Abstract
Objective:
Evaluate efficacy and safety of varenicline, bupropion, and nicotine replacement therapy (NRT) for smoking cessation in smokers with schizophrenia spectrum disorders in post-hoc analyses of EAGLES data.
Methods:
Smokers with a schizophrenia spectrum disorder (n=390) and without a psychiatric illness (controls) (n=4,028) were randomly assigned to varenicline, bupropion, NRT, or placebo for 12 weeks. Outcomes included abstinence rates during treatment and follow-up, number needed to treat (NNT) for abstinence, incidence of neuropsychiatric adverse events, and temporal relationship between neuropsychiatric adverse events and abstinence status.
Results:
Smokers with schizophrenia smoked more, had greater dependence, and fewer prior trials of cessation pharmacotherapy. At each timepoint, on varenicline, smokers with schizophrenia had significantly greater odds of abstinence than placebo and comparable NNT with controls. Bupropion and NRT increased odds of abstinence, though confidence intervals included one for some comparisons and produced greater NNT in smokers with schizophrenia than controls. No treatment was associated with significantly more neuropsychiatric adverse events than placebo in either cohort. The estimated neuropsychiatric adverse event rate was 5% (95% CI=3.0–7.7) in smokers with schizophrenia and 1% (95% CI=.6–2.1) in controls. Over one-third of neuropsychiatric adverse events occurred during partial or full abstinence, suggesting a multifactorial nature of neuropsychiatric adverse events.
Conclusions:
In smokers with schizophrenia, varenicline was associated with significantly higher abstinence rates than NRT, bupropion, and placebo, and had NNT comparable with smokers without psychiatric disorders. A significant proportion of neuropsychiatric adverse events occurred during early abstinence. No treatment significantly increased neuropsychiatric adverse event prevalence.
BACKGROUND
People with schizophrenia spectrum disorders are more likely to smoke tobacco, smoke heavily (1), and have severe dependence than those without psychiatric illness (2–4). Smoking rates are not decreasing for those with schizophrenia as they are in the general population (5), and smoking-related disease contributes disproportionately to a comparative 29-year mortality gap in adults (6–9). Though quitting smoking by middle age reduces the risk of death associated with continued smoking by 90% (10), smokers with schizophrenia are less likely than those in the general population to be offered effective pharmacotherapeutic smoking cessation aids, particularly varenicline (1, 11, 12). Consistent reports of abstinence rates <5% in smokers with schizophrenia with behavioral smoking cessation treatment alone (13–19) suggest that this group particularly needs pharmacotherapeutic cessation aids to quit smoking.
Despite evidence for safety and efficacy of first-line pharmacotherapeutic cessation aids in this population (19, 20), clinicians report negative attitudes toward providing smoking cessation treatment for smokers with schizophrenia (21), with pharmacotherapy—particularly non-nicotine pharmacotherapy—being particularly underutilized (1, 11, 12, 22, 23). Additionally, Medicaid coverage of the most effective cessation treatments remains limited in many US states despite legislation barring state Medicaid programs from excluding US Food and Drug Administration (FDA)-approved cessation medications from coverage (24). High co-pays and prior authorization requirements remain common barriers to obtaining smoking cessation medication through Medicaid and Medicare plans that insure most people with schizophrenia spectrum disorders (25). Additionally, limits on access to effective smoking cessation treatment, through low rates of prescribing and financial barriers, place people with schizophrenia at increased risk for smoking-related disease and death.
The neuropsychiatric safety and efficacy trial of varenicline, bupropion, and nicotine replacement therapy (NRT) in smokers with and without psychiatric disorders (EAGLES) estimated the incidence of moderate to severe neuropsychiatric adverse events (NPSAEs) during a 12-week treatment period and 12-week follow-up, and assessed tobacco abstinence rates (26). Neuropsychiatric adverse event incidence and continuous abstinence rates have been reported for the schizophrenia spectrum disorders subcohort relative to the mood and anxiety disorders subcohorts (27). To address safety concerns that may drive the particular underuse of effective smoking cessation medications in smokers with schizophrenia, we undertook a post-hoc analysis of weekly patterns of neuropsychiatric adverse events and abstinence rates in the EAGLES schizophrenia spectrum disorders subcohort versus smokers without psychiatric disorders, including analysis of timing of neuropsychiatric adverse events relative to start of study medication and change in weekly 7-day point prevalence abstinence (PPA) status, and analysis of end-of-treatment abstinence rates by baseline psychiatric symptom severity rating.
METHODS
EAGLES was a multinational, multicenter, randomized, double-blind, placebo- and active (NRT)-controlled trial, conducted from November 30, 2011 to January 13, 2015. The primary report provides details of the design and primary outcomes (26). Study procedures and consent forms were approved by the institutional review boards at participating institutions. All participants signed informed consent.
Eligible participants were motivated-to-quit adults, aged 18–75 years, who smoked ≥10 cigarettes per day, with exhaled carbon monoxide (CO) >10 parts per million (ppm) at screening. Eligible smokers with schizophrenia spectrum disorders met Diagnostic and Statistical Manual of Mental Disorders, Fourth Edition, Text Revision (DSM-IV-TR) (28) diagnostic criteria for current or lifetime psychotic disorders including schizophrenia and schizoaffective disorders and were not excluded for other prespecified psychiatric comorbidities that excluded alcohol and other substance use disorders within the previous 12 months. Smokers with schizophrenia spectrum disorders made up approximately 10% of the psychiatric cohort (n=390) in EAGLES. Enrolment criteria required a score of <5 on the 7-point Clinical Global Impression–Severity (CGI-S)(29), indicating moderate severity of symptoms or less. The control cohort without psychiatric disorders (n=4,028) had no Axis I diagnosis.
