Acessibilidade / Reportar erro

Small Molecules: Substrate Inhibitors, Chaperones, Stop-Codon Read Through, and Beyond

Abstract

Lysosomal storage disorders are rare genetic disorders due to deficient lysosomal activity, which leads to progressive accumulation of nonmetabolized substrates. Patient’s clinical outcomes have significantly improved since the advent of enzyme replacement therapy, even though this therapeutic approach presents important limitations, such as immune reactions, low bioavailability of recombinant enzymes, and incapability to reach the central nervous system. New strategies based on gene therapy or small molecules have been proposed and tested as an alternative to enzyme replacement therapy or to complement it. Small molecules are orally administrated, no antigenic compound that can diffuse across cell membranes and distribute in steady-state concentrations, also reaching the central nervous system. Substrate reduction therapy, pharmacological chaperones, and stop-codon read-through enhancers are small molecules currently available for the treatment of lysosomal storage disorders. This article describes the characteristics of this class of compounds and the possible strategies to improve their efficiency in future development.

Keywords
lysosomal storage disorders; substrate reduction therapy; pharmacological chaperones; stop-codon read through; cotreatment with enzyme replacement therapy

Introduction

Lysosomal storage disorders (LSDs) are a group of more than 50 different diseases due to a functional deficiency of a lysosomal protein (ie, acidic hydrolases, activator transporters, or nonlysosomal proteins necessary for lysosomal function), which leads to the accumulation of a variety of substrates, such as glycosaminoglycans (GAGs), glycosphingolipids, glycogen, oligosaccharides, cholesterol, peptides, and glycoproteins.11 Kingma, SD, Bodamer, OA, Wijburg, FA. Epidemiology and diagnosis of lysosomal storage disorders; challenges of screening. Best Pract Res Clin Endocrinol Metab. 2015;29(2):145–157. A broad clinical presentation is associated with LSDs, which includes visceral, ocular, hematological, skeletal, and neurological manifestations. These outcomes eventually affect lifespan, as well as physical and intellectual performance of the patients, determining an important challenge in terms of public health, social rights, and economic costs for the society. Even if LSDs are considered singularly as rare diseases (incidence ranging between 1:7000 and 1:1 000 000), collectively these pathologies are as frequent as 1:5000 to 1:7000 live births.22 Fuller, M, Meikle, PJ, Hopwood, JJ. Epidemiology of lysosomal storage diseases: an overview. In: Metha, A, Beck, M, Sunder-Plassmann, G, eds. Fabry Diseases: Perspectives From 5 Years of FOS. Oxford, UK: Oxford Pharmagenesis; 2006.

Nowadays, the most frequent LSDs (ie, Gaucher disease [GD], Fabry disease [FD], Pompe disease [PD], mucopolysaccharidosis [MPS], type I, II, IV, VI, and acid lipase deficiency) are treated by enzyme replacement therapy (ERT) or substrate reduction therapy (SRT).33 Beck, M. New therapeutic options for lysosomal storage disorders: enzyme replacement, small molecule and gene therapy. Hum Genet. 2007;121(1):1–22.

The ERT consists of periodic intravenous administration of the recombinant form of the human wild-type enzyme, which can be internalized by the cells with deficient lysosomal activity, through the mannose 6-phosphate receptor (MP6). The ERT has been used in the clinic for around 15 years with relevant benefits in patients’ quality of life, allowing, as an example, the normalization of hematologic parameters and organ volumes in GD or the decrease in neuropathic pain crisis in patients with FD.44 Charrow, J, Scott, CR. Long-term treatment outcomes in Gaucher disease. Am J Hematol. 2015;90(suppl 1):S19–S24.,55 Arends, M., Hollak, CE, Biegstraaten, M. Quality of life in patients with Fabry disease: a systematic review of the literature. Orphanet J Rare Dis. 2015;10:77. Nevertheless, ERT still presents important limitations, such as the low bioavailability and half-life of the drugs, the possibility to induce immunologic intolerance, and the incapability to cross the blood–brain barrier (BBB).66 Ortolano, S, Vieitez, I, Navarro, C, Spuch, C. Treatment of lysosomal storage disease: recent patents and future strategies. Rec Pat Endocr Metab Immune Drug Discov. 2014;8(1):9–25.

Figura
Mechanism of action of SRT drugs for GD and other LSDs. The SRT drugs like miglustat and eliglustat are competitive inhibitors ofceramide glucosyltransferase. By reducing glucocerebroside concentration, these drugs facilitate the degradation of the substrate by theb-glucosidase, the enzyme affected in GD. Glucocerebroside is a precursor of gangliosides accumulated in TSD, MPS III, SD, and NiemannPick C diseases. ERT enhances the activity of b-glucosidase. GD indicates Gaucher disease; LSD, lysosomal storage disorder; SRT, substrate reduction therapy; TSD, Tay-Sachs disorder; MPS III, mucopolysaccharidosis type III; SD, Sandhoff disorder; ERT, enzyme replacement therapy.

Alternative therapeutic approaches, based on both in vivo or ex vivo gene therapy, are being tested to meliorate enzyme biodistribution and thus increase the therapeutic window for treatment administration and the possibilities of delivering into the central nervous system (CNS). The LSDs are particularly prone to be cured through gene therapy for the possibility of cross-correction of the enzyme defect,77 Rastal, DP, Amalfitano, A. Recent advances in gene therapy for lysosomal storage disorders. Appl Clin Genet. 2015;8:157–169. and positive results have already been reported for metachromatic leukodystrophy.88 Biffi, A, Montini, E, Lorioli, L. Lentiviral hematopoietic stem cell gene therapy benefits metachromatic leukodystrophy. Science. 2013;341(6148):1233158. Nevertheless, gene therapy still needs to be optimized for clinical applications, especially in terms of immunogenicity and control of viral genome integration sites.

A second alternative to ERT is represented by the use of orally administrated, no antigenic small molecules that can diffuse across multiple cell membranes and distribute in steady-state concentration through different tissues including the CNS. In this article, we will describe the characteristic of the different class of small molecules in use or in development for the treatment of LSDs.

Substrate Reduction Therapy

Small molecules can be used in LSDs to reduce accumulated substrate concentration by inhibiting the enzyme that synthesizes these substrates or one of their precursors. In this way, accumulation rate is lowered, facilitating the clearance of the storage by the defective, but yet partially functional, enzyme (Figure 1).99 Weinreb, NJ. Oral small molecule therapy for lysosomal storage diseases. Pediatr Endocr Rev. 2013;11(supp 1):77–90.

Figure 1. Mechanism of action of SRT drugs for GD and other LSDs. The SRT drugs like miglustat and eliglustat are competitive inhibitors of ceramide glucosyltransferase. By reducing glucocerebroside concentration, these drugs facilitate the degradation of the substrate by the β-glucosidase, the enzyme affected in GD. Glucocerebroside is a precursor of gangliosides accumulated in TSD, MPS III, SD, and Niemann-Pick C diseases. ERT enhances the activity of β-glucosidase. GD indicates Gaucher disease; LSD, lysosomal storage disorder; SRT, substrate reduction therapy; TSD, Tay-Sachs disorder; MPS III, mucopolysaccharidosis type III; SD, Sandhoff disorder; ERT, enzyme replacement therapy.

The SRT-based drugs are now available for the treatment of GD type I (Table 1), an LSD in which patients usually present with anemia, thrombocytopenia, hepatosplenomegaly, and skeletal deformations. However, in the most severe manifestations of the disease (GD type III), these symptoms are associated with neurological involvement. The GD is characterized by a deficit of β-glucosidase (also called glucocerebrosidase), which removes glucose moieties from glucosylceramide, causing the accumulation of the substrate, predominately in macrophages.1010 Linari, S, Castaman, G. Clinical manifestation and management of Gaucher disease. Clin Cases Miner Bone Metab. 2015;12(2):157–164.

Table 1
SRT for GD Type I. a a Comparative description of the 2 small molecules used in clinical practice for GD type I treatment, which are based on SRT.

Miglustat (N-butyldeoxynojirimycin), a competitive inhibitor of glucosylceramide synthetase, was the first SRT medicament to be approved with a therapeutic indication for patients with GD who cannot be treated by ERT (ie, for immunological reaction or low compliance).99 Weinreb, NJ. Oral small molecule therapy for lysosomal storage diseases. Pediatr Endocr Rev. 2013;11(supp 1):77–90. currently use this drug, which is an oral agent with good gastrointestinal adsorption and wide tissue distribution, including the CNS. It seems that naive patients may respond later and with less efficiency to miglustat compared to ERT in terms of hematological and visceral values normalization1212 Pastores, GM, Giraldo, P, Cherin, P, Mehta, A. Goal-oriented therapy with miglustat in Gaucher disease. Curr Med Res Opin. 2009;25(1):23–37.; however, patients who achieved therapeutic response through ERT may be switched to miglustat without compromising previous benefits.1313 Elstein, D, Dweck, A, Attias, D. Oral maintenance clinical trial with miglustat for type I Gaucher disease: switch from or combination with intravenous enzyme replacement. Blood. 2007;110(7):2296–2301.,1414 Giraldo, P, Alfonso, P, Atutxa, K. Real world clinical experience with long-term miglustat maintenance therapy in type I Gaucher disease: the ZAGAL project. Haematologica. 2009;94(12):1771–1775.

