1
|
McKinney WS, Schmitt LM, De Stefano LA, Ethridge L, Norris JE, Horn PS, Dauterman S, Rosselot H, Pedapati EV, Reisinger DL, Dominick KC, Shaffer RC, Chin D, Friedman NR, Hong M, Sweeney JA, Erickson C. Results from a Double-Blind, Randomized, Placebo-Controlled, Single-Dose, Crossover Trial of Lovastatin or Minocycline in Fragile X Syndrome. J Child Adolesc Psychopharmacol 2024. [PMID: 39651602 DOI: 10.1089/cap.2024.0103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
Introduction: Treatment studies in FMR1 knockout rodent models have found that minocycline and lovastatin each improve synaptic, neurological, and behavioral functioning, and open-label chronic dosing studies in human patients with fragile X syndrome (FXS) have demonstrated modest clinical improvements. Findings from blinded studies are mixed, and there is a limited understanding of electrophysiological target engagement that would facilitate cross-species translational studies. Smaller-scale, acute (e.g., single-dose) drug studies may speed treatment identification by detecting subtle electrophysiological and behavioral changes. Materials and Methods: Twenty-nine participants with FXS (31% female) ages 15-45 years completed a randomized, double-blind, crossover study in which they received a single oral dose of 40 mg of lovastatin, 270 mg of minocycline, or placebo, with a 2-week washout period between dosing visits. Participants completed a comprehensive neuropsychological battery and three EEG paradigms (resting state; auditory chirp; auditory habituation) before and 4 hours after dosing. Results: No serious adverse events were reported, and both drugs were well-tolerated. Compared with placebo, there were no overall treatment effects for any outcomes, including EEG, but several modest drug responses varied as a function of sex and age. Lovastatin treatment was associated with improved spatial awareness in older participants and females compared with minocycline and placebo. Discussion: We show that single-dose drug studies are highly feasible in FXS and that patients with FXS can complete a range of EEG and behavioral tasks, many of which have been shown to be reliable and may therefore be sensitive to subtle drug target engagement. Conclusions: Acute single doses of lovastatin or minocycline did not lead to changes in electrophysiological or performance-based measures. This may be due to the limited effects of these drugs in human patients or limited acute effects relative to chronic dosing. However, the study design was further validated for use in neurodevelopmental populations.
Collapse
Affiliation(s)
- Walker S McKinney
- Department of Behavioral Medicine and Clinical Psychology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Lauren M Schmitt
- Department of Behavioral Medicine and Clinical Psychology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Lisa A De Stefano
- Division of Child and Adolescent Psychiatry, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Lauren Ethridge
- Department of Psychology, University of Oklahoma, Norman, Oklahoma, USA
- Department of Pediatrics, Section of Developmental and Behavioral Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Jordan E Norris
- Department of Psychology, University of Oklahoma, Norman, Oklahoma, USA
| | - Paul S Horn
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Shelby Dauterman
- Division of Child and Adolescent Psychiatry, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Neuroscience Graduate Program, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | | | - Ernest V Pedapati
- Division of Child and Adolescent Psychiatry, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Debra L Reisinger
- Department of Behavioral Medicine and Clinical Psychology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Kelli C Dominick
- Division of Child and Adolescent Psychiatry, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Rebecca C Shaffer
- Department of Behavioral Medicine and Clinical Psychology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Danielle Chin
- The Heidt Center of Excellence, Cincinnati, Ohio, USA
| | - Nicole R Friedman
- Department of Psychology, University of Alabama, Tuscaloosa, Alabama, USA
| | - Michael Hong
- Department of Radiology, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - John A Sweeney
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Craig Erickson
- Division of Child and Adolescent Psychiatry, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| |
Collapse
|
2
|
Kaufmann WE, Luu S, Budimirovic DB. Drug Treatments for Neurodevelopmental Disorders: Targeting Signaling Pathways and Homeostasis. Curr Neurol Neurosci Rep 2024; 25:7. [PMID: 39641900 DOI: 10.1007/s11910-024-01394-3] [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] [Accepted: 10/08/2024] [Indexed: 12/07/2024]
Abstract
PURPOSE OF THE REVIEW Preclinical and clinical evidence support the notion that neurodevelopmental disorders (NDDs) are synaptic disorders, characterized by excitatory-inhibitory imbalance. Despite this, NDD drug development programs targeting glutamate or gamma-aminobutyric acid (GABA) receptors have been largely unsuccessful. Nonetheless, recent drug trials in Rett syndrome (RTT), fragile X syndrome (FXS), and other NDDs targeting other mechanisms have met their endpoints. The purpose of this review is to identify the basis of these successful studies. RECENT FINDINGS Despite increasing evidence of disruption in synaptic homeostasis, most genetic variants associated with NDDs implicate proteins involved in cell regulation and not in neurotransmission. Metabolic processes, in particular mitochondrial function, appear to play a role in NDD pathophysiology. NDDs are also characterized by distinctive cell signaling abnormalities, which link cellular and synaptic homeostasis. Recent successful trials in NDDs, including those of trofinetide, the first drug specifically approved for one of these disorders (i.e., RTT), implicate the targeting of downstream processes (i.e., signaling pathways) rather than neurotransmitter receptors. Recent positive drug studies in NDDs and their underlying mechanisms, in conjunction with new knowledge on the pathophysiology of these disorders, support the concept that targeting signaling and cellular and synaptic homeostasis may be a preferred approach for ameliorating synaptic abnormalities in many NDDs.
Collapse
Affiliation(s)
- Walter E Kaufmann
- Boston Children's Hospital, Boston, MA, 02115, USA.
- Emory University School of Medicine, Atlanta, GA, 30322, USA.
| | - Skylar Luu
- Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Dejan B Budimirovic
- Kennedy Krieger Institute and Department of Psychiatry & Behavioral Sciences-Child Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
3
|
Nelson MA, Schmitt LM, Horn PS, Berry-Kravis E, Hessl D, Shaffer RC, Carpenter R, Budimirovic DB, Wang P, Reisinger DL, Walton-Bowen K, Erickson CA. Parent-Reported Outcome Measures for Individuals with Fragile X Syndrome: Clinically Meaningful Change Thresholds. J Autism Dev Disord 2024:10.1007/s10803-024-06634-6. [PMID: 39579284 DOI: 10.1007/s10803-024-06634-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/29/2024] [Indexed: 11/25/2024]
Abstract
Estimating meaningful change thresholds (MCT) on clinical outcome assessments is an important consideration when evaluating treatments. In fragile X syndrome (FXS) research, there has been no consensus on how to define MCT's on several commonly used outcome measures. The purpose of the current study was to determine clinically relevant MCT's of caregiver-rated assessments using data from a phase 3 clinical trials of arbaclofen (Berry-Kravis et al., 2017). Data were collected as a part of previous phase 3, double-blind, placebo-controlled studies of arbaclofen in individuals with FXS (Berry-Kravis et al., 2017). The two studies enrolled age groups of 5-11-years (n = 159) and 12-50-years (n = 119). The current study examines meaningful within-patient change thresholds from baseline to treatment week 8 across several measures: ABC-CFXS; PSI; Vineland-II; and a Visual Analog Scale (VAS) of Anxiety and Disruptive Behaviors. MCT's were established by using anchor-based methods, using the CGI-S and CGI-I as anchors. Examining the results of the anchor-based analyses and visual CDF plots, MCT's were observed for the pediatric study for the ABC-CFXS subscales (with a range depending on use of CGI-S or CGI-I as anchor): Irritability: 11.1-14.8 points; Hyperactivity: 6.7-8.9 points; and Socially Unresponsive/Lethargic: 6.6-8.1 points; as well both VAS subscales: Anxiety: 28.3-36.2 mm; and Disruptive Behavior: 22.4-27.4 mm. Such thresholds were not observed for the Vineland-II and PSI subscales. Our analysis of MCT's helps set the stage for interpreting clinical trial results in FXS. This may include use of relevant subscales of the ABC-CFXS and VAS as primary outcomes using the MCT's for response definition. This work may help define future study inclusion criteria and enable future interpretation of treatment outcome results in the field.
Collapse
Affiliation(s)
- Meredith A Nelson
- Cincinnati Children's Hospital Medical Center, Cincinnati, USA.
- Department of Pediatrics , University of Cincinnati College of Medicine, Cincinnati, USA.
| | - Lauren M Schmitt
- Cincinnati Children's Hospital Medical Center, Cincinnati, USA
- Department of Pediatrics , University of Cincinnati College of Medicine, Cincinnati, USA
| | - Paul S Horn
- Cincinnati Children's Hospital Medical Center, Cincinnati, USA
- Department of Pediatrics , University of Cincinnati College of Medicine, Cincinnati, USA
| | | | - David Hessl
- MIND Institute, University of California Davis, Davis, USA
| | - Rebecca C Shaffer
- Cincinnati Children's Hospital Medical Center, Cincinnati, USA
- Department of Pediatrics , University of Cincinnati College of Medicine, Cincinnati, USA
| | | | - Dejan B Budimirovic
- Department of Psychiatry, Fragile X Clinic, Kennedy Krieger Institute, Baltimore, USA
| | - Paul Wang
- Clinical Research Associates, LLC, New York, USA
| | - Debra L Reisinger
- Cincinnati Children's Hospital Medical Center, Cincinnati, USA
- Department of Pediatrics , University of Cincinnati College of Medicine, Cincinnati, USA
| | | | - Craig A Erickson
- Cincinnati Children's Hospital Medical Center, Cincinnati, USA
- Department of Psychiatry , University of Cincinnati, Cincinnati, USA
| |
Collapse
|
4
|
Mohammed HE, Bady Z, Haseeb ME, Aboeldahab H, Sharaf-Eldin WE, Zaki MS. Is trofinetide a future treatment for Rett syndrome? A comprehensive systematic review and meta-analysis of randomized controlled trials. BMC Med 2024; 22:299. [PMID: 39020317 PMCID: PMC11256568 DOI: 10.1186/s12916-024-03506-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Accepted: 06/27/2024] [Indexed: 07/19/2024] Open
Abstract
BACKGROUND Rett syndrome (RTT) is a rare, life-threatening, genetic neurodevelopmental disorder. Treatment in RTT encounters many challenges. Trofinetide, a modified amino-terminal tripeptide of insulin-like growth factor 1, has demonstrated clinically promising results in RTT. In this study, trofinetide efficacy and safety in RTT are systematically reviewed and meta-analyzed. METHODS A systematic search of five electronic databases was conducted until January 2024. Review Manager 5.4 software was used for the analysis. The analysis was based on a weighted mean difference and standard error with a confidence interval (CI) of 95%, and a statistically significant P-value was considered if it was < 0.05. The study was registered on PROSPERO with registration number CRD42024499849. Quality of evidence was assessed using GRADE. RESULTS Three randomized controlled trials (RCTs) with 276 patients were included in the analysis. Trofinetide improved both caregiver outcomes and clinical scales by improving the Rett Syndrome Behavior Questionnaire (RSBQ) (mean difference (MD): - 3.46 points, 95% CI: - 5.63 to - 1.27, P = 0.0002) and Clinical Global Impression Scale-Improvement (CGI-I) (MD: - 0.35, 95% CI: - 0.51 to - 0.18, P < 0.0001), respectively. However, trofinetide neither improved the Caregiver Top 3 Concerns Visual Analog Scale nor the Rett Motor Behavioral Assessment. Regarding safety, trofinetide was significantly associated with vomiting compared to placebo (odds ratio (OR): 3.17, 95% CI: 1.57 to 6.43, P = 0.001). After solving heterogeneity, results showed a statistically significant incidence of diarrhea in the trofinetide (200 mg) group compared to placebo (OR: 18.51, 95% CI: 9.30 to 36.84, P ≤ 0.00001). CONCLUSIONS Trofinetide demonstrated statistically significant improvements in CGI-I and RSBQ in pediatrics and adult patients with Rett. Side effects are limited to vomiting and diarrhea. Although diarrhea yielded an insignificant result in our analysis, it emerged as a cause for treatment discontinuation in the participating trials, and a statistically significant risk for diarrhea emerged when excluding the study using a lower dose of the drug, hence causing heterogeneity, in the meta-analysis. Given the diverse genetic landscape of RTT, future RCTs investigating correlations between RTT genotype and phenotypic improvements by trofinetide will be beneficial. RCTs encompassing male patients with larger and longer cohorts are recommended.
Collapse
Affiliation(s)
| | - Zeyad Bady
- Faculty of Medicine, Assiut University, Assiut, Egypt
| | | | - Heba Aboeldahab
- Medical Research Group of Egypt (MRGE), Negida Academy, Cairo, Egypt
- Clinical Research Department, El-Gomhoria General Hospital, MOHP, Alexandria, Egypt
- Biomedical Informatics and Medical Statistics Department, Medical Research Institute, Alexandria University, Alexandria, Egypt
| | - Wessam E Sharaf-Eldin
- Medical Molecular Genetics Department, Human Genetics and Genome Research Institute, National Research Centre, Cairo, Egypt
| | - Maha S Zaki
- Clinical Genetics Department, Human Genetics and Genome Research Institute, National Research Centre, Cairo, Egypt.
- Medical Genetics Department, Armed Forces College of Medicine (AFCM), Cairo, Egypt.
| |
Collapse
|
5
|
Protic D, Hagerman R. State-of-the-art therapies for fragile X syndrome. Dev Med Child Neurol 2024; 66:863-871. [PMID: 38385885 PMCID: PMC11144093 DOI: 10.1111/dmcn.15885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 01/22/2024] [Accepted: 01/24/2024] [Indexed: 02/23/2024]
Abstract
Fragile X syndrome (FXS) is a neurodevelopmental disorder caused by a full mutation (> 200 CGG repeats) in the FMR1 gene. FXS is the leading cause of inherited intellectual disabilities and the most commonly known genetic cause of autism spectrum disorder. Children with FXS experience behavioral and sleep problems, anxiety, inattention, learning difficulties, and speech and language delays. There are no approved medications for FXS; however, there are several interventions and treatments aimed at managing the symptoms and improving the quality of life of individuals with FXS. A combination of non-pharmacological therapies and pharmacotherapy is currently the most effective treatment for FXS. Currently, several targeted treatments, such as metformin, sertraline, and cannabidiol, can be used by clinicians to treat FXS. Gene therapy is rapidly developing and holds potential as a prospective treatment option. Soon its efficacy and safety in patients with FXS will be demonstrated. WHAT THIS PAPER ADDS: Targeted treatment of fragile X syndrome (FXS) is the best current therapeutic approach. Gene therapy holds potential as a prospective treatment for FXS in the future.
Collapse
Affiliation(s)
- Dragana Protic
- Department of Pharmacology, Clinical Pharmacology and Toxicology, Faculty of Medicine University of Belgrade, Belgrade, Serbia
- Fragile X Clinic, Special Hospital for Cerebral Palsy and Developmental Neurology, Belgrade, Serbia
| | - Randi Hagerman
- Medical Investigation of Neurodevelopmental Disorders Institute, University of California, Davis, CA, USA
- Department of Pediatrics, University of California, Davis School of Medicine, Sacramento, CA, USA
| |
Collapse
|
6
|
Watkins LV, Moon S, Burrows L, Tromans S, Barwell J, Shankar R. Pharmacological management of fragile X syndrome: a systematic review and narrative summary of the current evidence. Expert Opin Pharmacother 2024; 25:301-313. [PMID: 38393835 DOI: 10.1080/14656566.2024.2323605] [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: 12/17/2023] [Accepted: 02/22/2024] [Indexed: 02/25/2024]
Abstract
INTRODUCTION Fragile X syndrome (FXS) is the most common inherited cause of Intellectual Disability. There is a broad phenotype that includes deficits in cognition and behavioral changes, alongside physical characteristics. Phenotype depends upon the level of mutation in the FMR1 (fragile X messenger ribonucleoprotein 1) gene. The molecular understanding of the impact of the FMR1 gene mutation provides an opportunity to target treatment not only at symptoms but also on a molecular level. METHODS We conducted a systematic review to provide an up-to-date narrative summary of the current evidence for pharmacological treatment in FXS. The review was restricted to randomized, blinded, placebo-controlled trials. RESULTS The outcomes from these studies are discussed and the level of evidence assessed against validated criteria. The initial search identified 2377 articles, of which 16 were included in the final analysis. CONCLUSION Based on this review to date there is limited data to support any specific pharmacological treatments, although the data for cannabinoids are encouraging in those with FXS and in future developments in gene therapy may provide the answer to the search for precision medicine. Treatment must be person-centered and consider the combination of medical, genetic, cognitive, and emotional challenges.
