Abstract
Background
The treatment of inborn errors of metabolism (IEM) has seen significant advances over the last decade. Many medicines have been developed and the survival rates of some patients with IEM have improved. Dosages of drugs used for the treatment of various IEM can be obtained from a range of sources but tend to vary among these sources. Moreover, the published dosages are not usually supported by the level of existing evidence, and they are commonly based on personal experience.
Methods
A literature search was conducted to identify key material published in English in relation to the dosages of medicines used for specific IEM. Textbooks, peer reviewed articles, papers and other journal items were identified. The PubMed and Embase databases were searched for material published since 1947 and 1974, respectively. The medications found and their respective dosages were graded according to their level of evidence, using the grading system of the Oxford Centre for Evidence-Based Medicine.
Results
83 medicines used in various IEM were identified. The dosages of 17 medications (21%) had grade 1 level of evidence, 61 (74%) had grade 4, two medications were in level 2 and 3 respectively, and three had grade 5.
Conclusions
To the best of our knowledge, this is the first review to address this matter and the authors hope that it will serve as a quickly accessible reference for medications used in this important clinical field.
Keywords: Inborn errors of metabolism, IEM, evidence based medicine, treatment, dosage
Introduction
Inborn errors of metabolism (IEM) are defined as monogenic diseases resulting in deficient activity in a single enzyme in a pathway of intermediary metabolism.1 Although IEM are individually rare, they are collectively common (incidence likely to be more than 1/1000).2 Over 500 human diseases due to IEM are now recognised, and this number is constantly increasing as new concepts and techniques become available for identifying biochemical phenotypes. The treatment of these disorders has seen significant advances over the past decade.2 The progress in understanding the pathophysiology of the majority of these disorders has led to the discovery of several new therapies that have made it possible to attenuate the severity of the clinical manifestations associated with many IEM. Despite the rarity of these disorders, there is growing emphasis on the use of evidence-based medicine (EBM) in the treatment of such conditions. However, EBM faces significant practical difficulties in the field of IEM, and experts have pointed out some of these challenges.3 4 Dosages of medications used for the treatment of various IEM can be obtained from different sources. However, these dosages vary between different sources1–7 and are not usually supported by the level of existing evidence.
This article intends to summarise the dosages of medications used in the treatment of IEM as supported by the best level of evidence that currently exists in the literature. To the best of our knowledge, this is the first review article that addresses this issue, and the authors hope that it will provide quick and easy access to a comprehensive list of medications used in this important clinical field.
Methods
A literature search was conducted to identify key material published in English in relation to dosages of medications for specific IEM. The medications found and their respective dosages were graded according to their level of evidence, using the grading system defined by the Oxford Centre for Evidence-based Medicine (OCEBM),8 which, in brief, assigns level 1 to randomised controlled trials (RCT), level 2 to cohort studies, level 3 to case–control studies, level 4 to case series and level 5 to expert opinion.8
Search strategy
We systemically identified all known metabolic disorders or IEM as defined in well established text books in the field, namely: The Metabolic and Molecular Bases of Inherited Disease,1 Inborn Metabolic Diseases: Diagnosis and Treatment,5 and the Physician’s Guide to the Treatment and Follow-up of Metabolic Diseases.6 The references for each medication dosage mentioned in the books were reviewed. The PubMed and Embase databases were then searched for published material not covered by these textbooks. Different search terms with appropriate subheadings and keywords were used. Database searches were constructed based on two concepts: specific IEM and treatment. Using boolean operators, subject headings and text words were combined in all permutations for each individual disorder. The results from searches were combined with studies identified from the textbooks mentioned above.
Inclusion criteria
Studies considered in this review are RCT and observational studies including cohort, case–control and cross-sectional studies and case reports. Textbooks and grey literature as far back in time as possible were also included. The selection of literature for inclusion in the review was based on examination of abstracts and indexing (subject headings) where available, and on full text or the table of contents if accessible. When different dose regimens were suggested by different sources, the authors chose the doses with the best available evidence. Non-English studies and duplicated papers were excluded.
Results
Eighty-three medications used in various IEM were identified. The dosages of 17 medications (21%) had grade 1 level of evidence, 61 (74%) had grade 4, two medications were in level 2 and 3 respectively and three had grade 5. Unsurprisingly, the majority of medications that achieved grade 1 level of evidence (8/17, 47%) were enzyme replacement therapies for various lysosomal storage disorders (see table 1). These medications were novel and therefore required Food and Drug Administration (FDA) approval, which in turn required a high level of evidence. Most of the medications classified as grade 4 were approved for other indications not related to IEM before their use for specific metabolic disorders. For example, arginine was already approved for the treatment of growth hormone deficiency in children prior to the discovery of its usefulness in the management of hyperammonaemia in several urea cycle disorders.9 Detailed information about representative examples of medications used in the treatment of IEM is given in tables 1–3. These include medications used in the treatment of lysosomal storage disorders, disorders of organic acids and amino acid metabolism or transport, and vitamins and co-factors used in the treatment of IEM. For each medication, the tables also provide information about indication in IEM, supply information, routes, dosages and level of evidence supporting its use. An additional list of medications used in the treatment of IEM is provided in the online supplementary table.
Table 1.
Drug | Indication(s) | How supplied* | Dose | Route | Evidence level | |
---|---|---|---|---|---|---|
1 | Agalsidase-α (Replagal) | Fabry disease | 1 mg/ml solution for infusion | 0.2 mg/kg every 2 weeks as IV infusion over 40 min11 16 | IV | 1b |
2 | Agalsidase-β (Fabrazyme) | Fabry disease | 5 mg and 35 mg single-use vials for reconstitution to yield (5 mg/ml) | 1 mg/kg every 2 weeks as IV infusion over 2–4 h11 17 | IV | 1b |
3 | Alglucosidase-α (Myozyme) | Pompe disease (GSD II) | 50 mg single-use vials for reconstitution to yield (5 mg/ml) | 20 mg/kg every 2 weeks as IV infusion over 4 h18–20 | IV | 1b |
4 | Cysteamine bitartrate (Cystagon) | Cystinosis | 50 mg and 150 mg capsules | Begin with 10 mg/kg/day and increase weekly until the maintenance dose (60–90 mg of free base/kg/day) or (1.3–1.95 g/m2 per day) is reached The recommended adult dose is 500 mg free base q6h; however, for both children and adults, the dose is titrated to reduce, if possible, leukocyte cystine concentration (measured 5–6 h after a dose) to below 1 nmol half-cystine/mg protein21–23 |
PO | 4 |
5 | Cysteamine hydrochloride (Cystoran) | Cystinosis | Ophthalmic drops | 0.55% solution with benzalkonium chloride 0.01% as a preservative: 10–12 times/day in each eye24 25 | Eyes | 1b |
6 | Galsulfase (Neglazyme) | Mucopolysaccharidosis VI | 5 mg/ml solution for injection | 1 mg/kg/week†26–28 | IV | 1b |
7 | Idursulfase (Elaprase) | Hunter syndrome (mucopolysaccharidosis II) | IV solution must be diluted in 100 ml of 0.9 sodium chloride injection, each vial contains 2 mg/ml solution of idursulfase protein (6 mg) in an extractable volume of 3 ml and for single use only | 0.5 mg/kg weekly over 1–3 h29–31 | IV | 1b |
8 | Imiglucerase (Cerezyme) | GD | 200 U and 400 U powder for reconstitution | Various regimens for non-neuropathic Gaucher disease, chronic, symptomatic: Adults: Usual dosage, 60 U/kg IV over 1–2 h every 2 weeks; may range from 2.5 U/kg 3 times weekly to 60 U/kg once every 2 weeks Children(a) Safety and effectiveness have not been established in children younger than 2 years of age (b) 2 years and older: usual dosage, 60 U/kg IV over 1–2 h every 2 weeks; may range from 2.5 U/kg 3 times weekly to 60 U/kg once every 2 weeks‡32 33 The absence of an improvement in visceral, haematological and biochemical markers within 6 months may indicate that a higher dose is required. If bone crises continue, the dose should be increased by at least 50%32 33 For type III GD, some clinicians recommend a higher dosage: 120 U/kg/2 weeks5 |
IV infusion over 1–2 h | 1b |
9 | Laronidase (Aldurazyme) | Mucopolysaccharidosis type 1 | 2.9 mg/5 ml solution for injection | 100 U/kg/week34 | IV | 1b |
10 | Miglustat (Zavesca) | GD in patients unable to receive intravenous ERT, NPC | 100 mg capsule | GD: 100 mg/kg/day TID35–38 NPC: 200 mg/kg/day TID39 40 |
PO | 4 |
11 | Velaglucerase α | GD | Powder for solution for injection, 200 U/vial and 400 U/vial | 60 U/kg administered every other week over 1 h§ Adjust based on disease activity |
IV | 1b |
†Product information for Naglazyme.
‡Product information: Cerezyme injection, imiglucerase injection.
§Product information for velaglucerase α. *Available under different brand names; sometimes in various dosage forms and strengths (only a few examples are given).
ERT, enzyme replacement therapy; GD, Gaucher disease; IV, intravenous; NPC, Niemann-Pick disease type C; PO, per os (by mouth); q6h, every 6 h; TID, three times a day.
Table 2.
Medication | Indication(s) | How supplied | Dose | Route | Evidence level | |
---|---|---|---|---|---|---|
1 | Arginine hydrochloride (R-Gene) | Acute management of hyperammonaemic crises in suspected or confirmed urea cycle disorders, except arginase deficiency | 10% solution for injection (100 mg/ml) | For suspected urea cycle or ASS or ASL, give: 600 mg/kg if (<20 kg) or 12 g/m2 if (>20 kg) as a loading dose over 90 min followed by 600 mg/kg if (<20 kg) or 12 g/m2 if (>20 kg) as maintenance infusion over 24 h. For OTC and CPS: 200 mg/kg if (<20 kg) or 4 g/m2 if (>20 kg) as a loading dose over 90 min followed by 200 mg/kg if (<20 kg) or 4 g/m2 if (>20 kg) as maintenance infusion over 24 h41 |
IV | 4 |
2 | l-Citrulline | CPS deficiency OTC deficiency LPI |
500 mg capsules, powder |
CPS and OTC deficiency: 170 mg/kg/day or 3.8 g/m2/day42–45 LPI: 100 mg/kg/day, however, the aim is to keep the citrulline within normal ranges In some patients 400 mg/kg/day were used46–48 |
PO | 4 |
3 | Dextromethorphan | Non-ketotic hyperglycinaemia | 15 mg tablets 15 mg/5 ml syrup |
5–35 mg/kg/day in 4 divided doses. Blood concentration can be monitored; the therapeutic level is not defined, but should be greater than zero (0) and lower than 100 nmol/l49–53 | PO | 4 |
4 | Glycine | Isovaleric acidaemia -HMG-CoA lyase deficiency. May be used in 3-methylcrotonyl glycinuria |
Powder | 250 mg/kg/day (150–300 mg/kg/day) in 4 divided doses5 6 54 55 | PO | 4 |
5 | Ketamine | NKH | 10 mg/ml, 50 mg/ml and 100 mg/ml solution for injection (maybe be used orally after mixing the dose with 0.2–0.3 ml/kg of cola or other beverages) | 1 mg/kg/day in 4 divided doses. Titrate it up to 30 mg/kg/day according to clinical and biochemical response56–59 | Oral or IV or IM | 4 |
6 | l-Carnitine | Primary and secondary carnitine deficiency | 300 mg/ml oral liquid 300 mg capsules 500 mg tablets 200 mg/ml solution for injection |
Acute crises (carnitine boluses): 100 mg/kg/dose 3–4 times daily, that is (300–400 mg/kg/day) should be given. Urine output should be appropriate prior to dosing (or haemofiltration be ongoing) Chronic: 100–300 mg/kg/day in 3 divided doses5 12 60 61 |
PO or IV | 4 |
7 | l-Isoleucine | MSUD | Powder | With the help of a metabolic dietician: 20–120 mg/kg/day. Dose is adjusted as necessary to achieve normal plasma amino acids levels62 | PO | 4 |
8 | l-Serine | 3-PGDH deficiency PSAT deficiency PSPH deficiency |
Powder | 3-PGDH:Infantile form: 500–600 mg/kg/day in 3 divided dosesJuvenile form: 100–150 mg/kg/day in 3 divided dosesPSAT: 500 mg/kg/dayPSPH: 200–300 mg/kg/dayHowever, the doses are varied aiming to normalise CSF serine63 | PO | 4 |
9 | l-Valine* | MSUD | 600 mg capsules, powder |
With the help of a metabolic dietician: 20–120 mg/kg/day. Dose is adjusted as necessary to achieve normal plasma amino acid levels62 | PO | 4 |
10 | Mercaptopropionylglycine (Tiopronin) | Cystinuria | Tablet, 100 mg | The dosage is wide at 15–50 mg/kg/day in 2 or 3 divided doses, maximum 1000 mg/day. However, the dose depends on monitoring free urine cystine level, so modify the dose in order to maintain a level below 200 mmol/mmol of creatinine5 6 64 65 | PO | 4 |
11 | Methionine | Several remethylation defects | Available in different dosage forms: capsules, powder and tablets | 40–50 mg/kg per day, adjust the dose to maintain upper normal ranges of plasma and CSF methionine.66 However, some investigators argue against its usage in such disorders because it may result in sustained hyperhomocystinaemia67 | PO | 4 |
12 | N-carbamoylglutamate (Carbaglu) | Unknown hyperammonaemia, NAGS deficiency, CPS-1 deficiency, propionic acidaemia or methylmalonic acidaemia | 200 mg tablet | 100–250 mg/kg/day, then adjusted individually in order to maintain normal ammonia plasma levels and divided into 2–4 doses68–75 Based on limited unpublished data, the maintenance dose is less than 100 mg/kg/day |
PO | 4 |
*Available under different brand names; sometimes in various dosage forms and strengths (only a few examples are given).
3-PGDH, 3-phosphoglycerate dehydrogenase; ASL, argininosuccinic acid lyase; ASS, argininosuccinic acid synthetase; CPS, carbamoyl phosphate synthetase; LPI, lysinuric protein intolerance; IM, intramuscular; MSUD, maple syrup urine disease; NAGS, N-acetylglutamate synthase; NKH, non-ketotic hyperglycinemia; OTC, ornithine transcarbamylase; PSAT, phosphoserine aminotransferase; PSPH, phosphoserine phosphatase.
Table 3.
Vitamin/co-factor | Indication(s) | How supplied | Dose | Route | Evidence level | |
---|---|---|---|---|---|---|
1 | Biotin | Cofactor for carboxylases Biotinidase deficiency BRBGD Multiple carboxylase deficiency |
10 mg and 50 mg tablets/capsules 1 mg capsule IV (available as part of a multivitamin complex) |
Biotinidase deficiency, co-factor for carboxylases and multiple carboxylase deficiency: 5–20 mg/day5
6
76
77 BRBGD: 5–10 mg/kg/day78–80 |
PO | 4 |
2 | Folic acid | Long term supplementation to compensate for the so-called methylfolate trap in remethylation defect | 1 mg and 5 mg tablets | Variable, 5–30 mg/day66 67 | PO | 4 |
3 | Folinic acid | DHPR deficiency UMP synthase deficiency (hereditary orotic aciduria) Methylene synthase deficiency Methionine synthase deficiency Hereditary folate malabsorption Cerebral folate transporter Folinic acid responsive seizure Remethylation defect |
5 mg, 10 mg, 15 mg and 25 mg tablets 10 mg/ml injection solution 50 mg, 100 mg, 200 mg and 350 mg powder for reconstitution (injection) |
Hereditary folate malabsorption: Adult: 150–200 mg/day PO once daily; infants and children: 50 mg or 10–15 mg/kg PO once daily81
82 Or 1.5–7.5 mg IM once daily.82–84 However, the dose should be adjusted in the individual to achieve a CSF folate level that is normal for age85 Other indications: 5–15 mg/day PO or IV BID5 86 |
PO or IV or IM | 4 |
4 | Hydroxocobalamin (vitamin B12) | Disorders of cobalamin metabolism | 1000 μg/ml solution for injection Tablets |
1 mg IM daily or oral dose: 10 mg once or twice daily5 6 87–89 |
IM or PO | 4 |
5 | Pyridoxine | Pyridoxine responsive CBS, PDE, pyridoxine responsive OAT, PH1 |
25 mg, 40 mg, 50 mg, 100 mg, 250 mg and 500 mg tablet Liquid, oral: 200 mg/5 ml |
CBS: 200 mg/day or the lowest dose that produces the maximum biochemical benefit (ie, lowest plasma homocysteine and methionine concentrations), as determined by measurement of total homocysteine and amino acid levels7
90 PDE: 100 mg IV, additional doses may be administered over the course of 30 min while observing for both a clinical and a possible electrographic response. If IV administration of pyridoxine is not possible for a first trial, pyridoxine is given orally/enterally with 30 mg/kg/day. Long term treatment: there are no clear-cut dosing recommendations, generally 15–30 mg/kg/day have been used in infants or up to 200 mg/day in neonates and 500 mg/day in adults91 OAT deficiency: 300–600 mg/day5 7 92 PH1: 5–10 mg/kg/day. Monitor oxalate and glycolate excretion and titrate the dose accordingly93 94 |
PO or IV (PDE) PO in others |
4 |
6 | Pyridoxal phosphate (PLP) | Pyridoxal phosphate-dependent seizures | 50 mg tablet | 30 mg/kg/day divided into 3 or 4 doses enterally, for 3–5 days 30–50 mg/kg/day divided into 4–6 doses86 95 |
NGT or PO | 4 |
7 | Riboflavin | GA1, MAD, mitochondrial complex 1 deficiency | 25 mg, 50 mg and 100 mg tablet 400 mg capsule |
GA1: There is no firm evidence that riboflavin improves the neurological outcome of GA.12 However, responsiveness to 100–150 mg/day divided into 2–3 doses has been demonstrated in a few patients96
97 MAD: 100–400 mg/day in 2–3 divided doses98 99 SCAD: 10 mg/kg/ day, divided into 3 doses with a maximum of 150 mg/day100 Mitochondrial complex 1 deficiency: 3–20 mg/kg/day divided into 3 doses6 101 |
PO | 4 |
8 | Sapropterin dihydrochloride (Kuvan) | HPA due to BH4 responsive PKU | 100 mg tablet | BH4 loading test: 20 mg/kg/dose once daily for 2 consecutive days Others: 10–20 mg/kg/day once daily102–104 Monitor phenylalanine levels and adjust the dose accordingly |
Oral | 1b |
9 | BH4 | BH4 loading test, disorders of BH4 synthesis, BH4 responsive PKU. Currently replaced by Kuvan | 50 mg tablet | BH4 loading test: 20 mg/kg/dose once daily for 2 consecutive days Others: 5–20 mg/kg/day, monitor phenylalanine levels and adjust the dose accordingly102 105 |
PO | 4 |
10 | Thiamine | Thiamine responsive MSUD, thiamine responsive pyruvate dehydrogenase deficiency and complex I deficiency | 50 mg, 100 mg, 250 mg and 500 mg tablet 100 mg/ml injection |
Various dosage have been used: 100 mg/day,106 10 mg/kg/day2; the dose ranges between 10 and 1000 mg/day5 62 107 108 | PO | 4 |
11 | Ubiquinone (coenzyme Q10) |
Primary CoQ10 deficiency | 50 mg, 100 mg and 200 mg soft gel capsule | The dosage employed is highly variable Adult: 200–600 mg QID, paediatrics: 2–15 mg/kg/day BID109
Other used: 30 mg/kg/day.110 As high as 2000 mg/day has been used111 |
PO | 4 |
12 | Vitamin C | GS Hawkinsinuria, tyrosinaemia type III and transient tyrosinemia of newborn |
100 mg tablet 100 mg/ml drops 1 g effervescent tablet |
GS: 100 mg/kg/day112–114
Others: 200–1000 mg/day |
PO | 4 |
13 | Vitamin E | GS | 100 mg capsule 50 U/ml drops |
GS: 10 mg/kg/day112–114 | PO | 4 |
BH4, tetrahydrobiopterin; BID, twice daily; BRBGD, biotin responsive basal ganglia disease; CBS, cystathionine β synthase deficiency; CSF, cerebrospinal fluid; DHPR, dihydropteridine reductase; GA1, glutaric aciduria; GS, glutathione synthetase deficiency; HPA, hyperphenylalaninemia; IM, intramuscular; IV, intravenous; MAD, multiple acyl-CoA dehydrogenase deficiency; MSUD, maple syrup urine disease; NGT, nasogastric tube; PDE, pyridoxine dependent epilepsy; PH1, primary hyperoxaluria type 1; PKU, phenylketonuria; PO, per os (by mouth); OAT, ornithine aminotransferase deficiency; SCAD, short-chain acyl-CoA dehydrogenase deficiency; QID, four times daily.
Discussion
EBM is defined as conscientious, explicit and judicious use of current best evidence in making decisions about the care of individual patients.10 In this systematic literature review, we used the current best evidence to select the dosages for medications that have been used for the last decade in various IEM. This will hopefully provide quick and easy access to information for clinicians in this field when determining appropriate dosages of medications that may not be routinely used.
Interestingly, most of the medications used in the field of IEM and their respective dosages only have level 4 evidence (74%). The gold standard for EBM is the RCT. However, obstacles to using RCTs to evaluate the efficacy of these medications include the rare nature of specific IEM, which makes it difficult to achieve significant statistical power when evaluating a specific treatment modality. Moreover, surrogate measures are often used when the disease is so rare or the desired outcome is so far in the future that it would take an unreasonably long follow-up period in order to obtain a sufficient number of clinical outcomes. Although the association between the surrogate measure and the true outcome may be biologically plausible, using the surrogate measure may produce misleading results if the association with the true outcome is not based on hard endpoints.11 Additionally, since many treatments for IEM have been used for decades, it may be difficult to go back and perform RCTs to prove their efficacy. A clear example of this issue is arginine hydrochloride for the treatment of urea cycle disorders.
The above observations are also reflected in a few established guidelines for the treatment of some IEM. For example, l-carnitine supplementation for patients with glutaric aciduria type 1 (GA1) was among the recommendations considered to be good clinical practice in a recently published guideline for the diagnosis and management of GA1.12 However, this was largely based on biological plausibility and expert opinion.12 On the other hand, although the American College of Medical Genetics recommendation of enzyme replacement therapy in patients with Fabry disease relies on level 1b evidence, adjuvant therapy recommendations rely on evidence obtained from studies not carried out in patients with Fabry disease.11 13
IEM are considered orphan diseases, a situation that causes difficulties in conducting RCTs because of the small number of patients found and the paucity of funding from pharmaceutical companies relative to common diseases. The FDA relaxed their role in the approval of new drugs used for the treatment of specific IEM and considers them as orphan drugs to be prescribed under compassionate use. Some of the medications currently used in the treatment of IEM have benefited from this relaxation, including carglumic acid (Carbaglu) for the treatment of acute hyperammonaemia resulting from a deficiency of the enzyme N-acetylglutamate synthase, and alglucosidase α (Myozyme) which was approved in April 2006 as enzyme replacement therapy for Pompe disease.
Despite the various challenges that face the development of evidence-based practice in patients with IEM, therapeutic trials are still being conducted and may pave the way for more evidence-based therapeutic interventions for these disorders. Substrate reduction therapies and molecular chaperone therapies are examples of two therapeutic modalities with active ongoing clinical trials in some IEM (http://www.clinicaltrial.gov).
This review has several limitations and gaps and caution should be used when accessing the information in the online supplementary table. These limitations include the fact that knowledge of IEM is continuously and dynamically changing and it may not be long before the information in this review is outdated. Secondly, the existing tools frequently used for EBM such as the OCEBM grading system, were mainly designed for common disorders rather than rare diseases. This renders critical appraisal of the evidence very difficult and may be inaccurate or misleading. For example, although many studies reach level 1 evidence, all were measuring surrogate markers as the primary end point, which may not necessarily correlate with significant clinical outcomes. For example, dichloroacetate has been used for the treatment of congenital lactic acidosis in case series. When examined in an RCT, it was shown to reduce lactate, a surrogate marker, but was not associated with improved neurological or clinical outcome. Third, some medications have reached level 1 evidence when used for disorders other than IEM, but the evidence supporting their use in metabolic disorders is derived only from case reports. For example, the use of baclofen to treat spasticity in patients with glutaric aciduria type 112 is based on its use for treating spasticity in children with cerebral palsy.15
In summary, clinicians face several challenges and obstacles as they try to select the appropriate dosages of medications to treat their patients with IEM. Using the currently available evidence for these decisions may help resolve some of these difficulties until standard guidelines and recommendations are published.
Supplementary Material
Footnotes
Contributors: MA: carried out the majority of the work, and prepared and drafted the initial manuscript; KA-T: reviewed the manuscript and the dosages and classification of evidence for every medication; HM: prepared the dosage forms and critically reviewed the article; WE: edited the manuscript; MA-J: reviewed the dosage forms and reviewed the classification of evidence for every medication. All authors approved the final manuscript as submitted.
Competing interests: None.
Provenance and peer review: Not commissioned; externally peer reviewed.
References
- 1.Scriver CR. The metabolic and molecular bases of inherited disease. 8th edn New York; London: McGraw-Hill, 2001 [Google Scholar]
- 2.Campeau PM, Scriver CR, Mitchell JJ. A 25-year longitudinal analysis of treatment efficacy in inborn errors of metabolism. Mol Genet Metab 2008;95:11–16 [DOI] [PubMed] [Google Scholar]
- 3.Wilcken B. Rare diseases and the assessment of intervention: what sorts of clinical trials can we use? J Inherit Metab Dis 2001;24:291–8 [DOI] [PubMed] [Google Scholar]
- 4.Winter SC. Treatment of carnitine deficiency. J Inherit Metab Dis 2003;26:171–80 [DOI] [PubMed] [Google Scholar]
- 5.Fernandes J, Saudubray JM, Van den Berghe G. Inborn metabolic diseases: diagnosis and treatment. 3rd rev. edn Berlin; New York; London: Springer, 2000 [Google Scholar]
- 6.Blau N. Physician's guide to the treatment and follow-up of metabolic diseases. Berlin: Springer, 2006 [Google Scholar]
- 7.Liu H, Dong H, Robertson K, et al. DNA methylation suppresses expression of the urea cycle enzyme carbamoyl phosphate synthetase 1 (CPS1) in human hepatocellular carcinoma. Am J Pathol 2011;178:652–61 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Centre for Evidence-Based Medicine. Secondary Centre for Evidence-Based Medicine 2009. http://www.cebm.net/index.aspx?o=1025. [Google Scholar]
- 9.Loche S, Carta D, Muntoni AC, et al. Oral administration of arginine enhances the growth hormone response to growth hormone releasing hormone in short children. Acta Paediatr 1993;82:883–4 [DOI] [PubMed] [Google Scholar]
- 10.Sackett DL, Rosenberg WM, Gray JA, et al. Evidence based medicine: what it is and what it isn't. BMJ 1996;312:71–2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Alfadhel M, Sirrs S. Enzyme replacement therapy for Fabry disease: some answers but more questions. Ther Clin Risk Manag 2011;7:69–82 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Kolker S, Christensen E, Leonard JV, et al. Diagnosis and management of glutaric aciduria type I—revised recommendations. J Inherit Metab Dis 2011;34:677–94 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Eng CM, Germain DP, Banikazemi M, et al. Fabry disease: guidelines for the evaluation and management of multi-organ system involvement. Genet Med 2006;8:539–48 [DOI] [PubMed] [Google Scholar]
- 14.Mokhtarani M, Diaz GA, Rhead W, et al. Urinary phenylacetylglutamine as dosing biomarker for patients with urea cycle disorders. Mol Genet Metab 2012;107:308–14 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Scheinberg A, Hall K, Lam LT, et al. Oral baclofen in children with cerebral palsy: a double-blind cross-over pilot study. J Paediatr Child Health 2006;42:715–20 [DOI] [PubMed] [Google Scholar]
- 16.Schiffmann R, Kopp JB, Austin HA, III, et al. Enzyme replacement therapy in Fabry disease: a randomized controlled trial. JAMA 2001;285:2743–9 [DOI] [PubMed] [Google Scholar]
- 17.Banikazemi M, Bultas J, Waldek S, et al. Agalsidase-beta therapy for advanced Fabry disease: a randomized trial. Ann Intern Med 2007;146:77–86 [DOI] [PubMed] [Google Scholar]
- 18.Chien YH, Hwu WL. A review of treatment of Pompe disease in infants. Biol Targets Ther 2007;1:195–201 [PMC free article] [PubMed] [Google Scholar]
- 19.Kishnani PS, Corzo D, Nicolino M, et al. Recombinant human acid [alpha]-glucosidase: major clinical benefits in infantile-onset Pompe disease. Neurology 2007;68:99–109 [DOI] [PubMed] [Google Scholar]
- 20.van der Ploeg AT, Clemens PR, Corzo D, et al. A randomized study of alglucosidase alfa in late-onset Pompe's disease. N Engl J Med 2010;362:1396–406 [DOI] [PubMed] [Google Scholar]
- 21.Belldina EB, Huang MY, Schneider JA, et al. Steady-state pharmacokinetics and pharmacodynamics of cysteamine bitartrate in paediatric nephropathic cystinosis patients. Br J Clin Pharmacol 2003;56:520–5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Gahl WA, Thoene JG, Schneider JA. Cystinosis. N Engl J Med 2002;347:111–21 [DOI] [PubMed] [Google Scholar]
- 23.Kleta R, Gahl WA. Pharmacological treatment of nephropathic cystinosis with cysteamine. Expert Opin Pharmacother 2004;5:2255–62 [DOI] [PubMed] [Google Scholar]
- 24.Kaiser-Kupfer MI, Gazzo MA, Datiles MB, et al. A randomized placebo-controlled trial of cysteamine eye drops in nephropathic cystinosis. Arch Ophthalmol 1990;108:689–93 [DOI] [PubMed] [Google Scholar]
- 25.Tsilou E, Zhou M, Gahl W, et al. Ophthalmic manifestations and histopathology of infantile nephropathic cystinosis: report of a case and review of the literature. Surv Ophthalmol 2007;52:97–105 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Harmatz P, Giugliani R, Schwartz I, et al. Enzyme replacement therapy for mucopolysaccharidosis VI: a phase 3, randomized, double-blind, placebo-controlled, multinational study of recombinant human N-acetylgalactosamine 4-sulfatase (recombinant human arylsulfatase B or rhASB) and follow-on, open-label extension study. J Pediatr 2006;148:533–9 [DOI] [PubMed] [Google Scholar]
- 27.Harmatz P, Kramer WG, Hopwood JJ, et al. Pharmacokinetic profile of recombinant human N-acetylgalactosamine 4-sulphatase enzyme replacement therapy in patients with mucopolysaccharidosis VI (Maroteaux-Lamy syndrome): a phase I/II study. Acta Paediatr Suppl 2005;94:61–8 [DOI] [PubMed] [Google Scholar]
- 28.Harmatz P, Whitley CB, Waber L, et al. Enzyme replacement therapy in mucopolysaccharidosis VI (Maroteaux-Lamy syndrome). J Pediatr 2004;144:574–80 [DOI] [PubMed] [Google Scholar]
- 29.Muenzer J, Gucsavas-Calikoglu M, McCandless SE, et al. A phase I/II clinical trial of enzyme replacement therapy in mucopolysaccharidosis II (Hunter syndrome). Mol Genet Metab 2007;90:329–37 [DOI] [PubMed] [Google Scholar]
- 30.Muenzer J, Wraith JE, Beck M, et al. A phase II/III clinical study of enzyme replacement therapy with idursulfase in mucopolysaccharidosis II (Hunter syndrome). Genet Med 2006;8:465–73 [DOI] [PubMed] [Google Scholar]
- 31.Zareba G. Idursulfase in Hunter syndrome treatment. Drugs Today (Barc) 2007;43:759–67 [DOI] [PubMed] [Google Scholar]
- 32.Baldellou A, Andria G, Campbell PE, et al. Paediatric non-neuronopathic Gaucher disease: recommendations for treatment and monitoring. Eur J Pediatr 2004;163:67–75 [DOI] [PubMed] [Google Scholar]
- 33.Charrow J, Andersson HC, Kaplan P, et al. Enzyme replacement therapy and monitoring for children with type 1 Gaucher disease: consensus recommendations. J Pediatr 2004;144:112–20 [DOI] [PubMed] [Google Scholar]
- 34.Wraith JE, Clarke LA, Beck M, et al. Enzyme replacement therapy for mucopolysaccharidosis I: a randomized, double-blinded, placebo-controlled, multinational study of recombinant human alpha-L-iduronidase (laronidase). J Pediatr 2004;144:581–8 [DOI] [PubMed] [Google Scholar]
- 35.Cox T, Lachmann R, Hollak C, et al. Novel oral treatment of Gaucher's disease with N-butyldeoxynojirimycin (OGT 918) to decrease substrate biosynthesis. Lancet 2000;355:1481–5 [DOI] [PubMed] [Google Scholar]
- 36.Pastores GM, Barnett NL, Kolodny EH. An open-label, noncomparative study of miglustat in type I Gaucher disease: efficacy and tolerability over 24 months of treatment. Clin Ther 2005;27:1215–27 [DOI] [PubMed] [Google Scholar]
- 37.Weinreb NJ, Barranger JA, Charrow J, et al. Guidance on the use of miglustat for treating patients with type 1 Gaucher disease. Am J Hematol 2005;80:223–9 [DOI] [PubMed] [Google Scholar]
- 38.Zimran A, Elstein D. Gaucher disease and the clinical experience with substrate reduction therapy. Philos Trans R Soc Lond B Biol Sci 2003;358:961–6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Patterson MC, Vecchio D, Jacklin E, et al. Long-term miglustat therapy in children with Niemann-Pick disease type C. J Child Neurol 2010;25:300–5 [DOI] [PubMed] [Google Scholar]
- 40.Patterson MC, Vecchio D, Prady H, et al. Miglustat for treatment of Niemann-Pick C disease: a randomised controlled study. Lancet Neurol 2007;6:765–72 [DOI] [PubMed] [Google Scholar]
- 41.Batshaw ML, MacArthur RB, Tuchman M. Alternative pathway therapy for urea cycle disorders: twenty years later. J Pediatr 2001;138:S46–54; discussion S-5 [DOI] [PubMed] [Google Scholar]
- 42.Brusilow SW. Arginine, an indispensable amino acid for patients with inborn errors of urea synthesis. J Clin Invest 1984;74:2144–8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Maestri NE, Brusilow SW, Clissold DB, et al. Long-term treatment of girls with ornithine transcarbamylase deficiency. N Engl J Med 1996;335:855–9 [DOI] [PubMed] [Google Scholar]
- 44.Tuchman M, Ahrens M, Barsotti R, et al. Consensus statement from a conference for the management of patients with urea cycle disorders. J Pediatr 2001;138:S1–5 [DOI] [PubMed] [Google Scholar]
- 45.Berry GT, Steiner RD. Long-term management of patients with urea cycle disorders. J Pediatr 2001;138:S56–60; discussion S-1 [DOI] [PubMed] [Google Scholar]
- 46.Carpenter TO, Levy HL, Holtrop ME, et al. Lysinuric protein intolerance presenting as childhood osteoporosis. Clinical and skeletal response to citrulline therapy. N Engl J Med 1985;312:290–4 [DOI] [PubMed] [Google Scholar]
- 47.Sebastio G, Sperandeo MP, Andria G. Lysinuric protein intolerance: reviewing concepts on a multisystem disease. Am J Med Genet C Semin Med Genet 2011;157:54–62 [DOI] [PubMed] [Google Scholar]
- 48.Tanner LM, Nanto-Salonen K, Venetoklis J, et al. Nutrient intake in lysinuric protein intolerance. J Inherit Metab Dis 2007;30:716–21 [DOI] [PubMed] [Google Scholar]
- 49.Alemzadeh R, Gammeltoft K, Matteson K. Efficacy of low-dose dextromethorphan in the treatment of nonketotic hyperglycinemia. Pediatrics 1996;97:924–6 [PubMed] [Google Scholar]
- 50.Atay E, Bozaykut A, Sezer G. Four cases of neonatal non-ketotic hyperglycinaemia. Ann Trop Paediatr 2004;24:345–7 [DOI] [PubMed] [Google Scholar]
- 51.Chien YH, Hsu CC, Huang A, et al. Poor outcome for neonatal-type nonketotic hyperglycinemia treated with high-dose sodium benzoate and dextromethorphan. J Child Neurol 2004;19:39–42 [DOI] [PubMed] [Google Scholar]
- 52.Hamosh A, Maher JF, Bellus GA, et al. Long-term use of high-dose benzoate and dextromethorphan for the treatment of nonketotic hyperglycinemia. J Pediatr 1998;132:709–13 [DOI] [PubMed] [Google Scholar]
- 53.Hamosh A, McDonald JW, Valle D, et al. Dextromethorphan and high-dose benzoate therapy for nonketotic hyperglycinemia in an infant. J Pediatr 1992;121:131–5 [DOI] [PubMed] [Google Scholar]
- 54.Fries MH, Rinaldo P, Schmidt-Sommerfeld E, et al. Isovaleric acidemia: response to a leucine load after three weeks of supplementation with glycine, L-carnitine, and combined glycine-carnitine therapy. J Pediatr 1996;129:449–52 [DOI] [PubMed] [Google Scholar]
- 55.Chalmers RA, de Sousa C, Tracey BM, et al. L-carnitine and glycine therapy in isovaleric acidaemia. J Inherit Metab Dis 1985;8 Suppl 2:141–2 [DOI] [PubMed] [Google Scholar]
- 56.Korman SH, Wexler ID, Gutman A, et al. Treatment from birth of nonketotic hyperglycinemia due to a novel GLDC mutation. Ann Neurol 2006;59:411–15 [DOI] [PubMed] [Google Scholar]
- 57.Lu FL, Wang PJ, Hwu WL, et al. Neonatal type of nonketotic hyperglycinemia. Pediatr Neurol 1999;20:295–300 [DOI] [PubMed] [Google Scholar]
- 58.Ohya Y, Ochi N, Mizutani N, et al. Nonketotic hyperglycinemia: treatment with NMDA antagonist and consideration of neuropathogenesis. Pediatr Neurol 1991;7:65–8 [DOI] [PubMed] [Google Scholar]
- 59.Tegtmeyer-Metzdorf H, Roth B, Gunther M, et al. Ketamine and strychnine treatment of an infant with nonketotic hyperglycinaemia. Eur J Pediatr 1995;154:649–53 [DOI] [PubMed] [Google Scholar]
- 60.Chapman KA, Gropman A, Macleod E, et al. Acute management of propionic acidemia. Mol Genet Metab 2012;105:16–25 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Sutton VR, Chapman KA, Gropman AL, et al. Chronic management and health supervision of individuals with propionic acidemia. Mol Genet Metab 2012;105:26–33 [DOI] [PubMed] [Google Scholar]
- 62.Strauss KA, Puffenberger EG, Morton DH. Maple syrup urine disease. In: Pagon RA, Bird TD, Dolan CR, Stephens K, Adam MP, eds. GeneReviews [Internet]. Seattle (WA): University of Washington, Seattle; 1993–2006 [Google Scholar]
- 63.Tabatabaie L, Klomp LW, Berger R, et al. L-serine synthesis in the central nervous system: a review on serine deficiency disorders. Mol Genet Metab 2010;99:256–62 [DOI] [PubMed] [Google Scholar]
- 64.Strologo L Dello, Rizzoni G. Cystinuria. Acta Paediatr Suppl 2006;95:31–3 [DOI] [PubMed] [Google Scholar]
- 65.Knoll T, Zollner A, Wendt-Nordahl G, et al. Cystinuria in childhood and adolescence: recommendations for diagnosis, treatment, and follow-up. Pediatr Nephrol 2005;20:19–24 [DOI] [PubMed] [Google Scholar]
- 66.Schiff M, Benoist JF, Tilea B, et al. Isolated remethylation disorders: do our treatments benefit patients? J Inherit Metab Dis 2011;34:137–45 [DOI] [PubMed] [Google Scholar]
- 67.de Baulny H Ogier, Gerard M, Saudubray JM, et al. Remethylation defects: guidelines for clinical diagnosis and treatment. Eur J Pediatr 1998;157 Suppl 2:S77–83 [DOI] [PubMed] [Google Scholar]
- 68.Guffon N, Vianey-Saban C, Bourgeois J, et al. A new neonatal case of N-acetylglutamate synthase deficiency treated by carbamylglutamate. J Inherit Metab Dis 1995;18:61–5 [DOI] [PubMed] [Google Scholar]
- 69.Haberle J. Role of carglumic acid in the treatment of acute hyperammonemia due to N-acetylglutamate synthase deficiency. Ther Clin Risk Manag 2011;7:327–32 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Hinnie J, Colombo JP, Wermuth B, et al. N-Acetylglutamate synthetase deficiency responding to carbamylglutamate. J Inherit Metab Dis 1997;20:839–40 [DOI] [PubMed] [Google Scholar]
- 71.Levrat V, Forest I, Fouilhoux A, et al. Carglumic acid: an additional therapy in the treatment of organic acidurias with hyperammonemia? Orphanet J Rare Dis 2008;3:2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Morris AA, Richmond SW, Oddie SJ, et al. N-acetylglutamate synthetase deficiency: favourable experience with carbamylglutamate. J Inherit Metab Dis 1998;21:867–8 [DOI] [PubMed] [Google Scholar]
- 73.Plecko B, Erwa W, Wermuth B. Partial N-acetylglutamate synthetase deficiency in a 13-year-old girl: diagnosis and response to treatment with N-carbamylglutamate. Eur J Pediatr 1998;157:996–8 [DOI] [PubMed] [Google Scholar]
- 74.de Miguel S Fernandez, Gimeno Diaz de Atauri A, Peral R Torres, et al. N-carbamyl glutamate treatment in hyperammoniemia decompensated propionic acidaemia. An Pediatr (Barc) 2009;71:579–80 [DOI] [PubMed] [Google Scholar]
- 75.Schwahn BC, Pieterse L, Bisset WM, et al. Biochemical efficacy of N-carbamylglutamate in neonatal severe hyperammonaemia due to propionic acidaemia. Eur J Pediatr 2010;169:133–4 [DOI] [PubMed] [Google Scholar]
- 76.Wastell HJ, Bartlett K, Dale G, et al. Biotinidase deficiency: a survey of 10 cases. Arch Dis Child 1988;63:1244–9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Heron B, Gautier A, Dulac O, et al. Biotinidase deficiency. Progressive encephalopathy curable with biotin. Arch Fr Pediatr 1993;50:875–8 [PubMed] [Google Scholar]
- 78.El-Hajj TI, Karam PE, Mikati MA. Biotin-responsive basal ganglia disease: case report and review of the literature. Neuropediatrics 2008;39:268–71 [DOI] [PubMed] [Google Scholar]
- 79.Bindu PS, Noone ML, Nalini A, et al. Biotin-responsive basal ganglia disease: a treatable and reversible neurological disorder of childhood. J Child Neurol 2009;24:750–2 [DOI] [PubMed] [Google Scholar]
- 80.Ozand PT, Gascon GG, Al Essa M, et al. Biotin-responsive basal ganglia disease: a novel entity. Brain 1998;121(Pt 7):1267–79 [DOI] [PubMed] [Google Scholar]
- 81.Geller J, Kronn D, Jayabose S, et al. Hereditary folate malabsorption: family report and review of the literature. In: Pagon RA, Bird TD, Dolan CR, Stephens K, Adam MP, eds. SourceGeneReviews [Internet]. Seattle (WA): University of Washington, Seattle; 1993–2008 [Google Scholar]
- 82.Diop-Bove N, Kronn D, Goldman ID. Hereditary folate malabsorption. 1993. Published Online First: 20 March 2010. doi:NBK1673 [bookaccession]. [Google Scholar]
- 83.Poncz M, Cohen A. Long-term treatment of congenital folate malabsorption. J Pediatr 1996;129:948. [DOI] [PubMed] [Google Scholar]
- 84.Poncz M, Colman N, Herbert V, et al. Therapy of congenital folate malabsorption. J Pediatr 1981;98:76–9 [DOI] [PubMed] [Google Scholar]
- 85.Borzutzky A, Crompton B, Bergmann AK, et al. Reversible severe combined immunodeficiency phenotype secondary to a mutation of the proton-coupled folate transporter. Clin Immunol 2009;133:287–94 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Gospe SM., Jr Neonatal vitamin-responsive epileptic encephalopathies. Chang Gung Med J 2010;33:1–12 [PubMed] [Google Scholar]
- 87.Andersson HC, Shapira E. Biochemical and clinical response to hydroxocobalamin versus cyanocobalamin treatment in patients with methylmalonic acidemia and homocystinuria (cblC). J Pediatr 1998;132:121–4 [DOI] [PubMed] [Google Scholar]
- 88.Bartholomew DW, Batshaw ML, Allen RH, et al. Therapeutic approaches to cobalamin-C methylmalonic acidemia and homocystinuria. J Pediatr 1988;112:32–9 [DOI] [PubMed] [Google Scholar]
- 89.Ninan TK, Thom H, Russell G. Oral vitamin B12 treatment of cobalamin-responsive methylmalonic aciduria. J Inherit Metab Dis 1992;15:939–40 [DOI] [PubMed] [Google Scholar]
- 90.Picker JD, Levy HL. Homocystinuria caused by cystathionine Beta-synthase deficiency. In: Pagon RA, Bird TD, Dolan CR, Stephens K, Adam MP, eds. SourceGeneReviews [Internet]. Seattle (WA): University of Washington, Seattle; 1993–2004 [Google Scholar]
- 91.Stockler S, Plecko B, Gospe SM, Jr, et al. Pyridoxine dependent epilepsy and antiquitin deficiency: clinical and molecular characteristics and recommendations for diagnosis, treatment and follow-up. Mol Genet Metab 2011;104:48–60 [DOI] [PubMed] [Google Scholar]
- 92.Ohkubo Y, Ueta A, Ito T, et al. Vitamin B6-responsive ornithine aminotransferase deficiency with a novel mutation G237D. Tohoku J Exp Med 2005;205:335–42 [DOI] [PubMed] [Google Scholar]
- 93.Bobrowski AE, Langman CB. The primary hyperoxalurias. Semin Nephrol 2008;28:152–62 [DOI] [PubMed] [Google Scholar]
- 94.Monico CG, Rossetti S, Olson JB, et al. Pyridoxine effect in type I primary hyperoxaluria is associated with the most common mutant allele. Kidney Int 2005;67:1704–9 [DOI] [PubMed] [Google Scholar]
- 95.Clayton PT. B6-responsive disorders: a model of vitamin dependency. J Inherit Metab Dis 2006;29:317–26 [DOI] [PubMed] [Google Scholar]
- 96.Brandt NJ, Gregersen N, Christensen E, et al. Treatment of glutaryl-CoA dehydrogenase deficiency (glutaric aciduria). Experience with diet, riboflavin, and GABA analogue. J Pediatr 1979;94:669–73 [DOI] [PubMed] [Google Scholar]
- 97.Chalmers RA, Bain MD, Zschocke J. Riboflavin-responsive glutaryl CoA dehydrogenase deficiency. Mol Genet Metab 2006;88:29–37 [DOI] [PubMed] [Google Scholar]
- 98.Izumi R, Suzuki N, Nagata M, et al. A case of late onset riboflavin-responsive multiple acyl-CoA dehydrogenase deficiency manifesting as recurrent rhabdomyolysis and acute renal failure. Intern Med 2011;50:2663–8 [DOI] [PubMed] [Google Scholar]
- 99.Olsen RK, Olpin SE, Andresen BS, et al. ETFDH mutations as a major cause of riboflavin-responsive multiple acyl-CoA dehydrogenation deficiency. Brain 2007;130:2045–54 [DOI] [PubMed] [Google Scholar]
- 100.van Maldegem BT, Duran M, Wanders RJ, et al. Flavin adenine dinucleotide status and the effects of high-dose riboflavin treatment in short-chain acyl-CoA dehydrogenase deficiency. Pediatr Res 2010;67:304–8 [DOI] [PubMed] [Google Scholar]
- 101.Scholte HR, Busch HF, Bakker HD, et al. Riboflavin-responsive complex I deficiency. Biochim Biophys Acta 1995;1271:75–83 [DOI] [PubMed] [Google Scholar]
- 102.Blau N, Hennermann JB, Langenbeck U, et al. Diagnosis, classification, and genetics of phenylketonuria and tetrahydrobiopterin (BH4) deficiencies. Mol Genet Metab 2011;104 Suppl:S2–9 [DOI] [PubMed] [Google Scholar]
- 103.Levy HL, Milanowski A, Chakrapani A, et al. Efficacy of sapropterin dihydrochloride (tetrahydrobiopterin, 6R-BH4) for reduction of phenylalanine concentration in patients with phenylketonuria: a phase III randomised placebo-controlled study. Lancet 2007;370:504–10 [DOI] [PubMed] [Google Scholar]
- 104.Anon. Sapropterin (Kuvan) for phenylketonuria. Med Lett Drugs Ther 2008;50:43–4 [PubMed] [Google Scholar]
- 105.Blau N, van Spronsen FJ, Levy HL. Phenylketonuria. Lancet 2010;376:1417–27 [DOI] [PubMed] [Google Scholar]
- 106.Morton DH, Strauss KA, Robinson DL, et al. Diagnosis and treatment of maple syrup disease: a study of 36 patients. Pediatrics 2002;109:999–1008 [DOI] [PubMed] [Google Scholar]
- 107.Pastoris O, Savasta S, Foppa P, et al. Pyruvate dehydrogenase deficiency in a child responsive to thiamine treatment. Acta Paediatr 1996;85:625–8 [DOI] [PubMed] [Google Scholar]
- 108.Di Rocco M, Lamba LD, Minniti G, et al. Outcome of thiamine treatment in a child with Leigh disease due to thiamine-responsive pyruvate dehydrogenase deficiency. Eur J Paediatr Neurol 2000;4:115–17 [DOI] [PubMed] [Google Scholar]
- 109.Santa KM. Treatment options for mitochondrial myopathy, encephalopathy, lactic acidosis, and stroke-like episodes (MELAS) syndrome. Pharmacotherapy 2010;30:1179–96 [DOI] [PubMed] [Google Scholar]
- 110.Trevisson E, DiMauro S, Navas P, et al. Coenzyme Q deficiency in muscle. Current Opin Neurol 2011;24:449–56 [DOI] [PubMed] [Google Scholar]
- 111.Dimauro S, Rustin P. A critical approach to the therapy of mitochondrial respiratory chain and oxidative phosphorylation diseases. Biochim Biophys Acta 2009;1792:1159–67 [DOI] [PubMed] [Google Scholar]
- 112.Njalsson R. Glutathione synthetase deficiency. Cell Mol Life Sci 2005;62:1938–45 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Njalsson R, Ristoff E, Carlsson K, et al. Genotype, enzyme activity, glutathione level, and clinical phenotype in patients with glutathione synthetase deficiency. Hum Genet 2005;116:384–9 [DOI] [PubMed] [Google Scholar]
- 114.Ristoff E, Mayatepek E, Larsson A. Long-term clinical outcome in patients with glutathione synthetase deficiency. J Pediatr 2001;139:79–84 [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.