Miglustat causes nonsevere sides effects such as diarrhea, flatulence, abdominal pain, or weight loss, which are due to intestinal α-disaccharides malabsorption, probably related to the inhibition of α-glucosidase isomers.1515 Amiri, M, Naim, HY. Miglustat-induced intestinal carbohydrate malabsorption is due to the inhibition of α-glucosidases, but not β-galactosidase. J Inherit Metab Dis. 2012;35(6):949–954. Nevertheless, this effects can be minimized with continued treatment, dose escalation, and/or by implementing a low sucrose–maltose diet.1616 Champion, H, Ramaswami, U, Imrie, J. Dietary modifications in patients receiving miglustat. J Inher Metab Dis. 2010;33(suppl 3):S379–S383. Tremor was also reported in 30% of the patients during trials and in postapproval treated patients,1717 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(8):1215–1227.,1818 Hollak, CE, Hughes, D, van Schaik, IN, Schwierin, B, Bembi, B. Miglustat (Zavesca) in type I Gaucher disease: 5-year results of a post-authorization safety surveillance program. Pharmacoepidemiol Drug Saf. 2009;18(9):770–777. since miglustat is a potent agonist of the human glucose sensor SGLT3, which is expressed at the neuromuscular junction.1919 Voss, AA, Diez-Sampedro, A, Hirayama, BA, Loo, DD, Wright, EM. Iminosugars are potent agonists of the human glucose sensor SGLT3. Mol Pharmacol. 2007;71(2):628–634. However, tremor is generally mild and may be reverted with dose adjustment. Paresthesia and burning sensations were also reported, although it is not known whether they are drug dependent. The potential neurotoxicity of miglustat compromises its possible use in patients with GD type III with neurological involvement, who did not show any improvement during the clinical trials that were performed.2020 Schiffmann, R, Fitzgibbon, EJ, Harris, C. Randomized controlled trial of miglustat in Gaucher’s type 3. Ann Neurol. 2008;64(5):514–522.

Since miglustat can retard biosynthesis of glucosylceramide-based compounds, its efficacy was also tested for the treatment of Niemann-Pick disease type C (NPC), gangliosidosis, and MPS III (Sanfilippo syndrome).

The NPC1 and NPC2 are lipid storage disorders caused by mutations of genes involved in endosomal–lysosomal transport of lipids.2121 Vanier, MT. Complex lipid trafficking in Niemann-Pick disease type C. J Inherit Metab Dis. 2015;38(1):187–199. Cells, predominantly in the brain, accumulate unesterified cholesterol and other lipids with structural similarity to glycosphingolipids. Progressive accumulation determines that the disease manifestations may appear either at birth or later in life course, until the sixth decade of life. Onset in infants presents a more severe phenotype and may cause sudden failure, whereas late-onset patients develop neuropsychiatric manifestations and other neurological conditions, such as cerebellar ataxia, dystonia, dysarthria, seizure, cognitive problems, and progressive dementia.2222 Santos-Lozano, A, Vilamandos-Garcia, D, Sanchis-Gomar, F. Niemann-Pick disease treatment: a systematic review of clinical trials. Ann Trasl Med. 2015;3(22):360.

Treatment with miglustat managed to correct abnormal lipid trafficking in lymphocytes of patients with NPC-1 and to slow progression of neurological manifestations in animal models (mouse and cat) of the disease.2323 Lachmann, RH, te Vruchte, D, Lloyd-Evans, E. Treatment with miglustat reverses the lipid-trafficking defect in Niemann-Pick disease type C. Neurobiol Dis. 2004;16(3):654–658.,2424 Stein, VM, Crooks, A, Ding, W. Miglustat improves Purkinje cell survival and alters microglial phenotype in feline Niemann-Pick disease type C. J Neuropathol Exp Neurol. 2012;71(5):434–448. When used in clinical trial, miglustat improved saccadic eye movement and swallowing capacity of the patients, with better evidence in patients with later onset.2525 Patterson, MC, Vecchio, D, Jacklin, E. Long-term miglustat therapy in children with Niemann-Pick disease type C. J Child Neurol. 2010;25(3):300–305.,2626 Wraith, JE, Vecchio, D, Jacklin, E. Miglustat in adult and juvenile patients with Niemann-Pick disease type C: long-term data from a clinical trail. Mol Genet Metab. 2010;99(4):351–357. These results were confirmed in extended clinical trials, through assessments of cognitive functions and disability, and lead to the approval of miglustat for the treatment of NPC-1.2727 Chien, YH, Peng, SF, Yang, CC. Long-term efficacy of miglustat in pediatric patients with Niemann-Pick disease type C. J Inherit Metab Dis. 2013;36(1):129–137.

Preclinical studies also showed positive results for the potential use of miglustat in the treatment of Tay-Sachs disorder (TSD) and Sandhoff disorder (SD). Both SD and TSD are gangliosidoses caused by the deficiency of β-hexosaminidase (β-Hex). β-Hexosaminidase is a dimeric enzyme (α and/or β subunits) that is required for the removal of the terminal N-acetylgalactosamine from several glycosphingolipids, as well as from some glycoproteins and oligosaccharides. Genetic mutations in the β-subunit result in compromised activity of both β-HexA and β-HexB leading to the accumulation of glycosphingolipids, GAGs, acetylgalactosamine, and N-acetylglucosamine, which are responsible for SD. On the other end, TSD results from mutations in the α-subunit, leading to β-HexA deficiency and the accumulation of ganglioside GM-2 as major storage product.2828 Mahuran, DJ. The biochemistry of HEXA and HEXB gene mutations causing GM2 gangliosidosis. Biochim Biophys Acta. 1991;1096(2):87–94. In TSD, GM-2 accumulation is confined to CNS, whereas in SD, there are multisystemic manifestations.

In mouse model of SD, miglustat was able to reverse neuronal pathophysiological and biochemical abnormalities, as well as to delay symptom of onset and to increase life expectancy. Moreover, treatment with the same drug prevented accumulation of GM-2 in the brain of the TSD model.2929 Pelled, D, Lloyd-Evans, E, Riebeling, C, Jeyakumar, M, Platt, FM, Futerman, AH. Inhibition of calcium uptake via the sarco/endoplasmic reticulum Ca2+-ATPase in a mouse model of Sandhoff disease and prevention by treatment with N-butyldeoxnojirimycin. J Biol Chem. 2003;278(32):29496–29501.,3030 Platt, FM, Neises, GR, Reinkensmeir, G. Prevention of lysosomal storage in Tay-Sachs mice treated with N-butyldeoxynojirimycin. Science. 1997;276(5311):428–431.

Unfortunately, these results were not confirmed in a trial performed in 5 teenagers with GM-2 gangliosidosis observed over 2 years, failing to prevent neurological deterioration,3131 Maegawa, GH, Banwell, BL, Blaser, S. Substrate reduction therapy in juvenile GM2 gangliosidosis. Mol Genet Metab. 2009;98(1-2):215–224. nor it did with patients with TSD,3232 Shapiro, BE, Pastores, GM, Gianutsos, J, Luzy, C, Kolodny, EH. Miglustat in late onset Tay-Sachs disease: a 12-month, randomized, controlled clinical study with 24 months of extended treatment. Genet Med. 2009;11(6):425–433. in spite of previously reported positive data of an isolated study performed with patients with SD.3333 Wortmann, SB, Lefeber, DJ, Dekomien, G, Willemsen, MA, Wevers, RA, Morava, E. Substrate deprivation therapy in juvenile Sandhoff disease. J Inherit Metab Dis. 2009;32(suppl 1):S307–S311.

Treatment with miglustat also failed to show any improvement or stabilization in patients with Sanfilippo syndrome, either in terms of clinical condition or of ganglioside concentration in the brain.3434 Guffon, N, Bin-Dorel, S, Decullier, EP, Paillet, C, Guitton, J, Fouilhoux, A. Evaluation of miglustat treatment in patients with type III mucopolysaccharidoses: a randomized double-blind placebo-controlled study. J Pediatr. 2011;159(5):838–844. Genistain, another SRT-based compound, was also reported to improve cognitive and behavioral functions in MPS III,3535 Piotrowska, E, Jakobkiewicz-Banecka, J, Maryniak, A. Two-year follow-up of Sanfilippo disease patients treated with a genistein-rich isoflavone extract: assessment of effects on cognitive functions and general status of patients. Med Sci Monit. 2011;17(4):CR196–CR202. although a more extended trial did not confirm the initial results.3636 de Ruijter, J, Valstar, MJ, Narajczyc, M. Genistain in Sanfilippo disease: a randomized controlled crossover trial. Ann Neurol. 2012;71(1):110–120.

