A randomised controlled trial of bumetanide in the treatment of autism in children

E Lemonnier, C Degrez, M Phelep, R Tyzio, F Josse, M Grandgeorge, N Hadjikhani, Y Ben-Ari, E Lemonnier, C Degrez, M Phelep, R Tyzio, F Josse, M Grandgeorge, N Hadjikhani, Y Ben-Ari

Abstract

Gamma aminobutyric acid (GABA)-mediated synapses and the oscillations they orchestrate are altered in autism. GABA-acting benzodiazepines exert in some patients with autism paradoxical effects, raising the possibility that like in epilepsies, GABA excites neurons because of elevated intracellular concentrations of chloride. Following a successful pilot study,(1) we have now performed a double-blind clinical trial using the diuretic, chloride-importer antagonist bumetanide that reduces intracellular chloride reinforcing GABAergic inhibition. Sixty children with autism or Asperger syndrome (3-11 years old) received for 3 months placebo or bumetanide (1 mg daily), followed by 1-month wash out. Determination of the severity of autism was made with video films at day 0 (D0) and D90 by blind, independent evaluators. Bumetanide reduced significantly the Childhood Autism Rating Scale (CARS) (D90-D0; P<0.004 treated vs placebo), Clinical Global Impressions (P<0.017 treated vs placebo) and Autism Diagnostic Observation Schedule values when the most severe cases (CARS values above the mean ± s.d.; n=9) were removed (Wilcoxon test: P-value=0.031; Student's t-test: P-value=0.017). Side effects were restricted to an occasional mild hypokalaemia (3.0-3.5 mM l(-1) K(+)) that was treated with supplemental potassium. In a companion study, chronic bumetanide treatment significantly improved accuracy in facial emotional labelling, and increased brain activation in areas involved in social and emotional perception (Hadjikhani et al., submitted). Therefore, bumetanide is a promising novel therapeutic agent to treat autism. Larger trials are warranted to better determine the population best suited for this treatment.

Trial registration: ClinicalTrials.gov NCT01078714.

Figures

Figure 1
Figure 1
Schematic diagram of the protocol used. A total of 60 children were recruited for the trial, 6 withdrew for various reasons (see text) and the remaining 54 completed treatments and assessment. The video film was not available for one child and thus ADOS could not be performed. Tests included CARS at D0, 90 and 120 after 1-month wash out, CGI at D0, 90 and 120, and ADOS at D0 and 90.
Figure 2
Figure 2
All the values obtained with CARS are depicted. Note the significant differences between placebo and bumetanide treated patients and a partial return to pretreatment values after 1-month wash-out period. The number of items >3 was also significantly reduced as shown at the right side of the figure (number corresponding to D0, D90 and D120; ***p<0.005 for D90 and **p<0.05 for D120).
Figure 3
Figure 3
Top figure: Boxplot of ADOS total scores gains between the beginning and the end of the trial (90 days later) for patient receiving placebo (left panel) and treatment (right panel). Bottom: Boxplot of ADOS total scores gains but excluding patients with ‘above normal' CARS scores (that is, above mean±s.d.)—between the beginning and the end of the trial (90 days later) for patient receiving placebo (left panel) and treatment (right panel).

