1
|
Nie L, Irwin C, Geahchan S, Singh KK. Human pluripotent stem cell (hPSC)-derived models for autism spectrum disorder drug discovery. Expert Opin Drug Discov 2025; 20:233-251. [PMID: 39718245 DOI: 10.1080/17460441.2024.2416484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 10/10/2024] [Indexed: 12/25/2024]
Abstract
INTRODUCTION Autism spectrum disorder (ASD) is a prevalent and complex neurodevelopmental disorder (NDD) with genetic and environmental origins. Currently, there are no effective pharmacological treatments targeting core ASD features. This leads to unmet medical needs of individuals with ASD and requires relevant human disease models recapitulating genetic and clinical heterogeneity to better understand underlying mechanisms and identify potential pharmacological therapies. Recent advancements in stem cell technology have enabled the generation of human pluripotent stem cell (hPSC)-derived two-dimensional (2D) and three-dimensional (3D) neural models, which serve as powerful tools for ASD modeling and drug discovery. AREAS COVERED This article reviews the applications of hPSC-derived 2D and 3D neural models in studying various forms of ASD using pharmacological perturbation and drug screenings, highlighting the potential use of these models to develop novel pharmacological treatment strategies for ASD. EXPERT OPINION hPSC-derived models recapitulate early human brain development spatiotemporally and have allowed patient-specific mechanistic investigation and therapeutic development using advanced molecular technologies, which will contribute to precision medicine for ASD therapy. Improvements are still required in hPSC-based models to further enhance their physiological relevance, clinical translation, and scalability for ASD drug discovery.
Collapse
Affiliation(s)
- Lingdi Nie
- Donald K. Johnson Eye Institute, Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | - Courtney Irwin
- Donald K. Johnson Eye Institute, Krembil Research Institute, University Health Network, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Sarah Geahchan
- Donald K. Johnson Eye Institute, Krembil Research Institute, University Health Network, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Karun K Singh
- Donald K. Johnson Eye Institute, Krembil Research Institute, University Health Network, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Department of Ophthalmology and Vision Sciences, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
2
|
Mihailovich M, Tolinački M, Soković Bajić S, Lestarevic S, Pejovic-Milovancevic M, Golić N. The Microbiome-Genetics Axis in Autism Spectrum Disorders: A Probiotic Perspective. Int J Mol Sci 2024; 25:12407. [PMID: 39596472 PMCID: PMC11594817 DOI: 10.3390/ijms252212407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 11/15/2024] [Accepted: 11/15/2024] [Indexed: 11/28/2024] Open
Abstract
Autism spectrum disorder (commonly known as autism) is a complex and prevalent neurodevelopmental condition characterized by challenges in social behavior, restricted interests, and repetitive behaviors. It is projected that the annual cost of autism spectrum disorder in the US will reach USD 461 billion by 2025. However, despite being a major public health problem, effective treatment for the underlying symptoms remains elusive. As numerous literature data indicate the role of gut microbiota in autism prognosis, particularly in terms of alleviating gastrointestinal (GI) symptoms, high hopes have been placed on probiotics for autism treatment. Approximately twenty clinical studies have been conducted using single or mixed probiotic cultures. However, unequivocal results on the effect of probiotics on people with autism have not been obtained. The small sample sizes, differences in age of participants, choice of probiotics, dose and duration of treatment, outcome measures, and analytical methods used are largely inconsistent, making it challenging to draw distinctive conclusions. Here, we discuss the experimental evidence for specific gut bacteria and their metabolites and how they affect autism in light of the phenotypic and etiological complexity and heterogeneity. We propose a personalized medicine approach for using probiotics to increase the quality of life of individuals with autism by selecting specific probiotics to improve particular features of the condition.
Collapse
Affiliation(s)
- Marija Mihailovich
- Institute of Molecular Genetics and Genetic Engineering (IMGGE), University of Belgrade, 11042 Belgrade, Serbia; (M.T.); (S.S.B.); (N.G.)
- Human Technopole, 20157 Milan, Italy
| | - Maja Tolinački
- Institute of Molecular Genetics and Genetic Engineering (IMGGE), University of Belgrade, 11042 Belgrade, Serbia; (M.T.); (S.S.B.); (N.G.)
| | - Svetlana Soković Bajić
- Institute of Molecular Genetics and Genetic Engineering (IMGGE), University of Belgrade, 11042 Belgrade, Serbia; (M.T.); (S.S.B.); (N.G.)
| | - Sanja Lestarevic
- Institute of Mental Health, 11000 Belgrade, Serbia; (S.L.); (M.P.-M.)
| | - Milica Pejovic-Milovancevic
- Institute of Mental Health, 11000 Belgrade, Serbia; (S.L.); (M.P.-M.)
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Nataša Golić
- Institute of Molecular Genetics and Genetic Engineering (IMGGE), University of Belgrade, 11042 Belgrade, Serbia; (M.T.); (S.S.B.); (N.G.)
| |
Collapse
|
3
|
Saldaris JM, Ayalde J, Kankanange S, Keeley J, Leonard H, Jacoby P, Marsh ED, Benke TA, Demarest ST, Downs J. Parent-reported outcome measures evaluating communication in individuals with rare neurodevelopmental disorders: A systematic review. INTERNATIONAL JOURNAL OF LANGUAGE & COMMUNICATION DISORDERS 2024; 59:2528-2553. [PMID: 39141588 PMCID: PMC11649302 DOI: 10.1111/1460-6984.13100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 07/25/2024] [Indexed: 08/16/2024]
Abstract
BACKGROUND Communication impairments are a leading concern for parent caregivers of individuals with rare neurodevelopmental disorders (RNDDs). Clinical trials of disease modifying therapies require valid and responsive outcome measures that are relevant to individuals with RNDDs. Identifying and evaluating current psychometric properties for communication measures is a critical step towards the selection and use of appropriate instruments. AIMS This systematic review offers (1) a description of parent-reported communication measures and (2) evidence for their psychometric properties, in RNDDs. METHODS The systematic review protocol was registered in the International Prospective Register of Systematic Reviews (PROSPERO; CRD42022334649). MEDLINE (Ovid), Embase, PsychINFO, Web of Science, CINAHL Plus, Cochrane Library, ClinicalTrials.gov, the Australian New Zealand Clinical Trials Registry were searched from inception to August 2023. Methodological assessment of quality was completed using the COnsensus-based Standards for the selection of health status Measurement INstruments (COSMIN) checklist. Parent-reported measures used in observational studies and clinical trials were identified. Data on utility, reliability and validity for RNDDs were extracted. MAIN CONTRIBUTION Sixteen parent-reported communication measures were used in RNDD research, the Vineland Adaptive Behavior Scales being most commonly used. Validation data in RNDDs were identified for six of these measures. Limitations related to sample size or the scope of psychometric testing. CONCLUSIONS Many communication measures have been used for RNDDs but there are few data validating their use. Valid and reliable methods of measuring communication in persons with RNDDs is a priority for future high-quality clinical trials. WHAT THIS PAPER ADDS What is already known on the subject Communication is a critical domain for families with a child with a rare neurodevelopmental disorder (RNDD). Validated outcome measures are essential for accurate evaluation and interpretation of responses to treatments in clinical trials. What this paper adds to existing knowledge We identified 16 parent-reported communication measures that have been used with RNDDs, but only six measures had validation data for at least one RNDD. High quality evidence is accumulating, with all validation studies in this review published between 2020 to 2023. Modifications of existing measures may be required to assess communication for RNDDs. What are the clinical implications of this work? This systematic review catalogues the available psychometric data for communication measures and indicates an ongoing need for new validation studies to ensure they are fit-for-purpose for upcoming clinical trials in RNDDs. This review will inform the selection of communication measures for clinical trials and research studies.
Collapse
Affiliation(s)
- Jacinta M Saldaris
- Telethon Kids Institute, Nedlands, Western Australia, Australia
- The University of Western Australia, Centre for Child Health Research, Crawley, Western Australia, Australia
| | - Jeremiah Ayalde
- Telethon Kids Institute, Nedlands, Western Australia, Australia
| | | | - Jessica Keeley
- Telethon Kids Institute, Nedlands, Western Australia, Australia
| | - Helen Leonard
- Telethon Kids Institute, Nedlands, Western Australia, Australia
- The University of Western Australia, Centre for Child Health Research, Crawley, Western Australia, Australia
| | - Peter Jacoby
- Telethon Kids Institute, Nedlands, Western Australia, Australia
| | - Eric D Marsh
- Children's Hospital of Philadelphia, Division of Child Neurology, Philadelphia, Pennsylvania, USA
- Departments of Neurology and Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Tim A Benke
- Children's Hospital Colorado, Neurology and Pharmacology, Aurora, Colorado, USA
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Scott T Demarest
- Children's Hospital Colorado, Neurology and Pharmacology, Aurora, Colorado, USA
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Jenny Downs
- Telethon Kids Institute, Nedlands, Western Australia, Australia
- The University of Western Australia, Centre for Child Health Research, Crawley, Western Australia, Australia
- Curtin University, School of Allied Health, Bentley, Western Australia, Australia
| |
Collapse
|
4
|
Dionne O, Abolghasemi A, Corbin F, Çaku A. Implication of the endocannabidiome and metabolic pathways in fragile X syndrome pathophysiology. Psychiatry Res 2024; 337:115962. [PMID: 38763080 DOI: 10.1016/j.psychres.2024.115962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 05/10/2024] [Accepted: 05/11/2024] [Indexed: 05/21/2024]
Abstract
Fragile X Syndrome (FXS) results from the silencing of the FMR1 gene and is the most prevalent inherited cause of intellectual disability and the most frequent monogenic cause of autism spectrum disorder. It is well established that Fragile X individuals are subjected to a wide array of comorbidities, ranging from cognitive, behavioural, and medical origin. Furthermore, recent studies have also described metabolic impairments in FXS individuals. However, the molecular mechanisms linking FMRP deficiency to improper metabolism are still misunderstood. The endocannabinoidome (eCBome) is a lipid-based signalling system that regulates several functions across the body, ranging from cognition, behaviour and metabolism. Alterations in the eCBome have been described in FXS animal models and linked to neuronal hyperexcitability, a core deficit of the disease. However, the potential link between dysregulation of the eCBome and altered metabolism observed in FXS remains unexplored. As such, this review aims to overcome this issue by describing the most recent finding related to eCBome and metabolic dysfunctions in the context of FXS. A better comprehension of this association will help deepen our understanding of FXS pathophysiology and pave the way for future therapeutic interventions.
Collapse
Affiliation(s)
- Olivier Dionne
- Biochemistry and Functional Genomic Department, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Canada.
| | - Armita Abolghasemi
- Biochemistry and Functional Genomic Department, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Canada
| | - François Corbin
- Biochemistry and Functional Genomic Department, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Canada
| | - Artuela Çaku
- Biochemistry and Functional Genomic Department, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Canada
| |
Collapse
|
5
|
Chakraborty P, Dey A, Gopalakrishnan AV, Swati K, Ojha S, Prakash A, Kumar D, Ambasta RK, Jha NK, Jha SK, Dewanjee S. Glutamatergic neurotransmission: A potential pharmacotherapeutic target for the treatment of cognitive disorders. Ageing Res Rev 2023; 85:101838. [PMID: 36610558 DOI: 10.1016/j.arr.2022.101838] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 12/27/2022] [Accepted: 12/29/2022] [Indexed: 01/06/2023]
Abstract
In the mammalian brain, glutamate is regarded to be the primary excitatory neurotransmitter due to its widespread distribution and wide range of metabolic functions. Glutamate plays key roles in regulating neurogenesis, synaptogenesis, neurite outgrowth, and neuron survival in the brain. Ionotropic and metabotropic glutamate receptors, neurotransmitters, neurotensin, neurosteroids, and others co-ordinately formulate a complex glutamatergic network in the brain that maintains optimal excitatory neurotransmission. Cognitive activities are potentially synchronized by the glutamatergic activities in the brain via restoring synaptic plasticity. Dysfunctional glutamate receptors and other glutamatergic components are responsible for the aberrant glutamatergic activity in the brain that cause cognitive impairments, loss of synaptic plasticity, and neuronal damage. Thus, controlling the brain's glutamatergic transmission and modifying glutamate receptor function could be a potential therapeutic strategy for cognitive disorders. Certain drugs that regulate glutamate receptor activities have shown therapeutic promise in improving cognitive functions in preclinical and clinical studies. However, several issues regarding precise functional information of glutamatergic activity are yet to be comprehensively understood. The present article discusses the scope of developing glutamatergic systems as prospective pharmacotherapeutic targets to treat cognitive disorders. Special attention has been given to recent developments, challenges, and future prospects.