The second SRT-based treatment for GD type I to be approved was eliglustat, a highly specific oral glucosylceramide synthase inhibitor structurally different from miglustat.3737 Scott, LJ. Eliglustat: a review in Gaucher disease Type I. Drugs. 2015;75(14):1669–1678. In a double-blind 9-month phase III multinational clinical trial (ENGAGE, Phase III, randomized double-blind, placebo controlled, multicentre study confirming the efficacy and safety of Genz-112638 in patients with Gaucher disease Type I, NCT00891202), the drug significantly improved hematological parameters, bone marrow burden score, and also reduced organomegaly in naive patients with GD type I.3838 Mistry, P, Amato, DJ, Dasouki, M. ENGAGE—a phase III randomized, double-blind, placebo controlled, multi-center study to investigate the efficacy and safety of eliglustat in adults with Gaucher disease type I: results after 11 months. Mol Genet Metab. 2015;114(2):S81–S82. Moreover, in a second clinical trial, comparing eliglustat to ERT (a study of Eliglustat tartrate (Genz-112638) in patients with Gaucher disease, who have reached therapeutic goals with enzyme replacement therapy, NCT00943111), the end points that were reached proved that this drug is not inferior to imiglucerase after 12 months of treatment.3939 Cox, TM, Drelichman, GI, Cravo, R. ENCORE a randomized controlled, open-label no inferiority study comparing eliglustat to imiglucerase in Gaucher disease type I patients stabilized on enzyme replacement therapy: 24 months results. Mol Genet Metab. 2015;114(2):S33–S34. Eliglustat does not inhibit intestinal glucosidases and had no neurotoxic effect, since it is a substrate for P-glycoprotein transporter and is extruded from the CNS.4040 Shayman, JA. Eliglustat tartrate: glucosyl ceramide synthase inhibitor treatment of type 1 Gaucher disease. Drugs Future. 2010;35(8):613–620. On the other end, other P-glycoprotein substrates such as digoxin, phenytoin, and colchicine can compete with eliglustat, affecting its pharmacokinetics. This active compound is metabolized through the cytochrome P450 complex, therefore, it can potentially interact with other drugs that are degraded through the same pathway. Pharmacokinetics of eliglustat can also be affected by the genetic heterogeneity of the patients with respect to the CYP2D6 enzyme of the cytochrome P450 complex. Prior to initiating eliglustat therapy, patients should be genotyped for CYP2D6 to determine their CYP2D6 metabolizer status and to find the adequate individual dosage. For this reason, the medicament obtained therapeutic indication for patients with GD type I, who are CYP2D6 extensive metabolizers, intermediate metabolizers, or poor metabolizers.4242 Genzyme Ireland Ltd. Cerdelga (eliglustat) capsules for oral use: US prescribing information. 2014. http://www.cerdelga.com/ Accessed August 22, 2016.
http://www.cerdelga.com/...

The SRT is also used for the treatment of infantile nephropathic cystinosis, a rare genetic disease due to dysfunction of cystinosine transporter expressed in lysosomal membrane. Cysteamine converts accumulated cystine into cysteine and cysteine–cysteamine mixed disulfide, both of which can pass through the lysosomal membrane of patients with cystinosis.4343 Tsilou, ET., Rubin, BI., Reed, G. Nephropathic cystinosis: posterior segment manifestations and effects of cysteamine therapy. Ophtalmology. 2006;113(6):1002–1009. Cysteamine bitartrate has been approved by the Food and Drug Administration since 1994, and in 2013, a delayed release formulation, no inferior to the previous one, was also approved.4545 Langman, C, Greenbaum, LA, Sarwal, M. A randomized controlled crossover trial with delayed release cysteamine bitartrate in nephropathic cystinosis: effectiveness on white blood cell cystine levels and comparison of safety. Clin J Am Soc Nephrol. 2012;7(7):1112–1120. In spite of its use in clinical practice, long-term studies indicate that cysteamine treatment even when started early (before 5) is not curative and 30% of the patients require renal transplant before 16 years.4646 Brodin-Sartorius, A, Tete, MJ, Niaudet, P. Cysteamine therapy delays the progression of nephropatic cystinosis in late adolescent and adults. Kidney Int. 2012;81(2):179–189. The same active compound was also tested for the treatment of neuronal ceroid lipofuscinosis caused by PPT1, which is involved in lysosomal degradation of S-fatty acylated proteins.4747 Gavin, M, Wen, GY, Messing, J. Substrate reduction therapy in four patients with milder CLN1 mutations and juvenile-onset Batten disease using cysteamine bitartrate. JIMD Rep. 2013;11:87–92.

Pharmacological Chaperones

In several LSDs, substrate accumulation that determines clinical manifestations occurs when residual enzyme activity decays below a certain threshold. It was determined that an activity >10% significantly prevents storage in many LSDs and even values between 3% and 5% could be sufficient to slow down the disease progression.4848 Hopwood, JJ, Muller, V. Biochemical discrimination of Hurler and Schie syndromes. Clin Sci. 1979;57(3):265–272.,4949 Leinekugel, P, Michel, S, Conzelmann, E, Sandhoff, K. Quantitative correlation between the residual activity of beta-hexosaminidase A and arylsulphatase A and the severity of the resulting lysosomal storage disease. Hum Genet. 1992;88(5):513–523. In terms of treatment, this means that even a low improvement in enzyme activity could be effective to meliorate clinical evidence. A pharmacological chaperone (PC) is a small molecule that stabilizes the tertiary structure of a mutant enzyme, avoiding its degradation by the quality control mechanisms of the endoplasmic reticulum (ER) and allowing its trafficking to the lysosome through the secretory pathway (Figure 2).5050 Parenti, G, Andria, G, Valenzano, KJ. Pharmacological chaperone therapy: preclinical development, clinical translation and prospects for the treatment of lysosomal storage disorders. Mol Ther. 2015;23(7):1138–1148. Once the complex has reached the target organelle, the PC is released from the enzyme upon acid hydrolysis and the protein can, at least partially, catalyze substrate degradation, still in the presence of the mutation. Even though lysosomal enzymes are active at acid pH, their synthesis normally occurs at neutral pH; therefore, these proteins may be thermodynamically less stable during synthesis process, especially in the presence of a mutation. The PCs stabilize protein conformation, inhibit premature misfolding, and prevent aggregation.

Figure 2
Pharmacological chaperones: mechanism of action. A, Mutated lysosomal enzyme that does not fold correctly is degraded at ER level. B, Pharmacological chaperone diffuses through the plasma membrane into the cell and binds to the mutated enzyme, allowing proper folding of the protein. The complex traffics through the secretory pathway and enters the lysosome, where the PC is released and the enzyme can catalyze substrate degradation. ER indicates endoplasmic reticulum; PC, pharmacological chaperone.

Several PCs were designed and synthesized based on the structure of the natural substrate (iminosugars, azasugars, carbasugars, etc) or alternatively identified by high-throughput screening (HTS) studies. These compounds, potentially active for LSDs therapy, showed encouraging results in preclinical studies for the treatment of FD, GD, PD, or gangliosidosis 1 and 2 (Table 2).

Table 2
Clinical Trials of PCs.a a PC has been or is being tested in clinical trials, either in monotherapy or in combination with ERT. The identification number of the clinical trials corresponds to the database clinicaltrial.gov (https://clinicaltrials.gov).

In 2016, the first PC-based therapy (Galafold, Amicus Therapeutics) obtained marketing approval by the European Medicines Agency for the treatment of patients with FD with specific missense mutations. The active compound of this drug is 1-deoxygalactonojirimycin (DGJ), an iminosugar mimicking the structure of d-galactose, which is also denominated migalastat.5757 Fan, JQ, Ishii, S, Asano, N, Suzuki, Y. Accelerated transport and maturation of lysosomal alpha-galactosidase A in Fabry lymphoblasts by an enzyme inhibitor. Nat Med. 1999;5(1):112–115. The FD is an X-linked LSD, caused by the impairment of α-galactosidase A (α-GalA), which leads to the progressive accumulation of glycosphingolipids such as globotriaosylceramide (GL-3) and Lyso-GL3 in vascular endothelium, smooth muscle, kidney, dorsal ganglia, brain, gastrointestinal tract, and so on.5858 El-Abassi, R, Singhal, D, England, JD. Fabry’s disease. J Neurol Sci. 2014;344(1-2):5–19

The chaperoning action of DGJ depends on the interaction of its amino group with the carboxylate of the aspartic acid 170 in the active site of the enzyme, as shown by crystallography.5959 Guce, AI, Clark, NE, Rogich, JJ, Garman, SC. The molecular basis of pharmacological chaperoning in human alpha-galactosidase. Chem Biol. 2011;18(12):1521–1526. Exposure to DGJ (0.2-20 μmol/L) of primary lymphoblast cultures from patients with FD caused a concentration-dependent increase in α-Gal A activity, which was maintained for 5 days.5757 Fan, JQ, Ishii, S, Asano, N, Suzuki, Y. Accelerated transport and maturation of lysosomal alpha-galactosidase A in Fabry lymphoblasts by an enzyme inhibitor. Nat Med. 1999;5(1):112–115. In vivo studies with DGJ on FD transgenic mouse models showed significant increase in α-Gal A activity in heart, kidney, spleen, and liver of the treated animals, as well as decreased levels of GL-3 in the affected tissues.5959 Guce, AI, Clark, NE, Rogich, JJ, Garman, SC. The molecular basis of pharmacological chaperoning in human alpha-galactosidase. Chem Biol. 2011;18(12):1521–1526. A phase 2 clinical trial performed with migalastat hydrochloride as monotherapy showed increased α-Gal A activity and decreased GL-3 concentration in skin, urine, and kidneys. Reduction in renal peritubular capillary inclusions was also found in heterozygous women.6060 Giugliani, R, Waldek, S, Germain, DP. A phase 2 study of migalastat hydrochloride in female with Fabry disease: selection of population, safety and pharmacodynamic effects. Mol Gen Metab. 2013;109(1):86–92. In phase 3 studies, stabilization of renal function, reduction in left ventricular mass, and improvement in gastrointestinal symptoms were demonstrated, as well as a generally good tolerability.6161 Nicholls, K, Germain, DP, Feliciani, C. Phase III study of migalastat HCl for Fabry disease: stage 1 results [abstract 168]. Mol Genet Metab. 2013;108:S70. Long-term effects of the drugs as well as its possible synergistic effect in the cotreatment with ERT are currently being evaluated.

