The American Psychiatric Association (APA) has updated its Privacy Policy and Terms of Use, including with new information specifically addressed to individuals in the European Economic Area. As described in the Privacy Policy and Terms of Use, this website utilizes cookies, including for the purpose of offering an optimal online experience and services tailored to your preferences.

Please read the entire Privacy Policy and Terms of Use. By closing this message, browsing this website, continuing the navigation, or otherwise continuing to use the APA's websites, you confirm that you understand and accept the terms of the Privacy Policy and Terms of Use, including the utilization of cookies.

×
Published Online:https://doi.org/10.1176/appi.ps.201800515

Abstract

Objective:

The objective of this study was to evaluate rates and correlates of stimulant medication adherence in a sample of pediatric patients using data derived from electronic medical records (EMRs) from a large health care organization in a large metropolitan area. The study relied on a novel definition of medication adherence as a timely renewal of an index prescription determined using the electronically recorded issuance of a stimulant prescription in the EMR (“refill”).

Methods:

Prescription and sociodemographic data were extracted from the Partners HealthCare Research Patient Data Registry to calculate adherence to stimulant medication treatment.

Results:

In the EMR, 2,206 patients with prescriptions for central nervous system stimulant medication were identified. Results showed that 46% of the index prescriptions were refilled within the timeframe necessary for the patient to be considered consistently medicated. A multivariable logistic regression model predicting medication adherence from patient demographic and treatment characteristics yielded an area-under-the-curve statistic of 0.57, indicating that these characteristics predicted adherence only modestly better than chance.

Conclusions:

EMR data from a large health care organization showed that 46% of pediatric patients were adherent to treatment with stimulants. Rates of medication adherence were worse among patients receiving care from a primary care provider than among those receiving care from a psychiatrist, in older patients, and in female patients and did not appear to be influenced by racial-ethnic group, economic class, stimulant type, or medication formulation (short or long acting). These findings, which show low rates of medication adherence among children and adolescents with ADHD, suggest the need for efforts to improve these rates.

HIGHLIGHTS

  • The objective of this study was to evaluate rates and correlates of adherence to stimulant medication among children and adolescents by using data derived from the electronic medical record (EMR) of a large health care organization in a large metropolitan area.

  • A novel definition of medication adherence was operationalized as a timely renewal of an index prescription of a stimulant as documented in the EMR.

  • EMR data showed that only 46% of more than 2,000 pediatric patients were adherent to treatment with stimulants.

  • Rates of treatment adherence were lower in the primary care setting, in older patients, and in female patients and did not appear to be influenced by racial-ethnic group, economic class, stimulant type, or medication formulation (short or long acting).

Attention-deficit hyperactivity disorder (ADHD) is a prevalent and morbid neurobiological disorder estimated to affect up to 11% of children in the United States (14). ADHD has been associated with a wide range of adverse outcomes, including educational and occupational underattainment (5); family disruption; interpersonal deficits (68); mood and anxiety disorders (9); addictions to cigarettes, drugs, and alcohol (10); injuries and accidents; automobile accidents (11); traumatic brain injury (1214); posttraumatic stress disorder (9); premature death (5, 15); and suicide (16, 17).

At the same time, ADHD is among the most treatable of all psychiatric disorders, with the documented safety and efficacy of stimulants (1827). Emerging data also document that treatment with stimulants decreases the risks for many ADHD-associated adverse outcomes, including addictions (28, 29), smoking initiation (7), mood and anxiety disorders (30), criminality (31), accidents and injuries (32, 33), and traumatic brain injury (34).

A limited literature suggests that compliance with stimulants is low (35, 36). Likely contributors to poor compliance include parental biases (37) and inadequate parental supervision (38). Perwien et al. (39) showed that patients with ADHD took stimulants consistently for 2 months or less, indicating that pharmacologic treatment in newly treated ADHD patients may be suboptimal and may adversely affect outcomes, including effectiveness of the treatment. Unfortunately, most of the literature on the subject of nonadherence to stimulants in ADHD does not provide specific rates in the general population, focusing instead on specific subpopulations (40, 41), such as those receiving public assistance (4245); specific ethnic groups, such as African Americans (46); and specific stimulant formulations, such as long- versus short-acting medications (4750). These findings call for improved efforts to better quantify the magnitude of the problem of low adherence to stimulants in broader populations of patients with ADHD.