Collapse
Affiliation(s)
- Pratik Chakraborty
- Advanced Pharmacognosy Research Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, India
| | - Abhijit Dey
- Department of Life Sciences, Presidency University, Kolkata 700073, West Bengal, India
| | - Abilash Valsala Gopalakrishnan
- Department of Biomedical Sciences, School of Bio Sciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu 632014, India
| | - Kumari Swati
- Department of Biotechnology, School of Life Science, Mahatma Gandhi Central University, Motihari, Bihar, India
| | - Shreesh Ojha
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, Abu Dhabi, United Arab Emirates
| | - Anand Prakash
- Department of Biotechnology, School of Life Science, Mahatma Gandhi Central University, Motihari, Bihar, India
| | - Dhruv Kumar
- School of Health Sciences & Technology, UPES University, Dehradun, Uttarakhand 248007, India
| | - Rashmi K Ambasta
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Delhi 110042, India
| | - Niraj Kumar Jha
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida 201310, UP, India; School of Bioengineering & Biosciences, Lovely Professional University, Phagwara, Punjab 144411, India.
| | - Saurabh Kumar Jha
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida 201310, UP, India; Department of Biotechnology Engineering and Food Technology, Chandigarh University, Mohali 140413, India; Department of Biotechnology, School of Applied & Life Sciences (SALS), Uttaranchal University, Dehradun 248007, India.
| | - Saikat Dewanjee
- Advanced Pharmacognosy Research Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, India.
| |
Collapse
|
6
|
Lacroix A, Proteau-Lemieux M, Côté S, Near J, Hui SC, Edden RA, Lippé S, Çaku A, Corbin F, Lepage JF. Multimodal assessment of the GABA system in patients with fragile-X syndrome and neurofibromatosis of type 1. Neurobiol Dis 2022; 174:105881. [DOI: 10.1016/j.nbd.2022.105881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 09/12/2022] [Accepted: 10/02/2022] [Indexed: 11/24/2022] Open
|
7
|
Kenny A, Wright D, Stanfield AC. EEG as a translational biomarker and outcome measure in fragile X syndrome. Transl Psychiatry 2022; 12:34. [PMID: 35075104 PMCID: PMC8786970 DOI: 10.1038/s41398-022-01796-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 12/01/2021] [Accepted: 01/12/2022] [Indexed: 01/08/2023] Open
Abstract
Targeted treatments for fragile X syndrome (FXS) have frequently failed to show efficacy in clinical testing, despite success at the preclinical stages. This has highlighted the need for more effective translational outcome measures. EEG differences observed in FXS, including exaggerated N1 ERP amplitudes, increased resting gamma power and reduced gamma phase-locking in the sensory cortices, have been suggested as potential biomarkers of the syndrome. These abnormalities are thought to reflect cortical hyper excitability resulting from an excitatory (glutamate) and inhibitory (GABAergic) imbalance in FXS, which has been the target of several pharmaceutical remediation studies. EEG differences observed in humans also show similarities to those seen in laboratory models of FXS, which may allow for greater translational equivalence and better predict clinical success of putative therapeutics. There is some evidence from clinical trials showing that treatment related changes in EEG may be associated with clinical improvements, but these require replication and extension to other medications. Although the use of EEG characteristics as biomarkers is still in the early phases, and further research is needed to establish its utility in clinical trials, the current research is promising and signals the emergence of an effective translational biomarker.
Collapse
Affiliation(s)
- Aisling Kenny
- Patrick Wild Centre, Division of Psychiatry, Kennedy Tower, Royal Edinburgh Hospital, University of Edinburgh, EH10 5HF, Edinburgh, UK.
| | - Damien Wright
- grid.4305.20000 0004 1936 7988Patrick Wild Centre, Division of Psychiatry, Kennedy Tower, Royal Edinburgh Hospital, University of Edinburgh, EH10 5HF Edinburgh, UK
| | - Andrew C. Stanfield
- grid.4305.20000 0004 1936 7988Patrick Wild Centre, Division of Psychiatry, Kennedy Tower, Royal Edinburgh Hospital, University of Edinburgh, EH10 5HF Edinburgh, UK
| |
Collapse
|
8
|
Roysommuti S, Wyss JM. Brain-Derived Neurotrophic Factor Potentiates Entorhinal-Dentate but not Hippocampus CA1 Pathway in Adult Male Rats: A Mechanism of Taurine-Modulated BDNF on Learning and Memory. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1370:369-379. [PMID: 35882811 PMCID: PMC9467516 DOI: 10.1007/978-3-030-93337-1_35] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Taurine plays an important role in neural growth and function from early to adult life, particularly in learning and memory via BDNF action. This study tested the hypothesis that BDNF differentially potentiates entorhinal-hippocampal synaptic transmission in vivo in adult rats. In anesthetized male Sprague-Dawley rats, a stainless steel recording electrode with an attached microinjector was placed into CA1 and the dentate gyrus to record fEPSP, and a paired stainless steel electrode was inserted into entorhinal cortex for continuous paired-pulse stimulation of that brain region. In the dentate gyrus, microinjection of BDNF resulted in a gradual increase in the peak slope of the fEPSP. Following the infusion, the peak fEPSP began to rise in about 8 min, reached a maximum of 120 ± 2% (from baseline) by about 20 min, and remained near peak elevation (~115%) for more than 30 min. In contrast, the same dose of BDNF when injected into CA1 had no consistent effect on fEPSP slopes in the CA1. Further, an equimolar cytochrome C (horse heart) infusion had no significant effect on fEPSP slopes in either the dentate gyrus or CA1. The potentiation effect of BDNF in the dentate gyrus is consistent with a significant increase in power spectral density of dentate gyrus field potentials at 70-200 Hz, but not at frequencies below 70 Hz. In addition, the CA1 power spectral density was not affected by BDNF (compared to cytochrome C). These data indicate that in vivo BDNF potentiates entorhinal-hippocampal synaptic transmission in dentate gyrus, but not in CA1.
Collapse
Affiliation(s)
- Sanya Roysommuti
- Department of Cell, Developmental and Integrative Biology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA.
- Department of Physiology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand.
| | - James Michael Wyss
- Department of Cell, Developmental and Integrative Biology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
9
|
Vallés AS, Barrantes FJ. Dendritic spine membrane proteome and its alterations in autistic spectrum disorder. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2021; 128:435-474. [PMID: 35034726 DOI: 10.1016/bs.apcsb.2021.09.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Dendritic spines are small protrusions stemming from the dendritic shaft that constitute the primary specialization for receiving and processing excitatory neurotransmission in brain synapses. The disruption of dendritic spine function in several neurological and neuropsychiatric diseases leads to severe information-processing deficits with impairments in neuronal connectivity and plasticity. Spine dysregulation is usually accompanied by morphological alterations to spine shape, size and/or number that may occur at early pathophysiological stages and not necessarily be reflected in clinical manifestations. Autism spectrum disorder (ASD) is one such group of diseases involving changes in neuronal connectivity and abnormal morphology of dendritic spines on postsynaptic neurons. These alterations at the subcellular level correlate with molecular changes in the spine proteome, with alterations in the copy number, topography, or in severe cases in the phenotype of the molecular components, predominantly of those proteins involved in spine recognition and adhesion, reflected in abnormally short lifetimes of the synapse and compensatory increases in synaptic connections. Since cholinergic neurotransmission participates in the regulation of cognitive function (attention, memory, learning processes, cognitive flexibility, social interactions) brain acetylcholine receptors are likely to play an important role in the dysfunctional synapses in ASD, either directly or indirectly via the modulatory functions exerted on other neurotransmitter receptor proteins and spine-resident proteins.
Collapse
Affiliation(s)
- Ana Sofía Vallés
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (UNS-CONICET), Bahía Blanca, Argentina
| | - Francisco J Barrantes
- Instituto de Investigaciones Biomédicas (BIOMED), UCA-CONICET, Buenos Aires, Argentina.
| |
Collapse
|
10
|
Hagerman RJ, Hagerman PJ. Fragile X Syndrome: Lessons Learned and What New Treatment Avenues Are on the Horizon. Annu Rev Pharmacol Toxicol 2021; 62:365-381. [PMID: 34499526 DOI: 10.1146/annurev-pharmtox-052120-090147] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Fragile X syndrome (FXS) is the most common form of inherited intellectual disability and the leading single-gene form of autism spectrum disorder, encompassing cognitive, behavioral, and physical forms of clinical involvement. FXS is caused by large expansions of a noncoding CGG repeat (>200 repeats) in the FMR1 gene, at which point the gene is generally silenced. Absence of FMR1 protein (FMRP), important for synaptic development and maintenance, gives rise to the neurodevelopmental disorder. There is, at present, no therapeutic approach that directly reverses the loss of FMRP; however, there is an increasing number of potential treatments that target the pathways dysregulated in FXS, including those that address the enhanced activity of the mGluR5 pathway and deficits in GABA pathways. Based on studies of targeted therapeutics to date, the prospects are good for one or more effective therapies for FXS in the near future. Expected final online publication date for the Annual Review of Pharmacology and Toxicology, Volume 62 is January 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Randi J Hagerman
- Department of Pediatrics, University of California, Davis, School of Medicine, Sacramento, California 95817, USA; .,MIND Institute, University of California Davis Health, Sacramento, California 95817, USA
| | - Paul J Hagerman
- MIND Institute, University of California Davis Health, Sacramento, California 95817, USA.,Department of Biochemistry and Molecular Medicine, University of California, Davis, School of Medicine, Davis, California 95616, USA;
| |
Collapse
|
11
|
Soorya LV, Fogg L, Ocampo E, Printen M, Youngkin S, Halpern D, Kolevzon A, Lee S, Grodberg D, Anagnostou E. Neurocognitive Outcomes from Memantine: A Pilot, Double-Blind, Placebo-Controlled Trial in Children with Autism Spectrum Disorder. J Child Adolesc Psychopharmacol 2021; 31:475-484. [PMID: 34543081 DOI: 10.1089/cap.2021.0010] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Objective: Studies interrogating therapeutics which alter the excitation-inhibition balance in the treatment of autism spectrum disorder (ASD) have reported mixed results on social and behavioral outcomes. Methods: The aim of this randomized, double-blind placebo-controlled pilot trial was to evaluate neurocognitive effects of memantine over a 24-week trial. Twenty-three children ages 6-12 years old with ASD were randomized to memantine or placebo. Primary outcomes included measures of apraxia and expressive language with evaluations at midpoint (week 12) and endpoint (week 24). Secondary outcomes included memory and adaptive behavior measures. Exploratory outcomes included changes in overall cognitive functioning and behavior (e.g., Aberrant Behavior Checklist). Results: Results suggest that memantine was well-tolerated. Dropout rates were high across groups with only 14 participants completing the 6-month trial. Memantine was not associated with improvements in apraxia and expressive language. Treatment with memantine was associated with improvements in verbal recognition memory as measured by the Narrative Memory-Recognition (NEPSY-II) (F = 5.05, p = .03). In addition, exploratory analyses of changes in Intelligence quotient (IQ) suggest improvements on verbal IQ (d = 1.8). Conclusions: Results suggest future studies of memantine in ASD may benefit from shifting treatment targets from social and behavioral outcomes to exploration of effects of memantine on cognition, potentially as an adjunct to learning and educational interventions. ClinicalTrials.gov: NCT01372449.
Collapse
Affiliation(s)
- Latha Valluripalli Soorya
- Department of Psychiatry and Behavioral Sciences, Rush University Medical Center, Chicago, Illinois, USA
| | - Louis Fogg
- Department of Nursing, Rush University Medical Center, Chicago, Illinois, USA
| | - Edith Ocampo
- Department of Psychiatry and Behavioral Sciences, Rush University Medical Center, Chicago, Illinois, USA
| | - Madison Printen
- Department of Psychiatry and Behavioral Sciences, Rush University Medical Center, Chicago, Illinois, USA
| | - Sarah Youngkin
- Department of Psychiatry and Behavioral Sciences, Rush University Medical Center, Chicago, Illinois, USA
| | - Danielle Halpern
- Department of Psychiatry, Icahn School of Medicine, New York, New York, USA
| | - Alexander Kolevzon
- Department of Psychiatry, Icahn School of Medicine, New York, New York, USA
| | - Soo Lee
- Department of Psychiatry and Behavioral Sciences, Rush University Medical Center, Chicago, Illinois, USA
| | - David Grodberg
- Child Study Center, Yale University, New Haven, Connecticut, USA
| | | |
Collapse
|
12
|
The Role of Taurine in Mitochondria Health: More Than Just an Antioxidant. Molecules 2021; 26:molecules26164913. [PMID: 34443494 PMCID: PMC8400259 DOI: 10.3390/molecules26164913] [Citation(s) in RCA: 96] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 08/08/2021] [Accepted: 08/10/2021] [Indexed: 12/21/2022] Open
Abstract
Taurine is a naturally occurring sulfur-containing amino acid that is found abundantly in excitatory tissues, such as the heart, brain, retina and skeletal muscles. Taurine was first isolated in the 1800s, but not much was known about this molecule until the 1990s. In 1985, taurine was first approved as the treatment among heart failure patients in Japan. Accumulating studies have shown that taurine supplementation also protects against pathologies associated with mitochondrial defects, such as aging, mitochondrial diseases, metabolic syndrome, cancer, cardiovascular diseases and neurological disorders. In this review, we will provide a general overview on the mitochondria biology and the consequence of mitochondrial defects in pathologies. Then, we will discuss the antioxidant action of taurine, particularly in relation to the maintenance of mitochondria function. We will also describe several reported studies on the current use of taurine supplementation in several mitochondria-associated pathologies in humans.
Collapse
|
13
|
Dionne O, Corbin F. A new strategy to uncover fragile X proteomic biomarkers using the nascent proteome of peripheral blood mononuclear cells (PBMCs). Sci Rep 2021; 11:15148. [PMID: 34312401 PMCID: PMC8313568 DOI: 10.1038/s41598-021-94027-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 07/05/2021] [Indexed: 02/06/2023] Open
Abstract
Fragile X syndrome (FXS) is the most prevalent inherited cause of intellectual disabilities and autism spectrum disorders. FXS result from the loss of expression of the FMRP protein, an RNA-binding protein that regulates the expression of key synaptic effectors. FXS is also characterized by a wide array of behavioural, cognitive and metabolic impairments. The severity and penetrance of those comorbidities are extremely variable, meaning that a considerable phenotypic heterogeneity is found among fragile X individuals. Unfortunately, clinicians currently have no tools at their disposal to assay a patient prognosis upon diagnosis. Since the absence of FMRP was repeatedly associated with an aberrant protein synthesis, we decided to study the nascent proteome in order to screen for potential proteomic biomarkers of FXS. We used a BONCAT (Biorthogonal Non-canonical Amino Acids Tagging) method coupled to label-free mass spectrometry to purify and quantify nascent proteins of peripheral blood mononuclear cells (PBMCs) from 7 fragile X male patients and 7 age-matched controls. The proteomic analysis identified several proteins which were either up or downregulated in PBMCs from FXS individuals. Eleven of those proteins were considered as potential biomarkers, of which 5 were further validated by Western blot. The gene ontology enrichment analysis highlighted molecular pathways that may contribute to FXS physiopathology. Our results suggest that the nascent proteome of PBMCs is well suited for the discovery of FXS biomarkers.