Two potential PCs for GD, isofagamin and ambroxol, were identified through HTS.6262 Chang, HH, Asano, N, Ishii, S, Ichikawa, Y, Fan, JQ. Hydrophilic iminosugar active site-specific chaperones increase residual glucocerebrosidase activity in fibroblasts from Gaucher patients. FEBS J. 2006;273(17):4082–4092.,6363 Maegawa, GH, Tropak, MB, Buttner, JD. Identification and characterization of ambroxol as an enzyme enhancement agent for Gaucher disease. J Biol Chem. 2009;284(35):23502–23516. Isofagamin increased β-glucosidase activity in cells derived from p.N370S- and p.L444P-affected patients.6464 Sun, Y, Liou, B, Xu, YH. Ex vivo and in vivo effects of isofagomine on acid β-glucosidase variants and substrate levels in Gaucher disease. J Biol Chem. 2012;287(6):4275–4287. These results were confirmed in transgenic mice with different mutations including the p.L444P6565 Khanna, R, Benjiamin, ER, pellegrino, L. The pharmacological chaperone isofagomine increases the activity of the Gaucher disease L444P mutant form of beta-glucosidase. FEBS J. 2010;277(7):1618–1638.,6666 Sanders, A, Hemmelgarn, H, Melrose, HL, Hein, L, Fuller, M, Clarke, LA. Transgenic mice expressing human glucocerebrosidase variants, utility for the study of Gaucher disease. Blood Cell Mol Dis. 2013;51(2):109–115. and put the basis for phase 1 and 2 clinical trials, which unfortunately were discontinued, failing to show a clinically meaningful end point in most of the involved patients (NCT00446550 and NTC00875160). Ambroxol was also able to stabilize p.N370S-mutated β-glucosidase in patient’s derived cells and animal models.6767 Bendikov-Bar, I, Maor, G, Filocamo, M, Horowitz, M. Ambroxol as a pharmacological chaperone for mutant glucocerebrosidase. Blood Cell Mol Dis. 2013;50(2):141–155.,6868 Luan, Z, Li, L, Higaki, K, Nanba, E, Suzuki, Y, Ohno, K. The chaperone activity and toxicity of ambroxol on Gaucher cells and normal mice. Brain Dev. 2013;35(4):317–322. In clinical trials, ambroxol showed reduction of 15% to 40% in spleen volume and increased platelet counts after more than 6 months therapy in 1 patient.5353 Zimran, A, Altarescu, G, Elstein, D. Pilot study using ambroxol as a pharmacological chaperone in type I Gaucher disease blood cell. Mol Dis. 2013;50(2):134–137.

The PCs have been proposed to complement ERT also for PD. The PD is an LSD caused due to mutations in GAA gene, which encodes acid α-glucosidase. The enzymatic deficit produces accumulation of glycogen predominantly in heart and skeletal muscles.6969 van der Ploeg, AT, Rauser, AJ. Pompe’s disease. Lancet. 2008;372(9646):1342–1353. Both 1-deoxynojirimycin (DNJ, duvoglustat) and miglustat can increase acid α-glucosidase activity in cells expressing different mutant forms of the enzyme and in animal models, since these iminosugars are 10- to 250-fold more selective for α-glucosidase compared to β-glucosidase.7070 Parenti, G, Zuppaldi, A, Pittis, GM. Pharmacological enhancement of mutated alpha-glucosidase activity in fibroblasts from patients with Fabry disease. Mol Ther. 2007;15(3):508–514.,7171 Tajima, Y, Saito, S, Ohno, K, Tsukimura, T, Tsujino, S, Sakuraba, H. Biochemical and structural study on a S529V mutant acid alpha-glucosidase responsive to pharmacological chaperones. J Hum Genet. 2011;56(6):440–446. In particular, DNJ administration to PD mouse model showed daily important reduction of substrate in heart, diaphragm, gastrocnemius, soleus, and brain.7272 Khanna, R, Flanagan, JJ, Feng, J. The pharmacological chaperone AT2220 increases recombinant human alpha-glucosidase uptake and glycogen reduction in a mouse model of Pompe disease. PLoS One. 2012;7(7):e40776. In spite of the positive tolerability results of phase 1 studies, duvoglustat showed severe adverse events in 2 patients during phase 2 clinical trial.5555 Web site: https://clinicaltrials.gov/Reference:NCT01380743, Accessed August 22, 2016.
https://clinicaltrials.gov/Reference:NCT...
A follow-up study, showing the pharmacokinetic profile of the drug in the muscle, demonstrated that the dose selected for phase 2 study was excessive (>Half Maximal Inhibitory Concentration [IC]5050 Parenti, G, Andria, G, Valenzano, KJ. Pharmacological chaperone therapy: preclinical development, clinical translation and prospects for the treatment of lysosomal storage disorders. Mol Ther. 2015;23(7):1138–1148. for inhibition of the enzyme), and therefore, appropriate balance between chaperoning and inhibition was not achieved, causing the adverse events.5656 Kishnani, P, Tarnopolski, M, Sivakumar, K. A phase 2a study to investigate drug–drug interactions between escalating doses of AT2220 (duvoglustat hydrochloride) and acid alpha-glucosidase in subjects with Pompe disease. Mol Genet Metab. 2013;108(2):S54.

On the other end, miglustat was tested as a combination therapy for PD to enhance the effects of ERT in patients with infantile and late onsets in a phase 2 trial. In the whole population (13 patients), acid α-glucosidase activity was significantly increased with the cotreatment.7373 Parenti, G, Fecarotta, S, La Marca, G. A chaperone enhance blood alpha-glucosidase activity in Pompe disease patients treated with enzyme replacement therapy. Mol Ther. 2014;22(11):2004–2012.

Since PCs can diffuse to the CNS, they are a potential good treatment for LSDs with important neurological manifestation such as SD and TSD, since ERT drugs do not cross the BBB. Among the tested compounds for SD and TSD, pyrimethamine, an approved antimalarial agent, significantly increased enzymatic activity in all the β-subunit mutants evaluated; however, it determined an insufficient increase in enzymatic activity of the α-subunit mutants.5454 Tropak, MB, Blanchard, JE, Withers, SG, Brown, ED, Mahuran, D. High-throughput screening for human lysosomal beta-.-acetyl hexosaminidase inhibitors acting as pharmacological chaperones. Chem Biol. 2007;14(2):153–164. In spite of this limitation, a clinical trial phase 1/2 was initiated in patients with SD or TSD. The 8 treated patients showed significant enzymatic activity increase; however, severe side effects, such as ataxia or blurred vision, among others, appeared during the trial, causing the suspension of the study.7474 Clarke, JT, Mahuran, DJ, Sathe, S. An open-label Phase I/II clinical trial of pyrimethamine for the treatment of patients affected with chronic GM2 gangliosidosis (Tay-Sachs or Sandhoff variants). Mol Genet Metab. 2011;102(1):6–12. Likewise, in the case of duvoglustat trial in patients with PD, it is possible that the dose of pyrimethamine selected for the study was too elevated for appropriate balance between chaperoning and inhibitory activity of the molecule. The majority of PCs that have been proposed binds to the active center of the target enzyme, therefore, they may act as inhibitors if the affinity is too high. This represents a major limitation for PC therapies for which the concentration has to be finely tuned to determine a net gain in chaperoning activity. Turnover is favored by high substrate concentration and by binders that present shorter half-life than the enzyme, as well as reduced binding affinity at lysosomal pH.

The increase in activity, however, depends on the disease and on the affected enzyme maturation process and its lysosomal trafficking. Moreover, the efficacy of PC-based drugs is mutation dependent, and a single compound may have different effects on patients with the same LSD, but different mutations. The PCs are more prone to stabilize proteins with missense mutations (either inside or outside the active site), which affect protein folding, thermodynamic stability, or lysosomal trafficking, while they are not suitable for large deletions, insertions, splice variants, or frameshift mutations.

