492
Views
0
CrossRef citations to date
0
Altmetric
Review

Pharmacological management of fragile X syndrome: a systematic review and narrative summary of the current evidence

, , , , &
Pages 301-313 | Received 17 Dec 2023, Accepted 22 Feb 2024, Published online: 18 Mar 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.

1. Introduction

The FMR1 (fragile X messenger ribonucleoprotein 1) gene (full mutation > 200 CGG repeats) is located on chromosome X (q27.3) [Citation1] Mutations in the FMR1 gene cause large expansion of the CGG trinucleotide repeat. This leads to decreased or absent levels of FMRP (fragile X messenger ribonucleoprotein) [Citation2]. Fragile X syndrome (FXS) is considered the most common inherited cause of Intellectual Disability (ID). A systematic review and meta-analysis found the prevalence of full mutation to be 1.4 (95% CI: 0.1–3.1) per 10,000 males and 0.9 (95% CI: 0.0–2.9) per 10,000 females [Citation3]. FXS has a broad phenotypic spectrum and is associated with cognitive deficits, behavioral challenges, and physical characteristics [Citation4]. The penetrance of the physical and behavioral phenotype is dependent on the level of mutation (full mutation/premutation/premutation carrier/carrier/methylation mosaicism) and the subsequent impact upon FMRP levels.

Males with FXS generally have a behavioral phenotype including a level of ID, anxiety, and traits consistent with other neurodevelopmental conditions, including autism (shyness/repetitive stereotyped behaviors/hyper-arousal/sensory needs) and attention deficit hyperactivity disorder (ADHD) (attention deficit/impulsivity). For females with FXS, the phenotype is generally milder (25% ID), with less behavioral challenges [Citation5]. Almost one in three males with FXS may meet the formal criteria for autism, and almost all will have notable traits [Citation6]. The etiology for autism is multifactorial, with numerous candidate genes and genetic variants that may contribute. However, given the level of association of autism with FXS, it has been considered as a surrogate to target autistic features.

FXS has been the target for investigations into pharmacological treatments for a number of reasons [Citation6]. Firstly, there was focus on supplementing identified deficits (folic acid) through dietary intake. Secondly, to target the associated neurodevelopmental/neuropsychiatric disorder (FXAND). This also extends to behaviors that challenge (BtC), with a systematic review identifying prevalence estimates of self-injurious behavior (SIB) at 10–81%, and aggression at 12.5–60.9%, higher than rates estimated in the general ID population [Citation7]. Furthermore, a study of children and adults with FXS (58 males, 39 females) found that 86% of males and 77% of females met the DSM-IV (Diagnostic and Statistical Manual of Mental Disorders IV) criteria for any anxiety disorder [Citation8]. Thirdly, there has been the development of molecules targeted at normalizing/reducing the impact of FMR1 on synaptic connectivity based upon molecular understanding. The impact of these molecules is often measured against more fundamental cognitive functions [Citation6].

The aim of this review was to systematically consider the current evidence base for pharmacological treatment in FXS and provide a narrative summary. This review focused strictly on FXS populations only and aimed to identify high-quality interventional studies. The focus of this review was on efficacy of the pharmacological intervention for clinically relevant outcomes.

2. Methods

We conducted a systematic search strategy of electronic databases Embase, Medline, CINAHL, and PsychINFO (Appendix 1. Example search strategy). The search dates were 1 January 1980 to 2March 2023. The search did not include any other registers or gray literature. The ancestry method was utilized, to identify any potentially eligible articles within the references of articles selected for inclusion. The search was limited to English language journal articles.

2.1. Inclusion criteria

Study design: Randomized, blinded, placebo controlled clinical trials. (RCTs)

Population: Fragile X (only) Excluding FXTAS (Fragile-X Associated Tremor/Ataxia Syndrome)

Intervention: Pharmacological only

Outcomes: Validated objective psychological or behavioral assessment tools with clinical relevance, excluding basic physiological parameters

Focus on efficacy over safety and tolerability.

Published in a peer-reviewed journal (excluding conference abstracts/posters)

We did not include studies with mixed population (unless data could be separated), reviews (unless systematic and meeting inclusion criteria), studies with multiple simultaneous interventions, post-hoc analyses of already considered data, or basic biological parameters.

2.2. Study selection

The search strategy was conducted by an information specialist and clinical academic. The initial abstract results were screened by two reviewers against the inclusion criteria. Full-text reviews were conducted blind (each reviewer blind to results of the other reviewer) by the same two reviewers. Any conflicts were then discussed against the inclusion criteria with the team. The references of included studies were screened for any missed studies that met the inclusion criteria. Final decisions on inclusion were made by the information specialist and clinical academic.

The results were presented in a descriptive narrative. The level of evidence assessed against the Oxford Centre for Evidence Based Medicine (OCEBM) Levels of evidence [Citation9]. For the context of this review, Level 1 evidence consists of good-quality RCTs. A systematic review of RCTs with homogeneity being the highest level of evidence (1a). Level 2 evidence includes cohort studies or lower powered individual RCTs (2b).

3. Results

The literature search identified 2377 articles for screening. Duplicates were removed (364 articles), leaving 1733 abstracts. Following screening against the including criteria, 42 articles were identified for full-text screening (1731 removed). Full text screening identified 22 articles for potential inclusion, of which 16 studies were finally included. Articles were excluded at full-text review for not being a peer reviewed journal article (7); editorials (5); no clinical application of results (5); post-hoc data already considered (3); wrong population (3); no randomization (1); dual intervention (1); pharmaceutical company communication (1) ().

Figure 1. PRISMA flow diagram.

Figure 1. PRISMA flow diagram.

3.1. Included studies

The 16 studies included in this review are summarized in . The focus of this review was to consider the efficacy of the pharmacological interventions on meaningful clinical parameters using validated measures. The results are reported alongside appraisal of the evidence.

Table 1. Summary of studies included in final narrative review (n = 16).

3.1.1. Folic acid

Level of Evidence: 2b

Risk of Bias: Small sample size, no ITT, crossover study design.