Randomization, masking, and study treatment
Randomization to varenicline 1 mg twice-daily, bupropion sustained-release 150 mg twice-daily, NRT transdermal patch 21 mg per day with taper, or placebo was done in a 1:1:1:1 ratio with block size of eight for each diagnostic subcohort by region in a double-blind, triple-dummy, parallel-group design.
Participants set a target quit-date 1 week post-randomization, coinciding with the end of varenicline and bupropion up-titration and initiation of NRT. Study visits were weekly for 6 weeks, biweekly for 6 weeks, then at weeks 13, 16, 20, and 24. Ten-minute individual smoking cessation counseling was provided at each visit (30). Telephone contacts to determine smoking status were conducted weekly between visits.
Assessments
Psychiatric diagnosis was assessed at screening with the Structured Clinical Interviews for DSM-IV-TR Axis I and II Disorders (SCID-I and SCID-II) (28, 31). Severity of cigarette dependence was assessed with the Fagerström Test for Cigarette Dependence (FTCD) (32).
Abstinence was assessed weekly, defined as self-report of tobacco abstinence since the previous study assessment. Expired CO ≤10 ppm was used to validate self-reported abstinence. CO was collected at study weeks 1–6, 8, 10, 12, 16, and 24. Participants who discontinued the study or were lost to follow-up or had missing CO data at weeks 12 or 24 were considered non-abstinent.
A neuropsychiatric adverse event was an adverse event in one of 16 neuropsychiatric symptom categories volunteered, observed, or solicited during treatment or 30-day follow-up that was new or increased in severity from baseline, irrespective of whether the adverse event was considered causally related to study medication, that met a priori severity criteria. Neuropsychiatric adverse events expected to be more common (anxiety, depression, feeling abnormal, or hostility) that were rated as severe, and those in agitation, aggression, delusions, hallucinations, homicidal ideation, mania, panic, paranoia, psychosis, suicidal ideation, behavior, or suicide categories rated as moderate or severe met criteria as a neuropsychiatric adverse event.
Neuropsychiatric adverse events were assessed at each study visit with open-ended questions, direct observation, and the semi-structured Neuropsychiatric Adverse Event Interview (26, 33). Positive responses on the Neuropsychiatric Adverse Event Interview were evaluated for frequency, duration, and severity to determine if they qualified. Investigators evaluated whether positive responses on the Columbia Suicide Severity Rating Scale (C-SSRS) or proxy reports from family members or others were NPSAEs. Psychiatric symptoms were assessed at each study visit with the Hospital Anxiety and Depression Scale (34) and C-SSRS (35). Tobacco and nicotine use were assessed with a structured questionnaire and expired CO measurement.
Analysis
Generalized linear models (GLMs) were conducted to test the effect of treatment (varenicline, bupropion, NRT, placebo), and cohort (schizophrenia spectrum disorders, no psychiatric disorders) on 7-day PPA and continuous abstinence rates during the treatment and follow-up periods. Observed rates of NPSAEs were reported and GLMs were conducted to test effects of treatment, cohort, and their interaction on the NPSAE primary endpoint for 16 weeks following treatment initiation (12 weeks treatment and 4 weeks follow-up). All models included region and age, race, body mass index (BMI), smoking characteristics, and past cessation medication use if associated with outcome (36). For each reported NPSAE the week of the event was plotted together with change in weekly 7-day PPA status: smoker, partial abstainer, abstainer, by treatment assignment and cohort.
RESULTS
Participants
The efficacy cohort consisted of all 390 and 4,028 participants enrolled in the schizophrenia and control cohorts. The safety cohort consisted of 386 (99%) and 3,984 (99%) smokers in the schizophrenia and control cohorts who received ≥1 dose of study medication. The 12-week treatment and 12-week follow-up phases, respectively, were completed by 350 (90%) and 336 (86%) smokers with schizophrenia and 3,404 (85%) and 3,124 (78%) smokers without psychiatric disorders (CONSORT diagram available online).
In the schizophrenia spectrum disorders subcohort, 303 (78%) met DSM-IV-TR criteria for schizophrenia, and 87 (22%) for schizoaffective disorder, while 142 (36%) met SCID criteria for comorbid Axis I disorder, including 105 (27%) with a prior alcohol or substance abuse disorder. Those in the schizophrenia subcohort were more likely to report suicidal ideation or behavior in their lifetime (32% vs. 5% in those without psychiatric disorders), to report greater symptoms of anxiety, depression, and aggression at baseline, and to be treated with psychotropic medications (Table 1).
Table 1.
