In women who have stress incontinence and intrinsic sphincter deficiency, which midurethral sling produces the best long-term results?

Article Type
Changed
Tue, 08/28/2018 - 10:59
Display Headline
In women who have stress incontinence and intrinsic sphincter deficiency, which midurethral sling produces the best long-term results?

RELATED ARTICLE WITH VIDEOS

3 video clips illustrating midurethral sling procedures

These videos were selected by Dr. Walters and presented courtesy of the International Academy of Pelvic Surgery (IAPS)

When ISD is present, the urethra cannot coaptate and loses its ability to maintain a watertight seal. Women who have this condition often are severely incontinent, leaking urine at low volumes and pressures and with minimal exertion.

In this randomized trial, Schierlitz and colleagues hypothesized that TOT would produce higher objective and subjective failure rates than the TVT. This was confirmed by 6-month data published in 2008.

Details of the trial

Women who had SUI were included in the trial if they had ISD based on urodynamic findings (i.e., maximum urethral closure pressure ≤20 cm H2O or Valsalva leak-point pressure ≤60 cm H2O, or both) and were randomly assigned to TVT or TOT. The primary endpoint was symptomatic SUI (confirmed by repeat urodynamic testing) that required a second procedure upon patient request.

Participants were followed for 3 years. If a patient reported symptoms, urodynamic testing was repeated. In addition, the patient was offered another surgery, usually involving placement of a TVT sling.

Schierlitz and colleagues concluded that, if TVT were used in all patients, repeat surgery would be avoided in one in every six patients. The risk of repeat surgery was 15 times greater for TOT, compared with the TVT sling. The median time to failure was 15.6 months for the TOT sling, compared with 43.7 months for the TVT.

Of the 16 patients who underwent repeat surgery, 56% were cured, 25% reported minimal leakage, and 19% remained unchanged.

Quality-of-life scores were similar between groups at the 6-month follow-up.

Why did the TVT outperform the TOT in this population?

Investigators theorized that there is a difference in sling axis, with the TVT placed at a more acute angle than the TOT sling. In addition, the location of the TOT sling is more distal than that of the TVT, based on ultrasonographic imaging. As a result, more effective urethral kinking and support are likely with the TVT sling, improving continence rates.

Strengths and limitations of the trial

The randomization of participants and long-term follow-up bolster the trial’s credibility.

Weaknesses include unblinded participation and postoperative surgical assessment.

Although the sample size was underpowered, there was a significant difference in the primary outcome between the two groups.

WHAT THIS EVIDENCE MEANS FOR PRACTICE

Long-term success is more likely with placement of a TVT sling in women who have SUI with ISD.

Urodynamic assessment still serves an important role in the diagnosis of ISD, and aids in preoperative planning.

LADIN A. YURTERI-KAPLAN, MD, AND AMY J. PARK, MD

We want to hear from you! Tell us what you think.

Article PDF
Author and Disclosure Information

The TVT sling. Among 164 women who had urodynamically confirmed stress urinary incontinence (SUI) and intrinsic sphincter deficiency (ISD), the tension-free vaginal tape (TVT) produced significantly greater long-term cure rates, compared with the transobturator tape (TOT). After 3 years, 15 of 75 women (20%) in the TOT group underwent repeat surgery to correct SUI, compared with one woman of 72 (1.4%) in the TVT group (P<.001).

Schierlitz L, Dwyer PL, Rosamilia A, et al. Three-year follow-up of tension-free vaginal tape compared with transobturator tape in women with stress urinary incontinence and intrinsic sphincter deficiency. Obstet Gynecol. 2012;119(2 Part 1):321–327.

EXPERT COMMENTARY

Ladin A. Yurteri-Kaplan, MD
Clinical Fellow, Section of Female Pelvic Medicine and Reconstructive Surgery, MedStar Washington Hospital Center and Georgetown University School of Medicine, Washington, DC

Amy J. Park, MD
Assistant Professor, Section of Female Pelvic Medicine and Reconstructive Surgery, MedStar Washington Hospital Center and Georgetown University School of Medicine.

Issue
OBG Management - 24(06)
Publications
Topics
Page Number
52-51
Sections
Author and Disclosure Information

The TVT sling. Among 164 women who had urodynamically confirmed stress urinary incontinence (SUI) and intrinsic sphincter deficiency (ISD), the tension-free vaginal tape (TVT) produced significantly greater long-term cure rates, compared with the transobturator tape (TOT). After 3 years, 15 of 75 women (20%) in the TOT group underwent repeat surgery to correct SUI, compared with one woman of 72 (1.4%) in the TVT group (P<.001).

Schierlitz L, Dwyer PL, Rosamilia A, et al. Three-year follow-up of tension-free vaginal tape compared with transobturator tape in women with stress urinary incontinence and intrinsic sphincter deficiency. Obstet Gynecol. 2012;119(2 Part 1):321–327.

EXPERT COMMENTARY

Ladin A. Yurteri-Kaplan, MD
Clinical Fellow, Section of Female Pelvic Medicine and Reconstructive Surgery, MedStar Washington Hospital Center and Georgetown University School of Medicine, Washington, DC

Amy J. Park, MD
Assistant Professor, Section of Female Pelvic Medicine and Reconstructive Surgery, MedStar Washington Hospital Center and Georgetown University School of Medicine.

Author and Disclosure Information

The TVT sling. Among 164 women who had urodynamically confirmed stress urinary incontinence (SUI) and intrinsic sphincter deficiency (ISD), the tension-free vaginal tape (TVT) produced significantly greater long-term cure rates, compared with the transobturator tape (TOT). After 3 years, 15 of 75 women (20%) in the TOT group underwent repeat surgery to correct SUI, compared with one woman of 72 (1.4%) in the TVT group (P<.001).

Schierlitz L, Dwyer PL, Rosamilia A, et al. Three-year follow-up of tension-free vaginal tape compared with transobturator tape in women with stress urinary incontinence and intrinsic sphincter deficiency. Obstet Gynecol. 2012;119(2 Part 1):321–327.

EXPERT COMMENTARY

Ladin A. Yurteri-Kaplan, MD
Clinical Fellow, Section of Female Pelvic Medicine and Reconstructive Surgery, MedStar Washington Hospital Center and Georgetown University School of Medicine, Washington, DC

Amy J. Park, MD
Assistant Professor, Section of Female Pelvic Medicine and Reconstructive Surgery, MedStar Washington Hospital Center and Georgetown University School of Medicine.

Article PDF
Article PDF

RELATED ARTICLE WITH VIDEOS

3 video clips illustrating midurethral sling procedures

These videos were selected by Dr. Walters and presented courtesy of the International Academy of Pelvic Surgery (IAPS)

When ISD is present, the urethra cannot coaptate and loses its ability to maintain a watertight seal. Women who have this condition often are severely incontinent, leaking urine at low volumes and pressures and with minimal exertion.

In this randomized trial, Schierlitz and colleagues hypothesized that TOT would produce higher objective and subjective failure rates than the TVT. This was confirmed by 6-month data published in 2008.

Details of the trial

Women who had SUI were included in the trial if they had ISD based on urodynamic findings (i.e., maximum urethral closure pressure ≤20 cm H2O or Valsalva leak-point pressure ≤60 cm H2O, or both) and were randomly assigned to TVT or TOT. The primary endpoint was symptomatic SUI (confirmed by repeat urodynamic testing) that required a second procedure upon patient request.

Participants were followed for 3 years. If a patient reported symptoms, urodynamic testing was repeated. In addition, the patient was offered another surgery, usually involving placement of a TVT sling.

Schierlitz and colleagues concluded that, if TVT were used in all patients, repeat surgery would be avoided in one in every six patients. The risk of repeat surgery was 15 times greater for TOT, compared with the TVT sling. The median time to failure was 15.6 months for the TOT sling, compared with 43.7 months for the TVT.

Of the 16 patients who underwent repeat surgery, 56% were cured, 25% reported minimal leakage, and 19% remained unchanged.

Quality-of-life scores were similar between groups at the 6-month follow-up.

Why did the TVT outperform the TOT in this population?

Investigators theorized that there is a difference in sling axis, with the TVT placed at a more acute angle than the TOT sling. In addition, the location of the TOT sling is more distal than that of the TVT, based on ultrasonographic imaging. As a result, more effective urethral kinking and support are likely with the TVT sling, improving continence rates.

Strengths and limitations of the trial

The randomization of participants and long-term follow-up bolster the trial’s credibility.

Weaknesses include unblinded participation and postoperative surgical assessment.

Although the sample size was underpowered, there was a significant difference in the primary outcome between the two groups.

WHAT THIS EVIDENCE MEANS FOR PRACTICE

Long-term success is more likely with placement of a TVT sling in women who have SUI with ISD.

Urodynamic assessment still serves an important role in the diagnosis of ISD, and aids in preoperative planning.

LADIN A. YURTERI-KAPLAN, MD, AND AMY J. PARK, MD

We want to hear from you! Tell us what you think.

RELATED ARTICLE WITH VIDEOS

3 video clips illustrating midurethral sling procedures

These videos were selected by Dr. Walters and presented courtesy of the International Academy of Pelvic Surgery (IAPS)

When ISD is present, the urethra cannot coaptate and loses its ability to maintain a watertight seal. Women who have this condition often are severely incontinent, leaking urine at low volumes and pressures and with minimal exertion.

In this randomized trial, Schierlitz and colleagues hypothesized that TOT would produce higher objective and subjective failure rates than the TVT. This was confirmed by 6-month data published in 2008.

Details of the trial

Women who had SUI were included in the trial if they had ISD based on urodynamic findings (i.e., maximum urethral closure pressure ≤20 cm H2O or Valsalva leak-point pressure ≤60 cm H2O, or both) and were randomly assigned to TVT or TOT. The primary endpoint was symptomatic SUI (confirmed by repeat urodynamic testing) that required a second procedure upon patient request.

Participants were followed for 3 years. If a patient reported symptoms, urodynamic testing was repeated. In addition, the patient was offered another surgery, usually involving placement of a TVT sling.

Schierlitz and colleagues concluded that, if TVT were used in all patients, repeat surgery would be avoided in one in every six patients. The risk of repeat surgery was 15 times greater for TOT, compared with the TVT sling. The median time to failure was 15.6 months for the TOT sling, compared with 43.7 months for the TVT.

Of the 16 patients who underwent repeat surgery, 56% were cured, 25% reported minimal leakage, and 19% remained unchanged.

Quality-of-life scores were similar between groups at the 6-month follow-up.

Why did the TVT outperform the TOT in this population?

Investigators theorized that there is a difference in sling axis, with the TVT placed at a more acute angle than the TOT sling. In addition, the location of the TOT sling is more distal than that of the TVT, based on ultrasonographic imaging. As a result, more effective urethral kinking and support are likely with the TVT sling, improving continence rates.

Strengths and limitations of the trial

The randomization of participants and long-term follow-up bolster the trial’s credibility.

Weaknesses include unblinded participation and postoperative surgical assessment.

Although the sample size was underpowered, there was a significant difference in the primary outcome between the two groups.

WHAT THIS EVIDENCE MEANS FOR PRACTICE

Long-term success is more likely with placement of a TVT sling in women who have SUI with ISD.

Urodynamic assessment still serves an important role in the diagnosis of ISD, and aids in preoperative planning.

LADIN A. YURTERI-KAPLAN, MD, AND AMY J. PARK, MD

We want to hear from you! Tell us what you think.

Issue
OBG Management - 24(06)
Issue
OBG Management - 24(06)
Page Number
52-51
Page Number
52-51
Publications
Publications
Topics
Article Type
Display Headline
In women who have stress incontinence and intrinsic sphincter deficiency, which midurethral sling produces the best long-term results?
Display Headline
In women who have stress incontinence and intrinsic sphincter deficiency, which midurethral sling produces the best long-term results?
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media

PTSD nightmares: Prazosin and atypical antipsychotics

Article Type
Changed
Tue, 12/11/2018 - 14:48
Display Headline
PTSD nightmares: Prazosin and atypical antipsychotics

 

Practice Points

• Prazosin is recommended as a first-line therapy for nighttime PTSD symptoms, such as nightmares or sleep disturbances—especially among veterans—because of superior long-term effectiveness.

Risk of metabolic syndrome, which has been reported with low-dose atypical antipsychotics used for treating insomnia, limits their use for PTSD-related nightmares.

Mr. S, a 45-year-old veteran, was diagnosed with posttraumatic stress disorder (PTSD) 18 years ago after a tour of duty in the Persian Gulf. He had combat-related flashbacks triggered by the smell of gasoline or smoke from a fire, was easily startled, and began to isolate himself socially. However, his symptoms improved when he started volunteering at his local Veterans Affairs Medical Center. After he lost his job 3 years ago, Mr. S started experiencing flashbacks. He was irritable, easily startled, and avoided things that reminded him of his time in the Persian Gulf. His psychiatrist prescribed sertraline, titrated to 200 mg/d. The drug reduced the severity of his avoidance and hyperarousal symptoms and improved his mood.

During a clinic visit, Mr. S says he is doing well and can fall asleep at night but is having recurring nightmares about traumatic events that occurred during combat. These nightmares wake him up and have become more frequent, occurring once per night for the past month. Mr. S says he has been watching more news programs about conflicts in Afghanistan and Iraq since the nightmares began. His psychiatrist starts quetiapine, 50 mg at bedtime for 7 nights then 100 mg at bedtime, but after 6 weeks Mr. S says his nightmares continue.

PTSD occurs in approximately 19% of Vietnam war combat veterans1 and 14% of service members returning from Iraq and Afghanistan.2 PTSD symptoms are classified into clusters: intrusive/re-experiencing; avoidant/numbing; and hyperarousal.3 Nightmares are part of the intrusive/re-experiencing cluster, which is Criterion B in DSM-IV-TR. See Table 1 for a description of DSM-IV-TR PTSD criteria. Among PTSD patients, 50% to 70% report PTSD-associated nightmares.4 Despite adequate treatment targeted to improve PTSD’s core symptoms, symptoms such as sleep disturbances or nightmares often persist.

Table 1

DSM-IV-TR diagnostic criteria for posttraumatic stress disorder

 

  1. The person has been exposed to a traumatic event in which both of the following were present:
    1. The person experienced, witnessed, or was confronted with an event or events that involved actual or threatened death or serious injury, or a threat to the physical integrity of self or others
    2. The person’s response involved intense fear, helplessness, or horror
  2. The traumatic event is persistently reexperienced in ≥1 of the following ways:
    1. Recurrent and intrusive distressing recollections of the event
    2. Recurrent distressing dreams of the event
    3. Acting or feeling as if the traumatic event were recurring
    4. Intense psychological distress at exposure to internal or external cues that symbolize or resemble an aspect of the traumatic event
    5. Physiological reactivity on exposure to internal or external cues that symbolize or resemble an aspect of the traumatic event
  3. Persistent avoidance of stimuli associated with the trauma and numbing of general responsiveness (not present before the trauma), as indicated by ≥3 of the following:
    1. Efforts to avoid thoughts, feelings, or conversations associated with the trauma
    2. Efforts to avoid activities, places, or people that arouse recollections of the trauma
    3. Inability to recall an important aspect of the trauma
    4. Markedly diminished interest or participation in significant activities
    5. Feeling of detachment or estrangement from others
    6. Restricted range of affect
    7. Sense of a foreshortened future
  4. Persistent symptoms of increased arousal (not present before the trauma), as indicated by ≥2 of the following:
    1. Difficulty falling or staying asleep
    2. Irritability or outbursts of anger
    3. Difficulty concentrating
    4. Hypervigilance
    5. Exaggerated startle response
  5. Duration of disturbance (symptoms in Criteria B, C, and D) is >1 month
  6. The disturbance causes clinically significant distress or impairment of social, occupational, or other important areas of functioning
Source: Diagnostic and statistical manual of mental disorders, 4th ed, text rev. Washington, DC: American Psychiatric Association; 2000

Nightmares and other sleep disturbances are associated with significant distress and daytime impairment and can interfere with PTSD recovery4-8 by disrupting sleep-dependent processing of emotional experiences and causing repeated resensitization to trauma cues (Table 2).8

Table 2

Psychosocial consequences of sleep disruption in PTSD

 

Increased reactivity to emotional cues
Compromised ability to function in social and occupational roles
Negative psychiatric outcomes, including suicidal ideation or worsening of depression or psychosis
Interference of natural recovery from trauma exposure
Repeated resensitization to trauma cues
Neurocognitive deficits
Neuroendocrine abnormalities
PTSD: posttraumatic stress disorder
Source: Adapted from reference 8

Few randomized controlled medication trials specifically address PTSD-related nightmares. Most PTSD studies do not examine sleep outcomes as a primary measure, and comprehensive literature reviews could not offer evidence-based recommendations.9,10 The American Academy of Sleep Medicine (AASM) also noted a paucity of PTSD studies that identified nightmares as a primary outcome measure.11 See Table 3 for a list of recommended medication options for PTSD-associated nightmares.

 

 

Table 3

Recommended medication treatments for PTSD-associated nightmares

 

Evidence levelMedicationEvidence
Recommended for treating PTSD-associated nightmares
1, 4PrazosinIn 3 level 1 studies, adding prazosin (mean dose 3 mg/d) significantly decreased trauma-related nightmares according to the CAPS “recurrent distressing dreams” item after 3 to 9 weeks of treatment vs placebo in veteran and civilian patients (N = 57)
Not suggested for treating PTSD-associated nightmares
1VenlafaxineNo difference between extended-release venlafaxine (37.5 to 300 mg/d) and placebo in the CAPS-SX17 “distressing dreams” item at 12 weeks in 340 PTSD patients
May be considered for treating PTSD-associated nightmares
4ClonidineReduced the number of nightmares in 11 of 13 refugees for 2 weeks to 3 months (dose: 0.2 to 0.6 mg/d)
May be considered for treating PTSD-associated nightmares, but data are low grade and sparse
4TrazodoneAlthough trazodone (25 to 600 mg) significantly decreased nightmare frequency in veteran patients during an 8-week hospital stay (N = 60), 19% discontinued therapy because of side effects
4OlanzapineAdjunctive olanzapine (10 to 20 mg) rapidly improved sleep in a case series of combat-related PTSD patients resistant to SSRIs and benzodiazepines (N = 5)
4RisperidoneIn case series, risperidone (0.5 to 3 mg) significantly decreased CAPS scores for recurrent distressing dreams and proportion of traumatic dreams documented in diaries of combat veterans over 6 weeks (N = 17), and improved nightmares in adult burn patients taking pain medications after 1 to 2 days (N = 10)
4AripiprazoleIn a case series, aripiprazole (15 to 30 mg at bedtime) with CBT or sertraline significantly improved nightmares in 4 of 5 combat-related PTSD patients
4TopiramateTopiramate reduced nightmares in 79% of civilians with PTSD and fully suppressed nightmares in 50% of patients in a case series (N = 35)
4Low-dose cortisolSignificant decrease in frequency but not intensity of nightmares with low-dose cortisol (10 mg/d) in civilians with PTSD (N = 3)
4FluvoxamineIn 2 case series, fluvoxamine (up to 300 mg/d) significantly decreased the IES-R level of “dreams about combat trauma” but not the SRRS “bad dreams” rating at 10 weeks (N = 21). During 4 to 12 weeks of follow-up there was a qualitative decrease in reported nightmares in veteran patients (n = 12)
2Triazolam/nitrazepamLimited data showed triazolam (0.5 mg) and nitrazepam (5 mg) provide equal efficacy in decreasing the number of patients who experience unpleasant dreams over 1 night
4PhenelzineOne study showed phenelzine monotherapy (30 to 90 mg) resulted in elimination of nightmares within 1 month (N = 5); another reported “moderately reduced traumatic dreams” (N = 21) in veterans. Therapy was discontinued because of short-lived efficacy or plateau effect
4GabapentinAdjunctive gabapentin (300 to 3,600 mg/d) improved insomnia and decreased nightmare frequency and/or intensity over 1 to 36 months in 30 veterans with PTSD
4CyproheptadineConflicting data ranges from eliminating nightmares to no changes in the presence or intensity of nightmares
4TCAsAmong 10 Cambodian concentration camp survivors treated with TCAs, 4 reported their nightmares ceased and 4 reported improvement after 1-year follow-up
4NefazodoneReduced nightmare occurrence in 3 open-label studies as monotherapy (386 to 600 mg/d). Not recommended first line because of hepatotoxicity risk
No recommendation because of sparse data
2ClonazepamClonazepam (1 to 2 mg/d) was ineffective in decreasing frequency or intensity of combat-related PTSD nightmares in veterans (N = 6)
Evidence levels:
  1. High-quality randomized clinical trials with narrow confidence intervals
  2. Low-quality randomized clinical trials or high-quality cohort studies
  3. Case-control studies
  4. Case series; poor case-control studies; poor cohort studies; case reports
CAPS: Clinician-Administered PTSD Scale; CAPS-SX17: 17-item Clinician-Administered PTSD Scale; CBT: cognitive-behavioral therapy; IES-R: Impact of Event Scale-Revised; PTSD: posttraumatic stress disorder; SRRS: Stress Response Rating Scale; SSRI: selective serotonin reuptake inhibitor; TCAs: tricyclic antidepressants
Source: Adapted from Aurora RN, Zak RS, Auerbach SH, et al. Best practice guide for the treatment of nightmare disorder in adults. J Clin Sleep Med. 2010;6(4):389-401

CASE CONTINUED: Medication change, improvement

After reviewing AASM’s treatment recommendations, we prescribe prazosin, 1 mg at bedtime for 7 nights, then increase by 1 mg at bedtime each week until Mr. S’s nightmares improve. He reports a substantial improvement in nightmare severity and frequency after a few weeks of treatment with prazosin, 5 mg at bedtime.

Prazosin

 

Prazosin is an α1-adrenergic receptor antagonist with good CNS penetrability. The rationale for reducing adrenergic activity to address intrusive PTSD symptoms has been well documented.12,13 In open-label trials,14-18 a chart review,19 and placebo-controlled trials,20-22prazosin reduced trauma nightmares and improved sleep quality and global clinical status more than placebo (Table 4). In these studies, prazosin doses ranged from 1 to 20 mg/d, with an average of 3 mg at bedtime and a starting dose of 1 mg. Prazosin is the only agent recommended in the AASM’s Best Practice Guide for treating PTSD-related nightmares.11

 

 

Table 4

RCTs of prazosin for trauma-related nightmares

 

StudyDesignPatientsResults
Raskind et al, 20032020-week, double-blind, placebo-controlled, crossover study (mean dose 9.5 mg/d at bedtime)10 Vietnam veterans with chronic PTSD and severe trauma-related nightmaresPrazosin was superior to placebo on scores on the recurrent distressing dreams item and difficulty falling/staying asleep item of the CAPS and change in PTSD severity and functional status on the CGI-C
Raskind et al, 2007218-week, placebo-controlled, parallel study (mean dose 13.3 ± 3 mg/d in the evening)40 veterans with chronic PTSD, distressing trauma nightmares, and sleep disturbancePrazosin was superior to placebo in reducing trauma nightmares and improving sleep quality and global clinical status; prazosin also shifted dream characteristics of trauma-related nightmares to those typical of normal dreams
Taylor et al, 2008227-week, randomized, placebo-controlled, crossover trial (mean dose 3.1 ± 1.3 mg)13 outpatients with chronic civilian trauma PTSD, frequent nightmares, and sleep disturbancePrazosin significantly increased total sleep time and REM sleep time; reduced trauma-related nightmares, distressed awakenings, and total PCL-C scores; improved CGI-I scores; and changed PDRS scores toward normal dreaming
CAPS: Clinician-Administered PTSD Scale; CGI-C: Clinical Global Impression of Change; CGI-I: Clinical Global Impression of Improvement; PCL-C: PTSD Checklist-Civilian; PDRS: PTSD Dream Rating Scale; PTSD: posttraumatic stress disorder; RCTs: randomized controlled trials; REM: rapid eye movement

Atypical antipsychotics

Atypical antipsychotics have been used to reduce nightmares in PTSD; however, most of the evidence from studies evaluated in the AASM’s Best Practice Guide were considered to be low quality.11 Quetiapine and ziprasidone were not included in the AASM review. See (Table 5) for a review of the evidence for atypical antipsychotics for treating PTSD nightmares.