Although the advent of electronic medical records (EMRs) offers a unique opportunity to investigate adherence to stimulants in pediatric populations, this technology has not been adequately utilized in the scientific literature. One of the few studies that used EMR data to quantify rates of adherence to stimulants among patients with ADHD reported that 84% of children were nonadherent to prescribed stimulant medication therapy during the first 2 months of treatment (39), suggesting that nonadherence can be observed shortly after initiation of stimulant treatment. This finding is noteworthy because short-term compliance with treatment may reflect the patient’s overall adherence to treatment. As shown in the literature, patients who adhere to treatment have better clinical outcomes than those who do not (51).

Further efforts to quantify and better understand rates of patient adherence to stimulant treatments could have important implications. If new insights can be gleaned regarding risk factors for low adherence, then efforts could be made to help mitigate this problem.

The main aim of this study was to quantify rates and correlates of patient adherence to stimulant treatment for ADHD. We conducted a systematic search of EMR from a large health care organization for children ages 4–17 years who had been prescribed an amphetamine or methylphenidate product between January 1, 2015, and December 31, 2016. We defined the children as adherent to stimulant medication treatment if the first prescription (hereafter referred to as the index prescription) was renewed in a timely fashion. On the basis of the literature, we hypothesized that adherence to stimulant medication for ADHD would be low.

Methods

Sample

Prescription and sociodemographic data were extracted from the Partners Health Care Research Patient Data Registry (RPDR) of Massachusetts General Hospital (MGH) for patients ages 4–17 years who had been prescribed a stimulant from January 1, 2015, to December 31, 2016. This period was based on the time when the newest EMR system from Epic Systems Corporation was implemented. Epic is one of the largest providers of health information technology used to access, organize, store, and share EMRs. Prescription information in Epic before January 1, 2015, was considered incomplete. This study was approved by the institutional review board at MGH.

Patients were included in the study sample if they had been prescribed any of the following formulations of stimulants (short- or long-acting): amphetamine/dextroamphetamine, dextroamphetamine or lisdexamfetamine, dexmethylphenidate, or methylphenidate. We included medications prescribed as a single prescription (30-day supply) and medications with prescriptions postdated by up to three months (60- or 90-day supply).

Index Prescription

We defined a patient’s index prescription as the first time a stimulant was prescribed for him or her during the study period of January 1, 2015, to December 31, 2016. This period was chosen because prescription data prior to January 1, 2015, were incomplete in the system, prohibiting differentiation between newly treated patients and patients who had received any treatment prior to the study period.

Prescription Refills and Medication Adherence

We defined a stimulant prescription refill as receipt of another prescription for any stimulant medication regardless of the medication type (amphetamine or methylphenidate), dose, or formulation (short- versus long-acting). We defined a patient as being adherent if the index stimulant prescription was refilled in one of the following three ways: a single index prescription was followed by a second prescription issued within 90 days; index prescriptions postdated by 2 months (i.e., two prescriptions in the system with the same prescribing date for medications of the same dose, formulation, and type) were followed by a third prescription issued between 31 and 120 days after the date of the index prescription; or index prescriptions postdated by 3 months (i.e., three prescriptions in the system with the same prescribing date for medications of the same dose, formulation, and type) were followed by a fourth prescription issued between 61 and 150 days after the date of the index prescription. If patients had more than one medication prescribed on the date of the index prescription (i.e., medications with different dosages or formulations), they had to refill only one of the medications to be considered adherent. Additionally, if patients had more than one medication prescribed on the date of the index prescription and one was postdated, we followed the rules for postdated prescriptions. Patients were excluded if the window of time to refill their prescriptions extended beyond the endpoint of our study.

Variable Derivation for Correlates and Prediction Analysis

We used patients’ zip codes to identify the median incomes for the towns in which they lived (52). We split the median incomes into terciles to classify patients as lower (≤$61,391), middle (>$61,391 to <$89,271), or upper (≥$89,271) class. We categorized index prescriptions as prescribed from a psychiatric or nonpsychiatric clinic on the basis of the clinic reported in the EMR. If patients had more than one medication prescribed on the date of the index prescription and at least one was from a nonpsychiatric clinic, we categorized that prescription as having a nonpsychiatric source .

Statistical Methods

Of the available pool of 2,206 participants, a minority (N=741, 34%) had unspecified data for at least one of the following demographic or treatment characteristics: race (N=81, 4% missing), clinic source (N=678, 31% missing), primary language (N=20, <1% missing), and economic status (N=2, <1% missing). Therefore, we used multivariate imputation with chained equations to impute the missing data for these independent variables. Using Stata’s “mi impute” command with 20 imputations, we implemented logistic regression imputation methods for race, clinic source, and primary language (binary variables) and ordered logistic regression imputation methods for economic status (ordinal variable). In addition to the imputed variables, we included the following predictors in our imputation models: sex, age, medication formulation (long- versus short-acting), medication type (methylphenidate versus amphetamine), and medication adherence.

