BACKGROUND:

Approximately 25% of children with concussion have persistent postconcussive symptoms (PPCS) with resultant significant impacts on quality of life. Melatonin has significant neuroprotective properties, and promising preclinical data suggest its potential to improve outcomes after traumatic brain injury. We hypothesized that treatment with melatonin would result in a greater decrease in PPCS symptoms when compared with a placebo.

METHODS:

We conducted a randomized, double-blind trial of 3 or 10 mg of melatonin compared with a placebo (NCT01874847). We included youth (ages 8–18 years) with PPCS at 4 to 6 weeks after mild traumatic brain injury. Those with significant medical or psychiatric histories or a previous concussion within the last 3 months were excluded. The primary outcome was change in the total youth self-reported Post-Concussion Symptom Inventory score measured after 28 days of treatment. Secondary outcomes included change in health-related quality of life, cognition, and sleep.

RESULTS:

Ninety-nine children (mean age: 13.8 years; SD = 2.6 years; 58% girls) were randomly assigned. Symptoms improved over time with a median Post-Concussion Symptom Inventory change score of −21 (95% confidence interval [CI]: −16 to −27). There was no significant effect of melatonin when compared with a placebo in the intention-to-treat analysis (3 mg melatonin, −2 [95% CI: −13 to 6]; 10 mg melatonin, 4 [95% CI: −7 to 14]). No significant group differences in secondary outcomes were observed. Side effects were mild and similar to the placebo.

CONCLUSIONS:

Children with PPCS had significant impairment in their quality of life. Seventy-eight percent demonstrated significant recovery between 1 and 3 months postinjury. This clinical trial does not support the use of melatonin for the treatment of pediatric PPCS.

What’s Known on This Subject:

Approximately 25% of children have persistent postconcussive problems after a mild traumatic brain injury with significant impairment in their quality of life. Evidence-based approaches for their management are limited. Melatonin is a promising neuroprotective agent that is commonly used in clinical practice.

What This Study Adds:

The evidence from this randomized trial does not support the use of melatonin for the treatment of persistent postconcussive symptoms at 4 weeks postinjury in typically developing children even in the context of sleep disturbance.

Persistent postconcussive symptoms (PPCS) are a common and significant problem among children after mild traumatic brain injury (mTBI), yet few evidence-based treatments are available.1,2  mTBI and concussion account for 90% of all traumatic brain injuries (TBIs), affecting as many as 1 in 10 children and adolescents before 16 years of age.3,4  Although the majority of children recover quickly, a significant proportion (11% to 30%) have PPCS one month later.1,5,6  PPCS have a significant and detrimental effect on children’s quality of life, participation in school and recreational activities, and family functioning.7,8 

More evidence-based treatments are needed after mTBI. Early management is directed by sport-related concussion guidelines, which recommend an initial limited period of rest followed by gradual reintegration into school and activities.9,10  For children with persistent symptoms, the evidence for treatment is more limited.9,11,12  Small studies suggest that specialized therapies, graded-exercise programs, and multimodal collaborative care may be helpful.2,1315  However, these treatments are often expensive and not readily available in most communities.

Melatonin is a promising, well-tolerated, neuroprotective agent in TBI. It has antioxidant and antiinflammatory properties16,17  and is associated with improved behavioral and pathologic outcomes in animal TBI models.17  It also has therapeutic potential in relieving symptoms that are common in PPCS11,1820  and is often recommended as a part of the management plan.21  Although it is often considered a nutritional supplement, rigorous evaluation of such nutraceuticals is nevertheless important to ensure best evidence-based practices.22  Our aim was to determine if treatment with melatonin improves PPCS after mTBI and concussion in youth. We hypothesized that treatment with melatonin (3 or 10 mg) for 28 days would result in a greater decrease in PPCS when compared with a placebo, with the null hypothesis being that melatonin and a placebo are equally effective or ineffective. Furthermore, we investigated whether the effect of melatonin was independent of its effects on sleep.

We conducted a single-center, randomized, double-blind, placebo-controlled trial at Alberta Children’s Hospital. The rationale for the trial, design, and analysis plan have been published previously.23  This study was approved by the university research ethics board. Enrollment occurred between February 2014 and April 2017. Changes to inclusion criteria (ie, lower age limit decreased from 13 to 8 years and time since last concussion decreased from 12 to 3 months) were made in December 2014 to improve recruitment. The trial was conducted in accordance with the standards of Good Clinical Practice. Potential participants were children seen in the emergency department with a medically diagnosed concussion and/or mTBI who consented to telephone follow-up. Concussion and/or mTBI was defined by using the American Academy of Neurology criteria.24  Outcome assessments were performed just before treatment commenced, weekly during treatment, at the end of treatment (posttreatment), and again at 4 and 6 months postinjury. Follow-up was completed in October 2017.

Participants were enrolled by using a 2-step process: first by telephone at 2 to 4 weeks and then in person 2 weeks later. We enrolled children ages 8 to 18 years if they had PPCS and a ≥10-point increase in their total symptom score on the Post-Concussion Symptom Inventory (PCSI) postinjury when compared with their preinjury score (assessed at enrollment).25  Children were ineligible if they had a significant medical or psychiatric history, a previous concussion within the last 3 months, persistent symptoms after a previous concussion, or a more severe TBI previously. Other exclusions included lactose intolerance, use of neuroactive drugs, and inability to complete questionnaires. All participants and their guardians provided written consent. At the time of enrollment, a standardized interview and medical examination were performed. Figure 1 outlines the trial details. An independent trial monitoring board periodically reviewed safety data. No interim analyses of efficacy were performed.

FIGURE 1

Consolidated Standards of Reporting Trials diagram demonstrating the enrollment, interventions, and follow-up of study participants. ED, emergency department; hx, history.

FIGURE 1

Consolidated Standards of Reporting Trials diagram demonstrating the enrollment, interventions, and follow-up of study participants. ED, emergency department; hx, history.

Close modal

After enrollment, participants were randomly assigned by using a random-block-size design (block sizes 3, 6, and 9) to 3 parallel treatment groups with a 1:1:1 allocation: placebo, melatonin 3 mg, and melatonin 10 mg. The computer-generated randomization list was created and held by an external statistician. An independent pharmacist created sequentially numbered, identical treatment packages. Melatonin and placebo tablets were identical in appearance and flavor. All investigators, outcome assessors, parents, and children were blinded to treatment groups.

The study drug was taken sublingually one hour before sleep time at night for 28 days and was continued even if symptom resolution occurred. No restrictions were placed on the use of other medications. Participants were advised to avoid analgesia overuse, abstain from contact sports, perform light exercise, and gradually return to school. Compliance and adverse events were monitored weekly.

The primary outcome was the change in Post-Concussion Symptom Inventory–Youth (PSCI-Y) total score for youth self-report at the end of 28 days of treatment, which was calculated as pretreatment score minus end-of-treatment score. This standardized, 26-item questionnaire provides an overall rating of PPCS (total score range: 0–156). The PCSI-Y has 4 specific domains derived from factor analysis (physical, cognitive, emotional, and fatigue) and a high level of internal consistency and reliability (α = .92).26,27 

Secondary outcomes were behavioral, cognitive, and sleep problems and functional impairment, which are common complaints among PPCS. The Post-Concussion Symptom Inventory–Parent (PCSI-P), a parent-proxy questionnaire, was completed (total score range: 0–104).26  Health-related quality of life was assessed by using the Child Health Questionnaire (Parent Form 50 [parent]; child form 87 [child]).28  Behavioral adjustment and everyday executive functioning were assessed by using the parent-report versions of the Behavior Assessment System for Children, Second Edition and the Behavior Rating Inventory of Executive Function, respectively. Scores for the Behavior Assessment System for Children, Second Edition and the Behavior Rating Inventory of Executive Function are age- and sex-adjusted T-scores (mean = 50; SD = 10). Neurocognitive ability was assessed by using a computerized assessment battery (CNS Vital Signs). The Neurocognition Index was used as a global score of neurocognition. The validity of test performance was assessed by using the Test of Memory Malingering (TOMM).29  These measures have demonstrated validity in TBI and concussion.3033 

Sleep was assessed by using a wrist-worn accelerometer (Actiwatch2 ) on the nondominant wrist for 5 to 7 days before and after treatment.34  Actigraphy was used to measure sleep-activity patterns, including total sleep time, onset latency, efficiency, and wake after sleep onset (WASO) (amount of time). Epochs were set at 15 seconds.

