Autism spectrum disorder (ASD) is a complex neurodevelopmental disorder with an unknown etiology and currently few effective therapies. Immune system alterations have being demonstrated in ASD, both in humans and via animal models; immune imbalance thus arises as a possible pathway for drug intervention. In this review, the studies were classified into 2 major groups: (1) clinical research whose authors classify therapies with primary anti-inflammatory and immunomodulatory actions, making use of: sulforaphane, celecoxib, lenalidomide, pentoxifylline, spironolactone, flavonoid luteolin, corticosteroids, oral immunoglobulin, intravenous immunoglobulin, cell therapy, dialyzable lymphocyte extracts, minocycline, and pioglitazone; and (2) other ASD therapies already used or currently under study whose initial characteristics were neither anti-inflammatory nor immunomodulatory initially, but displayed a capacity for immunomodulation throughout the treatment: risperidone, vitamin D, omega-3, Ginkgo biloba, L-carnosine, N-acetylcysteine, and microbiome restoration. These studies used various data acquisition methodologies. Questions arose such the need for randomized and placebo-controlled studies with greater numbers of participants as well as the use of biomarkers to refine the treatment of autistic subjects.

1.
American Psychiatric Association: Diagnostic and Statistical Manual of Mental Disorders, ed 5 (DSM-5TM). Arlington, American Psychiatric Publishing, 2013.
2.
Gottfried C, Bambini-Junior V, Francis F, Riesgo R, Savino W: The impact of neuroimmune alterations in autism spectrum disorder. Front Psychiatry 2015; 6: 121.
3.
Noriega DB, Savelkoul HFJ: Immune dysregulation in autism spectrum disorder. Eur J Pediatr 2014; 173: 33–43.
4.
Baio J, Wiggins L, Christensen DL, Maenner MJ, Daniels J, Warren Z, et al: Prevalence of autism spectrum disorder among children aged 8 years – Autism and Developmental Disabilities Monitoring Network, 11 Sites, United States, 2014. MMWR Surveill Summ 2018; 67: 1–23.
5.
Brentani H, Paula CS, Bordini D, Rolim D, Sato F, Portolese J, et al: Autism spectrum disorders: an overview on diagnosis and treatment. Rev Bras Psiquiatr 2013; 35:S62–S72.
6.
Baxter AJ, Brugha TS, Erskine HE, Scheurer RW, Vos T, Scott JG: The epidemiology and global burden of autism spectrum disorders. Psychol Med 2015; 45: 601–613.
7.
Benvenuto A, Battan B, Porfirio MC, Curatolo P: Pharmacotherapy of autism spectrum disorders. Brain Dev 2013; 35: 119–127.
8.
Mead J, Ashwood P: Evidence supporting an altered immune response in ASD. Immunol Lett 2015; 163: 49–55.
9.
Rossignol DA: Novel and emerging treatments for autism spectrum disorders: a systematic review. Ann Clin Psychiatry 2009; 21: 213–236.
10.
Kaplan G, McCracken JT: Psychopharma-cology of autism spectrum disorders. Pediatr Clin North Am 2012; 59: 175–187.
11.
Marcus RN, Owen R, Kamen L, Manos G, McQuade RD, Carson WH, et al: A placebo-controlled, fixed-dose study of aripiprazole in children and adolescents with irritability associated with autistic disorder. J Am Acad Child Adolesc Psychiatry 2009; 48: 1110–1119.
12.
Owen R, Sikich L, Marcus RN, Corey-Lisle P, Manos G, McQuade RD, et al: Aripiprazole in the treatment of irritability in children and adolescents with autistic disorder. Pediatrics 2009; 124: 1533–1540.
13.
Research Units on Pediatric Psychopharmacology Autism Network: Randomized, controlled, crossover trial of methylphenidate in pervasive developmental disorders with hyperactivity. Arch Gen Psychiatry 2005; 62: 1266–1274.
14.
LeClerc S, Easley D: Pharmacological therapies for autism spectrum disorder: a review. P T. 2015; 40: 389–397.
15.
Masi A, Glozier N, Dale R, Guastella AJ: The immune system, cytokines, and biomarkers in autism spectrum disorder. Neurosci Bull 2017; 33: 194–204.
16.
Buescher AVS, Cidav Z, Knapp M, Mandell DS: Costs of autism spectrum disorders in the United Kingdom and the United States. JAMA Pediatr 2014; 168: 721.
17.
Karst JS, Van Hecke AV: Parent and family impact of autism spectrum disorders: a review and proposed model for intervention evaluation. Clin Child Fam Psychol Rev 2012; 15: 247–277.
18.
Smith LE, Greenberg JS, Mailick MR: The family context of autism spectrum disorders: influence on the behavioral phenotype and quality of life. Child Adolesc Psychiatr Clin N Am 2014; 23: 143–155.
19.
Weiss JA, Cappadocia MC, MacMullin JA, Viecili M, Lunsky Y: The impact of child problem behaviors of children with ASD on parent mental health: the mediating role of acceptance and empowerment. Autism Int J Res Pract 2012; 16: 261–274.
20.
Marchezan J, Becker M, Schwartsmann G, Ohlweiler L, Roesler R, Renck LB, et al: A placebo-controlled crossover trial of gastrin-releasing peptide in childhood autism. Clin Neuropharmacol 2017; 40: 108–112.
21.
Verkhratsky A, Rodríguez JJ, Parpura V: Neuroglia in ageing and disease. Cell Tissue Res 2014; 357: 493–503.
22.
Careaga M, Van de Water J, Ashwood P: Immune dysfunction in autism: a pathway to treatment. Neurotherapeutics 2010; 7: 283–292.
23.
McDougle CJ, Landino SM, Vahabzadeh A, O’Rourke J, Zurcher NR, Finger BC, et al: Toward an immune-mediated subtype of autism spectrum disorder. Brain Res 2015; 1617: 72–92.
24.
Chez MG, Guido-Estrada N: Immune therapy in autism: historical experience and future directions with immunomodulatory therapy. Neurotherapeutics 2010; 7: 293–301.
25.
Masi A, Quintana DS, Glozier N, Lloyd AR, Hickie IB, Guastella AJ: Cytokine aberrations in autism spectrum disorder: a systematic review and meta-analysis. Mol Psychiatry 2015; 20: 440–446.
26.
Gładysz D, Krzywdzińska A, Hozyasz KK: Immune abnormalities in autism spectrum disorder – could they hold promise for causative treatment? Mol Neurobiol 2018; 55: 6387–6435.
27.
Stubbs EG: Autistic children exhibit undetectable hemagglutination-inhibition antibody titers despite previous Rubella vaccination. J Autism Child Schizophr 1976; 6: 269–274.
28.
Yang C-J, Liu C-L, Sang B, Zhu X-M, Du Y-J: The combined role of serotonin and interleukin-6 as biomarker for autism. Neuroscience 2015; 284: 290–296.
29.
Ashwood P, Krakowiak P, Hertz-Picciotto I, Hansen R, Pessah I, Van de Water J: Elevated plasma cytokines in autism spectrum disorders provide evidence of immune dysfunction and are associated with impaired behavioral outcome. Brain Behav Immun 2011; 25: 40–45.
30.
Jyonouchi H, Geng L, Ruby A, Zimmerman-Bier B: Dysregulated innate immune responses in young children with autism spectrum disorders: their relationship to gastrointestinal symptoms and dietary intervention. Neuropsychobiology 2005; 51: 77–85.
31.
Jyonouchi H, Geng L, Davidow AL: Cytokine profiles by peripheral blood monocytes are associated with changes in behavioral symptoms following immune insults in a subset of ASD subjects: an inflammatory subtype? J Neuroinflammation 2014; 11: 187.
32.
El-Ansary A, Hassan WM, Qasem H, Das UN: Identification of biomarkers of impaired sensory profiles among autistic patients. PLoS One 2016; 11:e0164153.