Stop-Codon Read-Through Enhancement

Many patients with LSD present nonsense mutations in one or both alleles of the gene encoding for the lysosomal enzyme, which generate a premature stop codon. As an example, mucopolysaccharidosis type I (Hurler and Scheie syndromes), an LSD caused by the deficit of α-l-iduronidase, is often due to the frequent mutations p.Q70X and p.W402X in the IDUA gene.7575 Wraith, JE, Jones, S. Mucopolysaccharidosis type I. Pediatr Endocrinol Rev. 2014;12(suppl 1):102–106. In this disease, as well as in neuronal ceroid lipofuscinosis type I, over 50% of the patients carry a nonsense mutation.7676 Hein, LK, Bawden, M, Muller, VJ, Sillence, D, Hopwood, JJ, Brooks, DA. alpha-l-iduronidase premature stop codons and potential read-through in mucopolysaccharidosis type I patients. J Mol Biol. 2004;338(3):453–462.,7777 Brooks, DA, Muller, VJ, Hopwood, JJ. Stop-codon read-through for patients affected by a lysosomal storage disorder. Trends Mol Med. 2006;12(8):367–373. Premature stop-codon mutations are usually associated with more severe clinical phenotypes, since truncated messenger RNA (mRNA) or protein is generally eliminated with resulting null or minimal net enzyme activity in the cells. Most mRNA transcripts containing stop-codon mutations are preferentially targeted for degradation by a cellular quality control system called nonsense-mediated decay, which limits the synthesis of potentially toxic or unwanted protein fragments that might interfere with normal cellular function.7878 Maquat, LE . Nonsense mediated mRNA decay, in mammals. J Cell Sci. 2005;118(pt 9):1773–1776. Normally, during the splicing process, an exon–exon junction complex is recruited to mRNA and binds 20 to 24 nucleotides upstream of the exon–exon junction. A premature stop codon, upstream of this junction can elicit nonsense-mediated decay.7979 Dreyfuss, G., Kim, VN., Kataoka, N. Messenger-RNA-binding protein and the message they carry. Nat Rev Mol Cell Biol. 2002;3(3):195–205. If a protein escapes this degradation mechanism, a truncated polypeptide is produced and often degraded by the control mechanisms of the ER. However, the read-through process may originate normal or mutated full-length proteins by incorporating an amino acid instead of terminating the synthesis at the premature stop codon. These products can be enough stable to bypass the ER degradation system and perform its function within the cell (Figure 3).

Figure 3
Stop-codon read-through process. The association of 3 proteins at the messenger ribonucleoprotein (mRNP) complex leads to a circular messenger RNA (mRNA) structure. The cap-binding protein eukaryotic initiation factor 4E (eIF4E) binds the 5′ extremity of mRNA; the poly(A)-binding protein (PABP) binds the 3′ end of mRNA; and the eIF4G protein binds both eIF4E and PABP. In mRNAs with wild-type stop codons, PABP is close to the termination complex and can interact with termination factor eukaryotic release factor 3 (eRF3) to stimulate polypeptide chain release. In the presence of a premature stop codon, PABP is at a larger distance and PABP interaction with eRF3 is more difficult, which increases the possibility of activate aminoacyl-transfer RNA to reach the complex and follow the translation (read-through process). Termination factor eRF1 directly recognize any of the 3 stop codons.

The read-through process can also be induced by drugs, such as gentamicin or other aminoglycosides, which bind to ribosomal RNA and affect the stop-codon transfer RNA recognition process.8080 Francois, B, Russell, RJ, Murray, JB. Crystal structures of complexes between aminoglycosides and decoding A site oligonucleotides: role of the number of rings and positive charges in the specific binding leading to miscoding. Nucleic Acids Res. 2005;33(17):5677–5690.,8181 Howard, MT, Shirts, Bh, Petros, LM, Flanigan, KM, Gesteland, RF, Atkins, JF. Sequence specificity of aminoglycoside-induced stop codon read-through: potential implications for treatment of Duchenne muscular dystrophy. Ann Neurol. 2000;48(2):164–169. The degree of read-through process will depend on the amounts of available RNA, the kind of stop codon of the nonsense mutation (UAA has high fidelity and little read-through potential, the UAG has intermediate fidelity, and the UGA sequence has the lowest fidelity and higher potential), the surrounding sequence (ie, nucleotide plus 4 is determinant), and correlates with the efficacy of nonsense-mediated decay.

The amino acid that is inserted in read-through process is not always the correct one, but the protein that is produced may still be stable. The amino acids that are inserted with the highest probability during read through are the glutamine and the tryptophan, which is an advantage in the case of patients with MPS I carrying p.Q70X or p.W402X mutations.8282 Harrell, L, Melker, U, Atkins, JF. Predominance of six different hexanucleotide recording signals 3’ of read-through stop codons. Nucleic Acid Res. 2002;30(9):2011–2017. Gentamicin gave promising results when tested in vitro for cystic fibrosis or Duchenne muscular dystrophy and also for MPS I, among the LSDs.8181 Howard, MT, Shirts, Bh, Petros, LM, Flanigan, KM, Gesteland, RF, Atkins, JF. Sequence specificity of aminoglycoside-induced stop codon read-through: potential implications for treatment of Duchenne muscular dystrophy. Ann Neurol. 2000;48(2):164–169.,8383 Wilschanski, M., Yahav, Y., Yaacov, Y. Gentamicin-induced correction of CFTR functions in patients with cystic fibrosis and CFTR stop mutations. N Engl J Med. 2003;349(15):1433–1441.,8484 Keeling, KM, Brooks, DA, Hopwood, JJ, Li, P, Thompson, JN, Bedwell, DM. Gentamicin mediated suppression of Hurler syndrome stop mutations restores a low level of α-l-iduronidase activity and reduces lysosomal glycosaminoglycan accumulation. Hum Mol Genet. 2001;10(3):291–299.

The principal limitations for the use of gentamicin as read-through inducer are its low cell permeability and its toxic side effects, including kidney damage and hearing loss. For this reason, different analogues of this compound have been proposed. It has been shown that the C1a-subunit of gentamicin (amikacin) preserves efficacy but has reduced side effects.8585 Du, M, Keeling, KM, Fan, L. Clinical doses of amikacin provide more effective suppression of the human CFTR-G542X stop mutation than gentamicin in a transgenic CF mouse model. J Mol Med. 2006;84(7):573–582. Another stop-codon read-through active compound, which was identified during HTS, is PTC124. This active molecule, which inhibits nonsense-mediated decay and allows read through without affecting normal termination codons, presents high permeability and no toxic side effects.8686 Welch, EM, Barton, ER, Zhuo, J. PTC124 targets genetic disorders caused by nonsense mutations. Nature. 2007;447(7140):87–91.

PTC124 proved to be more efficient than gentamicin in cystic fibrosis and Duchenne muscular dystrophy in preclinical studies. Phase 3 clinical trials were performed in patients with cystic fibrosis and Duchenne muscular dystrophy (NCT00803205 and NCT0182647), leading to the conditional approval of the compound for Duchenne muscular dystrophy.8787 Konstan, M, Accurso, F, De Boeck, K. Targeting class 1 mutations: update on ataluren as a promising treatment for nonsense mutation cystic fibrosis. Pediatr Pulmonol. 2012;47(S35):108–109.,8888 Bushby, K., Finkel, R., Wong, B; PTC124-GD-007-DMD STUDY GROUP. Ataluren treatment of patients with nonsense mutation dystrophinopathy. Muscle Nerve. 2014;50(4):477–487.

PTC124 was also tested in cells derived from patients with MPS VI (Maroteaux-Lamy). The MPS VI is an LSD without CNS involvement, which is caused by arylsulfatase B (ARSB, gene) deficiency. It is characterized by dysostosis multiplex, corneal clouding, heart valve defects, and urinary excretion of GAGs. A subgroup of patients with MPS VI carries nonsense mutations (p.R315X, p.R327X, p.Q456X, and p.Q503X) and therefore is eligible for enhancing of stop-codon read through, using small molecules. PTC124, but not gentamicin, showed to significantly increase the level of arylsulfatase B activity, resulting in a significant reduction in lysosomal size.8989 Bartolomeo, R, Polishchuk, EV, Volpi, N, Polishchuk, RS, Auricchio, A. Pharmacological read-through of nonsense ARSB mutations as a potential therapeutic approach for mucopolysaccharidosis VI. J Inherit Metab Dis. 2013;36(2):363–371.

In a second study, gentamicin, geneticin, PTC124, and 4 no aminoglycosidic compounds (RTC13, RTC14, BZ6, and Bz16) were tested in fibroblast derived from patients with MPS VI and MPS III B or C (Sanfilippo) with premature stop-codon mutations. Cells from 1 patient with Maroteaux-Lamy carrying the p.W322X mutation responded to treatment with gentamicin (2- to 3-fold increase in enzyme activity); however, the effects of the other compounds were not tested.

In Sanfilippo B and C patients’ cells, an increase in mRNA levels of NAGLU and HGSNAT was found after treatment with, respectively, geneticin and RTC14 or PTC124, although no enzyme activity was recovered.9090 Gomez-Grau, M, Garrido, E, Cozar, M. Evaluation of aminoglycoside and non-aminoglycoside compound for stop-codon read-through therapy in four lysosomal storage diseases. PLoS One. 2015;10(8):e0135873.

Conclusions and Future Perspectives

The use of small molecules, comprising SRT, PCs, and read-through enhancer, could represent a great advantage in the treatment of LSDs compared to ERT, since they can be administrated orally, achieve homogenous steady-state concentrations, and possibly lower production costs. Nevertheless, important challenges have still to be faced to advance in the development of these drugs.

Small molecules can potentially diffuse in the CNS; however, up to date, the only commercialized treatment for LSDs with neurological involvement is miglustat, which has a therapeutic indication for NPC, and there is still no available treatment for a considerable number of LSDs. Indeed, the clinical trials performed in GD type III with miglustat or in gangliosidoses with pyrimethamine did not give the expected results due to toxic side effects, which were probably related to unoptimized doses.