Overnight urinary 6-sulphatoxymelatonin (aMT6s), the major metabolite of melatonin, was analyzed before treatment, midtreatment, and at the end of treatment by using a solid-phase enzyme-linked immunosorbent assay (Cat#RE54031; IBL International, Hamburg, Germany). Individual levels were converted to a ratio normalized by using urinary creatinine concentration.35 

Data from a previous epidemiological study1  were used to calculate a Reliable Change Index.36  A 10-point change (SD = 14.7) in PCSI score indicated a reliable change for subjects who have PPCS at one month and is similar to previous reports.37,38  On the basis of medical record review, the relevance of 10-point improvement in PCSI score is a difference of ∼30% in overall clinical symptoms. Clinically meaningful examples of this would be a 50% reduction in posttraumatic headache burden (ie, daily headaches becoming intermittent and less severe or a 50% decrease in analgesic use), returning to full-time education with modifications, or a significant decrease in fatigue with associated increased participation in activities. The sample size (33 per group) was calculated a priori and powered (80%) to detect a 10-point (SD = 14.7) PCSI-Y score change at a significance level of α = .05.1 

Masked analysis in the differences in the change in PCSI-Y and PCSI-P scores between groups was performed by using the nonparametric Kruskal-Wallis test (KW) because parametric assumptions were not met. Bias-corrected and accelerated bootstrapped 95% confidence intervals (CIs) (1000 samples) for means and medians were computed.39  All analyses were performed with an intent-to-treat principle. Time to symptom resolution (as defined by a PCSI-Y total score equal to or less than preinjury) was examined using Kaplan-Meier survival curves. Hierarchical regression was used to predict symptom improvement by entering sleep efficiency (step 1) and treatment group (step 2) to determine how predictive treatment was above and beyond any effects of sleep efficiency. A secondary efficacy per-protocol analysis was performed, excluding participants with protocol violations. The per-protocol analysis included only participants who missed no more than 2 consecutive doses or <5 doses in total (n = 92).

Secondary outcomes were analyzed by using repeated-measures analysis of variance or the KW when assumptions were violated (change in sleep parameters) with significance α set to .01 to allow for multiple comparisons. Frequency and severity of side effects were evaluated by using Fisher’s exact test. Subsequent sensitivity analyses using quantile regression were performed to examine any effect of outliers, protocol deviations, and compliance on the primary outcome. All assumptions were checked and sufficed. Statistical analyses were performed by using IBM SPSS version 24 (IBM SPSS Statistics, IBM Corporation) and Stata release 14 (Stata Corp, College Station, TX).

The funding sources had no input in the study design, data analysis, interpretation, report generation, or submission for publication.

Ninety-nine participants (mean age = 13.8 years; SD = 2.6; 58% girls) were enrolled and randomly assigned to the placebo,33  melatonin 3-mg,33  or melatonin 10-mg33  groups. Enrollment details are displayed in Fig 1. Two participants withdrew after random assignment without starting treatment, and 3 were lost to follow-up. Ninety-four participants completed assessments immediately after intervention, and 92 completed the trial according to protocol.

Participant demographics, injury details, and clinical characteristics are summarized in Table 1. The majority of injuries (65%) were sport related, and 95% were witnessed. Treatment started at a mean of 38 days (SD = 6) postinjury. The median PCSI-Y total score before treatment was 36 (range: 6–127); Supplemental Table 5 presents details of symptom complaints.

TABLE 1

Demographic and Clinical Characteristics of Trial Participants

Placebo (n = 33)Melatonin 3 mg (n = 33)Melatonin 10 mg (n = 33)
Demographic characteristics    
 Age, y, mean (SD) 14.37 (2.55) 13.31 (2.53) 13.8 (2.61) 
 Male sex, n 13 14 15 
 Right-handedness, n 28 29 31 
 BMI (SD) 21.09 (2.93) 21.15 (4.37) 19.76 (5.15) 
 Family income, Canadian $, median (IQR) 122 409 (111 877–152 941) 112 720 (92 013–150 015) 123 536 (91 170–152 587) 
Clinical and educational history    
 Previous concussion, n (%) 15 (45) 13 (39) 16 (48) 
  Prolonged recovery (>7 d) 11 (33) 7 (22) 7 (22) 
 Migraine, n (%) 11 (33) 11 (33) 14 (42) 
 ADHD, n (%) 2 (6) 1 (3) 5 (15) 
 Learning support, n (%)  5 (15) 2 (6) 8 (24) 
 Previously seen by a counselor, n (%) 11 (33) 9 (3.6) 9 (3.6) 
 PCSI-Y total preinjury score, median (95% CI) 6 (4 to 8) 3 (2 to 6) 5 (2 to 10) 
Mechanism of injury, n (%)    
 MVA 4 (12) 1 (3) 2 (6) 
 Sport 23 (70) 21 (64) 20 (61) 
 Fall 2 (6) 4 (12) 5 (15) 
 Witnessed 32 (97) 30 (90) 32 (97) 
Acute symptoms, n (%)    
 Loss of consciousness 7 (22) 9 (27) 5 (15) 
 Retrograde amnesia 9 (27) 7 (22) 6 (18) 
 Anterograde amnesia 11 (33) 8 (25) 9 (27) 
 Confusion or disorientation 23 (70) 24 (73) 21 (64) 
 Slow to answer questions 12 (36) 15 (45) 36.4 
 Acute headache 27 (2) 30 (90) 31 (94) 
 Nausea and/or vomiting 19 (57) 15 (45) 57.6 
 Double vision 12 (36) 10 (30) 12 (36) 
 Dizziness 25 (76) 22 (67) 27 (82) 
Postconcussive symptoms    
 Time postinjury, d, mean (SD)  37.5 (6.7) 38.8 (6.2) 38.0 (5.2) 
 PCSI-Y postinjury score, median (95% CI)    
  Total score 33 (27 to 41) 36 (26 to 47) 36 (27 to 46) 
  Physical 12 (9.3 to 15.7) 14 (10 to 19) 14 (8.3 to 15.7) 
  Cognitive 9 (6 to 14) 12 (7 to 14.7) 8 (7 to 12.7) 
  Emotional 6 (2 to 9) 6 (2 to 7.7) 7 (3 to 8) 
  Fatigue 5 (3 to 6.7) 4 (3 to 6) 6 (4 to 7.7) 
Sleep parameter, median (95% CI)    
 Total sleep time, h:min 7:46 (7:10 to 8:15) 7:27 (7:06 to 7:45) 7:58 (7:42 to 8:15) 
 Onset latency, min 20.46 (14.63 to 29.78) 18.38 (10.96 to 23.31) 16.37 (12.52 to 23.59) 
 WASO, min 34.4 (25.8 to 43.2) 42.6 (34.9 to 45.2) 42.9 (38.2 to 46.4) 
 Efficiency, % 82.2 (80.5 to 84.3) 82.6 (81.1 to 85.7) 82.4 (80.4 to 84.9) 
Placebo (n = 33)Melatonin 3 mg (n = 33)Melatonin 10 mg (n = 33)
Demographic characteristics    
 Age, y, mean (SD) 14.37 (2.55) 13.31 (2.53) 13.8 (2.61) 
 Male sex, n 13 14 15 
 Right-handedness, n 28 29 31 
 BMI (SD) 21.09 (2.93) 21.15 (4.37) 19.76 (5.15) 
 Family income, Canadian $, median (IQR) 122 409 (111 877–152 941) 112 720 (92 013–150 015) 123 536 (91 170–152 587) 
Clinical and educational history    
 Previous concussion, n (%) 15 (45) 13 (39) 16 (48) 
  Prolonged recovery (>7 d) 11 (33) 7 (22) 7 (22) 
 Migraine, n (%) 11 (33) 11 (33) 14 (42) 
 ADHD, n (%) 2 (6) 1 (3) 5 (15) 
 Learning support, n (%)  5 (15) 2 (6) 8 (24) 
 Previously seen by a counselor, n (%) 11 (33) 9 (3.6) 9 (3.6) 
 PCSI-Y total preinjury score, median (95% CI) 6 (4 to 8) 3 (2 to 6) 5 (2 to 10) 
Mechanism of injury, n (%)    
 MVA 4 (12) 1 (3) 2 (6) 
 Sport 23 (70) 21 (64) 20 (61) 
 Fall 2 (6) 4 (12) 5 (15) 
 Witnessed 32 (97) 30 (90) 32 (97) 
Acute symptoms, n (%)    
 Loss of consciousness 7 (22) 9 (27) 5 (15) 
 Retrograde amnesia 9 (27) 7 (22) 6 (18) 
 Anterograde amnesia 11 (33) 8 (25) 9 (27) 
 Confusion or disorientation 23 (70) 24 (73) 21 (64) 
 Slow to answer questions 12 (36) 15 (45) 36.4 
 Acute headache 27 (2) 30 (90) 31 (94) 
 Nausea and/or vomiting 19 (57) 15 (45) 57.6 
 Double vision 12 (36) 10 (30) 12 (36) 
 Dizziness 25 (76) 22 (67) 27 (82) 
Postconcussive symptoms    
 Time postinjury, d, mean (SD)  37.5 (6.7) 38.8 (6.2) 38.0 (5.2) 
 PCSI-Y postinjury score, median (95% CI)    
  Total score 33 (27 to 41) 36 (26 to 47) 36 (27 to 46) 
  Physical 12 (9.3 to 15.7) 14 (10 to 19) 14 (8.3 to 15.7) 
  Cognitive 9 (6 to 14) 12 (7 to 14.7) 8 (7 to 12.7) 
  Emotional 6 (2 to 9) 6 (2 to 7.7) 7 (3 to 8) 
  Fatigue 5 (3 to 6.7) 4 (3 to 6) 6 (4 to 7.7) 
Sleep parameter, median (95% CI)    
 Total sleep time, h:min 7:46 (7:10 to 8:15) 7:27 (7:06 to 7:45) 7:58 (7:42 to 8:15) 
 Onset latency, min 20.46 (14.63 to 29.78) 18.38 (10.96 to 23.31) 16.37 (12.52 to 23.59) 
 WASO, min 34.4 (25.8 to 43.2) 42.6 (34.9 to 45.2) 42.9 (38.2 to 46.4) 
 Efficiency, % 82.2 (80.5 to 84.3) 82.6 (81.1 to 85.7) 82.4 (80.4 to 84.9) 