Table 5

Combat-related nightmares: Evidence for atypical antipsychotics

 

StudyDesignPatients/dosageResults
Aripiprazole
Lambert, 2006 aCase report4 veterans with combat-related PTSD (3 male, 1 female; age 22 to 24); dose: 15 to 30 mg; concurrent treatment sertraline or CBTDecreased frequency of weekly nightmares and agitated sleep by at least 50%
Olanzapine
Stein et al, 2002 b8-week, double-blind, placebo-controlled study19 male veterans with combat-related PTSD (olanzapine group mean age: 55.2 ± 6.6; placebo group 51.1 ± 8.1); mean dose: 15 mg/dSignificantly greater reduction in sleep disturbances (PSQI: -3.29 vs 1.57; P = .01); significantly higher weight gain (13.2 lbs vs -3 lbs; P = .001)
Jakovljevic et al, 2003 cCase reports5 veterans with combat-related PTSD for 6 to 7 years (age: 28 to 50); dose: 10 to 20 mg; adjunct treatmentDecreased frequency of nightmares within 3 days
Labbate et al, 2000 dCase report1 male veteran (age: 58) with a 20-year history of combat-related PTSD; dose: 5 mg at bedtime; concurrent treatment with sertraline (200 mg/d), bupropion (150 mg/d), and diazepam (15 mg/d)Eliminated nightmares after 1 week and improved sleep quality
Quetiapine
Ahearn et al, 2006 e8-week, open-label trial15 PTSD patients (8 male; 7 female; 5 with combat-related PTSD; mean age: 49); mean dose: 216 mg/d (100 to 400 mg/d)Significantly improved re-experiencing (CAPS: 10 vs 23; P = .0012) and sleep (PSQI: 17.5 vs 30; P = .0044) at 8 weeks compared with baseline
Robert et al, 2005 f6-week, open-label trial19 combat veterans; mean dose: 100 ± 70 mg/d (25 to 300 mg/d); adjunct treatmentSignificantly improved sleep quality (PSQI: 1.67 vs 2.41; P = .006), latency (PSQI: 1.5 vs 2.65; P = .002), duration (PSQI: 1.31 vs 2.71; P < .001), and sleep disturbances (PSQI: 1.22 vs 1.71; P = .034) and decreased terror episodes (PSQI-A: 0.73 vs 0.91; P = .040) and acting out dreams (PSQI-A: 1.07 vs 1.35; P = .013); however, no difference in nightmares caused by trauma (PSQI-A: 1.53 vs 2.06)
Sokolski et al, 2003 gRetrospective chart review68 male Vietnam War combat veterans (mean age: 55 ± 3.5); mean dose: 155 ± 130 mg (25 to 700 mg); adjunct treatmentImproved sleep disturbances in 62% and nightmares in 25% of patients
Ahearn et al, 2003 hCase report2 male patients with combat-related PTSD (age 53, 72); dose: 25 to 50 mg; adjunct to SSRI therapyDecreased frequency of nightmares with increased sleep duration
Risperidone
David et al, 2006 i6-week, open-label trial17 male veterans with combat-related PTSD (mean age: 53.7 ± 3.8); mean maximum dose: 2.3 ± 0.6 mg (range: 1 to 3 mg)Improved recurrent distressing dreams (CAPS B-2: 3.8 vs 5.4; P = .04), but not with the PSQI subscale (PSQI bad dreams: 2.5 vs 2.7; NS). Decreased nighttime awakenings (1.9 vs 2.8; P = .003) and trauma dreams (19% vs 38%; P = .04)
Leyba et al, 1998 jCase reports3 male patients (age 43 to 46); dose: 1 to 3 mg; adjunct therapyDecreased occurrence of nightmares
Ziprasidone
Siddiqui et al, 2005 kCase report1 male veteran with chronic combat-related PTSD (age 55); dose: 80 to 120 mg/d; adjunct with trazodone (100 mg) and topiramateImproved occurrence of nightmares up to 4 months
CAPS: Clinician-Administered PTSD Scale; CAPS B-2: Clinician-Administered PTSD Scale B-2 (recurrent distressing dreams of the event); CBT: cognitive-behavioral therapy; PSQI: Pittsburgh Sleep Quality Index; PSQI-A: Pittsburgh Sleep Quality Index Addendum for PTSD; NS: not significant; PTSD: posttraumatic stress disorder; SSRI: selective serotonin reuptake inhibitor References
  1. Lambert MT. Aripiprazole in the management of post-traumatic stress disorder symptoms in returning Global War on Terrorism veterans. Int Clin Psychopharmacol. 2006;21(3):185-187.
  2. Stein MB, Kline NA, Matloff JL. Adjunctive olanzapine for SSRI-resistant combat-related PTSD: a double-blind, placebo-controlled study. Am J Psychiatry. 2002;159(10):1777-1779.
  3. Jakovljevic M, Sagud M, Mihaljevic-Peles A. Olanzapine in the treatment-resistant, combat-related PTSD—a series of case reports. Acta Psychiatr Scand. 2003;107(5):394-396.
  4. Labbate LA, Douglas S. Olanzapine for nightmares and sleep disturbance in posttraumatic stress disorder (PTSD). Can J Psychiatry. 2000;45(7):667-668.
  5. Ahearn EP, Mussey M, Johnson C, et al. Quetiapine as an adjunctive treatment for post-traumatic stress disorder: an 8-week open-label study. Int Clin Psychopharmacol. 2006;21(1):29-33.
  6. Robert S, Hamner MB, Kose S, et al. Quetiapine improves sleep disturbances in combat veterans with PTSD: sleep data from a prospective, open-label study. J Clin Psychopharmacol. 2005;25(4):387-388.
  7. Sokolski KN, Denson TF, Lee RT, et al. Quetiapine for treatment of refractory symptoms of combat-related post-traumatic stress disorder. Mil Med. 2003;168(6):486-489.
  8. Ahearn EP, Winston E, Mussey M, et al. Atypical antipsychotics, improved intrusive symptoms in patients with posttraumatic stress disorder. Mil Med. 2003;168(9):x-xi.
  9. David D, De Faria L, Mellman TA. Adjunctive risperidone treatment and sleep symptoms in combat veterans with chronic PTSD. Depress Anxiety. 2006;23(8):489-491.
  10. Leyba CM, Wampler TP. Risperidone in PTSD. Psychiatr Serv. 1998;49(2):245-246.
  11. Siddiqui Z, Marcil WA, Bhatia SC, et al. Ziprasidone therapy for post-traumatic stress disorder. J Psychiatry Neurosci. 2005;30(6):430-431.
 

 

 

Comparing prazosin and quetiapine. A historical prospective cohort study of 237 veterans with PTSD receiving prazosin or quetiapine for nighttime PTSD symptoms demonstrated that although the 2 drugs have similar efficacy (defined as symptomatic improvement) for short-term, 6-month treatment (61% vs 62%; P=.54), a higher percentage of patients continued prazosin long-term (3 to 6 years) than those taking quetiapine (48% vs 24%; P < .001).23 Twenty-five percent of patients taking quetiapine switched to prazosin during the study, and approximately one-half of these patients remained on prazosin until the study’s end. Only 8% of prazosin patients switched to quetiapine, and none continued this therapy until study end.23 Patients in the quetiapine group were more likely to discontinue the drug because of lack of efficacy (13% vs 3%; P=.03) and adverse effects (35% vs 18%; P=.008), specifically sedation (21% vs 2%; P < .001) and metabolic effects (9% vs 0%; P=.014), compared with prazosin. Although this trial may be the only published comparison study of prazosin and quetiapine, its methodological quality has been questioned, which makes it difficult to draw definitive conclusions.

Metabolic syndrome—elevated diastolic blood pressure, increased waist circumference, and low high-density lipoprotein cholesterol—is common among PTSD patients treated with antipsychotics.24 This syndrome may be caused by medications, lifestyle factors, or long-term overactivation of stress-response pathways. A retrospective chart review at a community mental health center revealed that patients taking even low doses of quetiapine for insomnia gained an average of 5 lbs (P=.037).25 Another retrospective chart review at 2 military hospitals reported that patients receiving low-dose quetiapine (≤100 mg/d) gained an average of slightly less than 1 lb per month, which adds up to approximately 10 lbs per year (P < .001).26 The benefit of using atypical antipsychotics may be outweighed by metabolic risks such as obesity, new-onset diabetes, and dyslipidemia.27

 

Prazosin is considered a first-line treatment for sleep disturbances and nightmares in PTSD because of its superior long-term efficacy and decreased adverse effects compared with quetiapine.

Related Resources

 

  • American Psychiatric Association. Practice guidelines for the treatment of patients with acute stress disorder and posttraumatic stress disorder. Arlington, VA: American Psychiatric Publishing, Inc.; 2004.
  • Veterans Affairs/Department of Defense clinical practice guidelines. Management of traumatic stress disorder and acute stress reaction. www.healthquality.va.gov/Post_Traumatic_Stress_Disorder_PTSD.asp.

Drug Brand Names

 

  • Prazosin • Minipress
  • Quetiapine • Seroquel
  • Sertraline • Zoloft
  • Ziprasidone • Geodon

Disclosure

The authors report no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.

References

 

1. Dohrenwend BP, Turner JB, Turse NA, et al. The psychological risks of Vietnam for U.S. veterans: a revisit with new data and methods. Science. 2006;313(5789):979-982.

2. Tanielian T, Jaycox L. eds. Invisible wounds of war: psychological and cognitive injuries, their consequences, and services to assist recovery. Santa Monica, CA: RAND Corporation; 2008.

3. Diagnostic and statistical manual of mental disorders, 4th ed, text rev. Washington DC: American Psychiatric Association; 2000.

4. Wittmann L, Schredl M, Kramer M. Dreaming in posttraumatic stress disorder: a critical review of phenomenology psychophysiology and treatment. Psychother Psychosom. 2007;76(1):25-39.

5. Clum GA, Nishith P, Resick PA. Trauma-related sleep disturbance and self-reported physical health symptoms in treatment-seeking female rape victims. J Nerv Ment Dis. 2001;189(9):618-622.

6. Kramer TL, Booth BM, Han X, et al. Service utilization and outcomes in medically ill veterans with posttraumatic stress and depressive disorders. J Trauma Stress. 2003;16(3):211-219.

7. Neylan TC, Marmar CR, Metzler TJ, et al. Sleep disturbances in the Vietnam generation: findings from a nationally representative sample of male Vietnam veterans. Am J Psychiatry. 1998;155(7):929-933.

8. Nappi CM, Drummond SP, Hall JM. Treating nightmares and insomnia in posttraumatic stress disorder: a review of current evidence. Neuropharmacology. 2012;62(2):576-585.

9. Maher MJ, Rego SA, Asnis GM. Sleep disturbances in patients with post-traumatic stress disorder: epidemiology impact and approaches to management. CNS Drugs. 2006;20(7):567-590.

10. van Liempt S, Vermetten E, Geuze E, et al. Pharmacotherapy for disordered sleep in post-traumatic stress disorder: a systematic review. Int Clin Psychopharmacol. 2006;21(4):193-202.

11. Aurora RN, Zak RS, Auerbach SH, et al. Best practice guide for the treatment of nightmare disorder in adults. J Clin Sleep Med. 2010;6(4):389-401.

12. Boehnlein JK, Kinzie JD. Pharmacologic reduction of CNS noradrenergic activity in PTSD: the case for clonidine and prazosin. J Psychiatr Pract. 2007;13(2):72-78.

13. Strawn JR, Geracioti TD, Jr. Noradrenergic dysfunction and the psychopharmacology of posttraumatic stress disorder. Depress Anxiety. 2008;25(3):260-271.

14. Calohan J, Peterson K, Peskind ER, et al. Prazosin treatment of trauma nightmares and sleep disturbance in soldiers deployed in Iraq. J Trauma Stress. 2010;23(5):645-648.

15. Daly CM, Doyle ME, Radkind M, et al. Clinical case series: the use of Prazosin for combat-related recurrent nightmares among Operation Iraqi Freedom combat veterans. Mil Med. 2005;170(6):513-515.

16. Peskind ER, Bonner LT, Hoff DJ, et al. Prazosin reduces trauma-related nightmares in older men with chronic posttraumatic stress disorder. J Geriatr Psychiatry Neurol. 2003;16(3):165-171.

17. Raskind MA, Dobie DJ, Kanter ED, et al. The alpha1-adrenergic antagonist prazosin ameliorates combat trauma nightmares in veterans with posttraumatic stress disorder: a report of 4 cases. J Clin Psychiatry. 2000;61(2):129-133.

18. Taylor F, Raskind MA. The alpha1-adrenergic antagonist prazosin improves sleep and nightmares in civilian trauma posttraumatic stress disorder. J Clin Psychopharmacol. 2002;22(1):82-85.

19. Raskind MA, Thompson C, Petrie EC, et al. Prazosin reduces nightmares in combat veterans with posttraumatic stress disorder. J Clin Psychiatry. 2002;63(7):565-568.

20. Raskind MA, Peskind ER, Kanter ED, et al. Reduction of nightmares and other PTSD symptoms in combat veterans by prazosin: a placebo-controlled study. Am J Psychiatry. 2003;160(2):371-373.

21. Raskind MA, Peskind ER, Hoff DJ, et al. A parallel group placebo controlled study of prazosin for trauma nightmares and sleep disturbance in combat veterans with post-traumatic stress disorder. Biol Psychiatry. 2007;61(8):928-934.

22. Taylor FB, Martin P, Thompson C, et al. Prazosin effects on objective sleep measures and clinical symptoms in civilian trauma posttraumatic stress disorder: a placebo-controlled study. Biol Psychiatry. 2008;63(6):629-632.

23. Byers MG, Allison KM, Wendel CS, et al. Prazosin versus quetiapine for nighttime posttraumatic stress disorder symptoms in veterans: an assessment of long-term comparative effectiveness and safety. J Clin Psychopharmacol. 2010;30(3):225-229.

24. Jin H, Lanouette NM, Mudaliar S, et al. Association of posttraumatic stress disorder with increased prevalence of metabolic syndrome. J Clin Psychopharmacol. 2009;29(3):210-215.

25. Cates ME, Jackson CW, Feldman JM, et al. Metabolic consequences of using low-dose quetiapine for insomnia in psychiatric patients. Community Ment Health J. 2009;45(4):251-254.

26. Williams SG, Alinejad NA, Williams JA, et al. Statistically significant increase in weight caused by low-dose quetiapine. Pharmacotherapy. 2010;30(10):1011-1015.

27. American Diabetes Association; American Psychiatric Association; American Association of Clinical Endocrinologists; North American Association for the Study of Obesity. Consensus development conference on antipsychotic drugs and obesity and diabetes. J Clin Psychiatry. 2004;65(2):267-272.

Article PDF
Author and Disclosure Information

 

Rebecca L. Graham, PharmD
Dr. Graham is a Second-Year Psychiatric Pharmacy Resident, Veterans Affairs San Diego Healthcare System (VASDHS)
Susan G. Leckband, RPh, BCPP
Ms. Leckband is a Clinical Psychiatric Pharmacist Specialist, VASDHS, and Assistant Clinical Professor through Skaggs School of Pharmacy and Pharmaceutical Sciences and Department of Psychiatry, University of California, San Diego
Rene A. Endow-Eyer, PharmD, BCPP
Dr. Endow-Eyer is Psychiatric Clinical Pharmacy Specialist, VASDHS, and Assistant Clinical Professor through Skaggs School of Pharmacy and Pharmaceutical Sciences and Department of Psychiatry, University of California, San Diego, San Diego, CA

Vicki L. Ellingrod, PharmD, BCPP, FCCP
Series Editor

Issue
Current Psychiatry - 11(06)
Publications
Topics
Page Number
59-67
Sections
Author and Disclosure Information

 

Rebecca L. Graham, PharmD
Dr. Graham is a Second-Year Psychiatric Pharmacy Resident, Veterans Affairs San Diego Healthcare System (VASDHS)
Susan G. Leckband, RPh, BCPP
Ms. Leckband is a Clinical Psychiatric Pharmacist Specialist, VASDHS, and Assistant Clinical Professor through Skaggs School of Pharmacy and Pharmaceutical Sciences and Department of Psychiatry, University of California, San Diego
Rene A. Endow-Eyer, PharmD, BCPP
Dr. Endow-Eyer is Psychiatric Clinical Pharmacy Specialist, VASDHS, and Assistant Clinical Professor through Skaggs School of Pharmacy and Pharmaceutical Sciences and Department of Psychiatry, University of California, San Diego, San Diego, CA

Vicki L. Ellingrod, PharmD, BCPP, FCCP
Series Editor

Author and Disclosure Information

 

Rebecca L. Graham, PharmD
Dr. Graham is a Second-Year Psychiatric Pharmacy Resident, Veterans Affairs San Diego Healthcare System (VASDHS)
Susan G. Leckband, RPh, BCPP
Ms. Leckband is a Clinical Psychiatric Pharmacist Specialist, VASDHS, and Assistant Clinical Professor through Skaggs School of Pharmacy and Pharmaceutical Sciences and Department of Psychiatry, University of California, San Diego
Rene A. Endow-Eyer, PharmD, BCPP
Dr. Endow-Eyer is Psychiatric Clinical Pharmacy Specialist, VASDHS, and Assistant Clinical Professor through Skaggs School of Pharmacy and Pharmaceutical Sciences and Department of Psychiatry, University of California, San Diego, San Diego, CA

Vicki L. Ellingrod, PharmD, BCPP, FCCP
Series Editor

Article PDF
Article PDF

 

Practice Points

• Prazosin is recommended as a first-line therapy for nighttime PTSD symptoms, such as nightmares or sleep disturbances—especially among veterans—because of superior long-term effectiveness.

Risk of metabolic syndrome, which has been reported with low-dose atypical antipsychotics used for treating insomnia, limits their use for PTSD-related nightmares.

Mr. S, a 45-year-old veteran, was diagnosed with posttraumatic stress disorder (PTSD) 18 years ago after a tour of duty in the Persian Gulf. He had combat-related flashbacks triggered by the smell of gasoline or smoke from a fire, was easily startled, and began to isolate himself socially. However, his symptoms improved when he started volunteering at his local Veterans Affairs Medical Center. After he lost his job 3 years ago, Mr. S started experiencing flashbacks. He was irritable, easily startled, and avoided things that reminded him of his time in the Persian Gulf. His psychiatrist prescribed sertraline, titrated to 200 mg/d. The drug reduced the severity of his avoidance and hyperarousal symptoms and improved his mood.

During a clinic visit, Mr. S says he is doing well and can fall asleep at night but is having recurring nightmares about traumatic events that occurred during combat. These nightmares wake him up and have become more frequent, occurring once per night for the past month. Mr. S says he has been watching more news programs about conflicts in Afghanistan and Iraq since the nightmares began. His psychiatrist starts quetiapine, 50 mg at bedtime for 7 nights then 100 mg at bedtime, but after 6 weeks Mr. S says his nightmares continue.

PTSD occurs in approximately 19% of Vietnam war combat veterans1 and 14% of service members returning from Iraq and Afghanistan.2 PTSD symptoms are classified into clusters: intrusive/re-experiencing; avoidant/numbing; and hyperarousal.3 Nightmares are part of the intrusive/re-experiencing cluster, which is Criterion B in DSM-IV-TR. See Table 1 for a description of DSM-IV-TR PTSD criteria. Among PTSD patients, 50% to 70% report PTSD-associated nightmares.4 Despite adequate treatment targeted to improve PTSD’s core symptoms, symptoms such as sleep disturbances or nightmares often persist.

Table 1

DSM-IV-TR diagnostic criteria for posttraumatic stress disorder

 

  1. The person has been exposed to a traumatic event in which both of the following were present:
    1. The person experienced, witnessed, or was confronted with an event or events that involved actual or threatened death or serious injury, or a threat to the physical integrity of self or others
    2. The person’s response involved intense fear, helplessness, or horror
  2. The traumatic event is persistently reexperienced in ≥1 of the following ways:
    1. Recurrent and intrusive distressing recollections of the event
    2. Recurrent distressing dreams of the event
    3. Acting or feeling as if the traumatic event were recurring
    4. Intense psychological distress at exposure to internal or external cues that symbolize or resemble an aspect of the traumatic event
    5. Physiological reactivity on exposure to internal or external cues that symbolize or resemble an aspect of the traumatic event
  3. Persistent avoidance of stimuli associated with the trauma and numbing of general responsiveness (not present before the trauma), as indicated by ≥3 of the following:
    1. Efforts to avoid thoughts, feelings, or conversations associated with the trauma
    2. Efforts to avoid activities, places, or people that arouse recollections of the trauma
    3. Inability to recall an important aspect of the trauma
    4. Markedly diminished interest or participation in significant activities
    5. Feeling of detachment or estrangement from others
    6. Restricted range of affect
    7. Sense of a foreshortened future
  4. Persistent symptoms of increased arousal (not present before the trauma), as indicated by ≥2 of the following:
    1. Difficulty falling or staying asleep
    2. Irritability or outbursts of anger
    3. Difficulty concentrating
    4. Hypervigilance
    5. Exaggerated startle response
  5. Duration of disturbance (symptoms in Criteria B, C, and D) is >1 month
  6. The disturbance causes clinically significant distress or impairment of social, occupational, or other important areas of functioning
Source: Diagnostic and statistical manual of mental disorders, 4th ed, text rev. Washington, DC: American Psychiatric Association; 2000

Nightmares and other sleep disturbances are associated with significant distress and daytime impairment and can interfere with PTSD recovery4-8 by disrupting sleep-dependent processing of emotional experiences and causing repeated resensitization to trauma cues (Table 2).8

Table 2

Psychosocial consequences of sleep disruption in PTSD

 

Increased reactivity to emotional cues
Compromised ability to function in social and occupational roles
Negative psychiatric outcomes, including suicidal ideation or worsening of depression or psychosis
Interference of natural recovery from trauma exposure
Repeated resensitization to trauma cues
Neurocognitive deficits
Neuroendocrine abnormalities
PTSD: posttraumatic stress disorder
Source: Adapted from reference 8

Few randomized controlled medication trials specifically address PTSD-related nightmares. Most PTSD studies do not examine sleep outcomes as a primary measure, and comprehensive literature reviews could not offer evidence-based recommendations.9,10 The American Academy of Sleep Medicine (AASM) also noted a paucity of PTSD studies that identified nightmares as a primary outcome measure.11 See Table 3 for a list of recommended medication options for PTSD-associated nightmares.

 

 

Table 3

Recommended medication treatments for PTSD-associated nightmares

 

Evidence levelMedicationEvidence
Recommended for treating PTSD-associated nightmares
1, 4PrazosinIn 3 level 1 studies, adding prazosin (mean dose 3 mg/d) significantly decreased trauma-related nightmares according to the CAPS “recurrent distressing dreams” item after 3 to 9 weeks of treatment vs placebo in veteran and civilian patients (N = 57)
Not suggested for treating PTSD-associated nightmares
1VenlafaxineNo difference between extended-release venlafaxine (37.5 to 300 mg/d) and placebo in the CAPS-SX17 “distressing dreams” item at 12 weeks in 340 PTSD patients
May be considered for treating PTSD-associated nightmares
4ClonidineReduced the number of nightmares in 11 of 13 refugees for 2 weeks to 3 months (dose: 0.2 to 0.6 mg/d)
May be considered for treating PTSD-associated nightmares, but data are low grade and sparse
4TrazodoneAlthough trazodone (25 to 600 mg) significantly decreased nightmare frequency in veteran patients during an 8-week hospital stay (N = 60), 19% discontinued therapy because of side effects
4OlanzapineAdjunctive olanzapine (10 to 20 mg) rapidly improved sleep in a case series of combat-related PTSD patients resistant to SSRIs and benzodiazepines (N = 5)
4RisperidoneIn case series, risperidone (0.5 to 3 mg) significantly decreased CAPS scores for recurrent distressing dreams and proportion of traumatic dreams documented in diaries of combat veterans over 6 weeks (N = 17), and improved nightmares in adult burn patients taking pain medications after 1 to 2 days (N = 10)
4AripiprazoleIn a case series, aripiprazole (15 to 30 mg at bedtime) with CBT or sertraline significantly improved nightmares in 4 of 5 combat-related PTSD patients
4TopiramateTopiramate reduced nightmares in 79% of civilians with PTSD and fully suppressed nightmares in 50% of patients in a case series (N = 35)
4Low-dose cortisolSignificant decrease in frequency but not intensity of nightmares with low-dose cortisol (10 mg/d) in civilians with PTSD (N = 3)
4FluvoxamineIn 2 case series, fluvoxamine (up to 300 mg/d) significantly decreased the IES-R level of “dreams about combat trauma” but not the SRRS “bad dreams” rating at 10 weeks (N = 21). During 4 to 12 weeks of follow-up there was a qualitative decrease in reported nightmares in veteran patients (n = 12)
2Triazolam/nitrazepamLimited data showed triazolam (0.5 mg) and nitrazepam (5 mg) provide equal efficacy in decreasing the number of patients who experience unpleasant dreams over 1 night
4PhenelzineOne study showed phenelzine monotherapy (30 to 90 mg) resulted in elimination of nightmares within 1 month (N = 5); another reported “moderately reduced traumatic dreams” (N = 21) in veterans. Therapy was discontinued because of short-lived efficacy or plateau effect
4GabapentinAdjunctive gabapentin (300 to 3,600 mg/d) improved insomnia and decreased nightmare frequency and/or intensity over 1 to 36 months in 30 veterans with PTSD
4CyproheptadineConflicting data ranges from eliminating nightmares to no changes in the presence or intensity of nightmares
4TCAsAmong 10 Cambodian concentration camp survivors treated with TCAs, 4 reported their nightmares ceased and 4 reported improvement after 1-year follow-up
4NefazodoneReduced nightmare occurrence in 3 open-label studies as monotherapy (386 to 600 mg/d). Not recommended first line because of hepatotoxicity risk
No recommendation because of sparse data
2ClonazepamClonazepam (1 to 2 mg/d) was ineffective in decreasing frequency or intensity of combat-related PTSD nightmares in veterans (N = 6)
Evidence levels:
  1. High-quality randomized clinical trials with narrow confidence intervals
  2. Low-quality randomized clinical trials or high-quality cohort studies
  3. Case-control studies
  4. Case series; poor case-control studies; poor cohort studies; case reports
CAPS: Clinician-Administered PTSD Scale; CAPS-SX17: 17-item Clinician-Administered PTSD Scale; CBT: cognitive-behavioral therapy; IES-R: Impact of Event Scale-Revised; PTSD: posttraumatic stress disorder; SRRS: Stress Response Rating Scale; SSRI: selective serotonin reuptake inhibitor; TCAs: tricyclic antidepressants
Source: Adapted from Aurora RN, Zak RS, Auerbach SH, et al. Best practice guide for the treatment of nightmare disorder in adults. J Clin Sleep Med. 2010;6(4):389-401

CASE CONTINUED: Medication change, improvement

After reviewing AASM’s treatment recommendations, we prescribe prazosin, 1 mg at bedtime for 7 nights, then increase by 1 mg at bedtime each week until Mr. S’s nightmares improve. He reports a substantial improvement in nightmare severity and frequency after a few weeks of treatment with prazosin, 5 mg at bedtime.

Prazosin

 

Prazosin is an α1-adrenergic receptor antagonist with good CNS penetrability. The rationale for reducing adrenergic activity to address intrusive PTSD symptoms has been well documented.12,13 In open-label trials,14-18 a chart review,19 and placebo-controlled trials,20-22prazosin reduced trauma nightmares and improved sleep quality and global clinical status more than placebo (Table 4). In these studies, prazosin doses ranged from 1 to 20 mg/d, with an average of 3 mg at bedtime and a starting dose of 1 mg. Prazosin is the only agent recommended in the AASM’s Best Practice Guide for treating PTSD-related nightmares.11

 

 

Table 4

RCTs of prazosin for trauma-related nightmares

 

StudyDesignPatientsResults
Raskind et al, 20032020-week, double-blind, placebo-controlled, crossover study (mean dose 9.5 mg/d at bedtime)10 Vietnam veterans with chronic PTSD and severe trauma-related nightmaresPrazosin was superior to placebo on scores on the recurrent distressing dreams item and difficulty falling/staying asleep item of the CAPS and change in PTSD severity and functional status on the CGI-C
Raskind et al, 2007218-week, placebo-controlled, parallel study (mean dose 13.3 ± 3 mg/d in the evening)40 veterans with chronic PTSD, distressing trauma nightmares, and sleep disturbancePrazosin was superior to placebo in reducing trauma nightmares and improving sleep quality and global clinical status; prazosin also shifted dream characteristics of trauma-related nightmares to those typical of normal dreams
Taylor et al, 2008227-week, randomized, placebo-controlled, crossover trial (mean dose 3.1 ± 1.3 mg)13 outpatients with chronic civilian trauma PTSD, frequent nightmares, and sleep disturbancePrazosin significantly increased total sleep time and REM sleep time; reduced trauma-related nightmares, distressed awakenings, and total PCL-C scores; improved CGI-I scores; and changed PDRS scores toward normal dreaming
CAPS: Clinician-Administered PTSD Scale; CGI-C: Clinical Global Impression of Change; CGI-I: Clinical Global Impression of Improvement; PCL-C: PTSD Checklist-Civilian; PDRS: PTSD Dream Rating Scale; PTSD: posttraumatic stress disorder; RCTs: randomized controlled trials; REM: rapid eye movement

Atypical antipsychotics

Atypical antipsychotics have been used to reduce nightmares in PTSD; however, most of the evidence from studies evaluated in the AASM’s Best Practice Guide were considered to be low quality.11 Quetiapine and ziprasidone were not included in the AASM review. See (Table 5) for a review of the evidence for atypical antipsychotics for treating PTSD nightmares.