We first compared rates of medication adherence between patients with different demographic (e.g., boys versus girls) and treatment (e.g., short- versus long-acting formulation) characteristics by using logistic regression models. We then used a multivariable logistic regression model to predict medication adherence from the demographic characteristics that were associated with adherence at the level of p≤0.10 in the bivariate analyses and all of the treatment characteristics. We generated an area-under-the-curve (AUC) statistic for the multivariable model to examine how well these characteristics predicted medication adherence. An AUC of 0.5 means the combined characteristics do not exceed chance in predicting adherence, and an AUC of 1.0 means they predict adherence perfectly.

To assess the robustness of the findings in our primary analysis, we performed five sensitivity analyses: the first excluded patients who had an unspecified prescribing clinic; the second excluded patients with an unclear ADHD diagnosis; the third excluded patients who had index prescriptions prior to January 1, 2016, thus ensuring 1 year of lead-in time without stimulant medication for all included patients; the fourth used a 45-day window to define medication adherence; and the fifth used a 37-day window to define medication adherence.

We also analyzed the data using a discovery and replication set approach, which allowed us to build a model to predict medication adherence in one data set and evaluate how well the model worked in a separate, unbiased data set. To do this, we randomly selected 75% of the patients for the discovery set and 25% of the patients for the replication set. Using the same approach as stated above, we built a multivariable logistic regression model and generated an AUC statistic using the discovery set. We then applied the multivariable logistic regression model developed in the discovery set to the replication set to see how well it predicted adherence in the replication set. This approach is used routinely in predictive analytics to protect against drawing conclusions from false-positive results.

For all analyses, standard errors were adjusted by using Stata’s “mi estimate” command to account for the variability between imputations. All tests were two-tailed and performed at the 0.05 alpha level in Stata (version 15.1).

Results

Sample

We identified 2,206 children and adolescents with prescriptions for stimulant medication; of these, a putative diagnosis of ADHD could be confirmed in 1,355 (61%). Of the 2,206 patients, 95% had single index prescriptions, 4.9% had prescriptions that were postdated by 2 months, and 0.5% had prescriptions postdated by 3 months.

Medication Adherence

Only 46% (N=1,023) of patients met our definition of medication adherence, indicating that they refilled their stimulant prescriptions quickly enough to be considered consistently medicated.

Sociodemographic Correlates of Medication Adherence

As shown in Table 1, there were small but statistically significant differences in the rates of medication adherence by age, sex, racial-ethnic group, and economic status. Patients who were older, female, Caucasian, or middle or upper class were less likely to adhere to the medication regimen. Patients who were taking methylphenidate instead of amphetamines and those who received prescriptions from psychiatric clinics rather than nonpsychiatric clinics had a small but significantly greater likelihood of adherence to treatment (Table 1). There were no other meaningful differences.

TABLE 1. Rates of stimulant medication adherence among 2,206 children and adolescents, by demographic and treatment characteristic

Nonadherent to treatment (N=1,183)Adherent to treatment (N=1,023)
CharacteristicTotal NN%aN%ap
Age (M±SD)12.5±3.012.1±3.0.003
Age group.04
 Child (age <12) 8974575144049
 Adolescent (age ≥12) 1,3097265558345
Sex.009
 Male 1,6248445278048
 Female 5823395824342
Racial-ethnic groupb.01
 Caucasian 1,5688695569945
 Not Caucasian 6383144932451
Primary language.51
 English 2,0151,0855493046
 Not English 19198519349
Economic class.03
 Lower 7393675037250
 Middle7304015532945
 Upper7374155632244
Medication formulation.29
 Long-acting 1,6398685377147
 Short-acting 5673155625244
Medication type.02
 Methylphenidate 1,5738195275448
 Amphetamine 6333645826942
Prescription source.001
 Psychiatric clinic 9264494847752
 Nonpsychiatric clinic 1,2807345754643

aRow percentages. Denominator is total N for each characteristic.

bRacial-ethnic group combined into Caucasian versus non-Caucasian for analysis. Breakdown was as follows: Caucasian, N=1,511 (68.5%); black, N=138 (6.3%); Asian American, N=50 (2.3%); Hispanic, N=352 (15.8%); Native American: N=5 (.3%); other: N=69 (3.1%), not reported, N=81 (3.6%).

