovaleric academia,
(ORPHA:33, OMIM: 243500)
P: 1-9/100,000
IVD, 15q14, isovaleric acid CoA dehydrogenase.
Autosomal recessive
Intermittent Newborn screening of plasma acylcarnitine profile (↑ C5), urine organic acids (↑↑ isovalerylglycine, OH-isovaleric acid) Ataxia, other movement disordersDevelopmental delay, cognitive impairment, aversion to dietary protein, episodes of massive metabolic acidosis in infancy (lethargy, vomiting, dehydration, impaired consciousness, seizures, odor of ‘sweaty feet’) moderate hepatomegaly, depressed platelets and leukocytesLow protein diet, administration of glycine or l-carnitine, restriction of leucine intakePrevention of further complications
Lesch-Nyhan disease,
(ORPHA:510, OMIM: 300322)
P: 1-9/1,000,000
HGPRT1, Xq26, Hypoxanthine-guanine phosphoribosyl-transferase.
X-linked recessive
Chronic progressive Prenatal diagnosis autoradiographic test for HPRT activity applied to cells obtained by amniocentesisDystonic movement disorder, and ataxia, spastic cerebral palsy, choreoathetosisCompulsive self-injury behaviour with self-mutilation, Intellectual disability, dysarthria, ocular motor apraxia, impaired smooth pursuit, increased risk for nephrolithiasis, renal insufficiency, crystalluria, gout with tophi
  • Protective equipment (restraints), dental management.
  • Atypical antipsychotics, e.g., risperidone, antidepressants, carbamazepine, gabapentin.
  • 20S-adenosylmethionine (SAMe), a physiological intermediate in methylation and transsulfuration is beneficial in selected patients.
  • DBS to reduce self-injury and/or modify dystonia
Reduction in self-injury and aggression, pharmacological management focuses on mood and anxiety stabilization, modification of dystonia
Maple syrup urine disease
(ORPHA:268145, OMIM: 248600)
P: 1-9/1,000,000
BCKDHA, 19q13.2, BCKDHB, 6q14.1, DBT, 1p21.2 or DLD, 7q31.1, Branched-chain amino acids-dehydrogenase.
Autosomal recessive
Intermittent or Chronic progressive
  • Elevations of the branched-chain amino acids (BCAA) in plasma,
  • α-ketoacids in urine
  • Alloisoleucine in plasma
Characteristic “bicycling” movements in neonates, dystonia, spasticity, choreoathethosis, ataxiaBehavioural changes, nausea, vomiting, coma (attributed to cerebral edema), cognitive impairment, hyperactivity, sleep disturbance, hallucinations, up-gaze supranuclear palsy and adduction deficit, absence of voluntary eve movements, absent VOR
  • Low-protein die
  • Leucine- free, isoleucine-, and valine-controlled amino acid supplements.
  • Thiamine supplementation in thiamine-responsive MSUD
  • Phenylbutyrate in MSUD
  • Emergency treatment during metabolic stress, such as intercurrent illnesses
Improved outcome with timely introduction of specific diet treatment. Prevention of decompensation
Metachromatic leukodystrophy
(ORPHA:512)
P: 1-9/1,000,000
ARSA, 22q13, Arylsulfatase A.