Table 5

Combat-related nightmares: Evidence for atypical antipsychotics

 

StudyDesignPatients/dosageResults
Aripiprazole
Lambert, 2006 aCase report4 veterans with combat-related PTSD (3 male, 1 female; age 22 to 24); dose: 15 to 30 mg; concurrent treatment sertraline or CBTDecreased frequency of weekly nightmares and agitated sleep by at least 50%
Olanzapine
Stein et al, 2002 b8-week, double-blind, placebo-controlled study19 male veterans with combat-related PTSD (olanzapine group mean age: 55.2 ± 6.6; placebo group 51.1 ± 8.1); mean dose: 15 mg/dSignificantly greater reduction in sleep disturbances (PSQI: -3.29 vs 1.57; P = .01); significantly higher weight gain (13.2 lbs vs -3 lbs; P = .001)
Jakovljevic et al, 2003 cCase reports5 veterans with combat-related PTSD for 6 to 7 years (age: 28 to 50); dose: 10 to 20 mg; adjunct treatmentDecreased frequency of nightmares within 3 days
Labbate et al, 2000 dCase report1 male veteran (age: 58) with a 20-year history of combat-related PTSD; dose: 5 mg at bedtime; concurrent treatment with sertraline (200 mg/d), bupropion (150 mg/d), and diazepam (15 mg/d)Eliminated nightmares after 1 week and improved sleep quality
Quetiapine
Ahearn et al, 2006 e8-week, open-label trial15 PTSD patients (8 male; 7 female; 5 with combat-related PTSD; mean age: 49); mean dose: 216 mg/d (100 to 400 mg/d)Significantly improved re-experiencing (CAPS: 10 vs 23; P = .0012) and sleep (PSQI: 17.5 vs 30; P = .0044) at 8 weeks compared with baseline
Robert et al, 2005 f6-week, open-label trial19 combat veterans; mean dose: 100 ± 70 mg/d (25 to 300 mg/d); adjunct treatmentSignificantly improved sleep quality (PSQI: 1.67 vs 2.41; P = .006), latency (PSQI: 1.5 vs 2.65; P = .002), duration (PSQI: 1.31 vs 2.71; P < .001), and sleep disturbances (PSQI: 1.22 vs 1.71; P = .034) and decreased terror episodes (PSQI-A: 0.73 vs 0.91; P = .040) and acting out dreams (PSQI-A: 1.07 vs 1.35; P = .013); however, no difference in nightmares caused by trauma (PSQI-A: 1.53 vs 2.06)
Sokolski et al, 2003 gRetrospective chart review68 male Vietnam War combat veterans (mean age: 55 ± 3.5); mean dose: 155 ± 130 mg (25 to 700 mg); adjunct treatmentImproved sleep disturbances in 62% and nightmares in 25% of patients
Ahearn et al, 2003 hCase report2 male patients with combat-related PTSD (age 53, 72); dose: 25 to 50 mg; adjunct to SSRI therapyDecreased frequency of nightmares with increased sleep duration
Risperidone
David et al, 2006 i6-week, open-label trial17 male veterans with combat-related PTSD (mean age: 53.7 ± 3.8); mean maximum dose: 2.3 ± 0.6 mg (range: 1 to 3 mg)Improved recurrent distressing dreams (CAPS B-2: 3.8 vs 5.4; P = .04), but not with the PSQI subscale (PSQI bad dreams: 2.5 vs 2.7; NS). Decreased nighttime awakenings (1.9 vs 2.8; P = .003) and trauma dreams (19% vs 38%; P = .04)
Leyba et al, 1998 jCase reports3 male patients (age 43 to 46); dose: 1 to 3 mg; adjunct therapyDecreased occurrence of nightmares
Ziprasidone
Siddiqui et al, 2005 kCase report1 male veteran with chronic combat-related PTSD (age 55); dose: 80 to 120 mg/d; adjunct with trazodone (100 mg) and topiramateImproved occurrence of nightmares up to 4 months
CAPS: Clinician-Administered PTSD Scale; CAPS B-2: Clinician-Administered PTSD Scale B-2 (recurrent distressing dreams of the event); CBT: cognitive-behavioral therapy; PSQI: Pittsburgh Sleep Quality Index; PSQI-A: Pittsburgh Sleep Quality Index Addendum for PTSD; NS: not significant; PTSD: posttraumatic stress disorder; SSRI: selective serotonin reuptake inhibitor References
  1. Lambert MT. Aripiprazole in the management of post-traumatic stress disorder symptoms in returning Global War on Terrorism veterans. Int Clin Psychopharmacol. 2006;21(3):185-187.
  2. Stein MB, Kline NA, Matloff JL. Adjunctive olanzapine for SSRI-resistant combat-related PTSD: a double-blind, placebo-controlled study. Am J Psychiatry. 2002;159(10):1777-1779.
  3. Jakovljevic M, Sagud M, Mihaljevic-Peles A. Olanzapine in the treatment-resistant, combat-related PTSD—a series of case reports. Acta Psychiatr Scand. 2003;107(5):394-396.
  4. Labbate LA, Douglas S. Olanzapine for nightmares and sleep disturbance in posttraumatic stress disorder (PTSD). Can J Psychiatry. 2000;45(7):667-668.
  5. Ahearn EP, Mussey M, Johnson C, et al. Quetiapine as an adjunctive treatment for post-traumatic stress disorder: an 8-week open-label study. Int Clin Psychopharmacol. 2006;21(1):29-33.
  6. Robert S, Hamner MB, Kose S, et al. Quetiapine improves sleep disturbances in combat veterans with PTSD: sleep data from a prospective, open-label study. J Clin Psychopharmacol. 2005;25(4):387-388.
  7. Sokolski KN, Denson TF, Lee RT, et al. Quetiapine for treatment of refractory symptoms of combat-related post-traumatic stress disorder. Mil Med. 2003;168(6):486-489.
  8. Ahearn EP, Winston E, Mussey M, et al. Atypical antipsychotics, improved intrusive symptoms in patients with posttraumatic stress disorder. Mil Med. 2003;168(9):x-xi.
  9. David D, De Faria L, Mellman TA. Adjunctive risperidone treatment and sleep symptoms in combat veterans with chronic PTSD. Depress Anxiety. 2006;23(8):489-491.
  10. Leyba CM, Wampler TP. Risperidone in PTSD. Psychiatr Serv. 1998;49(2):245-246.
  11. Siddiqui Z, Marcil WA, Bhatia SC, et al. Ziprasidone therapy for post-traumatic stress disorder. J Psychiatry Neurosci. 2005;30(6):430-431.
 

 

 

Comparing prazosin and quetiapine. A historical prospective cohort study of 237 veterans with PTSD receiving prazosin or quetiapine for nighttime PTSD symptoms demonstrated that although the 2 drugs have similar efficacy (defined as symptomatic improvement) for short-term, 6-month treatment (61% vs 62%; P=.54), a higher percentage of patients continued prazosin long-term (3 to 6 years) than those taking quetiapine (48% vs 24%; P < .001).23 Twenty-five percent of patients taking quetiapine switched to prazosin during the study, and approximately one-half of these patients remained on prazosin until the study’s end. Only 8% of prazosin patients switched to quetiapine, and none continued this therapy until study end.23 Patients in the quetiapine group were more likely to discontinue the drug because of lack of efficacy (13% vs 3%; P=.03) and adverse effects (35% vs 18%; P=.008), specifically sedation (21% vs 2%; P < .001) and metabolic effects (9% vs 0%; P=.014), compared with prazosin. Although this trial may be the only published comparison study of prazosin and quetiapine, its methodological quality has been questioned, which makes it difficult to draw definitive conclusions.

Metabolic syndrome—elevated diastolic blood pressure, increased waist circumference, and low high-density lipoprotein cholesterol—is common among PTSD patients treated with antipsychotics.24 This syndrome may be caused by medications, lifestyle factors, or long-term overactivation of stress-response pathways. A retrospective chart review at a community mental health center revealed that patients taking even low doses of quetiapine for insomnia gained an average of 5 lbs (P=.037).25 Another retrospective chart review at 2 military hospitals reported that patients receiving low-dose quetiapine (≤100 mg/d) gained an average of slightly less than 1 lb per month, which adds up to approximately 10 lbs per year (P < .001).26 The benefit of using atypical antipsychotics may be outweighed by metabolic risks such as obesity, new-onset diabetes, and dyslipidemia.27

 

Prazosin is considered a first-line treatment for sleep disturbances and nightmares in PTSD because of its superior long-term efficacy and decreased adverse effects compared with quetiapine.

Related Resources

 

  • American Psychiatric Association. Practice guidelines for the treatment of patients with acute stress disorder and posttraumatic stress disorder. Arlington, VA: American Psychiatric Publishing, Inc.; 2004.
  • Veterans Affairs/Department of Defense clinical practice guidelines. Management of traumatic stress disorder and acute stress reaction. www.healthquality.va.gov/Post_Traumatic_Stress_Disorder_PTSD.asp.

Drug Brand Names

 

  • Prazosin • Minipress
  • Quetiapine • Seroquel
  • Sertraline • Zoloft
  • Ziprasidone • Geodon

Disclosure

The authors report no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.

 

Practice Points

• Prazosin is recommended as a first-line therapy for nighttime PTSD symptoms, such as nightmares or sleep disturbances—especially among veterans—because of superior long-term effectiveness.

Risk of metabolic syndrome, which has been reported with low-dose atypical antipsychotics used for treating insomnia, limits their use for PTSD-related nightmares.

Mr. S, a 45-year-old veteran, was diagnosed with posttraumatic stress disorder (PTSD) 18 years ago after a tour of duty in the Persian Gulf. He had combat-related flashbacks triggered by the smell of gasoline or smoke from a fire, was easily startled, and began to isolate himself socially. However, his symptoms improved when he started volunteering at his local Veterans Affairs Medical Center. After he lost his job 3 years ago, Mr. S started experiencing flashbacks. He was irritable, easily startled, and avoided things that reminded him of his time in the Persian Gulf. His psychiatrist prescribed sertraline, titrated to 200 mg/d. The drug reduced the severity of his avoidance and hyperarousal symptoms and improved his mood.

During a clinic visit, Mr. S says he is doing well and can fall asleep at night but is having recurring nightmares about traumatic events that occurred during combat. These nightmares wake him up and have become more frequent, occurring once per night for the past month. Mr. S says he has been watching more news programs about conflicts in Afghanistan and Iraq since the nightmares began. His psychiatrist starts quetiapine, 50 mg at bedtime for 7 nights then 100 mg at bedtime, but after 6 weeks Mr. S says his nightmares continue.

PTSD occurs in approximately 19% of Vietnam war combat veterans1 and 14% of service members returning from Iraq and Afghanistan.2 PTSD symptoms are classified into clusters: intrusive/re-experiencing; avoidant/numbing; and hyperarousal.3 Nightmares are part of the intrusive/re-experiencing cluster, which is Criterion B in DSM-IV-TR. See Table 1 for a description of DSM-IV-TR PTSD criteria. Among PTSD patients, 50% to 70% report PTSD-associated nightmares.4 Despite adequate treatment targeted to improve PTSD’s core symptoms, symptoms such as sleep disturbances or nightmares often persist.

Table 1

DSM-IV-TR diagnostic criteria for posttraumatic stress disorder

 

  1. The person has been exposed to a traumatic event in which both of the following were present:
    1. The person experienced, witnessed, or was confronted with an event or events that involved actual or threatened death or serious injury, or a threat to the physical integrity of self or others
    2. The person’s response involved intense fear, helplessness, or horror
  2. The traumatic event is persistently reexperienced in ≥1 of the following ways:
    1. Recurrent and intrusive distressing recollections of the event
    2. Recurrent distressing dreams of the event
    3. Acting or feeling as if the traumatic event were recurring
    4. Intense psychological distress at exposure to internal or external cues that symbolize or resemble an aspect of the traumatic event
    5. Physiological reactivity on exposure to internal or external cues that symbolize or resemble an aspect of the traumatic event
  3. Persistent avoidance of stimuli associated with the trauma and numbing of general responsiveness (not present before the trauma), as indicated by ≥3 of the following:
    1. Efforts to avoid thoughts, feelings, or conversations associated with the trauma
    2. Efforts to avoid activities, places, or people that arouse recollections of the trauma
    3. Inability to recall an important aspect of the trauma
    4. Markedly diminished interest or participation in significant activities
    5. Feeling of detachment or estrangement from others
    6. Restricted range of affect
    7. Sense of a foreshortened future
  4. Persistent symptoms of increased arousal (not present before the trauma), as indicated by ≥2 of the following:
    1. Difficulty falling or staying asleep
    2. Irritability or outbursts of anger
    3. Difficulty concentrating
    4. Hypervigilance
    5. Exaggerated startle response
  5. Duration of disturbance (symptoms in Criteria B, C, and D) is >1 month
  6. The disturbance causes clinically significant distress or impairment of social, occupational, or other important areas of functioning
Source: Diagnostic and statistical manual of mental disorders, 4th ed, text rev. Washington, DC: American Psychiatric Association; 2000

Nightmares and other sleep disturbances are associated with significant distress and daytime impairment and can interfere with PTSD recovery4-8 by disrupting sleep-dependent processing of emotional experiences and causing repeated resensitization to trauma cues (Table 2).8

Table 2

Psychosocial consequences of sleep disruption in PTSD

 

Increased reactivity to emotional cues
Compromised ability to function in social and occupational roles
Negative psychiatric outcomes, including suicidal ideation or worsening of depression or psychosis
Interference of natural recovery from trauma exposure
Repeated resensitization to trauma cues
Neurocognitive deficits
Neuroendocrine abnormalities
PTSD: posttraumatic stress disorder
Source: Adapted from reference 8

Few randomized controlled medication trials specifically address PTSD-related nightmares. Most PTSD studies do not examine sleep outcomes as a primary measure, and comprehensive literature reviews could not offer evidence-based recommendations.9,10 The American Academy of Sleep Medicine (AASM) also noted a paucity of PTSD studies that identified nightmares as a primary outcome measure.11 See Table 3 for a list of recommended medication options for PTSD-associated nightmares.

 

 

Table 3

Recommended medication treatments for PTSD-associated nightmares

 

Evidence levelMedicationEvidence
Recommended for treating PTSD-associated nightmares
1, 4PrazosinIn 3 level 1 studies, adding prazosin (mean dose 3 mg/d) significantly decreased trauma-related nightmares according to the CAPS “recurrent distressing dreams” item after 3 to 9 weeks of treatment vs placebo in veteran and civilian patients (N = 57)
Not suggested for treating PTSD-associated nightmares
1VenlafaxineNo difference between extended-release venlafaxine (37.5 to 300 mg/d) and placebo in the CAPS-SX17 “distressing dreams” item at 12 weeks in 340 PTSD patients
May be considered for treating PTSD-associated nightmares
4ClonidineReduced the number of nightmares in 11 of 13 refugees for 2 weeks to 3 months (dose: 0.2 to 0.6 mg/d)
May be considered for treating PTSD-associated nightmares, but data are low grade and sparse
4TrazodoneAlthough trazodone (25 to 600 mg) significantly decreased nightmare frequency in veteran patients during an 8-week hospital stay (N = 60), 19% discontinued therapy because of side effects
4OlanzapineAdjunctive olanzapine (10 to 20 mg) rapidly improved sleep in a case series of combat-related PTSD patients resistant to SSRIs and benzodiazepines (N = 5)
4RisperidoneIn case series, risperidone (0.5 to 3 mg) significantly decreased CAPS scores for recurrent distressing dreams and proportion of traumatic dreams documented in diaries of combat veterans over 6 weeks (N = 17), and improved nightmares in adult burn patients taking pain medications after 1 to 2 days (N = 10)
4AripiprazoleIn a case series, aripiprazole (15 to 30 mg at bedtime) with CBT or sertraline significantly improved nightmares in 4 of 5 combat-related PTSD patients
4TopiramateTopiramate reduced nightmares in 79% of civilians with PTSD and fully suppressed nightmares in 50% of patients in a case series (N = 35)
4Low-dose cortisolSignificant decrease in frequency but not intensity of nightmares with low-dose cortisol (10 mg/d) in civilians with PTSD (N = 3)
4FluvoxamineIn 2 case series, fluvoxamine (up to 300 mg/d) significantly decreased the IES-R level of “dreams about combat trauma” but not the SRRS “bad dreams” rating at 10 weeks (N = 21). During 4 to 12 weeks of follow-up there was a qualitative decrease in reported nightmares in veteran patients (n = 12)
2Triazolam/nitrazepamLimited data showed triazolam (0.5 mg) and nitrazepam (5 mg) provide equal efficacy in decreasing the number of patients who experience unpleasant dreams over 1 night
4PhenelzineOne study showed phenelzine monotherapy (30 to 90 mg) resulted in elimination of nightmares within 1 month (N = 5); another reported “moderately reduced traumatic dreams” (N = 21) in veterans. Therapy was discontinued because of short-lived efficacy or plateau effect
4GabapentinAdjunctive gabapentin (300 to 3,600 mg/d) improved insomnia and decreased nightmare frequency and/or intensity over 1 to 36 months in 30 veterans with PTSD
4CyproheptadineConflicting data ranges from eliminating nightmares to no changes in the presence or intensity of nightmares
4TCAsAmong 10 Cambodian concentration camp survivors treated with TCAs, 4 reported their nightmares ceased and 4 reported improvement after 1-year follow-up
4NefazodoneReduced nightmare occurrence in 3 open-label studies as monotherapy (386 to 600 mg/d). Not recommended first line because of hepatotoxicity risk
No recommendation because of sparse data
2ClonazepamClonazepam (1 to 2 mg/d) was ineffective in decreasing frequency or intensity of combat-related PTSD nightmares in veterans (N = 6)
Evidence levels:
  1. High-quality randomized clinical trials with narrow confidence intervals
  2. Low-quality randomized clinical trials or high-quality cohort studies
  3. Case-control studies
  4. Case series; poor case-control studies; poor cohort studies; case reports
CAPS: Clinician-Administered PTSD Scale; CAPS-SX17: 17-item Clinician-Administered PTSD Scale; CBT: cognitive-behavioral therapy; IES-R: Impact of Event Scale-Revised; PTSD: posttraumatic stress disorder; SRRS: Stress Response Rating Scale; SSRI: selective serotonin reuptake inhibitor; TCAs: tricyclic antidepressants
Source: Adapted from Aurora RN, Zak RS, Auerbach SH, et al. Best practice guide for the treatment of nightmare disorder in adults. J Clin Sleep Med. 2010;6(4):389-401

CASE CONTINUED: Medication change, improvement

After reviewing AASM’s treatment recommendations, we prescribe prazosin, 1 mg at bedtime for 7 nights, then increase by 1 mg at bedtime each week until Mr. S’s nightmares improve. He reports a substantial improvement in nightmare severity and frequency after a few weeks of treatment with prazosin, 5 mg at bedtime.

Prazosin

 

Prazosin is an α1-adrenergic receptor antagonist with good CNS penetrability. The rationale for reducing adrenergic activity to address intrusive PTSD symptoms has been well documented.12,13 In open-label trials,14-18 a chart review,19 and placebo-controlled trials,20-22prazosin reduced trauma nightmares and improved sleep quality and global clinical status more than placebo (Table 4). In these studies, prazosin doses ranged from 1 to 20 mg/d, with an average of 3 mg at bedtime and a starting dose of 1 mg. Prazosin is the only agent recommended in the AASM’s Best Practice Guide for treating PTSD-related nightmares.11

 

 

Table 4

RCTs of prazosin for trauma-related nightmares

 

StudyDesignPatientsResults
Raskind et al, 20032020-week, double-blind, placebo-controlled, crossover study (mean dose 9.5 mg/d at bedtime)10 Vietnam veterans with chronic PTSD and severe trauma-related nightmaresPrazosin was superior to placebo on scores on the recurrent distressing dreams item and difficulty falling/staying asleep item of the CAPS and change in PTSD severity and functional status on the CGI-C
Raskind et al, 2007218-week, placebo-controlled, parallel study (mean dose 13.3 ± 3 mg/d in the evening)40 veterans with chronic PTSD, distressing trauma nightmares, and sleep disturbancePrazosin was superior to placebo in reducing trauma nightmares and improving sleep quality and global clinical status; prazosin also shifted dream characteristics of trauma-related nightmares to those typical of normal dreams
Taylor et al, 2008227-week, randomized, placebo-controlled, crossover trial (mean dose 3.1 ± 1.3 mg)13 outpatients with chronic civilian trauma PTSD, frequent nightmares, and sleep disturbancePrazosin significantly increased total sleep time and REM sleep time; reduced trauma-related nightmares, distressed awakenings, and total PCL-C scores; improved CGI-I scores; and changed PDRS scores toward normal dreaming
CAPS: Clinician-Administered PTSD Scale; CGI-C: Clinical Global Impression of Change; CGI-I: Clinical Global Impression of Improvement; PCL-C: PTSD Checklist-Civilian; PDRS: PTSD Dream Rating Scale; PTSD: posttraumatic stress disorder; RCTs: randomized controlled trials; REM: rapid eye movement

Atypical antipsychotics

Atypical antipsychotics have been used to reduce nightmares in PTSD; however, most of the evidence from studies evaluated in the AASM’s Best Practice Guide were considered to be low quality.11 Quetiapine and ziprasidone were not included in the AASM review. See (Table 5) for a review of the evidence for atypical antipsychotics for treating PTSD nightmares.

Table 5

Combat-related nightmares: Evidence for atypical antipsychotics

 

StudyDesignPatients/dosageResults
Aripiprazole
Lambert, 2006 aCase report4 veterans with combat-related PTSD (3 male, 1 female; age 22 to 24); dose: 15 to 30 mg; concurrent treatment sertraline or CBTDecreased frequency of weekly nightmares and agitated sleep by at least 50%
Olanzapine
Stein et al, 2002 b8-week, double-blind, placebo-controlled study19 male veterans with combat-related PTSD (olanzapine group mean age: 55.2 ± 6.6; placebo group 51.1 ± 8.1); mean dose: 15 mg/dSignificantly greater reduction in sleep disturbances (PSQI: -3.29 vs 1.57; P = .01); significantly higher weight gain (13.2 lbs vs -3 lbs; P = .001)
Jakovljevic et al, 2003 cCase reports5 veterans with combat-related PTSD for 6 to 7 years (age: 28 to 50); dose: 10 to 20 mg; adjunct treatmentDecreased frequency of nightmares within 3 days
Labbate et al, 2000 dCase report1 male veteran (age: 58) with a 20-year history of combat-related PTSD; dose: 5 mg at bedtime; concurrent treatment with sertraline (200 mg/d), bupropion (150 mg/d), and diazepam (15 mg/d)Eliminated nightmares after 1 week and improved sleep quality
Quetiapine
Ahearn et al, 2006 e8-week, open-label trial15 PTSD patients (8 male; 7 female; 5 with combat-related PTSD; mean age: 49); mean dose: 216 mg/d (100 to 400 mg/d)Significantly improved re-experiencing (CAPS: 10 vs 23; P = .0012) and sleep (PSQI: 17.5 vs 30; P = .0044) at 8 weeks compared with baseline
Robert et al, 2005 f6-week, open-label trial19 combat veterans; mean dose: 100 ± 70 mg/d (25 to 300 mg/d); adjunct treatmentSignificantly improved sleep quality (PSQI: 1.67 vs 2.41; P = .006), latency (PSQI: 1.5 vs 2.65; P = .002), duration (PSQI: 1.31 vs 2.71; P < .001), and sleep disturbances (PSQI: 1.22 vs 1.71; P = .034) and decreased terror episodes (PSQI-A: 0.73 vs 0.91; P = .040) and acting out dreams (PSQI-A: 1.07 vs 1.35; P = .013); however, no difference in nightmares caused by trauma (PSQI-A: 1.53 vs 2.06)
Sokolski et al, 2003 gRetrospective chart review68 male Vietnam War combat veterans (mean age: 55 ± 3.5); mean dose: 155 ± 130 mg (25 to 700 mg); adjunct treatmentImproved sleep disturbances in 62% and nightmares in 25% of patients
Ahearn et al, 2003 hCase report2 male patients with combat-related PTSD (age 53, 72); dose: 25 to 50 mg; adjunct to SSRI therapyDecreased frequency of nightmares with increased sleep duration
Risperidone
David et al, 2006 i6-week, open-label trial17 male veterans with combat-related PTSD (mean age: 53.7 ± 3.8); mean maximum dose: 2.3 ± 0.6 mg (range: 1 to 3 mg)Improved recurrent distressing dreams (CAPS B-2: 3.8 vs 5.4; P = .04), but not with the PSQI subscale (PSQI bad dreams: 2.5 vs 2.7; NS). Decreased nighttime awakenings (1.9 vs 2.8; P = .003) and trauma dreams (19% vs 38%; P = .04)
Leyba et al, 1998 jCase reports3 male patients (age 43 to 46); dose: 1 to 3 mg; adjunct therapyDecreased occurrence of nightmares
Ziprasidone
Siddiqui et al, 2005 kCase report1 male veteran with chronic combat-related PTSD (age 55); dose: 80 to 120 mg/d; adjunct with trazodone (100 mg) and topiramateImproved occurrence of nightmares up to 4 months
CAPS: Clinician-Administered PTSD Scale; CAPS B-2: Clinician-Administered PTSD Scale B-2 (recurrent distressing dreams of the event); CBT: cognitive-behavioral therapy; PSQI: Pittsburgh Sleep Quality Index; PSQI-A: Pittsburgh Sleep Quality Index Addendum for PTSD; NS: not significant; PTSD: posttraumatic stress disorder; SSRI: selective serotonin reuptake inhibitor References
  1. Lambert MT. Aripiprazole in the management of post-traumatic stress disorder symptoms in returning Global War on Terrorism veterans. Int Clin Psychopharmacol. 2006;21(3):185-187.
  2. Stein MB, Kline NA, Matloff JL. Adjunctive olanzapine for SSRI-resistant combat-related PTSD: a double-blind, placebo-controlled study. Am J Psychiatry. 2002;159(10):1777-1779.
  3. Jakovljevic M, Sagud M, Mihaljevic-Peles A. Olanzapine in the treatment-resistant, combat-related PTSD—a series of case reports. Acta Psychiatr Scand. 2003;107(5):394-396.
  4. Labbate LA, Douglas S. Olanzapine for nightmares and sleep disturbance in posttraumatic stress disorder (PTSD). Can J Psychiatry. 2000;45(7):667-668.
  5. Ahearn EP, Mussey M, Johnson C, et al. Quetiapine as an adjunctive treatment for post-traumatic stress disorder: an 8-week open-label study. Int Clin Psychopharmacol. 2006;21(1):29-33.
  6. Robert S, Hamner MB, Kose S, et al. Quetiapine improves sleep disturbances in combat veterans with PTSD: sleep data from a prospective, open-label study. J Clin Psychopharmacol. 2005;25(4):387-388.
  7. Sokolski KN, Denson TF, Lee RT, et al. Quetiapine for treatment of refractory symptoms of combat-related post-traumatic stress disorder. Mil Med. 2003;168(6):486-489.
  8. Ahearn EP, Winston E, Mussey M, et al. Atypical antipsychotics, improved intrusive symptoms in patients with posttraumatic stress disorder. Mil Med. 2003;168(9):x-xi.
  9. David D, De Faria L, Mellman TA. Adjunctive risperidone treatment and sleep symptoms in combat veterans with chronic PTSD. Depress Anxiety. 2006;23(8):489-491.
  10. Leyba CM, Wampler TP. Risperidone in PTSD. Psychiatr Serv. 1998;49(2):245-246.
  11. Siddiqui Z, Marcil WA, Bhatia SC, et al. Ziprasidone therapy for post-traumatic stress disorder. J Psychiatry Neurosci. 2005;30(6):430-431.
 