Collapse
Affiliation(s)
- Olivier Dionne
- Department of Biochemistry and Functional Genomic, Faculty of Medicine and Health Sciences, Université de Sherbrooke and Centre de Recherche du CHUS, CIUSSS de l'Estrie-CHUS, Sherbrooke, QC, Canada.
| | - François Corbin
- Department of Biochemistry and Functional Genomic, Faculty of Medicine and Health Sciences, Université de Sherbrooke and Centre de Recherche du CHUS, CIUSSS de l'Estrie-CHUS, Sherbrooke, QC, Canada.
| |
Collapse
|
14
|
Dionne O, Corbin F. An "Omic" Overview of Fragile X Syndrome. BIOLOGY 2021; 10:433. [PMID: 34068266 PMCID: PMC8153138 DOI: 10.3390/biology10050433] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 05/01/2021] [Accepted: 05/08/2021] [Indexed: 01/16/2023]
Abstract
Fragile X syndrome (FXS) is a neurodevelopmental disorder associated with a wide range of cognitive, behavioral and medical problems. It arises from the silencing of the fragile X mental retardation 1 (FMR1) gene and, consequently, in the absence of its encoded protein, FMRP (fragile X mental retardation protein). FMRP is a ubiquitously expressed and multifunctional RNA-binding protein, primarily considered as a translational regulator. Pre-clinical studies of the past two decades have therefore focused on this function to relate FMRP's absence to the molecular mechanisms underlying FXS physiopathology. Based on these data, successful pharmacological strategies were developed to rescue fragile X phenotype in animal models. Unfortunately, these results did not translate into humans as clinical trials using same therapeutic approaches did not reach the expected outcomes. These failures highlight the need to put into perspective the different functions of FMRP in order to get a more comprehensive understanding of FXS pathophysiology. This work presents a review of FMRP's involvement on noteworthy molecular mechanisms that may ultimately contribute to various biochemical alterations composing the fragile X phenotype.
Collapse
Affiliation(s)
- Olivier Dionne
- Department of Biochemistry and Functional Genomics, Faculty of Medicine and Health Sciences, Université de Sherbrooke and Centre de Recherche du CHUS, CIUSSS de l’Estrie-CHUS, Sherbrooke, QC J1H 5H4, Canada;
| | | |
Collapse
|
15
|
Salcedo-Arellano MJ, Cabal-Herrera AM, Punatar RH, Clark CJ, Romney CA, Hagerman RJ. Overlapping Molecular Pathways Leading to Autism Spectrum Disorders, Fragile X Syndrome, and Targeted Treatments. Neurotherapeutics 2021; 18:265-283. [PMID: 33215285 PMCID: PMC8116395 DOI: 10.1007/s13311-020-00968-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/04/2020] [Indexed: 02/06/2023] Open
Abstract
Autism spectrum disorders (ASD) are subdivided into idiopathic (unknown) etiology and secondary, based on known etiology. There are hundreds of causes of ASD and most of them are genetic in origin or related to the interplay of genetic etiology and environmental toxicology. Approximately 30 to 50% of the etiologies can be identified when using a combination of available genetic testing. Many of these gene mutations are either core components of the Wnt signaling pathway or their modulators. The full mutation of the fragile X mental retardation 1 (FMR1) gene leads to fragile X syndrome (FXS), the most common cause of monogenic origin of ASD, accounting for ~ 2% of the cases. There is an overlap of molecular mechanisms in those with idiopathic ASD and those with FXS, an interaction between various signaling pathways is suggested during the development of the autistic brain. This review summarizes the cross talk between neurobiological pathways found in ASD and FXS. These signaling pathways are currently under evaluation to target specific treatments in search of the reversal of the molecular abnormalities found in both idiopathic ASD and FXS.
Collapse
Affiliation(s)
- Maria Jimena Salcedo-Arellano
- Department of Pediatrics, University of California Davis School of Medicine, Sacramento, CA, 95817, USA.
- Department of Pathology and Laboratory Medicine, UC Davis School of Medicine, Sacramento, CA, 95817, USA.
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute UCDHS, University of California Davis, 2825 50th Street, Sacramento, CA, 95817, USA.
| | - Ana Maria Cabal-Herrera
- Group on Congenital Malformations and Dysmorphology, Faculty of Health, Universidad del Valle, Cali, 00000, Colombia
| | - Ruchi Harendra Punatar
- Department of Pediatrics, University of California Davis School of Medicine, Sacramento, CA, 95817, USA
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute UCDHS, University of California Davis, 2825 50th Street, Sacramento, CA, 95817, USA
| | - Courtney Jessica Clark
- Department of Pediatrics, University of California Davis School of Medicine, Sacramento, CA, 95817, USA
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute UCDHS, University of California Davis, 2825 50th Street, Sacramento, CA, 95817, USA
| | - Christopher Allen Romney
- Department of Pediatrics, University of California Davis School of Medicine, Sacramento, CA, 95817, USA
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute UCDHS, University of California Davis, 2825 50th Street, Sacramento, CA, 95817, USA
| | - Randi J Hagerman
- Department of Pediatrics, University of California Davis School of Medicine, Sacramento, CA, 95817, USA.
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute UCDHS, University of California Davis, 2825 50th Street, Sacramento, CA, 95817, USA.
| |
Collapse
|
16
|
Verdura E, Pérez-Cano L, Sabido-Vera R, Guney E, Hyvelin JM, Durham L, Gomez-Mancilla B. Heterogeneity in Fragile X Syndrome Highlights the Need for Precision Medicine-Based Treatments. Front Psychiatry 2021; 12:722378. [PMID: 34658958 PMCID: PMC8514715 DOI: 10.3389/fpsyt.2021.722378] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 09/01/2021] [Indexed: 11/21/2022] Open
Abstract
Fragile X syndrome (FXS) is the most frequent monogenic cause of autism or intellectual disability, and research on its pathogenetic mechanisms has provided important insights on this neurodevelopmental condition. Nevertheless, after 30 years of intense research, efforts to develop treatments have been mostly unsuccessful. The aim of this review is to compile evidence from existing research pointing to clinical, genetic, and therapeutic response heterogeneity in FXS and highlight the need of implementing precision medicine-based treatments. We comment on the high genetic and phenotypic heterogeneity present in FXS, as a contributing factor to the difficulties found during drug development. Given that several clinical trials have showed a non-negligeable fraction of positive responders to drugs targeting core FXS symptoms, we propose that success of clinical trials can be achieved by tackling the underlying heterogeneity in FXS by accurately stratifying patients into drug-responder subpopulations. These precision medicine-based approaches, which can be first applied to well-defined monogenic diseases such as FXS, can also serve to define drug responder profiles based on specific biomarkers or phenotypic features that can associate patients with different genetic backgrounds to a same candidate drug, thus repositioning a same drug for a larger number of patients with NDDs.
Collapse
Affiliation(s)
- Edgard Verdura
- Discovery and Data Science (DDS) Unit, Sociedad Limitada (STALICLA SL), Barcelona, Spain
| | - Laura Pérez-Cano
- Discovery and Data Science (DDS) Unit, Sociedad Limitada (STALICLA SL), Barcelona, Spain
| | - Rubén Sabido-Vera
- Discovery and Data Science (DDS) Unit, Sociedad Limitada (STALICLA SL), Barcelona, Spain
| | - Emre Guney
- Discovery and Data Science (DDS) Unit, Sociedad Limitada (STALICLA SL), Barcelona, Spain.,Research Programme on Biomedical Informatics, Hospital del Mar Medical Research Institute (IMIM), Departament de Ciències Experimentals i de la Salut (DCEXS), Pompeu Fabra University (UPF), Barcelona, Spain
| | - Jean-Marc Hyvelin
- Drug Development Unit (DDU), Société Anonyme (STALICLA SA), Geneva, Switzerland
| | - Lynn Durham
- Discovery and Data Science (DDS) Unit, Sociedad Limitada (STALICLA SL), Barcelona, Spain.,Drug Development Unit (DDU), Société Anonyme (STALICLA SA), Geneva, Switzerland
| | - Baltazar Gomez-Mancilla
- Drug Development Unit (DDU), Société Anonyme (STALICLA SA), Geneva, Switzerland.,Department Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| |
Collapse
|
17
|
Glutamatergic Dysfunction and Synaptic Ultrastructural Alterations in Schizophrenia and Autism Spectrum Disorder: Evidence from Human and Rodent Studies. Int J Mol Sci 2020; 22:ijms22010059. [PMID: 33374598 PMCID: PMC7793137 DOI: 10.3390/ijms22010059] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 12/15/2020] [Accepted: 12/22/2020] [Indexed: 12/12/2022] Open
Abstract
The correlation between dysfunction in the glutamatergic system and neuropsychiatric disorders, including schizophrenia and autism spectrum disorder, is undisputed. Both disorders are associated with molecular and ultrastructural alterations that affect synaptic plasticity and thus the molecular and physiological basis of learning and memory. Altered synaptic plasticity, accompanied by changes in protein synthesis and trafficking of postsynaptic proteins, as well as structural modifications of excitatory synapses, are critically involved in the postnatal development of the mammalian nervous system. In this review, we summarize glutamatergic alterations and ultrastructural changes in synapses in schizophrenia and autism spectrum disorder of genetic or drug-related origin, and briefly comment on the possible reversibility of these neuropsychiatric disorders in the light of findings in regular synaptic physiology.
Collapse
|
18
|
Improving the Diagnosis of Autism Spectrum Disorder in Fragile X Syndrome by Adapting the Social Communication Questionnaire and the Social Responsiveness Scale-2. J Autism Dev Disord 2020; 50:3276-3295. [PMID: 31342442 DOI: 10.1007/s10803-019-04148-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
We carried out a psychometric assessment of the Social Communication Questionnaire (SCQ) and the Social Responsiveness Scale (SRS-2) in fragile X syndrome (FXS), relative to clinician DSM5-based diagnosis of autism spectrum disorder (ASD) in FXS. This was followed by instrument revisions that included: removal of non-discriminating and/or low face validity items for FXS; use of receiver operating characteristic (ROC) curves to determine optimal cut points for the original and revised measures; an exploratory factor analysis to outline subscales better representing ASD in FXS; and creation of a "triple criteria" diagnosis to better delineate ASD subgroups in FXS. These methods improved the sensitivity and/or specificity of the SCQ and SRS-2, but diagnostic accuracy of ASD remains problematic in FXS.
Collapse
|
19
|
Recent Advances in the Pharmacological Management of Behavioral Disturbances Associated with Autism Spectrum Disorder in Children and Adolescents. Paediatr Drugs 2020; 22:473-483. [PMID: 32686015 DOI: 10.1007/s40272-020-00408-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Autism spectrum disorder (ASD) is a heterogeneous neuropsychiatric condition affecting an estimated one in 36 children. Youth with ASD may have severe behavioral disturbances including irritability, aggression, and hyperactivity. Currently, there are only two medications (risperidone and aripiprazole) approved by the US Food and Drug Administration (FDA) for the treatment of irritability associated with ASD. Pharmacologic treatments are commonly used to target ASD-associated symptoms including irritability, mood lability, anxiety, and hyperactivity. However, evidence for the efficacy of many commonly used treatments is limited by the lack of large placebo-controlled trials of these medications in this population. Research into the pathophysiology of ASD has led to new targets for pharmacologic therapy including the neuroimmune system, the endocannabinoid system, and the glutamatergic neurotransmitter system. The goal of this review is to provide an overview of the current evidence base for commonly used treatments, as well as emerging treatment options for common behavioral disturbances seen in youth with ASD.
Collapse
|
20
|
Telias M. Pharmacological Treatments for Fragile X Syndrome Based on Synaptic Dysfunction. Curr Pharm Des 2020; 25:4394-4404. [PMID: 31682210 DOI: 10.2174/1381612825666191102165206] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Accepted: 10/31/2019] [Indexed: 12/29/2022]
Abstract
BACKGROUND Fragile X syndrome (FXS) is the most common form of monogenic hereditary cognitive impairment, including intellectual disability, autism, hyperactivity, and epilepsy. METHODS This article reviews the literature pertaining to the role of synaptic dysfunction in FXS. RESULTS In FXS, synaptic dysfunction alters the excitation-inhibition ratio, dysregulating molecular and cellular processes underlying cognition, learning, memory, and social behavior. Decades of research have yielded important hypotheses that could explain, at least in part, the development of these neurological disorders in FXS patients. However, the main goal of translating lab research in animal models to pharmacological treatments in the clinic has been so far largely unsuccessful, leaving FXS a still incurable disease. CONCLUSION In this concise review, we summarize and analyze the main hypotheses proposed to explain synaptic dysregulation in FXS, by reviewing the scientific evidence that led to pharmaceutical clinical trials and their outcome.
