Polytherapy with a combination of three repurposed drugs (PXT3003) down-regulates Pmp22 over-expression and improves myelination, axonal and functional parameters in models of CMT1A neuropathy

Ilya Chumakov, Aude Milet, Nathalie Cholet, Gwenaël Primas, Aurélie Boucard, Yannick Pereira, Esther Graudens, Jonas Mandel, Julien Laffaire, Julie Foucquier, Fabrice Glibert, Viviane Bertrand, Klaus-Armin Nave, Michael W Sereda, Emmanuel Vial, Mickaël Guedj, Rodolphe Hajj, Serguei Nabirotchkin, Daniel Cohen, Ilya Chumakov, Aude Milet, Nathalie Cholet, Gwenaël Primas, Aurélie Boucard, Yannick Pereira, Esther Graudens, Jonas Mandel, Julien Laffaire, Julie Foucquier, Fabrice Glibert, Viviane Bertrand, Klaus-Armin Nave, Michael W Sereda, Emmanuel Vial, Mickaël Guedj, Rodolphe Hajj, Serguei Nabirotchkin, Daniel Cohen

Abstract

Charcot-Marie-Tooth disease type 1A (CMT1A) is the most common inherited sensory and motor peripheral neuropathy. It is caused by PMP22 overexpression which leads to defects of peripheral myelination, loss of long axons, and progressive impairment then disability. There is no treatment available despite observations that monotherapeutic interventions slow progression in rodent models. We thus hypothesized that a polytherapeutic approach using several drugs, previously approved for other diseases, could be beneficial by simultaneously targeting PMP22 and pathways important for myelination and axonal integrity. A combination of drugs for CMT1A polytherapy was chosen from a group of authorised drugs for unrelated diseases using a systems biology approach, followed by pharmacological safety considerations. Testing and proof of synergism of these drugs were performed in a co-culture model of DRG neurons and Schwann cells derived from a Pmp22 transgenic rat model of CMT1A. Their ability to lower Pmp22 mRNA in Schwann cells relative to house-keeping genes or to a second myelin transcript (Mpz) was assessed in a clonal cell line expressing these genes. Finally in vivo efficacy of the combination was tested in two models: CMT1A transgenic rats, and mice that recover from a nerve crush injury, a model to assess neuroprotection and regeneration. Combination of (RS)-baclofen, naltrexone hydrochloride and D-sorbitol, termed PXT3003, improved myelination in the Pmp22 transgenic co-culture cellular model, and moderately down-regulated Pmp22 mRNA expression in Schwannoma cells. In both in vitro systems, the combination of drugs was revealed to possess synergistic effects, which provided the rationale for in vivo clinical testing of rodent models. In Pmp22 transgenic CMT1A rats, PXT3003 down-regulated the Pmp22 to Mpz mRNA ratio, improved myelination of small fibres, increased nerve conduction and ameliorated the clinical phenotype. PXT3003 also improved axonal regeneration and remyelination in the murine nerve crush model. Based on these observations in preclinical models, a clinical trial of PTX3003 in CMT1A, a neglected orphan disease, is warranted. If the efficacy of PTX3003 is confirmed, rational polytherapy based on novel combinations of existing non-toxic drugs with pleiotropic effects may represent a promising approach for rapid drug development.

Figures

Figure 1
Figure 1
Pharmacology network-based drug repurposing for CMT1A disease. For detailed explanations and abbreviations, see Additional file 1. (A) Three principal pathways regulating expression of PMP22 gene through extracellular GPCR signalling in Schwann cells. The functional interaction between cAMP pathway, neurosteroid-mediated signalling and PI3K-AKT/ERK kinase cascades, activated by receptor tyrosine kinases, control the expression of PMP22 gene in Schwann cells. Individual drugs are shown in green. Blue shading: cAMP pathway; green shading: neurosteroid-mediated signalling; yellow shading: RTK/PI3K-AKT/ERK kinase cascades. (B) Cytoprotective and neuromodulator actions of PXT3003 drug combination in peripheral neurons. Dysfunction of CMT1A Schwann cells could affect membrane excitability of neuronal cells, leading to abnormal processing of neuronal information, cytotoxicity and axonal loss. GPCRs, modulated by PXT3003 drugs, are well-known regulators of neuronal excitability and pain sensation, are able to activate cytoprotective signalling pathways in different experimental settings, and could preserve functional integrity of peripheral neurons in CMT1 patients. Baclofen, naltrexone and sorbitol are shown in green. Red symbols: established or putative functional targets for PTX3003 drugs. Blue arrows: activation links; red lines: inhibition links; dashed lines: functional effect link is receptor-type specific.