Autosomal recessive
Chronic progressive
  • Reduced nerve conduction velocity
  • Elevated cerebrospinal fluid protein
  • Genetics: pathogenic ARSA variant,
  • Deficient arylsulfatase A (ARSA) enzyme activity in leukocytes. In patients with MLD, ARSA activity levels typically range from undetectable to less than 10 percent of normal values. CAVE: ARSA pseudodeficiency in ~1% of general population
Rigidity, muscle weakness and unsteady gait, ataxia, spasticityMental deterioration, convulsion, megacolon, hypotonia, schizophrenia, gallbladder nonfunctional, progressive apathy, memory disturbances, neuropathyBone marrow transplant, umbilical blood transplantation
ERT,
Clinical trials with lentiviral gene transfer
Continued developmental progress, improvement in neurophysiologic function and sulfatide metabolism
New problems arise with treated phenotype
Mitochondrial disorders, including MERRF (P: unknown), MELAS (P: 1-9/1,000,000), NARP (P: 1-9/100,000), MILS (P: unknown), KSSmtDNA, nuclear genes
mitochondrial DNA (heteroplasmia), but also nuclear DNA variants
Intermittent, non-progressive, chronic progressive
  • ↑ lactic acid (+/− pyruvat and other related metabolites of energy metabolism, including Krebs cycle) in plasma, urine, CSF and MR spectroscopy
  • Paradoxical postprandial ketosis and other markers
  • Abnormal OXPHOS/Oxymetric analyses in fibroblasts (or other cells, including urothelium) and muscle (or liver, kidney) biopsy
  • histology
Any movement disorder, in any combinationAny organ may be affected: mental retardation, epilepsy, myopathy, cardiomyopathy, conduction defect (e.g., WPW syndrome), nephropathy, liver disease, retinitis pigmentosa, diabetes, exocrine pancreas insufficiencyTrial with co-factor substitution, e.g., thiamine, riboflavine, coenzyme Q10, carnitine, vitamine C, vitamine E, etc. [12,13] apart from specific defects, little evidence for efficacy of any specific intervention
Multiple carboxylate deficiency (ORPHA:148), Biotin responsive and Biotidinase deficiency,
P: 1-9/100,000
HLCS, 21q22, Holocarboxylase synthetase
BTD, 3p25, leads to secondary deficiency of:
  • Pyruvate carboxylase
  • Propionyl-CoA carboxylase
  • 3-methylcrotonyl-CoA carboxylase
  • Acetyl-CoA carboxylase.
  • Autosomal recessive
Intermittent or chronic progressive
  • NBS in some countries
  • Biotinidase activity measured in plasma, (or in fibroblasts and leukocytes and other tissue extracts by radioassay [14].)
  • Untreated patients: metabolic ketoacidosis, lactic acidosis, hyperammonemia; Elevation of 3-hydroxyisovaleric, 3-hydroxypropionic, lactic acid, and 3-methylcrotonylglycine in urine organic acid analysis. Urinary excretion of 3-hydroxyisovaleric acid- an indicator of biotin status.
Ataxia, Ketoacidosis, lactic acidosis, dermatitis, alopecia, severe hypotonia, developmental regression, seizures, infantile spasms, infections, atrophy of the superior vermis of the cerebellum, hearing loss, optic atrophy
Age at manifestation, for MCD usually neonatal, for biotinidase deficiency usually in childhood, or any time
Oral supplementation with biotin 5–20 (−40) mg/day
(higher doses needed in HLCS deficiency)
Improvement or prevention with pharmacological doses of oral biotin [15]
Niemann-Pick type A and B (ASMD = acid sphingomyelinase deficiency)
(ORPHA:618899)
P: unknown
SMPD1, 11p15.4-p15.1, Acid sphingomyelinase.
Autosomal recessive
Chronic progressive
  • ↑ oxysterols (as in NPC, somewhat less consistently)
  • Elevated serum triglycerides and LDL-cholesterol
  • Low HDL-cholesterol
  • ASM enzyme activity
Spasticity, rigidity, ataxia
  • Neurodevelopmental delay
  • Type A: failure to thrive in neonatal period, hypotonia, failure to reach developmental milestones, rarely survival beyond 2–3 years
  • Type B: little neurological involvement, pulmonary disease, frequent infections, hepatosplenomegaly, frequently living into adulthood
Treatment with olipudase alfa i.v. up to 3.0 mg/kgImprovements in disease pathology
Niemann-Pick type C,
(ORPHA:646)
P: 1-9/100,000
NPC1: 18q11-q12, NPC1 protein
NPC2: 14q24.3, NPC2 protein.