TABLE 1. Rates of stimulant medication adherence among 2,206 children and adolescents, by demographic and treatment characteristic

Enlarge table

Predictors of Medication Adherence

We used a multivariable logistic regression model to predict medication adherence from all demographic characteristics that had p≤0.10 in the bivariate analyses and all treatment characteristics (Table 2). After the analyses controlled for all other variables, the characteristics that remained significantly associated with adherence to treatment were age, sex, and prescription source. Older patients were significantly less likely to adhere to treatment, while male patients and patients who got their prescriptions from a psychiatry clinic were significantly likelier to adhere to treatment. Overall, however, this model yielded an AUC statistic of 0.57, indicating that these demographic and treatment characteristics were only modestly better than chance at predicting medication adherence.

TABLE 2. Predictors of adherence to stimulant medication among 2,206 children and adolescents

CharacteristicOR95% CIp
Age (continuous).97.94–.99.03
Male (reference: female)1.231.01–1.49.04
Caucasian (reference: non-Caucasian).83.67–1.03.09
Economic class.07
 Middle (reference: lower).82.65–1.02
 Upper (reference: lower).77.61–.97
 Upper (reference: middle).95.77–1.17
Long-acting formulation (reference: short-acting formulation)1.13.93–1.38.21
Methylphenidate (reference: amphetamine)1.17.97–1.42.10
Prescription from psychiatric clinic (reference: nonpsychiatric clinic)1.541.24–1.91<.001

TABLE 2. Predictors of adherence to stimulant medication among 2,206 children and adolescents

Enlarge table

Discovery and Replication Set Analysis

We randomly assigned 1,662 patients (75%) to the discovery set and 544 patients (25%) to the replication set. Medication adherence rates in the discovery and replication sets were 46% and 48%, respectively. The multivariable logistic regression model predicting medication adherence from age, sex, race, medication formulation, medication type, and prescribing clinic yielded an AUC statistic of 0.59 in the discovery set and 0.58 in the replication set (see the online supplement).

Sensitivity Analysis Results

To assess the robustness of the results in our primary analysis, we reran our analyses after excluding 678 patients with unspecified prescribing clinics. Medication adherence remained low (53%) and our model predicting adherence from age, sex, medication formulation, medication type, and prescribing clinic yielded a low AUC statistic of 0.57. As a second sensitivity analysis, we reran our analyses after excluding 851 patients with no recorded psychiatric diagnoses in the EMR. Adherence remained low (59%), and our model predicting medication adherence from age, economic status, medication formulation, medication type, and prescribing clinic yielded a low AUC statistic of 0.56. In a third sensitivity analysis, we excluded 1,035 patients with index prescriptions prescribed prior to January 1, 2016, to help mitigate the problem of being unable to differentiate between newly treated patients and patients who had received treatment before the study period. This exclusion ensured that all patients included in the analysis had at least one year of lead-in time without stimulant prescriptions. Again, adherence to medication remained low (59%), and our model predicting adherence from age, sex, race, primary language, economic status, medication formulation, medication type, and prescribing clinic yielded a low AUC statistic of 0.60.

In the fourth sensitivity analysis, we narrowed the prescription refill window from 90 days to between 7 and 45 days of the index prescription, making an additional assumption that prescriptions refilled within the first 7 days were not actually refills, but likely reprints of lost prescriptions. We decided to give a 15-day window beyond the date the medication ran out to allow some margin of error for minor delays or problems with obtaining the new prescription exactly when the medication ran out. With this new definition, medication adherence dropped to 30%, and our model predicting adherence from age, race, medication formulation, medication type, and prescribing clinic yielded a low AUC statistic of 0.54.

In our last sensitivity analysis, we narrowed the prescription refill window even more, from 90 days to between 7 and 37 days of the index prescription. Adherence was the lowest with this definition (25%), and our model predicting medication adherence based on age, race, medication formulation, medication type, and prescribing clinic yielded a low AUC statistic of 0.55. As in the primary analysis, the low AUC statistics in all sensitivity analyses indicate that these demographic and treatment characteristics were only modestly better than chance at predicting medication adherence.

Discussion

Using a novel definition of medication adherence that was based on the renewal of the index prescription, a study of 2 years of EMR data from a large health care organization demonstrated that only 46% of 2,206 patients adhered to the medication regimen for treatment with stimulants. These findings provide new and generalizable estimates of low rates of medication adherence among children and adolescents with ADHD.

Strengths of this study included the use of a large EMR data set from a well-defined, representative population of patients seen at a large health care organization in a large metropolitan area representing all socioeconomic and racial-ethnic strata and the use of an objective metric of adherence based on the issuance of a prescription for a stimulant as documented in the EMR. Additionally, data were analyzed by using differing definitions of medication adherence (sensitivity analyses) as well as through a discovery and replication set analysis (see the online supplement), with all analyses leading to similar results showing poor rates of medication adherence.