Figure 2
Figure 2
BCL, NTX, SRB and their mix (PXT3003) improve myelination in DRG co-cultures. (A) and (B) PXT3003 treatment (Dose 2) of myelinating DRG co-cultures of neurons and Schwann cells derived from CMT1A transgenic rats (TG) demonstrated an increased length of MBP-positive fibres (red staining in (B)) compared to the untreated control (A). Scale bar: 56 μm. Blue staining: DAPI. (C) to (F) After 10–11 days of treatment of TG DRG co-cultures with single compounds BCL (C), NTX (D), SRB (E) or their PXT3003 (F) combination at four doses (Dose 1: 0.32 nM of BCL, 0.32 nM of NTX and 32 nM of SRB; Dose 2: 1.6 nM of BCL, 1.6 nM of NTX and 160 nM of SRB; Dose 3: 8 nM of BCL, 8 nM of NTX and 800 nM of SRB; Dose 4: 40 nM of BCL, 40 nM of NTX and 4 μM of SRB), myelination was significantly improved (featured by an increased length of MBP-stained segments). (G) The synergistic potential of PXT3003 (Dose 2: 1.6 nM of BCL, 1.6 nM of NTX and 160 nM of SRB) was displayed by an isobologram plotted from dose-effect curves of each drug. The calculated Combination Index (CI) of synergy was 0.36; Synergism is characterized by a CI < 1. Greyed surface represents the concentrations of the mixes at which CI = 1. Three independent DRG co-cultures with 6 replicates each were performed and analysed. +, *P < 0.05; ++, **P < 0.01; ***P < 0.001 vs Vehicle; ANOVA with Dunnett’s test (*) and t-test (+). Data are shown as mean + SEM. Vhc: Vehicle.
Figure 3
Figure 3
BCL, NTX, SRB and their mix (PXT3003) down-regulatePmp22mRNA expression in RT4 Schwannoma cells. (A) After a treatment of 8 hours of RT4 Schwannoma cells with single compounds (100 nM of BCL, 100 nM of NTX or 10 μM of SRB) and with PXT3003 combination (100 nM of BCL, 100 nM of NTX and 10 μM of SRB; same drug ratio that was used for all doses in DRG co-cultures), a significant decrease of Pmp22 mRNA expression level was observed when compared to vehicle (Actb and Rps9 used together as reference genes) with a significant observed higher efficacy of PXT3003 over single compounds. (B) The same drugs had no effect on Mpz expression (Actb and Rps9 used as reference genes). (C) The expression of Pmp22 with respect to Mpz was significantly decreased after PXT3003 treatment, unlike single compounds. Three different experiments using 2 cultures each with 3 replicates were performed and analysed. *P < 0.05, ***P < 0.001 vs Vehicle; ANOVA with Dunnett’s test. #P < 0.05 vs the most active compound; post-hoc contrast test. Data are shown as mean + SEM.
Figure 4
Figure 4
Daily oral treatment of CMT1A rats with PXT3003 down-regulatesPmp22expression and reduces signs of motor and sensory neuropathy. (A) A 9-week treatment with PXT3003 (BCL 30 μg/kg, NTX 3.5 μg/kg and SRB 1.05 mg/kg) decreased Pmp22 to Mpz mRNA ratio in the sciatic nerve. n = 18, 20 and 18 animals for respectively WT vehicle, TG vehicle and TG PXT3003 groups. (B) Bar test. The latency to fall for transgenic rats was significantly improved after 9 weeks of treatment with PXT3003. (C) Kaplan-Meier representation of the data set in (B) demonstrated a positive effect of PXT3003. (D) After 9 weeks of treatment, the inclined plane score was significantly improved for transgenic rats. (B) to (D)n = 36, 38 and 36 animals for respectively WT vehicle, TG vehicle and TG PXT3003 groups. (E) A 4-month long PXT3003 treatment normalised the impaired thermal sensitivity of transgenic rats in the hot plate test. n = 12, 10 and 7 animals respectively for WT, vehicle, TG vehicle and TG PXT3003 groups. *P < 0.05; **P < 0.01; ***P < 0.001 vs TG vehicle; ANOVA with Dunnett’s test (except in (C), logrank). Data are shown as mean + SEM.
