Low-dose suramin in autism spectrum disorder: a small, phase I/II, randomized clinical trial

Robert K Naviaux, Brooke Curtis, Kefeng Li, Jane C Naviaux, A Taylor Bright, Gail E Reiner, Marissa Westerfield, Suzanne Goh, William A Alaynick, Lin Wang, Edmund V Capparelli, Cynthia Adams, Ji Sun, Sonia Jain, Feng He, Deyna A Arellano, Lisa E Mash, Leanne Chukoskie, Alan Lincoln, Jeanne Townsend, Robert K Naviaux, Brooke Curtis, Kefeng Li, Jane C Naviaux, A Taylor Bright, Gail E Reiner, Marissa Westerfield, Suzanne Goh, William A Alaynick, Lin Wang, Edmund V Capparelli, Cynthia Adams, Ji Sun, Sonia Jain, Feng He, Deyna A Arellano, Lisa E Mash, Leanne Chukoskie, Alan Lincoln, Jeanne Townsend

Abstract

Objective: No drug is yet approved to treat the core symptoms of autism spectrum disorder (ASD). Low-dose suramin was effective in the maternal immune activation and Fragile X mouse models of ASD. The Suramin Autism Treatment-1 (SAT-1) trial was a double-blind, placebo-controlled, translational pilot study to examine the safety and activity of low-dose suramin in children with ASD.

Methods: Ten male subjects with ASD, ages 5-14 years, were matched by age, IQ, and autism severity into five pairs, then randomized to receive a single, intravenous infusion of suramin (20 mg/kg) or saline. The primary outcomes were ADOS-2 comparison scores and Expressive One-Word Picture Vocabulary Test (EOWPVT). Secondary outcomes were the aberrant behavior checklist, autism treatment evaluation checklist, repetitive behavior questionnaire, and clinical global impression questionnaire.

Results: Blood levels of suramin were 12 ± 1.5 μmol/L (mean ± SD) at 2 days and 1.5 ± 0.5 μmol/L after 6 weeks. The terminal half-life was 14.7 ± 0.7 days. A self-limited, asymptomatic rash was seen, but there were no serious adverse events. ADOS-2 comparison scores improved by -1.6 ± 0.55 points (n = 5; 95% CI = -2.3 to -0.9; Cohen's d = 2.9; P = 0.0028) in the suramin group and did not change in the placebo group. EOWPVT scores did not change. Secondary outcomes also showed improvements in language, social interaction, and decreased restricted or repetitive behaviors.

Interpretation: The safety and activity of low-dose suramin showed promise as a novel approach to treatment of ASD in this small study.

Figures

Figure 1
Figure 1
Trial profile.
Figure 2
Figure 2
Safety monitoring. (A) Free cortisol, (B) proteinuria, (C) creatinine, (D) hemoglobin, (E) white blood cells (WBC), (F) platelets, (G) aspartate aminotransferase (AST), (H) rash – antecubital fossa, (I) chest. Data were analyzed by two‐way ANOVA to test for treatment, time, and treatment × time interaction effects. P and F values reflect the treatment effect. Only the rash was significantly different between suramin and placebo groups.
Figure 3
Figure 3
Pharmacokinetics of single‐dose suramin in children with autism spectrum disorders. (A) Two‐compartment model of suramin blood concentrations. The first 48 h were dominated by the distribution phase. Over 90% of the model is described by the elimination phase. (B) Plasma suramin concentrations. (C) A two‐compartment model correlated well with measured values. (D) Pediatric PK parameters of suramin.
Figure 4
Figure 4
Suramin pharmacometabolomics. Rank order of metabolites and pathways that were changed by suramin at 6 weeks after treatment.
Figure 5
Figure 5
Shared biochemical pathways. 75% of the pathways that were altered by suramin in children with ASD were also altered in the mouse models. Asterisks (*) indicate pathways that were changed at 2 days, but not at 6 weeks after treatment.