33.
Ferguson BJ, Marler S, Altstein LL, Lee EB, Mazurek MO, McLaughlin A, et al: Associations between cytokines, endocrine stress response, and gastrointestinal symptoms in autism spectrum disorder. Brain Behav Immun 2016; 58: 57–62.
34.
Careaga M, Rogers S, Hansen RL, Amaral DG, Van de Water J, Ashwood P: Immune endophenotypes in children with autism spectrum disorder. Biol Psychiatry 2017; 81: 434–441.
35.
Al-Ayadhi LY: Pro-inflammatory cytokines in autistic children in central Saudi Arabia. Neurosciences (Riyadh) 2005; 10: 155–158.
36.
Ricci S, Businaro R, Ippoliti F, Lo Vasco VR, Massoni F, Onofri E, et al: Altered cytokine and BDNF levels in autism spectrum disorder. Neurotox Res 2013; 24: 491–501.
37.
Makinodan M, Iwata K, Ikawa D, Yamashita Y, Yamamuro K, Toritsuka M, et al: Tumor necrosis factor-α expression in peripheral blood mononuclear cells correlates with early childhood social interaction in autism spectrum disorder. Neurochem Int 2017; 104: 1–5.
38.
Han YMY, Cheung WKY, Wong CK, Sze SL, Cheng TWS, Yeung MK, et al: Distinct cytokine and chemokine profiles in autism spectrum disorders. Front Immunol 2017; 8: 11.
39.
Ashwood P, Krakowiak P, Hertz-Picciotto I, Hansen R, Pessah IN, Van de Water J: Associations of impaired behaviors with elevated plasma chemokines in autism spectrum disorders. J Neuroimmunol 2011; 232: 196–199.
40.
Ashwood P, Enstrom A, Krakowiak P, Hertz-Picciotto I, Hansen RL, Croen LA, et al: Decreased transforming growth factor β1 in autism: a potential link between immune dysregulation and impairment in clinical behavioral outcomes. J Neuroimmunol 2008; 204: 149–153.
41.
El Gohary TM, El Aziz NA, Darweesh M, Sadaa ES: Plasma level of transforming growth factor β1 in children with autism spectrum disorder. Egypt J Ear Nose Throat Allied Sci 2015; 16: 69–73.
42.
AL-Ayadhi LY, Mostafa GA: Elevated serum levels of macrophage-derived chemokine and thymus and activation-regulated chemokine in autistic children. J Neuroinflamm 2013; 10: 846.
43.
Al-Ayadhi LY, Mostafa GA: Elevated serum levels of interleukin-17A in children with autism. J Neuroinflamm 2012; 9: 158.
44.
Enstrom A, Onore C, Hertz-Picciotto I, Hansen R, Croen L, Van de Water J, et al: Detection of IL-17 and IL-23 in plasma samples of children with autism. Am J Biochem Biotechnol 2008; 4: 114–120.
45.
Rose D, Van de Water J, Ashwood P: Abstract # 1787 Unique immune profiles in children with autism who experience gastrointestinal co-morbidity. Brain Behav Immun 2016; 57:e23–e24.
46.
Napolioni V, Ober-Reynolds B, Szelinger S, Corneveaux JJ, Pawlowski T, Ober-Reynolds S, et al: Plasma cytokine profiling in sibling pairs discordant for autism spectrum disorder. J Neuroinflamm 2013; 10: 38.
47.
Heuer L, Ashwood P, Schauer J, Goines P, Krakowiak P, Hertz-Picciotto I, et al: Reduced levels of immunoglobulin in children with autism correlates with behavioral symptoms. Autism Res Off J Int Soc Autism Res 2008; 1: 275–283.
48.
Hollander E, DelGiudice-Asch G, Simon L, Schmeidler J, Cartwright C, DeCaria CM, et al: B lymphocyte antigen D8/17 and repetitive behaviors in autism. Am J Psychiatry 1999; 156: 317–320.
49.
Mostafa GA, Al-Ayadhi LY: The relationship between the increased frequency of serum antineuronal antibodies and the severity of autism in children. Eur J Paediatr Neurol 2012; 16: 464–468.
50.
Goines P, Haapanen L, Boyce R, Duncanson P, Braunschweig D, Delwiche L, et al: Autoantibodies to cerebellum in children with autism associate with behavior. Brain Behav Immun 2011; 25: 514–523.
51.
Piras IS, Haapanen L, Napolioni V, Sacco R, Van de Water J, Persico AM: Anti-brain antibodies are associated with more severe cognitive and behavioral profiles in Italian children with autism spectrum disorder. Brain Behav Immun 2014; 38: 91–99.
52.
Mostafa GA, Al-Ayadhi LY: Increased serum levels of anti-ganglioside M1 auto-antibodies in autistic children: relation to the disease severity. J Neuroinflammation 2011; 8: 39.
53.
Braunschweig D, Duncanson P, Boyce R, Hansen R, Ashwood P, Pessah IN, et al: Behavioral correlates of maternal antibody status among children with autism. J Autism Dev Disord 2012; 42: 1435–1445.
54.
Lacivita E, Perrone R, Margari L, Leopoldo M: Targets for drug therapy for autism spectrum disorder: challenges and future directions. J Med Chem 2017; 60: 9114–9141.
55.
Estes ML, McAllister AK: Immune mediators in the brain and peripheral tissues in autism spectrum disorder. Nat Rev Neurosci 2015; 16: 469–486.
56.
Meltzer A, Van de Water J: The role of the immune system in autism spectrum dis order. Neuropsychopharmacology 2017; 42: 284–298.
57.
Tarozzi A, Angeloni C, Malaguti M, Morroni F, Hrelia S, Hrelia P: Sulforaphane as a potential protective phytochemical against neurodegenerative diseases. Oxid Med Cell Longev 2013; 2013: 415078.
58.
Juge N, Mithen RF, Traka M: Molecular basis for chemoprevention by sulforaphane: a comprehensive review. Cell Mol Life Sci 2007; 64: 1105–1127.
59.
Singh K, Connors SL, Macklin EA, Smith KD, Fahey JW, Talalay P, et al: Sulforaphane treatment of autism spectrum disorder (ASD). Proc Natl Acad Sci USA 2014; 111: 15550–15555.
60.
Lynch R, Diggins EL, Connors SL, Zimmerman AW, Singh K, Liu H, et al: Sulforaphane from broccoli reduces symptoms of autism: a follow-up case series from a randomized double-blind study. Glob Adv Heal Med 2017; 6: 2164957X17735826.
61.
Sareddy GR, Geeviman K, Ramulu C, Babu PP: The nonsteroidal anti-inflammatory drug celecoxib suppresses the growth and induces apoptosis of human glioblastoma cells via the NF-κB pathway. J Neurooncol 2012; 106: 99–109.
62.
Arias-Negrete S, Keller K, Chadee K: Proinflammatory cytokines regulate cyclooxygenase-2 mRNA expression in human macrophages. Biochem Biophys Res Commun 1995; 208: 582–589.
63.
Asadabadi M, Mohammadi M-R, Ghanizadeh A, Modabbernia A, Ashrafi M, Hassanzadeh E, et al: Celecoxib as adjunctive treatment to risperidone in children with autistic disorder: a randomized, double-blind, placebo-controlled trial. Psychopharmacology 2013; 225: 51–59.
64.
Pulte ED, Dmytrijuk A, Nie L, Goldberg KB, McKee AE, Farrell AT, et al: FDA approval summary: lenalidomide as maintenance therapy after autologous stem cell transplant in newly diagnosed multiple myeloma. Oncologist 2018; 23: 734–739.
65.
Chez M, Low R, Parise C, Donnel T: Safety and observations in a pilot study of lenalidomide for treatment in autism. Autism Res Treat 2012; 2012: 291601.