We found some small but statistically significant demographic and clinical differences between patients who did and did not adhere to stimulant treatment. Children tended to adhere to the medication regimen somewhat more than adolescents (perhaps indicating effects of parental surveillance), adherence was slightly better in boys than in girls, and adherence was somewhat better for patients receiving prescriptions from psychiatric versus nonpsychiatric clinics, which may reflect either the severity of these patient populations or a greater ability of psychiatrists to motivate adherence. The findings show that low adherence to stimulant treatment in ADHD affects all ages, both sexes, and all economic class strata.

Our finding that rates of medication adherence were slightly higher among patients who received prescriptions from psychiatric clinics is novel. This finding may reflect better familiarity with ADHD within psychiatry that may help patients become more adherent to stimulant treatment. These findings suggest that efforts to improve medication adherence for ADHD may be most needed in the primary care setting.

While uncertainty remains as to why medication adherence is low, it is possible that low adherence is driven by the unique complexity of renewing prescriptions for stimulants, which are schedule II medicines; poor tolerability to stimulants, such as lack of appetite and difficulty sleeping; as well as ambivalence of parents about using medications to treat their children. Low adherence may also stem from misinformation or biases about stimulants in the media. Some patients may take stimulant medications on schooldays only; thus, failure to refill a prescription on time may not reflect nonadherence. It is also possible that the diagnosis of ADHD may have been inaccurate, thereby the index prescription may have been inappropriate. More research is needed to clarify the causes of low medication adherence in ADHD to develop appropriate measures to mitigate them (35, 36).

Our results should be seen in the context of methodological limitations. Our definition of medication adherence was based on renewal of the index prescription . Thus we do not know whether other measures of medication adherence, such as those proposed by Simmons et al. (53), would have led to different results. Additionally, no consideration was given to time of year; some pediatric patients do not take their medication during the summer. Because our sample was derived from a single health care organization in New England, the extent to which the results can be generalized to other regions or countries merits further investigation. Another limitation stems from not having access to data about co-occurring conditions, making it difficult to assess the impact of comorbid conditions on our findings. As well, in 30% of cases, the clinic source of prescriber was not noted and was imputed. However, an analysis of our findings excluding this subsample led to the same results.

Notwithstanding these caveats, these EMR-derived data relying on a novel definition of medication adherence and on an objective metric of adherence (issuance of a prescription) indicate that only 46% of 2,206 patients were adherent to stimulant treatment. These findings provide strong evidence of low rates of medication adherence among youths, underscoring the need for active efforts to better understand the problem and develop approaches to help mitigate it, especially in primary care settings.

Clinical and Research Programs in Pediatric Psychopharmacology and Adult ADHD, Massachusetts General Hospital, Boston (J. Biederman, Fried, DiSalvo, Storch, Pulli, Woodworth, I. Biederman); Department of Psychiatry, Harvard Medical School, Boston (J. Biederman, Fried, Perlis); Department of Neuroscience and Physiology, State University of New York Upstate Medical University, Syracuse, New York, and K.G. Jebsen Centre for Psychiatric Disorders, Department of Biomedicine, University of Bergen, Bergen, Norway (Faraone); Center for Quantitative Health, Massachusetts General Hospital, Boston (Perlis).
Send correspondence to Dr. Biederman ().

This work was sponsored in part by the Massachusetts General Hospital (MGH) Pediatric Psychopharmacology Council Fund. The Council Fund had no role in the design and conduct of the study; collection, management, analysis, and interpretation of the data; preparation, review, or approval of the manuscript; or the decision to submit the manuscript for publication.

Dr. Biederman receives research support from the American Academy of Child and Adolescent Psychiatry, Feinstein Institute for Medical Research, Headspace, Lundbeck AS, National Institute of Health Neurocentria, National Institute on Drug Abuse, Pfizer, Roche Translational and Clinical Research Center, Shire, Sunovion, and U.S. Food and Drug Administration. He has a financial interest in Avekshan LLC, a company that develops treatments for attention-deficit hyperactivity disorder (ADHD). His interests were reviewed and are managed by MGH and Partners Health Care in accordance with their conflict of interest policies. His program has received departmental royalties from a copyrighted rating scale used for ADHD diagnoses, paid by Bracket Global, Ingenix, ProPhase Labs, Shire, Sunovion, and Theravance; these royalties were paid to the Department of Psychiatry at MGH. He has served on the scientific advisory board for Akili and Shire. Through MGH corporate licensing, he has a U.S. patent (14/027,676) for a non-stimulant treatment for ADHD and a patent pending (61/233,686) on a method to prevent stimulant abuse. Dr. Fried receives research support from Shire and Roche Pharmaceuticals. Dr. Faraone has received income, potential income, travel expenses for continuing education support, and/or research support from Akili Interactive Labs, Arbor, Genomind, Ironshore, Otsuka, Shire, Sunovion, Supernus Tris, and VAYA. With his institution, he has a U.S. patent (US20130217707 A1) for the use of sodium-hydrogen exchange inhibitors in the treatment of ADHD. Dr. Perlis has served on scientific advisory boards or consulted for Genomind, Psy Therapeutics, and RID Ventures. He holds equity in Psy Therapeutics. The other authors report no financial relationships with commercial interests.