Collapse
Affiliation(s)
- Lance V Watkins
- Epilepsy Specialist Service, Swansea Bay University Health Board, Cardiff, UK
- Unit for Development in Intellectual and Developmental Disabilities, University of South Wales, Pontypridd, UK
- Cornwall Intellectual Disability Equitable Research (CIDER), University of Plymouth Peninsula School of Medicine, Truro, UK
| | - Seungyoun Moon
- Epilepsy Specialist Service, Swansea Bay University Health Board, Cardiff, UK
| | - Lisa Burrows
- Cornwall Intellectual Disability Equitable Research (CIDER), University of Plymouth Peninsula School of Medicine, Truro, UK
- Adult Neurodevelopmental Psychiatry, Cornwall Partnership NHS Trust, Truro, UK
| | - Samuel Tromans
- Department of Population Health Sciences, University of Leicester, Leicester, UK
- Adult Learning Disability Service, Leicestershire Partnership NHS Trust, Leicester, UK
| | - Julian Barwell
- Clinical Genetics Department, University Hospitals of Leicester, Leicester, UK
| | - Rohit Shankar
- Cornwall Intellectual Disability Equitable Research (CIDER), University of Plymouth Peninsula School of Medicine, Truro, UK
| |
Collapse
|
7
|
Dakopolos A, Condy E, Smith E, Harvey D, Kaat AJ, Coleman J, Riley K, Berry-Kravis E, Hessl D. Developmental Associations between Cognition and Adaptive Behavior in Intellectual and Developmental Disability. RESEARCH SQUARE 2024:rs.3.rs-3684708. [PMID: 38260292 PMCID: PMC10802716 DOI: 10.21203/rs.3.rs-3684708/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Background Intellectual and developmental disabilities (IDDs) are associated with both cognitive challenges and difficulties in conceptual, social, and practical areas of living (DSM-5). Individuals with IDD often present with an intellectual disability in addition to a developmental disability such as autism or Down syndrome. Those with IDD may present with deficits in intellectual functioning as well as adaptive functioning that interfere with independence and living skills. The present study sought to examine associations of longitudinal developmental change in domains of cognition (NIH Toolbox Cognition Battery, NIHTB-CB) and adaptive behavior domains (Vineland Adaptive Behavior Scales-3; VABS-3) including Socialization, Communication, and Daily Living Skills (DLS) over a two-year period. Methods Eligible participants for this multisite longitudinal study included those who were between 6 and 26 years at Visit 1, and who had a diagnosis of, or suspected intellectual disability (ID), including borderline ID. Three groups were recruited, including those with fragile X syndrome, Down syndrome, and other/idiopathic intellectual disability. In order to examine the association of developmental change between cognitive and adaptive behavior domains, bivariate latent change score (BLCS) models were fit to compare change in the three cognitive domains measured by the NIHTB-CB (Fluid, Crystallized, Composite) and the three adaptive behavior domains measured by the VABS-3 (Communication, DLS, and Socialization). Results Over a two-year period, change in cognition (both Crystalized and Composite) was significantly and positively associated with change in daily living skills. Also, baseline cognition level predicted growth in adaptive behavior, however baseline adaptive behavior did not predict growth in cognition in any model. Conclusions The present study demonstrated that developmental improvements in cognition and adaptive behavior are associated in children and young adults with IDD, indicating the potential for cross-domain effects of intervention. Notably, improvements in Daily Living Skills on the VABS-3 emerged as a primary area of adaptive behavior that positively related to improvements in cognition. This work provides evidence for the clinical, "real life" meaningfulness of the NIHTB-CB in IDD, and important empirical support for the NIHTB-CB as a fit-for-purpose performance-based outcome measure for this population.
Collapse
Affiliation(s)
| | | | - Elizabeth Smith
- Cincinnati Children's Hospital Medical Center Burnet Campus: Cincinnati Children's Hospital Medical Center
| | | | - Aaron J Kaat
- Northwestern University Feinberg School of Medicine
| | | | | | | | | |
Collapse
|
8
|
Müller AR, den Hollander B, van de Ven PM, Roes KCB, Geertjens L, Bruining H, van Karnebeek CDM, Jansen FE, de Wit MCY, Ten Hoopen LW, Rietman AB, Dierckx B, Wijburg FA, Boot E, Brands MMG, van Eeghen AM. Cannabidiol (Epidyolex®) for severe behavioral manifestations in patients with tuberous sclerosis complex, mucopolysaccharidosis type III and fragile X syndrome: protocol for a series of randomized, placebo-controlled N-of-1 trials. BMC Psychiatry 2024; 24:23. [PMID: 38177999 PMCID: PMC10768432 DOI: 10.1186/s12888-023-05422-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 11/29/2023] [Indexed: 01/06/2024] Open
Abstract
BACKGROUND Many rare genetic neurodevelopmental disorders (RGNDs) are characterized by intellectual disability (ID), severe cognitive and behavioral impairments, potentially diagnosed as a comorbid autism spectrum disorder or attention-deficit hyperactivity disorder. Quality of life is often impaired due to irritability, aggression and self-injurious behavior, generally refractory to standard therapies. There are indications from previous (case) studies and patient reporting that cannabidiol (CBD) may be an effective treatment for severe behavioral manifestations in RGNDs. However, clear evidence is lacking and interventional research is challenging due to the rarity as well as the heterogeneity within and between disease groups and interindividual differences in treatment response. Our objective is to examine the effectiveness of CBD on severe behavioral manifestations in three RGNDs, including Tuberous Sclerosis Complex (TSC), mucopolysaccharidosis type III (MPS III), and Fragile X syndrome (FXS), using an innovative trial design. METHODS We aim to conduct placebo-controlled, double-blind, block-randomized, multiple crossover N-of-1 studies with oral CBD (twice daily) in 30 patients (aged ≥ 6 years) with confirmed TSC, MPS III or FXS and severe behavioral manifestations. The treatment is oral CBD up to a maximum of 25 mg/kg/day, twice daily. The primary outcome measure is the subscale irritability of the Aberrant Behavior Checklist. Secondary outcome measures include (personalized) patient-reported outcome measures with regard to behavioral and psychiatric outcomes, disease-specific outcome measures, parental stress, seizure frequency, and adverse effects of CBD. Questionnaires will be completed and study medication will be taken at the participants' natural setting. Individual treatment effects will be determined based on summary statistics. A mixed model analysis will be applied for analyzing the effectiveness of the intervention per disorder and across disorders combining data from the individual N-of-1 trials. DISCUSSION These N-of-1 trials address an unmet medical need and will provide information on the effectiveness of CBD for severe behavioral manifestations in RGNDs, potentially generating generalizable knowledge at an individual-, disorder- and RGND population level. TRIAL REGISTRATION EudraCT: 2021-003250-23, registered 25 August 2022, https://www.clinicaltrialsregister.eu/ctr-search/trial/2021-003250-23/NL .
Collapse
Affiliation(s)
- A R Müller
- Department of Pediatrics, Emma Children's Hospital, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
- 's Heeren Loo Care Group, Amersfoort, The Netherlands
- Emma Center for Personalized Medicine, Amsterdam UMC, Amsterdam, the Netherlands
| | - B den Hollander
- Department of Pediatrics, Emma Children's Hospital, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
- Emma Center for Personalized Medicine, Amsterdam UMC, Amsterdam, the Netherlands
- United for Metabolic Diseases, Amsterdam, The Netherlands
| | - P M van de Ven
- Department of Data Science and Biostatistics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - K C B Roes
- Department of Health Evidence, Biostatistics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - L Geertjens
- Child and Adolescent Psychiatry and Psychosocial Care, Amsterdam UMC Location Vrije Universiteit, Amsterdam, The Netherlands
- Amsterdam UMC, Amsterdam Neuroscience, Amsterdam Reproduction and Development, N=You Neurodevelopmental Precision Center, Amsterdam, The Netherlands
| | - H Bruining
- Emma Center for Personalized Medicine, Amsterdam UMC, Amsterdam, the Netherlands
- Child and Adolescent Psychiatry and Psychosocial Care, Amsterdam UMC Location Vrije Universiteit, Amsterdam, The Netherlands
- Amsterdam UMC, Amsterdam Neuroscience, Amsterdam Reproduction and Development, N=You Neurodevelopmental Precision Center, Amsterdam, The Netherlands
- Levvel, Center for Child and Adolescent Psychiatry, Amsterdam, The Netherlands
| | - C D M van Karnebeek
- Department of Pediatrics, Emma Children's Hospital, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
- Emma Center for Personalized Medicine, Amsterdam UMC, Amsterdam, the Netherlands
- United for Metabolic Diseases, Amsterdam, The Netherlands
- Department of Human Genetics, Amsterdam UMC, Amsterdam, The Netherlands
| | - F E Jansen
- Department of Pediatric Neurology, Brain, University Medical Center Utrecht, Utrecht, The Netherlands
| | - M C Y de Wit
- ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus University Medical Center, Rotterdam, The Netherlands
- Department of Neurology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - L W Ten Hoopen
- ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus University Medical Center, Rotterdam, The Netherlands
- Department of Child and Adolescent Psychiatry/Psychology, Sophia Children's Hospital, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - A B Rietman
- ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus University Medical Center, Rotterdam, The Netherlands
- Department of Child and Adolescent Psychiatry/Psychology, Sophia Children's Hospital, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - B Dierckx
- ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus University Medical Center, Rotterdam, The Netherlands
- Department of Child and Adolescent Psychiatry/Psychology, Sophia Children's Hospital, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - F A Wijburg
- Department of Pediatrics, Emma Children's Hospital, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
| | - E Boot
- 's Heeren Loo Care Group, Amersfoort, The Netherlands
- The Dalglish Family 22Q Clinic, Toronto, ON, Canada
- Department of Psychiatry & Neuropsychology, Maastricht University Medical Center, Maastricht, the Netherlands
| | - M M G Brands
- Department of Pediatrics, Emma Children's Hospital, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
- Emma Center for Personalized Medicine, Amsterdam UMC, Amsterdam, the Netherlands
- United for Metabolic Diseases, Amsterdam, The Netherlands
| | - A M van Eeghen
- Department of Pediatrics, Emma Children's Hospital, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands.
- 's Heeren Loo Care Group, Amersfoort, The Netherlands.
- Emma Center for Personalized Medicine, Amsterdam UMC, Amsterdam, the Netherlands.
| |
Collapse
|
9
|
Müller AR, van Silfhout NY, den Hollander B, Kampman DHC, Bakkum L, Brands MMMG, Haverman L, Terwee CB, Schuengel C, Daams J, Hessl D, Wijburg FA, Boot E, van Eeghen AM. Navigating the outcome maze: a scoping review of outcomes and instruments in clinical trials in genetic neurodevelopmental disorders and intellectual disability. THERAPEUTIC ADVANCES IN RARE DISEASE 2024; 5:26330040241245721. [PMID: 38681798 PMCID: PMC11047260 DOI: 10.1177/26330040241245721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 03/14/2024] [Indexed: 05/01/2024]
Abstract
Background Individuals with genetic neurodevelopmental disorders (GNDs) or intellectual disability (ID) are often affected by complex neuropsychiatric comorbidities. Targeted treatments are increasingly available, but due to the heterogeneity of these patient populations, choosing a key outcome and corresponding outcome measurement instrument remains challenging. Objectives The aim of this scoping review was to describe the research on outcomes and instruments used in clinical trials in GNDs and ID. Eligibility criteria Clinical trials in individuals with GNDs and ID for any intervention over the past 10 years were included in the review. Sources of evidence MEDLINE, PsycINFO, and Cochrane CENTRAL were searched. Titles and abstracts were independently screened for eligibility with a subsample of 10% double-screening for interrater reliability. Data from full texts were independently reviewed. Discrepancies were discussed until consensus was reached. Charting methods Information was recorded on patient populations, interventions, designs, outcomes, measurement instruments, and type of reporter when applicable. Qualitative and descriptive analyses were performed. Results We included 312 studies reporting 91 different outcomes, with cognitive function most frequently measured (28%). Various outcome measurement instruments (n = 457) were used, with 288 in only a single clinical trial. There were 18 genetic condition-specific instruments and 16 measures were designed ad-hoc for one particular trial. Types of report included proxy-report (39%), self-report (22%), clinician-report (16%), observer-report (6%), self-assisted report (1%), or unknown (16%). Conclusion This scoping review of current practice reveals a myriad of outcomes and outcome measurement instruments for clinical trials in GNDs and ID. This complicates generalization, evidence synthesis, and evaluation. It underlines the need for consensus on suitability, validity, and relevancy of instruments, ultimately resulting in a core outcome set. A series of steps is proposed to move from the myriad of measures to a more unified approach.
Collapse
Affiliation(s)
- Annelieke R. Müller
- Department of Pediatrics, Emma Children’s Hospital, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
- Advisium, ’s Heeren Loo, Amersfoort, Utrecht, The Netherlands
- Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
- Emma Center for Personalized Medicine, Amsterdam UMC, Amsterdam, The Netherlands
| | - Nadia Y. van Silfhout
- Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
- Amsterdam Reproduction & Development, Child Development, Amsterdam, The Netherlands
- Emma Children’s Hospital, Child and Adolescent Psychiatry & Psychosocial Care, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
| | - Bibiche den Hollander
- Department of Pediatrics, Emma Children’s Hospital, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
- Emma Center for Personalized Medicine, Amsterdam UMC, Amsterdam, The Netherlands
- United for Metabolic Diseases, Amsterdam, The Netherlands
| | - Dick H. C. Kampman
- Faculty of Science, Operational Management, ICT Department, Utrecht University, Utrecht, The Netherlands
| | - Lianne Bakkum
- Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
- Department of Clinical Child and Family Studies, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Marion M. M. G. Brands
- Department of Pediatrics, Emma Children’s Hospital, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
- Emma Center for Personalized Medicine, Amsterdam UMC, Amsterdam, The Netherlands
- Amsterdam Reproduction & Development, Child Development, Amsterdam, The Netherlands
- United for Metabolic Diseases, Amsterdam, The Netherlands
| | - Lotte Haverman
- Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
- Amsterdam Reproduction & Development, Child Development, Amsterdam, The Netherlands
- Emma Children’s Hospital, Child and Adolescent Psychiatry & Psychosocial Care, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
| | - Caroline B. Terwee
- Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
- Epidemiology and Data Science, Amsterdam UMC Location Vrije Universiteit, Amsterdam, The Netherlands
| | - Carlo Schuengel
- Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
- Department of Clinical Child and Family Studies, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Joost Daams
- Medical Library, Research Support, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
| | - David Hessl
- MIND Institute and Department of Psychiatry and Behavioral Sciences, University of California Davis, Sacramento, CA, USA
| | - Frits A. Wijburg
- Department of Pediatrics, Emma Children’s Hospital, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
| | - Erik Boot
- Advisium, ’s Heeren Loo, Amersfoort, Utrecht, The Netherlands
- The Dalglish Family 22q Clinic, Toronto, ON, Canada
- Department of Psychiatry & Neuropsychology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Agnies M. van Eeghen
- Emma Center for Personalized Medicine, Amsterdam UMC, Meibergdreef 9, Amsterdam, 1105 AZ, The Netherlands
- Advisium, ’s Heeren Loo, Berkenweg 11, 3818 LA, Amersfoort, The Netherlands
- Amsterdam Public Health Research Institute, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
- Amsterdam Reproduction & Development, Child Development, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| |
Collapse
|
10
|
Stasolla F, Passaro A, Di Gioia M, Curcio E, Zullo A. Combined extended reality and reinforcement learning to promote healthcare and reduce social anxiety in fragile X syndrome: a new assessment tool and a rehabilitative strategy. Front Psychol 2023; 14:1273117. [PMID: 38179497 PMCID: PMC10765535 DOI: 10.3389/fpsyg.2023.1273117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 11/30/2023] [Indexed: 01/06/2024] Open
Affiliation(s)
| | - Anna Passaro
- University “Giustino Fortunato” of Benevento, Benevento, Italy
| | | | - Enza Curcio
- University “Giustino Fortunato” of Benevento, Benevento, Italy
| | | |
Collapse
|
11
|
Li R, Lightbody AA, Lee CH, Bartholomay KL, Marzelli MJ, Reiss AL. Association of Intrinsic Functional Brain Network and Longitudinal Development of Cognitive Behavioral Symptoms in Young Girls With Fragile X Syndrome. Biol Psychiatry 2023; 94:814-822. [PMID: 37004849 PMCID: PMC10544666 DOI: 10.1016/j.biopsych.2023.03.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 03/01/2023] [Accepted: 03/19/2023] [Indexed: 04/04/2023]
Abstract
BACKGROUND Fragile X syndrome (FXS) is an X chromosome-linked genetic disorder characterized by increased risk for behavioral, social, and neurocognitive deficits. Because males express a more severe phenotype than females, research has focused largely on identifying neural abnormalities in all-male or both-sex populations with FXS. Therefore, very little is known about the neural alterations that contribute to cognitive behavioral symptoms in females with FXS. This cross-sectional study aimed to elucidate the large-scale resting-state brain networks associated with the multidomain cognitive behavioral phenotype in girls with FXS. METHODS We recruited 38 girls with full-mutation FXS (11.58 ± 3.15 years) and 32 girls without FXS (11.66 ± 2.27 years). Both groups were matched on age, verbal IQ, and multidomain cognitive behavioral symptoms. Resting-state functional magnetic resonance imaging data were collected. RESULTS Compared with the control group, girls with FXS showed significantly greater resting-state functional connectivity of the default mode network, lower nodal strength at the right middle temporal gyrus, stronger nodal strength at the left caudate, and higher global efficiency of the default mode network. These aberrant brain network characteristics map directly onto the cognitive behavioral symptoms commonly observed in girls with FXS. An exploratory analysis suggested that brain network patterns at a prior time point (time 1) were predictive of the longitudinal development of participants' multidomain cognitive behavioral symptoms. CONCLUSIONS These findings represent the first examination of large-scale brain network alterations in a large sample of girls with FXS, expanding our knowledge of potential neural mechanisms underlying the development of cognitive behavioral symptoms in girls with FXS.