66.
Kreth S, Ledderose C, Luchting B, Weis F, Thiel M: Immunomodulatory properties of pentoxifylline are mediated via adenosine-dependent pathways. Shock 2010; 34: 10–16.
67.
Dolatabadi HRD, Zarrindast MR, Reisi P, Nasehi M: The effects of pentoxifylline on serum levels of interleukin 10 and interferon gamma and memory function in lipopolysaccharide-induced inflammation in rats. Adv Biomed Res 2017; 6: 110.
68.
Gupta S, Rimland B, Shilling PD: Pentoxifylline: brief review and rationale for its possible use in the treatment of autism. J Child Neurol 1996; 11: 501–504.
69.
Gupta S: Immunological treatments for autism. J Autism Dev Disord 2000; 30: 475–479.
70.
Sogame S: Clinical experiences with the administration of pentoxifylline against autism and behavior abnormalities. Jpn J Child Psych 1978; 137–144.
71.
Nakane A: Effect of pentoxifylline in autistic children. Tokyoto Eisei Gakkashi 1980; 104–105.
72.
Shimoide M: Effect of pentoxifylline (Trental) on infantile autism. Clin Exp Med 1981; 285–288.
73.
Turek S: Treatment of psychotic and autistic children with pentoxifylline. ASANA 1981; 51–60.
74.
Suzuki M, Nakashita Y, Ogawa T: Sugiyama A, et al: On the effects of pentoxifylline upon the EEG activity of early infantile autism. Brain Dev 1984; 6: 156.
75.
Akhondzadeh S, Fallah J, Mohammadi M-R, Imani R, Mohammadi M, Salehi B, et al: Double-blind placebo-controlled trial of pentoxifylline added to risperidone: effects on aberrant behavior in children with autism. Prog Neuropsychopharmacol Biol Psychiatry 2010; 34: 32–36.
76.
Pérez JCJ, Ramírez AC, González LT, Espinosa LEM, Quintana MMP, Galván GA, et al: Spironolactone effect in hepatic ischemia/reperfusion injury in Wistar rats. Oxid Med Cell Longev 2016; 2016: 3196431.
77.
Miura R, Nakamura K, Miura D, Miura A, Hisamatsu K, Kajiya M, et al: Anti-inflammatory effect of spironolactone on human peripheral blood mononuclear cells. J Pharmacol Sci 2006; 101: 256–259.
78.
Miura R, Nakamura K, Miura D, Miura A, Hisamatsu K, Kajiya M, et al: Aldosterone synthesis and cytokine production in human peripheral blood mononuclear cells. J Pharmacol Sci 2006; 102: 288–295.
79.
Bendtzen K, Hansen PR, Rieneck K: Spironolactone inhibits production of proinflammatory cytokines, including tumour necrosis factor-alpha and interferon-gamma, and has potential in the treatment of arthritis. Clin Exp Immunol 2003; 134: 151–158.
80.
Kato Y, Kamiya H, Koide N, Odkhuu E, Ko-matsu T, Dagvadorj J, et al: Spironolactone inhibits production of proinflammatory mediators in response to lipopolysaccharide via inactivation of nuclear factor-κB. Immunopharmacol Immunotoxicol 2014; 36: 237–241.
81.
Bradstreet JJ, Smith S, Granpeesheh D, El-Dahr JM, Rossignol D: Spironolactone might be a desirable immunologic and hormonal intervention in autism spectrum disorders. Med Hypotheses 2007; 68: 979–987.
82.
Middleton E, Kandaswami C, Theoharides TC: The effects of plant flavonoids on mammalian cells: implications for inflammation, heart disease, and cancer. Pharmacol Rev 2000; 52: 673–751.
83.
López-Lázaro M: Distribution and biological activities of the flavonoid luteolin. Mini Rev Med Chem 2009; 9: 31–59.
84.
Kempuraj D, Madhappan B, Christodoulou S, Boucher W, Cao J, Papadopoulou N, et al: Flavonols inhibit proinflammatory mediator release, intracellular calcium ion levels and protein kinase C theta phosphorylation in human mast cells. Br J Pharmacol 2005; 145: 934–944.
85.
Theoharides TC, Asadi S, Panagiotidou S: A case series of a luteolin formulation (NeuroProtek®) in children with autism spectrum disorders. Int J Immunopathol Pharmacol 2012; 25: 317–323.
86.
Taliou A, Zintzaras E, Lykouras L, Francis K: An open-label pilot study of a formulation containing the anti-inflammatory flavonoid luteolin and its effects on behavior in children with autism spectrum disorders. Clin Ther 2013; 35: 592–602.
87.
Tsilioni I, Taliou A, Francis K, Theoharides TC: Children with autism spectrum disorders, who improved with a luteolin-containing dietary formulation, show reduced serum levels of TNF and IL-6. Transl Psychiatry 2015; 5:e647.
88.
Bertolino B, Crupi R, Impellizzeri D, Bruschetta G, Cordaro M, Siracusa R, et al: Beneficial effects of co-ultramicronized palmitoylethanolamide/luteolin in a mouse model of autism and in a case report of autism. CNS Neurosci Ther 2017; 23: 87–98.
89.
Schweingruber N, Reichardt SD, Lühder F, Reichardt HM: Mechanisms of glucocorticoids in the control of neuroinflammation. J Neuroendocrinol 2012; 24: 174–182.
90.
Duffy FH, Shankardass A, McAnulty GB, Eksioglu YZ, Coulter D, Rotenberg A, et al: Corticosteroid therapy in regressive autism: a retrospective study of effects on the frequen cy modulated auditory evoked response (FMAER), language, and behavior. BMC Neurol 2014; 14: 70.
91.
Shenoy S, Arnold S, Chatila T: Response to steroid therapy in autism secondary to autoimmune lymphoproliferative syndrome. J Pediatr 2000; 136: 682–687.
92.
Stefanatos GA, Grover W, Geller E: Case study: corticosteroid treatment of language regression in pervasive developmental disorder. J Am Acad Child Adolesc Psychiatry 1995; 34: 1107–1111.
93.
Mordekar SR, Prendergast M, Chattopadhyay AK, Baxter PS: Corticosteroid treatment of behaviour, language and motor regression in childhood disintegrative disorder. Eur J Paediatr Neurol 2009; 13: 367–369.
94.
Buitelaar JK, Dekker ME, van Ree JM, van Engeland H: A controlled trial with ORG 2766, an ACTH-(4–9) analog, in 50 relatively able children with autism. Eur Neuropsychopharmacol 1996; 6: 13–19.
95.
Buitelaar JK, van Engeland H, de Kogel K, de Vries H, van Hooff J, van Ree J: The adrenocorticotrophic hormone (4–9) analog ORG 2766 benefits autistic children: report on a second controlled clinical trial. J Am Acad Child Adolesc Psychiatry 1992; 31: 1149–1156.
96.
Buitelaar JK, van Engeland H, de Kogel KH, de Vries H, van Hooff JA, van Ree JM: The use of adrenocorticotrophic hormone (4–9) analog ORG 2766 in autistic children: effects on the organization of behavior. Biol Psychiatry 1992; 31: 1119–1129.
97.
Buitelaar JK, van Engeland H, van Ree JM, de Wied D: Behavioral effects of Org 2766, a synthetic analog of the adrenocorticotrophic hormone (4–9), in 14 outpatient autistic children. J Autism Dev Disord 1990; 20: 467–478.
98.
Verbaten MN, Kemner C, Buitelaar JK, van Ree JM, van Beijsterveld CE, van Engeland H: Effects of ORG-2766 on brain event-related potentials of autistic children. Psychiatry Res 1996; 63: 33–45.
99.
Fung LK, Libove RA, Phillips J, Haddad F, Hardan AY: Brief report: an open-label study of the neurosteroid pregnenolone in adults with autism spectrum disorder. J Autism Dev Disord 2014; 44: 2971–2977.