Collapse
Affiliation(s)
- Michael Telias
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA, United States
| |
Collapse
|
21
|
Zafarullah M, Tassone F. Molecular Biomarkers in Fragile X Syndrome. Brain Sci 2019; 9:E96. [PMID: 31035599 PMCID: PMC6562871 DOI: 10.3390/brainsci9050096] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 04/22/2019] [Accepted: 04/24/2019] [Indexed: 01/01/2023] Open
Abstract
Fragile X syndrome (FXS) is the most common inherited form of intellectual disability (ID) and a known monogenic cause of autism spectrum disorder (ASD). It is a trinucleotide repeat disorder, in which more than 200 CGG repeats in the 5' untranslated region (UTR) of the fragile X mental retardation 1 (FMR1) gene causes methylation of the promoter with consequent silencing of the gene, ultimately leading to the loss of the encoded fragile X mental retardation 1 protein, FMRP. FMRP is an RNA binding protein that plays a primary role as a repressor of translation of various mRNAs, many of which are involved in the maintenance and development of neuronal synaptic function and plasticity. In addition to intellectual disability, patients with FXS face several behavioral challenges, including anxiety, hyperactivity, seizures, repetitive behavior, and problems with executive and language performance. Currently, there is no cure or approved medication for the treatment of the underlying causes of FXS, but in the past few years, our knowledge about the proteins and pathways that are dysregulated by the loss of FMRP has increased, leading to clinical trials and to the path of developing molecular biomarkers for identifying potential targets for therapies. In this paper, we review candidate molecular biomarkers that have been identified in preclinical studies in the FXS mouse animal model and are now under validation for human applications or have already made their way to clinical trials.
Collapse
Affiliation(s)
- Marwa Zafarullah
- Department of Biochemistry and Molecular Medicine, University of California Davis, School of Medicine, Sacramento, 95817 CA, USA.
| | - Flora Tassone
- Department of Biochemistry and Molecular Medicine, University of California Davis, School of Medicine, Sacramento, 95817 CA, USA.
- MIND Institute, University of California Davis Medical Center, Sacramento, 95817 CA, USA.
| |
Collapse
|
22
|
Altered steady state and activity-dependent de novo protein expression in fragile X syndrome. Nat Commun 2019; 10:1710. [PMID: 30979884 PMCID: PMC6461708 DOI: 10.1038/s41467-019-09553-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 03/15/2019] [Indexed: 12/22/2022] Open
Abstract
Whether fragile X mental retardation protein (FMRP) target mRNAs and neuronal activity contributing to elevated basal neuronal protein synthesis in fragile X syndrome (FXS) is unclear. Our proteomic experiments reveal that the de novo translational profile in FXS model mice is altered at steady state and in response to metabotropic glutamate receptor (mGluR) stimulation, but the proteins expressed differ under these conditions. Several altered proteins, including Hexokinase 1 and Ras, also are expressed in the blood of FXS model mice and pharmacological treatments previously reported to ameliorate phenotypes modify their abundance in blood. In addition, plasma levels of Hexokinase 1 and Ras differ between FXS patients and healthy volunteers. Our data suggest that brain-based de novo proteomics in FXS model mice can be used to find altered expression of proteins in blood that could serve as disease-state biomarkers in individuals with FXS. Elevated protein synthesis, and dysregulated mGluR signalling, are documented in fragile X syndrome (FXS) Here the authors use proteomic analysis in a mouse model of FXS, and following mGluR5 stimulation, to identify potential biomarkers for the disease.
Collapse
|
23
|
Jalnapurkar I, Cochran DM, Frazier JA. New Therapeutic Options for Fragile X Syndrome. Curr Treat Options Neurol 2019; 21:12. [PMID: 30809735 DOI: 10.1007/s11940-019-0551-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
PURPOSE OF REVIEW The purpose of this review is to provide an overview of current research and clinical practice guidelines in fragile X syndrome (FXS) with regard to therapeutic approaches in the management of this condition. The authors summarize and discuss findings from relevant preclinical studies and results from clinical trials in human subjects with FXS. Additionally, we provide an outline of the basic framework for understanding and providing educational and psychosocial supports for these individuals. RECENT FINDINGS Current treatments in FXS are largely symptom based and focused on managing associated psychiatric and behavioral co-morbidities. While data from animal studies has been promising in providing targeted treatments to correct the underlying deficits at the cellular level, there have not been as robust findings in human trials. There are several targeted treatments for FXS currently under development. Individuals with FXS present with several behavioral challenges including anxiety, social withdrawal, ADHD, hyperarousal, self-injury, and aggression. Therapeutic services are often necessary, such as behavioral intervention, speech and language therapy, occupational therapy, and individualized educational support; adjunctive psychopharmacologic treatment is often helpful as well. It is important to address these symptoms and weigh the evidence for the use of medications that target the underlying neurobiology and pathophysiology of the syndrome.
Collapse
Affiliation(s)
- Isha Jalnapurkar
- Eunice Kennedy Shriver Center, University of Massachusetts Medical School, 55 Lake Ave N, Worcester, MA, 01655, USA. .,Department of Psychiatry, University of Massachusetts Medical School, 55 Lake Ave N, Worcester, MA, 01655, USA.
| | - David M Cochran
- Eunice Kennedy Shriver Center, University of Massachusetts Medical School, 55 Lake Ave N, Worcester, MA, 01655, USA.,Department of Psychiatry, University of Massachusetts Medical School, 55 Lake Ave N, Worcester, MA, 01655, USA
| | - Jean A Frazier
- Eunice Kennedy Shriver Center, University of Massachusetts Medical School, 55 Lake Ave N, Worcester, MA, 01655, USA.,Department of Psychiatry, University of Massachusetts Medical School, 55 Lake Ave N, Worcester, MA, 01655, USA
| |
Collapse
|
24
|
Mooney LN, Dominick KC, Erickson CA. Psychopharmacology of neurobehavioral disorders. HANDBOOK OF CLINICAL NEUROLOGY 2019; 165:383-390. [PMID: 31727225 DOI: 10.1016/b978-0-444-64012-3.00023-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
At times psychotropic drug use is required to address behavioral and other interfering symptoms that accompany neurobehavioral disorders. We review such prescribing practice in autism spectrum disorder, fragile X syndrome, and Prader-Willi syndrome.
Collapse
Affiliation(s)
- Lindsey N Mooney
- Department of Psychiatry, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Kelli C Dominick
- Department of Psychiatry, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Craig A Erickson
- Department of Psychiatry, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States.
| |
Collapse
|
25
|
Lamy M, Erickson CA. Pharmacological management of behavioral disturbances in children and adolescents with autism spectrum disorders. Curr Probl Pediatr Adolesc Health Care 2018; 48:250-264. [PMID: 30262163 DOI: 10.1016/j.cppeds.2018.08.015] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Autism spectrum disorder (ASD) is a heterogeneous neuropsychiatric condition that, based on recent CDC estimates affects an estimated 1 in 59 American children. Behavioral treatments remain the mainstay of treatment for the core symptoms of ASD including communication deficits, social interaction deficits and repetitive behavior. However, youth with ASD may also have severe behavioral challenges including irritability, aggression, and hyperactivity. Currently there are only two medications (risperidone and aripiprazole) approved by the FDA for the treatment of irritability associated with ASD in children. Psychiatric comorbidities are common in youth with ASD, affecting up to 70% of affected children and adolescents. Given the burden of co-occurring disorders, medications are often employed to target symptoms such as irritability, anxiety, and hyperactivity. Other common co-occurring conditions including gastrointestinal disorders and sleep disorders may be improved with pharmacologic management. Evidence for the efficacy of many commonly used psychotropic medications in ASD is limited by the lack of large placebo-controlled trials in youth with ASD. This paper reviews the current literature regarding use of medications to address co-occurring conditions in children and adolescents with ASD as well as areas of emerging research.
Collapse
Affiliation(s)
- Martine Lamy
- Division of Child and Adolescent Psychiatry, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, United States.
| | - Craig A Erickson
- Division of Child and Adolescent Psychiatry, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, United States
| |
Collapse
|
26
|
Melancia F, Trezza V. Modelling fragile X syndrome in the laboratory setting: A behavioral perspective. Behav Brain Res 2018; 350:149-163. [DOI: 10.1016/j.bbr.2018.04.042] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 04/24/2018] [Accepted: 04/24/2018] [Indexed: 12/13/2022]
|
27
|
Fragile X syndrome and fragile X-associated tremor ataxia syndrome. HANDBOOK OF CLINICAL NEUROLOGY 2018; 147:377-391. [PMID: 29325626 DOI: 10.1016/b978-0-444-63233-3.00025-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Fragile X-associated disorders encompass several conditions, which are caused by expansion mutations in the fragile X mental retardation 1 (FMR1) gene. Fragile X syndrome is the most common inherited etiology of intellectual disability and results from a full mutation or >200 CGG repeats in FMR1. It is associated with developmental delay, autism spectrum disorder, and seizures. Fragile X-associated tremor/ataxia syndrome is a progressive neurodegenerative disease that occurs in premutation carriers of 55-200 CGG repeats in FMR1 and is characterized by kinetic tremor, gait ataxia, parkinsonism, executive dysfunction, and neuropathy. Fragile X-associated primary ovarian insufficiency also occurs in premutation carrier women and manifests with infertility and early menopause. The diseases constituting fragile X-associated disorders differ mechanistically, due to the distinct molecular properties of premutation versus full mutations. Fragile X syndrome occurs when there is a lack of fragile X mental retardation protein (FMRP) due to FMR1 methylation and silencing. In fragile X-associated tremor ataxia syndrome, a toxic gain of function is postulated with the production of excess CGG repeat-containing FMR1 mRNA, abnormal translation of the repeat sequence leading to production of polyglycine, polyalanine, and other polypeptides and to outright deficits in translation leading to reduced FMRP at larger premutation sizes. The changes in underlying brain chemistry due to FMR1 mutations have led to therapeutic studies in these disorders, with some progress being made in fragile X syndrome. This paper also summarizes indications for testing, genetic counseling issues, and what the future holds for these disorders.
Collapse
|
28
|
Tranfaglia MR, Thibodeaux C, Mason DJ, Brown D, Roberts I, Smith R, Guilliams T, Cogram P. Repurposing available drugs for neurodevelopmental disorders: The fragile X experience. Neuropharmacology 2018; 147:74-86. [PMID: 29792283 DOI: 10.1016/j.neuropharm.2018.05.004] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 04/30/2018] [Accepted: 05/02/2018] [Indexed: 10/17/2022]
Abstract
Many available drugs have been repurposed as treatments for neurodevelopmental disorders. In the specific case of fragile X syndrome, many clinical trials of available drugs have been conducted with the goal of disease modification. In some cases, detailed understanding of basic disease mechanisms has guided the choice of drugs for clinical trials, and several notable successes in fragile X clinical trials have led to common use of drugs such as minocycline in routine medical practice. Newer technologies like Disease-Gene Expression Matching (DGEM) may allow for more rapid identification of promising repurposing candidates. A DGEM study predicted that sulindac could be therapeutic for fragile X, and subsequent preclinical validation studies have shown promising results. The use of combinations of available drugs and nutraceuticals has the potential to greatly expand the options for repurposing, and may even be a viable business strategy. This article is part of the Special Issue entitled 'Drug Repurposing: old molecules, new ways to fast track drug discovery and development for CNS disorders'.
Collapse
Affiliation(s)
| | - Clare Thibodeaux
- Cures Within Reach, 125 S. Clark Street, 17th Floor, Chicago, IL 60603, USA.
| | - Daniel J Mason
- Healx Ltd., Park House, Castle Park, Cambridge, CB3 0DU, United Kingdom.
| | - David Brown
- Healx Ltd., Park House, Castle Park, Cambridge, CB3 0DU, United Kingdom
| | - Ian Roberts
- Healx Ltd., Park House, Castle Park, Cambridge, CB3 0DU, United Kingdom
| | - Richard Smith
- Healx Ltd., Park House, Castle Park, Cambridge, CB3 0DU, United Kingdom
| | - Tim Guilliams
- Healx Ltd., Park House, Castle Park, Cambridge, CB3 0DU, United Kingdom
| | - Patricia Cogram
- FRAXA-DVI, IEB, Las Encinas 3370, Ñuñoa, Santiago, Chile; Laboratory of Molecular Neuropsychiatry, Institute of Cognitive and Translational Neuroscience (INCyT), INECO Foundation, Favaloro University, National Scientific and Technical Research Council (CONICET), Pacheco de Melo 1854, CP 1126, Ciudad de Buenos Aires, Argentina; Institute of Ecology and Biodiversity, Faculty of Science, University of Chile, Las Palmeras 3425, Ñuñoa, Santiago, Chile.
| |
Collapse
|
29
|
Hutson RL, Thompson RL, Bantel AP, Tessier CR. Acamprosate rescues neuronal defects in the Drosophila model of Fragile X Syndrome. Life Sci 2018; 195:65-70. [PMID: 29317220 DOI: 10.1016/j.lfs.2018.01.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 01/04/2018] [Accepted: 01/05/2018] [Indexed: 10/18/2022]
Abstract
AIMS Several off-label studies have shown that acamprosate can provide some clinical benefits in youth with Fragile X Syndrome (FXS), an autism spectrum disorder caused by loss of function of the highly conserved FMR1 gene. This study investigated the ability of acamprosate to rescue cellular, molecular and behavioral defects in the Drosophila model of FXS. MAIN METHODS A high (100μM) and low (10μM) dose of acamprosate was fed to Drosophila FXS (dfmr1 null) or genetic control (w1118) larvae and then analyzed in multiple paradigms. A larval crawling assay was used to monitor aberrant FXS behavior, overgrowth of the neuromuscular junction (NMJ) was quantified to assess neuronal development, and quantitative RT-PCR was used to evaluate expression of deregulated cbp53E mRNA. KEY FINDINGS Acamprosate treatment partially or completely rescued all of the FXS phenotypes analyzed, according to dose. High doses rescued cellular overgrowth and dysregulated cbp53E mRNA expression, but aberrant crawling behavior was not affected. Low doses of acamprosate, however, did not affect synapse number at the NMJ, but could rescue NMJ overgrowth, locomotor defects, and cbp53E mRNA expression. This dual nature of acamprosate suggests multiple molecular mechanisms may be involved in acamprosate function depending on the dosage used. SIGNIFICANCE Acamprosate may be a useful therapy for FXS and potentially other autism spectrum disorders. However, understanding the molecular mechanisms involved with different doses of this drug will likely be necessary to obtain optimal results.