Collapse
Affiliation(s)
- Rihui Li
- Center for Interdisciplinary Brain Sciences Research, Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, California; Center for Cognitive and Brain Sciences, Institute of Collaborative Innovation, University of Macau, Taipa, Macau S.A.R., China.
| | - Amy A Lightbody
- Center for Interdisciplinary Brain Sciences Research, Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, California
| | - Cindy H Lee
- Center for Interdisciplinary Brain Sciences Research, Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, California
| | - Kristi L Bartholomay
- Center for Interdisciplinary Brain Sciences Research, Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, California
| | - Matthew J Marzelli
- Center for Interdisciplinary Brain Sciences Research, Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, California
| | - Allan L Reiss
- Center for Interdisciplinary Brain Sciences Research, Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, California; Department of Radiology, Stanford University, Stanford, California; Department of Pediatrics, Stanford University, Stanford, California
| |
Collapse
|
12
|
Talapatra D, Snider L, Coleman J, Thompson T, Reinhardt JS, Hessl D, Riley K. Deviation scores: An innovative approach to interpreting cognitive test results for individuals with intellectual disabilities. JOURNAL OF APPLIED RESEARCH IN INTELLECTUAL DISABILITIES 2023; 36:1218-1228. [PMID: 37553958 PMCID: PMC10591767 DOI: 10.1111/jar.13137] [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: 10/13/2022] [Revised: 03/31/2023] [Accepted: 06/08/2023] [Indexed: 08/10/2023]
Abstract
BACKGROUND Students with Intellectual Disability undergo frequent cognitive testing. Testing with this population is limited by insensitivity to relative strengths and weaknesses due to floor effects. AIM The study explored the utility of deviation scores via four case studies as a supplement to educational decision-making. METHODS Four students with Intellectual Disability completed cognitive testing. Deviation scores were calculated using age dependent raw z-score transformations to determine deviation from the standardization sample norms. RESULTS The application of deviation scores highlighted true relative strengths and weaknesses for students with Intellectual Disability rather than documenting previously known deficits. The four cases studies illustrated where deviation scores could, or could not, add value above and beyond traditional scoring. DISCUSSION Deviation scores can supplement placement and service decisions for students. Practical and psychometric considerations are reviewed. CONCLUSION The findings highlight the usefulness of deviation scores in providing meaningful information to school- and clinic-based practitioners.
Collapse
Affiliation(s)
- Devadrita Talapatra
- College of Education, Teaching and Learning Sciences Department, University of Denver, Denver, Colorado, USA
| | - Laurel Snider
- College of Education, The University of Alabama, Tuscaloosa, Alabama, USA
| | - Jeanine Coleman
- Office of the Provost, Regis University, Denver, Colorado, USA
| | - Talia Thompson
- School of Medicine, University of Colorado, Aurora, Colorado, USA
| | - Jessica S Reinhardt
- College of Education, Department of Psychological Studies in Education, Temple University, Philadelphia, Pennsylvania, USA
| | - David Hessl
- MIND Institute and Department of Psychiatry and Behavioral Sciences, University of California Davis, Davis, California, USA
| | - Karen Riley
- Office of the President, Slippery Rock University, Slippery Rock, PA, USA
| |
Collapse
|
13
|
Berry-Kravis E, Abbeduto L, Hagerman R, Coffey CS, Cudkowicz M, Erickson CA, McDuffie A, Hessl D, Ethridge L, Tassone F, Kaufmann WE, Friedmann K, Bullard L, Hoffmann A, Veenstra-VanderWeele J, Staley K, Klements D, Moshinsky M, Harkey B, Long J, Fedler J, Klingner E, Ecklund D, Costigan M, Huff T, Pearson B. Effects of AFQ056 on language learning in fragile X syndrome. J Clin Invest 2023; 134:e171723. [PMID: 37651202 PMCID: PMC10904045 DOI: 10.1172/jci171723] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 08/22/2023] [Indexed: 09/02/2023] Open
Abstract
BACKGROUNDFXLEARN, the first-ever large multisite trial of effects of disease-targeted pharmacotherapy on learning, was designed to explore a paradigm for measuring effects of mechanism-targeted treatment in fragile X syndrome (FXS). In FXLEARN, the effects of metabotropic glutamate receptor type 5 (mGluR5) negative allosteric modulator (NAM) AFQ056 on language learning were evaluated in 3- to 6-year-old children with FXS, expected to have more learning plasticity than adults, for whom prior trials of mGluR5 NAMs have failed.METHODSAfter a 4-month single-blind placebo lead-in, participants were randomized 1:1 to AFQ056 or placebo, with 2 months of dose optimization to the maximum tolerated dose, then 6 months of treatment during which a language-learning intervention was implemented for both groups. The primary outcome was a centrally scored videotaped communication measure, the Weighted Communication Scale (WCS). Secondary outcomes were objective performance-based and parent-reported cognitive and language measures.RESULTSFXLEARN enrolled 110 participants, randomized 99, and had 91 who completed the placebo-controlled period. Although both groups made language progress and there were no safety issues, the change in WCS score during the placebo-controlled period was not significantly different between the AFQ056 and placebo-treated groups, nor were there any significant between-group differences in change in any secondary measures.CONCLUSIONDespite the large body of evidence supporting use of mGluR5 NAMs in animal models of FXS, this study suggests that this mechanism of action does not translate into benefit for the human FXS population and that better strategies are needed to determine which mechanisms will translate from preclinical models to humans in genetic neurodevelopmental disorders.TRIAL REGISTRATIONClincalTrials.gov NCT02920892.FUNDING SOURCESNeuroNEXT network NIH grants U01NS096767, U24NS107200, U24NS107209, U01NS077323, U24NS107183, U24NS107168, U24NS107128, U24NS107199, U24NS107198, U24NS107166, U10NS077368, U01NS077366, U24NS107205, U01NS077179, and U01NS077352; NIH grant P50HD103526; and Novartis IIT grant AFQ056X2201T for provision of AFQ056.
Collapse
Affiliation(s)
- Elizabeth Berry-Kravis
- Departments of Pediatrics, Neurological Sciences, and Anatomy & Cell Biology, Rush University Medical Center, Chicago, Illinois, USA
| | - Leonard Abbeduto
- MIND Institute and Department of Psychiatry and Behavioral Sciences and
| | - Randi Hagerman
- MIND Institute and Department of Pediatrics, UCD, Sacramento, California, USA
| | | | - Merit Cudkowicz
- Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Craig A. Erickson
- Division of Child and Adolescent Psychiatry, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Andrea McDuffie
- MIND Institute and Department of Psychiatry and Behavioral Sciences and
| | - David Hessl
- MIND Institute and Department of Psychiatry and Behavioral Sciences and
| | - Lauren Ethridge
- Department of Psychology, University of Oklahoma, Norman, Oklahoma, and Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Flora Tassone
- MIND Institute and Department of Biochemistry and Molecular Medicine, UCD, Sacramento, California, USA
| | - Walter E. Kaufmann
- Department of Human Genetics, Emory University School of Medicine, Atlanta, Georgia, USA
| | | | - Lauren Bullard
- MIND Institute and Department of Psychiatry and Behavioral Sciences and
| | - Anne Hoffmann
- Departments of Pediatrics and Communication Disorders and Sciences, Rush University Medical Center, Chicago, Illinois, USA
| | - Jeremy Veenstra-VanderWeele
- Department of Psychiatry, Columbia University, and New York State Psychiatric Institute, New York, New York, USA
| | - Kevin Staley
- Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - David Klements
- Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Michael Moshinsky
- Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Brittney Harkey
- Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Jeff Long
- Department of Biostatistics, University of Iowa, Iowa City, Iowa, USA
| | - Janel Fedler
- Department of Biostatistics, University of Iowa, Iowa City, Iowa, USA
| | | | - Dixie Ecklund
- Department of Biostatistics, University of Iowa, Iowa City, Iowa, USA
| | - Michele Costigan
- Department of Biostatistics, University of Iowa, Iowa City, Iowa, USA
| | - Trevis Huff
- Department of Biostatistics, University of Iowa, Iowa City, Iowa, USA
| | - Brenda Pearson
- Department of Biostatistics, University of Iowa, Iowa City, Iowa, USA
| | | |
Collapse
|
14
|
Oya M, Matsuoka K, Kubota M, Fujino J, Tei S, Takahata K, Tagai K, Yamamoto Y, Shimada H, Seki C, Itahashi T, Aoki YY, Ohta H, Hashimoto RI, Sugihara G, Obata T, Zhang MR, Suhara T, Nakamura M, Kato N, Takado Y, Takahashi H, Higuchi M. Increased glutamate and glutamine levels and their relationship to astrocytes and dopaminergic transmissions in the brains of adults with autism. Sci Rep 2023; 13:11655. [PMID: 37468523 DOI: 10.1038/s41598-023-38306-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 07/06/2023] [Indexed: 07/21/2023] Open
Abstract
Increased excitatory neuronal tones have been implicated in autism, but its mechanism remains elusive. The amplified glutamate signals may arise from enhanced glutamatergic circuits, which can be affected by astrocyte activation and suppressive signaling of dopamine neurotransmission. We tested this hypothesis using magnetic resonance spectroscopy and positron emission tomography scan with 11C-SCH23390 for dopamine D1 receptors in the anterior cingulate cortex (ACC). We enrolled 18 male adults with high-functioning autism and 20 typically developed (TD) male subjects. The autism group showed elevated glutamate, glutamine, and myo-inositol (mI) levels compared with the TD group (p = 0.045, p = 0.044, p = 0.030, respectively) and a positive correlation between glutamine and mI levels in the ACC (r = 0.54, p = 0.020). In autism and TD groups, ACC D1 receptor radioligand binding was negatively correlated with ACC glutamine levels (r = - 0.55, p = 0.022; r = - 0.58, p = 0.008, respectively). The enhanced glutamate-glutamine metabolism might be due to astroglial activation and the consequent reinforcement of glutamine synthesis in autistic brains. Glutamine synthesis could underly the physiological inhibitory control of dopaminergic D1 receptor signals. Our findings suggest a high neuron excitation-inhibition ratio with astrocytic activation in the etiology of autism.
Collapse
Affiliation(s)
- Masaki Oya
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology (QST), 4-9-1 Anagawa, Inage-ku, Chiba-shi, Chiba, 263-8555, Japan
- Department of Psychiatry and Behavioral Sciences, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
| | - Kiwamu Matsuoka
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology (QST), 4-9-1 Anagawa, Inage-ku, Chiba-shi, Chiba, 263-8555, Japan.
- Department of Psychiatry, Nara Medical University, Kashihara-shi, Nara, Japan.
| | - Manabu Kubota
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology (QST), 4-9-1 Anagawa, Inage-ku, Chiba-shi, Chiba, 263-8555, Japan
- Department of Psychiatry, Graduate School of Medicine, Kyoto University, Kyoto-shi, Kyoto, Japan
- Medical Institute of Developmental Disabilities Research, Showa University, Setagaya-ku, Tokyo, Japan
| | - Junya Fujino
- Department of Psychiatry and Behavioral Sciences, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
- Medical Institute of Developmental Disabilities Research, Showa University, Setagaya-ku, Tokyo, Japan
| | - Shisei Tei
- Department of Psychiatry, Graduate School of Medicine, Kyoto University, Kyoto-shi, Kyoto, Japan
- Medical Institute of Developmental Disabilities Research, Showa University, Setagaya-ku, Tokyo, Japan
- Institute of Applied Brain Sciences, Waseda University, Tokorozawa-shi, Saitama, Japan
- School of Human and Social Sciences, Tokyo International University, Kawagoe-shi, Saitama, Japan
| | - Keisuke Takahata
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology (QST), 4-9-1 Anagawa, Inage-ku, Chiba-shi, Chiba, 263-8555, Japan
- Department of Neuropsychiatry, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Kenji Tagai
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology (QST), 4-9-1 Anagawa, Inage-ku, Chiba-shi, Chiba, 263-8555, Japan
| | - Yasuharu Yamamoto
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology (QST), 4-9-1 Anagawa, Inage-ku, Chiba-shi, Chiba, 263-8555, Japan
- Department of Neuropsychiatry, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Hitoshi Shimada
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology (QST), 4-9-1 Anagawa, Inage-ku, Chiba-shi, Chiba, 263-8555, Japan
- Center for Integrated Human Brain Science, Brain Research Institute, Niigata University, Niigata-shi, Niigata, Japan
| | - Chie Seki
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology (QST), 4-9-1 Anagawa, Inage-ku, Chiba-shi, Chiba, 263-8555, Japan
| | - Takashi Itahashi
- Medical Institute of Developmental Disabilities Research, Showa University, Setagaya-ku, Tokyo, Japan
| | - Yuta Y Aoki
- Medical Institute of Developmental Disabilities Research, Showa University, Setagaya-ku, Tokyo, Japan
| | - Haruhisa Ohta
- Medical Institute of Developmental Disabilities Research, Showa University, Setagaya-ku, Tokyo, Japan
- Department of Psychiatry, School of Medicine, Showa University, Setagaya-ku, Tokyo, Japan
| | - Ryu-Ichiro Hashimoto
- Medical Institute of Developmental Disabilities Research, Showa University, Setagaya-ku, Tokyo, Japan
- Department of Language Sciences, Graduate School of Humanities, Tokyo Metropolitan University, Hachioji-shi, Tokyo, Japan
| | - Genichi Sugihara
- Department of Psychiatry and Behavioral Sciences, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
| | - Takayuki Obata
- Department of Molecular Imaging and Theranostics, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, Chiba-shi, Chiba, Japan
| | - Ming-Rong Zhang
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, Chiba-shi, Chiba, Japan
| | - Tetsuya Suhara
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology (QST), 4-9-1 Anagawa, Inage-ku, Chiba-shi, Chiba, 263-8555, Japan
| | - Motoaki Nakamura
- Medical Institute of Developmental Disabilities Research, Showa University, Setagaya-ku, Tokyo, Japan
- Kanagawa Psychiatric Center, Yokohama-shi, Kanagawa, Japan
| | - Nobumasa Kato
- Medical Institute of Developmental Disabilities Research, Showa University, Setagaya-ku, Tokyo, Japan
| | - Yuhei Takado
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology (QST), 4-9-1 Anagawa, Inage-ku, Chiba-shi, Chiba, 263-8555, Japan
| | - Hidehiko Takahashi
- Department of Psychiatry and Behavioral Sciences, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
- Center for Brain Integration Research, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
| | - Makoto Higuchi
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology (QST), 4-9-1 Anagawa, Inage-ku, Chiba-shi, Chiba, 263-8555, Japan
| |
Collapse
|
15
|
Elhawary NA, AlJahdali IA, Abumansour IS, Azher ZA, Falemban AH, Madani WM, Alosaimi W, Alghamdi G, Sindi IA. Phenotypic variability to medication management: an update on fragile X syndrome. Hum Genomics 2023; 17:60. [PMID: 37420260 PMCID: PMC10329374 DOI: 10.1186/s40246-023-00507-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 07/03/2023] [Indexed: 07/09/2023] Open
Abstract
This review discusses the discovery, epidemiology, pathophysiology, genetic etiology, molecular diagnosis, and medication-based management of fragile X syndrome (FXS). It also highlights the syndrome's variable expressivity and common comorbid and overlapping conditions. FXS is an X-linked dominant disorder associated with a wide spectrum of clinical features, including but not limited to intellectual disability, autism spectrum disorder, language deficits, macroorchidism, seizures, and anxiety. Its prevalence in the general population is approximately 1 in 5000-7000 men and 1 in 4000-6000 women worldwide. FXS is associated with the fragile X messenger ribonucleoprotein 1 (FMR1) gene located at locus Xq27.3 and encodes the fragile X messenger ribonucleoprotein (FMRP). Most individuals with FXS have an FMR1 allele with > 200 CGG repeats (full mutation) and hypermethylation of the CpG island proximal to the repeats, which silences the gene's promoter. Some individuals have mosaicism in the size of the CGG repeats or in hypermethylation of the CpG island, both produce some FMRP and give rise to milder cognitive and behavioral deficits than in non-mosaic individuals with FXS. As in several monogenic disorders, modifier genes influence the penetrance of FMR1 mutations and FXS's variable expressivity by regulating the pathophysiological mechanisms related to the syndrome's behavioral features. Although there is no cure for FXS, prenatal molecular diagnostic testing is recommended to facilitate early diagnosis. Pharmacologic agents can reduce some behavioral features of FXS, and researchers are investigating whether gene editing can be used to demethylate the FMR1 promoter region to improve patient outcomes. Moreover, clustered regularly interspaced palindromic repeats (CRISPR)/Cas9 and developed nuclease defective Cas9 (dCas9) strategies have promised options of genome editing in gain-of-function mutations to rewrite new genetic information into a specified DNA site, are also being studied.