References

    1. Lemonnier E, Ben-Ari Y. The diuretic bumetanide decreases autistic behaviour in five infants treated during 3 months with no side effects. Acta Paediatr. 2010;99:1885–1888.
    1. Courchesne E, Mouton PR, Calhoun ME, Semendeferi K, hrens-Barbeau C, Hallet MJ, et al. Neuron number and size in prefrontal cortex of children with autism 1. JAMA. 2011;306:2001–2010.
    1. Khan NZ, Gallo LA, Arghir A, Budisteanu B, Budisteanu M, Dobrescu I, et al. Autism and the grand challenges in global mental health. Autism Res. 2012;5:156–159.
    1. Elsabbagh M, Divan G, Koh YJ, Kim YS, Kauchali S, Marcin C, et al. Global prevalence of autism and other pervasive developmental disorders. Autism Res. 2012;5:160–179.
    1. Lord C, Rutter M, Le CA. Autism diagnostic interview-revised: a revised version of a diagnostic interview for caregivers of individuals with possible pervasive developmental disorders. J Autism Dev Disord. 1994;24:659–685.
    1. Lord C, Risi S, Lambrecht L, Cook EH, Leventhal BL, DiLavore PC, et al. The autism diagnostic observation schedule-generic: a standard measure of social and communication deficits associated with the spectrum of autism. J Autism Dev Disord. 2000;30:205–223.
    1. Jamain S, Quach H, Betancur C, Rastam M, Colineaux C, Gillberg IC, et al. Mutations of the X-linked genes encoding neuroligins NLGN3 and NLGN4 are associated with autism. Nat Genet. 2003;34:27–29.
    1. Bourgeron T. A synaptic trek to autism. Curr Opin Neurobiol. 2009;19:231–234.
    1. Laumonnier F, Bonnet-Brilhault F, Gomot M, Blanc R, David A, Moizard MP, et al. X-linked mental retardation and autism are associated with a mutation in the NLGN4 gene, a member of the neuroligin family. Am J Hum Genet. 2004;74:552–557.
    1. Giannandrea M, Bianchi V, Mignogna ML, Sirri A, Carrabino S, D'Elia E, et al. Mutations in the Small GTPase Gene RAB39B are responsible for x-linked mental retardation associated with autism, epilepsy, and macrocephaly. Am J Hum Genet. 2010;86:185–195.
    1. Weiss LA. Autism genetics: emerging data from genome-wide copy-number and single nucleotide polymorphism scans. Expert Rev Mol Diagn. 2009;9:795–803.
    1. Tabuchi K, Blundell J, Etherton MR, Hammer RE, Liu X, Powell CM, et al. A neuroligin-3 mutation implicated in autism increases inhibitory synaptic transmission in mice. Science. 2007;318:71–76.
    1. Chubykin AA, Atasoy D, Etherton MR, Brose N, Kavalali ET, Gibson JR, et al. Activity-dependent validation of excitatory versus inhibitory synapses by neuroligin-1 versus neuroligin-2. Neuron. 2007;54:919–931.
    1. Croen L, Grether JK, Yoshida CK, Odouli R, Hendrick V. Antidepressant use during pregnancy and childhood autism spectrum disorders. Arch General Psychiatry. 2011;68:1104–1112.
    1. Patterson PH. Immune involvement in schizophrenia and autism: etiology, pathology and animal models. Behav Brain Res. 2009;204:313–321.
    1. Kemper TL, Bauman M. Neuropathology of infantile autism. J Neuropathol Exp Neurol. 1998;57:645–652.
    1. Dossche DM.GABA in autism. international review of neurobiology.vol. 71.Academic Press: Amsterdam; 2005 pp 1–481.
    1. Chao HT, Chen HM, Samaco RC, Xue MS, Chahrour M, Yoo J, et al. Dysfunction in GABA signalling mediates autism-like stereotypies and Rett syndrome phenotypes 1. Nature. 2010;468:263–269.
    1. Zhang C, Atasoy D, Arac D, Yang X, Fucillo MV, Robison AJ, et al. Neurexins physically and functionally interact with GABA(A) receptors. Neuron. 2010;66:403–416.
    1. Pizzarelli R, Cherubini E. Alterations of gabaergic signaling in autsim spectrum disorders. Neural Plast. 2011;297:1–12.
    1. Gogolla N, LeBlanc JJ, Quast KB, Sudhof TC, Fagiolini M, Hensch TK. Common circuit defect of excitatory-inhibitory balance in mouse models of autism 1. J Neurodev Disord. 2009;1:172–181.
    1. Singer W. Synchronization of cortical activity and its putative role in information-processing and learning 21. Annu Rev Physiol. 1993;55:349–374.