Baseline characteristics by treatment group and cohort
Schizophrenia spectrum disorders subcohort | No psychiatric disorders cohort | |||||||||
---|---|---|---|---|---|---|---|---|---|---|
|
||||||||||
All n=390 | Varenicline n=95 | Bupropion n=98 | NRT n=99 | Placebo n=98 | All n=4,028 | Varenicline n=1,005 | Bupropion n=1,001 | NRT n=1,013 | Placebo n=1,009 | |
|
||||||||||
% | % | % | % | % | % | % | % | % | % | |
| ||||||||||
Demographic characteristics | ||||||||||
Female sex* | 36 | 36 | 34 | 38 | 36 | 50 | 49 | 49 | 50 | 51 |
Age, years (M±SD) | 44.5±10.6 | 44.6±11.6 | 44.5±10.7 | 43.3±10.2 | 45.4±9.8 | 45.9±12.9 | 45.8±12.9 | 46.0±13.0 | 46.1±12.8 | 45.9±12.8 |
Race* | ||||||||||
White | 64 | 62 | 61 | 64 | 68 | 83 | 82 | 83 | 83 | 82 |
Black | 32 | 35 | 37 | 31 | 27 | 13 | 14 | 12 | 13 | 13 |
Other | 4 | 3 | 2 | 5 | 5 | 5 | 4 | 5 | 4 | 6 |
BMI*, kg/m2 (M±SD) | 29.8±6.9 | 30.6±7.5 | 29.6±7.0 | 29.3±6.6 | 29.5±6.6 | 27.6±6.1 | 27.4±6.0 | 27.6±6.2 | 27.8±6.3 | 27.8±6.0 |
Smoking characteristics | ||||||||||
FTCD score*,a (M±SD) | 6.9±1.8 | 6.8±1.9 | 6.9±1.7 | 6.8±2.0 | 7.0±1.7 | 5.5±2.0 | 5.5±2.0 | 5.5±2.0 | 5.6±2.0 | 5.5±2.0 |
Cigarettes smoked per day in past month*, n (M±SD) | 23.2±10.7 | 22.5±9.4 | 22.2±7.4 | 23.9±14.9 | 24.3±9.5 | 20.7±8.0 | 20.7±8.3 | 20.7±7.9 | 20.8±8.2 | 20.5±7.9 |
Previous quit attempts, n (M±SD) | 2.4±4.0 | 2.2±3.2 | 1.9±2.0 | 2.2±2.8 | 3.3±6.4 | 3.2±9.7 | 3.2±13.7 | 3.3±10.2 | 3.2±5.1 | 3.1±7.4 |
Prior cessation aid trials | ||||||||||
Varenicline* | 8 | 9 | 8 | 8 | 6 | 14 | 13 | 15 | 16 | 14 |
Bupropion*,b | 4 | 3 | 6 | 2 | 3 | 9 | 9 | 9 | 9 | 9 |
NRT* | 15 | 18 | 13 | 12 | 17 | 25 | 22 | 26 | 25 | 25 |
Psychiatric characteristics | ||||||||||
Past alcohol use disorder | 13 | 15 | 11 | 9 | 16 | <1 | <1 | <1 | 0 | <1 |
Past substance abuse disorderc | 23 | 25 | 21 | 18 | 28 | <1 | <1 | <1 | 0 | <1 |
Suicidal ideation*,d | 29 | 26 | 29 | 31 | 28 | 5 | 5 | 4 | 5 | 5 |
Suicidal behavior*,d | 18 | 16 | 20 | 22 | 15 | <1 | <1 | <1 | <1 | <1 |
HADS scoree | ||||||||||
Anxiety subscale score* (M±SD) | 5.1±4.1 | 4.6±4.0 | 5.4±4.2 | 5.5±4.5 | 5.0±3.8 | 2.8±2.7 | 2.8±2.8 | 2.7±2.7 | 2.7±2.6 | 2.9±2.8 |
Depression subscale score* (M±SD) | 3.9±3.2 | 4.0±3.6 | 4.2±2.8 | 3.8±3.3 | 3.8±3.0 | 1.5±2.1 | 1.5±2.1 | 1.4±2.0 | 1.5±2.0 | 1.6±2.1 |
BPAQ score*, f (M±SD) | 62.7±20.1 | 61.7±19.8 | 63.4±20.3 | 63.8±20.7 | 61.6±19.7 | 52.2±15.4 | 52.4±15.5 | 51.9±15.3 | 52.2±15.6 | 52.3±15.4 |
Psychotropic medication * | ||||||||||
Antipsychotics | 94 | 97 | 93 | 93 | 95 | <1 | <1 | <1 | <1 | <1 |
First-generation | 24 | 23 | 30 | 18 | 25 | <1 | <1 | 0 | <1 | <1 |
Second-generation | 88 | 93 | 80 | 91 | 89 | <1 | <1 | <1 | 0 | <1 |
Clozapine | 8 | 5 | 10 | 10 | 6 | <1 | 0 | 0 | 0 | <1 |
Antidepressants | 31 | 32 | 28 | 36 | 25 | 3 | 2 | 2 | 3 | 4 |
Anxiolytics, hypnotics | 17 | 19 | 17 | 21 | 11 | 6 | 5 | 5 | 6 | 6 |
Mood stabilizers | 4 | 4 | 3 | 6 | 2 | <1 | <1 | <1 | <1 | 1 |
BMI, body mass index; BPAQ, Buss–Perry Aggression Questionnaire; C-SSRS, Columbia Suicide Severity Rating Scale; FTCD, Fagerström Test for Cigarette Dependence; HADS, Hospital Anxiety and Depression Scale; NRT, nicotine replacement therapy (transdermal nicotine patch); SD, standard deviation.
Possible FTCD scores range from 0–10, with higher scores indicating more intense physical dependence on nicotine.
Bupropion prior use for smoking cessation or other indications.
Substance use disorder other than alcohol, caffeine or nicotine.