Collapse
Affiliation(s)
- Nasser A. Elhawary
- Department of Medical Genetics, College of Medicine, Umm Al-Qura University, Mecca, 21955 Saudi Arabia
| | - Imad A. AlJahdali
- Department of Community Medicine, College of Medicine, Umm Al-Qura University, Mecca, Saudi Arabia
| | - Iman S. Abumansour
- Department of Medical Genetics, College of Medicine, Umm Al-Qura University, Mecca, 21955 Saudi Arabia
| | - Zohor A. Azher
- Department of Medical Genetics, College of Medicine, Umm Al-Qura University, Mecca, 21955 Saudi Arabia
| | - Alaa H. Falemban
- Department of Pharmacology and Toxicology, College of Medicine, Umm Al-Qura University, Mecca, 24382 Saudi Arabia
| | - Wefaq M. Madani
- Department of Hematology and Immunology, Faculty of Medicine, Umm Al-Qura University, Mecca, Saudi Arabia
| | - Wafaa Alosaimi
- Department of Hematology, Maternity and Children Hospital, Mecca, Saudi Arabia
| | - Ghydda Alghamdi
- Department of Medical Genetics, College of Medicine, Umm Al-Qura University, Mecca, 21955 Saudi Arabia
| | - Ikhlas A. Sindi
- Department of Biology, Faculty of Science, King Abdulaziz University, Jeddah, 21589 Saudi Arabia
- Preparatory Year Program, Batterjee Medical College, Jeddah, 21442 Saudi Arabia
| |
Collapse
|
16
|
Vockley J, Burton B, Jurecka A, Ganju J, Leiro B, Zori R, Longo N. Challenges and strategies for clinical trials in propionic and methylmalonic acidemias. Mol Genet Metab 2023; 139:107612. [PMID: 37245378 DOI: 10.1016/j.ymgme.2023.107612] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 05/19/2023] [Accepted: 05/19/2023] [Indexed: 05/30/2023]
Abstract
Clinical trial development in rare diseases poses significant study design and methodology challenges, such as disease heterogeneity and appropriate patient selection, identification and selection of key endpoints, decisions on study duration, choice of control groups, selection of appropriate statistical analyses, and patient recruitment. Therapeutic development in organic acidemias (OAs) shares many challenges with other inborn errors of metabolism, such as incomplete understanding of natural history, heterogenous disease presentations, requirement for sensitive outcome measures and difficulties recruiting a small sample of participants. Here, we review strategies for the successful development of a clinical trial to evaluate treatment response in propionic and methylmalonic acidemias. Specifically, we discuss crucial decisions that may significantly impact success of the study, including patient selection, identification and selection of endpoints, determination of the study duration, consideration of control groups including natural history controls, and selection of appropriate statistical analyses. The significant challenges associated with designing a clinical trial in rare disease can sometimes be successfully met through strategic engagement with experts in the rare disease, seeking regulatory and biostatistical guidance, and early involvement of patients and families.
Collapse
Affiliation(s)
- Jerry Vockley
- Division Medical Genetics, Department of Pediatrics, University of Pittsburgh, School of Medicine, Center for Rare Disease Therapy, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Barbara Burton
- Ann & Robert H. Lurie Children's Hospital, Chicago, IL, USA
| | - Agnieszka Jurecka
- CoA Therapeutics, Inc., a BridgeBio company, San Francisco, CA, USA.
| | - Jitendra Ganju
- Independent Consultant to BridgeBio, San Francisco, CA, USA
| | - Beth Leiro
- Independent Consultant to BridgeBio, San Francisco, CA, USA
| | - Roberto Zori
- Department of Pediatrics, Division of Genetics and Metabolism, University of Florida, Gainesville, FL, USA
| | - Nicola Longo
- Division of Medical Genetics, Department of Pediatrics, University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
17
|
Abbeduto L, Del Hoyo Soriano L, Berry-Kravis E, Sterling A, Edgin JO, Abdelnur N, Drayton A, Hoffmann A, Hamilton D, Harvey DJ, Thurman AJ. Expressive language sampling and outcome measures for treatment trials in fragile X and down syndromes: composite scores and psychometric properties. Sci Rep 2023; 13:9267. [PMID: 37286643 PMCID: PMC10247708 DOI: 10.1038/s41598-023-36087-3] [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: 08/14/2022] [Accepted: 05/29/2023] [Indexed: 06/09/2023] Open
Abstract
The lack of psychometrically sound outcome measures has been a barrier to evaluating the efficacy of treatments proposed for core symptoms of intellectual disability (ID). Research on Expressive Language Sampling (ELS) procedures suggest it is a promising approach to measuring treatment efficacy. ELS entails collecting samples of a participant's talk in interactions with an examiner that are naturalistic but sufficiently structured to ensure consistency and limit examiner effects on the language produced. In this study, we extended previous research on ELS by analyzing an existing dataset to determine whether psychometrically adequate composite scores reflecting multiple dimensions of language can be derived from ELS procedures administered to 6- to 23-year-olds with fragile X syndrome (n = 80) or Down syndrome (n = 78). Data came from ELS conversation and narration procedures administered twice in a 4-week test-retest interval. We found that several composites emerged from variables indexing syntax, vocabulary, planning processes, speech articulation, and talkativeness, although there were some differences in the composites for the two syndromes. Evidence of strong test-retest reliability and construct validity of two of three composites were obtained for each syndrome. Situations in which the composite scores would be useful in evaluating treatment efficacy are outlined.
Collapse
Affiliation(s)
- Leonard Abbeduto
- MIND Institute and Department of Psychiatry and Behavioral Sciences, University of California Davis Health, 2828 50Th St., Sacramento, CA, 95817, USA.
| | - Laura Del Hoyo Soriano
- MIND Institute and Department of Psychiatry and Behavioral Sciences, University of California Davis Health, 2828 50Th St., Sacramento, CA, 95817, USA
| | | | - Audra Sterling
- Waisman Center and Department of Communication Sciences and Disorders, University of Wisconsin-Madison, Madison, WI, USA
| | - Jamie O Edgin
- Department of Psychology, Sonoran UCEDD, UA Family and Community Medicine, University of Arizona, Phoenix, AZ, USA
| | - Nadia Abdelnur
- MIND Institute and Department of Psychiatry and Behavioral Sciences, University of California Davis Health, 2828 50Th St., Sacramento, CA, 95817, USA
| | - Andrea Drayton
- MIND Institute and Department of Psychiatry and Behavioral Sciences, University of California Davis Health, 2828 50Th St., Sacramento, CA, 95817, USA
| | - Anne Hoffmann
- Department of Communication Disorders and Sciences, Rush University, Chicago, IL, USA
- Department of Pediatrics, Rush University, Chicago, IL, USA
| | - Debra Hamilton
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Danielle J Harvey
- Department of Public Health Sciences, University of California, Davis, USA
| | - Angela John Thurman
- MIND Institute and Department of Psychiatry and Behavioral Sciences, University of California Davis Health, 2828 50Th St., Sacramento, CA, 95817, USA
| |
Collapse
|
18
|
Neul JL, Percy AK, Benke TA, Berry-Kravis EM, Glaze DG, Marsh ED, Lin T, Stankovic S, Bishop KM, Youakim JM. Trofinetide for the treatment of Rett syndrome: a randomized phase 3 study. Nat Med 2023; 29:1468-1475. [PMID: 37291210 PMCID: PMC10287558 DOI: 10.1038/s41591-023-02398-1] [Citation(s) in RCA: 65] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 05/12/2023] [Indexed: 06/10/2023]
Abstract
Rett syndrome is a rare, genetic neurodevelopmental disorder. Trofinetide is a synthetic analog of glycine-proline-glutamate, the N-terminal tripeptide of the insulin-like growth factor 1 protein, and has demonstrated clinical benefit in phase 2 studies in Rett syndrome. In this phase 3 study ( https://clinicaltrials.gov identifier NCT04181723 ), females with Rett syndrome received twice-daily oral trofinetide (n = 93) or placebo (n = 94) for 12 weeks. For the coprimary efficacy endpoints, least squares mean (LSM) change from baseline to week 12 in the Rett Syndrome Behaviour Questionnaire for trofinetide versus placebo was -4.9 versus -1.7 (P = 0.0175; Cohen's d effect size, 0.37), and LSM Clinical Global Impression-Improvement at week 12 was 3.5 versus 3.8 (P = 0.0030; effect size, 0.47). For the key secondary efficacy endpoint, LSM change from baseline to week 12 in the Communication and Symbolic Behavior Scales Developmental Profile Infant-Toddler Checklist Social Composite score was -0.1 versus -1.1 (P = 0.0064; effect size, 0.43). Common treatment-emergent adverse events included diarrhea (80.6% for trofinetide versus 19.1% for placebo), which was mostly mild to moderate in severity. Significant improvement for trofinetide compared with placebo was observed for the coprimary efficacy endpoints, suggesting that trofinetide provides benefit in treating the core symptoms of Rett syndrome.
Collapse
Affiliation(s)
- Jeffrey L Neul
- Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Alan K Percy
- University of Alabama at Birmingham, Birmingham, AL, USA
| | - Timothy A Benke
- Children's Hospital of Colorado and University of Colorado School of Medicine, Aurora, CO, USA
| | | | - Daniel G Glaze
- Texas Children's Hospital and Baylor College of Medicine, Houston, TX, USA
| | - Eric D Marsh
- Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Tim Lin
- Acadia Pharmaceuticals Inc., San Diego, CA, USA
| | | | | | | |
Collapse
|
19
|
Saggar M, Bruno JL, Hall SS. Brief intensive social gaze training reorganizes functional brain connectivity in boys with fragile X syndrome. Cereb Cortex 2023; 33:5218-5227. [PMID: 36376964 PMCID: PMC10151883 DOI: 10.1093/cercor/bhac411] [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: 06/20/2022] [Revised: 09/19/2022] [Accepted: 09/20/2022] [Indexed: 11/16/2022] Open
Abstract
Boys with fragile X syndrome (FXS), the leading known genetic cause of autism spectrum disorder (ASD), demonstrate significant impairments in social gaze and associated weaknesses in communication, social interaction, and other areas of adaptive functioning. Little is known, however, concerning the impact of behavioral treatments for these behaviors on functional brain connectivity in this population. As part of a larger study, boys with FXS (mean age 13.23 ± 2.31 years) and comparison boys with ASD (mean age 12.15 ± 2.76 years) received resting-state functional magnetic resonance imaging scans prior to and following social gaze training administered by a trained behavior therapist in our laboratory. Network-agnostic connectome-based predictive modeling of pretreatment resting-state functional connectivity data revealed a set of positive (FXS > ASD) and negative (FXS < ASD) edges that differentiated the groups significantly and consistently across all folds of cross-validation. Following administration of the brief training, the FXS and ASD groups demonstrated reorganization of connectivity differences. The divergence in the spatial pattern of reorganization response, based on functional connectivity differences pretreatment, suggests a unique pattern of response to treatment in the FXS and ASD groups. These results provide further support for implementing targeted behavioral treatments to ameliorate syndrome-specific behavioral features in FXS.
Collapse
Affiliation(s)
- Manish Saggar
- Division of Interdisciplinary Brain Sciences, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA 94304, United States
| | - Jennifer L Bruno
- Division of Interdisciplinary Brain Sciences, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA 94304, United States
| | - Scott S Hall
- Division of Interdisciplinary Brain Sciences, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA 94304, United States
| |
Collapse
|
20
|
Byiers BJ, Merbler AM, Burkitt CC, Beisang A, Symons FJ. Preliminary assessment of the reliability and validity of infrared skin temperature measurements in Rett syndrome. JOURNAL OF INTELLECTUAL DISABILITY RESEARCH : JIDR 2023; 67:387-395. [PMID: 36744445 PMCID: PMC10251747 DOI: 10.1111/jir.13010] [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: 06/06/2022] [Revised: 12/02/2022] [Accepted: 01/04/2023] [Indexed: 06/11/2023]
Abstract
BACKGROUND As clinical trials for Rett syndrome are underway, there is a need to validate potential supplemental outcome measures that reflect important signs and symptoms. Autonomic dysfunction, particularly vasomotor dysfunction, is one potential area for which biomarkers could be developed. METHODS In the current study, infrared thermal images of hands and feet from 26 females with Rett syndrome (aged 62 months to 39 years), and 17 females without known intellectual, genetic or neurological disorders (aged 55 months to 39 years) were collected. Between-group differences in skin temperature, and temporal stability of skin temperature measures in the Rett syndrome group, and relationships between skin temperature measures and parent-reported and researcher-evaluated indicators of autonomic dysfunction were evaluated. RESULTS Between-group differences showed lower hand and foot temperatures in the Rett syndrome group. Hand temperature measurements were stable over time and were moderately correlated with parent-reported autonomic symptoms. Foot temperature measurements were more variable than hand temperatures but showed stronger correlations with parent-reported autonomic symptoms. CONCLUSIONS The results provide preliminary support for the reliability and validity of hand and foot skin temperature measures in Rett syndrome. Additional research is needed to replicate these results and evaluate the temporal stability of these measures over shorter time scales.
Collapse
Affiliation(s)
- B J Byiers
- Department of Educational Psychology, University of Minnesota, Minneapolis, MN, USA
| | - A M Merbler
- Department of Educational Psychology, University of Minnesota, Minneapolis, MN, USA
| | - C C Burkitt
- Gillette Children's Specialty Healthcare, St. Paul, MN, USA
| | - A Beisang
- Gillette Children's Specialty Healthcare, St. Paul, MN, USA
| | - F J Symons
- Department of Educational Psychology, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
21
|
Li R, Bruno JL, Jordan T, Miller JG, Lee CH, Bartholomay KL, Marzelli MJ, Piccirilli A, Lightbody AA, Reiss AL. Aberrant Neural Response During Face Processing in Girls With Fragile X Syndrome: Defining Potential Brain Biomarkers for Treatment Studies. BIOLOGICAL PSYCHIATRY. COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2023; 8:311-319. [PMID: 34555563 PMCID: PMC8964834 DOI: 10.1016/j.bpsc.2021.09.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 09/02/2021] [Accepted: 09/07/2021] [Indexed: 02/02/2023]
Abstract
BACKGROUND Children and adolescents with fragile X syndrome (FXS) manifest significant symptoms of anxiety, particularly in response to face-to-face social interaction. In this study, we used functional near-infrared spectroscopy to reveal a specific pattern of brain activation and habituation in response to face stimuli in young girls with FXS, an important but understudied clinical population. METHODS Participants were 32 girls with FXS (age: 11.8 ± 2.9 years) and a control group of 28 girls without FXS (age: 10.5 ± 2.3 years) matched for age, general cognitive function, and autism symptoms. Functional near-infrared spectroscopy was used to assess brain activation during a face habituation task including repeated upright/inverted faces and greeble (nonface) objects. RESULTS Compared with the control group, girls with FXS showed significant hyperactivation in the frontopolar and dorsal lateral prefrontal cortices in response to all face stimuli (upright + inverted). Lack of neural habituation (and significant sensitization) was also observed in the FXS group in the frontopolar cortex in response to upright face stimuli. Finally, aberrant frontopolar sensitization in response to upright faces in girls with FXS was significantly correlated with notable cognitive-behavioral and social-emotional outcomes relevant to this condition, including executive function, autism symptoms, depression, and anxiety. CONCLUSIONS These findings strongly support a hypothesis of neural hyperactivation and accentuated sensitization during face processing in FXS, a phenomenon that could be developed as a biomarker end point for improving treatment trial evaluation in girls with this condition.
Collapse
Affiliation(s)
- Rihui Li
- Center for Interdisciplinary Brain Sciences Research, Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, California.
| | - Jennifer L Bruno
- Center for Interdisciplinary Brain Sciences Research, Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, California
| | - Tracy Jordan
- Center for Interdisciplinary Brain Sciences Research, Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, California
| | - Jonas G Miller
- Center for Interdisciplinary Brain Sciences Research, Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, California
| | - Cindy H Lee
- Center for Interdisciplinary Brain Sciences Research, Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, California
| | - Kristi L Bartholomay
- Center for Interdisciplinary Brain Sciences Research, Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, California
| | - Matthew J Marzelli
- Center for Interdisciplinary Brain Sciences Research, Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, California
| | - Aaron Piccirilli
- Center for Interdisciplinary Brain Sciences Research, Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, California
| | - Amy A Lightbody
- Center for Interdisciplinary Brain Sciences Research, Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, California
| | - Allan L Reiss
- Center for Interdisciplinary Brain Sciences Research, Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, California; Departments of Radiology and Pediatrics, Stanford University, Stanford, California
| |
Collapse
|
22
|
Shields RH, Kaat A, Sansone SM, Michalak C, Coleman J, Thompson T, McKenzie FJ, Dakopolos A, Riley K, Berry-Kravis E, Widaman KF, Gershon RC, Hessl D. Sensitivity of the NIH Toolbox to Detect Cognitive Change in Individuals With Intellectual and Developmental Disability. Neurology 2023; 100:e778-e789. [PMID: 36460468 PMCID: PMC9984222 DOI: 10.1212/wnl.0000000000201528] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 09/21/2022] [Indexed: 12/04/2022] Open
Abstract
BACKGROUND AND OBJECTIVE Individuals with intellectual disability (ID) experience protracted cognitive development compared with typical youth. Sensitive measurement of cognitive change in this population is a critical need for clinical trials and other intervention studies, but well-validated outcome measures are scarce. This study's aim was to evaluate the sensitivity of the NIH Toolbox Cognition Battery (NIHTB-CB) to detect developmental changes in groups with ID-fragile X syndrome (FXS), Down syndrome (DS), and other ID (OID)-and to provide further support for its use as an outcome measure for treatment trials. METHODS We administered the NIHTB-CB and a reference standard cross-validation measure (Stanford-Binet Intelligence Scales, Fifth Edition [SB5]) to 256 individuals with FXS, DS, and OID (ages 6-27 years). After 2 years of development, we retested 197 individuals. Group developmental changes in each cognitive domain of the NIHTB-CB and SB5 were assessed using latent change score models, and 2-year growth was evaluated at 3 age points (10, 16, and 22 years). RESULTS Overall, effect sizes of growth measured by the NIHTB-CB tests were comparable with or exceeded those of the SB5. The NIHTB-CB showed significant gains in almost all domains in OID at younger ages (10 years), with continued gains at 16 years and stability in early adulthood (22 years). The FXS group showed delayed gains in attention and inhibitory control compared with OID. The DS group had delayed gains in receptive vocabulary compared with OID. Unlike the other groups, DS had significant growth in early adulthood in 2 domains (working memory and attention/inhibitory control). Notably, each group's pattern of NIHTB-CB growth across development corresponded to their respective pattern of SB5 growth. DISCUSSION The NIHTB-CB is sensitive to developmental changes in individuals with ID. Comparison with levels and timing of growth on the cross-validation measure shows that the NIHTB-CB has potential to identify meaningful trajectories across cognitive domains and ID etiologies. Sensitivity to change within the context of treatment studies and delineation of clinically meaningful changes in NIHTB-CB scores, linked to daily functioning, must be established in future research to evaluate the battery more completely as a key outcome measure.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | - David Hessl
- From the MIND Institute and Department of Psychiatry and Behavioral Sciences (R.H.S., S.M.S., F.J.M., A.D., D.H.), University of California Davis, Sacramento; Northwestern University Feinberg School of Medicine (A.K., R.C.G.), Chicago, IL; Rush University Medical Center Departments of Pediatrics (C.M., E.B.), Neurological Sciences and Biochemistry, Chicago, IL; University of Denver Morgridge College of Education (J.C.), Denver, CO; University of Colorado School of Medicine (T.T.), Aurora; Regis University (K.R.), Denver, CO; and University of California Riverside Graduate School of Education (K.F.W.), Riverside.