 

 

Comparing prazosin and quetiapine. A historical prospective cohort study of 237 veterans with PTSD receiving prazosin or quetiapine for nighttime PTSD symptoms demonstrated that although the 2 drugs have similar efficacy (defined as symptomatic improvement) for short-term, 6-month treatment (61% vs 62%; P=.54), a higher percentage of patients continued prazosin long-term (3 to 6 years) than those taking quetiapine (48% vs 24%; P < .001).23 Twenty-five percent of patients taking quetiapine switched to prazosin during the study, and approximately one-half of these patients remained on prazosin until the study’s end. Only 8% of prazosin patients switched to quetiapine, and none continued this therapy until study end.23 Patients in the quetiapine group were more likely to discontinue the drug because of lack of efficacy (13% vs 3%; P=.03) and adverse effects (35% vs 18%; P=.008), specifically sedation (21% vs 2%; P < .001) and metabolic effects (9% vs 0%; P=.014), compared with prazosin. Although this trial may be the only published comparison study of prazosin and quetiapine, its methodological quality has been questioned, which makes it difficult to draw definitive conclusions.

Metabolic syndrome—elevated diastolic blood pressure, increased waist circumference, and low high-density lipoprotein cholesterol—is common among PTSD patients treated with antipsychotics.24 This syndrome may be caused by medications, lifestyle factors, or long-term overactivation of stress-response pathways. A retrospective chart review at a community mental health center revealed that patients taking even low doses of quetiapine for insomnia gained an average of 5 lbs (P=.037).25 Another retrospective chart review at 2 military hospitals reported that patients receiving low-dose quetiapine (≤100 mg/d) gained an average of slightly less than 1 lb per month, which adds up to approximately 10 lbs per year (P < .001).26 The benefit of using atypical antipsychotics may be outweighed by metabolic risks such as obesity, new-onset diabetes, and dyslipidemia.27

 

Prazosin is considered a first-line treatment for sleep disturbances and nightmares in PTSD because of its superior long-term efficacy and decreased adverse effects compared with quetiapine.

Related Resources

 

  • American Psychiatric Association. Practice guidelines for the treatment of patients with acute stress disorder and posttraumatic stress disorder. Arlington, VA: American Psychiatric Publishing, Inc.; 2004.
  • Veterans Affairs/Department of Defense clinical practice guidelines. Management of traumatic stress disorder and acute stress reaction. www.healthquality.va.gov/Post_Traumatic_Stress_Disorder_PTSD.asp.

Drug Brand Names

 

  • Prazosin • Minipress
  • Quetiapine • Seroquel
  • Sertraline • Zoloft
  • Ziprasidone • Geodon

Disclosure

The authors report no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.

References

 

1. Dohrenwend BP, Turner JB, Turse NA, et al. The psychological risks of Vietnam for U.S. veterans: a revisit with new data and methods. Science. 2006;313(5789):979-982.

2. Tanielian T, Jaycox L. eds. Invisible wounds of war: psychological and cognitive injuries, their consequences, and services to assist recovery. Santa Monica, CA: RAND Corporation; 2008.

3. Diagnostic and statistical manual of mental disorders, 4th ed, text rev. Washington DC: American Psychiatric Association; 2000.

4. Wittmann L, Schredl M, Kramer M. Dreaming in posttraumatic stress disorder: a critical review of phenomenology psychophysiology and treatment. Psychother Psychosom. 2007;76(1):25-39.

5. Clum GA, Nishith P, Resick PA. Trauma-related sleep disturbance and self-reported physical health symptoms in treatment-seeking female rape victims. J Nerv Ment Dis. 2001;189(9):618-622.

6. Kramer TL, Booth BM, Han X, et al. Service utilization and outcomes in medically ill veterans with posttraumatic stress and depressive disorders. J Trauma Stress. 2003;16(3):211-219.

7. Neylan TC, Marmar CR, Metzler TJ, et al. Sleep disturbances in the Vietnam generation: findings from a nationally representative sample of male Vietnam veterans. Am J Psychiatry. 1998;155(7):929-933.

8. Nappi CM, Drummond SP, Hall JM. Treating nightmares and insomnia in posttraumatic stress disorder: a review of current evidence. Neuropharmacology. 2012;62(2):576-585.

9. Maher MJ, Rego SA, Asnis GM. Sleep disturbances in patients with post-traumatic stress disorder: epidemiology impact and approaches to management. CNS Drugs. 2006;20(7):567-590.

10. van Liempt S, Vermetten E, Geuze E, et al. Pharmacotherapy for disordered sleep in post-traumatic stress disorder: a systematic review. Int Clin Psychopharmacol. 2006;21(4):193-202.

11. Aurora RN, Zak RS, Auerbach SH, et al. Best practice guide for the treatment of nightmare disorder in adults. J Clin Sleep Med. 2010;6(4):389-401.

12. Boehnlein JK, Kinzie JD. Pharmacologic reduction of CNS noradrenergic activity in PTSD: the case for clonidine and prazosin. J Psychiatr Pract. 2007;13(2):72-78.

13. Strawn JR, Geracioti TD, Jr. Noradrenergic dysfunction and the psychopharmacology of posttraumatic stress disorder. Depress Anxiety. 2008;25(3):260-271.

14. Calohan J, Peterson K, Peskind ER, et al. Prazosin treatment of trauma nightmares and sleep disturbance in soldiers deployed in Iraq. J Trauma Stress. 2010;23(5):645-648.

15. Daly CM, Doyle ME, Radkind M, et al. Clinical case series: the use of Prazosin for combat-related recurrent nightmares among Operation Iraqi Freedom combat veterans. Mil Med. 2005;170(6):513-515.

16. Peskind ER, Bonner LT, Hoff DJ, et al. Prazosin reduces trauma-related nightmares in older men with chronic posttraumatic stress disorder. J Geriatr Psychiatry Neurol. 2003;16(3):165-171.

17. Raskind MA, Dobie DJ, Kanter ED, et al. The alpha1-adrenergic antagonist prazosin ameliorates combat trauma nightmares in veterans with posttraumatic stress disorder: a report of 4 cases. J Clin Psychiatry. 2000;61(2):129-133.

18. Taylor F, Raskind MA. The alpha1-adrenergic antagonist prazosin improves sleep and nightmares in civilian trauma posttraumatic stress disorder. J Clin Psychopharmacol. 2002;22(1):82-85.

19. Raskind MA, Thompson C, Petrie EC, et al. Prazosin reduces nightmares in combat veterans with posttraumatic stress disorder. J Clin Psychiatry. 2002;63(7):565-568.

20. Raskind MA, Peskind ER, Kanter ED, et al. Reduction of nightmares and other PTSD symptoms in combat veterans by prazosin: a placebo-controlled study. Am J Psychiatry. 2003;160(2):371-373.

21. Raskind MA, Peskind ER, Hoff DJ, et al. A parallel group placebo controlled study of prazosin for trauma nightmares and sleep disturbance in combat veterans with post-traumatic stress disorder. Biol Psychiatry. 2007;61(8):928-934.

22. Taylor FB, Martin P, Thompson C, et al. Prazosin effects on objective sleep measures and clinical symptoms in civilian trauma posttraumatic stress disorder: a placebo-controlled study. Biol Psychiatry. 2008;63(6):629-632.

23. Byers MG, Allison KM, Wendel CS, et al. Prazosin versus quetiapine for nighttime posttraumatic stress disorder symptoms in veterans: an assessment of long-term comparative effectiveness and safety. J Clin Psychopharmacol. 2010;30(3):225-229.

24. Jin H, Lanouette NM, Mudaliar S, et al. Association of posttraumatic stress disorder with increased prevalence of metabolic syndrome. J Clin Psychopharmacol. 2009;29(3):210-215.

25. Cates ME, Jackson CW, Feldman JM, et al. Metabolic consequences of using low-dose quetiapine for insomnia in psychiatric patients. Community Ment Health J. 2009;45(4):251-254.

26. Williams SG, Alinejad NA, Williams JA, et al. Statistically significant increase in weight caused by low-dose quetiapine. Pharmacotherapy. 2010;30(10):1011-1015.

27. American Diabetes Association; American Psychiatric Association; American Association of Clinical Endocrinologists; North American Association for the Study of Obesity. Consensus development conference on antipsychotic drugs and obesity and diabetes. J Clin Psychiatry. 2004;65(2):267-272.

References

 

1. Dohrenwend BP, Turner JB, Turse NA, et al. The psychological risks of Vietnam for U.S. veterans: a revisit with new data and methods. Science. 2006;313(5789):979-982.

2. Tanielian T, Jaycox L. eds. Invisible wounds of war: psychological and cognitive injuries, their consequences, and services to assist recovery. Santa Monica, CA: RAND Corporation; 2008.

3. Diagnostic and statistical manual of mental disorders, 4th ed, text rev. Washington DC: American Psychiatric Association; 2000.

4. Wittmann L, Schredl M, Kramer M. Dreaming in posttraumatic stress disorder: a critical review of phenomenology psychophysiology and treatment. Psychother Psychosom. 2007;76(1):25-39.

5. Clum GA, Nishith P, Resick PA. Trauma-related sleep disturbance and self-reported physical health symptoms in treatment-seeking female rape victims. J Nerv Ment Dis. 2001;189(9):618-622.

6. Kramer TL, Booth BM, Han X, et al. Service utilization and outcomes in medically ill veterans with posttraumatic stress and depressive disorders. J Trauma Stress. 2003;16(3):211-219.

7. Neylan TC, Marmar CR, Metzler TJ, et al. Sleep disturbances in the Vietnam generation: findings from a nationally representative sample of male Vietnam veterans. Am J Psychiatry. 1998;155(7):929-933.

8. Nappi CM, Drummond SP, Hall JM. Treating nightmares and insomnia in posttraumatic stress disorder: a review of current evidence. Neuropharmacology. 2012;62(2):576-585.

9. Maher MJ, Rego SA, Asnis GM. Sleep disturbances in patients with post-traumatic stress disorder: epidemiology impact and approaches to management. CNS Drugs. 2006;20(7):567-590.

10. van Liempt S, Vermetten E, Geuze E, et al. Pharmacotherapy for disordered sleep in post-traumatic stress disorder: a systematic review. Int Clin Psychopharmacol. 2006;21(4):193-202.

11. Aurora RN, Zak RS, Auerbach SH, et al. Best practice guide for the treatment of nightmare disorder in adults. J Clin Sleep Med. 2010;6(4):389-401.

12. Boehnlein JK, Kinzie JD. Pharmacologic reduction of CNS noradrenergic activity in PTSD: the case for clonidine and prazosin. J Psychiatr Pract. 2007;13(2):72-78.

13. Strawn JR, Geracioti TD, Jr. Noradrenergic dysfunction and the psychopharmacology of posttraumatic stress disorder. Depress Anxiety. 2008;25(3):260-271.

14. Calohan J, Peterson K, Peskind ER, et al. Prazosin treatment of trauma nightmares and sleep disturbance in soldiers deployed in Iraq. J Trauma Stress. 2010;23(5):645-648.

15. Daly CM, Doyle ME, Radkind M, et al. Clinical case series: the use of Prazosin for combat-related recurrent nightmares among Operation Iraqi Freedom combat veterans. Mil Med. 2005;170(6):513-515.

16. Peskind ER, Bonner LT, Hoff DJ, et al. Prazosin reduces trauma-related nightmares in older men with chronic posttraumatic stress disorder. J Geriatr Psychiatry Neurol. 2003;16(3):165-171.

17. Raskind MA, Dobie DJ, Kanter ED, et al. The alpha1-adrenergic antagonist prazosin ameliorates combat trauma nightmares in veterans with posttraumatic stress disorder: a report of 4 cases. J Clin Psychiatry. 2000;61(2):129-133.

18. Taylor F, Raskind MA. The alpha1-adrenergic antagonist prazosin improves sleep and nightmares in civilian trauma posttraumatic stress disorder. J Clin Psychopharmacol. 2002;22(1):82-85.

19. Raskind MA, Thompson C, Petrie EC, et al. Prazosin reduces nightmares in combat veterans with posttraumatic stress disorder. J Clin Psychiatry. 2002;63(7):565-568.

20. Raskind MA, Peskind ER, Kanter ED, et al. Reduction of nightmares and other PTSD symptoms in combat veterans by prazosin: a placebo-controlled study. Am J Psychiatry. 2003;160(2):371-373.

21. Raskind MA, Peskind ER, Hoff DJ, et al. A parallel group placebo controlled study of prazosin for trauma nightmares and sleep disturbance in combat veterans with post-traumatic stress disorder. Biol Psychiatry. 2007;61(8):928-934.

22. Taylor FB, Martin P, Thompson C, et al. Prazosin effects on objective sleep measures and clinical symptoms in civilian trauma posttraumatic stress disorder: a placebo-controlled study. Biol Psychiatry. 2008;63(6):629-632.

23. Byers MG, Allison KM, Wendel CS, et al. Prazosin versus quetiapine for nighttime posttraumatic stress disorder symptoms in veterans: an assessment of long-term comparative effectiveness and safety. J Clin Psychopharmacol. 2010;30(3):225-229.

24. Jin H, Lanouette NM, Mudaliar S, et al. Association of posttraumatic stress disorder with increased prevalence of metabolic syndrome. J Clin Psychopharmacol. 2009;29(3):210-215.

25. Cates ME, Jackson CW, Feldman JM, et al. Metabolic consequences of using low-dose quetiapine for insomnia in psychiatric patients. Community Ment Health J. 2009;45(4):251-254.

26. Williams SG, Alinejad NA, Williams JA, et al. Statistically significant increase in weight caused by low-dose quetiapine. Pharmacotherapy. 2010;30(10):1011-1015.

27. American Diabetes Association; American Psychiatric Association; American Association of Clinical Endocrinologists; North American Association for the Study of Obesity. Consensus development conference on antipsychotic drugs and obesity and diabetes. J Clin Psychiatry. 2004;65(2):267-272.

Issue
Current Psychiatry - 11(06)
Issue
Current Psychiatry - 11(06)
Page Number
59-67
Page Number
59-67
Publications
Publications
Topics
Article Type
Display Headline
PTSD nightmares: Prazosin and atypical antipsychotics
Display Headline
PTSD nightmares: Prazosin and atypical antipsychotics
Sections
Disallow All Ads
Alternative CME
Article PDF Media

‘Curbside’ consults: Know your liability

Article Type
Changed
Thu, 03/28/2019 - 16:18
Display Headline
‘Curbside’ consults: Know your liability

Discuss this article at www.facebook.com/CurrentPsychiatry

Dear Dr. Mossman:
Could providing a “curbside” consultation to a colleague leave me medico legally vulnerable if an adverse event leads to a malpractice lawsuit? If so, what can I do to address this risk?—Submitted by “Dr. W”

Medicine is a collaborative profession. Surgeons often combine skills to perform complex operations together, and specialists pool their expertise when they collectively manage patients with several medical problems. Doctors share their knowledge when they give lectures to medical audiences, write reports to referring physicians, or respond verbally to colleagues’ requests for information or advice.1

Doctors use the phrase “curbside consult” to refer (with humor and self-deprecation) to informal conversations with colleagues about patients’ medical management—advice-seeking that falls short of asking a colleague to make recommendations based on a formal, personal examination. Many physicians seek or provide curbside advice several times a month.2 Curbside consults transmit knowledge and cement professional bonds among physicians, making them “an integral part of our medical culture.”3

More than a dozen legal decisions mention curbside consultations. Judges think informal information-sharing improves medical practice and don’t want doctors to stop soliciting ideas or offering suggestions because they fear lawsuits.4,5 However, courts have found that, under certain conditions, giving advice can create liability for a bad outcome, even though the doctor never met the patient who was harmed.

In this article, we’ll look at:

  • when such liability might occur, and
  • what you can do to minimize it.

A doctor-patient relationship?

Legally, doctors are obligated to provide competent care for just 1 group of people: their patients. Therefore, to decide if plaintiffs could pursue malpractice claims in cases where doctors offered comments about patients they did not personally examine, courts have asked whether the circumstances, actions undertaken, or nature of information that was exchanged created a professional relationship.

Reynolds v Decatur Memorial Hospital4 describes an informal consultation that did not create a physician-patient relationship. In this case, a boy was admitted to a hospital after he had fallen. The treating pediatrician telephoned a neurosurgeon, who asked whether the boy’s neck was stiff, discussed diagnostic possibilities with the pediatrician, and suggested doing a lumbar puncture. The neurosurgeon offered to see the boy if requested, but he never did, and he did not bill for the telephone consultation. Guillain-Barré syndrome was first suspected, but a spinal cord injury was discovered after the boy—who developed quadriplegia—was transferred to another hospital.

In a subsequent lawsuit, the boy’s mother claimed her son’s paralysis resulted from negligence by the first hospital and its doctors, but the trial court dismissed the case against the neurosurgeon. Affirming the trial court’s ruling, an Illinois appeals court explained that the neurosurgeon had not been asked to provide medical services, conduct tests, or interpret test results. “A doctor who gives an informal opinion at the request of a treating physician does not owe a duty of care to the patient whose case was discussed,” the Reynolds court said.

Campbell v Haber6 describes circumstances that differed slightly from those described in the Reynolds decision but appeared to create a doctor-patient relationship. Campbell concerned a patient who came to an emergency room (ER) complaining of chest pain. The ER physician’s findings indicated possible heart muscle damage, so he telephoned a cardiologist (whom the ER doctor believed was “on call”) and described the patient’s symptoms and test results. The cardiologist thought the test results were not consistent with a cardiac event. The ER physician told the patient and his wife about the cardiologist’s opinion and, relying on what the cardiologist said, discharged the patient. Shortly after, the patient had a heart attack.

The patient sued not just the ER physician, but the cardiologist, who sought dismissal from the suit because he never saw the patient, had no treatment relationship with him, and never billed for services. However, the trial judge ruled that the patient could sue the cardiologist and the appellate court agreed, saying that a jury had to decide whether the cardiologist had incurred a doctor-patient relationship and might be liable. “An implied physician-patient relationship may arise when a physician gives advice to a patient,” the appeals court said, “even if that advice is communicated through another health care professional.”

Telling the difference

So what differentiates a no-liability curbside consult from a medical discussion that creates a doctor-patient duty and potential for liability for adverse results?

 

 

You create a physician-patient relationship when you assume responsibility to diagnose or treat someone.7 Although typically this requires an in-person encounter with a patient, it can happen indirectly—electronically (through e-mail), by telephone, or through a family member or another professional. But if you do nothing that implies consent to act for the patient’s benefit, you should have no actual malpractice liability if something goes wrong.3,8 As a Kansas Supreme Court decision explains, you “cannot be liable for medical malpractice” if you “merely consult with a treating physician and [do] nothing more.”5

Several legal cases discuss doctors’ efforts to extricate themselves from lawsuits arising from clinical encounters that the doctors mistakenly thought were just curbside consults. Table 18-12 lists situations in which talking about patients goes beyond just being “curbsided.”

Table 1

When it’s not a ‘curbside consultation’

SituationWhy it’s not a curbside consultation
On callIf you are “on call” for an emergency room, get called about a patient with an emergency condition, and discuss the patient’s symptoms, possible diagnosis, or treatment, you have a relationship with the patient that entails a duty of care8,9
CoveringIf you have agreed to “cover” patients for a colleague, you have assumed a duty to properly care for the colleague’s patients: they’re your patients during the colleague’s absence. Getting asked questions about managing those patients is not a curbside consultation, even if you’ve never met or spoken to the patient10,11
SupervisingPhysician assistants, residents in training, and nurse practitioners do not practice independently of their supervising physicians. If you’re a supervisor and get a call about managing a patient, you may bear vicarious liability for adverse results12
Specifics and relianceIf responding to the informal consult requires you to give specific advice that the consulting colleague will rely on to make a diagnosis or select treatment, you are participating in the patient’s care11

How to respond

Should you decline to provide curbside consultations to keep yourself out of lawsuits? Some authors think so, pointing out that informally transmitted clinical data may be faulty, which means you may give bad advice based on incomplete information or a verbal misunderstanding.13-16 These authors suggest that if you’re curbsided you should ask to see the patient for a formal consultation, decline to give informal advice, or provide a response that lacks specifics.

Other authors feel that these approaches are needlessly cautious and would harm patients by impeding doctors’ ability to help and learn from each other.3,17 These authors think the risk of incurring liability from a curbside consult is low. Also, getting advice from a colleague is a valuable risk management strategy; it helps you make sure you’re on the right track, and it shows you are a thoughtful clinician whose patients benefit from your own and your colleagues’ medical expertise.

Even if you’re comfortable soliciting and providing curbside advice, sometimes circumstances make it wise to follow-up an informal initial inquiry with a formal consultation. Table 23,17 lists examples of when you should follow-up with a formal consultation.

Table 2

Considerations that favor formal consultation

Complicated diagnostic situations
The consulted or requesting physician feels that giving good advice requires a personal examination
Advice is based on a detailed discussion and is specific to a patient’s situation
The patient requested the consultation
The consultant will make a report for the patient’s record
The consult bills for the consultation

Documentation

Experts disagree about whether the requesting or receiving physician should document a curbside consultation, and if so, how. On one hand, making a notation in a patient’s record documents the treating doctor’s diligence and may provide a measure of liability protection in a malpractice action. Doing this, however, exposes the identity of the consultant, who might be named among the defendants in a lawsuit.

One commonly recommended strategy is to request the consultant’s permission before identifying him or her in the record,13,16,17 a position that is defensible on grounds of courtesy alone. But omitting a consultant’s name from record does not guarantee that the consultant’s involvement won’t be discovered in the course of litigation.3 For example, treating doctors who get sued often are asked during their depositions about whether they talked with anyone about the case, and they have to answer honestly.

If a consulted doctor makes written notes, it might suggest that the consultation was more than the sort of informal information-sharing implied by the term “curbside.” However, in the unlikely event that a lawsuit arose and included the consultant as a defendant, documentation of advice given would help the consultant recall and defend what was said.

 

 

Related Resources

  • Grant-Kels JM, Kels BD. The curbside consultation: legal, moral, and ethical considerations. J Am Acad Dermatol. 2012;66(5):827-829.
  • Kreichelt R, Hilbert ML, Shinn D. Minimizing the legal risk with ‘curbside’ consultation. J Healthc Risk Manag. 2008;28(1):27-29.
  • Atkinson L. Curbside consults: what is your liability risk? Iowa Med. 2003;93(4):15.

Disclosure

Dr. Mossman reports no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.

References

1. Perley CM. Physician use of the curbside consultation to address information needs: report on a collective case study. J Med Libr Assoc. 2006;94(2):137-144.

2. Kuo D, Gifford DR, Stein MD. Curbside consultation practices and attitudes among primary care physicians and medical subspecialists. JAMA. 1998;280(10):905-909.

3. Cotton VR. Legal risks of “curbside” consults. Am J Cardiol. 2010;106(1):135-138.

4. Reynolds v Decatur Memorial Hospital, 277 Ill App 3d 80 (Ill App Ct 4th Dist 1996).

5. Irvin v Smith, 272 Kan 112 (Kan 2001).

6. Campbell v Haber, 274 A.D.2d 946 (NY App Div 4th Dep’t 2000).

7. Sterling v Johns Hopkins Hospital, 802 A.2d 440 (Md Ct Spec App 2002), cert den, 808 A.2d 808 (Md 2002).

8. Emergency Medical Treatment and Active Labor Act, 42 USC § 1395DD.

9. Lownsbury v VanBuren, 94 Ohio St 3d 231, 762 NE 2d 354 (2002).

10. Blazo v McLaren Regional Medical Center, 2002 Mich App LEXIS 752 (Mich Ct App 2002).

11. Kelley v Middle Tennessee Emergency Physicians, PC, 133 SW3d 587 (Tenn 2004).

12. Hammonds v Jewish Hospital, 899 SW2d 527 (Mo Ct App 1995).

13. MAG Mutual Insurance Company, Inc. Curbing the curbside consult—a risk management perspective. J Med Assoc Ga. 2008;97(1):50.-

14. Burns CD. Death of the curbside consult? J Ky Med Assoc. 2006;104(1):27.-

15. Hendel T. Informal consultations: do new risks exist with this age-old tradition? J Med Pract Manage. 2002;17(6):308-311.

16. Manian FA, Janssen DA. Curbside consultations. A closer look at a common practice. JAMA. 1996;275(2):145-147.

17. Curbside consultations. Psychiatry (Edgmont). 2010;7(5):51-53.

Article PDF
Author and Disclosure Information


Douglas Mossman, MD
Dr. Mossman is Administrative Director, Glenn M. Weaver Institute of Law and Psychiatry, University of Cincinnati College of Law, and Adjunct Professor of Clinical Psychiatry, and Training Director, University of Cincinnati Forensic Psychiatry Fellowship, Cincinnati, OH

Issue
Current Psychiatry - 11(06)
Publications
Topics
Page Number
42-45
Sections
Author and Disclosure Information


Douglas Mossman, MD
Dr. Mossman is Administrative Director, Glenn M. Weaver Institute of Law and Psychiatry, University of Cincinnati College of Law, and Adjunct Professor of Clinical Psychiatry, and Training Director, University of Cincinnati Forensic Psychiatry Fellowship, Cincinnati, OH

Author and Disclosure Information


Douglas Mossman, MD
Dr. Mossman is Administrative Director, Glenn M. Weaver Institute of Law and Psychiatry, University of Cincinnati College of Law, and Adjunct Professor of Clinical Psychiatry, and Training Director, University of Cincinnati Forensic Psychiatry Fellowship, Cincinnati, OH

Article PDF
Article PDF

Discuss this article at www.facebook.com/CurrentPsychiatry

Dear Dr. Mossman:
Could providing a “curbside” consultation to a colleague leave me medico legally vulnerable if an adverse event leads to a malpractice lawsuit? If so, what can I do to address this risk?—Submitted by “Dr. W”

Medicine is a collaborative profession. Surgeons often combine skills to perform complex operations together, and specialists pool their expertise when they collectively manage patients with several medical problems. Doctors share their knowledge when they give lectures to medical audiences, write reports to referring physicians, or respond verbally to colleagues’ requests for information or advice.1

Doctors use the phrase “curbside consult” to refer (with humor and self-deprecation) to informal conversations with colleagues about patients’ medical management—advice-seeking that falls short of asking a colleague to make recommendations based on a formal, personal examination. Many physicians seek or provide curbside advice several times a month.2 Curbside consults transmit knowledge and cement professional bonds among physicians, making them “an integral part of our medical culture.”3

More than a dozen legal decisions mention curbside consultations. Judges think informal information-sharing improves medical practice and don’t want doctors to stop soliciting ideas or offering suggestions because they fear lawsuits.4,5 However, courts have found that, under certain conditions, giving advice can create liability for a bad outcome, even though the doctor never met the patient who was harmed.