Reviewing patents databases66 Ortolano, S, Vieitez, I, Navarro, C, Spuch, C. Treatment of lysosomal storage disease: recent patents and future strategies. Rec Pat Endocr Metab Immune Drug Discov. 2014;8(1):9–25. (https://worldwide.espacenet.com/), many registered molecules claim PC activity for LSDs, nevertheless, at the moment the only PC to be commercialized is migalastat. The delay in the translation to the clinic could be related to the difficulty to select an appropriate dose of administration for PC-based treatment, which should allow maximal substrate turnover without inhibitory effect. An option to reach this goal is to administer the PC in alternative intervals, since the turnover of the small molecule is generally faster than that of the defective enzyme. In hR301Q α-GalA transgenic mice, oral administration of migalastat in cycle of 4 days during a week resulted in greater substrate clearance, compared to daily administration.9090 Gomez-Grau, M, Garrido, E, Cozar, M. Evaluation of aminoglycoside and non-aminoglycoside compound for stop-codon read-through therapy in four lysosomal storage diseases. PLoS One. 2015;10(8):e0135873.

Alternatively, next-generation PC should be directed to the domains of the lysosomal enzyme, which do not include the active site but have a crucial role in the stabilization of the protein (allosteric sites). This will preserve PC scaffolding function without affecting its catalytic action. Potential PCs binding to allosteric sites have already been identified for acid α-glucosidase5252 Porto, C, Ferrara, MC, Meli, M. Pharmacological enhancement of α-glucosidase by allosteric chaperone.-acetylcysteine. Mol Ther. 2012;20(12):2201–2211.,9191 Marugan, JJ, Zheng, W, Ferrer, M. Discovery, SAR and biological evaluation of a non-inhibitory chaperone for acid alpha-glucosidase. Probe reports from the NIH Molecular libraries program. MD, USA: National Center for Biotechnology Information Bethesda. http://www.ncbi.nlm.nih.gov/books/NBK153221.
http://www.ncbi.nlm.nih.gov/books/NBK153...
and glucocerebrosidase.9292 Zheng, W, Padia, J, Urban, DJ. Three classes of glucocerebrosidase inhibitors identified by quantitative high-throughput screening are chaperone leads for GD. Proc Nat Acad Sci U S A. 2007;104(32):13192–13197.,9393 Landon, MR, Lieberman, RL, Hoang, QQW. Detection of ligand binding hot spots on protein surfaces via fragment-based methods: application to DJ-1 and glucocerebrosidase. J Comput Aided Mol Des. 2009;23(8):491–500.

Finally, a therapeutic option that is being explored for the treatment of LSDs is the coadministration of ERT with small molecules. Among the limitations of ERT, low half-life and uneven distribution of the recombinant enzyme are crucial in lowering the efficacy of the treatment. As an example, ERT for PD has to be administered at higher frequency and dose compared to other enzyme, due to the low expression of MP6 receptors in the muscle and the higher adaptive immune system activation in these patients. Nevertheless, it has been shown that wild-type lysosomal enzymes like the ones affected in FD, PD, and GD respond to PCs, increasing the rate of substrate degradation. Interestingly, only some of the wild-type enzymes related to LSDs respond to chaperons, depending on the folding efficiency of the enzyme, which may also be different across cell types and species. As mentioned previously, cotreatment was tested in patients with PD, achieving significant increase in blood α-glucosidase activity by administering miglustat or eliglustat prior to ERT.5555 Web site: https://clinicaltrials.gov/Reference:NCT01380743, Accessed August 22, 2016.
https://clinicaltrials.gov/Reference:NCT...
,5656 Kishnani, P, Tarnopolski, M, Sivakumar, K. A phase 2a study to investigate drug–drug interactions between escalating doses of AT2220 (duvoglustat hydrochloride) and acid alpha-glucosidase in subjects with Pompe disease. Mol Genet Metab. 2013;108(2):S54. Similar results were obtained in patients with FD treated with migalastat before ERT infusion, increasing α-Gal A activity in the blood and the skin of all the patients.9494 Warnock, D, Bichet, D, Holida, M. A phase 2A study to investigate the effect of a single dose of migalastat HCl, a pharmacological chaperone, on agalsidase activity in subjects with Fabry disease [abstract 248]. Mol Genet Metab. 2013;108:S96.,9595 Hugues, D, Adera, M, Castelli, J. Preliminary long-term safety, tolerability and assessments of renal function of adult Fabry patients receiving treatment with AT1001 (migalastat, hydrochloride), a pharmacological chaperone, for up to 3 years [abstract 473-O]. J Inher Metab Dis. 2010;33(suppl 1):S148.

For their action mechanism, SRT and read-through enhancer can also potentially contribute to higher storage metabolization, when used in combination with ERT. Therefore, cotreatment could be the key to achieve clinical improvement in patients with LSDs.

Due to the heterogeneity of LSDs and the variability of the expressed phenotypes, the ideal treatment for all affected patients has not been developed up to date. Nevertheless, thanks to the recent advances exposed in this review, crucial problems and possible solutions have been identified and will allow to design new tools and specific management protocols based on the association of different therapeutic agents to face individual diversity.

Funding

  • The author(s) received no financial support for the research, authorship, and/or publication of this article.