ADHD, attention-deficit/hyperactivity disorder; IQR, interquartile range; MVA, motor vehicle accident.

There was, on average, a decrease in scores in all groups over time, but there was no significant difference between the groups (Table 2). Participants reported an overall mean decrease in PCSI-Y score of 21 points (95% CI: 16 to 27; P < .001) after 28 days of treatment. Parents also reported an overall mean decrease in PCSI-P score of 13 points (95% CI: 10 to 16; P < .001). Placebo, melatonin 3-mg, and melatonin 10-mg groups showed a similar median change in PCSI-Y scores (16, 16, and 27, respectively; KW = 2.024; P = .36) and PCSI-P scores (12.5, 15, and 9.5, respectively; KW = 1.692; P = .43; Fig 2).

TABLE 2

Median Change in Total PCSI Score and Domain Scores After Treatment

PCSI ChangePlacebo (n = 32), Median (95% CI)Melatonin 3 mg (n = 33), Median (95% CI)Melatonin 10 mg (n = 32), Median (95% CI)Melatonin 3 mg to Placebo,a Median (95% CI)Melatonin 10 mg to Placebo,a Median (95% CI)χKW2, 95P
Total PCSI-Y 16 (13 to 35) 16 (12 to 26) 27 (24 to 34) −2 (−13 to 6) 4 (−7 to 14) 2.024 .36 
 Physical 8.0 (4 to 12) 8.0 (5 to 10) 10.0 (6 to 13) 0 (−4 to 4) 2 (−3 to 6)   
 Cognitive 6.0 (3 to 11) 4.0 (2 to 8) 7.0 (4 to 8) 1 (−5 to 2) 0 (3 to −3)   
 Emotional 2.0 (0 to 5) 2.0 (0 to 5) 5.0 (2 to 6) 0 (−3 to 2) 1 (−3 to 3)   
 Fatigue 2.0 (1 to 4) 3.00 (2 to 4) 4.0 (2 to 5) 0 (−2 to 2) 1 (−1 to 3)   
Total PCSI-P 12.50 (7 to 16) 15 (10 to 21) 9.5 (4.5 to 19.0) 3 (−3 to 11) −2 (−10 to 6) 1.692 .43 
 Physical 5.0 (2 to 9) 6.0 (2 to 8) 6.5 (4 to 8) 1 (−2 to 4) 0 (−2 to 3)   
 Cognitive 2.0 (0 to 6) 3.0 (2 to 4) 2.0 (2 to 6) 1 (−3 to 4) 1 (−1 to 2)   
 Emotional 2.0 (0 to 4) 1.0 (0 to 3) 2.5 (0 to 4) 0 (−2 to 1) 0 (−1 to 2)   
 Fatigue 2.0 (0 to 4) 3.00 (1 to 5) 2.0 (1 to 4) 0 (−1 to 2) 1 (−1 to 2)   
PCSI ChangePlacebo (n = 32), Median (95% CI)Melatonin 3 mg (n = 33), Median (95% CI)Melatonin 10 mg (n = 32), Median (95% CI)Melatonin 3 mg to Placebo,a Median (95% CI)Melatonin 10 mg to Placebo,a Median (95% CI)χKW2, 95P
Total PCSI-Y 16 (13 to 35) 16 (12 to 26) 27 (24 to 34) −2 (−13 to 6) 4 (−7 to 14) 2.024 .36 
 Physical 8.0 (4 to 12) 8.0 (5 to 10) 10.0 (6 to 13) 0 (−4 to 4) 2 (−3 to 6)   
 Cognitive 6.0 (3 to 11) 4.0 (2 to 8) 7.0 (4 to 8) 1 (−5 to 2) 0 (3 to −3)   
 Emotional 2.0 (0 to 5) 2.0 (0 to 5) 5.0 (2 to 6) 0 (−3 to 2) 1 (−3 to 3)   
 Fatigue 2.0 (1 to 4) 3.00 (2 to 4) 4.0 (2 to 5) 0 (−2 to 2) 1 (−1 to 3)   
Total PCSI-P 12.50 (7 to 16) 15 (10 to 21) 9.5 (4.5 to 19.0) 3 (−3 to 11) −2 (−10 to 6) 1.692 .43 
 Physical 5.0 (2 to 9) 6.0 (2 to 8) 6.5 (4 to 8) 1 (−2 to 4) 0 (−2 to 3)   
 Cognitive 2.0 (0 to 6) 3.0 (2 to 4) 2.0 (2 to 6) 1 (−3 to 4) 1 (−1 to 2)   
 Emotional 2.0 (0 to 4) 1.0 (0 to 3) 2.5 (0 to 4) 0 (−2 to 1) 0 (−1 to 2)   
 Fatigue 2.0 (0 to 4) 3.00 (1 to 5) 2.0 (1 to 4) 0 (−1 to 2) 1 (−1 to 2)   

Positive scores indicate improvement.

a

Hodges-Lehman median difference between treatment group and placebo; positive score indicates improvement with melatonin 3 mg or melatonin 10mg .