Possible HADS scores range from 0–21 for either anxiety or depression, with higher scores indicating higher levels of either anxiety or depression.
Lifetime history of suicidal ideation or behavior from C-SSRS. Possible C-SSRS scores range from 2–25, with a higher number indicating more intense ideation and greater risk.
Possible BPAQ scores range from 29–145, with higher scores indicating higher levels of aggression.
p<.001 for comparison of baseline variable by cohort.
Smokers with schizophrenia smoked more cigarettes per day (23.2 vs. 20.7) and had greater severity of cigarette dependence (FTCD total score: 6.9 vs. 5.5) than controls. Smokers with schizophrenia were not significantly less likely to have made a smoking cessation attempt (72% in the schizophrenia subcohort reported a mean 2.4 prior serious quit attempts and 82% of controls reported a mean of 3.2 quit attempts). However, they were significantly less likely than controls to report prior trials of smoking cessation treatment: varenicline (8% vs. 14%), bupropion (4% vs. 9%), and NRT (15% vs. 25%) (Table 1).
Tobacco-smoking abstinence
Seven-day PPA rates at end-of-treatment and follow-up
Weekly observed 7-day PPA rates by treatment are shown in Figure 1a. Odds of 7-day end-of-treatment PPA in smokers with schizophrenia were 6-fold higher with varenicline than placebo and >2-fold higher with varenicline than bupropion or NRT. Odds of 7-day end-of-treatment PPA in smokers with schizophrenia were >2-fold higher with bupropion and NRT than placebo (treatment comparison figure online). NNT in smokers with schizophrenia and controls for 7-day end-of-treatment PPA was: varenicline, five and four; bupropion, 17 and eight; NRT, 12 and eight (Figure 1b).
Figure 1. a) Observed 7-day point prevalence abstinence rates at each study visit, and b) number needed to treat for 7-day point prevalence abstinence at end of treatment and end of follow-up.
NRT, nicotine replacement therapy (transdermal nicotine patch); PPA, point prevalence abstinence.
At week 24, odds of 7-day PPA for active treatments vs. placebo in smokers with schizophrenia ranged from 2.8 for NRT to 5.2 for varenicline. In controls, odds ratios for 7-day PPA for all active treatments were superior to placebo and were superior for varenicline to bupropion and NRT (treatment comparison figure online). The relative efficacy of active treatments was similar across cohorts, and abstinence rates were higher in those without psychiatric disorders (Figure 1a). NNT in smokers with schizophrenia and controls for 7-day PPA at week 24 was: varenicline, seven and seven; bupropion, 13 and 12; NRT, 13 and 11 (Figure 1b).
Figures presenting weekly observed and estimated continuous abstinence rates for weeks 9–12 in a model including treatment, cohort, region, treatment-by-cohort, FTCD, cigarettes/day in the past month, race, age, years smoked, and BMI are available in the online supplement. NNT in smokers with schizophrenia and controls for continuous abstinence, weeks 9–12, was: varenicline, six and five; bupropion, 14 and nine; NRT, 12 and eight.
A table presenting 7-day end-of-treatment PPA rates by baseline CGI-S symptom severity category is online. While abstinence rate was highest among those few with the lowest possible baseline symptom severity, there was no progression to lower abstinence rates with increasing baseline psychiatric symptom burden.
Neuropsychiatric safety during 12 weeks’ treatment and 4 weeks’ follow-up
Neuropsychiatric adverse event rates were not significantly higher with any active treatment than placebo overall or in either cohort, and there were no significant treatment by diagnosis interactions (Figure 2); the point estimates are negative in smokers with schizophrenia for varenicline and NRT. There were significant effects for diagnostic cohort, region, and race. Smokers with schizophrenia were more likely than controls to experience a neuropsychiatric adverse event. Observed neuropsychiatric adverse event rates in smokers with schizophrenia were: varenicline, 6%; bupropion, 6%; NRT, 5%; placebo, 6% (table available online).
Figure 2. Risk differences for neuropsychiatric adverse events with varenicline, bupropion, NRT, and placebo.
Period for ascertainment of neuropsychiatric adverse events was during 12 weeks’ treatment and ≤30 days after last dose. Model terms included: treatment group (varenicline, bupropion, NRT, placebo), cohort (schizophrenia spectrum disorders no psychiatric disorders), treatment by cohort interaction, region (US or non-US), and race (White, Black, Other).
CI, confidence interval; NRT, nicotine replacement therapy (transdermal nicotine patch); RD, risk difference.
There were no observed effects of treatment on suicidal ideation or behavior, serious adverse events, or adverse events resulting in permanent discontinuations. In smokers with schizophrenia, NPSAEs in the primary endpoint that were severe, constituted a serious adverse event, or led to treatment discontinuation or an intervention occurred in 2% (varenicline), 1% (bupropion), 2% (NRT), and 2% (placebo). Serious adverse events were observed in <1% in both cohorts (table available online). In smokers with schizophrenia, 22% and in controls, 30% reported an adverse event in the Medical Dictionary for Regulatory Activities psychiatric disorders category of mild, moderate, or severe intensity.
In an exploratory analysis of the temporal relationship between neuropsychiatric adverse event onset with 7-day PPA status, by treatment assignment and cohort, 8/23 (35%) NPSAEs in smokers with schizophrenia and 40/84 (48%) in controls were reported at a study visit when the participant had been partially or fully abstinent in the prior week (figure available online).