Figure 5
Figure 5
Daily oral treatment of CMT1A rats with PXT3003 improves myelination and electrophysiology. A 4-month PXT3003 treatment (BCL 30 μg/kg, NTX 3.5 μg/kg and SRB 1.05 mg/kg) of TG rats significantly increased the number of myelinated axons in sciatic nerve cross sections (A) mostly in the small to medium sized axon class of myelinated axons (< 4 μm) (B). n = 12, 12 and 15 animals for respectively WT vehicle, TG vehicle and TG PXT3003 groups. (C) 8-month PXT3003 treatment increased the motor nerve conduction velocity of TG rats in sciatic nerve. n = 11, 9 and 7 animals for respectively WT vehicle, TG vehicle and TG PXT3003 groups. (D) Representative images of toluidine blue-stained sciatic nerve cross sections for each group. Scale bar: 25 μm. *,+ P < 0.05; ** P < 0.01; ***P < 0.001 vs TG vehicle; ANOVA with Dunnett’s test (except in (C), + t-test). Data are shown as mean + SEM.
Figure 6
Figure 6
Daily oral PXT3003 treatment of nerve-crushed mice restores nerve physiology, improves axonal and myelin integrity and functional behaviour. (A) A 21 and 30 day-treatment with PXT3003 (BCL 60 μg/kg, NTX 7 μg/kg and SRB 2.1 mg/kg) increased the amplitude of CMAP (measured in the gastrocnemius) of crushed nerves in male mice. n = 10 animals in each group. (B) to (D) A 42 day-treatment with PXT3003 significantly improved axonal calibre size (B), normalised the distribution of the number of myelinated axons in terms of axonal diameter (C) and improved the distribution of myelin g-ratio in tibial nerve cross sections (D). n = 6 for each group. (E) After 13 days of treatment, PXT3003 (BCL 600 μg/kg, NTX 70 μg/kg and SRB 21 mg/kg) improved significantly the functional activity of crushed mice as assessed by the paw surface pressure against the floor (paw surface bearing) of the affected paw normalised to the contralateral non-affected one. (F) to (H) In contrast to PXT3003, single drugs had no effect on surface bearing ratio. n = 10 for each group. *P < 0.05, **P < 0.01, ***P < 0.001 vs Crush Vehicle; t–test. Data are shown as mean ± SEM.

References

    1. Patzkó A, Shy ME. Update on Charcot-Marie-Tooth disease. Curr Neurol Neurosci Rep. 2011;11:78–88. doi: 10.1007/s11910-010-0158-7.
    1. Rossor AM, Polke JM, Houlden H, Reilly MM. Clinical implications of genetic advances in Charcot-Marie-Tooth disease. Nat Rev Neurol. 2013;9:562–571. doi: 10.1038/nrneurol.2013.179.
    1. Fledrich R, Stassart RM, Klink A, Rasch LM, Prukop T, Haag L, Czesnik D, Kungl T, Abdelaal TAM, Keric N, Stadelmann C, Brück W, Nave K-A, Sereda MW. Soluble neuregulin-1 modulates disease pathogenesis in rodent models of Charcot-Marie-Tooth disease 1A. Nat Med. 2014;20:1055–1061. doi: 10.1038/nm.3664.
    1. Nave K-A, Trapp BD. Axon-glial signaling and the glial support of axon function. Annu Rev Neurosci. 2008;31:535–561. doi: 10.1146/annurev.neuro.30.051606.094309.
    1. Sereda M, Griffiths I, Pühlhofer A, Stewart H, Rossner MJ, Zimmerman F, Magyar JP, Schneider A, Hund E, Meinck HM, Suter U, Nave KA. A transgenic rat model of Charcot-Marie-Tooth disease. Neuron. 1996;16:1049–1060. doi: 10.1016/S0896-6273(00)80128-2.
    1. Cosgaya JM, Chan JR, Shooter EM. The neurotrophin receptor p75NTR as a positive modulator of myelination. Science. 2002;298:1245–1248. doi: 10.1126/science.1076595.