Tolerability: Not formally measured

Efficacy: No significant difference to placebo

Disorders of the metabolism of folate and folate deficiency is known to be associated with a range of neurological problems, not specific to FXS. It was hypothesized that FXS was associated with folic acid deficiency/problems with metabolism. This review identified one randomized, double-blind, placebo-controlled trial including 25 males with FXS [Citation10]. This was a six-month cross over study. The dose of Folic acid was 5 mg twice daily. The results showed no significant difference to placebo in performance overall on any of the validated psychological or behavioral outcome measures. Two participants were removed from the study due to significant deterioration in functioning when folic acid was removed (treatment then continued). Four pre-pubertal participants in each treatment group completed psychological testing (IQ score) and showed significant improvement with folic acid compared to placebo. This suggests a possible response in younger children with FXS, however the magnitude/relevance of this IQ change is not clearly reported. Furthermore, the low IQ scores (average IQ 27) reduce the accuracy of testing.

3.1.2. Methylphenidate

Level of Evidence: 2b

Risk of Bias: Small sample size, no ITT, cross over design, sort intervention period.

Tolerability: No difference on side effects scores between methylphenidate and placebo.

Efficacy: Significant improvement on attention and social skills factor domains.

This review identified one randomized, double-blind, placebo-controlled trial [Citation18]. This was a crossover study design with three phases (placebo/methylphenidate/dextroamphetamine). Each intervention was assigned randomly for a one-week period. The methylphenidate dose was 0.3 mg/kg twice daily. The total number of participants was 15 (13 males, 2 females). The only statistically significant findings were improvement on the domains of attention (p = 0.025) and social skills factor (p = 0.034) improved on methylphenidate. There were no significant differences between dextroamphetamine and placebo. Ten of the 15 participants were considered responders and continued with intervention. However, there was a strong trend for improvement across all other variables.

3.1.3. Folinic acid (leucovorin)

Level of Evidence: 2b

Risk of Bias: Small sample size, crossover design.

Tolerability: No side effect reported with folinic acid

Efficacy: No significant difference with placebo on any psychological or behavioral assessment tool

Folinic acid is the active metabolite of folic acid. As a result, it skips the stages of folic acid activation. It is hypothesized that it may have therapeutic advantages over taking folic acid. This review identified one randomized, double-blind, placebo-controlled trial [Citation21]. A cross over design, with intervals of 12 weeks. The study included 21 male participants. Only 16 participants completed both phases of the study. There were no statistically significant differences between folinic acid and placebo on any of the validated assessment tools.

3.1.4. AMPA-Modulating CX516

Level of Evidence: 2b

Risk of Bias: Small sample size, no ITT, short intervention period.

Tolerability: No significant difference to placebo. No serious adverse events. One withdrawal on CX516 due to potentially serious rash.

Efficacy: No significant difference to placebo.

CX516 is an Ampakine compound with evidence to support potential utility for cognitive deficits. AMPA-receptor activity may be deficient in FXS. Ampakine’s may enhance AMPA-related neurotransmission and promote BDNF mediated synaptic maturation. Therefore, CX516 is a targeted treatment that may impact upon the behavioral phenotype of FXS [Citation23]. This review identified one study including 49 participants (38 males, 11 females). This study was a 4-week randomized, double-blind placebo-controlled trial. There were no significant differences between CX516 and placebo across any of the validated assessment tools.

3.1.5. L-acetylcarnitine

Level of Evidence: 2b

Risk of Bias: No ITT

Tolerability: No significant side effects identified.

Efficacy: Significant improvement across primary outcome measures (clinical global impression)

L-acetylcarnitine (LAC) has been considered as a possible alternative to stimulant treatment for ADHD symptoms in FXS. LAC has been shown to reduce hyperactivity, an impulsive behavior in association with ADHD, including within a small male FXS population [Citation44,Citation45]. LAC is thought to act on the FMR1 promotor. This review identified one study including 56 males with FXS and ADHD [Citation26]. This was a multicentre, randomized, double-blind, parallel, placebo-controlled trial over 12 months. Only 51 were included in the final analysis (24 LAC, 27 placebos). The dose of LAC was 1000mg/day. The results show significant improvement in the treatment group for the primary outcome measures. This study builds on a previous review from the same group; this was published as a letter to the editor and so did not meet the inclusion criteria for this review [Citation44].

3.1.6. Melatonin

Level of Evidence: 2b

Risk of Bias: No ITT, mixed population, crossover design, accuracy of technology, sleep measures

Tolerability: No significant side effects identified.

Efficacy: Significant improvement in sleep-onset time.

Melatonin is a pharmacological treatment for sleep disorders. Melatonin is secreted from the pineal gland at night and regulates sleep in accordance with circadian rhythm. Autism and FXS sleep disorders are common, and melatonin levels can vary resulting in abnormal circadian rhythm, due to a range of postulated mechanisms. This review identified one study including 18 participants (16 males, 2 females) with clinically reported sleep problem [Citation27]. This was a randomized, double-blind, placebo-controlled study, with a 4-week cross over design. The melatonin dose was 3 mg. The results show a significant difference in sleep onset time, although not necessarily to a clinically relevant level.

3.1.7. Donepezil

Level of Evidence: 1b

Risk of Bias: No ITT, short duration

Tolerability: No significant side effects identified.

Efficacy: No significant difference to placebo on cognitive or behavioral measures

It has been postulated that cholinergic channels may play a role in some of the phenotypic features of FXS. It has been demonstrated in animal models with FX that there is cholinergic dysfunction in memory and learning regions of the limbic system [Citation46]. This review identified two studies with Donepezil as the treatment.

The first study included 20 participants, all male [Citation28]. This was a randomized, double-blind, placebo controlled, parallel group study. The duration was 12 weeks. The dose of Donepezil was 2.5 mg daily for 4 weeks, increasing to 5 mg daily for 8 weeks. There was no difference to placebo on cognitive or behavioral measures.

The second study included 41 participants (27 males, 15 females), with a focus on basic cognitive functions and including functional MRI imaging [Citation38]. Participants were randomized to receive Donepezil (2.5–10 mg) or placebo for 12 weeks. There was no significant difference to placebo on cognitive or behavioral outcomes.