References

  • 1
    Kingma, SD, Bodamer, OA, Wijburg, FA. Epidemiology and diagnosis of lysosomal storage disorders; challenges of screening. Best Pract Res Clin Endocrinol Metab. 2015;29(2):145–157.
  • 2
    Fuller, M, Meikle, PJ, Hopwood, JJ. Epidemiology of lysosomal storage diseases: an overview. In: Metha, A, Beck, M, Sunder-Plassmann, G, eds. Fabry Diseases: Perspectives From 5 Years of FOS. Oxford, UK: Oxford Pharmagenesis; 2006.
  • 3
    Beck, M. New therapeutic options for lysosomal storage disorders: enzyme replacement, small molecule and gene therapy. Hum Genet. 2007;121(1):1–22.
  • 4
    Charrow, J, Scott, CR. Long-term treatment outcomes in Gaucher disease. Am J Hematol. 2015;90(suppl 1):S19–S24.
  • 5
    Arends, M., Hollak, CE, Biegstraaten, M. Quality of life in patients with Fabry disease: a systematic review of the literature. Orphanet J Rare Dis. 2015;10:77.
  • 6
    Ortolano, S, Vieitez, I, Navarro, C, Spuch, C. Treatment of lysosomal storage disease: recent patents and future strategies. Rec Pat Endocr Metab Immune Drug Discov. 2014;8(1):9–25.
  • 7
    Rastal, DP, Amalfitano, A. Recent advances in gene therapy for lysosomal storage disorders. Appl Clin Genet. 2015;8:157–169.
  • 8
    Biffi, A, Montini, E, Lorioli, L. Lentiviral hematopoietic stem cell gene therapy benefits metachromatic leukodystrophy. Science. 2013;341(6148):1233158.
  • 9
    Weinreb, NJ. Oral small molecule therapy for lysosomal storage diseases. Pediatr Endocr Rev. 2013;11(supp 1):77–90.
  • 10
    Linari, S, Castaman, G. Clinical manifestation and management of Gaucher disease. Clin Cases Miner Bone Metab. 2015;12(2):157–164.
  • 11
    McCormack, PL, Goa, KL. Miglustat. Drugs. 2003;63(22):2427–2434.
  • 12
    Pastores, GM, Giraldo, P, Cherin, P, Mehta, A. Goal-oriented therapy with miglustat in Gaucher disease. Curr Med Res Opin. 2009;25(1):23–37.
  • 13
    Elstein, D, Dweck, A, Attias, D. Oral maintenance clinical trial with miglustat for type I Gaucher disease: switch from or combination with intravenous enzyme replacement. Blood. 2007;110(7):2296–2301.
  • 14
    Giraldo, P, Alfonso, P, Atutxa, K. Real world clinical experience with long-term miglustat maintenance therapy in type I Gaucher disease: the ZAGAL project. Haematologica. 2009;94(12):1771–1775.
  • 15
    Amiri, M, Naim, HY. Miglustat-induced intestinal carbohydrate malabsorption is due to the inhibition of α-glucosidases, but not β-galactosidase. J Inherit Metab Dis. 2012;35(6):949–954.
  • 16
    Champion, H, Ramaswami, U, Imrie, J. Dietary modifications in patients receiving miglustat. J Inher Metab Dis. 2010;33(suppl 3):S379–S383.
  • 17
    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(8):1215–1227.
  • 18
    Hollak, CE, Hughes, D, van Schaik, IN, Schwierin, B, Bembi, B. Miglustat (Zavesca) in type I Gaucher disease: 5-year results of a post-authorization safety surveillance program. Pharmacoepidemiol Drug Saf. 2009;18(9):770–777.
  • 19
    Voss, AA, Diez-Sampedro, A, Hirayama, BA, Loo, DD, Wright, EM. Iminosugars are potent agonists of the human glucose sensor SGLT3. Mol Pharmacol. 2007;71(2):628–634.
  • 20
    Schiffmann, R, Fitzgibbon, EJ, Harris, C. Randomized controlled trial of miglustat in Gaucher’s type 3. Ann Neurol. 2008;64(5):514–522.
  • 21
    Vanier, MT. Complex lipid trafficking in Niemann-Pick disease type C. J Inherit Metab Dis. 2015;38(1):187–199.
  • 22
    Santos-Lozano, A, Vilamandos-Garcia, D, Sanchis-Gomar, F. Niemann-Pick disease treatment: a systematic review of clinical trials. Ann Trasl Med. 2015;3(22):360.
  • 23
    Lachmann, RH, te Vruchte, D, Lloyd-Evans, E. Treatment with miglustat reverses the lipid-trafficking defect in Niemann-Pick disease type C. Neurobiol Dis. 2004;16(3):654–658.
  • 24
    Stein, VM, Crooks, A, Ding, W. Miglustat improves Purkinje cell survival and alters microglial phenotype in feline Niemann-Pick disease type C. J Neuropathol Exp Neurol. 2012;71(5):434–448.
  • 25
    Patterson, MC, Vecchio, D, Jacklin, E. Long-term miglustat therapy in children with Niemann-Pick disease type C. J Child Neurol. 2010;25(3):300–305.
  • 26
    Wraith, JE, Vecchio, D, Jacklin, E. Miglustat in adult and juvenile patients with Niemann-Pick disease type C: long-term data from a clinical trail. Mol Genet Metab. 2010;99(4):351–357.
  • 27
    Chien, YH, Peng, SF, Yang, CC. Long-term efficacy of miglustat in pediatric patients with Niemann-Pick disease type C. J Inherit Metab Dis. 2013;36(1):129–137.
  • 28
    Mahuran, DJ. The biochemistry of HEXA and HEXB gene mutations causing GM2 gangliosidosis. Biochim Biophys Acta. 1991;1096(2):87–94.
  • 29
    Pelled, D, Lloyd-Evans, E, Riebeling, C, Jeyakumar, M, Platt, FM, Futerman, AH. Inhibition of calcium uptake via the sarco/endoplasmic reticulum Ca2+-ATPase in a mouse model of Sandhoff disease and prevention by treatment with N-butyldeoxnojirimycin. J Biol Chem. 2003;278(32):29496–29501.
  • 30
    Platt, FM, Neises, GR, Reinkensmeir, G. Prevention of lysosomal storage in Tay-Sachs mice treated with N-butyldeoxynojirimycin. Science. 1997;276(5311):428–431.
  • 31
    Maegawa, GH, Banwell, BL, Blaser, S. Substrate reduction therapy in juvenile GM2 gangliosidosis. Mol Genet Metab. 2009;98(1-2):215–224.
  • 32
    Shapiro, BE, Pastores, GM, Gianutsos, J, Luzy, C, Kolodny, EH. Miglustat in late onset Tay-Sachs disease: a 12-month, randomized, controlled clinical study with 24 months of extended treatment. Genet Med. 2009;11(6):425–433.
  • 33
    Wortmann, SB, Lefeber, DJ, Dekomien, G, Willemsen, MA, Wevers, RA, Morava, E. Substrate deprivation therapy in juvenile Sandhoff disease. J Inherit Metab Dis. 2009;32(suppl 1):S307–S311.
  • 34
    Guffon, N, Bin-Dorel, S, Decullier, EP, Paillet, C, Guitton, J, Fouilhoux, A. Evaluation of miglustat treatment in patients with type III mucopolysaccharidoses: a randomized double-blind placebo-controlled study. J Pediatr. 2011;159(5):838–844.
  • 35
    Piotrowska, E, Jakobkiewicz-Banecka, J, Maryniak, A. Two-year follow-up of Sanfilippo disease patients treated with a genistein-rich isoflavone extract: assessment of effects on cognitive functions and general status of patients. Med Sci Monit. 2011;17(4):CR196–CR202.
  • 36
    de Ruijter, J, Valstar, MJ, Narajczyc, M. Genistain in Sanfilippo disease: a randomized controlled crossover trial. Ann Neurol. 2012;71(1):110–120.
  • 37
    Scott, LJ. Eliglustat: a review in Gaucher disease Type I. Drugs. 2015;75(14):1669–1678.
  • 38
    Mistry, P, Amato, DJ, Dasouki, M. ENGAGE—a phase III randomized, double-blind, placebo controlled, multi-center study to investigate the efficacy and safety of eliglustat in adults with Gaucher disease type I: results after 11 months. Mol Genet Metab. 2015;114(2):S81–S82.
  • 39
    Cox, TM, Drelichman, GI, Cravo, R. ENCORE a randomized controlled, open-label no inferiority study comparing eliglustat to imiglucerase in Gaucher disease type I patients stabilized on enzyme replacement therapy: 24 months results. Mol Genet Metab. 2015;114(2):S33–S34.
  • 40
    Shayman, JA. Eliglustat tartrate: glucosyl ceramide synthase inhibitor treatment of type 1 Gaucher disease. Drugs Future. 2010;35(8):613–620.
  • 41
    European Medicine Agency. Cerdelga: summary of product characteristics. 2015. http://www.ema.europa.eu Accessed August 22, 2016.
    » http://www.ema.europa.eu
  • 42
    Genzyme Ireland Ltd. Cerdelga (eliglustat) capsules for oral use: US prescribing information. 2014. http://www.cerdelga.com/ Accessed August 22, 2016.
    » http://www.cerdelga.com/
  • 43
    Tsilou, ET., Rubin, BI., Reed, G. Nephropathic cystinosis: posterior segment manifestations and effects of cysteamine therapy. Ophtalmology. 2006;113(6):1002–1009.
  • 44
    Cerqui, S. Cysteamine therapy: a treatment for cystinosis, not a cure. Kidney Int. 2012;81(2):127–129.
  • 45
    Langman, C, Greenbaum, LA, Sarwal, M. A randomized controlled crossover trial with delayed release cysteamine bitartrate in nephropathic cystinosis: effectiveness on white blood cell cystine levels and comparison of safety. Clin J Am Soc Nephrol. 2012;7(7):1112–1120.
  • 46
    Brodin-Sartorius, A, Tete, MJ, Niaudet, P. Cysteamine therapy delays the progression of nephropatic cystinosis in late adolescent and adults. Kidney Int. 2012;81(2):179–189.
  • 47
    Gavin, M, Wen, GY, Messing, J. Substrate reduction therapy in four patients with milder CLN1 mutations and juvenile-onset Batten disease using cysteamine bitartrate. JIMD Rep. 2013;11:87–92.
  • 48
    Hopwood, JJ, Muller, V. Biochemical discrimination of Hurler and Schie syndromes. Clin Sci. 1979;57(3):265–272.
  • 49
    Leinekugel, P, Michel, S, Conzelmann, E, Sandhoff, K. Quantitative correlation between the residual activity of beta-hexosaminidase A and arylsulphatase A and the severity of the resulting lysosomal storage disease. Hum Genet. 1992;88(5):513–523.
  • 50
    Parenti, G, Andria, G, Valenzano, KJ. Pharmacological chaperone therapy: preclinical development, clinical translation and prospects for the treatment of lysosomal storage disorders. Mol Ther. 2015;23(7):1138–1148.
  • 51
    Khanna, R, Soska, R, Lun, Y. The pharmacological chaperone 1-deoxygalactonojirimycin reduces tissue globotriaosylceramide levels in a mouse model of Fabry disease. Mol Ther. 2010;18(1):23–33.
  • 52