    1. Rangaraju S, Madorsky I, Pileggi JG, Kamal A, Notterpek L. Pharmacological induction of the heat shock response improves myelination in a neuropathic model. Neurobiol Dis. 2008;32:105–115. doi: 10.1016/j.nbd.2008.06.015.
    1. Callizot N, Combes M, Steinschneider R, Poindron P. A new long term in vitro model of myelination. Exp Cell Res. 2011;317:2374–2383. doi: 10.1016/j.yexcr.2011.07.002.
    1. Sereda MW, Meyer zu Hörste G, Suter U, Uzma N, Nave K-A. Therapeutic administration of progesterone antagonist in a model of Charcot-Marie-Tooth disease (CMT-1A) Nat Med. 2003;9:1533–1537. doi: 10.1038/nm957.
    1. Tashiro H, Fukuda Y, Kimura A, Hoshino S, Ito H, Dohi K. Assessment of microchimerism in rat liver transplantation by polymerase chain reaction. Hepatology. 1996;23:828–834. doi: 10.1002/hep.510230425.
    1. Hirst JA, Howick J, Aronson JK, Roberts N, Perera R, Koshiaris C, Heneghan C. The need for randomization in animal trials: an overview of systematic reviews. PLoS One. 2014;9:e98856. doi: 10.1371/journal.pone.0098856.
    1. Kilkenny C, Browne WJ, Cuthill IC, Emerson M, Altman DG. Improving bioscience research reporting: the ARRIVE guidelines for reporting animal research. J Pharmacol Pharmacother. 2010;1:94–99. doi: 10.4103/0976-500X.72351.
    1. Meyer Zu Horste G, Prukop T, Liebetanz D, Mobius W, Nave K-A, Sereda MW. Antiprogesterone therapy uncouples axonal loss from demyelination in a transgenic rat model of CMT1A neuropathy. Ann Neurol. 2007;61:61–72. doi: 10.1002/ana.21026.
    1. Rivlin AS, Tator CH. Objective clinical assessment of motor function after experimental spinal cord injury in the rat. J Neurosurg. 1977;47:577–581. doi: 10.3171/jns.1977.47.4.0577.
    1. Beyreuther BK, Callizot N, Brot MD, Feldman R, Bain SC, Stöhr T. Antinociceptive efficacy of lacosamide in rat models for tumor- and chemotherapy-induced cancer pain. Eur J Pharmacol. 2007;565:98–104. doi: 10.1016/j.ejphar.2007.02.041.
    1. Tétreault P, Dansereau M-A, Doré-Savard L, Beaudet N, Sarret P. Weight bearing evaluation in inflammatory, neuropathic and cancer chronic pain in freely moving rats. Physiol Behav. 2011;104:495–502. doi: 10.1016/j.physbeh.2011.05.015.
    1. Bordet T, Buisson B, Michaud M, Drouot C, Gale P, Delaage P, Akentieva NP, Evers AS, Covey DF, Ostuni MA, Lacape J, Massaad C, Schumacher M, Steidl E, Maux D, Delaage M, Henderson CE, Pruss RM. Identification and characterization of cholest-4-en-3-one, oxime (TRO19622), a novel drug candidate for amyotrophic lateral sclerosis. J Pharmacol Exp Ther. 2007;322:709–720. doi: 10.1124/jpet.107.123000.
    1. Markowski C, Markowski E. Conditions for the effectiveness of a preliminary test of variance. Am Stat. 1990;44:322–326.
    1. Sawilowsky SS, Blair RC. A more realistic look at the robustness and Type II error properties of the t test to departures from population normality. Psychol Bull. 1992;111:352–360. doi: 10.1037/0033-2909.111.2.352.
    1. Grabovsky Y, Tallarida RJ. Isobolographic analysis for combinations of a full and partial agonist: curved isoboles. J Pharmacol Exp Ther. 2004;310:981–986. doi: 10.1124/jpet.104.067264.
    1. Chou T-C. Theoretical basis, experimental design, and computerized simulation of synergism and antagonism in drug combination studies. Pharmacol Rev. 2006;58:621–681. doi: 10.1124/pr.58.3.10.
    1. Geary N. Understanding synergy. Am J Physiol Metab. 2013;304:E237–E253.
    1. Tallarida RJ. An overview of drug combination analysis with isobolograms. J Pharmacol Exp Ther. 2006;319:1–7. doi: 10.1124/jpet.106.104117.