DISCUSSION
This analysis provides robust evidence for efficacy of first-line FDA-approved smoking cessation medications—particularly varenicline, in smokers with schizophrenia spectrum disorders and those without psychiatric disorders, where the NNT to obtain end-of-treatment abstinence was 5 and 4—with no clear relationship between baseline psychiatric symptom burden and attainment of abstinence, or between neuropsychiatric adverse events and treatment in either cohort. In both cohorts, odds of 7-day end-of-treatment PPA were higher with varenicline than bupropion, NRT, and placebo, and higher with NRT than placebo. Prior trials have shown efficacy of varenicline (37), bupropion (14, 15), and bupropion combined with NRT for smoking cessation in smokers with schizophrenia (16, 17). EAGLES is the first trial to report efficacy of NRT vs. placebo for smoking cessation in smokers with schizophrenia.
Considering the greater severity of nicotine dependence and psychiatric symptom burden in smokers with schizophrenia, these participants would be expected to be less likely to quit smoking than smokers without an Axis I psychiatric illness. Interestingly, we report no clear relationship in smokers with schizophrenia between abstinence rates and psychiatric symptom severity, and essentially equivalent NNT for varenicline in smokers with schizophrenia and those without psychiatric disorders (5 vs. 4). While the confidence intervals overlapped, point estimates for odds of abstinence with varenicline were nearly twice as high in smokers with schizophrenia versus controls without psychiatric disorders, likely due to very low abstinence rates for smokers with schizophrenia with placebo, which is consistent with prior reports of 4–5% abstinence rates with behavioral treatment alone (14–16, 19, 37). Such low success rates indicate that it is critical for smokers with schizophrenia to have access to pharmacotherapeutic cessation aids if they are to be successful in their efforts to quit smoking. Varenicline is underutilized in smokers with psychotic illness (11, 12), and in this trial, smokers with schizophrenia reported significantly fewer prior treatment trials with varenicline, bupropion, or NRT—without significantly fewer prior cessation attempts—than smokers without psychiatric disorders.
In light of greater psychiatric symptom burden, higher ratings of anxiety, depression, aggression and nicotine dependence severity, and more prior suicidal ideation/behavior, it is not surprising that those in the schizophrenia subcohort were more likely than controls to experience a neuropsychiatric adverse event during the treatment and follow-up periods. The baseline neuropsychiatric adverse event rate for smokers with schizophrenia outside the context of a smoking cessation attempt is unknown. In smokers with schizophrenia, neuropsychiatric adverse events appear to be multifactorial and sporadic as they did not cluster within any particular symptom domain or shortly after study medication initiation or the quit date, occurred independent of treatment assignment, rarely led to permanent discontinuation of smoking cessation medication, and approximately one-third were reported during study weeks with partial or complete tobacco abstinence. While serious adverse events were observed in <0.5% in the schizophrenia subcohort and 0.2% in the cohort without psychiatric disorders, this is worth weighing against the well-established metric that half of smokers who do not quit will die prematurely from a smoking-related illness.
It is now considered standard of care to offer effective smoking cessation pharmacotherapy to all smokers at every clinical visit (38), even in smokers who report they may not be ready to make a cessation attempt. In considering the risk–benefit ratio of providing smoking cessation treatment to smokers with schizophrenia, several points are worth considering. It is increasingly recognized that smoking cessation itself does not significantly exacerbate the symptoms or course of mental illness (39, 40), and that active treatments that significantly improve abstinence rates in smokers with schizophrenia do not exacerbate psychiatric symptoms (19, 41). In EAGLES, smokers with schizophrenia treated with placebo and behavioral support were as likely as those treated with varenicline, bupropion, or NRT to experience a moderate to severe neuropsychiatric adverse event, but were far less likely to attain abstinence. The life expectancy for people with schizophrenia is approximately 29 years shorter than those without psychiatric illness, and tobacco smoking is the single largest cause of this disparity in life expectancy (8, 42, 43), while smoking cessation effectively mitigates this risk (10, 44, 45). Because tobacco smoking is associated with increased hepatic clearance of many psychotropic drugs, particularly those metabolized by cytochrome P450 1A2 and 2E1, it is recommended that clinicians monitor patients who reduce or quit smoking for evidence of reduced clearance of psychotropic medications metabolized by these enzymes and consider dose adjustment accordingly (46–48).
Limitations
Though 27% of smokers with schizophrenia had a prior alcohol or substance use disorder, smokers with an active substance use disorder other than nicotine were excluded, so results cannot be expected to generalize to those with active substance use. Likewise, though participants were symptomatic at baseline, enrolment criteria required that psychiatric symptoms be stable, and 95% of smokers with schizophrenia were taking psychotropic medications, thus results cannot be expected to generalize to unstable or untreated smokers with schizophrenia. While increasingly considered standard clinical care, dual NRT (NRT patch plus NRT gum, lozenge, nasal spray, or inhaler) was not tested. Future research is needed to test the effects of dual NRT in smokers with schizophrenia and to compare efficacy and tolerability of dual NRT with varenicline and bupropion. The behavioral component of the intervention was brief; trials of pharmacotherapy plus more intensive behavioral treatment have shown higher abstinence rates in smokers with schizophrenia (49). Further research is needed to determine whether more intensive behavioral treatment improves efficacy of pharmacotherapy for nicotine dependence in smokers with schizophrenia. We report 24 weeks’ efficacy data and 16 weeks’ safety data, though clinicians and smokers will be interested in longer-term outcomes (49, 50).