References

1 Gillberg C: The ESSENCE in child psychiatry: early symptomatic syndromes eliciting neurodevelopmental clinical examinations. Res Dev Disabil 2010; 31:1543–1551Crossref, MedlineGoogle Scholar

2 Visser SN, Danielson ML, Bitsko RH, et al.: Trends in the parent-report of health care provider-diagnosed and medicated attention-deficit/hyperactivity disorder: United States, 2003-2011. J Am Acad Child Adolesc Psychiatry 2014; 53:34–46.e2, e2Crossref, MedlineGoogle Scholar

3 Jitendra AK, Dupaul GJ, Someki F, et al.: Enhancing academic achievement for children with attention-deficit hyperactivity disorder: evidence from school-based intervention research. Dev Disabil Res Rev 2008; 14:325–330Crossref, MedlineGoogle Scholar

4 Faraone SV, Sergeant J, Gillberg C, et al.: The worldwide prevalence of ADHD: is it an American condition? World Psychiatry 2003; 2:104–113MedlineGoogle Scholar

5 Barkley RA: Major life activity and health outcomes associated with attention-deficit/hyperactivity disorder. J Clin Psychiatry 2002; 63(suppl 12):10–15MedlineGoogle Scholar

6 Loe IM, Feldman HM: Academic and educational outcomes of children with ADHD. Ambul Pediatr 2007; 7(suppl):82–90Crossref, MedlineGoogle Scholar

7 Biederman J, Faraone S, Milberger S, et al.: A prospective 4-year follow-up study of attention-deficit hyperactivity and related disorders. Arch Gen Psychiatry 1996; 53:437–446Crossref, MedlineGoogle Scholar

8 Barkley RA, Fischer M, Edelbrock CS, et al.: The adolescent outcome of hyperactive children diagnosed by research criteria: I. an 8-year prospective follow-up study. J Am Acad Child Adolesc Psychiatry 1990; 29:546–557Crossref, MedlineGoogle Scholar

9 Biederman J, Monuteaux MC, Mick E, et al.: Young adult outcome of attention deficit hyperactivity disorder: a controlled 10-year follow-up study. Psychol Med 2006; 36:167–179Crossref, MedlineGoogle Scholar

10 Biederman J, Monuteaux MC, Mick E, et al.: Is cigarette smoking a gateway to alcohol and illicit drug use disorders? A study of youths with and without attention deficit hyperactivity disorder. Biol Psychiatry 2006; 59:258–264Crossref, MedlineGoogle Scholar

11 Jerome L, Habinski L, Segal A: Attention-deficit/hyperactivity disorder (ADHD) and driving risk: a review of the literature and a methodological critique. Curr Psychiatry Rep 2006; 8:416–426Crossref, MedlineGoogle Scholar

12 Segalowitz SJ, Lawson S: Subtle symptoms associated with self-reported mild head injury. J Learn Disabil 1995; 28:309–319Crossref, MedlineGoogle Scholar

13 McKinlay A, Grace R, Horwood J, et al.: Adolescent psychiatric symptoms following preschool childhood mild traumatic brain injury: evidence from a birth cohort. J Head Trauma Rehabil 2009; 24:221–227Crossref, MedlineGoogle Scholar

14 Max JE, Lansing AE, Koele SL, et al.: Attention deficit hyperactivity disorder in children and adolescents following traumatic brain injury. Dev Neuropsychol 2004; 25:159–177Crossref, MedlineGoogle Scholar

15 Biederman J, Faraone SV, Mick E: Psychiatric and functional outcome of adolescent females with ADHD: a large, controlled, five-year longitudinal follow-up study. Presented at the annual meeting of the American Psychiatric Association, Atlanta, May 21–26, 2005Google Scholar

16 Barbaresi WJ, Colligan RC, Weaver AL, et al.: Mortality, ADHD, and psychosocial adversity in adults with childhood ADHD: a prospective study. Pediatrics 2013; 131:637–644Crossref, MedlineGoogle Scholar