References

    1. Zablotsky B, Black LI, Maenner MJ, et al. Estimated prevalence of autism and other developmental disabilities following questionnaire changes in the 2014 National Health interview survey. Natl Health Stat Report 2015;13:1–20.
    1. Christensen DL, Baio J, Van Naarden Braun K, et al. Prevalence and Characteristics of Autism Spectrum Disorder Among Children Aged 8 Years–Autism and Developmental Disabilities Monitoring Network, 11 Sites, United States, 2012. MMWR Surveill Summ 2016;65:1–23.
    1. Pinto D, Pagnamenta AT, Klei L, et al. Functional impact of global rare copy number variation in autism spectrum disorders. Nature 2010;466:368–372.
    1. Richards C, Jones C, Groves L, et al. Prevalence of autism spectrum disorder phenomenology in genetic disorders: a systematic review and meta‐analysis. Lancet Psychiatry 2015;2:909–916.
    1. Talkowski ME, Minikel EV, Gusella JF. Autism spectrum disorder genetics: diverse genes with diverse clinical outcomes. Harv Rev Psychiatry 2014;22:65–75.
    1. Kalkbrenner AE, Schmidt RJ, Penlesky AC. Environmental chemical exposures and autism spectrum disorders: a review of the epidemiological evidence. Curr Probl Pediatr Adolesc Health Care 2014;44:277–318.
    1. Zerbo O, Iosif AM, Walker C, et al. Is Maternal Influenza or Fever During Pregnancy Associated with Autism or Developmental Delays? Results from the CHARGE (CHildhood Autism Risks from Genetics and Environment) Study. J Autism Dev Disord 2013;43(1):25–33.
    1. Naviaux RK. Metabolic features of the cell danger response. Mitochondrion 2014;16:7–17.
    1. Silva JM, Wong A, Carelli V, Cortopassi GA. Inhibition of mitochondrial function induces an integrated stress response in oligodendroglia. Neurobiol Dis 2009;34:357–365.
    1. Nikkanen J, Forsstrom S, Euro L, et al. Mitochondrial DNA Replication Defects Disturb Cellular dNTP Pools and Remodel One‐Carbon Metabolism. Cell Metab 2016;23:635–648.
    1. Green DR, Galluzzi L, Kroemer G. Mitochondria and the autophagy‐inflammation‐cell death axis in organismal aging. Science 2011;333:1109–1112.
    1. Naviaux JC, Wang L, Li K, et al. Antipurinergic therapy corrects the autism‐like features in the Fragile X (Fmr1 knockout) mouse model. Mol Autism 2015;6:1.
    1. Naviaux JC, Schuchbauer MA, Li K, et al. Reversal of autism‐like behaviors and metabolism in adult mice with single‐dose antipurinergic therapy. Transl Psychiat 2014;4:e400.
    1. Naviaux RK, Zolkipli‐Cunningham Z, Nakayama T, et al. Antipurinergic Therapy Corrects the Autism‐Like Features in the Poly(IC) Mouse Model. PLoS ONE 2013;8(3):e57380.
    1. Wikoff WR, Kalisak E, Trauger S, et al. Response and recovery in the plasma metabolome tracks the acute LCMV‐induced immune response. J Proteome Res 2009;8:3578–3587.
    1. Degtyar E, Zusman T, Ehrlich M, Segal G. A Legionella effector acquired from protozoa is involved in sphingolipids metabolism and is targeted to the host cell mitochondria. Cell Microbiol 2009;11:1219–1235.
    1. James SJ, Cutler P, Melnyk S, et al. Metabolic biomarkers of increased oxidative stress and impaired methylation capacity in children with autism. Am J Clin Nutr 2004;80:1611–1617.
    1. West AP, Shadel GS, Ghosh S. Mitochondria in innate immune responses. Nat Rev Immunol 2011;11:389–402.
    1. Naviaux RK. Oxidative shielding or oxidative stress? J Pharmacol Exp Ther 2012;342:608–618.
    1. Lohman AW, Isakson BE. Differentiating connexin hemichannels and pannexin channels in cellular ATP release. FEBS Lett 2014;588:1379–1388.
    1. Trautmann A. Extracellular ATP in the immune system: more than just a “danger signal”. Sci Signal 2009;2:pe6.
    1. Riteau N, Baron L, Villeret B, et al. ATP release and purinergic signaling: a common pathway for particle‐mediated inflammasome activation. Cell Death Dis 2012;3:e403.
    1. Burnstock G. The Paton Lecture: Purinergic signalling: from discovery to current developments. Exp Physiol 2014;99(1):16–34.
    1. Imamura H, Nhat KP, Togawa H, et al. Visualization of ATP levels inside single living cells with fluorescence resonance energy transfer‐based genetically encoded indicators. Proc Natl Acad Sci USA 2009;106:15651–15656.