3.1.8. Minocycline

Level of Evidence: 2b

Risk of Bias: Blinding inconsistent, crossover design

Tolerability: No serious adverse events, no significant difference to placebo

Efficacy: Significant improvement in primary outcome measure based on ITT, although difference modest and may not be clinically relevant.

Minocycline is thought to be a neuroprotective agent. In FXS, the lack of FMRP is associated with alterations in the expressions of a number of other proteins including MMP9 (matrix metalloproteinase 9). MMP9 plays a role in the synaptic function and plasticity in the hippocampus. Levels may be raised in association with FXS. Minocycline treatment has been found to lower these levels in animal models [Citation47]. This review identified one study including 55 participants [Citation29]. This was a randomized, double-blind, placebo-controlled, crossover trial, 3 months for each phase. The results showed a statistically significant improvement in clinical global impression; however, results may not be clinically relevant.

3.1.9. Mavoglurant

Level of Evidence: 2b

Risk of Bias: Short duration, validity of assessment tools

Tolerability: Adverse events were higher in the treatment group, and at higher dosing. Discontinuation was higher in the treatment group. The most common adverse effects were psychiatric.

Efficacy: No significant difference to placebo in any primary outcome measure in adolescent or adult cohort.

Metabotropic glutamate receptor (mGluR) signaling may be overactive in FXS and may lead to increased long-term depression in the hippocampus. This is a postulated theory for the FXS phenotype [Citation48]. This review identified one article including two studies or cohorts with similar design (adolescent cohort and adult cohort) [Citation30]. The studies included 175 adults (age 18–45 years) and 139 adolescents (age 12–17 years). The duration of both studies was 12 weeks. The dose of Mavoglurant was 25, 50, or 100 mg twice daily. There was no significant difference to placebo in either study in any of the primary outcome measures.

3.1.10. Sertraline

Level of Evidence: 2b

Risk of Bias: Inconsistent blinding, not all outcome measures completed at baseline, impact of normal development

Tolerability: No serious adverse events. No significant difference with placebo.

Efficacy: No significant difference to placebo in any primary outcome measure.

Selective Serotonin Re-uptake Inhibitors (SSRIs) such as Serotonin are commonly prescribed in association with FXS. This review identified one study that met the inclusion criteria. Sertraline was investigated in younger children, not with specific focus on the treatment of comorbid symptoms of psychiatric illness (anxiety or mood disorder). Serotonin levels may be lower in children with autism, and SSRIs may be beneficial to combat the lack of synaptic maturation in FXS and may also stimulate BDNFs’ role in the early development period. This review identified one study, a randomized double-blind, placebo-controlled trial, including 52 children (age 2–6 years) [Citation33]. The duration was 6 months. The dose of sertraline was low (between 2.5 and 5 mg per day, depending on age). There was no significant difference to placebo on primary outcome measures.

3.1.11. Arbaclofen

Level of Evidence: 2b

Risk of Bias:

Tolerability: No serious adverse events, similar in both placebo and treatment groups.

Efficacy: No significant difference to placebo in primary outcome measure in child or adolescent/adult cohort. Reduction in irritability measure in child group at the highest dose.

As we have discussed inhibition of the mGLuR signaling may correct the developmental phenotype associated with FXS in animal models (mGLuR5). The use of GABA agonists may be a mechanism to reduce glutaminergic activate through regulation. Arbaclofen is a potent GABA-B agonist with observed behavioral benefits in autistic people, and preliminary data suggest such benefits also have application in FXS [Citation49]. The review identified one article including two studies or cohorts with similar design (adolescent/adult cohort, 12–50 years and child cohort, 5–11 years) [Citation35]. The studies included 119 in the adolescent/adult study and 159 in the child study. These studies were multisite, randomized, double-blind, placebo controlled, parallel group in both cohorts. The dose of Arbaclofen was titrated up though trial (5 mg twice daily to 15 mg three times daily). There was no significant benefit to placebo on primary outcome measures (social avoidance). In the child group, the highest dose court had significant benefit on other secondary measures including irritability, with an effect size similar to SSRIs.

3.1.12. Basmimglurant

Level of Evidence: 2b

Risk of Bias: Short duration of study, validity of assessment tools

Tolerability: Well tolerated, but a far higher rate of adverse effects (including psychiatric effects) with treatment (dose-dependent).

Efficiency: No significant difference to placebo in primary outcome measures in either group. Outcomes were better in the placebo group (possible side-effect related).

Basmimglurant is an mGluR5 Negative Allosteric Modulator, again considered based upon the underlying ‘mGluR theory’ of FXS phenotype being driven by excessive mGluR-dependent protein synthesis. This review identified one study, including 183 adults/adolescents (14–50 years mean age 23.4) [Citation36]. This was a 12-week, multi-site, randomized, double-blind, placebo-controlled trial using two doses of Basmimglurant (0.5 mg and 1.5 mg). There was no difference to placebo in either group in primary outcome measures.

3.1.13. Trofinetide

Level of Evidence: 2b

Risk of Bias: Small sample size, short duration, new unvalidated assessment tools

Tolerability: No serious adverse events, similar in both placebo and treatment group.

Efficacy: No significant difference to placebo in primary outcome measure in child or adolescent/adult cohort. Reduction in irritability measure in child group at the highest dose.

Trofinetide is an analog of insulin-like growth factor 1 (IGF-1). Levels of IGF-1 have been found to be low in FMR1 knock out mice. Treatment with Trofinetide normalized the behavioral phenotype (and raised IGF-1) [Citation50]. This review identified one study including 70 participants, all male [Citation40]. This was a randomized, double-blind, placebo-controlled trial, over 28 days. Participants were randomly assigned to placebo, trofinetide 35 mg/kg, or trofinetide 70 mg/kg. There was no significant improvement compared to placebo on any of the outcome measures. Trend of improvement with treatment observed.