In this article, we’ll look at:

  • when such liability might occur, and
  • what you can do to minimize it.

A doctor-patient relationship?

Legally, doctors are obligated to provide competent care for just 1 group of people: their patients. Therefore, to decide if plaintiffs could pursue malpractice claims in cases where doctors offered comments about patients they did not personally examine, courts have asked whether the circumstances, actions undertaken, or nature of information that was exchanged created a professional relationship.

Reynolds v Decatur Memorial Hospital4 describes an informal consultation that did not create a physician-patient relationship. In this case, a boy was admitted to a hospital after he had fallen. The treating pediatrician telephoned a neurosurgeon, who asked whether the boy’s neck was stiff, discussed diagnostic possibilities with the pediatrician, and suggested doing a lumbar puncture. The neurosurgeon offered to see the boy if requested, but he never did, and he did not bill for the telephone consultation. Guillain-Barré syndrome was first suspected, but a spinal cord injury was discovered after the boy—who developed quadriplegia—was transferred to another hospital.

In a subsequent lawsuit, the boy’s mother claimed her son’s paralysis resulted from negligence by the first hospital and its doctors, but the trial court dismissed the case against the neurosurgeon. Affirming the trial court’s ruling, an Illinois appeals court explained that the neurosurgeon had not been asked to provide medical services, conduct tests, or interpret test results. “A doctor who gives an informal opinion at the request of a treating physician does not owe a duty of care to the patient whose case was discussed,” the Reynolds court said.

Campbell v Haber6 describes circumstances that differed slightly from those described in the Reynolds decision but appeared to create a doctor-patient relationship. Campbell concerned a patient who came to an emergency room (ER) complaining of chest pain. The ER physician’s findings indicated possible heart muscle damage, so he telephoned a cardiologist (whom the ER doctor believed was “on call”) and described the patient’s symptoms and test results. The cardiologist thought the test results were not consistent with a cardiac event. The ER physician told the patient and his wife about the cardiologist’s opinion and, relying on what the cardiologist said, discharged the patient. Shortly after, the patient had a heart attack.

The patient sued not just the ER physician, but the cardiologist, who sought dismissal from the suit because he never saw the patient, had no treatment relationship with him, and never billed for services. However, the trial judge ruled that the patient could sue the cardiologist and the appellate court agreed, saying that a jury had to decide whether the cardiologist had incurred a doctor-patient relationship and might be liable. “An implied physician-patient relationship may arise when a physician gives advice to a patient,” the appeals court said, “even if that advice is communicated through another health care professional.”

Telling the difference

So what differentiates a no-liability curbside consult from a medical discussion that creates a doctor-patient duty and potential for liability for adverse results?

 

 

You create a physician-patient relationship when you assume responsibility to diagnose or treat someone.7 Although typically this requires an in-person encounter with a patient, it can happen indirectly—electronically (through e-mail), by telephone, or through a family member or another professional. But if you do nothing that implies consent to act for the patient’s benefit, you should have no actual malpractice liability if something goes wrong.3,8 As a Kansas Supreme Court decision explains, you “cannot be liable for medical malpractice” if you “merely consult with a treating physician and [do] nothing more.”5

Several legal cases discuss doctors’ efforts to extricate themselves from lawsuits arising from clinical encounters that the doctors mistakenly thought were just curbside consults. Table 18-12 lists situations in which talking about patients goes beyond just being “curbsided.”

Table 1

When it’s not a ‘curbside consultation’

SituationWhy it’s not a curbside consultation
On callIf you are “on call” for an emergency room, get called about a patient with an emergency condition, and discuss the patient’s symptoms, possible diagnosis, or treatment, you have a relationship with the patient that entails a duty of care8,9
CoveringIf you have agreed to “cover” patients for a colleague, you have assumed a duty to properly care for the colleague’s patients: they’re your patients during the colleague’s absence. Getting asked questions about managing those patients is not a curbside consultation, even if you’ve never met or spoken to the patient10,11
SupervisingPhysician assistants, residents in training, and nurse practitioners do not practice independently of their supervising physicians. If you’re a supervisor and get a call about managing a patient, you may bear vicarious liability for adverse results12
Specifics and relianceIf responding to the informal consult requires you to give specific advice that the consulting colleague will rely on to make a diagnosis or select treatment, you are participating in the patient’s care11

How to respond

Should you decline to provide curbside consultations to keep yourself out of lawsuits? Some authors think so, pointing out that informally transmitted clinical data may be faulty, which means you may give bad advice based on incomplete information or a verbal misunderstanding.13-16 These authors suggest that if you’re curbsided you should ask to see the patient for a formal consultation, decline to give informal advice, or provide a response that lacks specifics.

Other authors feel that these approaches are needlessly cautious and would harm patients by impeding doctors’ ability to help and learn from each other.3,17 These authors think the risk of incurring liability from a curbside consult is low. Also, getting advice from a colleague is a valuable risk management strategy; it helps you make sure you’re on the right track, and it shows you are a thoughtful clinician whose patients benefit from your own and your colleagues’ medical expertise.

Even if you’re comfortable soliciting and providing curbside advice, sometimes circumstances make it wise to follow-up an informal initial inquiry with a formal consultation. Table 23,17 lists examples of when you should follow-up with a formal consultation.

Table 2

Considerations that favor formal consultation

Complicated diagnostic situations
The consulted or requesting physician feels that giving good advice requires a personal examination
Advice is based on a detailed discussion and is specific to a patient’s situation
The patient requested the consultation
The consultant will make a report for the patient’s record
The consult bills for the consultation

Documentation

Experts disagree about whether the requesting or receiving physician should document a curbside consultation, and if so, how. On one hand, making a notation in a patient’s record documents the treating doctor’s diligence and may provide a measure of liability protection in a malpractice action. Doing this, however, exposes the identity of the consultant, who might be named among the defendants in a lawsuit.

One commonly recommended strategy is to request the consultant’s permission before identifying him or her in the record,13,16,17 a position that is defensible on grounds of courtesy alone. But omitting a consultant’s name from record does not guarantee that the consultant’s involvement won’t be discovered in the course of litigation.3 For example, treating doctors who get sued often are asked during their depositions about whether they talked with anyone about the case, and they have to answer honestly.

If a consulted doctor makes written notes, it might suggest that the consultation was more than the sort of informal information-sharing implied by the term “curbside.” However, in the unlikely event that a lawsuit arose and included the consultant as a defendant, documentation of advice given would help the consultant recall and defend what was said.

 

 

Related Resources

  • Grant-Kels JM, Kels BD. The curbside consultation: legal, moral, and ethical considerations. J Am Acad Dermatol. 2012;66(5):827-829.
  • Kreichelt R, Hilbert ML, Shinn D. Minimizing the legal risk with ‘curbside’ consultation. J Healthc Risk Manag. 2008;28(1):27-29.
  • Atkinson L. Curbside consults: what is your liability risk? Iowa Med. 2003;93(4):15.

Disclosure

Dr. Mossman reports no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.

Discuss this article at www.facebook.com/CurrentPsychiatry

Dear Dr. Mossman:
Could providing a “curbside” consultation to a colleague leave me medico legally vulnerable if an adverse event leads to a malpractice lawsuit? If so, what can I do to address this risk?—Submitted by “Dr. W”

Medicine is a collaborative profession. Surgeons often combine skills to perform complex operations together, and specialists pool their expertise when they collectively manage patients with several medical problems. Doctors share their knowledge when they give lectures to medical audiences, write reports to referring physicians, or respond verbally to colleagues’ requests for information or advice.1

Doctors use the phrase “curbside consult” to refer (with humor and self-deprecation) to informal conversations with colleagues about patients’ medical management—advice-seeking that falls short of asking a colleague to make recommendations based on a formal, personal examination. Many physicians seek or provide curbside advice several times a month.2 Curbside consults transmit knowledge and cement professional bonds among physicians, making them “an integral part of our medical culture.”3

More than a dozen legal decisions mention curbside consultations. Judges think informal information-sharing improves medical practice and don’t want doctors to stop soliciting ideas or offering suggestions because they fear lawsuits.4,5 However, courts have found that, under certain conditions, giving advice can create liability for a bad outcome, even though the doctor never met the patient who was harmed.

In this article, we’ll look at:

  • when such liability might occur, and
  • what you can do to minimize it.

A doctor-patient relationship?

Legally, doctors are obligated to provide competent care for just 1 group of people: their patients. Therefore, to decide if plaintiffs could pursue malpractice claims in cases where doctors offered comments about patients they did not personally examine, courts have asked whether the circumstances, actions undertaken, or nature of information that was exchanged created a professional relationship.

Reynolds v Decatur Memorial Hospital4 describes an informal consultation that did not create a physician-patient relationship. In this case, a boy was admitted to a hospital after he had fallen. The treating pediatrician telephoned a neurosurgeon, who asked whether the boy’s neck was stiff, discussed diagnostic possibilities with the pediatrician, and suggested doing a lumbar puncture. The neurosurgeon offered to see the boy if requested, but he never did, and he did not bill for the telephone consultation. Guillain-Barré syndrome was first suspected, but a spinal cord injury was discovered after the boy—who developed quadriplegia—was transferred to another hospital.

In a subsequent lawsuit, the boy’s mother claimed her son’s paralysis resulted from negligence by the first hospital and its doctors, but the trial court dismissed the case against the neurosurgeon. Affirming the trial court’s ruling, an Illinois appeals court explained that the neurosurgeon had not been asked to provide medical services, conduct tests, or interpret test results. “A doctor who gives an informal opinion at the request of a treating physician does not owe a duty of care to the patient whose case was discussed,” the Reynolds court said.

Campbell v Haber6 describes circumstances that differed slightly from those described in the Reynolds decision but appeared to create a doctor-patient relationship. Campbell concerned a patient who came to an emergency room (ER) complaining of chest pain. The ER physician’s findings indicated possible heart muscle damage, so he telephoned a cardiologist (whom the ER doctor believed was “on call”) and described the patient’s symptoms and test results. The cardiologist thought the test results were not consistent with a cardiac event. The ER physician told the patient and his wife about the cardiologist’s opinion and, relying on what the cardiologist said, discharged the patient. Shortly after, the patient had a heart attack.

The patient sued not just the ER physician, but the cardiologist, who sought dismissal from the suit because he never saw the patient, had no treatment relationship with him, and never billed for services. However, the trial judge ruled that the patient could sue the cardiologist and the appellate court agreed, saying that a jury had to decide whether the cardiologist had incurred a doctor-patient relationship and might be liable. “An implied physician-patient relationship may arise when a physician gives advice to a patient,” the appeals court said, “even if that advice is communicated through another health care professional.”

Telling the difference

So what differentiates a no-liability curbside consult from a medical discussion that creates a doctor-patient duty and potential for liability for adverse results?

 

 

You create a physician-patient relationship when you assume responsibility to diagnose or treat someone.7 Although typically this requires an in-person encounter with a patient, it can happen indirectly—electronically (through e-mail), by telephone, or through a family member or another professional. But if you do nothing that implies consent to act for the patient’s benefit, you should have no actual malpractice liability if something goes wrong.3,8 As a Kansas Supreme Court decision explains, you “cannot be liable for medical malpractice” if you “merely consult with a treating physician and [do] nothing more.”5

Several legal cases discuss doctors’ efforts to extricate themselves from lawsuits arising from clinical encounters that the doctors mistakenly thought were just curbside consults. Table 18-12 lists situations in which talking about patients goes beyond just being “curbsided.”

Table 1

When it’s not a ‘curbside consultation’

SituationWhy it’s not a curbside consultation
On callIf you are “on call” for an emergency room, get called about a patient with an emergency condition, and discuss the patient’s symptoms, possible diagnosis, or treatment, you have a relationship with the patient that entails a duty of care8,9
CoveringIf you have agreed to “cover” patients for a colleague, you have assumed a duty to properly care for the colleague’s patients: they’re your patients during the colleague’s absence. Getting asked questions about managing those patients is not a curbside consultation, even if you’ve never met or spoken to the patient10,11
SupervisingPhysician assistants, residents in training, and nurse practitioners do not practice independently of their supervising physicians. If you’re a supervisor and get a call about managing a patient, you may bear vicarious liability for adverse results12
Specifics and relianceIf responding to the informal consult requires you to give specific advice that the consulting colleague will rely on to make a diagnosis or select treatment, you are participating in the patient’s care11

How to respond

Should you decline to provide curbside consultations to keep yourself out of lawsuits? Some authors think so, pointing out that informally transmitted clinical data may be faulty, which means you may give bad advice based on incomplete information or a verbal misunderstanding.13-16 These authors suggest that if you’re curbsided you should ask to see the patient for a formal consultation, decline to give informal advice, or provide a response that lacks specifics.

Other authors feel that these approaches are needlessly cautious and would harm patients by impeding doctors’ ability to help and learn from each other.3,17 These authors think the risk of incurring liability from a curbside consult is low. Also, getting advice from a colleague is a valuable risk management strategy; it helps you make sure you’re on the right track, and it shows you are a thoughtful clinician whose patients benefit from your own and your colleagues’ medical expertise.

Even if you’re comfortable soliciting and providing curbside advice, sometimes circumstances make it wise to follow-up an informal initial inquiry with a formal consultation. Table 23,17 lists examples of when you should follow-up with a formal consultation.

Table 2

Considerations that favor formal consultation

Complicated diagnostic situations
The consulted or requesting physician feels that giving good advice requires a personal examination
Advice is based on a detailed discussion and is specific to a patient’s situation
The patient requested the consultation
The consultant will make a report for the patient’s record
The consult bills for the consultation

Documentation

Experts disagree about whether the requesting or receiving physician should document a curbside consultation, and if so, how. On one hand, making a notation in a patient’s record documents the treating doctor’s diligence and may provide a measure of liability protection in a malpractice action. Doing this, however, exposes the identity of the consultant, who might be named among the defendants in a lawsuit.

One commonly recommended strategy is to request the consultant’s permission before identifying him or her in the record,13,16,17 a position that is defensible on grounds of courtesy alone. But omitting a consultant’s name from record does not guarantee that the consultant’s involvement won’t be discovered in the course of litigation.3 For example, treating doctors who get sued often are asked during their depositions about whether they talked with anyone about the case, and they have to answer honestly.

If a consulted doctor makes written notes, it might suggest that the consultation was more than the sort of informal information-sharing implied by the term “curbside.” However, in the unlikely event that a lawsuit arose and included the consultant as a defendant, documentation of advice given would help the consultant recall and defend what was said.

 

 

Related Resources

  • Grant-Kels JM, Kels BD. The curbside consultation: legal, moral, and ethical considerations. J Am Acad Dermatol. 2012;66(5):827-829.
  • Kreichelt R, Hilbert ML, Shinn D. Minimizing the legal risk with ‘curbside’ consultation. J Healthc Risk Manag. 2008;28(1):27-29.
  • Atkinson L. Curbside consults: what is your liability risk? Iowa Med. 2003;93(4):15.

Disclosure

Dr. Mossman reports no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.

References

1. Perley CM. Physician use of the curbside consultation to address information needs: report on a collective case study. J Med Libr Assoc. 2006;94(2):137-144.

2. Kuo D, Gifford DR, Stein MD. Curbside consultation practices and attitudes among primary care physicians and medical subspecialists. JAMA. 1998;280(10):905-909.

3. Cotton VR. Legal risks of “curbside” consults. Am J Cardiol. 2010;106(1):135-138.

4. Reynolds v Decatur Memorial Hospital, 277 Ill App 3d 80 (Ill App Ct 4th Dist 1996).

5. Irvin v Smith, 272 Kan 112 (Kan 2001).

6. Campbell v Haber, 274 A.D.2d 946 (NY App Div 4th Dep’t 2000).

7. Sterling v Johns Hopkins Hospital, 802 A.2d 440 (Md Ct Spec App 2002), cert den, 808 A.2d 808 (Md 2002).

8. Emergency Medical Treatment and Active Labor Act, 42 USC § 1395DD.

9. Lownsbury v VanBuren, 94 Ohio St 3d 231, 762 NE 2d 354 (2002).

10. Blazo v McLaren Regional Medical Center, 2002 Mich App LEXIS 752 (Mich Ct App 2002).

11. Kelley v Middle Tennessee Emergency Physicians, PC, 133 SW3d 587 (Tenn 2004).

12. Hammonds v Jewish Hospital, 899 SW2d 527 (Mo Ct App 1995).

13. MAG Mutual Insurance Company, Inc. Curbing the curbside consult—a risk management perspective. J Med Assoc Ga. 2008;97(1):50.-

14. Burns CD. Death of the curbside consult? J Ky Med Assoc. 2006;104(1):27.-

15. Hendel T. Informal consultations: do new risks exist with this age-old tradition? J Med Pract Manage. 2002;17(6):308-311.

16. Manian FA, Janssen DA. Curbside consultations. A closer look at a common practice. JAMA. 1996;275(2):145-147.

17. Curbside consultations. Psychiatry (Edgmont). 2010;7(5):51-53.

References

1. Perley CM. Physician use of the curbside consultation to address information needs: report on a collective case study. J Med Libr Assoc. 2006;94(2):137-144.

2. Kuo D, Gifford DR, Stein MD. Curbside consultation practices and attitudes among primary care physicians and medical subspecialists. JAMA. 1998;280(10):905-909.

3. Cotton VR. Legal risks of “curbside” consults. Am J Cardiol. 2010;106(1):135-138.

4. Reynolds v Decatur Memorial Hospital, 277 Ill App 3d 80 (Ill App Ct 4th Dist 1996).

5. Irvin v Smith, 272 Kan 112 (Kan 2001).

6. Campbell v Haber, 274 A.D.2d 946 (NY App Div 4th Dep’t 2000).

7. Sterling v Johns Hopkins Hospital, 802 A.2d 440 (Md Ct Spec App 2002), cert den, 808 A.2d 808 (Md 2002).

8. Emergency Medical Treatment and Active Labor Act, 42 USC § 1395DD.

9. Lownsbury v VanBuren, 94 Ohio St 3d 231, 762 NE 2d 354 (2002).

10. Blazo v McLaren Regional Medical Center, 2002 Mich App LEXIS 752 (Mich Ct App 2002).

11. Kelley v Middle Tennessee Emergency Physicians, PC, 133 SW3d 587 (Tenn 2004).

12. Hammonds v Jewish Hospital, 899 SW2d 527 (Mo Ct App 1995).

13. MAG Mutual Insurance Company, Inc. Curbing the curbside consult—a risk management perspective. J Med Assoc Ga. 2008;97(1):50.-

14. Burns CD. Death of the curbside consult? J Ky Med Assoc. 2006;104(1):27.-

15. Hendel T. Informal consultations: do new risks exist with this age-old tradition? J Med Pract Manage. 2002;17(6):308-311.

16. Manian FA, Janssen DA. Curbside consultations. A closer look at a common practice. JAMA. 1996;275(2):145-147.

17. Curbside consultations. Psychiatry (Edgmont). 2010;7(5):51-53.

Issue
Current Psychiatry - 11(06)
Issue
Current Psychiatry - 11(06)
Page Number
42-45
Page Number
42-45
Publications
Publications
Topics
Article Type
Display Headline
‘Curbside’ consults: Know your liability
Display Headline
‘Curbside’ consults: Know your liability
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media

High-dose donepezil or memantine: Next step for Alzheimer’s disease?

Article Type
Changed
Tue, 12/11/2018 - 15:04
Display Headline
High-dose donepezil or memantine: Next step for Alzheimer’s disease?

Although cholinesterase inhibitors (ChEIs) and memantine at standard doses may slow the progression of Alzheimer’s disease (AD) as assessed by cognitive, functional, and global measures, this effect is relatively modest. For the estimated 5.4 million Americans with AD1—more than one-half of whom have moderate to severe disease2—there is a great need for new approaches to slow AD progression.

High doses of donepezil or memantine may be the next step in achieving better results than standard pharmacologic treatments for AD. This article presents the possible benefits and indications for high doses of donepezil (23 mg/d) and memantine (28 mg/d) for managing moderate to severe AD and their safety and tolerability profiles.

Current treatments offer modest benefits

AD treatments comprise 2 categories: ChEIs (donepezil, rivastigmine, and galantamine) and the N-methyl-D-aspartate (NMDA) receptor antagonist memantine (Table 1).3,4 All ChEIs are FDA-approved for mild to moderate AD; donepezil also is approved for severe AD. Memantine is approved for moderate to severe AD, either alone or in combination with ChEIs. Until recently, the maximum FDA-approved doses were donepezil, 10 mg/d, and memantine, 20 mg/d. However, these dosages are associated with only modest beneficial effects in managing cognitive deterioration in patients with moderate to severe dementia.5,6 Studies have reported that combining a ChEI, such as donepezil, and memantine is well tolerated and may result in synergistic benefits by affecting different neurotransmitters in patients with moderate to severe AD.7,8

Recently, the FDA approved higher daily doses of donepezil (23 mg) and memantine (28 mg) for moderate to severe AD on the basis of positive phase III trial results.9-11 Donepezil, 23 mg/d, currently is marketed in the United States; the availability date for memantine, 28 mg/d, was undetermined at press time.

Table 1

FDA-approved treatments for Alzheimer’s disease

DrugMaximum daily doseMechanism of actionIndicationCommon side effects/comments
Tacrine160 mg/dChEIMild to moderate ADNausea, vomiting, loss of appetite, diarrhea. First ChEI to be approved, but rarely used because of associated possible hepatotoxicity
Donepezil10 mg/dChEIAll stages of ADNausea, vomiting, loss of appetite, diarrhea, sleep disturbance
Rivastigmine12 mg/dChEIMild to moderate ADNausea, vomiting, diarrhea, weight loss, loss of appetite
Galantamine24 mg/dChEIMild to moderate ADNausea, vomiting, diarrhea, weight loss, loss of appetite
Memantine20 mg/dNMDA receptor antagonistModerate to severe ADDizziness, headache, constipation, confusion
Galantamine ER24 mg/dChEIMild to moderate ADNausea, vomiting, diarrhea, weight loss, loss of appetite
Rivastigmine transdermal system9.5 mg/dChEIMild to moderate ADNausea, vomiting, diarrhea, weight loss, loss of appetite
Donepezil 2323 mg/dChEIModerate to severe ADNausea, vomiting, diarrhea
Memantine ER28 mg/dNMDA receptor antagonistModerate to severe ADDizziness, headache, constipation, confusion
AD: Alzheimer’s disease; ChEI: cholinesterase inhibitor; ER: extended release; NMDA: N-methyl-D-aspartate
Source: References 3,4

High-dose donepezil (23 mg/d)

Cognitive decline with AD has been associated with increasing loss of cholinergic neurons and cholinergic activities, particularly in areas associated with memory/cognition and learning, including cortical areas involving the temporal lobe, hippocampus, and nucleus basalis of Meynert.12-14 In addition, evidence suggests that increasing levels of acetylcholine by using ChEIs can enhance cognitive function.13,15

Donepezil is a selective, reversible ChEI believed to enhance central cholinergic function.15 Randomized clinical trials assessing dose-response with donepezil, 5 mg/d and 10 mg/d, have demonstrated more benefit in cognition with either dose than placebo. The 10 mg/d dose was more effective than 5 mg/d in patients with mild to moderate and severe AD.16-18 In patients with advanced AD who are stable on 5 mg/d, increasing to 10 mg/d could slow the progression of cognitive decline.18

Rationale for higher doses. Positron emission tomography studies have shown that at stable doses of donepezil, 5 mg/d or 10 mg/d, average cortical acetylcholinesterase (AChE) inhibition was <30%.19,20 Based on these findings, researchers thought that cortical AChE inhibition may be suboptimal with donepezil, 10 mg/d, and that higher doses of ChEI may be required in patients with more advanced AD—and therefore more cholinergic loss—for adequate cholinesterase inhibition. In a pilot study of patients with mild to moderate AD, higher doses of donepezil (15 mg/d and 20 mg/d) were reported to be safe and well tolerated.21

The 23-mg/d donepezil formulation was developed to provide a higher dose administered once daily without a sharp rise in peak concentration. The FDA approved donepezil, 23 mg/d, for patients with moderate to severe AD on the basis of phase III trial results.9,22 In a randomized, double-blind, multicenter, head-to-head clinical trial, >1,400 patients with moderate to severe AD (Mini-Mental State Exam [MMSE]: 0 to 20) on a stable dose of donepezil, 10 mg/d, for ≥3 months were randomly assigned to receive high-dose donepezil (23 mg/d) or standard-dose donepezil (10 mg/d) for 24 weeks.9,22 Patients in the 23-mg/d group showed a statistically significant improvement in cognition compared with the 10-mg/d group. The difference between groups on a measure of global improvement was not significant.9,22 However, in a post-hoc analysis, it was demonstrated that a subgroup of patients with more severe cognitive impairment (baseline MMSE: 0 to 16), showed significant improvement in cognition as well as global functioning.9

 

 

Overall, treatment-emergent adverse events (TEAEs) during the study were higher in patients receiving 23 mg/d (74%) than those receiving 10 mg/d (64%). The most common TEAEs in the 23-mg/d and 10-mg/d groups were nausea (12% vs 3%, respectively), vomiting (9% vs 3%), and diarrhea (8% vs 5%) (Table 2).22 These gastrointestinal adverse effects were more frequent during the first month of treatment and were relatively infrequent beyond 1 month. Serious TEAEs, such as falls, urinary tract infection, pneumonia, syncope, aggression, and confusional state, were noted in a similar proportion of patients in the 23-mg/d and 10-mg/d groups; most of these were considered unrelated to treatment. No drug-related deaths occurred during the study. High-dose (23 mg/d) donepezil generally was well tolerated, with a typical ChEI safety profile but superior efficacy.

A recent commentary discussed the issue of effect size and whether a 2.2-point difference on a 100-point scale (the Severe Impairment Battery [SIB]) is clinically meaningful.23 As with all anti-dementia therapies, in any cohort some patients will gain considerably more than 2.2 points on the SIB, which is clinically significant. A 6-month trial is recommended to identify these optimal responders.