100.
Marx CE, Keefe RSE, Buchanan RW, Hamer RM, Kilts JD, Bradford DW, et al: Proof-of-concept trial with the neurosteroid pregnenolone targeting cognitive and negative symptoms in schizophrenia. Neuropsychopharmacology 2009; 34: 1885–1903.
101.
Jasion VS, Burnett BP: Survival and digestibility of orally-administered immunoglobulin preparations containing IgG through the gastrointestinal tract in humans. Nutr J 2015; 14: 22.
102.
Schneider CK, Melmed RD, Barstow LE, Enriquez FJ, Ranger-Moore J, Ostrem JA: Oral human immunoglobulin for children with autism and gastrointestinal dysfunction: a prospective, open-label study. J Autism Dev Disord 2006; 36: 1053–1064.
103.
Handen BL, Melmed RD, Hansen RL, Aman MG, Burnham DL, Bruss JB, et al: A double-blind, placebo-controlled trial of oral human immunoglobulin for gastrointestinal dysfunction in children with autistic disorder. J Autism Dev Disord 2009; 39: 796–805.
104.
Rütter A, Luger TA: High-dose intravenous immunoglobulins: an approach to treat severe immune-mediated and autoimmune diseases of the skin. J Am Acad Dermatol 2001; 44: 1010–1024.
105.
Niederhofer H, Staffen W, Mair A: Immunoglobulins as an alternative strategy of psychopharmacological treatment of children with autistic disorder. Neuropsychopharmacology 2003; 28: 1014–1015.
106.
DelGiudice-Asch G, Simon L, Schmeidler J, Cunningham-Rundles C, Hollander E: Brief report: a pilot open clinical trial of intravenous immunoglobulin in childhood autism. J Autism Dev Disord 1999; 29: 157–160.
107.
Plioplys AV: Intravenous immunoglobulin treatment of children with autism. J Child Neurol 1998; 13: 79–82.
108.
Melamed IR, Heffron M, Testori A, Lipe K: A pilot study of high-dose intravenous immunoglobulin 5% for autism: impact on autism spectrum and markers of neuroinflammation. Autism Res 2018; 11: 421–433.
109.
Siniscalco D, Bradstreet JJ, Antonucci N: Therapeutic role of hematopoietic stem cells in autism spectrum disorder-related inflammation. Front Immunol 2013; 4: 140.
110.
Lv Y-T, Zhang Y, Liu M, Qiuwaxi J, Ashwood P, Cho SC, et al: Transplantation of human cord blood mononuclear cells and umbilical cord-derived mesenchymal stem cells in autism. J Transl Med 2013; 11: 196.
111.
Bradstreet JJ, Sych N, Antonucci N, Klunnik M, Ivankova O, Matyashchuk I, et al: Efficacy of fetal stem cell transplantation in autism spectrum disorders: an open-labeled pilot study. Cell Transplant 2014; 23(suppl 1):S105–S112.
112.
Sharma A, Badhe P, Gokulchandran N, Kulkarni P, Mishra P, Shetty A, et al: An improved case of autism as revealed by PET CT scan in patient transplanted with autologous bone marrow derived mononuclear cells. J Stem Cell Res Ther 2013; 3: 139.
113.
Sharma A, Gokulchandran N, Sane H, Nagrajan A, Paranjape A, Kulkarni P, et al: Autologous bone marrow mononuclear cell therapy for autism: an open label proof of concept study. Stem Cells Int 2013; 2013: 623875.
114.
Bansal H, Verma P, Agrawal A, Leon J, Sundell IB, Koka PS: A Short study report on bone marrow aspirate concentrate cell therapy in ten South Asian Indian patients with autism. J Stem Cells 2016; 11: 25–36.
115.
Dawson G, Sun JM, Davlantis KS, Murias M, Franz L, Troy J, et al: Autologous cord blood infusions are safe and feasible in young children with autism spectrum disorder: results of a single-center phase I open-label trial. Stem Cells Transl Med 2017; 6: 1332–1339.
116.
Shroff G: Human embryonic stem cells in the treatment of autism: a case series. Innov Clin Neurosci 2017; 14: 12–16.
117.
Arnaudov A, Kostova Z: Dialysable leukocyte extracts in immunotherapy. Biotechnol Biotechnol Equip 2015; 29: 1017–1023.
118.
Fudenberg HH: Dialysable lymphocyte extract (DLyE) in infantile onset autism: a pilot study. Biotherapy 1996; 9: 143–147.
119.
Soczynska JK, Mansur RB, Brietzke E, Swardfager W, Kennedy SH, Woldeyohannes HO, et al: Novel therapeutic targets in depression: minocycline as a candidate treatment. Behav Brain Res 2012; 235: 302–317.
120.
Rosenblat JD, McIntyre RS: Efficacy and tolerability of minocycline for depression: a systematic review and meta-analysis of clinical trials. J Affect Disord 2018; 227: 219–225.
121.
Kim H-S, Suh Y-H: Minocycline and neurodegenerative diseases. Behav Brain Res 2009; 196: 168–179.
122.
Orsucci D, Calsolaro V, Mancuso M, Siciliano G: Neuroprotective effects of tetracyclines: molecular targets, animal models and human disease. CNS Neurol Disord Drug Targets 2009; 8: 222–231.
123.
Yong VW, Wells J, Giuliani F, Casha S, Power C, Metz LM: The promise of minocycline in neurology. Lancet Neurol 2004; 3: 744–751.
124.
Paribello C, Tao L, Folino A, Berry-Kravis E, Tranfaglia M, Ethell IM, et al: Open-label add-on treatment trial of minocycline in fragile X syndrome. BMC Neurol 2010; 10: 91.
125.
Leigh MJS, Nguyen DV, Mu Y, Winarni TI, Schneider A, Chechi T, et al: A randomized double-blind, placebo-controlled trial of minocycline in children and adolescents with fragile X syndrome. J Dev Behav Pediatr JDBP 2013; 34: 147–155.
126.
Pardo CA, Buckley A, Thurm A, Lee L-C, Azhagiri A, Neville DM, et al: A pilot open-label trial of minocycline in patients with autism and regressive features. J Neurodev Disord 2013; 5: 9.
127.
Ghaleiha A, Alikhani R, Kazemi M-R, Mohammadi M-R, Mohammadinejad P, Zeinoddini A, et al: Minocycline as adjunctive treatment to risperidone in children with autistic disorder: a randomized, double-blind placebo-controlled trial. J Child Adolesc Psychopharmacol 2016; 26: 784–791.
128.
Derosa G, Sahebkar A, Maffioli P: The role of various peroxisome proliferator-activated receptors and their ligands in clinical practice. J Cell Physiol 2018; 233: 153–161.
129.
Yandrapalli S, Aronow WS: Cardiovascular benefits of the newer medications for treating type 2 diabetes mellitus. J Thorac Dis 2017; 9: 2124–2134.
130.
Yessoufou A, Wahli W: Multifaceted roles of peroxisome proliferator-activated receptors (PPARs) at the cellular and whole organism levels. Swiss Med Wkly 2010; 140:w13071.
131.
Rogliani P, Ora J, Di Daniele N, Lauro D: Pleiotropic effects of hypoglycemic agents: implications in asthma and COPD. Curr Opin Pharmacol 2018; 40: 34–38.
132.
Nierenberg AA, Ghaznavi SA, Sande Mathias I, Ellard KK, Janos JA, Sylvia LG: Peroxisome proliferator-activated receptor gamma coactivator-1 alpha as a novel target for bipolar disorder and other neuropsychiatric disorders. Biol Psychiatry 2018; 83: 761–769.
133.