3.1.14. BPN14770

Level of Evidence: 2b

Risk of Bias: Small sample size, recruitment, cross over design

Tolerability: No serious adverse events, no significant difference to placebo

Efficacy: Significant improvement on secondary outcome measures (language, communication, level of functioning)

BPN14770 is a phosphodiesterase-4D (PDE4D) allosteric inhibitor. PDE4D is a key modulator of cAMP activity, related to learning and memory. cAMP metabolism may be altered in FXS, based upon animal models. PDE4D missense mutations are associated with a neurodevelopmental disorder phenotype with ID. This review identified one study including 30 adult male patients from a single center [Citation41]. The dose of BPN14770 was 25 mg twice daily. Significant and clinically relevant improvement on secondary outcomes include language and daily functioning (parent/caregiver observation only).

3.1.15. Transdermal cannabidiol gel

Level of Evidence: 2b

Risk of Bias: Validity of assessment tools, Post-Hoc analysis (small sample size)

Tolerability: No serious adverse events, safe and well tolerated, application site pain main adverse event.

Efficacy: Significant improvement on secondary outcome measures (language, communication, level of functioning)

FXS is associated with dysregulation in endocannabinoid signaling. It is therefore postulated that treatment with cannabidiol may be effective. Preliminary clinical results with transdermal cannabidiol, get (ZYN002) have shown positive impact on anxiety and behavior [Citation51]. This review identified one study including 212 participants, 75% male, age 3 to <18 years [Citation43]. This was a randomized, double-blind, placebo controlled, multi-site trial, over 12 weeks. Treatment doses were 250 mg or 500 mg daily. No significant improvement with placebo in primary outcome measures. There was significant improvement in the subgroup with ≥90% methylation (generally more severe phenotype).

4. Discussion

A previous systematic review of the pharmacological treatments in FXS employed less restrictive inclusion criteria [Citation52]. The studies were controlled but not restricted to requiring randomization. The review identified 10 studies, with a crossover to those eligible for this review. In this updated, more restricted systematic review we can identify that the quality of methodology in the studies and level of evidence is improving over time. We can observe the development from controlled designs through to RCTs by the same research groups in those molecules identified as safe and tolerable. This review has also identified new pharmacological treatment options, and these have been investigated using a parallel design.

There are limitations to the literature search based upon the inclusion criteria and sources searched. A previous literature review conducted by experts in the field provides an overview of the evolving status of clinical trials focused on interventions in FXS [Citation53–56]. The authors indicate over 20 clinical trials completed or in progress. If we cross-reference there is significant overlap with the results of this systematic review. A small number of studies included would not have met the inclusion criteria for this review based upon methodology (no randomization, open-label trials), outcome measures (no assessment of global clinical impact), or no results data available. This review also only included peer reviewed journal articles (excluding conference abstracts). If we consider the additional studies, then there is no significant new information provided for the compounds already included in this review.

However, the review by the expert research team did identify one study that would have met the inclusion criteria for this review. A randomized double blind, placebo-controlled trial of Ganaxolone (GABA-A agonist) in children and adolescents (age 6–17 years) with FXS (n=59, male 50, female 9). The results found Ganaxolone to be safe and tolerable; however, there was no significant difference between treatment group and placebo group for the primary outcome measure (CGI). A post-hoc subgroup analysis suggests the potential for benefit for those with anxiety and lower IQ [Citation57].

Researchers remain restricted by the prevalence of FXS, which has led to the use of crossover studies of short duration that introduces the significant risk of bias. There is a need for multi-center collaborative project to ensure enough participants to demonstrate any significant effects. On observing the development of the research base, there are specific research centers with a focus on identifying treatment options for FXS, for which the FMR1 mutation was only identified around 35 years ago. There has been progression through the research pathway to more robust study designs with improved power that is improving the evidence rapidly.

One challenge posed to researchers is how to measure success. If we consider a model of behavior, there are multi-factorial influencers [Citation58]. Behavior is a crude representation, and broad characteristics (e.g. aggression) may be influenced by a plethora of genetic variants that then lead a range of consequences to different proteins and their function. There are a number of validated assessment tools that consider global impact and level of function [Citation55]. We can also observe that more specific validated assessment measures have been developed by the expert researchers in this field. Being a single gene disorder, FXS provides the opportunity to focus on interventions targeted at the behavioral phenotype, particularly for those with complete or near-complete methylation, associated with a more severe phenotype. Moreover, the strong association with autism and autistic traits may provide inside into treatment of core autistic features. This has led to the development of treatment strategies based upon our molecular understanding of the FMR1 gene and the effect on FMRP and wider impacts including synapse function, maturation, and plasticity. Disease-specific treatments targeted, and these changes have been successful in animal models (FMR1 KO mice). However, the findings from this review indicate that these results are not easily replicated in human studies. Perhaps highlighting the challenges in recruiting enough participants and in study methodology.

In this review, there has been a trend toward improvement with intervention driven by a focus on precision medicine. We can also learn that interventions may need to be targeted at particular populations. This includes a focus on those with complete or near-complete methylation, and the recurring theme that outcomes are better for younger children. This correlates with earlier intervention in the neurodevelopmental period but poses ethical considerations when developing study protocols.

4.1. Neurodevelopmental/Psychiatric Co-morbidities

There are a range of lower-level studies that consider treatment options for the most common co-morbid symptoms/diagnoses in FXS. This evidence base and clinical utility are summarized by Hagerman et al. (2009), as part of the leading research collaborative in FXS treatments [Citation6]. The focus of pharmacological treatments covers a range of key domains ().

Table 2. Evidence base for treatment considerations by domain in FXS [Citation6].

5. Conclusion

This systematic review has identified 16 studies that focus on pharmacological interventions for FXS across a range of indications. This review finds that there is limited higher level evidence available confirming the efficacy of any of the molecules tested. However, there is a clear research pathway of developing compounds based upon molecular understanding driving precision medicine, which must be the focus for the future and FXS researchers may be leading the way for other complex gene disorders.