| |
Collapse
|
23
|
Jones A, Kang S, Shaffer RC, Erickson CA, Schmitt LM. Behavioral inflexibility in fragile X syndrome: Accounts from caregivers and self-advocates. Front Psychol 2023; 14:1118652. [PMID: 36874826 PMCID: PMC9978519 DOI: 10.3389/fpsyg.2023.1118652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 01/25/2023] [Indexed: 02/18/2023] Open
Abstract
Introduction Behavioral difficulties in individuals with fragile X Syndrome (FXS) are one of the primary reasons families seek medical and psychological support. Among these, behavioral inflexibility is very common, and when left untreated, can negatively impact quality of life for the individuals with FXS and their families. Behavioral inflexibility refers to the difficulty in changing one's behaviors based on environmental demands or social contexts, thus impeding daily functioning, opportunities for learning, and social interactions. In addition to the individual and family impact, behavioral inflexibility is often recognized as a defining phenotype of FXS and appears to be specific to FXS when compared to other genetic forms of intellectual disability. Despite the pervasiveness and severity of behavioral inflexibility in FXS, there are limited measures that adequately assess behavioral inflexibility in FXS. Methods We conducted semi-structured virtual focus groups with 22 caregivers, 3 self-advocates, and 1 professional to gather key stakeholders' perspectives on and experiences of inflexible behavior observed in FXS. Audio-recordings from focus groups were transcribed using NVivo, then verified and coded. Two trained professionals reviewed codes to extract primary themes. Results Six themes were extracted: (1) Intolerance of change, (2) Intolerance to uncertainty, (3) Repetitive interests and behaviors, (4) Family impact, (5) Change in behavior across the lifespan, and (6) Impact of the COVID pandemic. Our findings show common examples of these themes included intolerance to disruption to routine, perseverative questioning, watching the same things over and over, and caregivers having to extensively pre-plan for events. Discussion The purpose of the current study was to gain key stakeholders' perspectives via focus groups to elicit information and understand patterns of inflexible behaviors in FXS, with the goal of developing a disorder-specific measure to accurately assess behavioral inflexibility across the lifespan and in response to treatment. We were able to capture several phenotypic examples of behavioral inflexibility in FXS as well as their impact on individuals with FXS and their families. The wealth of information gained through our study will aid in our next steps of item generation for measure development of Ratings of Inflexibility in Genetic Disorders associated with Intellectual Disability - Fragile X Syndrome (RIGID-FX).
Collapse
Affiliation(s)
- Angelina Jones
- Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
| | - Sungeun Kang
- Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
| | - Rebecca C. Shaffer
- Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Craig A. Erickson
- Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
- Department of Psychiatry and Behavioral Neuroscience, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
| | - Lauren M. Schmitt
- Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| |
Collapse
|
24
|
Farmer C, Thurm A, Troy JD, Kaat AJ. Comparing ability and norm-referenced scores as clinical trial outcomes for neurodevelopmental disabilities: a simulation study. J Neurodev Disord 2023; 15:4. [PMID: 36650450 PMCID: PMC9843928 DOI: 10.1186/s11689-022-09474-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 12/24/2022] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND For genetic conditions associated with neurodevelopmental disorder (GCAND), developmental domains such as motor ability, thinking and learning, social abilities, and communication are potential intervention targets. Performance on measures of developmental concepts can be expressed using several types of scores. Norm-referenced scores are intended for the diagnostic context, allowing for the identification of impairment relative to age-based expectations, and can exhibit dramatic floor effects when used in individuals with more significant limitations. Person ability scores, which are derived via Rasch analysis or item response theory, are available on many standardized tests and are intended to measure within-person change. However, they have not been used or evaluated as primary endpoints in GCAND clinical trials. In this study, we simulated a series of parallel-arm clinical trials under several chronological age and impairment conditions, to compare empirically the power and type I error rate of operationalizing test performance using ability scores rather than norm-referenced scores. RESULTS Using the Vineland Adaptive Behavior Scales as the example, we demonstrated an advantage in statistical power of ability scores over norm-referenced scores at extreme levels of impairment. This advantage was at least partially driven by floor effects in norm-referenced scores. For simulated conditions where impairment was less severe, ability scores outperformed norm-referenced scores, but they were more similar. The type I error rate closely approximated the nominal type I error rate of 5% for both scores. CONCLUSION The results of this simulation demonstrate a substantial power and interpretative advantage of ability scores over norm-referenced scores for studies of GCAND that will enroll participants with high levels of impairment. These results are expected to generalize to studies of developmental concepts, regardless of the etiology or specific test. However, the relative advantage of ability scores is expected to be even greater for tests with a higher floor than the Vineland.
Collapse
Affiliation(s)
- Cristan Farmer
- grid.416868.50000 0004 0464 0574Neurodevelopmental and Behavioral Phenotyping Service, National Institute of Mental Health, Bethesda, MD USA
| | - Audrey Thurm
- grid.416868.50000 0004 0464 0574Neurodevelopmental and Behavioral Phenotyping Service, National Institute of Mental Health, Bethesda, MD USA
| | - Jesse D. Troy
- grid.26009.3d0000 0004 1936 7961Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, NC USA
| | - Aaron J. Kaat
- grid.16753.360000 0001 2299 3507Feinberg School of Medicine, Northwestern University, Chicago, IL USA
| |
Collapse
|
25
|
Schmitt LM, Arzuaga AL, Dapore A, Duncan J, Patel M, Larson JR, Erickson CA, Sweeney JA, Ragozzino ME. Parallel learning and cognitive flexibility impairments between Fmr1 knockout mice and individuals with fragile X syndrome. Front Behav Neurosci 2023; 16:1074682. [PMID: 36688132 PMCID: PMC9849779 DOI: 10.3389/fnbeh.2022.1074682] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 12/14/2022] [Indexed: 01/07/2023] Open
Abstract
Introduction Fragile X Syndrome (FXS) is a monogenic condition that leads to intellectual disability along with behavioral and learning difficulties. Among behavioral and learning difficulties, cognitive flexibility impairments are among the most commonly reported in FXS, which significantly impacts daily living. Despite the extensive use of the Fmr1 knockout (KO) mouse to understand molecular, synaptic and behavioral alterations related to FXS, there has been limited development of translational paradigms to understand cognitive flexibility that can be employed in both animal models and individuals with FXS to facilitate treatment development. Methods To begin addressing this limitation, a parallel set of studies were carried out that investigated probabilistic reversal learning along with other behavioral and cognitive tests in individuals with FXS and Fmr1 KO mice. Fifty-five adolescents and adults with FXS (67% male) and 34 age- and sex-matched typically developing controls (62% male) completed an initial probabilistic learning training task and a probabilistic reversal learning task. Results In males with FXS, both initial probabilistic learning and reversal learning deficits were found. However, in females with FXS, we only observed reversal learning deficits. Reversal learning deficits related to more severe psychiatric features in females with FXS, whereas increased sensitivity to negative feedback (lose:shift errors) unexpectedly appear to be adaptive in males with FXS. Male Fmr1 KO mice exhibited both an initial probabilistic learning and reversal learning deficit compared to that of wildtype (WT) mice. Female Fmr1 KO mice were selectively impaired on probabilistic reversal learning. In a prepotent response inhibition test, both male and female Fmr1 KO mice were impaired in learning to choose a non-preferred spatial location to receive a food reward compared to that of WT mice. Neither male nor female Fmr1 KO mice exhibited a change in anxiety compared to that of WT mice. Discussion Together, our findings demonstrate strikingly similar sex-dependent learning disturbances across individuals with FXS and Fmr1 KO mice. This suggests the promise of using analogous paradigms of cognitive flexibility across species that may speed treatment development to improve lives of individuals with FXS.
Collapse
Affiliation(s)
- Lauren M. Schmitt
- Division of Behavioral Medicine and Clinical Psychology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Anna L. Arzuaga
- Department of Psychology, University of Illinois Chicago, Chicago, IL, United States
| | - Ashley Dapore
- Department of Psychiatry, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
| | - Jason Duncan
- Department of Psychology, University of Illinois Chicago, Chicago, IL, United States
| | - Maya Patel
- Department of Psychology, University of Illinois Chicago, Chicago, IL, United States
| | - John R. Larson
- Department of Psychiatry, University of Illinois Chicago, Chicago, IL, United States
| | - Craig A. Erickson
- Department of Psychiatry, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States,Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - John A. Sweeney
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Michael E. Ragozzino
- Department of Psychology, University of Illinois Chicago, Chicago, IL, United States,*Correspondence: Michael E. Ragozzino,
| |
Collapse
|
26
|
Busner J, Pandina G, Day S, Mahableshwarkar A, Kempf L, Sheean M, Dunn J. Patient Centricity: Design and Conduct of Clinical Trials in Orphan Diseases: Third of Three Sets of Expanded Proceedings from the 2020 ISCTM Autumn Conference on Pediatric Drug Development. INNOVATIONS IN CLINICAL NEUROSCIENCE 2023; 20:25-31. [PMID: 37122576 PMCID: PMC10132279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
This article expands on a session, titled "Patient Centricity: Design and Conduct of Clinical Trials in Orphan Diseases," that was presented as part of a two-day meeting on Pediatric Drug Development at the International Society for Central Nervous System (CNS) Clinical Trials and Methodology (ISCTM) Autumn Conference in Boston, Massachusetts, in October 2020. Speakers from various areas of pediatric drug development addressed a variety of implications of including children in drug development programs, including implications for rare/orphan diseases. The speakers have written summaries of their talks. The session's lead Chair was Dr. Joan Busner, who wrote introductory and closing comments. Dr. Simon Day, regulatory consultant, outlined some of the past mistakes that have plagued trials that did not consult with patient groups in the early design phase. Dr. Atul Mahableshwarkar provided an industry perspective of a recent trial that benefited from the inclusion of patient input. Drs. Lucas Kempf and Maria Sheean provided regulatory input from the perspectives of the United States (US) Food and Drug Administration (FDA) and European Medicines Agency (EMA), respectively. Dr. Judith Dunn outlined a novel approach for assessing and rank ordering patient and clinician clinical meaningfulness and the disconnect that may occur. Dr. Busner provided closing comments, tied together the presented issues, and provided a synopsis of the lively discussion that followed the session. In addition to the speakers above, the discussion included two representatives from patient advocacy groups, as well as an additional speaker who described the challenges of conducting a pediatric trial in the US and European Union (EU), given the often competing regulatory requirements. This article should serve as an expert-informed reference to those interested and involved in CNS drug development programs that are aimed at children and rare diseases and seek to ensure a patient-centric approach.
Collapse
Affiliation(s)
- Joan Busner
- Dr. Busner is with Signant Health in Blue Bell, Pennsylvania and Department of Psychiatry, Virginia Commonwealth University School of Medicine in Richmond, Virginia
| | - Gahan Pandina
- Dr. Pandina is with Janssen Pharmaceuticals in Titusville, New Jersey
| | - Simon Day
- Dr. Day is with Clinical Trials Consulting and Training, Ltd. in North Marston, England, United Kingdom. At the time of this writing
| | - Atul Mahableshwarkar
- Dr. Mahableshwarkar was with Emalex Biosciences; he is now with ARM Pharma Consulting in Deerfield, Illinois
| | | | - Maria Sheean
- Dr. Sheean is with the European Medicines Agency in Amsterdam, North Holland, Netherlands
| | - Judith Dunn
- Dr. Dunn is with Fulcrum Therapeutics in Cambridge, Massachusetts
| |
Collapse
|
27
|
Abstract
The histories of targeted treatment trials in fragile X syndrome (FXS) are reviewed in animal studies and human trials. Advances in understanding the neurobiology of FXS have identified a number of pathways that are dysregulated in the absence of FMRP and are therefore pathways that can be targeted with new medication. The utilization of quantitative outcome measures to assess efficacy in multiple studies has improved the quality of more recent trials. Current treatment trials including the use of cannabidiol (CBD) topically and metformin orally have positive preliminary data, and both of these medications are available clinically. The use of the phosphodiesterase inhibitor (PDE4D), BPN1440, which raised the level of cAMP that is low in FXS has very promising results for improving cognition in adult males who underwent a controlled trial. There are many more targeted treatments that will undergo trials in FXS, so the future looks bright for new treatments.
Collapse
Affiliation(s)
- Devon Johnson
- MIND Institute, University of California Davis Health, Sacramento, CA, USA
| | - Courtney Clark
- MIND Institute, University of California Davis Health, Sacramento, CA, USA
| | - Randi Hagerman
- MIND Institute, University of California Davis Health, Sacramento, CA, USA
- Department of Pediatrics, University of California Davis Health, Sacramento, CA, USA
| |
Collapse
|
28
|
Hoffmann A. Communication in fragile X syndrome: Patterns and implications for assessment and intervention. Front Psychol 2022; 13:929379. [PMID: 36619013 PMCID: PMC9817301 DOI: 10.3389/fpsyg.2022.929379] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 12/05/2022] [Indexed: 12/24/2022] Open
Abstract
Fragile X syndrome (FXS) is the most common cause of inherited intellectual disability and is associated with a high rate of autism diagnosis. Language delays have been noted in the areas of overall communication and the specific areas of receptive, expressive, and pragmatic language, as well as in development of speech sounds and literacy. It has been widely noted that those individuals with a diagnosis of both FXS and autism tend to have more significant intellectual disability and language disorder. In this study, the research exploring the FXS language phenotype is presented, and the roles of cognition, autistic symptomatology, and gender are highlighted as possible. Implications for assessment and intervention approaches based on the strengths and weaknesses of the FXS language phenotype are provided.
Collapse
Affiliation(s)
- Anne Hoffmann
- Department of Communication Disorders and Sciences, Rush University Medical Center, Chicago, IL, United States,Department of Pediatrics, Rush University Medical Center, Chicago, IL, United States,*Correspondence: Anne Hoffmann,
| |
Collapse
|
29
|
Jalnapurkar I, Frazier JA, Roth M, Cochran DM, Foley A, Merk T, Venuti L, Ronco L, Raines S, Cadavid D. The feasibility and utility of hair follicle sampling to measure FMRP and FMR1 mRNA in children with or without fragile X syndrome: a pilot study. J Neurodev Disord 2022; 14:57. [PMID: 36494616 PMCID: PMC9733195 DOI: 10.1186/s11689-022-09465-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 10/26/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Fragile X syndrome (FXS) is the most common inherited cause of intellectual disability in males and the most common single gene cause of autism. This X-linked disorder is caused by an expansion of a trinucleotide CGG repeat (> 200 base pairs) on the promotor region of the fragile X messenger ribonucleoprotein 1 gene (FMR1). This leads to the deficiency or absence of the encoded protein, fragile X messenger ribonucleoprotein 1 (FMRP). FMRP has a central role in the translation of mRNAs involved in synaptic connections and plasticity. Recent studies have demonstrated the benefit of therapeutics focused on reactivation of the FMR1 locus towards improving key clinical phenotypes via restoration of FMRP and ultimately disease modification. A key step in future studies directed towards this effort is the establishment of proof of concept (POC) for FMRP reactivation in individuals with FXS. For this, it is key to determine the feasibility of repeated collection of tissues or fluids to measure FMR1 mRNA and FMRP. METHODS Individuals, ages 3 to 22 years of age, with FXS and those who were typically developing participated in this single-site pilot clinical biomarker study. The repeated collection of hair follicles was compared with the collection of blood and buccal swabs for detection of FMR1 mRNA and FMRP and related molecules. RESULTS There were n = 15 participants, of whom 10 had a diagnosis of FXS (7.0 ± 3.56 years) and 5 were typically developing (8.2 ± 2.77 years). Absolute levels of FMRP and FMR1 mRNA were substantially higher in healthy participants compared to full mutation and mosaic FXS participants and lowest in the FXS boys. Measurement of FMR1 mRNA and FMRP levels by any method did not show any notable variation by collection location at home versus office across the various sample collection methodologies of hair follicle, blood sample, and buccal swab. CONCLUSION Findings demonstrated that repeated sampling of hair follicles in individuals with FXS, in both, home, and office settings, is feasible, repeatable, and can be used for measurement of FMR1 mRNA and FMRP in longitudinal studies.