FIGURE 2

Box plots demonstrating the PCSI-Y scores in each group before and immediately after treatment and at 4- and 6-month follow-up time points. There was no effect of melatonin on PCSI-Y change scores immediately after treatment compared with a placebo (χ2 = 2.024; P = .36).

FIGURE 2

Box plots demonstrating the PCSI-Y scores in each group before and immediately after treatment and at 4- and 6-month follow-up time points. There was no effect of melatonin on PCSI-Y change scores immediately after treatment compared with a placebo (χ2 = 2.024; P = .36).

Close modal

Kaplan-Meier survival curves in Fig 3 illustrate no significant group differences in the probability of symptom survival during treatment or 6-month follow-up. The Cox proportional hazards model showed no significant difference in hazard ratios of symptoms returning to baseline between the placebo and melatonin 3-mg (1.00 [95% CI: 0.49 to 2.06; P = .99]) groups and the melatonin 10-mg group (1.11 [95% CI: 0.54 to 2.29; P = .77]). The proportionality assumption was not violated. Regression revealed no effect of melatonin after any effect of sleep efficiency (pretreatment) was considered (F[1,84] = 0.158; P = .69). Per-protocol analysis did not show any evidence of a favorable effect of melatonin on PCSI-Y change (KW = 2.811; P = .24).

FIGURE 3

Kaplan-Meier curves demonstrating the probability of not returning to preinjury PCSI symptom scores after starting treatment with a placebo, melatonin 3 mg, and melatonin 10 mg for 4 weeks. The dotted lines represent 95% CIs.

FIGURE 3

Kaplan-Meier curves demonstrating the probability of not returning to preinjury PCSI symptom scores after starting treatment with a placebo, melatonin 3 mg, and melatonin 10 mg for 4 weeks. The dotted lines represent 95% CIs.

Close modal

Paired sleep actigraphy parameters before and after treatment were available for 66 participants: placebo,23  melatonin 3 mg,16  and melatonin 10 mg.25  Missing data were due to watch unavailability,16  watch malfunction,10  and loss of watch.2  No group differences on actigraphy sleep parameters were apparent after treatment (Table 3).

TABLE 3

Changes in Sleep Actigraphy Parameters Before and After Treatment (Posttreatment Minus Pretreatment) by Treatment Group

Change in Sleep ParameterPlacebo (n = 23), Median (95% CI)Melatonin 3 mg (n = 16), Median (95% CI)Melatonin 10 mg (n = 25), Median (95% CI)χKW(2)P
Total sleep time, min −17 (−33.2 to 2.7) −10 (−40.6 to 5.5) −8.5 (−29.6 to 22.4) 1.113 .57 
Sleep onset latency, min 1.0 (−3.5 to 9.2) 8.4 (−3.9 to 12.2) 1.9 (−7.7 to 12.0) 1.192 .55 
WASO, min 1.9 (−1.9 to 10.7) −0.4 (−3.2 to 3.8) −0.4 (−3.3 to 0.8) 3.565 .17 
Sleep efficiency, % −0.1 (−3.2 to 2.1) −4.1 (−10.3 to 1.9) −3.1 (−6.7 to 4.6) 0.070 .97 
Change in Sleep ParameterPlacebo (n = 23), Median (95% CI)Melatonin 3 mg (n = 16), Median (95% CI)Melatonin 10 mg (n = 25), Median (95% CI)χKW(2)P
Total sleep time, min −17 (−33.2 to 2.7) −10 (−40.6 to 5.5) −8.5 (−29.6 to 22.4) 1.113 .57 
Sleep onset latency, min 1.0 (−3.5 to 9.2) 8.4 (−3.9 to 12.2) 1.9 (−7.7 to 12.0) 1.192 .55 
WASO, min 1.9 (−1.9 to 10.7) −0.4 (−3.2 to 3.8) −0.4 (−3.3 to 0.8) 3.565 .17 
Sleep efficiency, % −0.1 (−3.2 to 2.1) −4.1 (−10.3 to 1.9) −3.1 (−6.7 to 4.6) 0.070 .97 

Cognitive and behavioral outcomes are reported in Table 4. Five participants displayed noncredible effort on the TOMM and were excluded from the analysis of cognitive function. Groups did not differ on changes in cognitive testing, executive function, or internalizing problems after treatment. Externalizing problems (hyperactivity, conduct, and aggression) were significantly different across groups after treatment (F[2, 84] = 4.928; P = .009). Post hoc paired comparisons demonstrated decreased externalizing problems in the 3-mg melatonin group (P = .008). This had a large effect size (−0.797 [95% CI: −1.327 to −0.266]) compared with the placebo.

TABLE 4

Cognitive and Behavioral Function Before and in the Week After Treatment in Trial Participants