    1. Lisman JE, Idiart MAP. Storage of 7+/−2 short-term memories in oscillatory subcycles 1. Science. 1995;267:1512–1515.
    1. Murthy VN, Fetz EE. Coherent 25-Hz to 35-Hz oscillations in the sensorimotor cortex of awake behaving monkeys 1. Proc Natl Acad Sci USA. 1992;89:5670–5674.
    1. Lewis DA, Hashimoto T, Volk DW. Cortical inhibitory neurons and schizophrenia. Nat Rev Neurosci. 2005;6:312–324.
    1. Lisman JE, Coyle JT, Green RW, Javitt DC, Benes FM, Heckers S, et al. Circuit-based framework for understanding neurotransmitter and risk gene interactions in schizophrenia. Trends Neurosci. 2008;31:234–242.
    1. Gogolla N, LeBlanc JJ, Quast KB, Sudhof TC, Fagiolini M, Hensch TK. Common circuit defect of excitatory-inhibitory balance in mouse models of autism. J Neurodev Disord. 2009;1:172–181.
    1. Grice SJ, Spratling MW, Karmiloff-Smith A, Halit H, Csibra G, de Haan M, et al. Disordered visual processing and oscillatory brain activity in autism and Williams syndrome. Neuroreport. 2001;12:2697–2700.
    1. Brown C, Gruber T, Boucher J, Rippon G, Brock J. Gamma abnormalities during perception of illusory figures in autism. Cortex. 2005;41:364–376.
    1. Wilson TW, Rojas DC, Reite ML, Teale PD, Rogers SJ. Children and adolescents with autism exhibit reduced MEG steady-state gamma responses. Biol Psychiatry. 2007;62:192–197.
    1. Tierney AL, Gabard-Durnam L, Vogel-Farley V, Tager-Flusberg H, Nelson CA. Developmental trajectories of resting EEG power: an endophenotype of autism spectrum disorder. Plos One. 2012;7 (6
    1. Marrosu F, Marrosu G, Rachel M, Biggio G. Paradoxical reactions elicited by diazepam in children with classic autism. Funct Neurol. 1987;2:355–361.
    1. Wing L, Shah A. Catatonia in autistic spectrum disorders. Br J Psychiatry. 2000;176:357–362.
    1. Hutton J, Goode S, Murphy M, Le Couteur A, Rutter M. New-onset psychiatric disorders in individuals with autism. Autism. 2008;12:373–390.
    1. Ungvari GS, Kau LS, Wai-Kwong T, Shing NF. The pharmacological treatment of catatonia: an overview. Eur Arch Psychiatry Clin Neurosci. 2001;251:I31–I34.
    1. Tibrewal P, Narayanaswamy J, Zutshi A, Srinivasaraju R, Math SB. Response rate of lorazepam in catatonia: A developing country's perspective. Prog Neuro-Psych Biol Psych. 2010;34:1520–1522.
    1. Fink M, Taylor MA, Ghaziuddin N. Catatonia in autistic spectrum disorders: a medical treatment algorithm. Int Rev Neurobiol. 2006;72:233–244.
    1. Ghaziuddin M, Quinlan P, Ghaziuddin N. Catatonia in autism: a distinct subtype. J Intellectual Disabil Res. 2005;49:102–105.
    1. Orekhova EV, Stroganova TA, Prokofiev AO, Nygren G, Gillberg C, Elam M. Right hemispheric failure to respond to temporal novelty in autism: evidence from an event-related potential study. Int J Psychophysiol. 2008;69:186.
    1. Enticott PG, Rinehart NJ, Tonge BJ, Bradshaw JL, Fitzgerald PB. Repetitive transcranial magnetic stimulation (rTMS) improves movement-related cortical potentials in autism spectrum disorders. Brain Stimulation. 2012;5:30–37.
    1. Painter MJ, Scher MS, Stein AD, Armatti S, Wang ZM, Gardiner JC, et al. Phenobarbital compared with phenytoin for the treatment of neonatal seizures. N Engl J Med. 1999;341:485–489.
    1. Boylan GB, Rennie JM, Pressler RM, Wilson G, Morton M, Binnie CD. Phenobarbitone neonatal seizures, and video-EEG. Arch Dis Child Fetal Neonatal Ed. 2002;86:F165–F170.
    1. Guillet R, Kwon J. Seizure recurrence and developmental disabilities after neonatal seizures: outcomes are unrelated to use of phenobarbital prophylaxis. J Child Neurol. 2007;22:389–395.
    1. Kaindl AM, Koppelstaetter A, Nebrich G, Stuwe J, Sifringer M, Zabel C, et al. Brief alteration of NMDA or GABAA receptor-mediated neurotransmission has long term effects on the developing cerebral cortex. Mol Cell Proteomics. 2008;7:2293–2310.
    1. Huberfeld G, Wittner L, Clemenceau S, Baulac M, Miles R, Kaila K, et al. Perturbed expression of the K-Cl cotransporter KCC2 and GABAergic signalling in human temporal lobe epilepsy. Epilepsia. 2006;47:9.