Boris M, Kaiser CC, Goldblatt A, Elice MW, Edelson SM, Adams JB, et al: Effect of pioglitazone treatment on behavioral symptoms in autistic children. J Neuroinflamm 2007; 4: 3.
134.
Ghaleiha A, Rasa SM, Nikoo M, Farokhnia M, Mohammadi M-R, Akhondzadeh S: A pilot double-blind placebo-controlled trial of pioglitazone as adjunctive treatment to risperidone: effects on aberrant behavior in children with autism. Psychiatry Res 2015; 229: 181–187.
135.
McCracken JT, McGough J, Shah B, Cronin P, Hong D, Aman MG, et al: Risperidone in children with autism and serious behavioral problems. N Engl J Med 2002; 347: 314–321.
136.
Noto C, Ota VK, Gouvea ES, Rizzo LB, Spindola LMN, Honda PHS, et al: Effects of risperidone on cytokine profile in drug-naïve first-episode psychosis. Int J Neuropsychopharmacol 2014; 18:pii: pyu042.
137.
Baumeister D, Ciufolini S, Mondelli V: Effects of psychotropic drugs on inflammation: consequence or mediator of therapeutic effects in psychiatric treatment? Psychopharmacology 2016; 233: 1575–1589.
138.
MacDowell KS, García-Bueno B, Madrigal JLM, Parellada M, Arango C, Micó JA, et al: Risperidone normalizes increased inflammatory parameters and restores anti-inflammatory pathways in a model of neuroinflammation. Int J Neuropsychopharmacol 2013; 16: 121–135.
139.
Choi JE, Widjaja F, Careaga M, Bent S, Ashwood P, Hendren RL: Change in plasma cytokine levels during risperidone treatment in children with autism. J Child Adolesc Psychopharmacol 2014; 24: 586–589.
140.
Tobiasova Z, van der Lingen KHB, Scahill L, Leckman JF, Zhang Y, Chae W, et al: Risperidone-related improvement of irritability in children with autism is not associated with changes in serum of epidermal growth factor and interleukin-13. J Child Adolesc Psychopharmacol 2011; 21: 555–564.
141.
Mostafa GA, Al-Ayadhi LY: Reduced serum concentrations of 25-hydroxy vitamin D in children with autism: relation to autoimmunity. J Neuroinflamm 2012; 9: 201.
142.
Saad K, Abdel-Rahman AA, Elserogy YM, Al-Atram AA, Cannell JJ, Bjørklund G, et al: Vitamin D status in autism spectrum disorders and the efficacy of vitamin D supplementation in autistic children. Nutr Neurosci 2016; 19: 346–351.
143.
Meguid NA, Hashish AF, Anwar M, Sidhom G: Reduced serum levels of 25-hydroxy and 1,25-dihydroxy vitamin D in Egyptian children with autism. J Altern Complement Med 2010; 16: 641–645.
144.
Tostes MHF de S, Polonini HC, Gattaz WF, Raposo NRB, Baptista EB: Low serum levels of 25-hydroxyvitamin D (25-OHD) in children with autism. Trends Psychiatry Psychother 2012; 34: 161–163.
145.
Kočovská E, Andorsdóttir G, Weihe P, Halling J, Fernell E, Stóra T, et al: Vitamin D in the general population of young adults with autism in the Faroe islands. J Autism Dev Disord 2014; 44: 2996–3005.
146.
Cannell JJ: Autism, will vitamin D treat core symptoms? Med Hypotheses 2013; 81: 195–198.
147.
Jia F, Wang B, Shan L, Xu Z, Staal WG, Du L: Core symptoms of autism improved af ter vitamin D supplementation. Pediatrics 2015; 135:e196–e198.
148.
Feng J, Shan L, Du L, Wang B, Li H, Wang W, et al: Clinical improvement following vitamin D3 supplementation in autism spectrum disorder. Nutr Neurosci 2017; 20: 284–290.
149.
Kerley CP, Power C, Gallagher L, Coghlan D: Lack of effect of vitamin D3 supplementation in autism: a 20-week, placebo-controlled RCT. Arch Dis Child 2017; 102: 1030–1036.
150.
Saad K, Abdel-Rahman AA, Elserogy YM, Al-Atram AA, El-Houfey AA, Othman HA-K, et al: Randomized controlled trial of vitamin D supplementation in children with autism spectrum disorder. J Child Psychol Psychiatry 2018; 59: 20–29.
151.
Mazahery H, Stonehouse W, Delshad M, Kruger MC, Conlon CA, Beck KL, et al: Relationship between long chain n-3 polyunsaturated fatty acids and autism spectrum disorder: systematic review and meta-analysis of case-control and randomised controlled trials. Nutrients 2017; 9:pii: E155.
152.
Layé S, Nadjar A, Joffre C, Bazinet RP: Anti-inflammatory effects of omega-3 fatty acids in the brain: physiological mechanisms and relevance to pharmacology. Pharmacol Rev 2018; 70: 12–38.
153.
Madore C, Leyrolle Q, Lacabanne C, Benmamar-Badel A, Joffre C, Nadjar A, et al: Neuroinflammation in autism: plausible role of maternal inflammation, dietary omega 3, and microbiota. Neural Plast 2016; 2016: 3597209.
154.
SalLam M, Motaleb F, Ahmed M, Mahmoud A: Anti-inflammatory effect of omega-3 polyunsaturated fatty acids in children with bronchial asthma; relation to nuclear factor-kappa B (NF-κB) and inflammatory cytokines IL-12 and IL-13. Egypt J Biochem Mol Biol 2010; 28.
155.
Zhang R, He G, Wang Y, Zhou K, Ma E: Omega-3 polyunsaturated fatty acids inhibit the increase in cytokines and chemotactic factors induced in vitro by lymph fluid from an intestinal ischemia-reperfusion injury model. Nutrition 2015; 31: 508–514.
156.
Farjadian S, Moghtaderi M, Kalani M, Gholami T, Hosseini Teshnizi S: Effects of omega-3 fatty acids on serum levels of T-helper cytokines in children with asthma. Cytokine 2016; 85: 61–66.
157.
Zhao Y, Joshi-Barve S, Barve S, Chen LH: Eicosapentaenoic acid prevents LPS-induced TNF-α expression by preventing NF-κB activation. J Am Coll Nutr 2004; 23: 71–78.
158.
Allam-Ndoul B, Guénard F, Barbier O, Vohl M-C: Effect of n-3 fatty acids on the expression of inflammatory genes in THP-1 macrophages. Lipids Health Dis 2016; 15: 69.
159.
Posar A, Visconti P: Omega-3 supplementation in autism spectrum disorders: a still open question? J Pediatr Neurosci 2016; 11: 225–227.
160.
Posar A, Visconti P: Complementary and alternative medicine in autism: the question of omega-3. Pediatr Ann 2016; 45:e103–e107.
161.
Sathe N, Andrews JC, McPheeters ML, Warren ZE: Nutritional and dietary interventions for autism spectrum disorder: a systematic review. Pediatrics 2017; 139:e20170346.
162.
Horvath A, Łukasik J, Szajewska H: ω-3 Fatty acid supplementation does not affect autism spectrum disorder in children: a systematic review and meta-analysis. J Nutr 2017; 147: 367–376.
163.
Li Y-J, Li Y-M, Xiang D-X: Supplement intervention associated with nutritional deficiencies in autism spectrum disorders: a systematic review. Eur J Nutr 2017, DOI: 10.1007/s00394-017-1528-6.
164.
Cheng Y-S, Tseng P-T, Chen Y-W, Stubbs B, Yang W-C, Chen T-Y, et al: Supplementation of omega 3 fatty acids may improve hyperactivity, lethargy, and stereotypy in children with autism spectrum disorders: a meta-analysis of randomized controlled trials. Neuropsychiatr Dis Treat 2017; 13: 2531–2543.
165.