Autosomal recessive
Chronic progressive
  • ↑↑ Oxysterols
  • Plasma bile acids: 3β,5α,6β-trihydroxycholanic acid, lysosphingolipids e.g., lyso-SM-509
  • Filipin staining
  • Vertical supranuclear saccade and gaze palsy
Early-late infancy cases: Cataplexy
Juvenile and adolescent cases: ataxia
  • Early-late infancy cases: Regression of already gained neurological skills in infancy, epilepsy, vertical supranuclear saccade, gaze palsy
  • With mutations in the NPC2 gene often respiratory insufficiency
  • Juvenile and adolescent cases: dysarthrophonia, cognitive decline, psychiatric involvement, vertical supranuclear saccade, gaze palsy, epilepsy
  • Treatment with miglustat 200 mg tid p.o. in patients > 12 years, or weight-tiered dosis in children < 12 years [16]
  • Side effects: diarrhoea (lack of selectivity affecting intestinal disaccharidases versus GCS and GbA2, tremor.
  • Not authorities-approved yet:
  • Acetyl-L-Leucine (ALL): Adult patients aged ≥13 years is 4 g per day (2 g in the morning, 1 g in the afternoon, and 1 g in the evening). Pediatric patients, the weight-tiered dosage; patients between 15 to <25 kg 2 g per day (1 g in the morning and 1 g in the evening); patients between 25 to <35 kg 3 g per day (1 g in the morning, 1 g in the afternoon, and 1 g in the evening), and patients > 35 kg 4 g per day at the same interval as adults
Miglustat: Stabilisation of disease, reduction in the progression rate [17]
ALL: Improvement in signs and symptoms, reduction in disease progression [18,19]
Ornithine transcarbamylase deficiency,
(ORPHA:664, OMIM: 311250)
P: 1-9/100,000
OTC, Xp21.1, Ornithine transcarbamylase.
X-linked inheritance.
Penetrance of disease in females depends on X-inactivation, generally less severe than in (hemizygous) males.
Intermittent
  • Elevated plasma ammonia concentration >100 to 150 micromol/L measured in arterial or venous blood sample.
  • Newborn screening in some countries
  • Plasma amino acid/urine orotic acid analyses to differentiate among UCDs: ↑ glutamine, ↑ alanine, ↓citrulline, ↓ arginine, in plasma; ↑↑ orotic acid in urine
  • Neuroimaging during acute presentation may show cerebral edema, during prolonged hyperammonemia cortical atrophy, white matter cystic changes, hypomyelination
  • Enzyme analysis: in liver (and intestinal) biopsies
Abnormal motor function, abnormal posturing, ataxia, spasticityFrequent vomiting, nausea, poor appetite, food refusal, protein aversion, diarrhea, constipation, decreased level of consciousness, altered mental status, seizures, central hyperventilation, cerebral edema, progressive encephalopathy, intellectual disability, developmental delayProtein-restricted diet, amino acid mixtures as needed,
Nitrogen scavengers as needed
Arginine- (or citrulline-) supplementation
Emergency procedure: avoidance of catabolic states, anti-catabolic treatment (i.v. glucose) [7,20]
Outcome depends on the severity of the enzyme defect, the extent and duration of initial and recurrent hyperammonemia, as well as diagnostic delay
Pyruvate dehydrogenase complex/pyruvate deficiency,
(ORPHA:79243, OMIM: 312170)
P: <1/1,000,000
PDHA1, Xp22.2-p22.1, E1-alpha subunit of the PDH enzyme complex.
X-linked dominant
Intermittent
  • Lactic acid & pyruvate ↑ in plasma
  • Postprandial increase in lactate & pyruvate, while normalization of lactate/pyruvate-ratio
  • urinary organic acids
  • Brain MRI
  • Enzyme analysis of skin fibroblasts, Western blots, etc.