CONCLUSION
Tobacco smokers, particularly smokers with schizophrenia, need help to quit. We provide robust evidence for efficacy and tolerability for smoking cessation treatments, particularly varenicline. These data, together with strong evidence that smoking cessation does not exacerbate mental illness (19, 39), strong consistent evidence for low abstinence rates in smokers with schizophrenia with behavioral treatment alone (19, 22), and benefit of smoking cessation on premature mortality (10, 44) should spur lowering of barriers, at the policy and practitioner level, to greater utilization of the most effective pharmacotherapeutic cessation aids for those with smokers with schizophrenia as standard of care.
Supplementary Material
Highlights.
Smokers with schizophrenia spectrum disorders smoked more cigarettes per day, had greater severity of cigarette dependence, and had fewer prior trials of smoking cessation pharmacotherapy than smokers without Axis I psychiatric disorders though they had not made significantly fewer quit attempts.
In smokers with schizophrenia spectrum disorders, smoking cessation pharmacotherapy with varenicline was associated with higher abstinence rates than with NRT or bupropion.
The number needed to treat with varenicline was comparable in smokers with schizophrenia spectrum disorders and those without a psychiatric disorder.
None of the active smoking cessation pharmacotherapies significantly increased the prevalence of neuropsychiatric adverse events in either smokers with schizophrenia spectrum disorders or those without a psychiatric disorder.
Acknowledgments:
Editorial support was provided by Anne Jakobsen, M.Sc., of Engage Scientific Solutions, Horsham, UK, and was funded by Pfizer.
Funding sources: This study was funded by Pfizer and GlaxoSmithKline.
Disclosures: Dr. Evins reports advisory board service for Alkermes, Scientific Consultant service for Charles River Analytics (NIDA SBIR), Data Safety Board service for Karuna Pharmaceuticals (Data Safety Monitoring Board), and research grants to her institution from Brain Solutions, LLC (NIDA SBIR). Dr. Evins’s contribution to this manuscript was supported by a National Institute on Drug Abuse Career Award in Patient-Oriented Addiction Research, K24 DA030443. Dr. West is a consultant to Pfizer, Johnson & Johnson, and GlaxoSmithKline, and has received research funding from Pfizer and Johnson & Johnson. Dr. West’s salary is funded by Cancer Research UK. Dr. Benowitz is a consultant to Pfizer and Achieve Life Sciences, and has been a paid expert witness in litigation against tobacco companies. Drs. Maravic and Heffner report no conflicts of interest. Dr. Anthenelli reports receiving grant support in the form of drug supply from Pfizer and providing consulting and/or advisory board services to Astraea Therapeutics and Pfizer. Dr. Anthenelli’s contribution to this manuscript was supported, in part, by National Institute on Alcohol Abuse and Alcoholism Grants # U10 AA008401 and # R44 AA024643, National Institute on Drug Abuse Grants # U01 DA041731, and UCOP TRDRP # T29IPO379. Dr. Russ, Dr. Lawrence, and Dr. McRae are employees and stockholders of Pfizer. The opinions expressed in this article are those of the authors and do not necessarily reflect the views of their employers.
Footnotes
Data-sharing statement
Upon request, and subject to certain criteria, conditions, and exceptions (see https://www.pfizer.com/science/clinical-trials/trial-data-and-results for more information), Pfizer will provide access to individual de-identified participant data from Pfizer-sponsored global interventional clinical studies conducted for medicines, vaccines, and medical devices: 1) for indications that have been approved in the USA and/or EU; or 2) in programs that have been terminated (i.e., development for all indications has been discontinued). Pfizer will also consider requests for the protocol, data dictionary, and statistical analysis plan. Data may be requested from Pfizer trials 24 months after study completion. The de-identified participant data will be made available to researchers whose proposals meet the research criteria and other conditions, and for which an exception does not apply, via a secure portal. To gain access, data requestors must enter into a data access agreement with Pfizer.
Previous presentation: Parts of this manuscript were presented at a symposium at the 24th Annual Meeting of the Society for Research on Nicotine and Tobacco, February 21–24, 2018, Baltimore, MD, USA.