Maclennan KM, Darlington CL, Smith PF: The CNS effects of Ginkgo biloba extracts and ginkgolide B. Prog Neurobiol 2002; 67: 235–257.
166.
Ernst E: The risk-benefit profile of commonly used herbal therapies: Ginkgo, St. John’s Wort, Ginseng, Echinacea, Saw Palmetto, and Kava. Ann Intern Med 2002; 136: 42–53.
167.
Niederhofer H: First preliminary results of an observation of Ginkgo biloba treating patients with autistic disorder. Phyther Res PTR 2009; 23: 1645–1646.
168.
Hasanzadeh E, Mohammadi M-R, Ghanizadeh A, Rezazadeh S-A, Tabrizi M, Rezaei F, et al: A double-blind placebo-controlled trial of Ginkgo biloba added to risperidone in patients with autistic disorders. Child Psychiatry Hum Dev 2012; 43: 674–682.
169.
Prokopieva VD, Yarygina EG, Bokhan NA, Ivanova SA: Use of carnosine for oxidative stress reduction in different pathologies. Oxid Med Cell Longev 2016; 2016: 2939087.
170.
Stvolinsky SL, Kukley ML, Dobrota D, Matejovicova M, Tkac I, Boldyrev AA: Carnosine: an endogenous neuroprotector in the is-chemic brain. Cell Mol Neurobiol 1999; 19: 45–56.
171.
Wang AM, Ma C, Xie ZH, Shen F: Use of carnosine as a natural anti-senescence drug for human beings. Biochemistry 2000; 65: 869–871.
172.
Fleisher-Berkovich S, Abramovitch-Dahan C, Ben-Shabat S, Apte R, Beit-Yannai E: Inhibitory effect of carnosine and N-acetyl carnosine on LPS-induced microglial oxidative stress and inflammation. Peptides 2009; 30: 1306–1312.
173.
Hajizadeh-Zaker R, Ghajar A, Mesgarpour B, Afarideh M, Mohammadi M-R, Akhondzadeh S: L-Carnosine as an adjunctive therapy to risperidone in children with autistic disorder: a randomized, double-blind, placebo-controlled trial. J Child Adolesc Psychopharmacol 2018; 28: 74–81.
174.
Chez MG, Buchanan CP, Aimonovitch MC, Becker M, Schaefer K, Black C, et al: Double-blind, placebo-controlled study of L-carnosine supplementation in children with autistic spectrum disorders. J Child Neurol 2002; 17: 833–837.
175.
Mehrazad-Saber Z, Kheirouri S, Noorazar S-G: Effects of L-carnosine supplementation on sleep disorders and disease severity in autistic children: a randomized, controlled clinical trial. Basic Clin Pharmacol Toxicol 2018; 123: 72–77.
176.
Atkuri KR, Mantovani JJ, Herzenberg LA, Herzenberg LA: N-acetylcysteine – a safe antidote for cysteine/glutathione deficiency. Curr Opin Pharmacol 2007; 7: 355–359.
177.
Deepmala null, Slattery J, Kumar N, Delhey L, Berk M, Dean O, et al: Clinical trials of N-acetylcysteine in psychiatry and neurology: a systematic review. Neurosci Biobehav Rev 2015; 55: 294–321.
178.
Pinar Karapinar S, Ulum YZA, Ozcelik B, Dogan Buzoglu H, Ceyhan D, Balci Peynircioglu B, et al: The effect of N-acetylcys teine and calcium hydroxide on TNF-α and TGF-β1 in lipopolysaccharide-activated macrophages. Arch Oral Biol 2016; 68: 48–54.
179.
Palacio JR, Markert UR, Martínez P: Anti-inflammatory properties of N-acetylcyste ine on lipopolysaccharide-activated macrophages. Inflamm Res 2011; 60: 695–704.
180.
Marler S, Sanders KB, Veenstra-VanderWeele J: N-acetylcysteine as treatment for self-injurious behavior in a child with autism. J Child Adolesc Psychopharmacol 2014; 24: 231–234.
181.
Stutzman D, Dopheide J: Acetylcysteine for treatment of autism spectrum disorder symptoms. Am J Heal Pharm 2015; 72: 1956–1959.
182.
Ghanizadeh A, Derakhshan N: N-acetylcysteine for treatment of autism, a case report. J Res Med Sci 2012; 17: 985–987.
183.
Hardan AY, Fung LK, Libove RA, Obu-khanych TV, Nair S, Herzenberg LA, et al: A randomized controlled pilot trial of oral N-acetylcysteine in children with autism. Biol Psychiatry 2012; 71: 956–961.
184.
Ghanizadeh A, Moghimi-Sarani E: A randomized double-blind placebo controlled clinical trial of N-acetylcysteine added to risperidone for treating autistic disorders. BMC Psychiatry 2013; 13: 196.
185.
Nikoo M, Radnia H, Farokhnia M, Mohammadi M-R, Akhondzadeh S: N-acetylcysteine as an adjunctive therapy to risperidone for treatment of irritability in autism: a randomized, double-blind, placebo-controlled clinical trial of efficacy and safety. Clin Neuropharmacol 2015; 38: 11–17.
186.
Wink LK, Adams R, Wang Z, Klaunig JE, Plawecki MH, Posey DJ, et al: A randomized placebo-controlled pilot study of N-acetylcysteine in youth with autism spectrum disorder. Mol Autism 2016; 7: 26.
187.
Dean OM, Gray KM, Villagonzalo K-A, Dodd S, Mohebbi M, Vick T, et al: A randomised, double blind, placebo-controlled trial of a fixed dose of N-acetyl cysteine in children with autistic disorder. Aust NZ J Psychiatry 2017; 51: 241–249.
188.
Li Q, Zhou J-M: The microbiota-gut-brain axis and its potential therapeutic role in autism spectrum disorder. Neuroscience 2016; 324: 131–139.
189.
Doenyas C: Gut microbiota, inflammation, and probiotics on neural development in autism spectrum disorder. Neuroscience 2018; 374: 271–286.
190.
Yang Y, Tian J, Yang B: Targeting gut microbiome: a novel and potential therapy for autism. Life Sci 2018; 194: 111–119.
191.
Sandler RH, Finegold SM, Bolte ER, Buchanan CP, Maxwell AP, Väisänen ML, et al: Short-term benefit from oral vancomycin treatment of regressive-onset autism. J Child Neurol 2000; 15: 429–435.
192.
Kałużna-Czaplińska J, Błaszczyk S: The level of arabinitol in autistic children after probiotic therapy. Nutrition 2012; 28: 124–126.
193.
Kang D-W, Adams JB, Gregory AC, Borody T, Chittick L, Fasano A, et al: Microbiota transfer therapy alters gut ecosystem and improves gastrointestinal and autism symptoms: an open-label study. Microbiome 2017; 5: 10.
194.
Pärtty A, Kalliomäki M, Wacklin P, Salmi-nen S, Isolauri E: A possible link between early probiotic intervention and the risk of neuropsychiatric disorders later in childhood: a randomized trial. Pediatr Res 2015; 77: 823–828.
195.
Seki Y, Kato TA, Monji A, Mizoguchi Y, Horikawa H, Sato-Kasai M, et al: Pretreatment of aripiprazole and minocycline, but not haloperidol, suppresses oligodendrocyte damage from interferon-γ-stimulated microglia in co-culture model. Schizophr Res 2013; 151: 20–28.
196.
Sobiś J, Rykaczewska-Czerwińska M, Świę tochowska E, Gorczyca P: Therapeutic effect of aripiprazole in chronic schizophrenia is accompanied by anti-inflammatory activity. Pharmacol Rep 2015; 67: 353–359.
197.
Leng Y, Chuang D-M: Endogenous alpha-synuclein is induced by valproic acid through histone deacetylase inhibition and participates in neuroprotection against glutamate-induced excitotoxicity. J Neurosci 2006; 26: 7502–7512.