Pretreatment, Mean (SD)Posttreatment, Mean (SD)Change After Treatment, Mean (SD)F(2, 84)P
PlaceboMelatonin 3 mgMelatonin 10 mgPlaceboMelatonin 3 mgMelatonin 10 mgPlaceboMelatonin 3 mgMelatonin 10 mg
CNS Vital Signs            
 Neurocognitive Index 98.73 (11.13) 93.64 (14.77) 98.04 (11.77) 102.13 (9.68) 99.32 (11.30) 102.14 (11.09) 3.4 (8.43) 5.6 (8.52) 4.11 (8.02) 0.563 .57 
 Composite memory 95.37 (15.75) 93.21 (24.30) 98.45 (17.75) 92.47 (16.81) 93.36 (20.75) 94.86 (20.81) −2.9 (16.53) .1 (16.67) −3.6 (18.89) — — 
 Verbal memory 94.84 (16.09) 92.04 (22.25) 98.66 (18.52) 96.06 (19.65) 97.36 (20.76) 94.0 (21.75) 1.2 (21.26) 5.3 (20.83) −4.7 (23.61) — — 
 Visual memory 97.0 (14.46) 96.75 (20.13) 96.75 (20.13) 92.6 (13.75) 92.57 (20.19) 97.41 (17.93) −4.4 (14.25) −4.2 (16.31) −1.6 (19.02) — — 
 Psychomotor speed 101.74 (17.32) 95.68 (17.07) 99.9 (14.22) 106.58 (13.56) 102.96 (15.35) 104.24 (13.64) 4.8 (11.63) 6.8 (11.98) 4.3 (7.74) — — 
 Reaction time 97.06 (14.929) 95.93 (15.64) 97.55 (14.87) 97.06 (15.64) 97.39 (15.48) 103.34 (16.91) 0.0 (13.09) 1.2 (13.50) 5.9 (13.80) — — 
 Complex attention 96.97 (14.19) 88.18 (23.99) 92.32 (22.12) 103.55 (15.05) 97.61 (18.01) 100.43 (14.53) 6.5 (16.12) 9.4 (14.16) 8.1 (16.62) — — 
 Cognitive flexibility 103.03 (13.37) 94.79 (17.03) 102.43 (15.53) 110.61 (11.40) 104.89 (14.10) 108.07 (14.29) −0.1 (0.25) 0.0 (0.19) −0.1 (0.31) — — 
 Processing speed 112.55 (12.87) 110.04 (20.12) 107.11 (17.83) 119.19 (15.83) 117.07 (19.87) 113.43 (17.37) 6.6 (9.82) 6.5 (14.33) 6.1 (13.2) — — 
 Executive function 104.13 (13.08) 97.54 (14.79) 104.25 (14.63) 104.13 (13.08) 97.54 (14.79) 104.25 (14.63) 6.9 (9.26) 5.9 (22.46) 6.4 (8.51) — — 
Behavior Assessment System for Children, Second Edition, Parent Report            
 Behavioral Symptoms Index 50.93 (8.47) 50.73 (7.72) 48.35 (8.89) 50.93 (8.26) 47.35 (6.91) 48.42 (9.95) 0.0 (4.18) −3.4 (6.26) 0.1 (6.02) 3.396 .04 
 Internalizing behaviors 55.48 (11.66) 55.73 (9.22) 53.35 (8.11) 54.14 (11.75) 51.81 (9.67) 51.04 (9.46) −1.3 (7.33) −3.9 (7.78) 2.3 (5.43) 0.959 .39 
 Externalizing behaviors 47.66 (6.90) 48.85 (7.87) 47.42 (9.23) 48.52 (7.46) 46.15 (7.67) 46.81 (9.73) 0.9 (4.10) −2.9 (5.35) −0.4 (4.65) 4.928 .01a 
Behavior Rating Inventory of Executive Function, Parent Report            
 Global executive composite 53.03 (9.23) 52.23 (10.12) 50.31 (8.92) 52.17 (11.26) 48.81 (9.76) 50.23 (11.35) −0.9 (6.93) −4.1 (6.84) −0.2 (4.78) 1.003 .37 
 Metacognition 53.79 (8.76) 52.04 (9.70) 51.38 (8.12) 53.14 (1.12) 49.23 (9.61) 51.35 (11.0) −0.7 (6.51) −3.3 (6.05) 0.1 (4.91) 2.768 .07 
 Behavioral regulation 51.76 (10.62) 52.12 (10.31) 48.5 (10.89) 50.83 (11.17) 48.54 (9.07) 48.62 (11.15) −0.9 (7.62) −4.3 (7.63) 0.4 (4.76) 0.771 .47 
Pretreatment, Mean (SD)Posttreatment, Mean (SD)Change After Treatment, Mean (SD)F(2, 84)P
PlaceboMelatonin 3 mgMelatonin 10 mgPlaceboMelatonin 3 mgMelatonin 10 mgPlaceboMelatonin 3 mgMelatonin 10 mg
CNS Vital Signs            
 Neurocognitive Index 98.73 (11.13) 93.64 (14.77) 98.04 (11.77) 102.13 (9.68) 99.32 (11.30) 102.14 (11.09) 3.4 (8.43) 5.6 (8.52) 4.11 (8.02) 0.563 .57 
 Composite memory 95.37 (15.75) 93.21 (24.30) 98.45 (17.75) 92.47 (16.81) 93.36 (20.75) 94.86 (20.81) −2.9 (16.53) .1 (16.67) −3.6 (18.89) — — 
 Verbal memory 94.84 (16.09) 92.04 (22.25) 98.66 (18.52) 96.06 (19.65) 97.36 (20.76) 94.0 (21.75) 1.2 (21.26) 5.3 (20.83) −4.7 (23.61) — — 
 Visual memory 97.0 (14.46) 96.75 (20.13) 96.75 (20.13) 92.6 (13.75) 92.57 (20.19) 97.41 (17.93) −4.4 (14.25) −4.2 (16.31) −1.6 (19.02) — — 
 Psychomotor speed 101.74 (17.32) 95.68 (17.07) 99.9 (14.22) 106.58 (13.56) 102.96 (15.35) 104.24 (13.64) 4.8 (11.63) 6.8 (11.98) 4.3 (7.74) — — 
 Reaction time 97.06 (14.929) 95.93 (15.64) 97.55 (14.87) 97.06 (15.64) 97.39 (15.48) 103.34 (16.91) 0.0 (13.09) 1.2 (13.50) 5.9 (13.80) — — 
 Complex attention 96.97 (14.19) 88.18 (23.99) 92.32 (22.12) 103.55 (15.05) 97.61 (18.01) 100.43 (14.53) 6.5 (16.12) 9.4 (14.16) 8.1 (16.62) — — 
 Cognitive flexibility 103.03 (13.37) 94.79 (17.03) 102.43 (15.53) 110.61 (11.40) 104.89 (14.10) 108.07 (14.29) −0.1 (0.25) 0.0 (0.19) −0.1 (0.31) — — 
 Processing speed 112.55 (12.87) 110.04 (20.12) 107.11 (17.83) 119.19 (15.83) 117.07 (19.87) 113.43 (17.37) 6.6 (9.82) 6.5 (14.33) 6.1 (13.2) — — 
 Executive function 104.13 (13.08) 97.54 (14.79) 104.25 (14.63) 104.13 (13.08) 97.54 (14.79) 104.25 (14.63) 6.9 (9.26) 5.9 (22.46) 6.4 (8.51) — — 
Behavior Assessment System for Children, Second Edition, Parent Report            
 Behavioral Symptoms Index 50.93 (8.47) 50.73 (7.72) 48.35 (8.89) 50.93 (8.26) 47.35 (6.91) 48.42 (9.95) 0.0 (4.18) −3.4 (6.26) 0.1 (6.02) 3.396 .04 
 Internalizing behaviors 55.48 (11.66) 55.73 (9.22) 53.35 (8.11) 54.14 (11.75) 51.81 (9.67) 51.04 (9.46) −1.3 (7.33) −3.9 (7.78) 2.3 (5.43) 0.959 .39 
 Externalizing behaviors 47.66 (6.90) 48.85 (7.87) 47.42 (9.23) 48.52 (7.46) 46.15 (7.67) 46.81 (9.73) 0.9 (4.10) −2.9 (5.35) −0.4 (4.65) 4.928 .01a 
Behavior Rating Inventory of Executive Function, Parent Report            
 Global executive composite 53.03 (9.23) 52.23 (10.12) 50.31 (8.92) 52.17 (11.26) 48.81 (9.76) 50.23 (11.35) −0.9 (6.93) −4.1 (6.84) −0.2 (4.78) 1.003 .37 
 Metacognition 53.79 (8.76) 52.04 (9.70) 51.38 (8.12) 53.14 (1.12) 49.23 (9.61) 51.35 (11.0) −0.7 (6.51) −3.3 (6.05) 0.1 (4.91) 2.768 .07 
 Behavioral regulation 51.76 (10.62) 52.12 (10.31) 48.5 (10.89) 50.83 (11.17) 48.54 (9.07) 48.62 (11.15) −0.9 (7.62) −4.3 (7.63) 0.4 (4.76) 0.771 .47 

Computerized cognition was assessed by using CNS Vital Signs, which provides an overall summary score of cognitive function: the Neurocognitive Index. Parents completed the assessments of behavioral adjustment and executive function. No significant differences between groups were seen in cognitive assessments and executive function before and after treatment. —, not applicable.

a

Group differences in change in externalizing behaviors were observed, and post hoc pairwise comparisons (Sidak) demonstrated decreased externalizing behaviors in children treated with 3 mg of melatonin (F[2,84] = 4.928; P =·.01). Positive F values indicate more favorable behavioral change.

Youth and parents in all groups reported improved quality of life at the end of treatment (Supplemental Table 6). Mean parental rating of physical functioning significantly improved from 34.55 to 45.46 (change 95% CI: 8.12 to 13.70; t[86] = 7.77; P < .001), and psychosocial functioning increased from 42.07 to 47.50 (change 95% CI: 3.78 to 7.99; t[86] = 5.55; P < .001). Changes in quality of life significantly correlated with changes in PCSI-P total scores (Supplemental Fig 4). However, the treatment groups did not differ significantly.

Pretreatment overnight urinary aMT6s levels were not different between groups (mean = 54.59; SD = 33.40; F[2,67] = 0.87; P = .42). Forty-two participants missed at least 1 dose, and 5 participants missed 5 doses or more. Midtreatment urinary aMT6s levels increased in the melatonin groups only: placebo (mean = 36.85; SD = 20.94), melatonin 3 mg (mean = 2595.82; SD = 2891.34), and melatonin 10 mg (mean = 9889.22; SD = 11 372.35; F[2,56] = 9.256; P < .001). Sensitivity analyses revealed no effect of compliance or urinary melatonin levels on outcome.