    Porto, C, Ferrara, MC, Meli, M. Pharmacological enhancement of α-glucosidase by allosteric chaperone.-acetylcysteine. Mol Ther. 2012;20(12):2201–2211.
  • 53
    Zimran, A, Altarescu, G, Elstein, D. Pilot study using ambroxol as a pharmacological chaperone in type I Gaucher disease blood cell. Mol Dis. 2013;50(2):134–137.
  • 54
    Tropak, MB, Blanchard, JE, Withers, SG, Brown, ED, Mahuran, D. High-throughput screening for human lysosomal beta-.-acetyl hexosaminidase inhibitors acting as pharmacological chaperones. Chem Biol. 2007;14(2):153–164.
  • 55
    Web site: https://clinicaltrials.gov/Reference:NCT01380743, Accessed August 22, 2016.
    » https://clinicaltrials.gov/Reference:NCT01380743
  • 56
    Kishnani, P, Tarnopolski, M, Sivakumar, K. A phase 2a study to investigate drug–drug interactions between escalating doses of AT2220 (duvoglustat hydrochloride) and acid alpha-glucosidase in subjects with Pompe disease. Mol Genet Metab. 2013;108(2):S54.
  • 57
    Fan, JQ, Ishii, S, Asano, N, Suzuki, Y. Accelerated transport and maturation of lysosomal alpha-galactosidase A in Fabry lymphoblasts by an enzyme inhibitor. Nat Med. 1999;5(1):112–115.
  • 58
    El-Abassi, R, Singhal, D, England, JD. Fabry’s disease. J Neurol Sci. 2014;344(1-2):5–19
  • 59
    Guce, AI, Clark, NE, Rogich, JJ, Garman, SC. The molecular basis of pharmacological chaperoning in human alpha-galactosidase. Chem Biol. 2011;18(12):1521–1526.
  • 60
    Giugliani, R, Waldek, S, Germain, DP. A phase 2 study of migalastat hydrochloride in female with Fabry disease: selection of population, safety and pharmacodynamic effects. Mol Gen Metab. 2013;109(1):86–92.
  • 61
    Nicholls, K, Germain, DP, Feliciani, C. Phase III study of migalastat HCl for Fabry disease: stage 1 results [abstract 168]. Mol Genet Metab. 2013;108:S70.
  • 62
    Chang, HH, Asano, N, Ishii, S, Ichikawa, Y, Fan, JQ. Hydrophilic iminosugar active site-specific chaperones increase residual glucocerebrosidase activity in fibroblasts from Gaucher patients. FEBS J. 2006;273(17):4082–4092.
  • 63
    Maegawa, GH, Tropak, MB, Buttner, JD. Identification and characterization of ambroxol as an enzyme enhancement agent for Gaucher disease. J Biol Chem. 2009;284(35):23502–23516.
  • 64
    Sun, Y, Liou, B, Xu, YH. Ex vivo and in vivo effects of isofagomine on acid β-glucosidase variants and substrate levels in Gaucher disease. J Biol Chem. 2012;287(6):4275–4287.
  • 65
    Khanna, R, Benjiamin, ER, pellegrino, L. The pharmacological chaperone isofagomine increases the activity of the Gaucher disease L444P mutant form of beta-glucosidase. FEBS J. 2010;277(7):1618–1638.
  • 66
    Sanders, A, Hemmelgarn, H, Melrose, HL, Hein, L, Fuller, M, Clarke, LA. Transgenic mice expressing human glucocerebrosidase variants, utility for the study of Gaucher disease. Blood Cell Mol Dis. 2013;51(2):109–115.
  • 67
    Bendikov-Bar, I, Maor, G, Filocamo, M, Horowitz, M. Ambroxol as a pharmacological chaperone for mutant glucocerebrosidase. Blood Cell Mol Dis. 2013;50(2):141–155.
  • 68
    Luan, Z, Li, L, Higaki, K, Nanba, E, Suzuki, Y, Ohno, K. The chaperone activity and toxicity of ambroxol on Gaucher cells and normal mice. Brain Dev. 2013;35(4):317–322.
  • 69
    van der Ploeg, AT, Rauser, AJ. Pompe’s disease. Lancet. 2008;372(9646):1342–1353.
  • 70
    Parenti, G, Zuppaldi, A, Pittis, GM. Pharmacological enhancement of mutated alpha-glucosidase activity in fibroblasts from patients with Fabry disease. Mol Ther. 2007;15(3):508–514.
  • 71
    Tajima, Y, Saito, S, Ohno, K, Tsukimura, T, Tsujino, S, Sakuraba, H. Biochemical and structural study on a S529V mutant acid alpha-glucosidase responsive to pharmacological chaperones. J Hum Genet. 2011;56(6):440–446.
  • 72
    Khanna, R, Flanagan, JJ, Feng, J. The pharmacological chaperone AT2220 increases recombinant human alpha-glucosidase uptake and glycogen reduction in a mouse model of Pompe disease. PLoS One. 2012;7(7):e40776.
  • 73
    Parenti, G, Fecarotta, S, La Marca, G. A chaperone enhance blood alpha-glucosidase activity in Pompe disease patients treated with enzyme replacement therapy. Mol Ther. 2014;22(11):2004–2012.
  • 74
    Clarke, JT, Mahuran, DJ, Sathe, S. An open-label Phase I/II clinical trial of pyrimethamine for the treatment of patients affected with chronic GM2 gangliosidosis (Tay-Sachs or Sandhoff variants). Mol Genet Metab. 2011;102(1):6–12.
  • 75
    Wraith, JE, Jones, S. Mucopolysaccharidosis type I. Pediatr Endocrinol Rev. 2014;12(suppl 1):102–106.
  • 76
    Hein, LK, Bawden, M, Muller, VJ, Sillence, D, Hopwood, JJ, Brooks, DA. alpha-l-iduronidase premature stop codons and potential read-through in mucopolysaccharidosis type I patients. J Mol Biol. 2004;338(3):453–462.
  • 77
    Brooks, DA, Muller, VJ, Hopwood, JJ. Stop-codon read-through for patients affected by a lysosomal storage disorder. Trends Mol Med. 2006;12(8):367–373.
  • 78
    Maquat, LE . Nonsense mediated mRNA decay, in mammals. J Cell Sci. 2005;118(pt 9):1773–1776.
  • 79
    Dreyfuss, G., Kim, VN., Kataoka, N. Messenger-RNA-binding protein and the message they carry. Nat Rev Mol Cell Biol. 2002;3(3):195–205.
  • 80
    Francois, B, Russell, RJ, Murray, JB. Crystal structures of complexes between aminoglycosides and decoding A site oligonucleotides: role of the number of rings and positive charges in the specific binding leading to miscoding. Nucleic Acids Res. 2005;33(17):5677–5690.
  • 81
    Howard, MT, Shirts, Bh, Petros, LM, Flanigan, KM, Gesteland, RF, Atkins, JF. Sequence specificity of aminoglycoside-induced stop codon read-through: potential implications for treatment of Duchenne muscular dystrophy. Ann Neurol. 2000;48(2):164–169.
  • 82
    Harrell, L, Melker, U, Atkins, JF. Predominance of six different hexanucleotide recording signals 3’ of read-through stop codons. Nucleic Acid Res. 2002;30(9):2011–2017.
  • 83
    Wilschanski, M., Yahav, Y., Yaacov, Y. Gentamicin-induced correction of CFTR functions in patients with cystic fibrosis and CFTR stop mutations. N Engl J Med. 2003;349(15):1433–1441.
  • 84
    Keeling, KM, Brooks, DA, Hopwood, JJ, Li, P, Thompson, JN, Bedwell, DM. Gentamicin mediated suppression of Hurler syndrome stop mutations restores a low level of α-l-iduronidase activity and reduces lysosomal glycosaminoglycan accumulation. Hum Mol Genet. 2001;10(3):291–299.
  • 85
    Du, M, Keeling, KM, Fan, L. Clinical doses of amikacin provide more effective suppression of the human CFTR-G542X stop mutation than gentamicin in a transgenic CF mouse model. J Mol Med. 2006;84(7):573–582.
  • 86
    Welch, EM, Barton, ER, Zhuo, J. PTC124 targets genetic disorders caused by nonsense mutations. Nature. 2007;447(7140):87–91.
  • 87
    Konstan, M, Accurso, F, De Boeck, K. Targeting class 1 mutations: update on ataluren as a promising treatment for nonsense mutation cystic fibrosis. Pediatr Pulmonol. 2012;47(S35):108–109.
  • 88
    Bushby, K., Finkel, R., Wong, B; PTC124-GD-007-DMD STUDY GROUP. Ataluren treatment of patients with nonsense mutation dystrophinopathy. Muscle Nerve. 2014;50(4):477–487.
  • 89
    Bartolomeo, R, Polishchuk, EV, Volpi, N, Polishchuk, RS, Auricchio, A. Pharmacological read-through of nonsense ARSB mutations as a potential therapeutic approach for mucopolysaccharidosis VI. J Inherit Metab Dis. 2013;36(2):363–371.
  • 90
    Gomez-Grau, M, Garrido, E, Cozar, M. Evaluation of aminoglycoside and non-aminoglycoside compound for stop-codon read-through therapy in four lysosomal storage diseases. PLoS One. 2015;10(8):e0135873.
  • 91
    Marugan, JJ, Zheng, W, Ferrer, M. Discovery, SAR and biological evaluation of a non-inhibitory chaperone for acid alpha-glucosidase. Probe reports from the NIH Molecular libraries program. MD, USA: National Center for Biotechnology Information Bethesda. http://www.ncbi.nlm.nih.gov/books/NBK153221
    » http://www.ncbi.nlm.nih.gov/books/NBK153221
  • 92
    Zheng, W, Padia, J, Urban, DJ. Three classes of glucocerebrosidase inhibitors identified by quantitative high-throughput screening are chaperone leads for GD. Proc Nat Acad Sci U S A. 2007;104(32):13192–13197.
  • 93
    Landon, MR, Lieberman, RL, Hoang, QQW. Detection of ligand binding hot spots on protein surfaces via fragment-based methods: application to DJ-1 and glucocerebrosidase. J Comput Aided Mol Des. 2009;23(8):491–500.
  • 94
    Warnock, D, Bichet, D, Holida, M. A phase 2A study to investigate the effect of a single dose of migalastat HCl, a pharmacological chaperone, on agalsidase activity in subjects with Fabry disease [abstract 248]. Mol Genet Metab. 2013;108:S96.
  • 95
    Hugues, D, Adera, M, Castelli, J. Preliminary long-term safety, tolerability and assessments of renal function of adult Fabry patients receiving treatment with AT1001 (migalastat, hydrochloride), a pharmacological chaperone, for up to 3 years [abstract 473-O]. J Inher Metab Dis. 2010;33(suppl 1):S148.

Publication Dates

  • Publication in this collection
    30 May 2019
  • Date of issue
    2016

History

  • Received
    23 June 2016
  • Reviewed
    06 July 2016
  • Accepted
    06 July 2016
Latin American Society Inborn Errors and Neonatal Screening (SLEIMPN); Instituto Genética para Todos (IGPT) Rua Ramiro Barcelos, 2350, CEP: 90035-903, Porto Alegre, RS - Brasil, Tel.: 55-51-3359-6338, Fax: 55-51-3359-8010 - Porto Alegre - RS - Brazil
E-mail: rgiugliani@hcpa.edu.br