    1. Cohen I, Navarro V, Clemenceau S, Baulac M, Miles R. On the origin of interictal activity in human temporal lobe epilepsy in vitro. Science. 2002;298:1418–1421.
    1. Nardou R, Yamamoto S, Chazal G, Bhar A, Ferrand N, Dulac O, et al. Neuronal chloride accumulation and excitatory GABA underlie aggravation of neonatal epileptiform activities by phenobarbital. Brain. 2011;134:987–1002.
    1. Ben Ari Y, Gaiarsa JL, Tyzio R, Khazipov R. GABA: a pioneer transmitter that excites immature neurons and generates primitive oscillations. Physiol Rev. 2007;87:1215–1284.
    1. Dzhala VI, Talos DM, Sdrulla DA, Brumback AC, Mathews GC, Benke TA, et al. NKCC1 transporter facilitates seizures in the developing brain. Nat Med. 2005;11:1205–1213.
    1. Payne JA, Rivera C, Voipio J, Kaila K. Cation-chloride co-transporters in neuronal communication, development and trauma. Trends Neurosci. 2003;26:199–206.
    1. Rivera C, Voipio J, Payne JA, Ruusuvuori E, Lahtinen H, Lamsa K, et al. The K+/Cl− co-transporter KCC2 renders GABA hyperpolarizing during neuronal maturation. Nature. 1999;397:251–255.
    1. Delpire E. Cation-chloride cotransporters in neuronal communication. News Physiol Sci. 2000;15:309–312.
    1. Feit PW. Bumetanide--the way to its chemical structure. J Clin Pharmacol. 1981;21:531–536.
    1. Delpire E, Mount DB. Human and murine phenotypes associated with defects in cation-chloride cotransport. Annu Rev Physiol. 2002;64:803–843.
    1. Blaesse P, Airaksinen MS, Rivera C, Kaila K. Cation-chloride cotransporters and neuronal function. Neuron. 2009;61:820–838.
    1. Sullivan JE, Witte MK, Yamashita TS, Myers CM, Blumer JL. Dose-ranging evaluation of bumetanide pharmacodynamics in critically ill infants. Clin Pharmacol Ther. 1996;60:424–434.
    1. Le Couteur A, Rutter M, Lord C, Rios P, Robertson S, Holdgrafer M, et al. Autism diagnostic interview: a standardized investigator-based instrument. J Autism Dev Disord. 1989;19:363–387.
    1. Schopler E, Reichler RJ, DeVellis RF, Daly K. Toward objective classification of childhood autism: Childhood Autism Rating Scale (CARS) J Autism Dev Disord. 1980;10:91–103.
    1. DiLalla DL, Rogers SJ. Domains of the Childhood Autism Rating Scale: relevance for diagnosis and treatment. J Autism Dev Disord. 1994;24:115–128.
    1. Masi G, Cosenza A, Millepiedi S, Muratori F, Pari C, Salvadori F. Aripiprazole monotherapy in children and young adolescents with pervasive developmental disorders: a retrospective study. CNS Drugs. 2009;23:511–521.
    1. Geier DA, Kern JK, Davis G, King PG, Adams JB, Young JL, et al. A prospective double-blind, randomized clinical trial of levocarnitine to treat autism spectrum disorders. Med Sci Monit. 2011;17:I15–I23.
    1. Canitano R, Scandurra V. Psychopharmacology in autism: an update. Prog Neuro-Psych Biol Psych. 2011;35:18–28.
    1. Guy W. Assessment Manual for Psychopharmacology. Early Clinical Drug Evaluation Unit. National institute of mental health: Washington, DC; 2000.
    1. Crowley JC, Katz LC. Development of ocular dominance columns in the absence of retinal input. Nat Neurosci. 1999;2:1125–1130.
    1. Owley T, McMahon W, Cook EH, Laulhere T, South M, Mays LZ, et al. Multisite, double-blind, placebo-controlled trial of porcine secretin in autism. J Am Acad Child Adolesc Psychiatry. 2001;40:1293–1299.
    1. Papauasiliou AS, Nikaina I, Rizou J, Alexandrou S. The effect of a psycho-educational program on CARS scores and short sensory profile in autistic children. Eur J Paediatr Neurol. 2011;15:338–344.
    1. Masi G, Cosenza A, Mucci M, Brovedani P. Open trial of risperidone in 24 young children with pervasive developmental disorders. J Am Acad Child Adolesc Psychiatry. 2001;40:1206–1214.
    1. Nagaraj R, Singhi P, Malhi P. Risperidone in children with autism: randomized, placebo-controlled, double-blind study. J Child Neurol. 2006;21:450–455.
    1. Canitano R, Scandurra V. Risperidone in the treatment of behavioral disorders associated with autism in children and adolescents. Neuropsychiatr Dis Treat. 2008;4:723–730.