Collapse
Affiliation(s)
- Russell L Hutson
- Department of Biological Sciences, University of Notre Dame, South Bend, IN, United States
| | - Rachel L Thompson
- Department of Biological Sciences, University of Notre Dame, South Bend, IN, United States
| | - Andrew P Bantel
- Department of Medical and Molecular Genetics, Indiana University School of Medicine-South Bend, South Bend, IN, United States
| | - Charles R Tessier
- Department of Medical and Molecular Genetics, Indiana University School of Medicine-South Bend, South Bend, IN, United States.
| |
Collapse
|
30
|
Drug development for neurodevelopmental disorders: lessons learned from fragile X syndrome. Nat Rev Drug Discov 2017; 17:280-299. [PMID: 29217836 DOI: 10.1038/nrd.2017.221] [Citation(s) in RCA: 236] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Neurodevelopmental disorders such as fragile X syndrome (FXS) result in lifelong cognitive and behavioural deficits and represent a major public health burden. FXS is the most frequent monogenic form of intellectual disability and autism, and the underlying pathophysiology linked to its causal gene, FMR1, has been the focus of intense research. Key alterations in synaptic function thought to underlie this neurodevelopmental disorder have been characterized and rescued in animal models of FXS using genetic and pharmacological approaches. These robust preclinical findings have led to the implementation of the most comprehensive drug development programme undertaken thus far for a genetically defined neurodevelopmental disorder, including phase IIb trials of metabotropic glutamate receptor 5 (mGluR5) antagonists and a phase III trial of a GABAB receptor agonist. However, none of the trials has been able to unambiguously demonstrate efficacy, and they have also highlighted the extent of the knowledge gaps in drug development for FXS and other neurodevelopmental disorders. In this Review, we examine potential issues in the previous studies and future directions for preclinical and clinical trials. FXS is at the forefront of efforts to develop drugs for neurodevelopmental disorders, and lessons learned in the process will also be important for such disorders.
Collapse
|
31
|
Munshi K, Pawlowski K, Gonzalez-Heydrich J, Picker JD. Review of Salient Investigational Drugs for the Treatment of Fragile X Syndrome. J Child Adolesc Psychopharmacol 2017; 27:850-863. [PMID: 28475355 DOI: 10.1089/cap.2016.0200] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
OBJECTIVES Fragile X syndrome (FXS) is the most common inherited cause of intellectual disability, in addition to being the commonest diagnosable cause of autism. The identification of the biochemical mechanism underlying this disorder has provided amenable targets for therapy. This review aims to provide an overview of investigational drug therapies for FXS. METHODS The authors carried out a search of clinical and preclinical trials for FXS in PubMed and on the U.S. National Institutes of Health index of clinical trials ( www.clinicaltrials.gov ). We limited our review to Phase II trials or more preliminary and reviewed the associated publications for these studies, complemented by a review of the literature on PubMed. RESULTS The review of the preclinical, Phase I, and Phase II trials of agents with therapeutic potential in FXS revolves around an understanding of the putative pathways in the pathogenesis of FXS. While there is significant overlap between some of these pathways, the agents can be categorized as modulators of the metabotropic glutamate receptor system, GABAergic agents, and miscellaneous modulators affecting other pathways. CONCLUSION As trials involving agents targeting different aspects of the molecular biology proceed, common themes have emerged. With the great hope came great disappointment as the initial trials failed to demonstrate sufficient significance. In particular, the differences in outcome between the animal models and humans have highlighted the unique challenges of carrying out trials in these cognitively and behaviorally challenged individuals, as well as a dearth of clinically relevant outcome measures for use in medication trials. However, in reviewing and reframing the studies of the last decade, many important lessons have been learned, which will ultimately have a greater impact on therapeutic research in the field of developmental delay as a whole.
Collapse
Affiliation(s)
- Kaizad Munshi
- 1 Department of Psychiatry, Boston Children's Hospital , Boston, Massachusetts.,2 Harvard Medical School , Boston, Massachusetts
| | - Katherine Pawlowski
- 3 Division of Genetics and Genomics, Boston Children's Hospital , Boston, Massachusetts.,4 Division of Developmental Medicine, Department of Medicine, Boston Children's Hospital , Boston, Massachusetts
| | - Joseph Gonzalez-Heydrich
- 1 Department of Psychiatry, Boston Children's Hospital , Boston, Massachusetts.,2 Harvard Medical School , Boston, Massachusetts
| | - Jonathan D Picker
- 1 Department of Psychiatry, Boston Children's Hospital , Boston, Massachusetts.,2 Harvard Medical School , Boston, Massachusetts.,3 Division of Genetics and Genomics, Boston Children's Hospital , Boston, Massachusetts
| |
Collapse
|
32
|
Chahrour M, Kleiman RJ, Manzini MC. Translating genetic and preclinical findings into autism therapies. DIALOGUES IN CLINICAL NEUROSCIENCE 2017. [PMID: 29398929 PMCID: PMC5789211 DOI: 10.31887/dcns.2017.19.4/cmanzini] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Autism spectrum disorder (ASD) is a complex neurodevelopmental disorder characterized by social deficits and repetitive/restrictive interests. ASD is associated with multiple comorbidities, including intellectual disability, anxiety, and epilepsy. Evidence that ASD is highly heritable has spurred major efforts to unravel its genetics, revealing possible contributions from hundreds of genes through rare and common variation and through copy-number changes. In this perspective, we provide an overview of the current state of ASD genetics and of how genetic research has spurred the development of in vivo and in vitro models using animals and patient cells to evaluate the impact of genetic mutations on cellular function leading to disease. Efforts to translate these findings into successful therapies have yet to bear fruit. We discuss how the valuable insight into the disorder provided by these new models can be used to better understand ASD and develop future clinical trials.
Collapse
Affiliation(s)
- Maria Chahrour
- Eugene McDermott Center for Human Growth and Development, Departments of Neuroscience and Psychiatry, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | | | - M Chiara Manzini
- Institute for Neuroscience, Autism and Neurodevelopmental Disorders Institute, and Department of Pharmacology and Physiology, The George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| |
Collapse
|
33
|
Abstract
Fragile X syndrome (FXS) is the leading inherited form of intellectual disability and autism spectrum disorder, and patients can present with severe behavioural alterations, including hyperactivity, impulsivity and anxiety, in addition to poor language development and seizures. FXS is a trinucleotide repeat disorder, in which >200 repeats of the CGG motif in FMR1 leads to silencing of the gene and the consequent loss of its product, fragile X mental retardation 1 protein (FMRP). FMRP has a central role in gene expression and regulates the translation of potentially hundreds of mRNAs, many of which are involved in the development and maintenance of neuronal synaptic connections. Indeed, disturbances in neuroplasticity is a key finding in FXS animal models, and an imbalance in inhibitory and excitatory neuronal circuits is believed to underlie many of the clinical manifestations of this disorder. Our knowledge of the proteins that are regulated by FMRP is rapidly growing, and this has led to the identification of multiple targets for therapeutic intervention, some of which have already moved into clinical trials or clinical practice.
Collapse
|
34
|
Benke D, Möhler H. Impact on GABA systems in monogenetic developmental CNS disorders: Clues to symptomatic treatment. Neuropharmacology 2017; 136:46-55. [PMID: 28764992 DOI: 10.1016/j.neuropharm.2017.07.030] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 07/26/2017] [Accepted: 07/28/2017] [Indexed: 12/26/2022]
Abstract
Animal studies of several single-gene disorders demonstrate that reversing the molecular signaling deficits can result in substantial symptomatic improvements in function. Focusing on the ratio of excitation to inhibition as a potential pathophysiological hallmark, seven single-gene developmental CNS disorders are reviewed which are characterized by a striking dysregulation of neuronal inhibition. Deficits in inhibition and excessive inhibition are found. The examples of developmental disorders encompass Neurofibromatosis type 1, Fragile X syndrome, Rett syndrome, Dravet syndrome including autism-like behavior, NONO-mutation-induced intellectual disability, Succinic semialdehyde dehydrogenase deficiency and Congenital nystagmus due to FRMD7 mutations. The phenotype/genotype correlations observed in animal models point to potential treatment options and will continue to inspire clinical research. Three drugs are presently in clinical trials: acamprosate and ganoxolon for Fragile X syndrome and SGS-742 for SSADH deficiency. This article is part of the "Special Issue Dedicated to Norman G. Bowery".
Collapse
Affiliation(s)
- Dietmar Benke
- Institute of Pharmacology and Toxicology, University of Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland; Neuroscience Center Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland; Drug Discovery Network Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland.
| | - Hanns Möhler
- Institute of Pharmacology and Toxicology, University of Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland; Neuroscience Center Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland; Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology (ETH), Vladimir-Prelog-Weg 10, 8023 Zurich, Switzerland.
| |
Collapse
|
35
|
McClellan L, Dominick KC, Pedapati EV, Wink LK, Erickson CA. Lurasidone for the treatment of irritability and anger in autism spectrum disorders. Expert Opin Investig Drugs 2017; 26:985-989. [DOI: 10.1080/13543784.2017.1353600] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Lynn McClellan
- Division of Child and Adolescent Psychiatry, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Kelli C. Dominick
- Division of Child and Adolescent Psychiatry, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Ernest V. Pedapati
- Division of Child and Adolescent Psychiatry, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Logan K. Wink
- Division of Child and Adolescent Psychiatry, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Craig A. Erickson
- Division of Child and Adolescent Psychiatry, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| |
Collapse
|
36
|
Erickson CA, Davenport MH, Schaefer TL, Wink LK, Pedapati EV, Sweeney JA, Fitzpatrick SE, Brown WT, Budimirovic D, Hagerman RJ, Hessl D, Kaufmann WE, Berry-Kravis E. Fragile X targeted pharmacotherapy: lessons learned and future directions. J Neurodev Disord 2017; 9:7. [PMID: 28616096 PMCID: PMC5467059 DOI: 10.1186/s11689-017-9186-9] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Accepted: 01/18/2017] [Indexed: 01/04/2023] Open
Abstract
Our understanding of fragile X syndrome (FXS) pathophysiology continues to improve and numerous potential drug targets have been identified. Yet, current prescribing practices are only symptom-based in order to manage difficult behaviors, as no drug to date is approved for the treatment of FXS. Drugs impacting a diversity of targets in the brain have been studied in recent FXS-specific clinical trials. While many drugs have focused on regulation of enhanced glutamatergic or deficient GABAergic neurotransmission, compounds studied have not been limited to these mechanisms. As a single-gene disorder, it was thought that FXS would have consistent drug targets that could be modulated with pharmacotherapy and lead to significant improvement. Unfortunately, despite promising results in FXS animal models, translational drug treatment development in FXS has largely failed. Future success in this field will depend on learning from past challenges to improve clinical trial design, choose appropriate outcome measures and age range choices, and find readily modulated drug targets. Even with many negative placebo-controlled study results, the field continues to move forward exploring both the new mechanistic drug approaches combined with ways to improve trial execution. This review summarizes the known phenotype and pathophysiology of FXS and past clinical trial rationale and results, and discusses current challenges facing the field and lessons from which to learn for future treatment development efforts.