Trial Registration: ClinicalTrials.gov Identifier: NCT01456936
REFERENCES
- 1.Harker K, Cheeseman H: The Stolen Years: The Mental Health and Smoking Action Report 2016. London, United Kingdom: Action on Smoking and Health, 2016 [Google Scholar]
- 2.de Leon J, Diaz FJ: A meta-analysis of worldwide studies demonstrates an association between schizophrenia and tobacco smoking behaviors. Schizophr Res 76:135–57, 2005 [DOI] [PubMed] [Google Scholar]
- 3.Tidey JW, Rohsenow DJ, Kaplan GB, et al. : Cigarette smoking topography in smokers with schizophrenia and matched non-psychiatric controls. Drug Alcohol Depend 80:259–65, 2005 [DOI] [PubMed] [Google Scholar]
- 4.Williams JM, Gandhi KK, Lu SE, et al. : Higher nicotine levels in schizophrenia compared with controls after smoking a single cigarette. Nicotine Tob Res 12:855–9, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Cook BL, Wayne GF, Kafali EN, et al. : Trends in smoking among adults with mental illness and association between mental health treatment and smoking cessation. JAMA 311:172–82, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Hennekens CH: Increasing global burden of cardiovascular disease in general populations and patients with schizophrenia. J Clin Psychiatry 68 Suppl 4:4–7, 2007 [PubMed] [Google Scholar]
- 7.Saha S, Chant D, McGrath J: A systematic review of mortality in schizophrenia: is the differential mortality gap worsening over time? Arch Gen Psychiatry 64:1123–31, 2007 [DOI] [PubMed] [Google Scholar]
- 8.Olfson M, Gerhard T, Huang C, et al. : Premature mortality among adults with schizophrenia in the United States. JAMA Psychiatry 72:1172–81, 2015 [DOI] [PubMed] [Google Scholar]
- 9.Goff DC, Sullivan LM, McEvoy JP, et al. : A comparison of ten-year cardiac risk estimates in schizophrenia patients from the CATIE study and matched controls. Schizophr Res 80:45–53, 2005 [DOI] [PubMed] [Google Scholar]
- 10.Jha P, Ramasundarahettige C, Landsman V, et al. : 21st-century hazards of smoking and benefits of cessation in the United States. N Engl J Med 368:341–50, 2013 [DOI] [PubMed] [Google Scholar]
- 11.Huang Y, Lewis S, Britton J: Use of varenicline for smoking cessation treatment in UK primary care: an association rule mining analysis. BMC Public Health 14:1024, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Taylor GMJ, Itani T, Thomas KH, et al. : Prescribing prevalence, effectiveness, and mental health safety of smoking cessation medicines in patients with mental disorders. Nicotine Tob Res, 2019 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Weiner E, Ball MP, Summerfelt A, et al. : Effects of sustained-release bupropion and supportive group therapy on cigarette consumption in patients with schizophrenia. The American journal of psychiatry 158:635–7, 2001 [DOI] [PubMed] [Google Scholar]
- 14.George TP, Vessicchio JC, Termine A, et al. : A placebo controlled trial of bupropion for smoking cessation in schizophrenia. Biol Psychiatry 52:53–61, 2002 [DOI] [PubMed] [Google Scholar]
- 15.Evins AE, Cather C, Deckersbach T, et al. : A double-blind placebo-controlled trial of bupropion sustained-release for smoking cessation in schizophrenia. J Clin Psychopharmacol 25:218–25, 2005 [DOI] [PubMed] [Google Scholar]
- 16.Evins AE, Cather C, Culhane MA, et al. : A 12-week double-blind, placebo-controlled study of bupropion sr added to high-dose dual nicotine replacement therapy for smoking cessation or reduction in schizophrenia. J Clin Psychopharmacol 27:380–6, 2007 [DOI] [PubMed] [Google Scholar]
- 17.George TP, Vessicchio JC, Sacco KA, et al. : A placebo-controlled trial of bupropion combined with nicotine patch for smoking cessation in schizophrenia. Biol Psychiatry 63:1092–6, 2008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Weiner E, Buchholz A, Coffay A, et al. : Varenicline for smoking cessation in people with schizophrenia: a double blind randomized pilot study. Schizophr Res 129:94–5, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Tsoi DT, Porwal M, Webster AC: Interventions for smoking cessation and reduction in individuals with schizophrenia. Cochrane Database Syst Rev:CD007253, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Tidey JW, Miller ME: Smoking cessation and reduction in people with chronic mental illness. BMJ 351:h4065, 2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Sheals K, Tombor I, McNeill A, et al. : A mixed-method systematic review and meta-analysis of mental health professionals’ attitudes toward smoking and smoking cessation among people with mental illnesses. Addiction 111:1536–53, 2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Pearsall R, Smith DJ, Geddes JR: Pharmacological and behavioural interventions to promote smoking cessation in adults with schizophrenia and bipolar disorders: a systematic review and meta-analysis of randomised trials. BMJ Open 9:e027389, 2019 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Cerimele JM, Durango A: Does varenicline worsen psychiatric symptoms in patients with schizophrenia or schizoaffective disorder? A review of published studies. J Clin Psychiatry 73:e1039–47, 2012 [DOI] [PubMed] [Google Scholar]
- 24.Protection Patient and Affordable Care Act; in Health-Related Portions of the Health Care and Education Reconciliation Act of 2010. Washington, D.C.: Office of the Legislative Counsel For the use of the U.S. House of Representatives, 2010 [Google Scholar]
- 25.DiGiulio A, Jump Z, Yu A, et al. : State Medicaid coverage for tobacco cessation treatments and barriers to accessing treatments - United States, 2015–2017. MMWR Morb Mortal Wkly Rep 67:390–5, 2018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Anthenelli RM, Benowitz NL, West R, et al. : Neuropsychiatric safety and efficacy of varenicline, bupropion, and nicotine patch in smokers with and without psychiatric disorders (EAGLES): a double-blind, randomised, placebo-controlled clinical trial. Lancet 387:2507–20, 2016 [DOI] [PubMed] [Google Scholar]