6. Expert opinion

The evidence base for pharmacological treatments in FXS is developing rapidly, driven by increased understanding of the molecular impact of the FMR1 mutation. FXS provide a single gene disorder with a defined phenotype to target treatment considerations that may potentially be useful in wider populations. Given changes to neuroplasticity [Citation59] over time and the impact on planning families [Citation60], there may well be possible benefit to early diagnosis as well as social, educational, and/or pharmaceutical intervention but access to diagnostics and care may be inconsistent or be associated with waiting lists.

Based on this review to date, there is some encouraging data but no specific findings to support the newer pharmacological treatment options, although the data for cannabinoids are encouraging in those with full expansions and there is hope that gene therapy may be developed using antisense oligonucleotides [Citation61]. We are currently reliant on the lower level evidence available in combination with expert clinical opinion to guide treatment choice. The treatment of co-morbidities has an evidence base specifically in FXS, and we need to extrapolate from wider populations and be mindful of the possibility of carriers suffering from under-diagnosed anxiety, ataxia, or premature ovarian insufficiency [Citation62].

This review identified a range of potential treatment options that have been investigated through methodologies that did not meet the strict inclusion criteria. In the main, the studies represented were open-label cohort studies. Many of these initial investigations show promising results and are being investigated through randomized placebo-controlled trials now or will be in the near future.

One drug of particular note for discussion is Metformin. Metformin is well studied in many settings, generally well tolerated, low cost, has a range of properties, and currently mainly utilized for the treatment of diabetes. Metformin also has anti-cancer properties and has been found to have beneficial effects in a number of neurological conditions including Alzheimer’s Dementia, depression, and FXS [Citation63].

Alongside other known effects of Metformin, it likely plays a role in regulation of the mTOR and AMPK signaling pathways and potentially has a range of direct and indirect neuroprotective effects [Citation63,Citation64]. Metformin has indicated positive results in mice models and a case series of seven people with FXS who were treated with Metformin clinically [Citation65]. Alongside weight loss, the results demonstrated consistent improvements in irritability, social responsiveness, hyperactivity, and social avoidance. There were no significant side-effects. Currently, a multi-center controlled trial is underway.

Any approach to treatment must be person-centered and holistic. This should involve a multidisciplinary team to undertake in-depth assessments of an individual’s needs given the combination of medical, genetic, cognitive, and emotional challenges. Databasing of research interests [Citation66], working with patient and family organizations and adoption of internationally approved metrics for tracking cognitive improvements [Citation67] could well be beneficial in improving access to future interventional research.

Article highlights

  • Mutation of the FMR1 (Fragile X Messenger Ribonucleoprotein 1) gene leading to Fragile X Syndrome (FXS) provides an opportunity to consider pharmacological interventions on a molecular level.

  • This systematic review of Randomised Controlled Trials identified 16 studies including 15 treatments specifically focused on FXS including specific symptoms and co-morbidities associated with the broad phenotype.

  • Many of the treatments still being developed show positive findings, but results are not always translated to a significant clinical benefit.

  • Further research will focus on instruments that measure outcomes specific to FXS, and require collaboration between research centres for larger cohorts.

  • There is promise in a number of the pharmacological treatments identified including Cannabidiol based products and the versatile drug Metformin.

  • FXS pharmacological research is leading the way in precision-based medicine, developments in gene therapy will likely be the future.

Declaration of interest

R Shankar has received institutional and research support from LivaNova, UCB, Eisai, Veriton Pharma, Bial, Angelini, UnEEG, and Jazz/GW pharma outside the submitted work. He holds grants from NIHR AI, SBRI, and other funding bodies all outside this work.

The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

Author contributions

All authors satisfy the ICMJE guidance by substantially contributing to the design, analysis and interpretation of the work, drafting of the manuscript, final approval of the manuscript, and all agree to be accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Data statement

The data that support the findings of this study are available from the corresponding author upon reasonable request.

Reviewer disclosures

Peer reviewers on this manuscript have no relevant financial or other relationships to disclose.

Additional information

Funding

This paper was not funded.