Collapse
Affiliation(s)
- Craig A Erickson
- Division of Child and Adolescent Psychiatry (MLC 4002), Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave., Cincinnati, OH 45229-3039 USA.,Department of Psychiatry, College of Medicine, University of Cincinnati, Cincinnati, OH USA
| | - Matthew H Davenport
- Division of Child and Adolescent Psychiatry (MLC 4002), Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave., Cincinnati, OH 45229-3039 USA.,Department of Biomedical Engineering, College of Engineering and Applied Science, University of Cincinnati, Cincinnati, OH USA
| | - Tori L Schaefer
- Division of Child and Adolescent Psychiatry (MLC 4002), Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave., Cincinnati, OH 45229-3039 USA
| | - Logan K Wink
- Division of Child and Adolescent Psychiatry (MLC 4002), Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave., Cincinnati, OH 45229-3039 USA.,Department of Psychiatry, College of Medicine, University of Cincinnati, Cincinnati, OH USA
| | - Ernest V Pedapati
- Division of Child and Adolescent Psychiatry (MLC 4002), Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave., Cincinnati, OH 45229-3039 USA.,Department of Psychiatry, College of Medicine, University of Cincinnati, Cincinnati, OH USA
| | - John A Sweeney
- Department of Psychiatry, College of Medicine, University of Cincinnati, Cincinnati, OH USA
| | - Sarah E Fitzpatrick
- Division of Child and Adolescent Psychiatry (MLC 4002), Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave., Cincinnati, OH 45229-3039 USA
| | - W Ted Brown
- Institute for Basic Research in Developmental Disabilities, New York, NY USA
| | - Dejan Budimirovic
- Clinical Research Center, Clinical Trials Unit, Fragile X Clinic, Kennedy Krieger Institute, The Johns Hopkins Medical Institutions, Baltimore, MD USA.,Departments of Psychiatry & Behavioral Sciences, Child Psychiatry, The Johns Hopkins Medical Institutions, Baltimore, MD USA
| | - Randi J Hagerman
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, Davis Medical Center, University of California, Sacramento, CA USA.,Department of Pediatrics, Davis Medical Center, University of California, Sacramento, California USA
| | - David Hessl
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, Davis Medical Center, University of California, Sacramento, CA USA.,Department of Psychiatry and Behavioral Sciences, Davis Medical Center, University of California, Sacramento, California USA
| | - Walter E Kaufmann
- Greenwood Genetic Center, Greenwood, SC USA.,Boston Children's Hospital, Boston, Massachusetts USA
| | - Elizabeth Berry-Kravis
- Departments of Pediatrics, Neurological Sciences, Biochemistry, Rush University Medical Center, Chicago, Illinois USA
| |
Collapse
|
37
|
Budimirovic DB, Berry-Kravis E, Erickson CA, Hall SS, Hessl D, Reiss AL, King MK, Abbeduto L, Kaufmann WE. Updated report on tools to measure outcomes of clinical trials in fragile X syndrome. J Neurodev Disord 2017; 9:14. [PMID: 28616097 PMCID: PMC5467057 DOI: 10.1186/s11689-017-9193-x] [Citation(s) in RCA: 114] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 02/22/2017] [Indexed: 12/27/2022] Open
Abstract
OBJECTIVE Fragile X syndrome (FXS) has been the neurodevelopmental disorder with the most active translation of preclinical breakthroughs into clinical trials. This process has led to a critical assessment of outcome measures, which resulted in a comprehensive review published in 2013. Nevertheless, the disappointing outcome of several recent phase III drug trials in FXS, and parallel efforts at evaluating behavioral endpoints for trials in autism spectrum disorder (ASD), has emphasized the need for re-assessing outcome measures and revising recommendations for FXS. METHODS After performing an extensive database search (PubMed, Food and Drug Administration (FDA)/National Institutes of Health (NIH)'s www.ClinicalTrials.gov, etc.) to determine progress since 2013, members of the Working Groups who published the 2013 Report evaluated the available outcome measures for FXS and related neurodevelopmental disorders using the COSMIN grading system of levels of evidence. The latter has also been applied to a British survey of endpoints for ASD. In addition, we also generated an informal classification of outcome measures for use in FXS intervention studies as instruments appropriate to detect shorter- or longer-term changes. RESULTS To date, a total of 22 double-blind controlled clinical trials in FXS have been identified through www.ClinicalTrials.gov and an extensive literature search. The vast majority of these FDA/NIH-registered clinical trials has been completed between 2008 and 2015 and has targeted the core excitatory/inhibitory imbalance present in FXS and other neurodevelopmental disorders. Limited data exist on reliability and validity for most tools used to measure cognitive, behavioral, and other problems in FXS in these trials and other studies. Overall, evidence for most tools supports a moderate tool quality grading. Data on sensitivity to treatment, currently under evaluation, could improve ratings for some cognitive and behavioral tools. Some progress has also been made at identifying promising biomarkers, mainly on blood-based and neurophysiological measures. CONCLUSION Despite the tangible progress in implementing clinical trials in FXS, the increasing data on measurement properties of endpoints, and the ongoing process of new tool development, the vast majority of outcome measures are at the moderate quality level with limited information on reliability, validity, and sensitivity to treatment. This situation is not unique to FXS, since reviews of endpoints for ASD have arrived at similar conclusions. These findings, in conjunction with the predominance of parent-based measures particularly in the behavioral domain, indicate that endpoint development in FXS needs to continue with an emphasis on more objective measures (observational, direct testing, biomarkers) that reflect meaningful improvements in quality of life. A major continuous challenge is the development of measurement tools concurrently with testing drug safety and efficacy in clinical trials.
Collapse
Affiliation(s)
- Dejan B. Budimirovic
- Departments of Psychiatry and Behavioral Sciences, Kennedy Krieger Institute and Child Psychiatry, Johns Hopkins University School of Medicine, 716 N. Broadway, Baltimore, MD 21205 USA
| | - Elizabeth Berry-Kravis
- Departments of Pediatrics, Neurological Sciences, Biochemistry, Rush University Medical Center, 1725 West Harrison, Suite 718, Chicago, IL 60612 USA
| | - Craig A. Erickson
- Division of Child and Adolescent Psychiatry, Cincinnati Children’s Hospital Medical Center and the University of Cincinnati College of Medicine, 3333 Burnet Avenue MLC 4002, Cincinnati, OH 45229 USA
| | - Scott S. Hall
- Division of Interdisciplinary Brain Sciences, Department of Psychiatry and Behavioral Sciences, Stanford University, 401 Quarry Road, Stanford, CA 94305 USA
| | - David Hessl
- MIND Institute and Department of Psychiatry and Behavioral Sciences, University of California Davis Medical Center, 2825 50th Street, Sacramento, CA 95817 USA
| | - Allan L. Reiss
- Division of Interdisciplinary Brain Sciences, Departments of Psychiatry and Behavioral Sciences, Radiology and Pediatrics, Stanford University, 401 Quarry Road, Stanford, CA 94305 USA
| | - Margaret K. King
- Autism & Developmental Medicine Institute, Geisinger Health System, Present address: Novartis Pharmaceuticals Corporation, US Medical, One Health Plaza, East Hanover, NJ 07936 USA
| | - Leonard Abbeduto
- MIND Institute and Department of Psychiatry and Behavioral Sciences, University of California Davis Medical Center, 2825 50th Street, Sacramento, CA 95817 USA
| | - Walter E. Kaufmann
- Center for Translational Research, Greenwood Genetic Center, 113 Gregor Mendel Circle, Greenwood, SC 29646 USA
- Department of Neurology, Boston Children’s Hospital, Boston, MA 02115 USA
| |
Collapse
|
38
|
Schaefer TL, Davenport MH, Grainger LM, Robinson CK, Earnheart AT, Stegman MS, Lang AL, Ashworth AA, Molinaro G, Huber KM, Erickson CA. Acamprosate in a mouse model of fragile X syndrome: modulation of spontaneous cortical activity, ERK1/2 activation, locomotor behavior, and anxiety. J Neurodev Disord 2017; 9:6. [PMID: 28616095 PMCID: PMC5467053 DOI: 10.1186/s11689-017-9184-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Accepted: 01/13/2017] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Fragile X Syndrome (FXS) occurs as a result of a silenced fragile X mental retardation 1 gene (FMR1) and subsequent loss of fragile X mental retardation protein (FMRP) expression. Loss of FMRP alters excitatory/inhibitory signaling balance, leading to increased neuronal hyperexcitability and altered behavior. Acamprosate (the calcium salt of N-acetylhomotaurinate), a drug FDA-approved for relapse prevention in the treatment of alcohol dependence in adults, is a novel agent with multiple mechanisms that may be beneficial for people with FXS. There are questions regarding the neuroactive effects of acamprosate and the significance of the molecule's calcium moiety. Therefore, the electrophysiological, cellular, molecular, and behavioral effects of acamprosate were assessed in the Fmr1-/y (knock out; KO) mouse model of FXS controlling for the calcium salt in several experiments. METHODS Fmr1 KO mice and their wild-type (WT) littermates were utilized to assess acamprosate treatment on cortical UP state parameters, dendritic spine density, and seizure susceptibility. Brain extracellular-signal regulated kinase 1/2 (ERK1/2) activation was used to investigate this signaling molecule as a potential biomarker for treatment response. Additional adult mice were used to assess chronic acamprosate treatment and any potential effects of the calcium moiety using CaCl2 treatment on behavior and nuclear ERK1/2 activation. RESULTS Acamprosate attenuated prolonged cortical UP state duration, decreased elevated ERK1/2 activation in brain tissue, and reduced nuclear ERK1/2 activation in the dentate gyrus in KO mice. Acamprosate treatment modified behavior in anxiety and locomotor tests in Fmr1 KO mice in which control-treated KO mice were shown to deviate from control-treated WT mice. Mice treated with CaCl2 were not different from saline-treated mice in the adult behavior battery or nuclear ERK1/2 activation. CONCLUSIONS These data indicate that acamprosate, and not calcium, improves function reminiscent of reduced anxiety-like behavior and hyperactivity in Fmr1 KO mice and that acamprosate attenuates select electrophysiological and molecular dysregulation that may play a role in the pathophysiology of FXS. Differences between control-treated KO and WT mice were not evident in a recognition memory test or in examination of acoustic startle response/prepulse inhibition which impeded conclusions from being made about the treatment effects of acamprosate in these instances.
Collapse
Affiliation(s)
- Tori L Schaefer
- Division of Psychiatry, MLC 7004, Cincinnati Children's Research Foundation, 3333 Burnet Ave., Cincinnati, OH 45229-3039 USA
| | - Matthew H Davenport
- Division of Psychiatry, MLC 7004, Cincinnati Children's Research Foundation, 3333 Burnet Ave., Cincinnati, OH 45229-3039 USA
| | - Lindsay M Grainger
- Division of Psychiatry, MLC 7004, Cincinnati Children's Research Foundation, 3333 Burnet Ave., Cincinnati, OH 45229-3039 USA
| | - Chandler K Robinson
- Division of Psychiatry, MLC 7004, Cincinnati Children's Research Foundation, 3333 Burnet Ave., Cincinnati, OH 45229-3039 USA
| | - Anthony T Earnheart
- Division of Psychiatry, MLC 7004, Cincinnati Children's Research Foundation, 3333 Burnet Ave., Cincinnati, OH 45229-3039 USA
| | - Melinda S Stegman
- Division of Psychiatry, MLC 7004, Cincinnati Children's Research Foundation, 3333 Burnet Ave., Cincinnati, OH 45229-3039 USA.,Present address: Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229 USA
| | - Anna L Lang
- Division of Psychiatry, MLC 7004, Cincinnati Children's Research Foundation, 3333 Burnet Ave., Cincinnati, OH 45229-3039 USA.,Present address: Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY 40202 USA
| | - Amy A Ashworth
- Division of Psychiatry, MLC 7004, Cincinnati Children's Research Foundation, 3333 Burnet Ave., Cincinnati, OH 45229-3039 USA.,Present address: BlackbookHR, Cincinnati, OH 45202 USA
| | - Gemma Molinaro
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390 USA
| | - Kimberly M Huber
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390 USA
| | - Craig A Erickson
- Division of Psychiatry, MLC 7004, Cincinnati Children's Research Foundation, 3333 Burnet Ave., Cincinnati, OH 45229-3039 USA
| |
Collapse
|
39
|
AlOlaby RR, Sweha SR, Silva M, Durbin-Johnson B, Yrigollen CM, Pretto D, Hagerman RJ, Tassone F. Molecular biomarkers predictive of sertraline treatment response in young children with fragile X syndrome. Brain Dev 2017; 39:483-492. [PMID: 28242040 PMCID: PMC5420478 DOI: 10.1016/j.braindev.2017.01.012] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Revised: 01/26/2017] [Accepted: 01/27/2017] [Indexed: 12/28/2022]
Abstract
OBJECTIVES Several neurotransmitters involved in brain development are altered in fragile X syndrome (FXS), the most common monogenic cause of autism spectrum disorder (ASD). Serotonin plays a vital role in synaptogenesis and postnatal brain development. Deficits in serotonin synthesis and abnormal neurogenesis were shown in young children with autism, suggesting that treating within the first years of life with a selective serotonin reuptake inhibitor might be the most effective time. In this study we aimed to identify molecular biomarkers involved in the serotonergic pathway that could predict the response to sertraline treatment in young children with FXS. METHODS Genotypes were determined for several genes involved in serotonergic pathway in 51 children with FXS, ages 24-72months. Correlations between genotypes and deviations from baseline in primary and secondary outcome measures were modeled using linear regression models. RESULTS A significant association was observed between a BDNF polymorphism and improvements for several clinical measures, including the Clinical Global Impression scale (P=0.008) and the cognitive T score (P=0.017) in those treated with sertraline compared to those in the placebo group. Additionally, polymorphisms in the MAOA, Cytochrome P450 2C19 and 2D6, and in the 5-HTTLPR gene showed a significant correlation with some of the secondary measures included in this study. CONCLUSION This study shows that polymorphisms of genes involved in the serotonergic pathway could play a potential role in predicting response to sertraline treatment in young children with FXS. Larger studies are warranted to confirm these initial findings.
Collapse
Affiliation(s)
- Reem Rafik AlOlaby
- Department of Biochemistry and Molecular Medicine, University of California, Davis, School of Medicine, Davis, CA, USA
| | - Stefan R Sweha
- Department of Biochemistry and Molecular Medicine, University of California, Davis, School of Medicine, Davis, CA, USA
| | - Marisol Silva
- Department of Biochemistry and Molecular Medicine, University of California, Davis, School of Medicine, Davis, CA, USA
| | - Blythe Durbin-Johnson
- Department of Biostatistics, University of California Davis, School of Medicine, Davis, CA, USA
| | - Carolyn M Yrigollen
- Department of Biochemistry and Molecular Medicine, University of California, Davis, School of Medicine, Davis, CA, USA
| | - Dalyir Pretto
- Department of Biochemistry and Molecular Medicine, University of California, Davis, School of Medicine, Davis, CA, USA
| | - Randi J Hagerman
- MIND Institute, University of California Davis Medical Center, Davis, CA, USA,Department of Pediatrics, University of California Davis, School of Medicine, Davis, CA, USA
| | - Flora Tassone
- Department of Biochemistry and Molecular Medicine, University of California, Davis, School of Medicine, Davis, CA, USA; MIND Institute, University of California Davis Medical Center, Davis, CA, USA.
| |
Collapse
|
40
|
Jensen KB, Kirsch I, Pontén M, Rosén A, Yang K, Gollub RL, des Portes V, Kaptchuk TJ, Curie A. Certainty of genuine treatment increases drug responses among intellectually disabled patients. Neurology 2017; 88:1912-1918. [PMID: 28424273 PMCID: PMC5444309 DOI: 10.1212/wnl.0000000000003934] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2016] [Accepted: 02/28/2017] [Indexed: 02/06/2023] Open
Abstract
Objective: To determine the placebo component of treatment responses in patients with intellectual disability (ID). Methods: A statistical meta-analysis comparing bias-corrected effect sizes (Hedges g) of drug responses in open-label vs placebo-controlled clinical trials was performed, as these trial types represent different certainty of receiving genuine treatment (100% vs 50%). Studies in fragile X, Down, Prader-Willi, and Williams syndrome published before June 2015 were considered. Results: Seventeen open-label trials (n = 261, 65% male; mean age 23.6 years; mean trial duration 38 weeks) and 22 placebo-controlled trials (n = 721, 62% male; mean age 17.1 years; mean trial duration 35 weeks) were included. The overall effect size from pre to post treatment in open-label studies was g = 0.602 (p = 0.001). The effect of trial type was statistically significant (p = 0.001), and revealed higher effect sizes in studies with 100% likelihood of getting active drug, compared to both the drug and placebo arm of placebo-controlled trials. We thus provide evidence for genuine placebo effects, not explainable by natural history or regression toward the mean, among patients with ID. Conclusions: Our data suggest that clinical trials in patients with severe cognitive deficits are influenced by the certainty of receiving genuine medication, and open-label design should thus not be used to evaluate the effect of pharmacologic treatments in ID, as the results will be biased by an enhanced placebo component.