Thirty-two adverse events were reported in 28 participants: placebo (n = 8), melatonin 3 mg (n = 13), and melatonin 10 mg (n = 11). One participant had a serious adverse event (appendicitis) unrelated to the study drug. Eight events involved a known melatonin side effect, and 5 were potentially related (Supplemental Table 7). Ten events were associated with a mild functional impact. The frequency (χ2 = 1.755; P = .42) and severity (χ6 = 6.619; P = .36) of adverse events did not differ between groups.

In children with PPCS, the administration of melatonin at 4 weeks postinjury for 28 days did not significantly improve postconcussion symptoms compared with a placebo nor was there any effect of melatonin apparent on cognitive or health-related quality of life outcomes. Similarly, most measures of behavior were unchanged. We observed wide CIs in the primary outcome measure, however, suggesting that the sample size was insufficient to definitively conclude an absence of effect. Interestingly, parents reported fewer externalizing problems (mainly decreased hyperactivity) in those participants treated with melatonin, as reported previously in children with neurodevelopmental disability.40  The mean change of 2.7 T-scores seen in our study, however, is not likely to be clinically significant. Melatonin is often recommended in the management of pediatric PPCS, and both melatonin and a derivative, ramelteon, have been reported to improve sleep complaints in moderate or severe adult TBI.12,19,21,4143  Pretreatment sleep actigraphy parameters in our study, however, were not predictive of PPCS improvement with melatonin treatment.

This study has several strengths. Although many studies have examined recovery from concussion, few have systematically examined treatments for PPCS. Previous trials have been small, examined a variety of therapies,2,1315  and compared interventions to routine care or no treatment and so are susceptible to attribution bias.44  This is the first randomized placebo-controlled trial of a pharmacologic intervention. It is also one of the largest studies evaluating treatment in children with PPCS. The results are generalizable to typically developing children with mTBI from a variety of etiologies, including sport-related concussion. Children were enrolled and carefully phenotyped at a similar and tight time frame postinjury. As well as recording postconcussive symptoms, we also examined other traditional neurocognitive and behavioral outcomes that may be more robust than symptom ratings and allow for comparisons between treatment groups and other populations.

This trial provides useful information about PPCS in children. Participants at enrollment had a high symptom burden and significant impairments in their quality of life. Despite this, neurocognitive, executive, and behavioral function remained well within normal ranges. Participants performed well on symptom validity testing, and only 5% failed to demonstrate credible effort on the TOMM, suggesting that aberrant psychological responses to injury, if present, were not likely to have affected neuropsychological performance.45,46 

A high proportion of participants displayed significant clinical improvement during the intervention period. This could be due to the natural history of PPCS, comprehensive standardized care, or a placebo effect. Given our previous experience in studies involving similar populations, this preponderance for improvement is unlikely to be due to natural history alone1,47  but could be artificially inflated by the exclusion of children with a significant psychiatric history.48  More likely, however, the potential treatment effect is due to the supportive multidisciplinary care model employed in the brain injury clinic and is congruent with outcomes in a collaborative care model for the management of PPCS.49  Although neither of these factors should affect the results of this trial, the placebo effect could lead to an underestimation of the effect of melatonin.50,51 

Melatonin doses were chosen to stay within common clinical practice parameters to minimize risks to participants and achieve supraphysiological levels sufficient to target both receptor-mediated and subcellular processes.52  We achieved this goal, observing excellent tolerability and treatment compliance while demonstrating supraphysiological overnight urinary melatonin levels. The observed melatonin levels and our sensitivity analyses, adjusting for compliance, suggested that treatment failure was unlikely to be due to insufficient melatonin dosing at night.

The lack of an observable effect of melatonin in our study was somewhat surprising, but several possibilities could explain this outcome. Posttraumatic oxidative stress peaks between 3 and 5 days postinjury, although its temporal course and role in chronic mTBI have not been fully elucidated.53,54  Melatonin may need to be used within the first few days of injury, when oxidative stress is at its highest, to capitalize on its antioxidant properties and reduce the biological consequences of mTBI.55,56  The excellent tolerability and side-effect profile of melatonin seen in this study provides reassurance that it could be administered early at low risk to the patient. Conversely, the antiinflammatory effects of melatonin may be more evident with longer treatment durations if persistent neuroinflammation is playing a role in PPCS.1,5  Future studies are needed to evaluate these hypotheses.

This study has some limitations. The sample size for this study was determined on the basis of 80% power, resulting in the 20% chance that our study was underpowered. The wide CIs found for our primary outcomes indicate that our sample size was too small. Therefore, the results of this study must be interpreted with caution and need to be replicated in a larger sample. Obtaining retrospective reports of preinjury symptoms at enrollment could have introduced a “good-old-days” bias57,58  that may result in recovery rates being underestimated; however, this should not have affected any response to the intervention. To minimize bias due to attrition, we used an intention-to-treat analysis, imputing missing data using the last observation carried forward approach. Although this technique could introduce bias into the analysis, because the primary outcome data were missing in only 3 participants (1 per treatment group), it is unlikely to have affected our results. A specific pediatric sleep questionnaire would have improved the characterization of sleep disturbances.

The administration of melatonin at 4 weeks postinjury did not significantly improve postconcussion symptoms in children with PPCS compared with a placebo. Seventy-eight percent of children with PPCS demonstrated significant recovery between 1 and 3 months postinjury. The evidence from this randomized controlled trial does not support the use of melatonin for the treatment of PPCS during this time period postinjury.

Deidentified participant data (including a data dictionary), the protocol, and the statistical analysis plan that underlie the reported results (text, tables, figures, and appendices) will be available on publication for 5 years. The data (including the clinical data report) will be available to any researchers with a methodologically sound proposal and relevant ethical approvals through the University of Queensland eSpace. Proposals should be directed to [email protected]; to gain access, data requestors will need to sign a data access agreement.

Drs Barlow and Dewey were involved in all parts of the study, including study design, obtaining funding, data collection, analysis, and writing of manuscript; Drs Brooks, Kirton, Zemek, MacMaster, Nettel-Aguirre, Yeates, Kirk, Esser, Hill, and Buchhalter, Ms Crawford, and Ms Cameron were involved in the study design, trial operation, analysis, and preparation of the manuscript; Ms Turley and Dr Samuel were involved in data acquisition, data cleaning, and manuscript preparation; Drs Richer, Platt, Boyd, and Hutchison were involved in trial supervision and manuscript preparation; and all authors approved the final manuscript as submitted and agree to be accountable for all aspects of the work.

This trial has been registered at clinicaltrials.gov (identifier NCT01874847).