Table 2

High-dose vs standard-dose donepezil: Treatment-emergent adverse events

Adverse eventDonepezil, 23 mg/dDonepezil,10 mg/d
Nausea12%3%
Vomiting9%3%
Diarrhea8%5%
Anorexia5%2%
Dizziness5%3%
Weight decrease5%3%
Headache4%3%
Insomnia3%2%
Urinary incontinence3%1%
Fatigue2%1%
Weakness2%1%
Somnolence2%1%
Contusion2%0%
Source: Reference 22

High-dose memantine

Memantine is an NMDA receptor antagonist, which works on glutamate, an ubiquitous neurotransmitter in the brain that serves many functions. For reasons that are not fully understood, in AD glutamate becomes excitotoxic and causes neuronal death.

Some researchers have hypothesized that if safe and well tolerated, a memantine dose >20 mg/d may have better efficacy than a lower dose. Memantine’s manufacturer has developed an extended-release (ER), once-daily formulation of memantine, 28 mg/d, to improve adherence and possibly increase efficacy.10,11 Because of memantine ER’s relatively slow absorption rate and longer median Tmax, of 12 hours, there is minimal fluctuation in plasma levels during steady-state dosing intervals compared with the immediate-release (IR) formulation.10

In a phase I study of 24 healthy volunteers that investigated the safety, tolerability, and pharmacokinetics of memantine ER, 28 mg/d, TEAEs were mild; the most common were headache, somnolence, and dizziness.10 During memantine treatment, there were no serious adverse events, potential significant changes in patients’ vital signs, or deaths.

Memantine ER plus ChEI. A multicenter, multinational, randomized, double-blind study compared memantine ER, 28 mg/d, and placebo in patients with moderate to severe AD (MMSE: 3 to 14).11 All patients were receiving concurrent, stable ChEI treatment (donepezil, rivastigmine, or galantamine) for ≥3 months before the study. Patients treated with memantine ER, 28 mg/d, and ChEI (n = 342) showed a significant improvement compared with the placebo/ChEI group (n = 335) in cognition and global functioning. Patients receiving memantine/ChEI also showed statistically significant benefits on behavior and verbal fluency testing compared with patients receiving placebo/ChEI. Memantine was well tolerated; most adverse events were mild or moderate. The most common adverse events in the memantine/ChEI group that occurred at a higher rate relative to the placebo/ChEI group were headache (5.6% vs 5.1%, respectively), diarrhea (5.0% vs 3.9%), and dizziness (4.7% vs 1.5%). There were no deaths related to memantine (Table 3).11

Memantine ER, 28 mg/d, may be tolerated better than the IR formulation because of less plasma level fluctuation during the steady-state dosing interval. Also, memantine ER, 28 mg/d, may offer better efficacy over memantine IR, 20 mg/d, because of dose-dependent cognitive, global, and behavioral effects. In addition, once-daily dosing of memantine ER may improve adherence compared with the IR formulation.24

In patients with severe renal impairment, dosage of memantine IR should be reduced from 20 mg/d to 10 mg/d.25 However, there is no available information regarding the dosing, safety, and tolerability of memantine ER, 28 mg/d, in patients with renal disease.

Table3

High-dose memantine: Treatment-emergent adverse eventsa

Adverse eventPlacebo (n = 335)Memantine ER (n = 341)
Any TEAE214 (63.9%)214 (62.8%)
Fall26 (7.8%)19 (5.6%)
Urinary tract infection24 (7.2%)19 (5.6%)
Headache17 (5.1%)19 (5.6%)
Diarrhea13 (3.9%)17 (5.0%)
Dizziness5 (1.5%)16 (4.7%)
Influenza9 (2.7%)15 (4.4%)
Insomnia16 (4.8%)14 (4.1%)
Agitation15 (4.5%)14 (4.1%)
Hypertension8 (2.4%)13 (3.8%)
Anxiety9 (2.7%)12 (3.5%)
Depression5 (1.5%)11 (3.2%)
Weight increased3 (0.9%)11 (3.2%)
Constipation4 (1.2%)10 (2.9%)
Somnolence4 (1.2%)10 (2.9%)
Back pain2 (0.6%)9 (2.6%)
Aggression5 (1.5%)8 (2.3%)
Hypotension5 (1.5%)7 (2.1%)
Vomiting4 (1.2%)7 (2.1%)
Abdominal pain2 (0.6%)7 (2.1%)
Nasopharyngitis10 (3.0%)6 (1.8%)
Confusional state7 (2.1%)6 (1.8%)
Weight decreased11 (3.3%)5 (1.5%)
Nausea7 (2.1%)5 (1.5%)
Irritability8 (2.4%)4 (1.2%)
Cough8 (2.4%)3 (0.9%)
aData [n (%)] include all adverse events experienced by ≥2% patients in either group (safety population). Adverse events that were experienced at twice the rate in 1 group compared with the other are indicated by bold type
ER: extended-release (28 mg); TEAE: treatment-emergent adverse event
Source: Reference 11
 

 

Recommendations

Because there are few FDA-approved treatments for AD, higher doses of donepezil or memantine may be an option for patients who have “maxed out” on their AD therapy or no longer respond to lower doses. Higher doses of donepezil (23 mg/d) and memantine (28 mg/d) could improve medication adherence because both are once-daily preparations. In clinical trials, donepezil, 23 mg/d, was more effective than donepezil, 10 mg/d.9 Whether memantine ER, 28 mg/d, is superior to memantine IR, 20 mg/d, needs to be investigated in head-to-head, double-blind, controlled studies.

For patients with moderate to severe AD, donepezil, 23 mg, is associated with greater benefits in cognition compared with donepezil, 10 mg/d.9 Similarly, because of potentially superior efficacy because of a higher dose, memantine ER, 28 mg, might best help patients with moderate to severe AD, specifically those who either don’t respond or lose response to memantine IR, 20 mg/d. Combining a ChEI, such as donepezil, with memantine is associated with slower cognitive decline and short and long-term benefits on measures of cognition, activities of daily living, global outcome, and behavior.7,26 However, additional clinical trials are needed to assess the safety, tolerability, and efficacy of combination therapy with higher doses of donepezil and memantine ER.

Related Resources

  • Alzheimer’s Disease Education and Referral Center. www.nia.nih.gov/Alzheimers.
  • Lleó A, Greenberg SM, Growdon JH. Current pharmacotherapy for Alzheimer’s disease. Annu Rev Med. 2006;57:513-533.

Drug Brand Names

  • Donepezil • Aricept
  • Galantamine • Razadyne
  • Memantine • Namenda
  • Rivastigmine • Exelon
  • Tacrine • Cognex

Disclosures

Dr. Grossberg’s academic department has received research funding from Forest Pharmaceuticals and Pfizer Inc. Dr. Grossberg has received grant/research support from Baxter BioScience, Forest Pharmaceuticals, Janssen, the National Institutes of Health, Novartis, and Pfizer, Inc.; is a consultant to Baxter BioScience, Forest Pharmaceuticals, Merck, Novartis, and Otsuka; and is on the Safety Monitoring Committee for Merck.

Dr. Singh reports no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.

References

1. Alzheimer’s Association, Thies W, Bleiler L. 2011 Alzheimer’s disease facts and figures. Alzheimers Dement. 2011;7(2):208-244.

2. Hebert LE, Scherr PA, Bienias JL, et al. Alzheimer disease in the US population: prevalence estimates using the 2000 census. Arch Neurol. 2003;60(8):1119-1122.

3. Alzheimer’s Disease Education and Referral Center. Alzheimer’s disease medications. http://www.nia.nih.gov/alzheimers/publication/alzheimers-disease-medications-fact-sheet. Accessed May 10 2012.

4. Osborn GG, Saunders AV. Current treatments for patients with Alzheimer disease. J Am Osteopath Assoc. 2010;110(9 suppl 8):S16-S26.

5. Raina P, Santaguida P, Ismaila A, et al. Effectiveness of cholinesterase inhibitors and memantine for treating dementia: evidence review for a clinical practice guideline. Ann Intern Med. 2008;148(5):379-397.

6. Cummings JL. Alzheimer’s disease. N Engl J Med. 2004;351(1):56-67.

7. Tariot PN, Farlow MR, Grossberg GT, et al. Memantine Study Group. Memantine treatment in patients with moderate to severe Alzheimer disease already receiving donepezil: a randomized controlled trial. JAMA. 2004;291(3):317-324.

8. Xiong G, Doraiswamy PM. Combination drug therapy for Alzheimer’s disease: what is evidence-based and what is not? Geriatrics. 2005;60(6):22-26.

9. Farlow MR, Salloway S, Tariot PN, et al. Effectiveness and tolerability of high (23 mg/d) versus standard-dose (10 mg/d) donepezil in moderate to severe Alzheimer’s disease: a 24-week, randomized, double-blind study. Clin Ther. 2010;32(7):1234-1251.

10. Periclou A, Hu Y. Extended-release memantine capsule (28 mg once daily): a multiple dose, open-label study evaluating steady-state pharmacokinetics in healthy volunteers. Poster presented at 11th International Conference on Alzheimer’s Disease; July 26-31, 2008; Chicago, IL.

11. Grossberg GT, Manes F, Allegri R, et al. A multinational, randomized, double-blind, placebo-controlled, parallel-group trial of memantine extended-release capsule (28 mg, once daily) in patients with moderate to severe Alzheimer’s disease. Poster presented at 11th International Conference on Alzheimer’s Disease; July 26-31, 2008; Chicago, IL.

12. Geula C, Mesulam MM. Systematic regional variations in the loss of cortical cholinergic fibers in Alzheimer’s disease. Cereb Cortex. 1996;6(2):165-177.

13. Whitehouse PJ. The cholinergic deficit in Alzheimer’s disease. J Clin Psychiatry. 1998;59(suppl 13):19-22.

14. Teipel SJ, Flatz WH, Heinsen H, et al. Measurement of basal forebrain atrophy in Alzheimer’s disease using MRI. Brain. 2005;128(11):2626-2644.

15. Shintani EY, Uchida KM. Donepezil: an anticholinesterase inhibitor for Alzheimer’s disease. Am J Health Syst Pharm. 1997;54(24):2805-2810.

16. Homma A, Imai Y, Tago H, et al. Donepezil treatment of patients with severe Alzheimer’s disease in a Japanese population: results from a 24-week, double-blind, placebo-controlled, randomized trial. Dement Geriatr Cogn Disord. 2008;25(5):399-407.

17. Whitehead A, Perdomo C, Pratt RD, et al. Donepezil for the symptomatic treatment of patients with mild to moderate Alzheimer’s disease: a meta-analysis of individual patient data from randomised controlled trials. Int J Geriatr Psychiatry. 2004;19(7):624-633.

18. Nozawa M, Ichimiya Y, Nozawa E, et al. Clinical effects of high oral dose of donepezil for patients with Alzheimer’s disease in Japan. Psychogeriatrics. 2009;9(2):50-55.

19. Kuhl DE, Minoshima S, Frey KA, et al. Limited donepezil inhibition of acetylcholinesterase measured with positron emission tomography in living Alzheimer cerebral cortex. Ann Neurol. 2000;48(3):391-395.

20. Bohnen NI, Kaufer DI, Hendrickson R, et al. Degree of inhibition of cortical acetylcholinesterase activity and cognitive effects by donepezil treatment in Alzheimer’s disease. J Neurol Neurosurg Psychiatry. 2005;76(3):315-319.

21. Doody RS, Corey-Bloom J, Zhang R, et al. Safety and tolerability of donepezil at doses up to 20 mg/day: results from a pilot study in patients with Alzheimer’s disease. Drugs Aging. 2008;25(2):163-174.

22. Aricept [package insert]. Woodcliff Lake NJ: Eisai Co.; 2012.

23. Schwartz LM, Woloshin S. How the FDA forgot the evidence: the case of donepezil 23 mg. BMJ. 2012;344:e1086.-doi: 10.1136/bmj.e1086.

24. Saini SD, Schoenfeld P, Kaulback K, et al. Effect of medication dosing frequency on adherence in chronic diseases. Am J Manag Care. 2009;15(6):e22-e33.

25. Periclou A, Ventura D, Rao N, et al. Pharmacokinetic study of memantine in healthy and renally impaired subjects. Clin Pharmacol Ther. 2006;79(1):134-143.

26. Atri A, Shaughnessy LW, Locascio JJ, et al. Long-term course and effectiveness of combination therapy in Alzheimer disease. Alzheimer Dis Assoc Disord. 2008;22(3):209-221.

Article PDF
Author and Disclosure Information

Indrapal Singh, MD
Assistant Professor, Department of Geriatric Mental Health, Chhattrapati Shahuji Maharaj Medical University, Lucknow, Uttar Pradesh, India
George T. Grossberg, MD
Samuel W. Fordyce Professor, Department of Neurology and Psychiatry, Division of Geriatric Psychiatry, Saint Louis University School of Medicine, St. Louis, MO

Issue
Current Psychiatry - 11(06)
Publications
Topics
Page Number
20-29
Sections
Author and Disclosure Information

Indrapal Singh, MD
Assistant Professor, Department of Geriatric Mental Health, Chhattrapati Shahuji Maharaj Medical University, Lucknow, Uttar Pradesh, India
George T. Grossberg, MD
Samuel W. Fordyce Professor, Department of Neurology and Psychiatry, Division of Geriatric Psychiatry, Saint Louis University School of Medicine, St. Louis, MO

Author and Disclosure Information

Indrapal Singh, MD
Assistant Professor, Department of Geriatric Mental Health, Chhattrapati Shahuji Maharaj Medical University, Lucknow, Uttar Pradesh, India
George T. Grossberg, MD
Samuel W. Fordyce Professor, Department of Neurology and Psychiatry, Division of Geriatric Psychiatry, Saint Louis University School of Medicine, St. Louis, MO

Article PDF
Article PDF

Although cholinesterase inhibitors (ChEIs) and memantine at standard doses may slow the progression of Alzheimer’s disease (AD) as assessed by cognitive, functional, and global measures, this effect is relatively modest. For the estimated 5.4 million Americans with AD1—more than one-half of whom have moderate to severe disease2—there is a great need for new approaches to slow AD progression.

High doses of donepezil or memantine may be the next step in achieving better results than standard pharmacologic treatments for AD. This article presents the possible benefits and indications for high doses of donepezil (23 mg/d) and memantine (28 mg/d) for managing moderate to severe AD and their safety and tolerability profiles.

Current treatments offer modest benefits

AD treatments comprise 2 categories: ChEIs (donepezil, rivastigmine, and galantamine) and the N-methyl-D-aspartate (NMDA) receptor antagonist memantine (Table 1).3,4 All ChEIs are FDA-approved for mild to moderate AD; donepezil also is approved for severe AD. Memantine is approved for moderate to severe AD, either alone or in combination with ChEIs. Until recently, the maximum FDA-approved doses were donepezil, 10 mg/d, and memantine, 20 mg/d. However, these dosages are associated with only modest beneficial effects in managing cognitive deterioration in patients with moderate to severe dementia.5,6 Studies have reported that combining a ChEI, such as donepezil, and memantine is well tolerated and may result in synergistic benefits by affecting different neurotransmitters in patients with moderate to severe AD.7,8

Recently, the FDA approved higher daily doses of donepezil (23 mg) and memantine (28 mg) for moderate to severe AD on the basis of positive phase III trial results.9-11 Donepezil, 23 mg/d, currently is marketed in the United States; the availability date for memantine, 28 mg/d, was undetermined at press time.

Table 1

FDA-approved treatments for Alzheimer’s disease

DrugMaximum daily doseMechanism of actionIndicationCommon side effects/comments
Tacrine160 mg/dChEIMild to moderate ADNausea, vomiting, loss of appetite, diarrhea. First ChEI to be approved, but rarely used because of associated possible hepatotoxicity
Donepezil10 mg/dChEIAll stages of ADNausea, vomiting, loss of appetite, diarrhea, sleep disturbance
Rivastigmine12 mg/dChEIMild to moderate ADNausea, vomiting, diarrhea, weight loss, loss of appetite
Galantamine24 mg/dChEIMild to moderate ADNausea, vomiting, diarrhea, weight loss, loss of appetite
Memantine20 mg/dNMDA receptor antagonistModerate to severe ADDizziness, headache, constipation, confusion
Galantamine ER24 mg/dChEIMild to moderate ADNausea, vomiting, diarrhea, weight loss, loss of appetite
Rivastigmine transdermal system9.5 mg/dChEIMild to moderate ADNausea, vomiting, diarrhea, weight loss, loss of appetite
Donepezil 2323 mg/dChEIModerate to severe ADNausea, vomiting, diarrhea
Memantine ER28 mg/dNMDA receptor antagonistModerate to severe ADDizziness, headache, constipation, confusion
AD: Alzheimer’s disease; ChEI: cholinesterase inhibitor; ER: extended release; NMDA: N-methyl-D-aspartate
Source: References 3,4

High-dose donepezil (23 mg/d)

Cognitive decline with AD has been associated with increasing loss of cholinergic neurons and cholinergic activities, particularly in areas associated with memory/cognition and learning, including cortical areas involving the temporal lobe, hippocampus, and nucleus basalis of Meynert.12-14 In addition, evidence suggests that increasing levels of acetylcholine by using ChEIs can enhance cognitive function.13,15

Donepezil is a selective, reversible ChEI believed to enhance central cholinergic function.15 Randomized clinical trials assessing dose-response with donepezil, 5 mg/d and 10 mg/d, have demonstrated more benefit in cognition with either dose than placebo. The 10 mg/d dose was more effective than 5 mg/d in patients with mild to moderate and severe AD.16-18 In patients with advanced AD who are stable on 5 mg/d, increasing to 10 mg/d could slow the progression of cognitive decline.18

Rationale for higher doses. Positron emission tomography studies have shown that at stable doses of donepezil, 5 mg/d or 10 mg/d, average cortical acetylcholinesterase (AChE) inhibition was <30%.19,20 Based on these findings, researchers thought that cortical AChE inhibition may be suboptimal with donepezil, 10 mg/d, and that higher doses of ChEI may be required in patients with more advanced AD—and therefore more cholinergic loss—for adequate cholinesterase inhibition. In a pilot study of patients with mild to moderate AD, higher doses of donepezil (15 mg/d and 20 mg/d) were reported to be safe and well tolerated.21

The 23-mg/d donepezil formulation was developed to provide a higher dose administered once daily without a sharp rise in peak concentration. The FDA approved donepezil, 23 mg/d, for patients with moderate to severe AD on the basis of phase III trial results.9,22 In a randomized, double-blind, multicenter, head-to-head clinical trial, >1,400 patients with moderate to severe AD (Mini-Mental State Exam [MMSE]: 0 to 20) on a stable dose of donepezil, 10 mg/d, for ≥3 months were randomly assigned to receive high-dose donepezil (23 mg/d) or standard-dose donepezil (10 mg/d) for 24 weeks.9,22 Patients in the 23-mg/d group showed a statistically significant improvement in cognition compared with the 10-mg/d group. The difference between groups on a measure of global improvement was not significant.9,22 However, in a post-hoc analysis, it was demonstrated that a subgroup of patients with more severe cognitive impairment (baseline MMSE: 0 to 16), showed significant improvement in cognition as well as global functioning.9

 

 

Overall, treatment-emergent adverse events (TEAEs) during the study were higher in patients receiving 23 mg/d (74%) than those receiving 10 mg/d (64%). The most common TEAEs in the 23-mg/d and 10-mg/d groups were nausea (12% vs 3%, respectively), vomiting (9% vs 3%), and diarrhea (8% vs 5%) (Table 2).22 These gastrointestinal adverse effects were more frequent during the first month of treatment and were relatively infrequent beyond 1 month. Serious TEAEs, such as falls, urinary tract infection, pneumonia, syncope, aggression, and confusional state, were noted in a similar proportion of patients in the 23-mg/d and 10-mg/d groups; most of these were considered unrelated to treatment. No drug-related deaths occurred during the study. High-dose (23 mg/d) donepezil generally was well tolerated, with a typical ChEI safety profile but superior efficacy.

A recent commentary discussed the issue of effect size and whether a 2.2-point difference on a 100-point scale (the Severe Impairment Battery [SIB]) is clinically meaningful.23 As with all anti-dementia therapies, in any cohort some patients will gain considerably more than 2.2 points on the SIB, which is clinically significant. A 6-month trial is recommended to identify these optimal responders.

Table 2

High-dose vs standard-dose donepezil: Treatment-emergent adverse events

Adverse eventDonepezil, 23 mg/dDonepezil,10 mg/d
Nausea12%3%
Vomiting9%3%
Diarrhea8%5%
Anorexia5%2%
Dizziness5%3%
Weight decrease5%3%
Headache4%3%
Insomnia3%2%
Urinary incontinence3%1%
Fatigue2%1%
Weakness2%1%
Somnolence2%1%
Contusion2%0%
Source: Reference 22

High-dose memantine

Memantine is an NMDA receptor antagonist, which works on glutamate, an ubiquitous neurotransmitter in the brain that serves many functions. For reasons that are not fully understood, in AD glutamate becomes excitotoxic and causes neuronal death.

Some researchers have hypothesized that if safe and well tolerated, a memantine dose >20 mg/d may have better efficacy than a lower dose. Memantine’s manufacturer has developed an extended-release (ER), once-daily formulation of memantine, 28 mg/d, to improve adherence and possibly increase efficacy.10,11 Because of memantine ER’s relatively slow absorption rate and longer median Tmax, of 12 hours, there is minimal fluctuation in plasma levels during steady-state dosing intervals compared with the immediate-release (IR) formulation.10

In a phase I study of 24 healthy volunteers that investigated the safety, tolerability, and pharmacokinetics of memantine ER, 28 mg/d, TEAEs were mild; the most common were headache, somnolence, and dizziness.10 During memantine treatment, there were no serious adverse events, potential significant changes in patients’ vital signs, or deaths.

Memantine ER plus ChEI. A multicenter, multinational, randomized, double-blind study compared memantine ER, 28 mg/d, and placebo in patients with moderate to severe AD (MMSE: 3 to 14).11 All patients were receiving concurrent, stable ChEI treatment (donepezil, rivastigmine, or galantamine) for ≥3 months before the study. Patients treated with memantine ER, 28 mg/d, and ChEI (n = 342) showed a significant improvement compared with the placebo/ChEI group (n = 335) in cognition and global functioning. Patients receiving memantine/ChEI also showed statistically significant benefits on behavior and verbal fluency testing compared with patients receiving placebo/ChEI. Memantine was well tolerated; most adverse events were mild or moderate. The most common adverse events in the memantine/ChEI group that occurred at a higher rate relative to the placebo/ChEI group were headache (5.6% vs 5.1%, respectively), diarrhea (5.0% vs 3.9%), and dizziness (4.7% vs 1.5%). There were no deaths related to memantine (Table 3).11

Memantine ER, 28 mg/d, may be tolerated better than the IR formulation because of less plasma level fluctuation during the steady-state dosing interval. Also, memantine ER, 28 mg/d, may offer better efficacy over memantine IR, 20 mg/d, because of dose-dependent cognitive, global, and behavioral effects. In addition, once-daily dosing of memantine ER may improve adherence compared with the IR formulation.24

In patients with severe renal impairment, dosage of memantine IR should be reduced from 20 mg/d to 10 mg/d.25 However, there is no available information regarding the dosing, safety, and tolerability of memantine ER, 28 mg/d, in patients with renal disease.

Table3

High-dose memantine: Treatment-emergent adverse eventsa

Adverse eventPlacebo (n = 335)Memantine ER (n = 341)
Any TEAE214 (63.9%)214 (62.8%)
Fall26 (7.8%)19 (5.6%)
Urinary tract infection24 (7.2%)19 (5.6%)
Headache17 (5.1%)19 (5.6%)
Diarrhea13 (3.9%)17 (5.0%)
Dizziness5 (1.5%)16 (4.7%)
Influenza9 (2.7%)15 (4.4%)
Insomnia16 (4.8%)14 (4.1%)
Agitation15 (4.5%)14 (4.1%)
Hypertension8 (2.4%)13 (3.8%)
Anxiety9 (2.7%)12 (3.5%)
Depression5 (1.5%)11 (3.2%)
Weight increased3 (0.9%)11 (3.2%)
Constipation4 (1.2%)10 (2.9%)
Somnolence4 (1.2%)10 (2.9%)
Back pain2 (0.6%)9 (2.6%)
Aggression5 (1.5%)8 (2.3%)
Hypotension5 (1.5%)7 (2.1%)
Vomiting4 (1.2%)7 (2.1%)
Abdominal pain2 (0.6%)7 (2.1%)
Nasopharyngitis10 (3.0%)6 (1.8%)
Confusional state7 (2.1%)6 (1.8%)
Weight decreased11 (3.3%)5 (1.5%)
Nausea7 (2.1%)5 (1.5%)
Irritability8 (2.4%)4 (1.2%)
Cough8 (2.4%)3 (0.9%)
aData [n (%)] include all adverse events experienced by ≥2% patients in either group (safety population). Adverse events that were experienced at twice the rate in 1 group compared with the other are indicated by bold type
ER: extended-release (28 mg); TEAE: treatment-emergent adverse event
Source: Reference 11
 

 

Recommendations

Because there are few FDA-approved treatments for AD, higher doses of donepezil or memantine may be an option for patients who have “maxed out” on their AD therapy or no longer respond to lower doses. Higher doses of donepezil (23 mg/d) and memantine (28 mg/d) could improve medication adherence because both are once-daily preparations. In clinical trials, donepezil, 23 mg/d, was more effective than donepezil, 10 mg/d.9 Whether memantine ER, 28 mg/d, is superior to memantine IR, 20 mg/d, needs to be investigated in head-to-head, double-blind, controlled studies.

For patients with moderate to severe AD, donepezil, 23 mg, is associated with greater benefits in cognition compared with donepezil, 10 mg/d.9 Similarly, because of potentially superior efficacy because of a higher dose, memantine ER, 28 mg, might best help patients with moderate to severe AD, specifically those who either don’t respond or lose response to memantine IR, 20 mg/d. Combining a ChEI, such as donepezil, with memantine is associated with slower cognitive decline and short and long-term benefits on measures of cognition, activities of daily living, global outcome, and behavior.7,26 However, additional clinical trials are needed to assess the safety, tolerability, and efficacy of combination therapy with higher doses of donepezil and memantine ER.

Related Resources

  • Alzheimer’s Disease Education and Referral Center. www.nia.nih.gov/Alzheimers.
  • Lleó A, Greenberg SM, Growdon JH. Current pharmacotherapy for Alzheimer’s disease. Annu Rev Med. 2006;57:513-533.

Drug Brand Names

  • Donepezil • Aricept
  • Galantamine • Razadyne
  • Memantine • Namenda
  • Rivastigmine • Exelon
  • Tacrine • Cognex

Disclosures

Dr. Grossberg’s academic department has received research funding from Forest Pharmaceuticals and Pfizer Inc. Dr. Grossberg has received grant/research support from Baxter BioScience, Forest Pharmaceuticals, Janssen, the National Institutes of Health, Novartis, and Pfizer, Inc.; is a consultant to Baxter BioScience, Forest Pharmaceuticals, Merck, Novartis, and Otsuka; and is on the Safety Monitoring Committee for Merck.