198.
Song R, Yu D, Yoon J, Park J: Valproic acid attenuates the expression of pro-inflammatory cytokines lipopolysaccharide-treated canine peripheral blood mononuclear cells (in vitro) and in a canine endotoxemia model (in vivo). Vet Immunol Immunopathol 2015; 166: 132–137.
199.
Suda S, Katsura K, Kanamaru T, Saito M, Katayama Y: Valproic acid attenuates is-chemia-reperfusion injury in the rat brain through inhibition of oxidative stress and inflammation. Eur J Pharmacol 2013; 707: 26–31.
200.
Ximenes JCM, de Oliveira Gonçalves D, Siqueira RMP, Neves KRT, Santos Cerqueira G, Correia AO, et al: Valproic acid: an anticonvulsant drug with potent antinociceptive and anti-inflammatory properties. Naunyn Schmiedebergs Arch Pharmacol 2013; 386: 575–587.
201.
Chugani DC, Chugani HT, Wiznitzer M, Parikh S, Evans PA, Hansen RL, et al: Efficacy of low-dose buspirone for restricted and repetitive behavior in young children with autism spectrum disorder: a randomized trial. J Pediatr 2016; 170: 45–53.e1–4.
202.
Ghanizadeh A, Ayoobzadehshirazi A: A randomized double-blind placebo-controlled clinical trial of adjuvant buspirone for irritability in autism. Pediatr Neurol 2015; 52: 77–81.
203.
McCormick LH: Treatment with buspirone in a patient with autism. Arch Fam Med 1997; 6: 368–370.
204.
Chantiluke K, Barrett N, Giampietro V, Brammer M, Simmons A, Rubia K: Disorder-dissociated effects of fluoxetine on brain function of working memory in attention deficit hyperactivity disorder and autism spectrum disorder. Psychol Med 2015; 45: 1195–1205.
205.
Hollander E, Soorya L, Chaplin W, Anagnostou E, Taylor BP, Ferretti CJ, et al: A double-blind placebo-controlled trial of fluoxetine for repetitive behaviors and global severity in adult autism spectrum disorders. Am J Psychiatry 2012; 169: 292–299.
206.
Hollander E, Phillips A, Chaplin W, Zagursky K, Novotny S, Wasserman S, et al: A placebo-controlled crossover trial of liquid fluoxetine on repetitive behaviors in childhood and adolescent autism. Neuropsychopharmacology 2005; 30: 582–589.
207.
Alcamí Pertejo M, Peral Guerra M, Gilaberte I: Open study of fluoxetine in children with autism. Actas Esp Psiquiatr 2000; 28: 353–356.
208.
Fatemi SH, Realmuto GM, Khan L, Thuras P: Fluoxetine in treatment of adolescent patients with autism: a longitudinal open trial. J Autism Dev Disord 1998; 28: 303–307.
209.
Owley T, Brune CW, Salt J, Walton L, Guter S, Ayuyao N, et al: A pharmacogenetic study of escitalopram in autism spectrum disorders. Autism Res 2010; 3: 1–7.
210.
Owley T, Walton L, Salt J, Guter SJ, Winnega M, Leventhal BL, et al: An open-label trial of escitalopram in pervasive developmental disorders. J Am Acad Child Adolesc Psychiatry 2005; 44: 343–348.
211.
King BH, Hollander E, Sikich L, McCracken JT, Scahill L, Bregman JD, et al: Lack of efficacy of citalopram in children with autism spectrum disorders and high levels of repetitive behavior: citalopram ineffective in children with autism. Arch Gen Psychiatry 2009; 66: 583–590.
212.
Hellings JA, Kelley LA, Gabrielli WF, Kilgore E, Shah P: Sertraline response in adults with mental retardation and autistic disorder. J Clin Psychiatry 1996; 57: 333–336.
213.
McDougle CJ, Brodkin ES, Naylor ST, Carlson DC, Cohen DJ, Price LH: Sertraline in adults with pervasive developmental disorders: a prospective open-label investigation. J Clin Psychopharmacol 1998; 18: 62–66.
214.
Steingard RJ, Zimnitzky B, DeMaso DR, Bauman ML, Bucci JP: Sertraline treatment of transition-associated anxiety and agitation in children with autistic disorder. J Child Adolesc Psychopharmacol 1997; 7: 9–15.
215.
Posey DJ, Kem DL, Swiezy NB, Sweeten TL, Wiegand RE, McDougle CJ: A pilot study of D-cycloserine in subjects with autistic disorder. Am J Psychiatry 2004; 161: 2115–2117.
216.
Minshawi NF, Wink LK, Shaffer R, Plawecki MH, Posey DJ, Liu H, et al: A randomized, placebo-controlled trial of D-cycloserine for the enhancement of social skills training in autism spectrum disorders. Mol Autism 2016; 7: 2.
217.
Urbano M, Okwara L, Manser P, Hartmann K, Deutsch SI: A trial of D-cycloserine to treat the social deficit in older adolescents and young adults with autism spectrum disorders. J Neuropsychiatry Clin Neurosci 2015; 27: 133–138.
218.
Urbano M, Okwara L, Manser P, Hartmann K, Herndon A, Deutsch SI: A trial of D-cycloserine to treat stereotypies in older adolescents and young adults with autism spectrum disorder. Clin Neuropharmacol 2014; 37: 69–72.
219.
King BH, Wright DM, Handen BL, Sikich L, Zimmerman AW, McMahon W, et al: Double-blind, placebo-controlled study of amantadine hydrochloride in the treatment of children with autistic disorder. J Am Acad Child Adolesc Psychiatry 2001; 40: 658–665.
220.
Owley T, Salt J, Guter S, Grieve A, Walton L, Ayuyao N, et al: A prospective, open-label trial of memantine in the treatment of cognitive, behavioral, and memory dysfunction in pervasive developmental disorders. J Child Adolesc Psychopharmacol 2006; 16: 517–524.
221.
Chez MG, Burton Q, Dowling T, Chang M, Khanna P, Kramer C: Memantine as adjunctive therapy in children diagnosed with autistic spectrum disorders: an observation of initial clinical response and maintenance tolerability. J Child Neurol 2007; 22: 574–579.
222.
Joshi G, Wozniak J, Faraone SV, Fried R, Chan J, Furtak S, et al: A prospective open-label trial of memantine hydrochloride for the treatment of social deficits in intellectually capable adults with autism spectrum disorder. J Clin Psychopharmacol 2016; 36: 262–271.
223.
Williams PG, Allard A, Sears L, Dalrymple N, Bloom AS: Brief report: case reports on naltrexone use in children with autism: controlled observations regarding benefits and practical issues of medication management. J Autism Dev Disord 2001; 31: 103–108.
224.
Willemsen-Swinkels SH, Buitelaar JK, van Berckelaer-Onnes IA, van Engeland H: Brief report: six months continuation treatment in naltrexone-responsive children with autism: an open-label case-control design. J Autism Dev Disord 1999; 29: 167–169.
225.
Campbell M, Anderson LT, Small AM, Adams P, Gonzalez NM, Ernst M: Naltrexone in autistic children: behavioral symptoms and attentional learning. J Am Acad Child Adolesc Psychiatry 1993; 32: 1283–1291.
226.
Zingarelli G, Ellman G, Hom A, Wymore M, Heidorn S, Chicz-DeMet A: Clinical effects of naltrexone on autistic behavior. Am J Ment Retard 1992; 97: 57–63.
227.
Leboyer M, Bouvard MP, Launay JM, Tabuteau F, Waller D, Dugas M, et al: Brief report: a double-blind study of naltrexone in infantile autism. J Autism Dev Disord 1992; 22: 309–319.
228.
Knabe R, Schulz P, Richard J: Initial aggravation of self-injurious behavior in autistic patients receiving naltrexone treatment. J Autism Dev Disord 1990; 20: 591–593.