Collapse
Affiliation(s)
- Isha Jalnapurkar
- grid.410516.70000 0001 0707 2056Department of Psychiatry, EK Shriver Center, University of Massachusetts Medical School, Worcester, MA USA
| | - Jean A. Frazier
- grid.410516.70000 0001 0707 2056Department of Psychiatry, EK Shriver Center, University of Massachusetts Medical School, Worcester, MA USA
| | - Mark Roth
- grid.509699.a0000 0004 5907 6392Fulcrum Therapeutics, Cambridge, MA USA
| | - David M. Cochran
- grid.410516.70000 0001 0707 2056Department of Psychiatry, EK Shriver Center, University of Massachusetts Medical School, Worcester, MA USA
| | - Ann Foley
- grid.410516.70000 0001 0707 2056Department of Psychiatry, EK Shriver Center, University of Massachusetts Medical School, Worcester, MA USA
| | - Taylor Merk
- grid.410516.70000 0001 0707 2056Department of Psychiatry, EK Shriver Center, University of Massachusetts Medical School, Worcester, MA USA
| | - Lauren Venuti
- grid.410516.70000 0001 0707 2056Department of Psychiatry, EK Shriver Center, University of Massachusetts Medical School, Worcester, MA USA
| | - Lucienne Ronco
- grid.509699.a0000 0004 5907 6392Fulcrum Therapeutics, Cambridge, MA USA
| | - Shane Raines
- grid.509699.a0000 0004 5907 6392Fulcrum Therapeutics, Cambridge, MA USA
| | - Diego Cadavid
- grid.509699.a0000 0004 5907 6392Fulcrum Therapeutics, Cambridge, MA USA
| |
Collapse
|
30
|
Alusi G, Berry-Kravis E, Nelson D, Orefice LL, Booker SA. Emerging Therapeutic Strategies for Fragile X Syndrome: Q&A. ACS Chem Neurosci 2022; 13:3544-3546. [PMID: 36475635 PMCID: PMC9782331 DOI: 10.1021/acschemneuro.2c00674] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Understanding how best to treat aspects of Fragile X syndrome has the potential to improve the quality of life of affected individuals. Such an effective therapy has, as yet, remained elusive. In this article, we ask those researching or affected by Fragile X syndrome their views on the current state of research and from where they feel the most likely therapy may emerge.
Collapse
Affiliation(s)
| | - Elizabeth Berry-Kravis
- Department
of Pediatrics, Neurological Sciences, and Anatomy and Cell Biology, Rush University Medical Center, Chicago, Illinois60612, United States
| | - David Nelson
- Molecular
and Human Genetics, Baylor College of Medicine, Houston, Texas77030, United States
| | - Lauren L. Orefice
- Department
of Molecular Biology, Massachusetts General
Hospital, Boston, Massachusetts02114, United States,Department
of Genetics, Harvard Medical School, Boston, Massachusetts02115, United States
| | - Sam A. Booker
- Simons
Initiative
for the Developing Brain, University of
Edinburgh, EdinburghEH8 9XD, U.K.,
| |
Collapse
|
31
|
Lightbody AA, Bartholomay KL, Jordan T, Lee CH, Miller JG, Reiss AL. Anxiety, Depression, and Social Skills in Girls with Fragile X Syndrome: Understanding the Cycle to Improve Outcomes. J Dev Behav Pediatr 2022; 43:e565-e572. [PMID: 36219483 PMCID: PMC9712149 DOI: 10.1097/dbp.0000000000001128] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 07/04/2022] [Indexed: 11/26/2022]
Abstract
OBJECTIVE Female patients with fragile X syndrome (FXS), a genetic condition associated with a mutation in the FMR1 gene, are at significantly elevated risk for developing anxiety and depression. This study is designed to better understand these symptoms in school-age girls, particularly as they relate to age, social skills, and functional outcomes. METHODS We compared 58 girls aged 6 to 16 years with FXS with 46 age-matched, sex-matched, and developmentally matched peers without FXS on measures of anxiety, depression, social skills, adaptive behavior, and quality of life. RESULTS Girls with FXS 10.5 years and older demonstrated significantly higher levels of depression, withdrawal, and social avoidance than girls younger than 10.5 years with FXS ( p -values < 0.01). Girls in the comparison group did not show any age-related differences on these measures. The older FXS cohort also showed associations between social communication and interaction skills, adaptive behavior, and measures of anxiety and depression ( p -values < 0.05) not seen in the comparison group, regardless of age. CONCLUSION We found that age seems to play an important role in the development of mood symptoms and that such symptoms are uniquely correlated with social communication and reciprocal social interaction behaviors and adaptive functioning in girls with FXS after puberty. These data suggest a critical window of intervention for girls with FXS in the improvement of social interaction skills and the prevention of social avoidance and symptoms of anxiety and depression, with the ultimate goal of improving quality of life and promoting greater independence.
Collapse
Affiliation(s)
- Amy A. Lightbody
- Department of Psychiatry and Behavioral Sciences, Division of Interdisciplinary Brain Sciences, Stanford University, Stanford, CA 94305
| | - Kristi L. Bartholomay
- Department of Psychiatry and Behavioral Sciences, Division of Interdisciplinary Brain Sciences, Stanford University, Stanford, CA 94305
| | - Tracy Jordan
- Department of Psychiatry and Behavioral Sciences, Division of Interdisciplinary Brain Sciences, Stanford University, Stanford, CA 94305
| | - Cindy H. Lee
- Department of Psychiatry and Behavioral Sciences, Division of Interdisciplinary Brain Sciences, Stanford University, Stanford, CA 94305
| | - Jonas G. Miller
- Department of Psychiatry and Behavioral Sciences, Division of Interdisciplinary Brain Sciences, Stanford University, Stanford, CA 94305
| | - Allan L. Reiss
- Department of Psychiatry and Behavioral Sciences, Division of Interdisciplinary Brain Sciences, Stanford University, Stanford, CA 94305
- Department of Radiology, Stanford University, Stanford, CA 94305
- Department of Pediatrics, Stanford University, Stanford, CA 94305
| |
Collapse
|
32
|
Arpone M, Bretherton L, Amor DJ, Hearps SJC, Rogers C, Field MJ, Hunter MF, Santa Maria L, Alliende AM, Slee J, Godler DE, Baker EK. Agreement between parents' and clinical researchers' ratings of behavioral problems in children with fragile X syndrome and chromosome 15 imprinting disorders. RESEARCH IN DEVELOPMENTAL DISABILITIES 2022; 131:104338. [PMID: 36179574 DOI: 10.1016/j.ridd.2022.104338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 08/04/2022] [Accepted: 09/12/2022] [Indexed: 06/16/2023]
Abstract
BACKGROUND Despite the increasing number of clinical trials involving children with neurodevelopmental disorders, appropriate and objective outcome measures for behavioral symptoms are still required. AIM This study assessed the agreement between parents' and clinical researchers' ratings of behavioral problem severity in children with fragile X syndrome (FXS) and chromosome 15 imprinting disorders. METHODS AND PROCEDURES The cohort comprised 123 children (64% males), aged 3-17 years, with FXS (n = 79), Prader-Willi (PWS; n = 19), Angelman (AS; n = 15), and Chromosome 15q duplication (n = 10) syndromes. Specific items from the Autism Diagnostic Observation Schedule-Second Edition and Aberrant Behavior Checklist-Community Edition mapping to corresponding behavioral domains were selected ad-hoc, to assess behavioral problems. OUTCOMES AND RESULTS Inter-rater agreement for the cohort was slight for self-injury (Intraclass Correlation Coefficient (ICC) = 0.12), fair for tantrums/aggression (0.24) and mannerisms/stereotypies (0.25), and moderate for hyperactivity (0.48). When stratified by diagnosis, ICC ranged from poor (0; self-injury, AS and PWS) to substantial (0.48; hyperactivity, females with FXS). CONCLUSIONS AND IMPLICATIONS The high level of inter-rater disagreement across most domains suggests that parents' and researchers' assessments led to discrepant appraisal of behavioral problem severity. These findings have implications for treatment targets and outcome measure selection in clinical trials, supporting a multi-informant approach.
Collapse
Affiliation(s)
- Marta Arpone
- Diagnosis and Development, Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, VIC, Australia; Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, VIC, Australia; Brain and Mind, Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, VIC, Australia
| | - Lesley Bretherton
- Brain and Mind, Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, VIC, Australia
| | - David J Amor
- Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, VIC, Australia; Neurodisability and Rehabilitation, Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, VIC, Australia
| | - Stephen J C Hearps
- Brain and Mind, Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, VIC, Australia; Department of Critical Care, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, VIC, Australia
| | - Carolyn Rogers
- Genetics of Learning Disability Service, Hunter Genetics, Waratah, NSW, Australia
| | - Michael J Field
- Genetics of Learning Disability Service, Hunter Genetics, Waratah, NSW, Australia
| | - Matthew F Hunter
- Monash Genetics, Monash Health, Melbourne, VIC, Australia; Department of Paediatrics, Monash University, Melbourne, VIC, Australia
| | - Lorena Santa Maria
- Laboratory of Molecular Cytogenetics, Department of Genetics and Metabolic Diseases, Institute of Nutrition and Food Technology (INTA), University of Chile, Santiago, Chile
| | - Angelica M Alliende
- Laboratory of Molecular Cytogenetics, Department of Genetics and Metabolic Diseases, Institute of Nutrition and Food Technology (INTA), University of Chile, Santiago, Chile
| | - Jennie Slee
- Department of Health, Government of Western Australia, Genetic Services of Western Australia, Perth, Australia
| | - David E Godler
- Diagnosis and Development, Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, VIC, Australia; Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, VIC, Australia
| | - Emma K Baker
- Diagnosis and Development, Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, VIC, Australia; Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, VIC, Australia; School of Psychology and Public Health, La Trobe University, Bundoora, VIC, Australia.
| |
Collapse
|
33
|
Berry-Kravis E, Hagerman R, Budimirovic D, Erickson C, Heussler H, Tartaglia N, Cohen J, Tassone F, Dobbins T, Merikle E, Sebree T, Tich N, Palumbo JM, O’Quinn S. A randomized, controlled trial of ZYN002 cannabidiol transdermal gel in children and adolescents with fragile X syndrome (CONNECT-FX). J Neurodev Disord 2022; 14:56. [PMID: 36434514 PMCID: PMC9700889 DOI: 10.1186/s11689-022-09466-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 11/03/2022] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Fragile X syndrome (FXS) is associated with dysregulated endocannabinoid signaling and may therefore respond to cannabidiol therapy. DESIGN CONNECT-FX was a double-blind, randomized phase 3 trial assessing efficacy and safety of ZYN002, transdermal cannabidiol gel, for the treatment of behavioral symptoms in children and adolescents with FXS. METHODS Patients were randomized to 12 weeks of ZYN002 (250 mg or 500 mg daily [weight-based]) or placebo, as add-on to standard of care. The primary endpoint assessed change in social avoidance (SA) measured by the Aberrant Behavior Checklist-Community Edition FXS (ABC-CFXS) SA subscale in a full cohort of patients with a FXS full mutation, regardless of the FMR1 methylation status. Ad hoc analyses assessed efficacy in patients with ≥ 90% and 100% methylation of the promoter region of the FMR1 gene, in whom FMR1 gene silencing is most likely. RESULTS A total of 212 patients, mean age 9.7 years, 75% males, were enrolled. A total of 169 (79.7%) patients presented with ≥ 90% methylation of the FMR1 promoter and full mutation of FMR1. Although statistical significance for the primary endpoint was not achieved in the full cohort, significant improvement was demonstrated in patients with ≥ 90% methylation of FMR1 (nominal P = 0.020). This group also achieved statistically significant improvements in Caregiver Global Impression-Change in SA and isolation, irritable and disruptive behaviors, and social interactions (nominal P-values: P = 0.038, P = 0.028, and P = 0.002). Similar results were seen in patients with 100% methylation of FMR1. ZYN002 was safe and well tolerated. All treatment-emergent adverse events (TEAEs) were mild or moderate. The most common treatment-related TEAE was application site pain (ZYN002: 6.4%; placebo: 1.0%). CONCLUSIONS In CONNECT-FX, ZYN002 was well tolerated in patients with FXS and demonstrated evidence of efficacy with a favorable benefit risk relationship in patients with ≥ 90% methylation of the FMR1 gene, in whom gene silencing is most likely, and the impact of FXS is typically most severe. TRIAL REGISTRATION The CONNECT-FX trial is registered on Clinicaltrials.gov (NCT03614663).
Collapse
Affiliation(s)
- Elizabeth Berry-Kravis
- grid.240684.c0000 0001 0705 3621Departments of Pediatrics and Neurological Sciences, Rush University Medical Center, Chicago, IL USA
| | - Randi Hagerman
- grid.413079.80000 0000 9752 8549Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, University of California-Davis Medical Center, Sacramento, CA USA ,grid.27860.3b0000 0004 1936 9684Department of Pediatrics, University of California Davis School of Medicine, Sacramento, CA USA
| | - Dejan Budimirovic
- grid.21107.350000 0001 2171 9311Departments of Psychiatry and Child Psychiatry, Fragile X Clinic, Kennedy Krieger Institute/the Johns Hopkins Medical Institutions, Baltimore, MD USA ,grid.21107.350000 0001 2171 9311Department of Psychiatry & Behavioral Sciences-Child Psychiatry, Johns Hopkins School of Medicine, Baltimore, MD USA
| | - Craig Erickson
- grid.24827.3b0000 0001 2179 9593Department of Psychiatry and Behavioral Neuroscience, Cincinnati Children’s Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH USA
| | - Helen Heussler
- grid.512914.a0000 0004 0642 3960Centre for Clinical Trials in Rare Neurodevelopmental Disorders, Children’s Health Queensland, Brisbane, Australia ,grid.1003.20000 0000 9320 7537Centre for Child Health Research, University of Queensland, Brisbane, Australia
| | - Nicole Tartaglia
- Department of Pediatrics, Developmental Pediatrics, University of Colorado School of Medicine, Children’s Hospital Colorado, Aurora, CO USA
| | - Jonathan Cohen
- Fragile X Alliance Inc, North Caulfield, VIC, Australia ,grid.1002.30000 0004 1936 7857Centre for Developmental Disability Health Victoria, Monash University, Clayton, VIC Australia
| | - Flora Tassone
- grid.413079.80000 0000 9752 8549Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, University of California-Davis Medical Center, Sacramento, CA USA ,grid.413079.80000 0000 9752 8549Department of Biochemistry and Molecular Medicine, School of Medicine, University of California-Davis, Sacramento, CA USA
| | | | | | - Terri Sebree
- grid.422480.80000 0004 8307 0679Zynerba Pharmaceuticals Inc., Devon, PA USA
| | - Nancy Tich
- grid.422480.80000 0004 8307 0679Zynerba Pharmaceuticals Inc., Devon, PA USA
| | - Joseph M. Palumbo
- grid.422480.80000 0004 8307 0679Zynerba Pharmaceuticals Inc., Devon, PA USA
| | - Stephen O’Quinn
- grid.422480.80000 0004 8307 0679Zynerba Pharmaceuticals Inc., Devon, PA USA
| |
Collapse
|
34
|
Wilkinson EH, Britton TC, Hall SS. Examining Phenotypic Differences in Gaze Avoidance Between Autism Spectrum Disorder and Fragile X Syndrome. AMERICAN JOURNAL ON INTELLECTUAL AND DEVELOPMENTAL DISABILITIES 2022; 127:435-454. [PMID: 36306410 PMCID: PMC9667749 DOI: 10.1352/1944-7558-127.6.435] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 08/31/2021] [Indexed: 05/22/2023]
Abstract
We examined potential phenotypic differences in eye gaze avoidance exhibited by boys with autism spectrum disorder (ASD) and boys with fragile X syndrome (FXS). In Study 1, the Eye Contact Avoidance Scale (ECAS) was administered to caregivers of boys aged 7-18 years with FXS (n = 148), ASD (n = 168), and mixed developmental disabilities (MDD; n = 128). In Study 2, subsets of boys with FXS (n = 31) and boys with ASD (n = 25) received a brief behavioral treatment probe to improve eye contact. Results showed that boys with FXS obtained significantly higher scores on the ECAS compared to boys with ASD and MDD. Exposure to the brief behavioral treatment probe resulted in significant decreases in scores for boys with FXS, but not for boys with ASD.
Collapse
Affiliation(s)
- Ellen H Wilkinson
- Ellen H. Wilkinson, Tobias C. Britton, and Scott S. Hall, Stanford University School of Medicine
| | - Tobias C Britton
- Ellen H. Wilkinson, Tobias C. Britton, and Scott S. Hall, Stanford University School of Medicine
| | - Scott S Hall
- Ellen H. Wilkinson, Tobias C. Britton, and Scott S. Hall, Stanford University School of Medicine
| |
Collapse
|
35
|
Budimirovic DB, Protic DD. Fragile X Syndrome: Recent Research Updates toward Capturing Treatments’ Improvement in Clinical Trials. Brain Sci 2022; 12:brainsci12101276. [PMID: 36291209 PMCID: PMC9599739 DOI: 10.3390/brainsci12101276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 09/19/2022] [Indexed: 11/18/2022] Open
Affiliation(s)
- Dejan B. Budimirovic
- Department of Psychiatry, Fragile X Clinic, Kennedy Krieger Institute, Baltimore, MD 21205, USA
- Department of Psychiatry & Behavioral Sciences-Child Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Correspondence:
| | | |
Collapse
|
36
|
Laroui A, Galarneau L, Abolghasemi A, Benachenhou S, Plantefève R, Bouchouirab FZ, Lepage JF, Corbin F, Çaku A. Clinical significance of matrix metalloproteinase-9 in Fragile X Syndrome. Sci Rep 2022; 12:15386. [PMID: 36100610 PMCID: PMC9470743 DOI: 10.1038/s41598-022-19476-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 08/30/2022] [Indexed: 11/12/2022] Open
Abstract
High plasma matrix metalloproteases-9 (MMP-9) levels have been reported in Fragile X Syndrome in a limited number of animal and human studies. Since the results obtained are method-dependent and not directly comparable, the clinical utility of MMP-9 measurement in FXS remains unclear. This study aimed to compare quantitative gel zymography and ELISA and to determine which method better discriminates abnormal MMP-9 levels of individuals with FXS from healthy controls and correlates with the clinical profile. The active and total forms of MMP-9 were quantified respectively, by gel zymography and ELISA in a cohort of FXS (n = 23) and healthy controls (n = 20). The clinical profile was assessed for the FXS group using the Aberrant Behavior Checklist FXS adapted version (ABC-CFX), Adaptive Behavior Assessment System (ABAS), Social Communication Questionnaire (SCQ), and Anxiety Depression and Mood Scale questionnaires. Method comparison showed a disagreement between gel zymography and ELISA with a constant error of − 0.18 [95% CI: − 0.35 to − 0.02] and a proportional error of 2.31 [95% CI: 1.53 to 3.24]. Plasma level of MMP-9 active form was significantly higher in FXS (n = 12) as compared to their age-sex and BMI matched controls (n = 12) (p = 0.039) and correlated with ABC-CFX (rs = 0.60; p = 0.039) and ADAMS (rs = 0.57; p = 0.043) scores. As compared to the plasma total form, the plasma MMP-9 active form better enables the discrimination of individuals with FXS from controls and correlates with the clinical profile. Our results highlight the importance of choosing the appropriate method to quantify plasma MMP-9 in future FXS clinical studies.