Collapse
Affiliation(s)
- Karin B Jensen
- From the Department of Clinical Neuroscience (K.B.J., M.P., A.R.), Karolinska Institute, Sweden; Program in Placebo Studies (I.K., T.J.K.), BIDMC, Harvard Medical School; Department of Psychiatry (K.Y., R.L.G.), Massachusetts General Hospital, Boston; Institut des Sciences Cognitives (V.d.P., A.C.), Bron; Université Claude Bernard Lyon 1 (V.d.P., A.C.); Centre de Référence Déficiences Intellectuelles de Causes Rares (V.d.P., A.C.), Hôpital Femmes Mères Enfants, Hospices Civils de Lyon; and EPICIME-CIC1407/INSERM (A.C.), Bron, France.
| | - Irving Kirsch
- From the Department of Clinical Neuroscience (K.B.J., M.P., A.R.), Karolinska Institute, Sweden; Program in Placebo Studies (I.K., T.J.K.), BIDMC, Harvard Medical School; Department of Psychiatry (K.Y., R.L.G.), Massachusetts General Hospital, Boston; Institut des Sciences Cognitives (V.d.P., A.C.), Bron; Université Claude Bernard Lyon 1 (V.d.P., A.C.); Centre de Référence Déficiences Intellectuelles de Causes Rares (V.d.P., A.C.), Hôpital Femmes Mères Enfants, Hospices Civils de Lyon; and EPICIME-CIC1407/INSERM (A.C.), Bron, France
| | - Moa Pontén
- From the Department of Clinical Neuroscience (K.B.J., M.P., A.R.), Karolinska Institute, Sweden; Program in Placebo Studies (I.K., T.J.K.), BIDMC, Harvard Medical School; Department of Psychiatry (K.Y., R.L.G.), Massachusetts General Hospital, Boston; Institut des Sciences Cognitives (V.d.P., A.C.), Bron; Université Claude Bernard Lyon 1 (V.d.P., A.C.); Centre de Référence Déficiences Intellectuelles de Causes Rares (V.d.P., A.C.), Hôpital Femmes Mères Enfants, Hospices Civils de Lyon; and EPICIME-CIC1407/INSERM (A.C.), Bron, France
| | - Annelie Rosén
- From the Department of Clinical Neuroscience (K.B.J., M.P., A.R.), Karolinska Institute, Sweden; Program in Placebo Studies (I.K., T.J.K.), BIDMC, Harvard Medical School; Department of Psychiatry (K.Y., R.L.G.), Massachusetts General Hospital, Boston; Institut des Sciences Cognitives (V.d.P., A.C.), Bron; Université Claude Bernard Lyon 1 (V.d.P., A.C.); Centre de Référence Déficiences Intellectuelles de Causes Rares (V.d.P., A.C.), Hôpital Femmes Mères Enfants, Hospices Civils de Lyon; and EPICIME-CIC1407/INSERM (A.C.), Bron, France
| | - Kathy Yang
- From the Department of Clinical Neuroscience (K.B.J., M.P., A.R.), Karolinska Institute, Sweden; Program in Placebo Studies (I.K., T.J.K.), BIDMC, Harvard Medical School; Department of Psychiatry (K.Y., R.L.G.), Massachusetts General Hospital, Boston; Institut des Sciences Cognitives (V.d.P., A.C.), Bron; Université Claude Bernard Lyon 1 (V.d.P., A.C.); Centre de Référence Déficiences Intellectuelles de Causes Rares (V.d.P., A.C.), Hôpital Femmes Mères Enfants, Hospices Civils de Lyon; and EPICIME-CIC1407/INSERM (A.C.), Bron, France
| | - Randy L Gollub
- From the Department of Clinical Neuroscience (K.B.J., M.P., A.R.), Karolinska Institute, Sweden; Program in Placebo Studies (I.K., T.J.K.), BIDMC, Harvard Medical School; Department of Psychiatry (K.Y., R.L.G.), Massachusetts General Hospital, Boston; Institut des Sciences Cognitives (V.d.P., A.C.), Bron; Université Claude Bernard Lyon 1 (V.d.P., A.C.); Centre de Référence Déficiences Intellectuelles de Causes Rares (V.d.P., A.C.), Hôpital Femmes Mères Enfants, Hospices Civils de Lyon; and EPICIME-CIC1407/INSERM (A.C.), Bron, France
| | - Vincent des Portes
- From the Department of Clinical Neuroscience (K.B.J., M.P., A.R.), Karolinska Institute, Sweden; Program in Placebo Studies (I.K., T.J.K.), BIDMC, Harvard Medical School; Department of Psychiatry (K.Y., R.L.G.), Massachusetts General Hospital, Boston; Institut des Sciences Cognitives (V.d.P., A.C.), Bron; Université Claude Bernard Lyon 1 (V.d.P., A.C.); Centre de Référence Déficiences Intellectuelles de Causes Rares (V.d.P., A.C.), Hôpital Femmes Mères Enfants, Hospices Civils de Lyon; and EPICIME-CIC1407/INSERM (A.C.), Bron, France
| | - Ted J Kaptchuk
- From the Department of Clinical Neuroscience (K.B.J., M.P., A.R.), Karolinska Institute, Sweden; Program in Placebo Studies (I.K., T.J.K.), BIDMC, Harvard Medical School; Department of Psychiatry (K.Y., R.L.G.), Massachusetts General Hospital, Boston; Institut des Sciences Cognitives (V.d.P., A.C.), Bron; Université Claude Bernard Lyon 1 (V.d.P., A.C.); Centre de Référence Déficiences Intellectuelles de Causes Rares (V.d.P., A.C.), Hôpital Femmes Mères Enfants, Hospices Civils de Lyon; and EPICIME-CIC1407/INSERM (A.C.), Bron, France
| | - Aurore Curie
- From the Department of Clinical Neuroscience (K.B.J., M.P., A.R.), Karolinska Institute, Sweden; Program in Placebo Studies (I.K., T.J.K.), BIDMC, Harvard Medical School; Department of Psychiatry (K.Y., R.L.G.), Massachusetts General Hospital, Boston; Institut des Sciences Cognitives (V.d.P., A.C.), Bron; Université Claude Bernard Lyon 1 (V.d.P., A.C.); Centre de Référence Déficiences Intellectuelles de Causes Rares (V.d.P., A.C.), Hôpital Femmes Mères Enfants, Hospices Civils de Lyon; and EPICIME-CIC1407/INSERM (A.C.), Bron, France
| |
Collapse
|
41
|
Singh R, Turner RC, Nguyen L, Motwani K, Swatek M, Lucke-Wold BP. Pediatric Traumatic Brain Injury and Autism: Elucidating Shared Mechanisms. Behav Neurol 2016; 2016:8781725. [PMID: 28074078 PMCID: PMC5198096 DOI: 10.1155/2016/8781725] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2016] [Accepted: 11/23/2016] [Indexed: 02/08/2023] Open
Abstract
Pediatric traumatic brain injury (TBI) and autism spectrum disorder (ASD) are two serious conditions that affect youth. Recent data, both preclinical and clinical, show that pediatric TBI and ASD share not only similar symptoms but also some of the same biologic mechanisms that cause these symptoms. Prominent symptoms for both disorders include gastrointestinal problems, learning difficulties, seizures, and sensory processing disruption. In this review, we highlight some of these shared mechanisms in order to discuss potential treatment options that might be applied for each condition. We discuss potential therapeutic and pharmacologic options as well as potential novel drug targets. Furthermore, we highlight advances in understanding of brain circuitry that is being propelled by improved imaging modalities. Going forward, advanced imaging will help in diagnosis and treatment planning strategies for pediatric patients. Lessons from each field can be applied to design better and more rigorous trials that can be used to improve guidelines for pediatric patients suffering from TBI or ASD.
Collapse
Affiliation(s)
- Rahul Singh
- Department of Neurosurgery, West Virginia University School of Medicine, Morgantown, WV 26505, USA
| | - Ryan C. Turner
- Department of Neurosurgery, West Virginia University School of Medicine, Morgantown, WV 26505, USA
| | - Linda Nguyen
- Department of Basic Pharmaceutical Sciences, West Virginia University School of Medicine, Morgantown, WV 26505, USA
| | - Kartik Motwani
- Department of Medical Sciences, University of Florida School of Medicine, Gainesville, FL 32611, USA
| | - Michelle Swatek
- Department of Psychology, North Carolina State University, Raleigh, NC 27695, USA
| | - Brandon P. Lucke-Wold
- Department of Neurosurgery, West Virginia University School of Medicine, Morgantown, WV 26505, USA
| |
Collapse
|
42
|
Davenport MH, Schaefer TL, Friedmann KJ, Fitzpatrick SE, Erickson CA. Pharmacotherapy for Fragile X Syndrome: Progress to Date. Drugs 2016; 76:431-45. [PMID: 26858239 DOI: 10.1007/s40265-016-0542-y] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
To date, no drug is approved for the treatment of Fragile X Syndrome (FXS) although many drugs are used to manage challenging behaviors from a symptomatic perspective in this population. While our understanding of FXS pathophysiology is expanding, efforts to devise targeted FXS-specific treatments have had limited success in placebo-controlled trials. Compounds aimed at rectifying excessive glutamate and deficient gamma-aminobutyric acid (GABA) neurotransmission, as well as other signaling pathways known to be affected by Fragile X Mental Retardation Protein (FMRP) are under various phases of development in FXS. With the failure of several metabotropic glutamate receptor subtype 5 (mGlur5) selective antagonists under clinical investigation, no clear single treatment appears to be greatly effective. These recent challenges call into question various aspects of clinical study design in FXS. More objective outcome measures are under development and validation. Future trials will likely be aimed at correcting multiple pathways known to be disrupted by the loss of FMRP. This review offers a brief summary of the prevalence, phenotypic characteristics, genetic causes and molecular functions of FMRP in the brain (as these have been extensively reviewed elsewhere), discusses the most recent finding in FXS drug development, and summarizes FXS trials utilizing symptomatic treatment.
Collapse
Affiliation(s)
- Matthew H Davenport
- Division of Child and Adolescent Psychiatry (MLC 4002), Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave., Cincinnati, OH, 45229-3039, USA
- Department of Biomedical Engineering, College of Engineering and Applied Science, University of Cincinnati, Cincinnati, OH, 45221, USA
| | - Tori L Schaefer
- Division of Child and Adolescent Psychiatry (MLC 4002), Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave., Cincinnati, OH, 45229-3039, USA
| | - Katherine J Friedmann
- Division of Child and Adolescent Psychiatry (MLC 4002), Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave., Cincinnati, OH, 45229-3039, USA
| | | | - Craig A Erickson
- Division of Child and Adolescent Psychiatry (MLC 4002), Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave., Cincinnati, OH, 45229-3039, USA.
| |
Collapse
|
43
|
Brondino N, Fusar-Poli L, Panisi C, Damiani S, Barale F, Politi P. Pharmacological Modulation of GABA Function in Autism Spectrum Disorders: A Systematic Review of Human Studies. J Autism Dev Disord 2016; 46:825-39. [PMID: 26443675 DOI: 10.1007/s10803-015-2619-y] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Autism spectrum disorders are an emerging health problem worldwide, but little is known about their pathogenesis. It has been hypothesized that autism may result from an imbalance between excitatory glutamatergic and inhibitory GABAergic pathways. Commonly used medications such as valproate, acamprosate, and arbaclofen may act on the GABAergic system and be a potential treatment for people with ASD. The present systematic review aimed at evaluating the state-of-the-art of clinical trials of GABA modulators in autism. To date there is insufficient evidence to suggest the use of these drugs in autistic subjects, even if data are promising. Of note, short-term use of all the reviewed medications appears to be safe. Future well designed trials are needed to elucidate these preliminary findings.
Collapse
Affiliation(s)
- Natascia Brondino
- Department of Brain and Behavioral Sciences, University of Pavia, Via Bassi 21, 27100, Pavia, Italy.
| | - Laura Fusar-Poli
- Department of Brain and Behavioral Sciences, University of Pavia, Via Bassi 21, 27100, Pavia, Italy
| | - Cristina Panisi
- Department of Brain and Behavioral Sciences, University of Pavia, Via Bassi 21, 27100, Pavia, Italy
| | - Stefano Damiani
- Department of Brain and Behavioral Sciences, University of Pavia, Via Bassi 21, 27100, Pavia, Italy
| | - Francesco Barale
- Department of Brain and Behavioral Sciences, University of Pavia, Via Bassi 21, 27100, Pavia, Italy
| | - Pierluigi Politi
- Department of Brain and Behavioral Sciences, University of Pavia, Via Bassi 21, 27100, Pavia, Italy
| |
Collapse
|
44
|
Lozano R, Martinez-Cerdeno V, Hagerman RJ. Advances in the Understanding of the Gabaergic Neurobiology of FMR1 Expanded Alleles Leading to Targeted Treatments for Fragile X Spectrum Disorder. Curr Pharm Des 2016; 21:4972-4979. [PMID: 26365141 DOI: 10.2174/1381612821666150914121038] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 09/11/2015] [Indexed: 12/15/2022]
Abstract
Fragile X spectrum disorder (FXSD) includes: fragile X syndrome (FXS), fragile X-associated tremor ataxia syndrome (FXTAS) and fragile X-associated primary ovarian insufficiency (FXPOI), as well as other medical, psychiatric and neurobehavioral problems associated with the premutation and gray zone alleles. FXS is the most common monogenetic cause of autism (ASD) and intellectual disability (ID). The understanding of the neurobiology of FXS has led to many targeted treatment trials in FXS. The first wave of phase II clinical trials in FXS were designed to target the mGluR5 pathway; however the results did not show significant efficacy and the trials were terminated. The advances in the understanding of the GABA system in FXS have shifted the focus of treatment trials to GABA agonists, and a new wave of promising clinical trials is under way. Ganaxolone and allopregnanolone (GABA agonists) have been studied in individuals with FXSD and are currently in phase II trials. Both allopregnanolone and ganaxolone may be efficacious in treatment of FXS and FXTAS, respectively. Allopregnanolone, ganaxolone, riluzole, gaboxadol, tiagabine, and vigabatrin are potential GABAergic treatments. The lessons learned from the initial trials have not only shifted the targeted system, but also have refined the design of clinical trials. The results of these new trials will likely impact further clinical trials for FXS and other genetic disorders associated with ASD.