17 Dalsgaard S, Leckman JF, Mortensen PB, et al.: Effect of drugs on the risk of injuries in children with attention deficit hyperactivity disorder: a prospective cohort study. Lancet Psychiatry 2015; 2:702–709Crossref, MedlineGoogle Scholar

18 Spencer T, Biederman J, Wilens T, et al.: A large, double-blind, randomized clinical trial of methylphenidate in the treatment of adults with attention-deficit/hyperactivity disorder. Biol Psychiatry 2005; 57:456–463Crossref, MedlineGoogle Scholar

19 Spencer TJ, Adler LA, McGough JJ, et al.: Efficacy and safety of dexmethylphenidate extended-release capsules in adults with attention-deficit/hyperactivity disorder. Biol Psychiatry 2007; 61:1380–1387Crossref, MedlineGoogle Scholar

20 Spencer TJ, Brown A, Seidman LJ, et al.: Effect of psychostimulants on brain structure and function in ADHD: a qualitative literature review of magnetic resonance imaging-based neuroimaging studies. J Clin Psychiatry 2013; 74:902–917Crossref, MedlineGoogle Scholar

21 Biederman J, Mick E, Surman C, et al.: A randomized, placebo-controlled trial of OROS methylphenidate in adults with attention-deficit/hyperactivity disorder. Biol Psychiatry 2006; 59:829–835Crossref, MedlineGoogle Scholar

22 Medori R, Ramos-Quiroga JA, Casas M, et al.: A randomized, placebo-controlled trial of three fixed dosages of prolonged-release OROS methylphenidate in adults with attention-deficit/hyperactivity disorder. Biol Psychiatry 2008; 63:981–989Crossref, MedlineGoogle Scholar

23 Faraone SV, Glatt SJ: A comparison of the efficacy of medications for adult attention-deficit/hyperactivity disorder using meta-analysis of effect sizes. J Clin Psychiatry 2010; 71:754–763Crossref, MedlineGoogle Scholar

24 Canadian ADHD Practice Guidelines. Toronto, Canadian ADHD Resource Alliance, 2008 http://www.caddra.caGoogle Scholar

25 Kendall T, Taylor E, Perez A, et al.: Diagnosis and management of attention-deficit/hyperactivity disorder in children, young people, and adults: summary of NICE guidance. BMJ 2008; 337:a1239Crossref, MedlineGoogle Scholar

26 Kooij SJ, Bejerot S, Blackwell A, et al.: European consensus statement on diagnosis and treatment of adult ADHD: the European Network Adult ADHD. BMC Psychiatry 2010; 10:67Crossref, MedlineGoogle Scholar

27 Faraone SV, Buitelaar J: Comparing the efficacy of stimulants for ADHD in children and adolescents using meta-analysis. Eur Child Adolesc Psychiatry 2010; 19:353–364Crossref, MedlineGoogle Scholar

28 Biederman J, Wilens T, Mick E, et al.: Pharmacotherapy of attention-deficit/hyperactivity disorder reduces risk for substance use disorder. Pediatrics 1999; 104:e20Crossref, MedlineGoogle Scholar

29 Rubia K, Alegria AA, Cubillo AI, et al.: Effects of stimulants on brain function in attention-deficit/hyperactivity disorder: a systematic review and meta-analysis. Biol Psychiatry 2014; 76:616–628Crossref, MedlineGoogle Scholar

30 Biederman J, Monuteaux MC, Spencer T, et al.: Do stimulants protect against psychiatric disorders in youth with ADHD? A 10-year follow-up study. Pediatrics 2009; 124:71–78Crossref, MedlineGoogle Scholar

31 Lichtenstein P, Halldner L, Zetterqvist J, et al.: Medication for attention deficit-hyperactivity disorder and criminality. N Engl J Med 2012; 367:2006–2014Crossref, MedlineGoogle Scholar

32 Lam LT: Attention deficit disorder and hospitalization due to injury among older adolescents in New South Wales, Australia. J Atten Disord 2002; 6:77–82Crossref, MedlineGoogle Scholar

33 Lam LT: Attention deficit disorder and hospitalization owing to intra- and interpersonal violence among children and young adolescents. J Adolesc Health 2005; 36:19–24Crossref, MedlineGoogle Scholar

34 Mikolajczyk R, Horn J, Schmedt N, et al.: Injury prevention by medication among children with attention-deficit/hyperactivity disorder: a case-only study. JAMA Pediatr 2015; 169:391–395Crossref, MedlineGoogle Scholar

35 Adler LD, Nierenberg AA: Review of medication adherence in children and adults with ADHD. Postgrad Med 2010; 122:184–191Crossref, MedlineGoogle Scholar