References

  • Verkerk AJ, Pieretti M, Sutcliffe JS, et al. Identification of a gene (FMR-1) containing a CGG repeat coincident with a break- point cluster region exhibiting length variation in fragile X syndrome. Cell. 1991;65(5):905–914. doi: 10.1016/0092-8674(91)90397-H
  • Pieretti M, Zhang F, Fu YH, et al. Absence of expression of the FMR-1 gene in fragile X syndrome. Cell. 1991;66(4):817–822. doi: 10.1016/0092-8674(91)90125-I
  • Hunter J, Rivero‐Arias O, Angelov A, et al. Epidemiology of fragile X syndrome: a systematic review and meta‐analysis. Am J Med Genet A. 2014;164(7):1648–1658. doi: 10.1002/ajmg.a.36511
  • Loesch DZ, Huggins RM, Hagerman RJ. Phenotypic variation and FMRP levels in fragile X. Ment Retard Dev Disabil Res Rev. 2004;10(1):31–41. doi:10.1002/mrdd.20006
  • De Vries BB, Jansen CC, Duits AA, et al. Variable FMR1 gene methylation of large expansions leads to variable phenotype in three males from one fragile X family. J Med Genet. 1996;33(12):1007–1010. doi: 10.1136/jmg.33.12.1007
  • Hagerman RJ, Berry-Kravis E, Kaufmann WE, et al. Advances in the treatment of fragile X syndrome. Pediatrics. 2009;123(1):378–390. doi: 10.1542/peds.2008-0317
  • Hardiman RL, McGill P. How common are challenging behaviors amongst individuals with fragile X syndrome? A systematic review. Res Dev Disabil. 2018;76:99–109. doi:10.1016/j.ridd.2018.02.020
  • Cordeiro L, Ballinger E, Hagerman R, et al. Clinical assessment of DSM-IV anxiety disorders in fragile X syndrome: prevalence and characterization. J Neurodev Disord. 2011;3(1):57–67. doi:10.1007/s11689-010-9067-y
  • Howick J, Chalmers I, Glasziou P, et al. Carl Heneghan, “explanation of the 2011 Oxford Centre for Evidence-Based Medicine (OCEBM) levels of Evidence (background document)”. Oxford Centre For Evidence-Based. 2011/Medicine. Available from: https://www.cebm.ox.ac.uk/resources/levels-of-evidence/explanation-of-the-2011-ocebm-levels-of-evidence/OxfordCentreforEvidence-BasedMedicine:LevelsofEvidence(March2009)—CentreforEvidence-BasedMedicine(CEBM),UniversityofOxford.
  • Hagerman RJ, Jackson AW, Levitas A, et al. Oral folic acid versus placebo in the treatment of males with the fragile X syndrome. Am J Med Genet A. 1986;23(1‐2):241–262. doi: 10.1002/ajmg.1320230119
  • Dunn LM, Dunn LM. Peabody picture vocabulary test–third edition (PPVT-III) [database record]. APA PsycTests; 1997. doi: 10.1037/t15145-000
  • Wong BY, Roadhouse A. The test of language development (TOLD): a validation study. Learn Disabil Quarterly. 1978;1(3):48–61. doi:10.2307/1510937
  • Frankenburg WK, Camp BW, Van Natta PA. Validity of the Denver developmental screening test. Child Dev. 1971;42(2):475–485. doi: 10.2307/1127481
  • Leiter RG, Arthur G. The Leiter international performance scale. Vol. 1. California: Santa Barbara State College Press; 1940.
  • Marteleto MRF, Pedromônico MRM. Validity of autism behavior checklist (ABC): preliminary study. Rev Bras Psiquiatr. 2005;27(4):295–301. doi: 10.1590/S1516-44462005000400008
  • Krug DA, Arick JR, Almond PJ. ASIEP: autism screening instrument for educational planning, Revised. Austin, TX: Pro-Ed; 1980.
  • Schopler E, Reichler RJ, DeVellis RF, et al. Toward objective classification of childhood autism: childhood autism rating scale (CARS). J Autism Dev Disord. 1980;10(1):91–103. doi: 10.1007/BF02408436
  • Hagerman RJ, Murphy MA, Wittenberger MD. A controlled trial of stimulant medication in children with the fragile X syndrome. Am J Med Genet A. 1988;30(1‐2):377–392. doi:10.1002/ajmg.1320300138
  • Conners CK, Sitarenios G, Parker JD, et al. Revision and restandardization of the Conners teacher rating scale (CTRS-R): factor structure, reliability, and criterion validity. J Abnorm Child Psychol. 1998;26(4):279–291. doi: 10.1023/A:1022606501530
  • Ullmann RK, Sleator EK, Sprague RL. A new rating scale for diagnosis and monitoring of ADD children. Psychopharmacol Bull. 1984;20(1):160–164.
  • Strom CM, Brusca RM, Pizzi WJ. Double‐blind, placebo‐controlled crossover study of folinic acid (Leucovorin®) for the treatment of fragile X syndrome. Am J Med Genet A. 1992;44(5):676–682. doi:10.1002/ajmg.1320440529
  • Sparow SS, Cicchetti DV, Balla DA. Vineland adaptive behavior scales. 2nd (VinelandTM-II) ed. Circle Pines: American Guidance Service; 2005.
  • Berry-Kravis E, Krause SE, Block SS, et al. Effect of CX516, an AMPA-modulating compound, on cognition and behavior in fragile X syndrome: a controlled trial. J Child Adolesc Psychopharmacol. 2006;16(5):525–540. doi: 10.1089/cap.2006.16.525
  • Guy W. ECDEU assessment manual for psychopharmacology. Rockville, Maryland: US Department of Health, Education, and Welfare, Public Health Service, Alcohol, Drug Abuse, and Mental Health Administration, National Institute of Mental Health, Psychopharmacology Research Branch, Division of Extramural Research Programs; 1976.
  • Swanson JM, Kraemer HC, Hinshaw SP, et al. Clinical relevance of the primary findings of the MTA: success rates based on severity of ADHD and ODD symptoms at the end of treatment. J Am Acad Child Adolesc Psychiatry. 2001;40(2):168–179. doi: 10.1097/00004583-200102000-00011
  • Torrioli MG, Vernacotola S, Peruzzi L, et al. A double‐blind, parallel, multicenter comparison of L‐acetylcarnitine with placebo on the attention deficit hyperactivity disorder in fragile X syndrome boys. Am J Med Genet A. 2008;146(7):803–812. doi: 10.1002/ajmg.a.32268
  • Wirojanan J, Jacquemont S, Diaz R, et al. The efficacy of melatonin for sleep problems in children with autism, fragile X syndrome, or autism and fragile X syndrome. J Clin Sleep Med. 2009;5(2):145–150. doi: 10.5664/jcsm.27443
  • Sahu JK, Gulati S, Sapra S, et al. Effectiveness and safety of donepezil in boys with fragile x syndrome: a double-blind, randomized, controlled pilot study. J Child Neurol. 2013;28(5):570–575. doi: 10.1177/0883073812449381
  • Leigh MJS, Nguyen DV, Mu Y, et al. A randomized double-blind, placebo-controlled trial of minocycline in children and adolescents with fragile x syndrome. J Dev Behav Pediatr. 2013;34(3):147. doi: 10.1097/DBP.0b013e318287cd17
  • Berry-Kravis E, Des Portes V, Hagerman R, et al. Mavoglurant in fragile X syndrome: results of two randomized, double-blind, placebo-controlled trials. Sci, trans med. 2016;8(321):ra3215–ra3215. doi: 10.1126/scitranslmed.aab4109
  • Aman MG, Singh NN, Stewart AW, et al. Psychometric characteristics of the aberrant behavior checklist. Am J Mental Defic. 1985;89(5):492–502.
  • Sansone SM, Widaman KF, Hall SS, et al. Psychometric study of the aberrant behavior checklist in fragile X syndrome and implications for targeted treatment. J Autism Dev Disord. 2012;42(7):1377–1392. doi: 10.1007/s10803-011-1370-2
  • Hess LG, Fitzpatrick SE, Nguyen DV, et al. A randomized, double-blind, placebo-controlled trial of low-dose sertraline in young children with fragile X syndrome. J Dev Behav Pediatr. 2016;37(8):619. doi: 10.1097/DBP.0000000000000334
  • Mullen EM. Mullen scales of early learning. Circle Pines (MN): AGS; 1995. p. 58–64.