    1. Suter U, Snipes GJ, Schoener-Scott R, Welcher AA, Pareek S, Lupski JR, Murphy RA, Shooter EM, Patel PI. Regulation of tissue-specific expression of alternative peripheral myelin protein-22 (PMP22) gene transcripts by two promoters. J Biol Chem. 1994;269:25795–25808.
    1. Sabéran-Djoneidi D, Sanguedolce V, Assouline Z, Lévy N, Passage E, Fontés M. Molecular dissection of the Schwann cell specific promoter of the PMP22 gene. Gene. 2000;248:223–231. doi: 10.1016/S0378-1119(00)00116-5.
    1. Ogata T, Iijima S, Hoshikawa S, Miura T, Yamamoto S, Oda H, Nakamura K, Tanaka S. Opposing extracellular signal-regulated kinase and Akt pathways control Schwann cell myelination. J Neurosci. 2004;24:6724–6732. doi: 10.1523/JNEUROSCI.5520-03.2004.
    1. Arthur-Farraj P, Wanek K, Hantke J, Davis CM, Jayakar A, Parkinson DB, Mirsky R, Jessen KR. Mouse schwann cells need both NRG1 and cyclic AMP to myelinate. Glia. 2011;59:720–733. doi: 10.1002/glia.21144.
    1. Pereira JA, Lebrun-Julien F, Suter U. Molecular mechanisms regulating myelination in the peripheral nervous system. Trends Neurosci. 2012;35:123–134. doi: 10.1016/j.tins.2011.11.006.
    1. Faroni A, Magnaghi V. The neurosteroid allopregnanolone modulates specific functions in central and peripheral glial cells. Front Endocrinol (Lausanne) 2011;2:103.
    1. Procacci P, Ballabio M, Castelnovo LF, Mantovani C, Magnaghi V. GABA-B receptors in the PNS have a role in Schwann cells differentiation? Front Cell Neurosci. 2012;6:68.
    1. Glenn TD, Talbot WS. Signals regulating myelination in peripheral nerves and the Schwann cell response to injury. Curr Opin Neurobiol. 2013;23:1041–1048. doi: 10.1016/j.conb.2013.06.010.
    1. Towers S, Princivalle A, Billinton A, Edmunds M, Bettler B, Urban L, Bowery NG. GABA B receptor protein and mRNA distribution in rat spinal cord and dorsal root ganglia. Eur J Neurosci. 2000;12:3201–3210. doi: 10.1046/j.1460-9568.2000.00237.x.
    1. Wang H-Y, Frankfurt M, Burns LH. High-affinity naloxone binding to filamin a prevents mu opioid receptor-Gs coupling underlying opioid tolerance and dependence. PLoS One. 2008;3:e1554. doi: 10.1371/journal.pone.0001554.
    1. Hytrek SD, McLaughlin PJ, Lang CM, Zagon IS. Inhibition of human colon cancer by intermittent opioid receptor blockade with naltrexone. Cancer Lett. 1996;101:159–164. doi: 10.1016/0304-3835(96)04119-5.
    1. Zhu M, Li RC. Receptor binding activities of Schefflera triterpenoids and oligosaccharides. Planta Med. 1999;65:99–103. doi: 10.1055/s-1999-13967.
    1. Singer MA, Lindquist S. Multiple effects of trehalose on protein folding in vitro and in vivo. Mol Cell. 1998;1:639–648. doi: 10.1016/S1097-2765(00)80064-7.
    1. Kumar R. Role of naturally occurring osmolytes in protein folding and stability. Arch Biochem Biophys. 2009;491:1–6. doi: 10.1016/j.abb.2009.09.007.
    1. Notterpek L, Ryan MC, Tobler AR, Shooter EM. PMP22 accumulation in aggresomes: implications for CMT1A pathology. Neurobiol Dis. 1999;6:450–460. doi: 10.1006/nbdi.1999.0274.
    1. Schlebach JP, Peng D, Kroncke BM, Mittendorf KF, Narayan M, Carter BD, Sanders CR. Reversible folding of human peripheral myelin protein 22, a tetraspan membrane protein. Biochemistry. 2013;52:3229–3241. doi: 10.1021/bi301635f.