Dr. Singh reports no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.

Although cholinesterase inhibitors (ChEIs) and memantine at standard doses may slow the progression of Alzheimer’s disease (AD) as assessed by cognitive, functional, and global measures, this effect is relatively modest. For the estimated 5.4 million Americans with AD1—more than one-half of whom have moderate to severe disease2—there is a great need for new approaches to slow AD progression.

High doses of donepezil or memantine may be the next step in achieving better results than standard pharmacologic treatments for AD. This article presents the possible benefits and indications for high doses of donepezil (23 mg/d) and memantine (28 mg/d) for managing moderate to severe AD and their safety and tolerability profiles.

Current treatments offer modest benefits

AD treatments comprise 2 categories: ChEIs (donepezil, rivastigmine, and galantamine) and the N-methyl-D-aspartate (NMDA) receptor antagonist memantine (Table 1).3,4 All ChEIs are FDA-approved for mild to moderate AD; donepezil also is approved for severe AD. Memantine is approved for moderate to severe AD, either alone or in combination with ChEIs. Until recently, the maximum FDA-approved doses were donepezil, 10 mg/d, and memantine, 20 mg/d. However, these dosages are associated with only modest beneficial effects in managing cognitive deterioration in patients with moderate to severe dementia.5,6 Studies have reported that combining a ChEI, such as donepezil, and memantine is well tolerated and may result in synergistic benefits by affecting different neurotransmitters in patients with moderate to severe AD.7,8

Recently, the FDA approved higher daily doses of donepezil (23 mg) and memantine (28 mg) for moderate to severe AD on the basis of positive phase III trial results.9-11 Donepezil, 23 mg/d, currently is marketed in the United States; the availability date for memantine, 28 mg/d, was undetermined at press time.

Table 1

FDA-approved treatments for Alzheimer’s disease

DrugMaximum daily doseMechanism of actionIndicationCommon side effects/comments
Tacrine160 mg/dChEIMild to moderate ADNausea, vomiting, loss of appetite, diarrhea. First ChEI to be approved, but rarely used because of associated possible hepatotoxicity
Donepezil10 mg/dChEIAll stages of ADNausea, vomiting, loss of appetite, diarrhea, sleep disturbance
Rivastigmine12 mg/dChEIMild to moderate ADNausea, vomiting, diarrhea, weight loss, loss of appetite
Galantamine24 mg/dChEIMild to moderate ADNausea, vomiting, diarrhea, weight loss, loss of appetite
Memantine20 mg/dNMDA receptor antagonistModerate to severe ADDizziness, headache, constipation, confusion
Galantamine ER24 mg/dChEIMild to moderate ADNausea, vomiting, diarrhea, weight loss, loss of appetite
Rivastigmine transdermal system9.5 mg/dChEIMild to moderate ADNausea, vomiting, diarrhea, weight loss, loss of appetite
Donepezil 2323 mg/dChEIModerate to severe ADNausea, vomiting, diarrhea
Memantine ER28 mg/dNMDA receptor antagonistModerate to severe ADDizziness, headache, constipation, confusion
AD: Alzheimer’s disease; ChEI: cholinesterase inhibitor; ER: extended release; NMDA: N-methyl-D-aspartate
Source: References 3,4

High-dose donepezil (23 mg/d)

Cognitive decline with AD has been associated with increasing loss of cholinergic neurons and cholinergic activities, particularly in areas associated with memory/cognition and learning, including cortical areas involving the temporal lobe, hippocampus, and nucleus basalis of Meynert.12-14 In addition, evidence suggests that increasing levels of acetylcholine by using ChEIs can enhance cognitive function.13,15

Donepezil is a selective, reversible ChEI believed to enhance central cholinergic function.15 Randomized clinical trials assessing dose-response with donepezil, 5 mg/d and 10 mg/d, have demonstrated more benefit in cognition with either dose than placebo. The 10 mg/d dose was more effective than 5 mg/d in patients with mild to moderate and severe AD.16-18 In patients with advanced AD who are stable on 5 mg/d, increasing to 10 mg/d could slow the progression of cognitive decline.18

Rationale for higher doses. Positron emission tomography studies have shown that at stable doses of donepezil, 5 mg/d or 10 mg/d, average cortical acetylcholinesterase (AChE) inhibition was <30%.19,20 Based on these findings, researchers thought that cortical AChE inhibition may be suboptimal with donepezil, 10 mg/d, and that higher doses of ChEI may be required in patients with more advanced AD—and therefore more cholinergic loss—for adequate cholinesterase inhibition. In a pilot study of patients with mild to moderate AD, higher doses of donepezil (15 mg/d and 20 mg/d) were reported to be safe and well tolerated.21

The 23-mg/d donepezil formulation was developed to provide a higher dose administered once daily without a sharp rise in peak concentration. The FDA approved donepezil, 23 mg/d, for patients with moderate to severe AD on the basis of phase III trial results.9,22 In a randomized, double-blind, multicenter, head-to-head clinical trial, >1,400 patients with moderate to severe AD (Mini-Mental State Exam [MMSE]: 0 to 20) on a stable dose of donepezil, 10 mg/d, for ≥3 months were randomly assigned to receive high-dose donepezil (23 mg/d) or standard-dose donepezil (10 mg/d) for 24 weeks.9,22 Patients in the 23-mg/d group showed a statistically significant improvement in cognition compared with the 10-mg/d group. The difference between groups on a measure of global improvement was not significant.9,22 However, in a post-hoc analysis, it was demonstrated that a subgroup of patients with more severe cognitive impairment (baseline MMSE: 0 to 16), showed significant improvement in cognition as well as global functioning.9

 

 

Overall, treatment-emergent adverse events (TEAEs) during the study were higher in patients receiving 23 mg/d (74%) than those receiving 10 mg/d (64%). The most common TEAEs in the 23-mg/d and 10-mg/d groups were nausea (12% vs 3%, respectively), vomiting (9% vs 3%), and diarrhea (8% vs 5%) (Table 2).22 These gastrointestinal adverse effects were more frequent during the first month of treatment and were relatively infrequent beyond 1 month. Serious TEAEs, such as falls, urinary tract infection, pneumonia, syncope, aggression, and confusional state, were noted in a similar proportion of patients in the 23-mg/d and 10-mg/d groups; most of these were considered unrelated to treatment. No drug-related deaths occurred during the study. High-dose (23 mg/d) donepezil generally was well tolerated, with a typical ChEI safety profile but superior efficacy.

A recent commentary discussed the issue of effect size and whether a 2.2-point difference on a 100-point scale (the Severe Impairment Battery [SIB]) is clinically meaningful.23 As with all anti-dementia therapies, in any cohort some patients will gain considerably more than 2.2 points on the SIB, which is clinically significant. A 6-month trial is recommended to identify these optimal responders.

Table 2

High-dose vs standard-dose donepezil: Treatment-emergent adverse events

Adverse eventDonepezil, 23 mg/dDonepezil,10 mg/d
Nausea12%3%
Vomiting9%3%
Diarrhea8%5%
Anorexia5%2%
Dizziness5%3%
Weight decrease5%3%
Headache4%3%
Insomnia3%2%
Urinary incontinence3%1%
Fatigue2%1%
Weakness2%1%
Somnolence2%1%
Contusion2%0%
Source: Reference 22

High-dose memantine

Memantine is an NMDA receptor antagonist, which works on glutamate, an ubiquitous neurotransmitter in the brain that serves many functions. For reasons that are not fully understood, in AD glutamate becomes excitotoxic and causes neuronal death.

Some researchers have hypothesized that if safe and well tolerated, a memantine dose >20 mg/d may have better efficacy than a lower dose. Memantine’s manufacturer has developed an extended-release (ER), once-daily formulation of memantine, 28 mg/d, to improve adherence and possibly increase efficacy.10,11 Because of memantine ER’s relatively slow absorption rate and longer median Tmax, of 12 hours, there is minimal fluctuation in plasma levels during steady-state dosing intervals compared with the immediate-release (IR) formulation.10

In a phase I study of 24 healthy volunteers that investigated the safety, tolerability, and pharmacokinetics of memantine ER, 28 mg/d, TEAEs were mild; the most common were headache, somnolence, and dizziness.10 During memantine treatment, there were no serious adverse events, potential significant changes in patients’ vital signs, or deaths.

Memantine ER plus ChEI. A multicenter, multinational, randomized, double-blind study compared memantine ER, 28 mg/d, and placebo in patients with moderate to severe AD (MMSE: 3 to 14).11 All patients were receiving concurrent, stable ChEI treatment (donepezil, rivastigmine, or galantamine) for ≥3 months before the study. Patients treated with memantine ER, 28 mg/d, and ChEI (n = 342) showed a significant improvement compared with the placebo/ChEI group (n = 335) in cognition and global functioning. Patients receiving memantine/ChEI also showed statistically significant benefits on behavior and verbal fluency testing compared with patients receiving placebo/ChEI. Memantine was well tolerated; most adverse events were mild or moderate. The most common adverse events in the memantine/ChEI group that occurred at a higher rate relative to the placebo/ChEI group were headache (5.6% vs 5.1%, respectively), diarrhea (5.0% vs 3.9%), and dizziness (4.7% vs 1.5%). There were no deaths related to memantine (Table 3).11

Memantine ER, 28 mg/d, may be tolerated better than the IR formulation because of less plasma level fluctuation during the steady-state dosing interval. Also, memantine ER, 28 mg/d, may offer better efficacy over memantine IR, 20 mg/d, because of dose-dependent cognitive, global, and behavioral effects. In addition, once-daily dosing of memantine ER may improve adherence compared with the IR formulation.24

In patients with severe renal impairment, dosage of memantine IR should be reduced from 20 mg/d to 10 mg/d.25 However, there is no available information regarding the dosing, safety, and tolerability of memantine ER, 28 mg/d, in patients with renal disease.

Table3

High-dose memantine: Treatment-emergent adverse eventsa

Adverse eventPlacebo (n = 335)Memantine ER (n = 341)
Any TEAE214 (63.9%)214 (62.8%)
Fall26 (7.8%)19 (5.6%)
Urinary tract infection24 (7.2%)19 (5.6%)
Headache17 (5.1%)19 (5.6%)
Diarrhea13 (3.9%)17 (5.0%)
Dizziness5 (1.5%)16 (4.7%)
Influenza9 (2.7%)15 (4.4%)
Insomnia16 (4.8%)14 (4.1%)
Agitation15 (4.5%)14 (4.1%)
Hypertension8 (2.4%)13 (3.8%)
Anxiety9 (2.7%)12 (3.5%)
Depression5 (1.5%)11 (3.2%)
Weight increased3 (0.9%)11 (3.2%)
Constipation4 (1.2%)10 (2.9%)
Somnolence4 (1.2%)10 (2.9%)
Back pain2 (0.6%)9 (2.6%)
Aggression5 (1.5%)8 (2.3%)
Hypotension5 (1.5%)7 (2.1%)
Vomiting4 (1.2%)7 (2.1%)
Abdominal pain2 (0.6%)7 (2.1%)
Nasopharyngitis10 (3.0%)6 (1.8%)
Confusional state7 (2.1%)6 (1.8%)
Weight decreased11 (3.3%)5 (1.5%)
Nausea7 (2.1%)5 (1.5%)
Irritability8 (2.4%)4 (1.2%)
Cough8 (2.4%)3 (0.9%)
aData [n (%)] include all adverse events experienced by ≥2% patients in either group (safety population). Adverse events that were experienced at twice the rate in 1 group compared with the other are indicated by bold type
ER: extended-release (28 mg); TEAE: treatment-emergent adverse event
Source: Reference 11
 

 

Recommendations

Because there are few FDA-approved treatments for AD, higher doses of donepezil or memantine may be an option for patients who have “maxed out” on their AD therapy or no longer respond to lower doses. Higher doses of donepezil (23 mg/d) and memantine (28 mg/d) could improve medication adherence because both are once-daily preparations. In clinical trials, donepezil, 23 mg/d, was more effective than donepezil, 10 mg/d.9 Whether memantine ER, 28 mg/d, is superior to memantine IR, 20 mg/d, needs to be investigated in head-to-head, double-blind, controlled studies.

For patients with moderate to severe AD, donepezil, 23 mg, is associated with greater benefits in cognition compared with donepezil, 10 mg/d.9 Similarly, because of potentially superior efficacy because of a higher dose, memantine ER, 28 mg, might best help patients with moderate to severe AD, specifically those who either don’t respond or lose response to memantine IR, 20 mg/d. Combining a ChEI, such as donepezil, with memantine is associated with slower cognitive decline and short and long-term benefits on measures of cognition, activities of daily living, global outcome, and behavior.7,26 However, additional clinical trials are needed to assess the safety, tolerability, and efficacy of combination therapy with higher doses of donepezil and memantine ER.

Related Resources

  • Alzheimer’s Disease Education and Referral Center. www.nia.nih.gov/Alzheimers.
  • Lleó A, Greenberg SM, Growdon JH. Current pharmacotherapy for Alzheimer’s disease. Annu Rev Med. 2006;57:513-533.

Drug Brand Names

  • Donepezil • Aricept
  • Galantamine • Razadyne
  • Memantine • Namenda
  • Rivastigmine • Exelon
  • Tacrine • Cognex

Disclosures

Dr. Grossberg’s academic department has received research funding from Forest Pharmaceuticals and Pfizer Inc. Dr. Grossberg has received grant/research support from Baxter BioScience, Forest Pharmaceuticals, Janssen, the National Institutes of Health, Novartis, and Pfizer, Inc.; is a consultant to Baxter BioScience, Forest Pharmaceuticals, Merck, Novartis, and Otsuka; and is on the Safety Monitoring Committee for Merck.

Dr. Singh reports no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.

References

1. Alzheimer’s Association, Thies W, Bleiler L. 2011 Alzheimer’s disease facts and figures. Alzheimers Dement. 2011;7(2):208-244.

2. Hebert LE, Scherr PA, Bienias JL, et al. Alzheimer disease in the US population: prevalence estimates using the 2000 census. Arch Neurol. 2003;60(8):1119-1122.

3. Alzheimer’s Disease Education and Referral Center. Alzheimer’s disease medications. http://www.nia.nih.gov/alzheimers/publication/alzheimers-disease-medications-fact-sheet. Accessed May 10 2012.

4. Osborn GG, Saunders AV. Current treatments for patients with Alzheimer disease. J Am Osteopath Assoc. 2010;110(9 suppl 8):S16-S26.

5. Raina P, Santaguida P, Ismaila A, et al. Effectiveness of cholinesterase inhibitors and memantine for treating dementia: evidence review for a clinical practice guideline. Ann Intern Med. 2008;148(5):379-397.

6. Cummings JL. Alzheimer’s disease. N Engl J Med. 2004;351(1):56-67.

7. Tariot PN, Farlow MR, Grossberg GT, et al. Memantine Study Group. Memantine treatment in patients with moderate to severe Alzheimer disease already receiving donepezil: a randomized controlled trial. JAMA. 2004;291(3):317-324.

8. Xiong G, Doraiswamy PM. Combination drug therapy for Alzheimer’s disease: what is evidence-based and what is not? Geriatrics. 2005;60(6):22-26.

9. Farlow MR, Salloway S, Tariot PN, et al. Effectiveness and tolerability of high (23 mg/d) versus standard-dose (10 mg/d) donepezil in moderate to severe Alzheimer’s disease: a 24-week, randomized, double-blind study. Clin Ther. 2010;32(7):1234-1251.

10. Periclou A, Hu Y. Extended-release memantine capsule (28 mg once daily): a multiple dose, open-label study evaluating steady-state pharmacokinetics in healthy volunteers. Poster presented at 11th International Conference on Alzheimer’s Disease; July 26-31, 2008; Chicago, IL.

11. Grossberg GT, Manes F, Allegri R, et al. A multinational, randomized, double-blind, placebo-controlled, parallel-group trial of memantine extended-release capsule (28 mg, once daily) in patients with moderate to severe Alzheimer’s disease. Poster presented at 11th International Conference on Alzheimer’s Disease; July 26-31, 2008; Chicago, IL.

12. Geula C, Mesulam MM. Systematic regional variations in the loss of cortical cholinergic fibers in Alzheimer’s disease. Cereb Cortex. 1996;6(2):165-177.

13. Whitehouse PJ. The cholinergic deficit in Alzheimer’s disease. J Clin Psychiatry. 1998;59(suppl 13):19-22.

14. Teipel SJ, Flatz WH, Heinsen H, et al. Measurement of basal forebrain atrophy in Alzheimer’s disease using MRI. Brain. 2005;128(11):2626-2644.

15. Shintani EY, Uchida KM. Donepezil: an anticholinesterase inhibitor for Alzheimer’s disease. Am J Health Syst Pharm. 1997;54(24):2805-2810.

16. Homma A, Imai Y, Tago H, et al. Donepezil treatment of patients with severe Alzheimer’s disease in a Japanese population: results from a 24-week, double-blind, placebo-controlled, randomized trial. Dement Geriatr Cogn Disord. 2008;25(5):399-407.

17. Whitehead A, Perdomo C, Pratt RD, et al. Donepezil for the symptomatic treatment of patients with mild to moderate Alzheimer’s disease: a meta-analysis of individual patient data from randomised controlled trials. Int J Geriatr Psychiatry. 2004;19(7):624-633.

18. Nozawa M, Ichimiya Y, Nozawa E, et al. Clinical effects of high oral dose of donepezil for patients with Alzheimer’s disease in Japan. Psychogeriatrics. 2009;9(2):50-55.

19. Kuhl DE, Minoshima S, Frey KA, et al. Limited donepezil inhibition of acetylcholinesterase measured with positron emission tomography in living Alzheimer cerebral cortex. Ann Neurol. 2000;48(3):391-395.

20. Bohnen NI, Kaufer DI, Hendrickson R, et al. Degree of inhibition of cortical acetylcholinesterase activity and cognitive effects by donepezil treatment in Alzheimer’s disease. J Neurol Neurosurg Psychiatry. 2005;76(3):315-319.

21. Doody RS, Corey-Bloom J, Zhang R, et al. Safety and tolerability of donepezil at doses up to 20 mg/day: results from a pilot study in patients with Alzheimer’s disease. Drugs Aging. 2008;25(2):163-174.

22. Aricept [package insert]. Woodcliff Lake NJ: Eisai Co.; 2012.

23. Schwartz LM, Woloshin S. How the FDA forgot the evidence: the case of donepezil 23 mg. BMJ. 2012;344:e1086.-doi: 10.1136/bmj.e1086.

24. Saini SD, Schoenfeld P, Kaulback K, et al. Effect of medication dosing frequency on adherence in chronic diseases. Am J Manag Care. 2009;15(6):e22-e33.

25. Periclou A, Ventura D, Rao N, et al. Pharmacokinetic study of memantine in healthy and renally impaired subjects. Clin Pharmacol Ther. 2006;79(1):134-143.

26. Atri A, Shaughnessy LW, Locascio JJ, et al. Long-term course and effectiveness of combination therapy in Alzheimer disease. Alzheimer Dis Assoc Disord. 2008;22(3):209-221.

References

1. Alzheimer’s Association, Thies W, Bleiler L. 2011 Alzheimer’s disease facts and figures. Alzheimers Dement. 2011;7(2):208-244.

2. Hebert LE, Scherr PA, Bienias JL, et al. Alzheimer disease in the US population: prevalence estimates using the 2000 census. Arch Neurol. 2003;60(8):1119-1122.

3. Alzheimer’s Disease Education and Referral Center. Alzheimer’s disease medications. http://www.nia.nih.gov/alzheimers/publication/alzheimers-disease-medications-fact-sheet. Accessed May 10 2012.

4. Osborn GG, Saunders AV. Current treatments for patients with Alzheimer disease. J Am Osteopath Assoc. 2010;110(9 suppl 8):S16-S26.

5. Raina P, Santaguida P, Ismaila A, et al. Effectiveness of cholinesterase inhibitors and memantine for treating dementia: evidence review for a clinical practice guideline. Ann Intern Med. 2008;148(5):379-397.

6. Cummings JL. Alzheimer’s disease. N Engl J Med. 2004;351(1):56-67.

7. Tariot PN, Farlow MR, Grossberg GT, et al. Memantine Study Group. Memantine treatment in patients with moderate to severe Alzheimer disease already receiving donepezil: a randomized controlled trial. JAMA. 2004;291(3):317-324.

8. Xiong G, Doraiswamy PM. Combination drug therapy for Alzheimer’s disease: what is evidence-based and what is not? Geriatrics. 2005;60(6):22-26.

9. Farlow MR, Salloway S, Tariot PN, et al. Effectiveness and tolerability of high (23 mg/d) versus standard-dose (10 mg/d) donepezil in moderate to severe Alzheimer’s disease: a 24-week, randomized, double-blind study. Clin Ther. 2010;32(7):1234-1251.

10. Periclou A, Hu Y. Extended-release memantine capsule (28 mg once daily): a multiple dose, open-label study evaluating steady-state pharmacokinetics in healthy volunteers. Poster presented at 11th International Conference on Alzheimer’s Disease; July 26-31, 2008; Chicago, IL.

11. Grossberg GT, Manes F, Allegri R, et al. A multinational, randomized, double-blind, placebo-controlled, parallel-group trial of memantine extended-release capsule (28 mg, once daily) in patients with moderate to severe Alzheimer’s disease. Poster presented at 11th International Conference on Alzheimer’s Disease; July 26-31, 2008; Chicago, IL.

12. Geula C, Mesulam MM. Systematic regional variations in the loss of cortical cholinergic fibers in Alzheimer’s disease. Cereb Cortex. 1996;6(2):165-177.

13. Whitehouse PJ. The cholinergic deficit in Alzheimer’s disease. J Clin Psychiatry. 1998;59(suppl 13):19-22.

14. Teipel SJ, Flatz WH, Heinsen H, et al. Measurement of basal forebrain atrophy in Alzheimer’s disease using MRI. Brain. 2005;128(11):2626-2644.

15. Shintani EY, Uchida KM. Donepezil: an anticholinesterase inhibitor for Alzheimer’s disease. Am J Health Syst Pharm. 1997;54(24):2805-2810.

16. Homma A, Imai Y, Tago H, et al. Donepezil treatment of patients with severe Alzheimer’s disease in a Japanese population: results from a 24-week, double-blind, placebo-controlled, randomized trial. Dement Geriatr Cogn Disord. 2008;25(5):399-407.

17. Whitehead A, Perdomo C, Pratt RD, et al. Donepezil for the symptomatic treatment of patients with mild to moderate Alzheimer’s disease: a meta-analysis of individual patient data from randomised controlled trials. Int J Geriatr Psychiatry. 2004;19(7):624-633.

18. Nozawa M, Ichimiya Y, Nozawa E, et al. Clinical effects of high oral dose of donepezil for patients with Alzheimer’s disease in Japan. Psychogeriatrics. 2009;9(2):50-55.

19. Kuhl DE, Minoshima S, Frey KA, et al. Limited donepezil inhibition of acetylcholinesterase measured with positron emission tomography in living Alzheimer cerebral cortex. Ann Neurol. 2000;48(3):391-395.

20. Bohnen NI, Kaufer DI, Hendrickson R, et al. Degree of inhibition of cortical acetylcholinesterase activity and cognitive effects by donepezil treatment in Alzheimer’s disease. J Neurol Neurosurg Psychiatry. 2005;76(3):315-319.

21. Doody RS, Corey-Bloom J, Zhang R, et al. Safety and tolerability of donepezil at doses up to 20 mg/day: results from a pilot study in patients with Alzheimer’s disease. Drugs Aging. 2008;25(2):163-174.

22. Aricept [package insert]. Woodcliff Lake NJ: Eisai Co.; 2012.

23. Schwartz LM, Woloshin S. How the FDA forgot the evidence: the case of donepezil 23 mg. BMJ. 2012;344:e1086.-doi: 10.1136/bmj.e1086.

24. Saini SD, Schoenfeld P, Kaulback K, et al. Effect of medication dosing frequency on adherence in chronic diseases. Am J Manag Care. 2009;15(6):e22-e33.

25. Periclou A, Ventura D, Rao N, et al. Pharmacokinetic study of memantine in healthy and renally impaired subjects. Clin Pharmacol Ther. 2006;79(1):134-143.

26. Atri A, Shaughnessy LW, Locascio JJ, et al. Long-term course and effectiveness of combination therapy in Alzheimer disease. Alzheimer Dis Assoc Disord. 2008;22(3):209-221.

Issue
Current Psychiatry - 11(06)
Issue
Current Psychiatry - 11(06)
Page Number
20-29
Page Number
20-29
Publications
Publications
Topics
Article Type
Display Headline
High-dose donepezil or memantine: Next step for Alzheimer’s disease?
Display Headline
High-dose donepezil or memantine: Next step for Alzheimer’s disease?
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media

Does bupropion exacerbate anxiety?

Article Type
Changed
Fri, 01/24/2020 - 11:00
Display Headline
Does bupropion exacerbate anxiety?

For many clinicians, bupropion is the “go-to” medication for treating depressed patients who smoke, have concerns about sexual dysfunction side effects, and/or worry about weight gain. Bupropion is FDA-approved for preventing seasonal major depressive episodes in patients with seasonal affective disorder and is indicated as a smoking cessation aid.

“Anxious depression”—defined as depression with high levels of anxiety—is associated with poorer outcomes than “non-anxious” depression.1 Prescribing medications for these patients can be challenging. Some clinicians believe that bupropion exacerbates anxiety and should not be used to treat patients who experience both anxiety and depression.

Reports from our patients and our cumulative clinical experience are key factors in developing expertise in selecting appropriate medications. When informing our patients about what to expect from medications, however, it can be useful to combine anecdotal evidence with knowledge of the facts or lack thereof. Are there data to support or contradict the idea that bupropion can cause anxiety while treating depression?

What the research shows

The drug manufacturer reports a “substantial proportion of patients treated with Wellbutrin experience some degree of increased restlessness, agitation, anxiety, and insomnia, especially shortly after initiation of treatment.”2

In 2001, Rush et al3 published the results of a 16-week study (n=248) assessing pre-treatment anxiety levels and response to sertraline or bupropion. The authors concluded that anxious and depressed patients who received sertraline didn’t experience a superior anxiolytic or antidepressant response compared with bupropion.3 The same authors came to similar conclusions in a retrospective analysis of a pair of 8-week randomized, controlled, double-blind trials of selective serotonin reuptake inhibitors (SSRIs) and bupropion.4

In 2001, Nieuwstraten et al5 compared bupropion with SSRIs for treating depression by reviewing several randomized, double-blind, controlled trials. The relative risk of developing “anxiety/agitation” was 1.32 (95% confidence interval, 0.85 to 2.04), which was not statistically significant.