    1. Luby J, Mrakotsky C, Stalets MM, Belden A, Heffelfinger A, Williams M, et al. Risperidone in preschool children with autistic spectrum disorders: an investigation of safety and efficacy. J Child Adolesc Psychopharmacol. 2006;16:575–587.
    1. Shea S, Turgay A, Carroll A, Schulz M, Orlik H, Smith I, et al. Risperidone in the treatment of disruptive behavioral symptoms in children with autistic and other pervasive developmental disorders. Pediatrics. 2004;114:E634–E641.
    1. McDougle CJ, Stigler KA, Posey DJ. Pharmacological treatment of behavioral symptoms in autism. Neuropsychopharmacology. 2005;30:S72.
    1. Pandina GJ, Aman MG, Findling RL. Risperidone in the management of disruptive behavior disorders. J Child Adolesc Psychopharmacol. 2006;16:379–392.
    1. Robb AS, Andersson C, Bellocchio EE, Manos G, Rojas-Fernandez C, Mathew S, et al. Safety and tolerability of aripiprazole in the treatment of irritability associated with autistic disorder in pediatric subjects (6-17 years old):results from a pooled analysis of 2 studies. Prim Care Companion CNS Disord. 2011;13 (1
    1. Benton TD. Aripiprazole to treat irritability associated with autism: a placebo-controlled, fixed-dose trial. Curr Psychiatry Rep. 2011;13:77–79.
    1. Marcus RN, Owen R, Manos G, Mankoski R, Kamen L, McQuade RD, et al. Safety and tolerability of aripiprazole for irritability in pediatric patients with autistic disorder: a 52-week, open-label, multicenter study. J Clin Psychiatry. 2011;72:1270–1276.
    1. Eaves RC, Milner B. The criterion-related validity of the childhood autism rating-scale and the autism behavior checklist. J Abnorm Child Psychol. 1993;21:481–491.
    1. Ventola PE, Kleinman J, Pandey J, Barton M, Allen S, Green J, et al. Agreement among four diagnostic instruments for autism spectrum disorders in toddlers. J Autism Dev Disord. 2006;36:839–847.
    1. Perry A, Condillac RA, Freeman NL, Dunn-Geier J, Belair J. Multi-site study of the childhood autism rating scale (CARS) in five clinical groups of young children. J Autism Dev Disord. 2005;35:625–634.
    1. Chen WG, Chang Q, Lin YX, Meissner A, West AE, Griffith EC, et al. Derepression of BDNF transcription involves calcium-dependent phosphorylation of MeCP2. Science. 2003;302:885–889.
    1. Rivera C, Li H, Thomas-Crusells J, Lahtinen H, Viitanen T, Nanobashvili A, et al. BDNF-induced TrkB activation down-regulates the K+-Cl- cotransporter KCC2 and impairs neuronal Cl- extrusion. J Cell Biol. 2002;159:747–752.
    1. Ludwig A, Uvarov P, Soni S, Thomas-Crusells J, Airaksinen MS, Rivera C. Early growth response 4 mediates bdnf induction of potassium chloride cotransporter 2 transcription. J Neurosci. 2011;31:644–649.
    1. Roullet FI, Wollaston L, deCatanzaro D, Foster JA. Behavioral and molecular changes in the mouse in response to prenatal exposure to the anti-epileptic drug valproic acid. Neuroscience. 2010;170:514–522.
    1. Martinowich K, Hattori D, Wu H, Fouse S, He F, Hu Y, et al. DNA methylation-related chromatin remodeling in activity-dependent Bdnf gene regulation. Science. 2003;302:890–893.
    1. Chahrour M, Jung SY, Shaw C, Zhou XB, Wong STC, Qin J, et al. MeCP2, a key contributor to neurological disease, activates and represses transcription. Science. 2008;320:1224–1229.
    1. Deeb TZ, Lee HHC, Walker JA, Davies PA, Moss SJ. Hyperpolarizing GABA ergic transmission depends on KCC2 function and membrane potential. Channels. 2011;5:475–481.
    1. Sipila ST, Schuchmann S, Voipio J, Yamada J, Kaila K. The cation-chloride cotransporter NKCC1 promotes sharp waves in the neonatal rat hippocampus. J Physiol. 2006;573:765–773.
    1. Li YJ, Cleary R, Kellogg M, Soul JS, Berry GT, Jensen FE. Sensitive isotope dilution liquid chromatography/tandem mass spectrometry method for quantitative analysis of bumetanide in serum and brain tissue. J Chromatogr B. 2011;879:998–1002.

Source: PubMed

3
Sottoscrivi