36 Gajria K, Lu M, Sikirica V, et al.: Adherence, persistence, and medication discontinuation in patients with attention-deficit/hyperactivity disorder—a systematic literature review. Neuropsychiatr Dis Treat 2014; 10:1543–1569MedlineGoogle Scholar

37 Coletti DJ, Pappadopulos E, Katsiotas NJ, et al.: Parent perspectives on the decision to initiate medication treatment of attention-deficit/hyperactivity disorder. J Child Adolesc Psychopharmacol 2012; 22:226–237Crossref, MedlineGoogle Scholar

38 Swanson J: Compliance with stimulants for attention-deficit/hyperactivity disorder: issues and approaches for improvement. CNS Drugs 2003; 17:117–131Crossref, MedlineGoogle Scholar

39 Perwien A, Hall J, Swensen A, et al.: Stimulant treatment patterns and compliance in children and adults with newly treated attention-deficit/hyperactivity disorder. J Manag Care Pharm 2004; 10:122–129Crossref, MedlineGoogle Scholar

40 Georgiopoulos AM, Hua LL: The diagnosis and treatment of attention deficit-hyperactivity disorder in children and adolescents with cystic fibrosis: a retrospective study. Psychosomatics 2011; 52:160–166Crossref, MedlineGoogle Scholar

41 Torres A, Whitney J, Rao S, et al.: Tolerability of atomoxetine for treatment of pediatric attention-deficit/hyperactivity disorder in the context of epilepsy. Epilepsy Behav 2011; 20:95–102Crossref, MedlineGoogle Scholar

42 Huskamp HA, Deverka PA, Epstein AM, et al.: Impact of 3-tier formularies on drug treatment of attention-deficit/hyperactivity disorder in children. Arch Gen Psychiatry 2005; 62:435–441Crossref, MedlineGoogle Scholar

43 Ji X, Druss BG, Lally C, et al.: Racial-ethnic differences in patterns of discontinuous medication treatment among Medicaid-insured youths with ADHD. Psychiatr Serv 2018; 69:322–331LinkGoogle Scholar

44 Moran A, Serban N, Danielson ML, et al.: Adherence to recommended care guidelines in the treatment of preschool-age Medicaid-enrolled children with a diagnosis of ADHD. Psychiatr Serv 2019; 70:26–34LinkGoogle Scholar

45 Joyce NR, Huskamp HA, Hadland SE, et al.: The alternative quality contract: impact on service use and spending for children with ADHD. Psychiatr Serv 2017; 68:1210–1212LinkGoogle Scholar

46 Hervey-Jumper H, Douyon K, Franco KN: Deficits in diagnosis, treatment and continuity of care in African-American children and adolescents with ADHD. J Natl Med Assoc 2006; 98:233–238MedlineGoogle Scholar

47 Setyawan J, Hodgkins P, Guérin A, et al.: Comparing treatment adherence of lisdexamfetamine and other medications for the treatment of attention deficit/hyperactivity disorder: a retrospective analysis. J Med Econ 2013; 16:962–975Crossref, MedlineGoogle Scholar

48 Grcevich S, Rowane WA, Marcellino B, et al.: Retrospective comparison of Adderall and methylphenidate in the treatment of attention deficit hyperactivity disorder. J Child Adolesc Psychopharmacol 2001; 11:35–41Crossref, MedlineGoogle Scholar

49 Kemner JE, Lage MJ: Impact of methylphenidate formulation on treatment patterns and hospitalizations: a retrospective analysis. Ann Gen Psychiatry 2006; 5:5Crossref, MedlineGoogle Scholar

50 Lage M, Hwang P: Effect of methylphenidate formulation for attention deficit hyperactivity disorder on patterns and outcomes of treatment. J Child Adolesc Psychopharmacol 2004; 14:575–581Crossref, MedlineGoogle Scholar

51 A Leadership Resource for Patient and Family Engagement Strategies. Chicago, American Hospital Association, Health Research & Educational Trust, 2013. http://www.hpoe.org/Patient-Family-engagementGoogle Scholar

52 American Community Survey, 2011–2015 American Community Survey 5-Year Estimates, Table S1903—Median Income in the Past 12 Months (In 2015 Inflation-Adjusted Dollars). Washington, DC, US Census Bureau. https://factfinder.census.gov/faces/nav/jsf/pages/community_facts.xhtmlGoogle Scholar

53 Simmons LA, Wolever RQ, Bechard EM, et al.: Patient engagement as a risk factor in personalized health care: a systematic review of the literature on chronic disease. Genome Med 2014; 6:16Crossref, MedlineGoogle Scholar