    1. Nobbio L, Mancardi G, Grandis M, Levi G, Suter U, Nave KA, Windebank AJ, Abbruzzese M, Schenone A. PMP22 transgenic dorsal root ganglia cultures show myelin abnormalities similar to those of human CMT1A. Ann Neurol. 2001;50:47–55. doi: 10.1002/ana.1034.
    1. Vandesompele J, De Preter K, Pattyn F, Poppe B, Van Roy N, De Paepe A, Speleman F. Accurate normalization of real-time quantitative RT-PCR data by geometric averaging of multiple internal control genes. Genome Biol. 2002;3:RESEARCH0034. doi: 10.1186/gb-2002-3-7-research0034.
    1. Fledrich R, Schlotter-Weigel B, Schnizer TJ, Wichert SP, Stassart RM, Meyer zu Hörste G, Klink A, Weiss BG, Haag U, Walter MC, Rautenstrauss B, Paulus W, Rossner MJ, Sereda MW. A rat model of Charcot-Marie-Tooth disease 1A recapitulates disease variability and supplies biomarkers of axonal loss in patients. Brain. 2012;135:72–87. doi: 10.1093/brain/awr322.
    1. Shy ME, Chen L, Swan ER, Taube R, Krajewski KM, Herrmann D, Lewis RA, McDermott MP. Neuropathy progression in Charcot-Marie-Tooth disease type 1A. Neurology. 2008;70:378–383. doi: 10.1212/01.wnl.0000297553.36441.ce.
    1. Pareyson D, Scaioli V, Laurà M. Clinical and electrophysiological aspects of Charcot-Marie-Tooth disease. Neuromolecular Med. 2006;8:3–22. doi: 10.1385/NMM:8:1-2:3.
    1. Martini R, Klein D, Groh J. Similarities between inherited demyelinating neuropathies and Wallerian degeneration: an old repair program may cause myelin and axon perturbation under nonlesion conditions. Am J Pathol. 2013;183:655–660. doi: 10.1016/j.ajpath.2013.06.002.
    1. Castorina A, Scuderi S, D’Amico AG, Drago F, D’Agata V. PACAP and VIP increase the expression of myelin-related proteins in rat schwannoma cells: involvement of PAC1/VPAC2 receptor-mediated activation of PI3K/Akt signaling pathways. Exp Cell Res. 2014;322:108–121. doi: 10.1016/j.yexcr.2013.11.003.
    1. Hai M, Muja N, DeVries GH, Quarles RH, Patel PI. Comparative analysis of Schwann cell lines as model systems for myelin gene transcription studies. J Neurosci Res. 2002;69:497–508. doi: 10.1002/jnr.10327.
    1. Nobbio L, Visigalli D, Radice D, Fiorina E, Solari A, Lauria G, Reilly MM, Santoro L, Schenone A, Pareyson D. PMP22 messenger RNA levels in skin biopsies: testing the effectiveness of a Charcot-Marie-Tooth 1A biomarker. Brain. 2014;137:1614–1620. doi: 10.1093/brain/awu071.
    1. Katona I, Wu X, Feely SME, Sottile S, Siskind CE, Miller LJ, Shy ME, Li J. PMP22 expression in dermal nerve myelin from patients with CMT1A. Brain. 2009;132:1734–1740. doi: 10.1093/brain/awp113.
    1. Hay M, Thomas DW, Craighead JL, Economides C, Rosenthal J. Clinical development success rates for investigational drugs. Nat Biotechnol. 2014;32:40–51. doi: 10.1038/nbt.2786.
    1. Attarian S, Vallat J-M, Magy L, Funalot B, Gonnaud P-M, Lacour A, Péréon Y, Dubourg O, Pouget J, Micallef J, Franques J, Lefèvre M-N, Ghorab K, Al-Moussawi M, Tiffreau V, Magot A, Leclair-Visonneau L, Stojkovic T, Bossi L, Lehert P, Walter G, Bertrand V, Mandel J, Milet A, Hajj R, Boudiaf L, Scart-Grès C, Nabirotchkin S, Guedj M, Chumakov I, et al: An Exploratory randomised double-blind and placebo-controlled phase 2 study of a combination of baclofen, naltrexone and sorbitol (PXT3003) in patients with Charcot-Marie-Tooth disease type 1A.Orphanet J Rare Dis 2014, 9:199.

Source: PubMed

3
Tilaa