  • Berry-Kravis E, Hagerman R, Visootsak J, et al. Arbaclofen in fragile X syndrome: results of phase 3 trials. J Neurodevelop Disord. 2017;9(1):1–18. doi: 10.1186/s11689-016-9181-6
  • Youssef EA, Berry-Kravis E, Czech C, et al. Effect of the mGlur5-NAM basimglurant on behavior in adolescents and adults with fragile X syndrome in a randomized, double-blind, placebo-controlled trial: FragXis phase 2 results. Neuropsychopharmacology. 2018;43(3):503–512. doi: 10.1038/npp.2017.177
  • Esbensen AJ, Rojahn J, Aman MG, et al. Reliability and validity of an assessment instrument for anxiety, depression, and mood among individuals with mental retardation. J Autism Dev Disord. 2003;33(6):617–629. doi: 10.1023/B:JADD.0000005999.27178.55
  • Bruno JL, Hosseini SH, Lightbody AA, et al. Brain circuitry, behavior, and cognition: a randomized placebo-controlled trial of donepezil in fragile X syndrome. J Psychopharmacol. 2019;33(8):975–985. doi: 10.1177/0269881119858304
  • Anderson P, Anderson V, Northam E, et al. Standardization of theContingency Naming Test for school-aged children: A new measure of reactiveflexibility. Clinical Neuropsychological Assessment. 2000;1:247–273.
  • Berry-Kravis E, Horrigan JP, Tartaglia N, et al. A double-blind, randomized, placebo-controlled clinical study of trofinetide in the treatment of fragile X syndrome. Pediatr Neurol. 2020;110:30–41. doi: 10.1016/j.pediatrneurol.2020.04.019
  • Berry-Kravis EM, Harnett MD, Reines SA, et al. Inhibition of phosphodiesterase-4D in adults with fragile X syndrome: a randomized, placebo-controlled, phase 2 clinical trial. Nature Med. 2021;27(5):862–870. doi: 10.1038/s41591-021-01321-w
  • Akshoomoff N, Beaumont J, Bauer L, et al. National Institutes of Health Toolbox Cognition Battery (NIH Toolbox CB): validation for children between 3 and 15 years: VIII. NIH Toolbox Cognition Battery (CB): composite scores of crystallized, fluid, and overall cognition. Monogr Soc Res Child Dev. 2013;78(4):119–132.
  • Berry-Kravis E, Hagerman R, Budimirovic D, et al. A randomized, controlled trial of ZYN002 cannabidiol transdermal gel in children and adolescents with fragile X syndrome (CONNECT-FX). J Neurodev Disord. 2022;14(1):56. doi: 10.1186/s11689-022-09466-6
  • Torrioli MG, Vernacotola S, Mariotti P, et al. Double‐blind, placebo‐controlled study of L‐acetylcarnitine for the treatment of hyperactive behavior in fragile X syndrome. Am J Med Genet A. 1999;87(4):366–368. doi: 10.1002/(SICI)1096-8628(19991203)87:4<366:AID-AJMG18>3.0.CO;2-F
  • Van Oudheusden LJ, Scholte HR. Efficacy of carnitine in the treatment of children with attention-deficit hyperactivity disorder. Prostaglandins, Leukot Essent Fat Acids. 2002;67(1):33–38. doi:10.1054/plef.2002.0378
  • D’Antuono M, Merlo D, Avoli M. Involvement of cholinergic and GABAergic systems in the fragile X knockout mice. Neuroscience. 2003;119(1):9–13. doi:10.1016/S0306-4522(03)00103-9
  • Bilousova TV, Dansie L, Ngo M, et al. Minocycline promotes dendritic spine maturation and improves behavioral performance in the fragile X mouse model. J Med Genet. 2009;46(2):94–102. doi: 10.1136/jmg.2008.061796
  • Huber KM, Gallagher SM, Warren ST, et al. Altered synaptic plasticity in a mouse model of fragile X mental retardation. Proc Nat Acad Sci. 2002;99(11):7746–7750. doi:10.1073/pnas.122205699
  • Berry-Kravis EM, Hessl D, Rathmell B, et al. Effects of STX209 (arbaclofen) on neurobehavioral function in children and adults with fragile X syndrome: a randomized, controlled, phase 2 trial. Sci Trans Med. 2012;4(152):ra152127–ra152127. doi: 10.1126/scitranslmed.3004214
  • Deacon RM, Glass L, Snape M, et al. NNZ-2566, a novel analog of (1–3) IGF-1, as a potential therapeutic agent for fragile X syndrome. Neuromol Med. 2015;17(1):71–82. doi: 10.1007/s12017-015-8341-2
  • Heussler H, Cohen J, Silove N, et al. A phase 1/2, open-label assessment of the safety, tolerability, and efficacy of transdermal cannabidiol (ZYN002) for the treatment of pediatric fragile X syndrome. J Neurodevelop Disord. 2019;11(1):1–9. doi: 10.1186/s11689-019-9277-x
  • Rueda JR, Ballesteros J, Tejada MI. Systematic review of pharmacological treatments in fragile X syndrome. BMC neurol. 2009;9(1):1–11. doi: 10.1186/1471-2377-9-53
  • Budimirović DB, Duy PQ. Neurobehavioral features and targeted treatments in fragile X syndrome: Current insights and future directions. Engrami. 2015;37(4):5–26. doi:10.5937/engrami1504005B
  • Ligsay A, Hagerman RJ. Review of targeted treatments in fragile X syndrome. Intractable Rare Dis. 2016;5(3):158–167.
  • Budimirovic DB, Berry-Kravis E, Erickson CA, et al. Updated report on tools to measure outcomes of clinical trials in fragile X syndrome. J Neurodev Disord. 2017;9(1):1–36. doi: 10.1186/s11689-017-9193-x
  • Berry-Kravis EM, Lindemann L, Jønch AE, et al. Drug development for neurodevelopmental disorders: lessons learned from fragile X syndrome. Nat Rev Drug Discov. 2018;17(4):280–299. doi: 10.1038/nrd.2017.221
  • Ligsay A, Van Dijck A, Nguyen DV, et al. A randomized double-blind, placebo-controlled trial of ganaxolone in children and adolescents with fragile X syndrome. J Neurodev Disord. 2017;9(1):1–13. doi: 10.1186/s11689-017-9207-8
  • Watkins LV, O’Dwyer M, Shankar R. A review of the pharmacotherapeutic considerations for managing epilepsy in people with autism. Expert Opin Pharmacother. 2022;23(7):841–851. doi:10.1080/14656566.2022.2055461
  • Richter JD, Zhao X. The molecular biology of FMRP: new insights into fragile X syndrome. Nat Rev Neurosci. 2021;22(4):209–222. doi:10.1038/s41583-021-00432-0
  • Hipp HS, Charen KH, Spencer JB, et al. Reproductive and gynecologic care of women with fragile X primary ovarian insufficiency (FXPOI). Menopause. 2016;23(9):993–9. doi: 10.1097/GME.0000000000000658
  • Shah S, Sharp KJ, Raju Ponny S, et al. Antisense oligonucleotide rescue of CGG expansion-dependent FMR1 mis-splicing in fragile X syndrome restores FMRP. Proc Natl Acad Sci U S A. 2023;120(27):e2302534120. doi: 10.1073/pnas.2302534120
  • Bourgeois JA. Neuropsychiatry of fragile X-premutation carriers with and without fragile X-associated tremor-ataxia syndrome: implications for neuropsychology. Clin Neuropsychol. 2016;30(6):913–928. doi:10.1080/13854046.2016.1192134
  • Li N, Zhou T, Fei E. Actions of metformin in the brain: a new perspective of metformin treatments in related neurological disorders. Int J Mol Sci. 2022;23(15):8281.2022. doi:10.3390/ijms23158281
  • Miranda I, Hagerman R. Molecular mechanisms for targeted treatments in fragile X syndrome. Rare Dis Orphan Drugs J. 2023;2:20. doi: 10.20517/rdodj.2023.21
  • Dy ABC, Tassone F, Eldeeb M, et al. Metformin as targeted treatment in fragile X syndrome. Clin Genet. 2018;93(2):216–222. doi: 10.1111/cge.13039
  • Sherman SL, Kidd SA, Riley C, et al. FORWARD: a registry and longitudinal clinical database to study fragile X syndrome. Pediatrics. 2016;139(Suppl 3):S183–S193. doi:10.1542/peds.2016-1159E
  • Hessl D, Sansone SM, Berry-Kravis E, et al. The NIH toolbox cognitive battery for intellectual disabilities: three preliminary studies and future directions. J Neurodev Disord. 2016;8(1):35. doi: 10.1186/s11689-016-9167-4