    1. Jacobson KA, Balasubramanian R, Deflorian F, Gao ZG. G protein‐coupled adenosine (P1) and P2Y receptors: ligand design and receptor interactions. Purinergic Signalling 2012;8:419–436.
    1. Adams JB, Audhya T, McDonough‐Means S, et al. Nutritional and metabolic status of children with autism vs. neurotypical children, and the association with autism severity. Nutr Metab 2011;8:34.
    1. Jia M, Li MX, Fields RD, Nelson PG. Extracellular ATP in activity‐dependent remodeling of the neuromuscular junction. Dev Neurobiol 2007;67:924–932.
    1. Naviaux RK. Mitochondria and Autism In: Buxbaum JD, Hof PR, eds. The Neuroscience of Autism Spectrum Disorders. Waltham, MA: Academic Press, Elsevier, 2012:179–193.
    1. Naviaux RK. Mitochondrial control of epigenetics. Cancer Biol Ther 2008;7:1191–1193.
    1. Wallace DC, Fan W. Energetics, epigenetics, mitochondrial genetics. Mitochondrion 2010;10:12–31.
    1. Burnstock G. Pathophysiology and therapeutic potential of purinergic signaling. Pharmacol Rev 2006;58:58–86.
    1. Stein CA. Suramin: a novel antineoplastic agent with multiple potential mechanisms of action. Can Res 1993;53(10 Suppl):2239–2248.
    1. World Medical A . World Medical Association Declaration of Helsinki: ethical principles for medical research involving human subjects. JAMA 2013;310:2191–2194.
    1. Schulz KF, Altman DG, Moher D, Group C . CONSORT 2010 Statement: updated guidelines for reporting parallel group randomised trials. Trials 2010;11:32.
    1. Gotham K, Risi S, Pickles A, Lord C. The Autism Diagnostic Observation Schedule: revised algorithms for improved diagnostic validity. J Autism Dev Disord 2007;37:613–627.
    1. Grondhuis SN, Mulick JA. Comparison of the Leiter International Performance Scale‐Revised and the Stanford‐Binet Intelligence Scales, 5th Edition, in children with autism spectrum disorders. Am J Intellect Dev Disabil 2013;118:44–54.
    1. Adams‐Chapman I, Bann C, Carter SL, et al. Language outcomes among ELBW infants in early childhood. Early Hum Dev 2015;91:373–379.
    1. Kaat AJ, Lecavalier L, Aman MG. Validity of the aberrant behavior checklist in children with autism spectrum disorder. J Autism Dev Disord 2014;44:1103–1116.
    1. Geier DA, Kern JK, Geier MR. A Comparison of the Autism Treatment Evaluation Checklist (ATEC) and the Childhood Autism Rating Scale (CARS) for the quantitative evaluation of autism. J Mental Health Res Intellect Disabil 2013;6:255–267.
    1. Rimland B, Edelson S. Autism treatment evaluation checklist. Autism Research Institute 2000. [cited 2016 June 15].
    1. Busner J, Targum SD. The clinical global impressions scale: applying a research tool in clinical practice. Psychiatry (Edgmont). 2007;4:28–37.
    1. Honey E, McConachie H, Turner M, Rodgers J. Validation of the repetitive behaviour questionnaire for use with children with autism spectrum disorder. Res Autism Spectr Disord 2012;6:355–364.
    1. Sheiner LB, Beal SL. Evaluation of methods for estimating population pharmacokinetic parameters. II. Biexponential model and experimental pharmacokinetic data. J Pharmacokinet Biopharm 1981;9:635–651.
    1. Naviaux RK, Naviaux JC, Li K, et al. Metabolic features of chronic fatigue syndrome. Proc Natl Acad Sci USA 2016;113(37):E5472–E5480.
    1. Xia J, Sinelnikov IV, Han B, Wishart DS. MetaboAnalyst 3.0‐making metabolomics more meaningful. Nucleic Acids Res 2015;43(W1):W251–W257.
    1. Hawking F. Suramin: with special reference to onchocerciasis. Advances Pharmacol Chemother 1978;15:289–322.
    1. Pickles A, Le Couteur A, Leadbitter K, et al. Parent‐mediated social communication therapy for young children with autism (PACT): long‐term follow‐up of a randomised controlled trial. Lancet 2016;388:2501–2509.
    1. Hawking F. Concentration of Bayer 205 (Germanin) in human blood and cerebrospinal fluid after treatment. Trans R Soc Trop Med Hyg 1940;34:37–52.
    1. Siso S, Jeffrey M, Gonzalez L. Sensory circumventricular organs in health and disease. Acta Neuropathol 2010;120:689–705.

Source: PubMed

3
Abonneren