Collapse
Affiliation(s)
- Reymundo Lozano
- Icahn School of Medicine at Mount Sinai, New York, NY USA; Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY USA; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY USA; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY USA; Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Veronica Martinez-Cerdeno
- Medical Investigation of Neurodevelopmental Disorders MIND Institute, UC Davis, CA, USA; Institute for Pediatric Regenerative Medicine and Shriners Hospital for Children of Northern California, Sacramento, CA, USA; Department of Pathology and Laboratory Medicine, UC Davis, Sacramento, USA
| | - Randi J Hagerman
- Medical Investigation of Neurodevelopmental Disorders MIND Institute, UC Davis, CA, USA; Department of Pediatrics, UC Davis, Sacramento, CA, USA
| |
Collapse
|
45
|
Abstract
Fragile X syndrome (FXS) is the most common inherited form of intellectual disability, and is the leading single-gene cause of autism spectrum disorders. It is due to a loss of the fragile X mental retardation protein, which leads to molecular, behavioral, and cognitive deficits in these patients. Improvements in our understanding of its pathophysiology have led to the development of numerous targeted treatments in FXS as highlighted by metabotropic glutamate receptor antagonists and gamma-Aminobutyric acid receptor modulators. This review will summarize relevant pre-clinical data and results from clinical trials in human subjects with FXS. It will also highlight upcoming studies and future directions for clinical trials as well.
Collapse
Affiliation(s)
- Andrew Ligsay
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, University of California, Davis Medical Center, Sacramento, CA, USA
- University of California, Davis School of Medicine, Sacramento, CA, USA
| | - Randi J Hagerman
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, University of California, Davis Medical Center, Sacramento, CA, USA
- Department of Pediatrics, University of California, Davis Medical Center, Sacramento, CA, USA
- Address correspondence to: Dr. Randi J Hagerman, Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, University of California, Davis Medical Center, 2825 50th Street, Sacramento, CA 95817, USA. E-mail:
| |
Collapse
|
46
|
Salcedo-Arellano MJ, Lozano R, Tassone F, Hagerman RJ, Saldarriaga W. Alcohol use dependence in fragile X syndrome. Intractable Rare Dis Res 2016; 5:207-13. [PMID: 27672544 PMCID: PMC4995423 DOI: 10.5582/irdr.2016.01046] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 06/28/2016] [Accepted: 06/29/2016] [Indexed: 11/05/2022] Open
Abstract
Alcohol use disorders (AUDs) have been reported in a limited number of individuals with cognitive impairment but rarely in those with fragile X syndrome (FXS). However, in Colombia, culturally, alcohol consumption is very common. Here, we report eight cases of patients with FXS who have frequent alcohol consumption in Ricaurte, Colombia. Some of these patients have also used tobacco and illegal substances, including cocaine, which use has not been previously reported in those with FXS. Alcohol and substance use dependence is associated with exacerbation of their behavioral problems, such as increased impulsivity and aggression, as well as of medical problems such as an increased frequency of seizures.
Collapse
Affiliation(s)
- María J Salcedo-Arellano
- School of Medicine, Universidad del Valle, Cali, Colombia
- Research Group in Congenital & Perinatal Malformations, Dysmorphology and Clinical Genetics (MACOS), Universidad del Valle, Cali, Colombia
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, University of California, Davis Medical Center, Sacramento, CA, USA
| | - Reymundo Lozano
- Seaver Autism Center. Departments of Genomic Sciences, Psychiatry and Pediatrics. Icahn School of Medicine, Mount Sinai Hospital, New York, NY, USA
| | - Flora Tassone
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, University of California, Davis Medical Center, Sacramento, CA, USA
- Department of Biochemistry and Molecular Medicine, University of California, Davis, USA
| | - Randi J Hagerman
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, University of California, Davis Medical Center, Sacramento, CA, USA
- Department of Pediatrics, University of California, Davis Medical Center, Sacramento, CA, USA
| | - Wilmar Saldarriaga
- School of Medicine, Universidad del Valle, Cali, Colombia
- Research Group in Congenital & Perinatal Malformations, Dysmorphology and Clinical Genetics (MACOS), Universidad del Valle, Cali, Colombia
- Departments of Morphology and Obstetrics & Gynecology, Universidad del Valle, Hospital Universitario Del Valle, Cali, Colombia
| |
Collapse
|
47
|
Luo SY, Wu LQ, Duan RH. Molecular medicine of fragile X syndrome: based on known molecular mechanisms. World J Pediatr 2016; 12:19-27. [PMID: 26547211 DOI: 10.1007/s12519-015-0052-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Accepted: 08/25/2014] [Indexed: 11/29/2022]
Abstract
BACKGROUND Extensive research on fragile X mental retardation gene knockout mice and mutant Drosophila models has largely expanded our knowledge on mechanism-based treatment of fragile X syndrome (FXS). In light of these findings, several clinical trials are now underway for therapeutic translation to humans. DATA SOURCES Electronic literature searches were conducted using the PubMed database and ClinicalTrials.gov. The search terms included "fragile X syndrome", "FXS and medication", "FXS and therapeutics" and "FXS and treatment". Based on the publications identified in this search, we reviewed the neuroanatomical abnormalities in FXS patients and the potential pathogenic mechanisms to monitor the progress of FXS research, from basic studies to clinical trials. RESULTS The pathological mechanisms of FXS were categorized on the basis of neuroanatomy, synaptic structure, synaptic transmission and fragile X mental retardation protein (FMRP) loss of function. The neuroanatomical abnormalities in FXS were described to motivate extensive research into the region-specific pathologies in the brain responsible for FXS behavioural manifestations. Mechanism-directed molecular medicines were classified according to their target pathological mechanisms, and the most recent progress in clinical trials was discussed. CONCLUSIONS Current mechanism-based studies and clinical trials have greatly contributed to the development of FXS pharmacological therapeutics. Research examining the extent to which these treatments provided a rescue effect or FMRP compensation for the developmental impairments in FXS patients may help to improve the efficacy of treatments.
Collapse
Affiliation(s)
- Shi-Yu Luo
- State Key Laboratory of Medical Genetics & School of Life Sciences, Central South University, Changsha, China
| | - Ling-Qian Wu
- State Key Laboratory of Medical Genetics & School of Life Sciences, Central South University, Changsha, China
| | - Ran-Hui Duan
- State Key Laboratory of Medical Genetics & School of Life Sciences, Central South University, Changsha, China.
| |
Collapse
|
48
|
Kazdoba TM, Leach PT, Yang M, Silverman JL, Solomon M, Crawley JN. Translational Mouse Models of Autism: Advancing Toward Pharmacological Therapeutics. Curr Top Behav Neurosci 2016; 28:1-52. [PMID: 27305922 PMCID: PMC5116923 DOI: 10.1007/7854_2015_5003] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Animal models provide preclinical tools to investigate the causal role of genetic mutations and environmental factors in the etiology of autism spectrum disorder (ASD). Knockout and humanized knock-in mice, and more recently knockout rats, have been generated for many of the de novo single gene mutations and copy number variants (CNVs) detected in ASD and comorbid neurodevelopmental disorders. Mouse models incorporating genetic and environmental manipulations have been employed for preclinical testing of hypothesis-driven pharmacological targets, to begin to develop treatments for the diagnostic and associated symptoms of autism. In this review, we summarize rodent behavioral assays relevant to the core features of autism, preclinical and clinical evaluations of pharmacological interventions, and strategies to improve the translational value of rodent models of autism.
Collapse
Affiliation(s)
- Tatiana M Kazdoba
- MIND Institute, Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Room 1001A Research 2 Building 96, 4625 2nd Avenue, Sacramento, CA 95817, USA
| | - Prescott T Leach
- MIND Institute, Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Room 1001A Research 2 Building 96, 4625 2nd Avenue, Sacramento, CA 95817, USA
| | - Mu Yang
- MIND Institute, Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Room 1001A Research 2 Building 96, 4625 2nd Avenue, Sacramento, CA 95817, USA
| | - Jill L Silverman
- MIND Institute, Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Room 1001A Research 2 Building 96, 4625 2nd Avenue, Sacramento, CA 95817, USA
| | - Marjorie Solomon
- MIND Institute, Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Room 1001A Research 2 Building 96, 4625 2nd Avenue, Sacramento, CA 95817, USA
| | - Jacqueline N Crawley
- MIND Institute, Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Room 1001A Research 2 Building 96, 4625 2nd Avenue, Sacramento, CA 95817, USA.
| |
Collapse
|
49
|
Gross C, Hoffmann A, Bassell GJ, Berry-Kravis EM. Therapeutic Strategies in Fragile X Syndrome: From Bench to Bedside and Back. Neurotherapeutics 2015; 12:584-608. [PMID: 25986746 PMCID: PMC4489963 DOI: 10.1007/s13311-015-0355-9] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Fragile X syndrome (FXS), an inherited intellectual disability often associated with autism, is caused by the loss of expression of the fragile X mental retardation protein. Tremendous progress in basic, preclinical, and translational clinical research has elucidated a variety of molecular-, cellular-, and system-level defects in FXS. This has led to the development of several promising therapeutic strategies, some of which have been tested in larger-scale controlled clinical trials. Here, we will summarize recent advances in understanding molecular functions of fragile X mental retardation protein beyond the well-known role as an mRNA-binding protein, and will describe current developments and emerging limitations in the use of the FXS mouse model as a preclinical tool to identify therapeutic targets. We will review the results of recent clinical trials conducted in FXS that were based on some of the preclinical findings, and discuss how the observed outcomes and obstacles will inform future therapy development in FXS and other autism spectrum disorders.
Collapse
Affiliation(s)
- Christina Gross
- />Division of Neurology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229 USA
| | - Anne Hoffmann
- />Department of Pediatrics, Rush University Medical Center, Chicago, IL 60612 USA
| | - Gary J. Bassell
- />Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322 USA
| | - Elizabeth M. Berry-Kravis
- />Departments of Pediatrics, Neurological Sciences, Biochemistry, Rush University Medical Center, Chicago, IL 60612 USA
| |
Collapse
|
50
|
Schaefer TL, Davenport MH, Erickson CA. Emerging pharmacologic treatment options for fragile X syndrome. APPLICATION OF CLINICAL GENETICS 2015; 8:75-93. [PMID: 25897255 PMCID: PMC4396424 DOI: 10.2147/tacg.s35673] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Fragile X syndrome (FXS) is the most common single gene cause of intellectual disability and autism spectrum disorder. Caused by a silenced fragile X mental retardation 1 gene and the subsequent deficiency in fragile X mental retardation protein, patients with FXS experience a range of physical, behavioral, and intellectual debilitations. The FXS field, as a whole, has recently met with some challenges, as several targeted clinical trials with high expectations of success have failed to elucidate significant improvements in a variety of symptom domains. As new clinical trials in FXS are planned, there has been much discussion about the use of the commonly used clinical outcome measures, as well as study design considerations, patient stratification, and optimal age range for treatment. The evidence that modification of these drug targets and use of these failed compounds would prove to be efficacious in human clinical study were rooted in years of basic and translational research. There are questions arising as to the use of the mouse models for studying FXS treatment development. This issue is twofold: many of the symptom domains and molecular and biochemical changes assessed and indicative of efficacy in mouse model study are not easily amenable to clinical trials in people with FXS because of the intolerability of the testing paradigm or a lack of noninvasive techniques (prepulse inhibition, sensory hypersensitivity, startle reactivity, or electrophysiologic, biochemical, or structural changes in the brain); and capturing subtle yet meaningful changes in symptom domains such as sociability, anxiety, and hyperactivity in human FXS clinical trials is challenging with the currently used measures (typically parent/caregiver rating scales). Clinicians, researchers, and the pharmaceutical industry have all had to take a step back and critically evaluate the way we think about how to best optimize future investigations into pharmacologic FXS treatments. As new clinical trials are coming down the drug discovery pipeline, it is clear that the field is moving in a direction that values the development of molecular biomarkers, less subjective quantitative measures of symptom improvement, and rating scales developed specifically for use in FXS in conjunction with drug safety. While summarizing preclinical evidence, where applicable, and discussing challenges in FXS treatment development, this review details both completed clinical trials for the targeted and symptomatic treatment of FXS and introduces novel projects on the cusp of clinical trial investigation.
Collapse
Affiliation(s)
- Tori L Schaefer
- Division of Child and Adolescent Psychiatry, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Matthew H Davenport
- Division of Child and Adolescent Psychiatry, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Craig A Erickson
- Division of Child and Adolescent Psychiatry, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| |
Collapse
|