Appendix 1

Search Strategy

  1. (Fragile X or FragileX).ab,ti,kf. 9998

  2. exp fragile X syndrome/ 9736

  3. ((pharmacological or clinical or medical) adj2 (treatment* or management)).ab,kf,ti. 326665

  4. pharmacotherapy.ab,kf,ti. 57391

  5. ‘medication*.’ab,kf,ti. 649360

  6. exp drug therapy/ 3414714

  7. sertraline.ab,kf,ti. 8037

  8. zoloft.ab,kf,ti. 177

  9. exp sertraline/ 29425

  10. metformin.ab,kf,ti. 45982

  11. exp metformin/ 81466

  12. cannabidiol.ab,kf,ti. 6845

  13. CBD.ab,kf,ti. 16534

  14. exp cannabidiol/ 8236

  15. acamprosate.ab,kf,ti. 1312

  16. exp acamprosate/ 2718

  17. lovastatin.ab,kf,ti. 5325

  18. minocycline.ab,kf,ti. 10716

  19. exp minocycline/ 27761

  20. anti-depressant*.ab,ti,kf. 4459

  21. antidepressant.ab,ti,kf. 72485

  22. exp antidepressant agent/ 553449

  23. ‘SSRI*.’ab,ti,kf. 19868

  24. selective serotonin reuptake inhibitor.ab,ti,kf. 6609

  25. ‘SNRI*.’ab,ti,kf. 3232

  26. Serotonin norepinephrine reuptake inhibitors.ab,ti,kf. 1198

  27. (Noradrenergic and specific serotonergic antidepressants).ab,ti,kf. 52

  28. NaSSa.ab,ti,kf. 187

  29. ‘tricyclic anti-depressant*.’ab,kf,ti. 252

  30. tricyclic antidepressant.ab,kf,ti. 4602

  31. ‘anti-psychotics*.’ab,kf,ti. 792

  32. antipsychotic.ab,ti,kf. 53568

  33. exp neuroleptic agent/ 284094

  34. ‘anti-seizure medication*.’ab,kf,ti. 1275

  35. ‘antiseizure medication*.’ab,kf,ti. 1803

  36. ASMs.ab,kf,ti. 1203

  37. ‘anti-epileptic medication*.’ab,kf,ti. 802

  38. ‘antiepileptic medication*.’ab,kf,ti. 2831

  39. AEMs.ab,kf,ti. 488

  40. exp anticonvulsive agent/ 472103

  41. ‘mood stabiliser*.’ab,kf,ti. 737

  42. ‘Mood stabilizer*.’ab,kf,ti. 6755

  43. exp mood stabilizer/ 136894

  44. ‘adhd medication*.’ab,kf,ti. 1436

  45. ‘Benzodiazepine*.’ab,kf,ti. 55426

  46. exp benzodiazepine derivative/ 253811

  47. 1 or 2 12,569

  48. 3 or 4 or 5 or 6 or 7 or 8 or 9 or 10 or 11 or 12 or 13 or 14 or 15 or 16 or 17 or 18 or 19 or 20 or 21 or 22 or 23 or 24 or 25 or 26 or 27 or 28 or 29 or 30 or 31 or 32 or 33 or 34 or 35 or 36 or 37 or 38 or 39 or 40 or 41 or 42 or 43 or 44 or 45 or 46 4,877,314

  49. 47 and 48 1546