- 27.Evins AE, Benowitz NL, West R, et al. : Neuropsychiatric safety and efficacy of varenicline, bupropion, and nicotine patch in smokers with psychotic, anxiety, and mood disorders in the EAGLES trial. J Clin Psychopharmacol 39:108–16, 2019 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.First MB, Spitzer RL, Gibbon M, et al. : Structured Clinical Interview for DSM-IV-TR Axis I Disorders, Research Version, Non-Patient Edition. New York, New York Biometrics Research, New York State Psychiatric Institute, 2002 [Google Scholar]
- 29.Guy W: ECDEU Assessment Manual for Psychopharmacology. Rockville, MD: US Department of Heath, Education, and Welfare Public Health Service Alcohol, Drug Abuse, and Mental Health Administration, 1976 [Google Scholar]
- 30.Fiore MC, Jaén CR, Baker TB, et al. : Treating Tobacco Use and Dependence: 2008 Update; in Clinical Practice Guideline. Rockville, MD: U.S. Department of Health and Human Services. Public Health Service, 2008 [Google Scholar]
- 31.First M, Gibbon M, Spitzer R, et al. : Structured Clinical Interview for DSM-IV Axis II Personality Disorders (SCID-II). Washington, D.C.: American Psychiatric Press, Inc, 1997 [Google Scholar]
- 32.Fagerström K: Determinants of tobacco use and renaming the FTND to the Fagerstrom Test for Cigarette Dependence. Nicotine Tob Res 14:75–8, 2012 [DOI] [PubMed] [Google Scholar]
- 33.Anthenelli RM, Morris C, Ramey TS, et al. : Effects of varenicline on smoking cessation in adults with stably treated current or past major depression: a randomized trial. Ann Intern Med 159:390–400, 2013 [DOI] [PubMed] [Google Scholar]
- 34.Zigmond AS, Snaith RP: The hospital anxiety and depression scale. Acta psychiatrica Scandinavica 67:361–70, 1983 [DOI] [PubMed] [Google Scholar]
- 35.Posner K, Brown GK, Stanley B, et al. : The Columbia-Suicide Severity Rating Scale: initial validity and internal consistency findings from three multisite studies with adolescents and adults. The American journal of psychiatry 168:1266–77, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.West R, Evins AE, Benowitz NL, et al. : Factors associated with the efficacy of smoking cessation treatments and predictors of smoking abstinence in EAGLES. Addiction 113:1507–16, 2018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Williams JM, Anthenelli RM, Morris CD, et al. : A randomized, double-blind, placebo-controlled study evaluating the safety and efficacy of varenicline for smoking cessation in patients with schizophrenia or schizoaffective disorder. J Clin Psychiatry 73:654–60, 2012 [DOI] [PubMed] [Google Scholar]
- 38.Barua RS, Rigotti NA, Benowitz NL, et al. : 2018. ACC Expert Consensus Decision Pathway on Tobacco Cessation Treatment: a report of the American College of Cardiology Task Force on clinical expert consensus documents. J Am Coll Cardiol 72:3332–65, 2018 [DOI] [PubMed] [Google Scholar]
- 39.Taylor G, McNeill A, Girling A, et al. : Change in mental health after smoking cessation: systematic review and meta-analysis. BMJ 348:g1151, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Hammett PJ, Lando HA, Taylor BC, et al. : The relationship between smoking cessation and binge drinking, depression, and anxiety symptoms among smokers with serious mental illness. Drug Alcohol Depend 194:128–35, 2019 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Cather C, Hoeppner S, Pachas G, et al. : Improved Depressive Symptoms in Adults with Schizophrenia During a Smoking Cessation Attempt with Varenicline and Behavioral Therapy. J Dual Diagn 13:168–78, 2017 [DOI] [PubMed] [Google Scholar]
- 42.Chang CK, Hayes RD, Perera G, et al. : Life expectancy at birth for people with serious mental illness and other major disorders from a secondary mental health care case register in London. PLoS One 6:e19590, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Druss BG, Zhao L, Von Esenwein S, et al. : Understanding excess mortality in persons with mental illness: 17-year follow up of a nationally representative US survey. Med Care 49:599–604, 2011 [DOI] [PubMed] [Google Scholar]
- 44.Pirie K, Peto R, Reeves GK, et al. : The 21st century hazards of smoking and benefits of stopping: a prospective study of one million women in the UK. Lancet 381:133–41, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Thorndike AN, Achtyes ED, Cather C, et al. : Weight gain and 10-year cardiovascular risk with sustained tobacco abstinence in smokers with serious mental illness: a subgroup analysis of a randomized trial. J Clin Psychiatry 77:e320–6, 2016 [DOI] [PubMed] [Google Scholar]
- 46.Evins AE, Cather C, Laffer A: Treatment of tobacco use disorders in smokers with serious mental illness: toward clinical best practices. Harv Rev Psychiatry 23:90–8, 2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Zevin S, Benowitz NL: Drug interactions with tobacco smoking. An update. Clin Pharmacokinet 36:425–38, 1999 [DOI] [PubMed] [Google Scholar]
- 48.Schrenk D, Brockmeier D, Morike K, et al. : A distribution study of CYP1A2 phenotypes among smokers and non-smokers in a cohort of healthy Caucasian volunteers. Eur J Clin Pharmacol 53:361–7, 1998 [DOI] [PubMed] [Google Scholar]
- 49.Evins AE, Cather C, Pratt SA, et al. : Maintenance treatment with varenicline for smoking cessation in patients with schizophrenia and bipolar disorder: a randomized clinical trial. JAMA 311:145–54, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Evins AE, Hoeppner SS, Schoenfeld DA, et al. : Maintenance pharmacotherapy normalizes the relapse curve in recently abstinent tobacco smokers with schizophrenia and bipolar disorder. Schizophr Res 183:124–9, 2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.