FUNDING: Funded by the Canadian Institutes of Health Research (grant 293375), the Alberta Children’s Hospital Research Institute, and the University of Calgary.

aMT6s

urinary 6-sulphatoxymelatonin

CI

confidence interval

KW

Kruskal-Wallis test

mTBI

mild traumatic brain injury

PCSI

Post-Concussion Symptom Inventory

PCSI-P

Post-Concussion Symptom Inventory–Parent

PCSI-Y

Post-Concussion Symptom Inventory–Youth

PPCS

persistent postconcussive symptoms

TBI

traumatic brain injury

TOMM

Test of Memory Malingering

WASO

wake after sleep onset

1
Barlow
KM
,
Crawford
S
,
Stevenson
A
,
Sandhu
SS
,
Belanger
F
,
Dewey
D
.
Epidemiology of postconcussion syndrome in pediatric mild traumatic brain injury
.
Pediatrics
.
2010
;
126
(
2
).
2
Makdissi
M
,
Schneider
KJ
,
Feddermann-Demont
N
, et al
.
Approach to investigation and treatment of persistent symptoms following sport-related concussion: a systematic review
.
Br J Sports Med
.
2017
;
51
(
12
):
958
968
3
Langlois
JA
,
Rutland-Brown
W
,
Thomas
KE
.
The incidence of traumatic brain injury among children in the United States: differences by race
.
J Head Trauma Rehabil
.
2005
;
20
(
3
):
229
238
4
McKinlay
A
,
Grace
RC
,
Horwood
LJ
,
Fergusson
DM
,
Ridder
EM
,
MacFarlane
MR
.
Prevalence of traumatic brain injury among children, adolescents and young adults: prospective evidence from a birth cohort
.
Brain Inj
.
2008
;
22
(
2
):
175
181
5
Zemek
R
,
Barrowman
N
,
Freedman
SB
, et al;
Pediatric Emergency Research Canada (PERC) Concussion Team
.
Clinical risk score for persistent postconcussion symptoms among children with acute concussion in the ED
.
JAMA
.
2016
;
315
(
10
):
1014
1025
6
Zemek
RL
,
Yeates
KO
.
Rates of persistent postconcussive symptoms
.
JAMA
.
2017
;
317
(
13
):
1375
1376
7
Novak
Z
,
Aglipay
M
,
Barrowman
N
, et al;
Pediatric Emergency Research Canada Predicting Persistent Postconcussive Problems in Pediatrics (PERC 5P) Concussion Team
.
Association of persistent postconcussion symptoms with pediatric quality of life
.
JAMA Pediatr
.
2016
;
170
(
12
):
e162900
8
Ganesalingam
K
,
Yeates
KO
,
Ginn
MS
, et al
.
Family burden and parental distress following mild traumatic brain injury in children and its relationship to post-concussive symptoms
.
J Pediatr Psychol
.
2008
;
33
(
6
):
621
629
9
McCrory
P
,
Meeuwisse
W
,
Dvořák
J
, et al
.
Consensus statement on concussion in sport-the 5th international conference on concussion in sport held in Berlin, October 2016
.
Br J Sports Med
.
2017
;
51
(
11
):
838
847
10
Grool
AM
,
Aglipay
M
,
Momoli
F
, et al;
Pediatric Emergency Research Canada (PERC) Concussion Team
.
Association between early participation in physical activity following acute concussion and persistent postconcussive symptoms in children and adolescents
.
JAMA
.
2016
;
316
(
23
):
2504
2514
11
Barlow
KM
.
Postconcussion syndrome: a review
.
J Child Neurol
.
2016
;
31
(
1
):
57
67
12
Hingley
S
,
Ross
J
.
Guidelines for diagnosing and managing paediatric concussion: Ontario Neurotrauma Foundation guideline
.
Arch Dis Child Educ Pract Ed
.
2016
;
101
(
2
):
58
60
13
Schneider
KJ
,
Meeuwisse
WH
,
Nettel-Aguirre
A
, et al
.
Cervicovestibular rehabilitation in sport-related concussion: a randomised controlled trial
.
Br J Sports Med
.
2014
;
48
(
17
):
1294
1298
14
Leddy
JJ
,
Cox
JL
,
Baker
JG
, et al
.
Exercise treatment for postconcussion syndrome: a pilot study of changes in functional magnetic resonance imaging activation, physiology, and symptoms
.
J Head Trauma Rehabil
.
2013
;
28
(
4
):
241
249
15
Gagnon
I
,
Galli
C
,
Friedman
D
,
Grilli
L
,
Iverson
GL
.
Active rehabilitation for children who are slow to recover following sport-related concussion
.
Brain Inj
.
2009
;
23
(
12
):
956
964
16
Maldonado
MD
,
Murillo-Cabezas
F
,
Terron
MP
, et al
.
The potential of melatonin in reducing morbidity-mortality after craniocerebral trauma
.
J Pineal Res
.
2007
;
42
(
1
):
1
11
17
Barlow
KM
,
Esser
MJ
,
Veidt
M
,
Boyd
R
.
Melatonin as a treatment after traumatic brain injury: a systematic review and meta-analysis of the pre-clinical and clinical literature
.
J Neurotrauma
.
2019
;
36
(
4
):
523
537
18
Maldonado
MD
,
Manfredi
M
,
Ribas-Serna
J
,
Garcia-Moreno
H
,
Calvo
JR
.
Melatonin administrated immediately before an intense exercise reverses oxidative stress, improves immunological defenses and lipid metabolism in football players
.
Physiol Behav
.
2012
;
105
(
5
):
1099
1103
19
Ponsford
JL
,
Ziino
C
,
Parcell
DL
, et al
.
Fatigue and sleep disturbance following traumatic brain injury–their nature, causes, and potential treatments
.
J Head Trauma Rehabil
.
2012
;
27
(
3
):
224
233
20
Kuczynski
A
,
Crawford
S
,
Bodell
L
,
Dewey
D
,
Barlow
KM
.
Characteristics of post-traumatic headaches in children following mild traumatic brain injury and their response to treatment: a prospective cohort
.
Dev Med Child Neurol
.
2013
;
55
(
7
):
636
641
21
Mahooti
N
.
Sports-related concussion: acute management and chronic postconcussive issues
.
Child Adolesc Psychiatr Clin N Am
.
2018
;
27
(
1
):
93
108
22
Santini
A
,
Cammarata
SM
,
Capone
G
, et al
.
Nutraceuticals: opening the debate for a regulatory framework
.
Br J Clin Pharmacol
.
2018
;
84
(
4
):
659
672
23
Barlow
KM
,
Brooks
BL
,
MacMaster
FP
, et al
.
A double-blind, placebo-controlled intervention trial of 3 and 10 mg sublingual melatonin for post-concussion syndrome in youths (PLAYGAME): study protocol for a randomized controlled trial
.
Trials
.
2014
;
15
:
271
24
Giza
CC
,
Kutcher
JS
,
Ashwal
S
, et al
.
Summary of evidence-based guideline update: evaluation and management of concussion in sports: report of the Guideline Development Subcommittee of the American Academy of Neurology
.
Neurology
.
2013
;
80
(
24
):
2250
2257
25
Teel
EF
,
Zemek
RL
,
Tang
K
, et al;
Pediatric Emergency Research Canada (PERC) Concussion Team
.
The stability of retrospective pre-injury symptom ratings following pediatric concussion
.
Front Neurol
.
2019
;
10
:
672
26
Janusz
JA
,
Sady
MD
,
Gioia
GA
. Postconcussion Symptom Assessment. In:
Kirkwood
MW
,
Yeates
KO
, eds.
Mild Traumatic Brain Injury in Children and Adolescents: From Basic Science to Clinical Management
.
New York, NY
:
Guilford Press
;
2012
:
241
263
27
Sady
MD
,
Vaughan
CG
,
Gioia
GA
.
Psychometric characteristics of the Postconcussion Symptom Inventory in children and adolescents
.
Arch Clin Neuropsychol
.
2014
;
29
(
4
):
348
363
28
Landgraf
JF
.
Child Health Questionnaire (CHQ): A User’s Manual
.
Boston, MA
:
HealthAct
;
1996
29
Donders
J
.
Performance on the Test of Memory Malingering in a mixed pediatric sample
.
Child Neuropsychol
.
2005
;
11
(
2
):
221
227
30
Mrazik
M
,
Brooks
BL
,
Jubinville
A
,
Meeuwisse
WH
,
Emery
CA
.