Ataxia, spasticitySeizures, impaired cognition
  • Ketogenic diet
  • Dichloroacetate or in some cases phenylbutyrate
  • Thiamine, carnitine or lipoic acid supplementation
Significant improvement in symptoms
Refsum disease,
(ORPHA:773, OMIM: 266500)
P: 1-9/1,000,000
PHYH, 10p13, Phytanoyl CoA hydroxylase.
Autosomal recessive
Chronic progressive
  • ↑ Serum phytanic acid
  • Phytanic acid oxidase activity in fibroblasts
Ataxia, limb paresis or atrophyNeuropathy, retinitis pigmentosa with night blindness and constriction of the visual fields, cataracts, skin changes, polyneuropathy, impaired reflexes, sensory disturbances, anosmia, progressive hearing loss, cardiac abnormalitiesDiet reduced in phytol and phytanic acids
Plasmapheresis
Prevent or reverse symptoms
Sandhoff disease (GM2-Gangliosidosis),
(ORPHA:796, OMIM: 268800)
P: 1-9/1,000,000
HEXB, 15q23-q24, Alpha subunit of hexosaminidase A and B.
Autosomal recessive
Chronic progressive
  • Chromatography of oligosaccharides (screening)
  • Enzymatic essay of β-hexosaminidase activities carried out in plasma and/or peripheral white blood cells
Ataxia with involvement of 2nd motoneuronSlowly developing intellectual disability, bipolar disorder, psychosis
  • No specific treatment, muscle-relaxing agents e. g. baclofen or tizanidine
  • ALL improves motor signs and symptoms [21], not yet approved
Chronic progressive at different rates
Tay–Sachs disease (GM2-Gangliosidosis),
(ORPHA:845, OMIM: 272800)
P: unknown
HEXA, 15q23-q24, Alpha subunit of hexosaminidase A.
Autosomal recessive
Chronic progressive
  • Chromatography of oligosaccharides (screening)
  • Enzymatic essay of β-hexosaminidase activities carried out in plasma and/or peripheral white blood cells
Ataxia with involvement of 2nd motoneuronSlowly developing intellectual disability, bipolar disorder, psychosis
  • No specific treatment, Baclofen, etc.
  • ALL improves motor signs and symptoms [21], not yet approved
Chronic progressive at different rates
Wilson disease,
(ORPHA:905, OMIM: 277900)
P: 1-9/100,000
ATPB7, 13q14, ATP7B-Copper transporting ATPase.
Autosomal recessive
Chronic progressive
  • Ceruloplasmin isoforms measurement in cord blood sample of neonates or venous blood sample of adults
  • Urinary copper excretion levels
  • Neuroophthalmologic slit-lamp assessment
  • Neuroimaging
Ataxia, spasticity, dystonia, parkinsonism, tremorManifestations of liver cirrhosisChelation agents e. g. penicillamine, trientine, reduction in intestinal copper uptake with zinc, TriheptanoinPrevention of neuronal damage, preservation of liver function
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Bremova-Ertl, T.; Hofmann, J.; Stucki, J.; Vossenkaul, A.; Gautschi, M. Inborn Errors of Metabolism with Ataxia: Current and Future Treatment Options. Cells 2023, 12, 2314. https://doi.org/10.3390/cells12182314

AMA Style

Bremova-Ertl T, Hofmann J, Stucki J, Vossenkaul A, Gautschi M. Inborn Errors of Metabolism with Ataxia: Current and Future Treatment Options. Cells. 2023; 12(18):2314. https://doi.org/10.3390/cells12182314

Chicago/Turabian Style

Bremova-Ertl, Tatiana, Jan Hofmann, Janine Stucki, Anja Vossenkaul, and Matthias Gautschi. 2023. "Inborn Errors of Metabolism with Ataxia: Current and Future Treatment Options" Cells 12, no. 18: 2314. https://doi.org/10.3390/cells12182314

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop