Abstract

Neurodegeneration with Brain Iron Accumulation (NBIA) is a heterogeneous group of progressive neurodegenerative diseases characterized by iron deposition in the globus pallidus and the substantia nigra. As of today, 15 distinct monogenetic disease entities have been identified. The four most common forms are pantothenate kinase-associated neurodegeneration (PKAN), phospholipase A2 group VI (PLA2G6)-associated neurodegeneration (PLAN), beta-propeller protein-associated neurodegeneration (BPAN) and mitochondrial membrane protein-associated neurodegeneration (MPAN). Neurodegeneration with Brain Iron Accumulation disorders present with a wide spectrum of clinical symptoms such as movement disorder signs (dystonia, parkinsonism, chorea), pyramidal involvement (e.g., spasticity), speech disorders, cognitive decline, psychomotor retardation, and ocular abnormalities. Treatment remains largely symptomatic but new drugs are in the pipeline. In this review, we discuss the rationale of new compounds, summarize results from clinical trials, provide an overview of important results in cell lines and animal models and discuss the future development of disease-modifying therapies for NBIA disorders. A general mechanistic approach for treatment of NBIA disorders is with iron chelators which bind and remove iron. Few studies investigated the effect of deferiprone in PKAN, including a recent placebo-controlled double-blind multicenter trial, demonstrating radiological improvement with reduction of iron load in the basal ganglia and a trend to slowing of disease progression. Disease-modifying strategies address the specific metabolic pathways of the affected enzyme. Such tailor-made approaches include provision of an alternative substrate (e.g., fosmetpantotenate or 4′-phosphopantetheine for PKAN) in order to bypass the defective enzyme. A recent randomized controlled trial of fosmetpantotenate, however, did not show any significant benefit of the drug as compared to placebo, leading to early termination of the trials' extension phase. 4′-phosphopantetheine showed promising results in animal models and a clinical study in patients is currently underway. Another approach is the activation of other enzyme isoforms using small molecules (e.g., PZ-2891 in PKAN). There are also compounds which counteract downstream cellular effects. For example, deuterated polyunsaturated fatty acids (D-PUFA) may reduce mitochondrial lipid peroxidation in PLAN. In infantile neuroaxonal dystrophy (a subtype of PLAN), desipramine may be repurposed as it blocks ceramide accumulation. Gene replacement therapy is still in a preclinical stage.

Highlights

  • Neurodegeneration with Brain Iron Accumulation (NBIA) comprises a heterogeneous group of disorders characterized by iron accumulation mainly in the globus pallidus and the substantia nigra, visible on MR imaging

  • Dysfunctions in several pathophysiological pathways have been identified to be involved in NBIA disorders, including [1] coenzyme A biosynthesis, [2] lipid metabolism, [3] iron homeostasis, [4] autophagy and [5] other pathways of yet unknown function [2]

  • In fibroblasts derived from pantothenate kinase-associated neurodegeneration (PKAN) patients, 24 hours of treatment with 4′phosphopantetheine led to recovery of the expression of CoA synthase (COASY) and Tfrc [80]

Read more

Summary

INTRODUCTION

Neurodegeneration with Brain Iron Accumulation (NBIA) comprises a heterogeneous group of disorders characterized by iron accumulation mainly in the globus pallidus and the substantia nigra, visible on MR imaging. Dysfunctions in several pathophysiological pathways have been identified to be involved in NBIA disorders, including [1] coenzyme A biosynthesis, [2] lipid metabolism, [3] iron homeostasis, [4] autophagy and [5] other pathways of yet unknown function [2]. This diversity in etiology and pathogenesis calls for different therapeutic approaches for the individual NBIA disorders. Disease-specific approaches are discussed including [1] provision of alternative substrates downstream of the defect enzyme, [2] activation of isoenzymes, [3] counteracting downstream cellular effects, [4] gene therapies and [5] enzyme replacement therapies

Iron and Iron Chelation Therapies
Iron Chelation
Fosmetpantothenate Alternative substrate
Inhibition of lipid peroxidation
Enzyme replacement therapy
Findings
DISCUSSION
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call