Collapse
|
37
|
Schmitt LM, Sweeney JA, Erickson CA, Shaffer R. Brief Report: Feasibility of the Probabilistic Reversal Learning Task as an Outcome Measure in an Intervention Trial for Individuals with Autism Spectrum Disorder. J Autism Dev Disord 2022; 52:4191-4199. [PMID: 34557984 PMCID: PMC8459822 DOI: 10.1007/s10803-021-05288-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/08/2021] [Indexed: 11/25/2022]
Abstract
Cognitive flexibility deficits are a hallmark feature of autism spectrum disorder (ASD), but few evidence-based behavioral interventions have successfully addressed this treatment target. Outcome measurement selection may help account for previous findings. The probabilistic reversal learning task (PRL) is a measure of cognitive flexibility previously validated for use in ASD, but its use as an outcome measure has not yet been assessed. The current study examined the feasibility, reproducibility, and sensitivity of PRL in a within-subjects trial of Regulating Together, a group-based intervention targeting emotion regulation. We demonstrated the PRL is highly feasible, showed test-retest reproducibility, and is sensitive to detect change following the intervention. Our findings demonstrate the PRL task may be a useful outcome measure of cognitive flexibility in future intervention trials in ASD.
Collapse
Affiliation(s)
- Lauren M Schmitt
- Cincinnati Children's Hospital Medical Center, Cincinnati, USA.
- College of Medicine, University of Cincinnati, Cincinnati, USA.
| | - John A Sweeney
- College of Medicine, University of Cincinnati, Cincinnati, USA
| | - Craig A Erickson
- Cincinnati Children's Hospital Medical Center, Cincinnati, USA
- College of Medicine, University of Cincinnati, Cincinnati, USA
| | - Rebecca Shaffer
- Cincinnati Children's Hospital Medical Center, Cincinnati, USA
- College of Medicine, University of Cincinnati, Cincinnati, USA
| |
Collapse
|
38
|
Li R, Bruno JL, Lee CH, Bartholomay KL, Sundstrom J, Piccirilli A, Jordan T, Miller JG, Lightbody AA, Reiss AL. Aberrant brain network and eye gaze patterns during natural social interaction predict multi-domain social-cognitive behaviors in girls with fragile X syndrome. Mol Psychiatry 2022; 27:3768-3776. [PMID: 35595977 DOI: 10.1038/s41380-022-01626-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 05/07/2022] [Accepted: 05/12/2022] [Indexed: 02/08/2023]
Abstract
Girls with fragile X syndrome (FXS) often manifest significant symptoms of avoidance, anxiety, and arousal, particularly in the context of social interaction. However, little is currently known about the associations among neurobiological, biobehavioral such as eye gaze pattern, and social-cognitive dysfunction in real-world settings. In this study, we sought to characterize brain network properties and eye gaze patterns in girls with FXS during natural social interaction. Participants included 42 girls with FXS and 31 age- and verbal IQ-matched girls (control). Portable functional near-infrared spectroscopy (fNIRS) and an eye gaze tracker were used to investigate brain network alterations and eye gaze patterns associated with social-cognitive dysfunction in girls with FXS during a structured face-to-face conversation. Compared to controls, girls with FXS showed significantly increased inter-regional functional connectivity and greater excitability within the prefrontal cortex (PFC), frontal eye field (FEF) and superior temporal gyrus (STG) during the conversation. Girls with FXS showed significantly less eye contact with their conversational partner and more unregulated eye gaze behavior compared to the control group. We also demonstrated that a machine learning approach based on multimodal data, including brain network properties and eye gaze patterns, was predictive of multiple domains of social-cognitive behaviors in girls with FXS. Our findings expand current knowledge of neural mechanisms and eye gaze behaviors underlying naturalistic social interaction in girls with FXS. These results could be further evaluated and developed as intermediate phenotypic endpoints for treatment trial evaluation in girls with FXS.
Collapse
Affiliation(s)
- Rihui Li
- Center for Interdisciplinary Brain Sciences Research, Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, 94305, USA.
| | - Jennifer L Bruno
- Center for Interdisciplinary Brain Sciences Research, Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, 94305, USA
| | - Cindy H Lee
- Center for Interdisciplinary Brain Sciences Research, Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, 94305, USA
| | - Kristi L Bartholomay
- Center for Interdisciplinary Brain Sciences Research, Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, 94305, USA
| | - Jamie Sundstrom
- Center for Interdisciplinary Brain Sciences Research, Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, 94305, USA
| | - Aaron Piccirilli
- Center for Interdisciplinary Brain Sciences Research, Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, 94305, USA
| | - Tracy Jordan
- Center for Interdisciplinary Brain Sciences Research, Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, 94305, USA
| | - Jonas G Miller
- Center for Interdisciplinary Brain Sciences Research, Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, 94305, USA
| | - Amy A Lightbody
- Center for Interdisciplinary Brain Sciences Research, Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, 94305, USA
| | - Allan L Reiss
- Center for Interdisciplinary Brain Sciences Research, Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, 94305, USA
- Departments of Radiology and Pediatrics, Stanford University, Stanford, CA, 94305, USA
- Department of Pediatrics, Stanford University, Stanford, CA, 94305, USA
| |
Collapse
|
39
|
Loveall SJ, Channell MM, Mattie LJ, Barkhimer AE. Inclusion of Individuals With Neurodevelopmental Disorders in Norm-Referenced Language Assessments. Front Psychol 2022; 13:929433. [PMID: 36033059 PMCID: PMC9412819 DOI: 10.3389/fpsyg.2022.929433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 06/09/2022] [Indexed: 11/13/2022] Open
Abstract
Standardized, norm-referenced language assessment tools are used for a variety of purposes, including in education, clinical practice, and research. Unfortunately, norm-referenced language assessment tools can demonstrate floor effects (i.e., a large percentage of individuals scoring at or near the lowest limit of the assessment tool) when used with some groups with neurodevelopmental disorders (NDDs), such as individuals with intellectual disability and neurogenetic syndromes. Without variability at the lower end of these assessment tools, professionals cannot accurately measure language strengths and difficulties within or across individuals. This lack of variability may be tied to poor representation of individuals with NDDs in normative samples. Therefore, the purpose of this study was to identify and examine common standardized, norm-referenced language assessment tools to report the representation of individuals with NDDs in normative samples and the range of standard/index scores provided. A systematic search identified 57 assessment tools that met inclusion criteria. Coding of the assessment manuals identified that most assessment tools included a “disability” or “exceptionality” group in their normative sample. However, the total number of individuals in these groups and the number of individuals with specific NDDs was small. Further, the characteristics of these groups (e.g., demographic information; disability type) were often poorly defined. The floor standard/index scores of most assessment tools were in the 40s or 50s. Only four assessment tools provided a standard score lower than 40. Findings of this study can assist clinicians, educators, and researchers in their selections of norm-referenced assessment tools when working with individuals with NDDs.
Collapse
Affiliation(s)
- Susan J. Loveall
- Department of Special Education and Communication Disorders, University of Nebraska–Lincoln, Lincoln, NE, United States
- *Correspondence: Susan J. Loveall,
| | - Marie Moore Channell
- Department of Speech and Hearing Science, University of Illinois at Urbana-Champaign, Champaign, IL, United States
| | - Laura J. Mattie
- Department of Speech and Hearing Science, University of Illinois at Urbana-Champaign, Champaign, IL, United States
| | - Alexandria E. Barkhimer
- Department of Speech and Hearing Science, University of Illinois at Urbana-Champaign, Champaign, IL, United States
| |
Collapse
|
40
|
Klusek J, O'Connor SL, Hickey A, Hills KJ, Abbeduto L, Roberts JE. Attention/Deficit Hyperactivity Disorder in Adolescent and Young Adult Males With Fragile X Syndrome. AMERICAN JOURNAL ON INTELLECTUAL AND DEVELOPMENTAL DISABILITIES 2022; 127:213-230. [PMID: 35443049 PMCID: PMC9414675 DOI: 10.1352/1944-7558-127.3.213] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 10/26/2021] [Indexed: 05/03/2023]
Abstract
This study characterized the rates of attention-deficit/hyperactivity disorder (ADHD) in adolescent and young adult males with fragile X syndrome (FXS) using a multi-method approach integrating a DSM-based parent interview (Children's Interview for Psychiatric Syndromes; P-ChIPS, Fristad et al., 1998) and a parent rating scale (Child Behavior Checklist; CBCL, Achenbach, 2001). Thirty-one males with FXS, aged 16-24 years, participated. Forty-two percent met DSM-5 criteria for ADHD and 35% exceeded the CBCL cut-offs. Agreement between the two classification methods was fair (κ = 0.38). Autism symptom severity and nonverbal cognitive ability did not predict ADHD diagnoses/symptoms. Results show high rates of ADHD in males with FXS during late adolescence and young adulthood, which are not accounted for by impaired nonverbal cognitive skills or autism symptom severity. DSM-based ADHD-specific scales are recommended over broadband symptom scales to improve accurate identification.
Collapse
Affiliation(s)
- Jessica Klusek
- Jessica Klusek, Shannon L. O'Connor, Alexandra Hickey and Kimberly J. Hills, University of South Carolina
| | - Shannon L O'Connor
- Jessica Klusek, Shannon L. O'Connor, Alexandra Hickey and Kimberly J. Hills, University of South Carolina
| | - Alexandra Hickey
- Jessica Klusek, Shannon L. O'Connor, Alexandra Hickey and Kimberly J. Hills, University of South Carolina
| | - Kimberly J Hills
- Jessica Klusek, Shannon L. O'Connor, Alexandra Hickey and Kimberly J. Hills, University of South Carolina
| | | | | |
Collapse
|
41
|
Sotoudeh Anvari M, Vasei H, Najmabadi H, Badv RS, Golipour A, Mohammadi-Yeganeh S, Salehi S, Mohamadi M, Goodarzynejad H, Mowla SJ. Identification of microRNAs associated with human fragile X syndrome using next-generation sequencing. Sci Rep 2022; 12:5011. [PMID: 35322102 PMCID: PMC8943156 DOI: 10.1038/s41598-022-08916-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 03/15/2022] [Indexed: 11/09/2022] Open
Abstract
Fragile X syndrome (FXS) is caused by a mutation in the FMR1 gene which can lead to a loss or shortage of the FMR1 protein. This protein interacts with specific miRNAs and can cause a range of neurological disorders. Therefore, miRNAs could act as a novel class of biomarkers for common CNS diseases. This study aimed to test this theory by exploring the expression profiles of various miRNAs in Iranian using deep sequencing-based technologies and validating the miRNAs affecting the expression of the FMR1 gene. Blood samples were taken from 15 patients with FXS (9 males, 6 females) and 12 controls. 25 miRNAs were differentially expressed in individuals with FXS compared to controls. Levels of 9 miRNAs were found to be significantly changed (3 upregulated and 6 downregulated). In Patients, the levels of hsa-miR-532-5p, hsa-miR-652-3p and hsa-miR-4797-3p were significantly upregulated while levels of hsa-miR-191-5p, hsa-miR-181-5p, hsa-miR-26a-5p, hsa-miR-30e-5p, hsa-miR-186-5p, and hsa-miR-4797-5p exhibited significant downregulation; and these dysregulations were confirmed by RT-qPCR. This study presents among the first evidence of altered miRNA expression in blood samples from patients with FXS, which could be used for diagnostic, prognostic, and treatment purposes. Larger studies are required to confirm these preliminary results.
Collapse
Affiliation(s)
- Maryam Sotoudeh Anvari
- Department of Molecular Pathology, School of Medicine, Children's Medical Center, Pediatrics Center of Excellence, Tehran University of Medical Sciences, Tehran, Iran.
| | - Hamed Vasei
- Department of Mathematical Science, Sharif University of Technology, Tehran, Iran
| | - Hossein Najmabadi
- Department of Genetics, School of Rehabilitation Sciences, Genetic Research Center, The University of Social Welfare and Rehabilitation Sciences, Tehran, Iran
| | - Reza Shervin Badv
- Department of Pediatrics, School of Medicine, Children's Medical Center, Pediatrics Center of Excellence, Tehran University of Medical Sciences, Tehran, Iran
| | - Akram Golipour
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Samira Mohammadi-Yeganeh
- Medical Nanotechnology and Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Saeede Salehi
- Cell-Based Therapies Research Center, Digestive Diseases Research Institute, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahmood Mohamadi
- Department of Pediatrics, School of Medicine, Children's Medical Center, Pediatrics Center of Excellence, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamidreza Goodarzynejad
- Department of Basic and Clinical Research, Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Javad Mowla
- Department of Molecular Genetics, Faculty of Biological Science, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
42
|
Brašić JR, Goodman JA, Nandi A, Russell DS, Jennings D, Barret O, Martin SD, Slifer K, Sedlak T, Mathur AK, Seibyl JP, Berry-Kravis EM, Wong DF, Budimirovic DB. Fragile X Mental Retardation Protein and Cerebral Expression of Metabotropic Glutamate Receptor Subtype 5 in Men with Fragile X Syndrome: A Pilot Study. Brain Sci 2022; 12:314. [PMID: 35326270 PMCID: PMC8946825 DOI: 10.3390/brainsci12030314] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/26/2022] [Accepted: 02/09/2022] [Indexed: 02/04/2023] Open
Abstract
Multiple lines of evidence suggest that a deficiency of Fragile X Mental Retardation Protein (FMRP) mediates dysfunction of the metabotropic glutamate receptor subtype 5 (mGluR5) in the pathogenesis of fragile X syndrome (FXS), the most commonly known single-gene cause of inherited intellectual disability (ID) and autism spectrum disorder (ASD). Nevertheless, animal and human studies regarding the link between FMRP and mGluR5 expression provide inconsistent or conflicting findings about the nature of those relationships. Since multiple clinical trials of glutamatergic agents in humans with FXS did not demonstrate the amelioration of the behavioral phenotype observed in animal models of FXS, we sought measure if mGluR5 expression is increased in men with FXS to form the basis for improved clinical trials. Unexpectedly marked reductions in mGluR5 expression were observed in cortical and subcortical regions in men with FXS. Reduced mGluR5 expression throughout the living brains of men with FXS provides a clue to examine FMRP and mGluR5 expression in FXS. In order to develop the findings of our previous study and to strengthen the objective tools for future clinical trials of glutamatergic agents in FXS, we sought to assess the possible value of measuring both FMRP levels and mGluR5 expression in men with FXS. We aimed to show the value of measurement of FMRP levels and mGluR5 expression for the diagnosis and treatment of individuals with FXS and related conditions. We administered 3-[18F]fluoro-5-(2-pyridinylethynyl)benzonitrile ([18F]FPEB), a specific mGluR5 radioligand for quantitative measurements of the density and the distribution of mGluR5s, to six men with the full mutation (FM) of FXS and to one man with allele size mosaicism for FXS (FXS-M). Utilizing the seven cortical and subcortical regions affected in neurodegenerative disorders as indicator variables, adjusted linear regression of mGluR5 expression and FMRP showed that mGluR5 expression was significantly reduced in the occipital cortex and the thalamus relative to baseline (anterior cingulate cortex) if FMRP levels are held constant (F(7,47) = 6.84, p < 0.001).These findings indicate the usefulness of cerebral mGluR5 expression measured by PET with [18F]FPEB and FMRP values in men with FXS and related conditions for assessments in community facilities within a hundred-mile radius of a production center with a cyclotron. These initial results of this pilot study advance our previous study regarding the measurement of mGluR5 expression by combining both FMRP levels and mGluR5 expression as tools for meaningful clinical trials of glutamatergic agents for men with FXS. We confirm the feasibility of this protocol as a valuable tool to measure FMRP levels and mGluR5 expression in clinical trials of individuals with FXS and related conditions and to provide the foundations to apply precision medicine to tailor treatment plans to the specific needs of individuals with FXS and related conditions.
Collapse
Affiliation(s)
- James Robert Brašić
- Section of High Resolution Brain Positron Emission Tomography Imaging, Division of Nuclear Medicine and Molecular Imaging, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; (A.N.); (S.D.M.); (T.S.); (A.K.M.); (D.F.W.)
| | - Jack Alexander Goodman
- Frank H. Netter MD School of Medicine, Quinnipiac University, North Haven, CT 06473, USA;
| | - Ayon Nandi
- Section of High Resolution Brain Positron Emission Tomography Imaging, Division of Nuclear Medicine and Molecular Imaging, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; (A.N.); (S.D.M.); (T.S.); (A.K.M.); (D.F.W.)
| | - David S. Russell
- Institute for Neurodegenerative Disorders, New Haven, CT 06510, USA; (D.S.R.); (D.J.); (O.B.); (J.P.S.)