229.
Walters AS, Barrett RP, Feinstein C, Mercurio A, Hole WT: A case report of naltrexone treatment of self-injury and social withdrawal in autism. J Autism Dev Disord 1990; 20: 169–176.
230.
Campbell M, Anderson LT, Small AM, Locascio JJ, Lynch NS, Choroco MC: Naltrexone in autistic children: a double-blind and placebo-controlled study. Psychopharmacol Bull 1990; 26: 130–135.
231.
Barrett RP, Feinstein C, Hole WT: Effects of naloxone and naltrexone on self-injury: a double-blind, placebo-controlled analysis. Am J Ment Retard 1989; 93: 644–651.
232.
Feldman HM, Kolmen BK, Gonzaga AM: Naltrexone and communication skills in young children with autism. J Am Acad Child Adolesc Psychiatry 1999; 38: 587–593.
233.
Kolmen BK, Feldman HM, Handen BL, Janosky JE: Naltrexone in young autistic children: replication study and learning measures. J Am Acad Child Adolesc Psychiatry 1997; 36: 1570–1578.
234.
Willemsen-Swinkels SH, Buitelaar JK, van Engeland H: The effects of chronic naltrexone treatment in young autistic children: a double-blind placebo-controlled crossover study. Biol Psychiatry 1996; 39: 1023–1031.
235.
Willemsen-Swinkels SH, Buitelaar JK, Weijnen FG, van Engeland H: Placebo-controlled acute dosage naltrexone study in young autistic children. Psychiatry Res 1995; 58: 203–215.
236.
Bouvard MP, Leboyer M, Launay JM, Recasens C, Plumet MH, Waller-Perotte D, et al: Low-dose naltrexone effects on plasma chemistries and clinical symptoms in autism: a double-blind, placebo-controlled study. Psychiatry Res 1995; 58: 191–201.
237.
Willemsen-Swinkels SH, Buitelaar JK, Nijhof GJ, van Engeland H: Failure of naltrexone hydrochloride to reduce self-injurious and autistic behavior in mentally retard ed adults. Double-blind placebo-controlled studies. Arch Gen Psychiatry 1995; 52: 766–773.
238.
Kolmen BK, Feldman HM, Handen BL, Janosky JE: Naltrexone in young autistic children: a double-blind, placebo-controlled crossover study. J Am Acad Child Adolesc Psychiatry 1995; 34: 223–231.
239.
Gonzalez NM, Campbell M, Small AM, Shay J, Bluhm LD, Adams PB, et al: Naltrexone plasma levels, clinical response and effect on weight in autistic children. Psychopharmacol Bull 1994; 30: 203–208.
240.
Scifo R, Cioni M, Nicolosi A, Batticane N, Tirolo C, Testa N, et al: Opioid-immune interactions in autism: behavioural and immunological assessment during a double-blind treatment with naltrexone. Ann Ist Super Sanita 1996; 32: 351–359.
241.
Al-Ayadhi LY, Halepoto DM, Al-Dress AM, Mitwali Y, Zainah R: Behavioral benefits of camel milk in subjects with autism spectrum disorder. J Coll Physicians Surg Pak 2015; 25: 819–823.
242.
Bashir S, Al-Ayadhi LY: Effect of camel milk on thymus and activation-regulated chemokine in autistic children: double-blind study. Pediatr Res 2014; 75: 559–563.
243.
Hu Y, Ehli EA, Boomsma DI: MicroRNAs as biomarkers for psychiatric disorders with a focus on autism spectrum disorder: current progress in genetic association studies, expression profiling, and translational research. Autism Res 2017; 10: 1184–1203.
244.
Fregeac J, Colleaux L, Nguyen LS: The emerging roles of microRNAs in autism spectrum disorders. Neurosci Biobehav Rev 2016; 71: 729–738.
245.
Inga Jácome MC, Morales Chacòn LM, Vera Cuesta H, Maragoto Rizo C, Whilby Santiesteban M, Ramos Hernandez L, et al: Peripheral inflammatory markers contributing to comorbidities in autism. Behav Sci 2016; 6:pii: E29.
246.
Ahmad SF, Nadeem A, Ansari MA, Bakheet SA, Attia SM, Zoheir KMA, et al: Imbalance between the anti- and pro-inflammatory milieu in blood leukocytes of autistic children. Mol Immunol 2017; 82: 57–65.
247.
Singh VK: Phenotypic expression of autoimmune autistic disorder (AAD): a major subset of autism. Ann Clin Psychiatry 2009; 21: 148–161.
248.
Kim J-W, Seung H, Kwon KJ, Ko MJ, Lee EJ, Oh HA, et al: Subchronic treatment of donepezil rescues impaired social, hyperactive, and stereotypic behavior in valproic acid-induced animal model of autism. PLoS One 2014; 9:e104927.
249.
Karvat G, Kimchi T: Acetylcholine elevation relieves cognitive rigidity and social deficiency in a mouse model of autism. Neuropsychopharmacology 2014; 39: 831–840.
250.
Bakheet SA, Alzahrani MZ, Ansari MA, Nadeem A, Zoheir KMA, Attia SM, et al: Resveratrol ameliorates dysregulation of Th1, Th2, Th17, and T regulatory cell-related transcription factor signaling in a BTBR T + tf/J mouse model of autism. Mol Neurobiol 2017; 54: 5201–5212.
251.
Bakheet SA, Alzahrani MZ, Nadeem A, Ansari MA, Zoheir KMA, Attia SM, et al: Resveratrol treatment attenuates chemokine receptor expression in the BTBR T + tf/J mouse model of autism. Mol Cell Neurosci 2016; 77: 1–10.
252.
Bhandari R, Kuhad A: Resveratrol suppresses neuroinflammation in the experimental paradigm of autism spectrum disorders. Neurochem Int 2017; 103: 8–23.
253.
Bambini-Junior V, Zanatta G, Della Flora Nunes G, Mueller de Melo G, Michels M, Fontes-Dutra M, et al: Resveratrol prevents social deficits in animal model of autism induced by valproic acid. Neurosci Lett 2014; 583: 176–181.
254.
Wu H, Wang X, Gao J, Liang S, Hao Y, Sun C, et al: Fingolimod (FTY720) attenuates social deficits, learning and memory impairments, neuronal loss and neuroinflammation in the rat model of autism. Life Sci 2017; 173: 43–54.
255.
Hirsch MM, Deckmann I, Fontes-Dutra M, Bauer-Negrini G, Della-Flora Nunes G, Nunes W, et al: Behavioral alterations in autism model induced by valproic acid and translational analysis of circulating micro-RNA. Food Chem Toxicol 2018; 115: 336–343.
Copyright / Drug Dosage / Disclaimer
Copyright: All rights reserved. No part of this publication may be translated into other languages, reproduced or utilized in any form or by any means, electronic or mechanical, including photocopying, recording, microcopying, or by any information storage and retrieval system, without permission in writing from the publisher.
Drug Dosage: The authors and the publisher have exerted every effort to ensure that drug selection and dosage set forth in this text are in accord with current recommendations and practice at the time of publication. However, in view of ongoing research, changes in government regulations, and the constant flow of information relating to drug therapy and drug reactions, the reader is urged to check the package insert for each drug for any changes in indications and dosage and for added warnings and precautions. This is particularly important when the recommended agent is a new and/or infrequently employed drug.
Disclaimer: The statements, opinions and data contained in this publication are solely those of the individual authors and contributors and not of the publishers and the editor(s). The appearance of advertisements or/and product references in the publication is not a warranty, endorsement, or approval of the products or services advertised or of their effectiveness, quality or safety. The publisher and the editor(s) disclaim responsibility for any injury to persons or property resulting from any ideas, methods, instructions or products referred to in the content or advertisements.
You do not currently have access to this content.