Psychosocial outcomes of sport concussions in youth hockey players
.
Arch Clin Neuropsychol
.
2016
;
31
(
4
):
297
304
31
Donders
J
,
DenBraber
D
,
Vos
L
.
Construct and criterion validity of the Behaviour Rating Inventory of Executive Function (BRIEF) in children referred for neuropsychological assessment after paediatric traumatic brain injury
.
J Neuropsychol
.
2010
;
4
(
pt 2
):
197
209
32
Brooks
BL
,
Khan
S
,
Daya
H
,
Mikrogianakis
A
,
Barlow
KM
.
Neurocognition in the emergency department after a mild traumatic brain injury in youth
.
J Neurotrauma
.
2014
;
31
(
20
):
1744
1749
33
Gualtieri
CT
,
Johnson
LG
.
Reliability and validity of a computerized neurocognitive test battery, CNS Vital Signs
.
Arch Clin Neuropsychol
.
2006
;
21
(
7
):
623
643
34
Jungquist
CR
,
Pender
JJ
,
Klingman
KJ
,
Mund
J
.
Validation of capturing sleep diary data via a wrist-worn device
.
Sleep Disord
.
2015
;
2015
:
758937
35
Klante
G
,
Brinschwitz
T
,
Secci
K
,
Wollnik
F
,
Steinlechner
S
.
Creatinine is an appropriate reference for urinary sulphatoxymelatonin of laboratory animals and humans
.
J Pineal Res
.
1997
;
23
(
4
):
191
197
36
Jacobson
NS
,
Truax
P
.
Clinical significance: a statistical approach to defining meaningful change in psychotherapy research
.
J Consult Clin Psychol
.
1991
;
59
(
1
):
12
19
37
Register-Mihalik
JK
,
Guskiewicz
KM
,
Mihalik
JP
,
Schmidt
JD
,
Kerr
ZY
,
McCrea
MA
.
Reliable change, sensitivity, and specificity of a multidimensional concussion assessment battery: implications for caution in clinical practice
.
J Head Trauma Rehabil
.
2013
;
28
(
4
):
274
283
38
Register-Mihalik
JK
,
Guskiewicz
KM
,
Marshall
SW
, et al
.
Methodology and implementation of a randomized controlled trial (RCT) for early rehabilitation post-concussion: the active rehab study
.
Front Neurol
.
2019
;
10
:
1176
39
Efron
B
.
Better bootstrap confidence intervals
.
J Am Stat Assoc
.
1987
;
82
(
397
):
171
185
40
Schroder
CM
,
Malow
BA
,
Maras
A
, et al
.
Pediatric prolonged-release melatonin for sleep in children with autism spectrum disorder: impact on child behavior and caregiver’s quality of life
.
J Autism Dev Disord
.
2019
;
49
(
8
):
3218
3230
41
Grima
NA
,
Rajaratnam
SMW
,
Mansfield
D
,
Sletten
TL
,
Spitz
G
,
Ponsford
JL
.
Efficacy of melatonin for sleep disturbance following traumatic brain injury: a randomised controlled trial
.
BMC Med
.
2018
;
16
(
1
):
8
42
Lequerica
A
,
Jasey
N
,
Portelli Tremont
JN
,
Chiaravalloti
ND
.
Pilot study on the effect of ramelteon on sleep disturbance after traumatic brain injury: preliminary evidence from a clinical trial
.
Arch Phys Med Rehabil
.
2015
;
96
(
10
):
1802
1809
43
Mannix
R
,
Zemek
R
,
Yeates
KO
, et al
.
Practice patterns in pharmacological and non-pharmacological therapies for children with mild traumatic brain injury: a survey of 15 Canadian and United States centers
.
J Neurotrauma
.
2019
;
36
(
20
):
2886
2894
44
Winkler
R
,
Taylor
NF
.
Do children and adolescents with mild traumatic brain injury and persistent symptoms benefit from treatment? A systematic review
.
J Head Trauma Rehabil
.
2015
;
30
(
5
):
324
333
45
Yeates
KO
,
Taylor
HG
,
Rusin
J
, et al
.
Premorbid child and family functioning as predictors of post-concussive symptoms in children with mild traumatic brain injuries
.
Int J Dev Neurosci
.
2012
;
30
(
3
):
231
237
46
Kirkwood
MW
,
Yeates
KO
,
Randolph
C
,
Kirk
JW
.
The implications of symptom validity test failure for ability-based test performance in a pediatric sample
.
Psychol Assess
.
2012
;
24
(
1
):
36
45
47
Barlow
KM
,
Crawford
S
,
Brooks
BL
,
Turley
B
,
Mikrogianakis
A
.
The incidence of postconcussion syndrome remains stable following mild traumatic brain injury in children
.
Pediatr Neurol
.
2015
;
53
(
6
):
491
497
48
Max
JE
,
Friedman
K
,
Wilde
EA
, et al
.
Psychiatric disorders in children and adolescents 24 months after mild traumatic brain injury
.
J Neuropsychiatry Clin Neurosci
.
2015
;
27
(
2
):
112
120
49
McCarty
CA
,
Zatzick
D
,
Stein
E
,
Wang
J
,
Hilt
R
,
Rivara
FP
;
Seattle Sports Concussion Research Collaborative
.
Collaborative care for adolescents with persistent postconcussive symptoms: a randomized trial
.
Pediatrics
.
2016
;
138
(
4
):
e20160459
50
Castelnuovo
G
,
Giusti
EM
,
Manzoni
GM
, et al
.
What is the role of the placebo effect for pain relief in neurorehabilitation? Clinical implications from the Italian consensus conference on pain in neurorehabilitation
.
Front Neurol
.
2018
;
9
:
310
51
Lewis
DW
,
Winner
P
,
Wasiewski
W
.
The placebo responder rate in children and adolescents
.
Headache
.
2005
;
45
(
3
):
232
239
52
Camins
A
,
Sureda
FX
,
Junyent
F
, et al
.
An overview of investigational antiapoptotic drugs with potential application for the treatment of neurodegenerative disorders
.
Expert Opin Investig Drugs
.
2010
;
19
(
5
):
587
604
53
Barkhoudarian
G
,
Hovda
DA
,
Giza
CC
.
The molecular pathophysiology of concussive brain injury - an update
.
Phys Med Rehabil Clin N Am
.
2016
;
27
(
2
):
373
393
54
Needham
EJ
,
Helmy
A
,
Zanier
ER
,
Jones
JL
,
Coles
AJ
,
Menon
DK
.
The immunological response to traumatic brain injury
.
J Neuroimmunol
.
2019
;
332
:
112
125
55
Fernández-Gajardo
R
,
Matamala
JM
,
Carrasco
R
,
Gutiérrez
R
,
Melo
R
,
Rodrigo
R
.
Novel therapeutic strategies for traumatic brain injury: acute antioxidant reinforcement
.
CNS Drugs
.
2014
;
28
(
3
):
229
248
56
Esposito
E
,
Cuzzocrea
S
.
Antiinflammatory activity of melatonin in central nervous system
.
Curr Neuropharmacol
.
2010
;
8
(
3
):
228
242
57
Brooks
BL
,
Kadoura
B
,
Turley
B
,
Crawford
S
,
Mikrogianakis
A
,
Barlow
KM
.
Perception of recovery after pediatric mild traumatic brain injury is influenced by the “good old days” bias: tangible implications for clinical practice and outcomes research
.
Arch Clin Neuropsychol
.
2014
;
29
(
2
):
186
193
58
Iverson
GL
,
Lange
RT
,
Brooks
BL
,
Rennison
VL
.
“Good old days” bias following mild traumatic brain injury
.
Clin Neuropsychol
.
2010
;
24
(
1
):
17
37

Competing Interests

POTENTIAL CONFLICT OF INTEREST: The authors have indicated they have no potential conflicts of interest to disclose.

FINANCIAL DISCLOSURE: Dr Brooks previously received in-kind support (free test credits for research) from the publisher of a computerized cognitive test (CNS Vital Signs, Morrisville, NC) for previous studies and receives royalties for 3 neuropsychological tests (Child and Adolescent Memory Profile, Multidimensional Everyday Memory Ratings for Youth, and Memory Validty Profile) that were not used in this study and for one textbook on pediatric forensic neuropsychology; the other authors have indicated they have no financial relationships relevant to this article to disclose.

Supplementary data