- Invicro, New Haven, CT 06510, USA
| | - Danna Jennings
- Institute for Neurodegenerative Disorders, New Haven, CT 06510, USA; (D.S.R.); (D.J.); (O.B.); (J.P.S.)
- Invicro, New Haven, CT 06510, USA
- Denali Therapeutics, Inc., South San Francisco, CA 94080, USA
| | - Olivier Barret
- Institute for Neurodegenerative Disorders, New Haven, CT 06510, USA; (D.S.R.); (D.J.); (O.B.); (J.P.S.)
- Invicro, New Haven, CT 06510, USA
- Laboratoire des Maladies Neurodégénératives, Molecular Imaging Research Center (MIRCen), Institut de Biologie François Jacob, Centre National de la Recherche Scientifique (CNRS), Commissariat à l’Énergie Atomique et aux Énergies Alternatives (CEA), Université Paris-Saclay, CEDEX, 92265 Fontenay-aux-Roses, France
| | - Samuel D. Martin
- Section of High Resolution Brain Positron Emission Tomography Imaging, Division of Nuclear Medicine and Molecular Imaging, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; (A.N.); (S.D.M.); (T.S.); (A.K.M.); (D.F.W.)
- Department of Neuroscience, Zanvyl Krieger School of Arts and Sciences, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Keith Slifer
- Department of Psychiatry and Behavioral Sciences-Child Psychiatry, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA;
- Department of Behavioral Psychology, Kennedy Krieger Institute, Baltimore, MD 21205, USA
| | - Thomas Sedlak
- Section of High Resolution Brain Positron Emission Tomography Imaging, Division of Nuclear Medicine and Molecular Imaging, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; (A.N.); (S.D.M.); (T.S.); (A.K.M.); (D.F.W.)
- Department of Psychiatry and Behavioral Sciences-General Psychiatry, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Anil Kumar Mathur
- Section of High Resolution Brain Positron Emission Tomography Imaging, Division of Nuclear Medicine and Molecular Imaging, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; (A.N.); (S.D.M.); (T.S.); (A.K.M.); (D.F.W.)
| | - John P. Seibyl
- Institute for Neurodegenerative Disorders, New Haven, CT 06510, USA; (D.S.R.); (D.J.); (O.B.); (J.P.S.)
- Invicro, New Haven, CT 06510, USA
| | - Elizabeth M. Berry-Kravis
- Departments of Pediatrics, Neurological Sciences, and Biochemistry, Rush University Medical Center, Chicago, IL 60612, USA;
| | - Dean F. Wong
- Section of High Resolution Brain Positron Emission Tomography Imaging, Division of Nuclear Medicine and Molecular Imaging, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; (A.N.); (S.D.M.); (T.S.); (A.K.M.); (D.F.W.)
- Laboratory of Central Nervous System (CNS) Neuropsychopharmacology and Multimodal, Imaging (CNAMI), Mallinckrodt Institute of Radiology, Washington University, Saint Louis, MO 63110, USA
| | - Dejan B. Budimirovic
- Department of Psychiatry and Behavioral Sciences-Child Psychiatry, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA;
- Department of Psychiatry, Kennedy Krieger Institute, Baltimore, MD 21205, USA
| |
Collapse
|
43
|
Limbu B, Deb S, Roy M, Lee R, Roy A, Taiwo O. Randomised controlled trials of mood stabilisers for people with autism spectrum disorder: systematic review and meta-analysis. BJPsych Open 2022; 8:e52. [PMID: 35197135 PMCID: PMC8935918 DOI: 10.1192/bjo.2022.18] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Revised: 01/08/2022] [Accepted: 01/25/2022] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Despite the widespread use of psychotropic medications in people with autism spectrum disorder (ASD), there is limited evidence to suggest that psychotropic medications including mood stabilisers are effective in individuals with ASD. AIMS To carry out a systematic review and meta-analysis of randomised controlled trials (RCTs) that assessed the effectiveness of mood stabilisers in people with ASD. METHOD We searched the following databases: Cochrane Library, MEDLINE, Embase, CINAHL, PsycINFO, ERIC, DARE, and ClinicalTrials.gov. In addition, we hand-searched 12 relevant journals. We used the Cochrane Risk of Bias and Jadad scores to assess the quality of included RCTs. We carried out a meta-analysis using a random-effects model. RESULTS We included eight RCTs (four on valproate, two on levetiracetam, and one each on lamotrigine and topiramate) that included a total of 310 people with ASD, primarily children. Outcomes were based on core and associated ASD symptoms including irritability and aggression but not bipolar disorder. Only two small studies (25%) from the same group showed definite superiority over placebo and one over psychoeducation alone. Meta-analysis of pooled data on the Aberrant Behaviour Checklist-irritability, Clinical Global Impression Scale-improvement, and Overt Aggression Scale (OAS)/OAS-modified did not show any significant inter-group difference. The rates of adverse effects did not show any significant inter-group difference. CONCLUSIONS Given the methodological flaws in the included studies and the contradictory findings, it is difficult to draw any definitive conclusion about the effectiveness of mood stabilisers to treat either ASD core symptoms or associated behaviours. Robust large-scale RCTs are needed in the future to address this issue.PROSPERO registration: CRD42021255467 on 18 May 2021.
Collapse
Affiliation(s)
- Bharati Limbu
- Research Assistant, Department of Brain Sciences, Faculty of Medicine, Imperial College London, UK
| | - Shoumitro Deb
- Visiting Professor of Neuropsychiatry, Department of Brain Sciences, Faculty of Medicine, Imperial College London, UK
| | - Meera Roy
- Honorary Consultant Psychiatrist, Hereford and Worcestershire Health and Care Trust, UK
| | - Rachel Lee
- Specialty Registrar in Psychiatry of Intellectual Disabilities, Coventry and Warwickshire Partnership NHS Foundation Trust, UK
| | - Ashok Roy
- Honorary Professorial Fellow, Warwick Medical School, University of Warwick, UK
| | - Oluwafemi Taiwo
- Core Trainee in Psychiatry, Coventry and Warwickshire Partnership NHS Foundation Trust, UK
| |
Collapse
|
44
|
Protic DD, Aishworiya R, Salcedo-Arellano MJ, Tang SJ, Milisavljevic J, Mitrovic F, Hagerman RJ, Budimirovic DB. Fragile X Syndrome: From Molecular Aspect to Clinical Treatment. Int J Mol Sci 2022; 23:1935. [PMID: 35216055 PMCID: PMC8875233 DOI: 10.3390/ijms23041935] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 02/01/2022] [Accepted: 02/02/2022] [Indexed: 02/01/2023] Open
Abstract
Fragile X syndrome (FXS) is a neurodevelopmental disorder caused by the full mutation as well as highly localized methylation of the fragile X mental retardation 1 (FMR1) gene on the long arm of the X chromosome. Children with FXS are commonly co-diagnosed with Autism Spectrum Disorder, attention and learning problems, anxiety, aggressive behavior and sleep disorder, and early interventions have improved many behavior symptoms associated with FXS. In this review, we performed a literature search of original and review articles data of clinical trials and book chapters using MEDLINE (1990-2021) and ClinicalTrials.gov. While we have reviewed the biological importance of the fragile X mental retardation protein (FMRP), the FXS phenotype, and current diagnosis techniques, the emphasis of this review is on clinical interventions. Early non-pharmacological interventions in combination with pharmacotherapy and targeted treatments aiming to reverse dysregulated brain pathways are the mainstream of treatment in FXS. Overall, early diagnosis and interventions are fundamental to achieve optimal clinical outcomes in FXS.
Collapse
Affiliation(s)
- Dragana D. Protic
- Department of Pharmacology, Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Belgrade, 11129 Belgrade, Serbia
| | - Ramkumar Aishworiya
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute UCDH, University of California Davis, 2825 50th Street, Sacramento, CA 95817, USA; (R.A.); (M.J.S.-A.); (S.J.T.); (R.J.H.)
- Khoo Teck Puat-National University Children’s Medical Institute, National University Health System, 5 Lower Kent Ridge Road, Singapore 119074, Singapore
| | - Maria Jimena Salcedo-Arellano
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute UCDH, University of California Davis, 2825 50th Street, Sacramento, CA 95817, USA; (R.A.); (M.J.S.-A.); (S.J.T.); (R.J.H.)
- Department of Pediatrics, University of California Davis School of Medicine, Sacramento, CA 95817, USA
- Department of Pathology and Laboratory Medicine, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| | - Si Jie Tang
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute UCDH, University of California Davis, 2825 50th Street, Sacramento, CA 95817, USA; (R.A.); (M.J.S.-A.); (S.J.T.); (R.J.H.)
| | - Jelena Milisavljevic
- Faculty of Medicine, University of Belgrade, 11129 Belgrade, Serbia; (J.M.); (F.M.)
| | - Filip Mitrovic
- Faculty of Medicine, University of Belgrade, 11129 Belgrade, Serbia; (J.M.); (F.M.)
| | - Randi J. Hagerman
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute UCDH, University of California Davis, 2825 50th Street, Sacramento, CA 95817, USA; (R.A.); (M.J.S.-A.); (S.J.T.); (R.J.H.)
- Department of Pediatrics, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| | - Dejan B. Budimirovic
- Department of Psychiatry, Fragile X Clinic, Kennedy Krieger Institute, Baltimore, MD 21205, USA
- Department of Psychiatry & Behavioral Sciences-Child Psychiatry, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
45
|
Design and outcome measures of LAVENDER, a phase 3 study of trofinetide for Rett syndrome. Contemp Clin Trials 2022; 114:106704. [PMID: 35149233 DOI: 10.1016/j.cct.2022.106704] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 02/01/2022] [Accepted: 02/02/2022] [Indexed: 11/23/2022]
Abstract
INTRODUCTION Rett syndrome (RTT) is a debilitating neurodevelopmental disorder with no approved treatments. Trofinetide is a synthetic analog of glycine-proline-glutamate, the N-terminal tripeptide of insulin-like growth factor 1. In a phase 2, placebo-controlled trial in 82 females with RTT aged 5-15 years, a significant (p ≤ 0.042) improvement over placebo was observed with the highest trofinetide dose (200 mg/kg twice daily [BID]) on three measures: Rett Syndrome Behavior Questionnaire (RSBQ), Clinical Global Impression-Improvement (CGI-I), and RTT-Clinician Domain Specific Concerns-Visual Analog Scale (RTT-DSC-VAS). Trofinetide was well tolerated at all doses (50, 100, and 200 mg/kg BID). A phase 3 trial utilizing disease-specific and novel scales was designed to investigate the efficacy and safety of trofinetide in girls and women with RTT. METHODS This 12-week, double-blind, randomized, placebo-controlled study (LAVENDER; NCT04181723) will evaluate trofinetide in 187 females, aged 5-20 years, with RTT. Co-primary endpoints are the RSBQ and CGI-I scales. Clinical domains of the CGI-I include communication, ambulation, hand use, seizures, attentiveness, and social (eye contact) and autonomic (breathing) aspects. Secondary endpoints will leverage four novel RTT-specific clinician ratings (derived from the RTT-DSC-VAS) of hand function, ambulation, ability to communicate, and verbal communication, and existing scales, to evaluate other core symptoms of RTT, quality of life and caregiver burden. A 40-week, open-label extension study will follow. DISCUSSION This study was designed using disease-specific scales optimized to demonstrate changes in core symptoms of RTT and may provide the first phase 3 data demonstrating drug efficacy in individuals with RTT. TRIAL REGISTRATION Clinicaltrials.govNCT04181723.
Collapse
|
46
|
del Hoyo Soriano L, Bullard L, Thurman AJ, Alvarez CH, Abbeduto L. Providing a parent-administered outcome measure in a bilingual family of a father and a mother of two adolescents with ASD: brief report. Dev Neurorehabil 2022; 25:140-144. [PMID: 34170787 PMCID: PMC8709875 DOI: 10.1080/17518423.2021.1942281] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 06/01/2021] [Accepted: 06/09/2021] [Indexed: 10/21/2022]
Abstract
There is a need for psychometrically sound outcome measures for treatment studies that have a low burden for families and that are available in multiple languages. We have developed a language assessment in English and Spanish that parents can administer to their children at home via telehealth-delivered procedures. The current case study presents descriptive data on a single family of two parent-child dyads. Both the mother and father from a single family were trained in their primary language (Spanish) on how to administer the Expressive Language Sampling - Narration (ELS-N) in their secondary language (English) to their two English-speaking monolingual sons with ASD through telehealth-delivered procedures. Both parents learned to administer the procedures to a predetermined level of fidelity. Extension to a larger sample of bilingual families is needed for this home-based, parent-administered test; however, the present results suggest feasibility even when the language of training and administration differ.
Collapse
Affiliation(s)
- Laura del Hoyo Soriano
- MIND Institute & Department of Psychiatry and Behavioral Sciences, University of California, Davis, Sacramento, CA, USA
| | - Lauren Bullard
- MIND Institute & Department of Psychiatry and Behavioral Sciences, University of California, Davis, Sacramento, CA, USA
| | - Angela John Thurman
- MIND Institute & Department of Psychiatry and Behavioral Sciences, University of California, Davis, Sacramento, CA, USA
| | - Cesar Hoyos Alvarez
- MIND Institute & Department of Psychiatry and Behavioral Sciences, University of California, Davis, Sacramento, CA, USA
- Department of Spanish and Portuguese, University of California Davis, Davis, USA
| | - Leonard Abbeduto
- MIND Institute & Department of Psychiatry and Behavioral Sciences, University of California, Davis, Sacramento, CA, USA
| |
Collapse
|
47
|
Personalized medicine for rare neurogenetic disorders: can we make it happen? Cold Spring Harb Mol Case Stud 2022; 8:mcs.a006200. [PMID: 35332073 PMCID: PMC8958924 DOI: 10.1101/mcs.a006200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Rare neurogenetic disorders are collectively common, affecting 3% of the population, and often manifest with complex multiorgan comorbidity. With advances in genetic, -omics, and computational analysis, more children can be diagnosed and at an earlier age. Innovations in translational research facilitate the identification of treatment targets and development of disease-modifying drugs such as gene therapy, nutraceuticals, and drug repurposing. This increasingly allows targeted therapy to prevent the often devastating manifestations of rare neurogenetic disorders. In this perspective, successes in diagnosis, prevention, and treatment are discussed with a focus on inherited disorders of metabolism. Barriers for the identification, development, and implementation of rare disease-specific therapies are discussed. New methodologies, care networks, and collaborative frameworks are proposed to optimize the potential of personalized genomic medicine to decrease morbidity and improve lives of these vulnerable patients.
Collapse
|
48
|
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: 11] [Impact Index Per Article: 3.7] [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
|
49
|
Shaffer R, Thurman AJ, Ronco L, Cadavid D, Raines S, Kim SH. Social communication in fragile X syndrome: pilot examination of the Brief Observation of Social Communication Change (BOSCC). J Neurodev Disord 2022; 14:4. [PMID: 35034602 PMCID: PMC8903546 DOI: 10.1186/s11689-021-09411-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 12/08/2021] [Indexed: 12/17/2022] Open
Abstract
Background Social communication is a key area of difficulty in fragile X syndrome (FXS) and there are not yet adequate outcome measurement tools. Appropriate outcome measures for FXS have been identified as a key area of research interest in order to evaluate future therapeutic trials. The Brief Observation of Social Communication Change-Minimally Verbal (BOSCC-MV), an outcome measure with strong psychometrics developed for autism spectrum disorder, has promise as an outcome measure to assess social communication change with FXS participants. Methods We examined the BOSCC-MV via central coders in this multi-site-trial to assess its appropriateness for FXS. Eighteen minimally verbal males ages 3–12 years were enrolled and assessed on two consecutive days and 7 participants completed a third visit 6 months later. We examined test-retest reliability, inter-rater reliability, and both convergent and divergent validity with standard clinical measures including the Autism Diagnostic and Observation Schedule-2, Vineland 3, Social Responsiveness Scale, and the Aberrant Behavior Checklist. Results The BOSCC-MV in FXS demonstrated strong inter-rater and test-retest reliability, comparable to previous trials in idiopathic ASD. Strong convergent validity was found with Autism Diagnostic Observation Schedule-2 and Vineland-3. Divergent validity was demonstrated between BOSCC-MV and unrelated measures. Conclusions The BOSCC-MV shows promise as a FXS social communication outcome measure, warranting further large-scale evaluation.
Collapse
Affiliation(s)
- Rebecca Shaffer
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati School of Medicine, 3333 Burnet Avenue, MLC 4002, Cincinnati, OH, 45229, USA.
| | - Angela John Thurman
- Department of Psychiatry and Behavioral Sciences, University of California Davis Health, MIND Institute, University of California Davis Health, Sacramento, CA, USA
| | - Lucienne Ronco
- Fulcrum Therapeutics, Cambridge, MA, USA.,Deep Genomics Therapeutics, Toronto, ON, Canada
| | - Diego Cadavid
- Fulcrum Therapeutics, Cambridge, MA, USA.,University of Massachusetts Medical School, Worcester, MA, USA
| | | | - So Hyun Kim
- Department of Psychiatry, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
50
|
van der Lei MB, Kooy RF. Therapeutic potential of GABAA receptor subunit expression abnormalities in fragile X syndrome. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2022. [DOI: 10.1080/23808993.2021.2008168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
| | - R. Frank Kooy
- Department of Medical Genetics, University of Antwerp, Antwerp, Belgium
| |
Collapse
|