In a 2008 meta-analysis, Papakostas et al6 pooled individual patient data from 10 randomized, double-blind, placebo-controlled trials. Their aim was to compare the efficacy of bupropion to SSRIs in treating “anxious depression.” They found no difference in timing or degree of improvement in anxiety symptoms between groups based on Hamilton Anxiety Scale or Hamilton Depression Rating Scale—Anxiety-Somatization (HDRS-AS) scores. The authors recommended that antidepressant choice should not be based on concerns about worsening anxiety symptoms in depressed patients.6

Another meta-analysis by Papakostas et al7 of the same 10 randomized, double-blind, placebo-controlled trials suggested SSRIs may confer an advantage over bupropion in treating a subset of patients with “anxious depression,” which they defined as a HDRS-AS score ≥7. The authors noted the advantage was statistically significant, although “modest.”

Other smaller studies suggest that bupropion does not increase anxiety.8,9 A pilot study (N = 24, no placebo control) concluded that bupropion XL was comparable to escitalopram in treating anxiety in outpatients with generalized anxiety disorder.8

Because designing and executing drug trials can be expensive, it is not surprising that most of the evidence cited above derives from pharmaceutical company-sponsored or industry-affiliated work. As such, we should evaluate available evidence within the context of what we hear from and observe in our patients.

Our opinion

When assessing patients with depression and anxiety, we must carefully evaluate symptoms to distinguish between depression with associated anxiety symptoms and depression with a comorbid anxiety disorder.

If a patient suffers from depression with associated anxiety symptoms (“anxious depression”), keep in mind that although some data demonstrate a superior response to SSRIs, other studies show no difference in effect. Some research—albeit smaller, less compelling studies—suggests that bupropion may decrease anxiety.

If your patient suffers from comorbid depression and an anxiety disorder, bupropion would not be a first-line choice because it is not FDA-approved to treat anxiety disorders. Although it is possible that anxiety/agitation could result from bupropion use, there is not sufficient data to support its reputation as ”anxiogenic.”

What is your experience?

Do you agree with the authors? Send comments to [email protected] or share your thoughts on http://www.facebook.com/CurrentPsychiatry.

 

Related Resource

 

  • American Psychiatric Association. Mixed anxiety-depressive disorder. Diagnostic and statistical manual of mental disorders, 4th ed, text rev. Washington, DC: American Psychiatric Association; 2000:780-781.

Drug Brand Names

 

  • Bupropion • Wellbutrin, Zyban
  • Escitalopram • Lexapro
  • Sertraline • Zoloft

Disclosure

The authors report no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.

References

 

1. Fava M, Rush AJ, Alpert JE, et al. Difference in treatment outcome in outpatients with anxious versus nonanxious depression: a STAR*D report. Am J Psychiatry. 2008;165(3):342-351.

2. Wellbutrin [package insert]. Research Triangle Park NC: GlaxoSmithKline; 2008.

3. Rush AJ, Trivedi MH, Carmody TJ, et al. Response in relation to baseline anxiety levels in major depressive disorder treated with bupropion sustained release or sertraline. Neuropsychopharmacology. 2001;25(1):131-138.

4. Trivedi MH, Rush AJ, Carmody TJ, et al. Do bupropion SR and sertraline differ in their effects on anxiety in depressed patients? J Clin Psychiatry. 2001;62(10):776-781.

5. Nieuwstraten CE, Dolovich LR. Bupropion versus selective serotonin-reuptake inhibitors for treatment of depression. Ann Pharmacother. 2001;35(12):1608-1613.

6. Papakostas GI, Trivedi MH, Alpert JE, et al. Efficacy of bupropion and the selective serotonin reuptake inhibitors in the treatment of anxiety symptoms in major depressive disorder: a meta-analysis of individual patient data from 10 double-blind, randomized clinical trials. J Psychiatr Res. 2008;42(2):134-140.

7. Papakostas GI, Stahl SM, Krishen A, et al. Efficacy of bupropion and the selective serotonin reuptake inhibitors in the treatment of major depressive disorder with high levels of anxiety (anxious depression): a pooled analysis of 10 studies. J Clin Psychiatry. 2008;69(8):1287-1292.

8. Bystritsky A, Kerwin L, Feusner JD, et al. A pilot controlled trial of bupropion XL versus escitalopram in generalized anxiety disorder. Psychopharmacol Bull. 2008;41(1):46-51.

9. Feighner JP, Gardner EA, Johnston JA, et al. Double-blind comparison of bupropion and fluoxetine in depressed outpatients. J Clin Psychiatry. 1991;52(8):329-335.

Article PDF
Author and Disclosure Information

 

Courtney Nemeth Wiseman, MD, MPH
Dr. Wiseman is Instructor, Clinical, Contributing Services Faculty, Department of Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL

Jessica L. Gören, PharmD, BCPP
Dr. Gören is Associate Professor, Department of Pharmacy Practice, University of Rhode Island, Kingston, RI; Senior Clinical Pharmacist Specialist, Department of Pharmacy, Cambridge Health Alliance; and Instructor in Psychiatry, Harvard Medical School, Boston, MA

Issue
Current Psychiatry - 11(06)
Publications
Topics
Page Number
E3-E4
Sections
Author and Disclosure Information

 

Courtney Nemeth Wiseman, MD, MPH
Dr. Wiseman is Instructor, Clinical, Contributing Services Faculty, Department of Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL

Jessica L. Gören, PharmD, BCPP
Dr. Gören is Associate Professor, Department of Pharmacy Practice, University of Rhode Island, Kingston, RI; Senior Clinical Pharmacist Specialist, Department of Pharmacy, Cambridge Health Alliance; and Instructor in Psychiatry, Harvard Medical School, Boston, MA

Author and Disclosure Information

 

Courtney Nemeth Wiseman, MD, MPH
Dr. Wiseman is Instructor, Clinical, Contributing Services Faculty, Department of Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL

Jessica L. Gören, PharmD, BCPP
Dr. Gören is Associate Professor, Department of Pharmacy Practice, University of Rhode Island, Kingston, RI; Senior Clinical Pharmacist Specialist, Department of Pharmacy, Cambridge Health Alliance; and Instructor in Psychiatry, Harvard Medical School, Boston, MA

Article PDF
Article PDF

For many clinicians, bupropion is the “go-to” medication for treating depressed patients who smoke, have concerns about sexual dysfunction side effects, and/or worry about weight gain. Bupropion is FDA-approved for preventing seasonal major depressive episodes in patients with seasonal affective disorder and is indicated as a smoking cessation aid.

“Anxious depression”—defined as depression with high levels of anxiety—is associated with poorer outcomes than “non-anxious” depression.1 Prescribing medications for these patients can be challenging. Some clinicians believe that bupropion exacerbates anxiety and should not be used to treat patients who experience both anxiety and depression.

Reports from our patients and our cumulative clinical experience are key factors in developing expertise in selecting appropriate medications. When informing our patients about what to expect from medications, however, it can be useful to combine anecdotal evidence with knowledge of the facts or lack thereof. Are there data to support or contradict the idea that bupropion can cause anxiety while treating depression?

What the research shows

The drug manufacturer reports a “substantial proportion of patients treated with Wellbutrin experience some degree of increased restlessness, agitation, anxiety, and insomnia, especially shortly after initiation of treatment.”2

In 2001, Rush et al3 published the results of a 16-week study (n=248) assessing pre-treatment anxiety levels and response to sertraline or bupropion. The authors concluded that anxious and depressed patients who received sertraline didn’t experience a superior anxiolytic or antidepressant response compared with bupropion.3 The same authors came to similar conclusions in a retrospective analysis of a pair of 8-week randomized, controlled, double-blind trials of selective serotonin reuptake inhibitors (SSRIs) and bupropion.4

In 2001, Nieuwstraten et al5 compared bupropion with SSRIs for treating depression by reviewing several randomized, double-blind, controlled trials. The relative risk of developing “anxiety/agitation” was 1.32 (95% confidence interval, 0.85 to 2.04), which was not statistically significant.

In a 2008 meta-analysis, Papakostas et al6 pooled individual patient data from 10 randomized, double-blind, placebo-controlled trials. Their aim was to compare the efficacy of bupropion to SSRIs in treating “anxious depression.” They found no difference in timing or degree of improvement in anxiety symptoms between groups based on Hamilton Anxiety Scale or Hamilton Depression Rating Scale—Anxiety-Somatization (HDRS-AS) scores. The authors recommended that antidepressant choice should not be based on concerns about worsening anxiety symptoms in depressed patients.6

Another meta-analysis by Papakostas et al7 of the same 10 randomized, double-blind, placebo-controlled trials suggested SSRIs may confer an advantage over bupropion in treating a subset of patients with “anxious depression,” which they defined as a HDRS-AS score ≥7. The authors noted the advantage was statistically significant, although “modest.”

Other smaller studies suggest that bupropion does not increase anxiety.8,9 A pilot study (N = 24, no placebo control) concluded that bupropion XL was comparable to escitalopram in treating anxiety in outpatients with generalized anxiety disorder.8

Because designing and executing drug trials can be expensive, it is not surprising that most of the evidence cited above derives from pharmaceutical company-sponsored or industry-affiliated work. As such, we should evaluate available evidence within the context of what we hear from and observe in our patients.

Our opinion

When assessing patients with depression and anxiety, we must carefully evaluate symptoms to distinguish between depression with associated anxiety symptoms and depression with a comorbid anxiety disorder.

If a patient suffers from depression with associated anxiety symptoms (“anxious depression”), keep in mind that although some data demonstrate a superior response to SSRIs, other studies show no difference in effect. Some research—albeit smaller, less compelling studies—suggests that bupropion may decrease anxiety.

If your patient suffers from comorbid depression and an anxiety disorder, bupropion would not be a first-line choice because it is not FDA-approved to treat anxiety disorders. Although it is possible that anxiety/agitation could result from bupropion use, there is not sufficient data to support its reputation as ”anxiogenic.”

What is your experience?

Do you agree with the authors? Send comments to [email protected] or share your thoughts on http://www.facebook.com/CurrentPsychiatry.

 

Related Resource

 

  • American Psychiatric Association. Mixed anxiety-depressive disorder. Diagnostic and statistical manual of mental disorders, 4th ed, text rev. Washington, DC: American Psychiatric Association; 2000:780-781.

Drug Brand Names

 

  • Bupropion • Wellbutrin, Zyban
  • Escitalopram • Lexapro
  • Sertraline • Zoloft

Disclosure

The authors report no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.

For many clinicians, bupropion is the “go-to” medication for treating depressed patients who smoke, have concerns about sexual dysfunction side effects, and/or worry about weight gain. Bupropion is FDA-approved for preventing seasonal major depressive episodes in patients with seasonal affective disorder and is indicated as a smoking cessation aid.

“Anxious depression”—defined as depression with high levels of anxiety—is associated with poorer outcomes than “non-anxious” depression.1 Prescribing medications for these patients can be challenging. Some clinicians believe that bupropion exacerbates anxiety and should not be used to treat patients who experience both anxiety and depression.

Reports from our patients and our cumulative clinical experience are key factors in developing expertise in selecting appropriate medications. When informing our patients about what to expect from medications, however, it can be useful to combine anecdotal evidence with knowledge of the facts or lack thereof. Are there data to support or contradict the idea that bupropion can cause anxiety while treating depression?

What the research shows

The drug manufacturer reports a “substantial proportion of patients treated with Wellbutrin experience some degree of increased restlessness, agitation, anxiety, and insomnia, especially shortly after initiation of treatment.”2

In 2001, Rush et al3 published the results of a 16-week study (n=248) assessing pre-treatment anxiety levels and response to sertraline or bupropion. The authors concluded that anxious and depressed patients who received sertraline didn’t experience a superior anxiolytic or antidepressant response compared with bupropion.3 The same authors came to similar conclusions in a retrospective analysis of a pair of 8-week randomized, controlled, double-blind trials of selective serotonin reuptake inhibitors (SSRIs) and bupropion.4

In 2001, Nieuwstraten et al5 compared bupropion with SSRIs for treating depression by reviewing several randomized, double-blind, controlled trials. The relative risk of developing “anxiety/agitation” was 1.32 (95% confidence interval, 0.85 to 2.04), which was not statistically significant.

In a 2008 meta-analysis, Papakostas et al6 pooled individual patient data from 10 randomized, double-blind, placebo-controlled trials. Their aim was to compare the efficacy of bupropion to SSRIs in treating “anxious depression.” They found no difference in timing or degree of improvement in anxiety symptoms between groups based on Hamilton Anxiety Scale or Hamilton Depression Rating Scale—Anxiety-Somatization (HDRS-AS) scores. The authors recommended that antidepressant choice should not be based on concerns about worsening anxiety symptoms in depressed patients.6

Another meta-analysis by Papakostas et al7 of the same 10 randomized, double-blind, placebo-controlled trials suggested SSRIs may confer an advantage over bupropion in treating a subset of patients with “anxious depression,” which they defined as a HDRS-AS score ≥7. The authors noted the advantage was statistically significant, although “modest.”

Other smaller studies suggest that bupropion does not increase anxiety.8,9 A pilot study (N = 24, no placebo control) concluded that bupropion XL was comparable to escitalopram in treating anxiety in outpatients with generalized anxiety disorder.8

Because designing and executing drug trials can be expensive, it is not surprising that most of the evidence cited above derives from pharmaceutical company-sponsored or industry-affiliated work. As such, we should evaluate available evidence within the context of what we hear from and observe in our patients.

Our opinion

When assessing patients with depression and anxiety, we must carefully evaluate symptoms to distinguish between depression with associated anxiety symptoms and depression with a comorbid anxiety disorder.

If a patient suffers from depression with associated anxiety symptoms (“anxious depression”), keep in mind that although some data demonstrate a superior response to SSRIs, other studies show no difference in effect. Some research—albeit smaller, less compelling studies—suggests that bupropion may decrease anxiety.

If your patient suffers from comorbid depression and an anxiety disorder, bupropion would not be a first-line choice because it is not FDA-approved to treat anxiety disorders. Although it is possible that anxiety/agitation could result from bupropion use, there is not sufficient data to support its reputation as ”anxiogenic.”

What is your experience?

Do you agree with the authors? Send comments to [email protected] or share your thoughts on http://www.facebook.com/CurrentPsychiatry.

 

Related Resource

 

  • American Psychiatric Association. Mixed anxiety-depressive disorder. Diagnostic and statistical manual of mental disorders, 4th ed, text rev. Washington, DC: American Psychiatric Association; 2000:780-781.

Drug Brand Names

 

  • Bupropion • Wellbutrin, Zyban
  • Escitalopram • Lexapro
  • Sertraline • Zoloft

Disclosure

The authors report no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.

References

 

1. Fava M, Rush AJ, Alpert JE, et al. Difference in treatment outcome in outpatients with anxious versus nonanxious depression: a STAR*D report. Am J Psychiatry. 2008;165(3):342-351.

2. Wellbutrin [package insert]. Research Triangle Park NC: GlaxoSmithKline; 2008.

3. Rush AJ, Trivedi MH, Carmody TJ, et al. Response in relation to baseline anxiety levels in major depressive disorder treated with bupropion sustained release or sertraline. Neuropsychopharmacology. 2001;25(1):131-138.

4. Trivedi MH, Rush AJ, Carmody TJ, et al. Do bupropion SR and sertraline differ in their effects on anxiety in depressed patients? J Clin Psychiatry. 2001;62(10):776-781.

5. Nieuwstraten CE, Dolovich LR. Bupropion versus selective serotonin-reuptake inhibitors for treatment of depression. Ann Pharmacother. 2001;35(12):1608-1613.

6. Papakostas GI, Trivedi MH, Alpert JE, et al. Efficacy of bupropion and the selective serotonin reuptake inhibitors in the treatment of anxiety symptoms in major depressive disorder: a meta-analysis of individual patient data from 10 double-blind, randomized clinical trials. J Psychiatr Res. 2008;42(2):134-140.

7. Papakostas GI, Stahl SM, Krishen A, et al. Efficacy of bupropion and the selective serotonin reuptake inhibitors in the treatment of major depressive disorder with high levels of anxiety (anxious depression): a pooled analysis of 10 studies. J Clin Psychiatry. 2008;69(8):1287-1292.

8. Bystritsky A, Kerwin L, Feusner JD, et al. A pilot controlled trial of bupropion XL versus escitalopram in generalized anxiety disorder. Psychopharmacol Bull. 2008;41(1):46-51.

9. Feighner JP, Gardner EA, Johnston JA, et al. Double-blind comparison of bupropion and fluoxetine in depressed outpatients. J Clin Psychiatry. 1991;52(8):329-335.

References

 

1. Fava M, Rush AJ, Alpert JE, et al. Difference in treatment outcome in outpatients with anxious versus nonanxious depression: a STAR*D report. Am J Psychiatry. 2008;165(3):342-351.

2. Wellbutrin [package insert]. Research Triangle Park NC: GlaxoSmithKline; 2008.

3. Rush AJ, Trivedi MH, Carmody TJ, et al. Response in relation to baseline anxiety levels in major depressive disorder treated with bupropion sustained release or sertraline. Neuropsychopharmacology. 2001;25(1):131-138.

4. Trivedi MH, Rush AJ, Carmody TJ, et al. Do bupropion SR and sertraline differ in their effects on anxiety in depressed patients? J Clin Psychiatry. 2001;62(10):776-781.

5. Nieuwstraten CE, Dolovich LR. Bupropion versus selective serotonin-reuptake inhibitors for treatment of depression. Ann Pharmacother. 2001;35(12):1608-1613.

6. Papakostas GI, Trivedi MH, Alpert JE, et al. Efficacy of bupropion and the selective serotonin reuptake inhibitors in the treatment of anxiety symptoms in major depressive disorder: a meta-analysis of individual patient data from 10 double-blind, randomized clinical trials. J Psychiatr Res. 2008;42(2):134-140.

7. Papakostas GI, Stahl SM, Krishen A, et al. Efficacy of bupropion and the selective serotonin reuptake inhibitors in the treatment of major depressive disorder with high levels of anxiety (anxious depression): a pooled analysis of 10 studies. J Clin Psychiatry. 2008;69(8):1287-1292.

8. Bystritsky A, Kerwin L, Feusner JD, et al. A pilot controlled trial of bupropion XL versus escitalopram in generalized anxiety disorder. Psychopharmacol Bull. 2008;41(1):46-51.

9. Feighner JP, Gardner EA, Johnston JA, et al. Double-blind comparison of bupropion and fluoxetine in depressed outpatients. J Clin Psychiatry. 1991;52(8):329-335.

Issue
Current Psychiatry - 11(06)
Issue
Current Psychiatry - 11(06)
Page Number
E3-E4
Page Number
E3-E4
Publications
Publications
Topics
Article Type
Display Headline
Does bupropion exacerbate anxiety?
Display Headline
Does bupropion exacerbate anxiety?
Sections
PURLs Copyright

Disallow All Ads
Alternative CME
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Article PDF Media

Managing Type 2 Diabetes in Men

Article Type
Changed
Tue, 05/03/2022 - 15:57
Display Headline
Managing Type 2 Diabetes in Men

 

DISCLOSURES

Dr. Aguilar has disclosed that he has ongoing relationships with the following companies: Amylin Pharmaceuticals; Eli Lilly; Janssen Pharmaceuticals, Inc; Novo Nordisk, Inc; and Takeda Pharmaceuticals USA, Inc.

SUPPORT

This program is sponsored by the PCEC and is supported by funding from Novo Nordisk, Inc.

 

The prevalence of type 2 diabetes mellitus (T2DM) is similar in men and women (11.8% vs 10.8%, respectively), however there are gender differences that should be considered when developing a treatment plan (eg, cardiovascular risk, psychosocial factors, coping strategies, and the perception of benefit from self-care) when managing those diagnosed with this disease and those at risk for developing it.1 This article describes these differences in the context of two patients—one at risk for T2DM being seen by his health care provider for a routine physical examination, and one who has been treated for several years for T2DM and is being seen for a follow-up office visit. For each patient, the implications for treatment are discussed.

Men at Risk for Type 2 Diabetes Mellitus

 

JW is a 48-year-old white male being seen for a routine physical examination; he last saw a physician 6 years ago, also for a routine physical. He has no complaints and is taking no medications. Having divorced 7 years ago, he lives alone in an apartment and eats many of his meals at fast food restaurants. JW drinks 2 to 3 beers a night several times a week and more when he socializes with his friends 2 to 3 evenings per week. He smokes socially. His father has a 12-year history of T2DM. His mother has a 4-year history of essential hypertension and a 9-year history of chronic obstructive pulmonary disease.

Physical examination shows that JW is 5’11” tall, weighs 207 pounds (body mass index (BMI), 29 kg/m2), and has a 41” waist circumference; his blood pressure (BP) is 138/86 mm Hg and respiratory rate is 17 breaths/min. The remainder of his physical examination, including eye and neurologic exams, is normal. Laboratory results, including a screening glycated homoglobin (A1C), are pending.

Key Risk Factors for Type 2 Diabetes Mellitus in Men

This case is not an uncommon presentation of a middle-aged male who has several risk factors for diabetes (see Case Study 1 continued ). JW also has key risk factors for T2DM in men. The Monitoring of Trends and Determinants in Cardiovascular Disease (MONICA) Augsburg surveys identified 128 men and 85 women with T2DM.2 Increasing age and BMI, positive parental history of T2DM, and a low high-density lipoprotein cholesterol (HDL-C) level were independent risk factors predicting the development of T2DM in both men and women. However, several other factors posed a higher risk in men relative to women, including systolic BP (hazard ratio [HR], 1.16 per 10-mm Hg increase), regular smoking (HR, 1.75), and alcohol intake ≥ 40 g/d (HR, 1.95). (Note: 1 fluid ounce 80 proof alcohol ≈ 11 g ethanol; 12 fluid ounces beer [~5% alcohol] ≈ 14 g ethanol). After adjusting for these factors, a separate analysis (4424 men, 4380 women) showed that men who lived alone were more likely to develop T2DM than either men or women who did not live alone (HR, 1.69 in men vs 0.85 in women; P = .006).3 While the number of people with T2DM in MONICA was small, the results suggest that measuring BP, particularly systolic BP, and taking a smoking and alcohol history may be especially important in men.

With respect to alcohol intake, epidemiologic and randomized clinical trials have generally demonstrated an inverse relationship between moderate alcohol consumption (20 to 30 g/d) and the long-term risk of T2DM.2,4-7 Differences among studies in how patients were grouped preclude determination of the daily alcohol consumption that confers the greatest risk benefit, although one recent study conducted over 4 years indicates that the greatest benefit in diabetes risk reduction may occur when men who previously consumed 8

Other nutrition and lifestyle patterns also seem to be particularly beneficial in reducing the risk of T2DM in men. Survey data involving 22,921 Japanese men and 29,759 Japanese women followed over 5 years showed that fish and seafood intake was significantly associated with a decreased risk of T2DM in men but not in women.9 The odds ratio of developing T2DM for the highest quartile versus the lowest quartile of fish and seafood intake was 0.73 (P = .04 for trend). Additional analysis did not identify any significant association with the fat content of fish.

 

 

Results of the Health Professionals Follow-up Study provide evidence of benefit in lowering the risk of T2DM in men who consume high amounts of low-fat dairy products, whole grains, and magnesium ( TABLE 1 ). With respect to dairy food consumption, after 12 years of follow-up involving 1243 incident cases of T2DM, the relative risk (RR) of developing T2DM in men in the top quintile of dairy intake was 0.77 compared with those in the lowest quintile (P = .003 for trend).10 Men in the highest quintile consumed 4.1 servings of dairy food per day compared with 0.5 servings per day in the lowest quintile. Each serving-per-day increase in total dairy intake was associated with a 9% lower risk for T2DM, with a lower risk seen with consumption of low-fat vs high-fat dairy food. With respect to whole-grain intake, the RR of developing T2DM was 0.58 in men in the upper vs lower quintiles (3.2 vs 0.4 servings/d), although the effect was attenuated with BMI (P = .0006 for trend).11 Similar observations were made with respect to magnesium consumption; a RR of 0.76 for T2DM was observed in men with a median magnesium consumption of 457 mg/d compared with those who consumed 270 mg/d.12

TABLE 1

Suggestions for Men Who Are at Risk of or Have Been Diagnosed with Type 2 Diabetes Mellitus (T2DM)*

 

For men who are at risk:
  • Key targets
    • -Systolic BP
    • -Smoking cessation
    • -Alcohol consumption (moderate)
  • Promote healthy diet
    • -Fish/seafood
    • -Low-fat dairy products
    • -Whole grains
    • -Magnesium
For men who have been diagnosed:
  • Key targets
    • -BP
    • -Blood glucose
    • -HDL-C
  • Emphasize the importance of self-management
  • Provide ongoing education/information regarding the progressive nature of T2DM and the need to adjust treatment over time, potentially adding both oral and injectable therapies
  • Recommend a diabetes support group
BP, blood pressure; HDL-C, high-density lipoprotein cholesterol.
*These suggestions are in addition to developing and fostering a collaborative, patient-centered approach.

 

JW has the following risk factors for T2DM:

  • Overweight with central adiposity
  • Physical inactivity
  • First-degree relative with T2DM
  • Possible cardiovascular disease (CVD; hypertension, smoking)
  • High daily alcohol intake (10 to 20 g alcohol/beer x 2-3 beers/d = 20 to 60 g alcohol/d)
  • Poor nutrition
  • Lives alone

Plan:

  • Discuss above risk factors with JW
  • Repeat BP measurement at next visit; implement treatment if BP >140/90 mm Hg (130/80 mm Hg if T2DM is diagnosed)
  • Consider evaluation for alcohol/substance abuse
  • Evaluate for smoking cessation program
  • Nutrition referral for lifestyle and dietary management intervention

Working with men to avoid the development of T2DM is an important objective for family physicians. It is essential to identify men who are at increased risk, including those with prediabetes, provide education about the disease and its risk factors, and implement appropriate risk reduction strategies. Risk reduction strategies should focus on modifiable factors, such as body weight, physical activity, BP, blood lipids, blood glucose, and smoking. With JW, his motivation to “get back into shape” will help move the conversation toward achievable goals that can be set and modified over time. Other strategies that may be helpful in reducing the risk of developing T2DM in men include a moderate daily alcohol intake and a diet high in fish and seafood, low-fat dairy products, whole grains, and magnesium ( TABLE 1 ).

Once diagnosed with T2DM, there are risk management strategies that can be particularly helpful in men. These include strategies that target cardiovascular health, as well as those that consider the psychosocial and coping behaviors of men.

Risk of Complications in Men With Type 2 Diabetes Mellitus

 

MR is a 57-year-old African American male diagnosed with hypertension 5 years ago and T2DM 3 years ago (A1C, 8.2%). Treatment with lifestyle modification and metformin 1000 mg twice daily had lowered his A1C to between 6.8% and 7.1%. However, 9 months ago, MR hurt his knee, which prevents him from walking his usual 1 to 1.5 miles several days a week and doing yard work on the weekends.

Physical examination: BP, 126/78 mm Hg; body weight, 183 pounds (a 13 to 17 pound increase since the knee injury); waist circumference, 38” (BMI, 28 kg/m2); grade 1 retinopathy bilaterally; neurologic exam normal.

Laboratory: A1C, 7.8%; lipids normal except triglyceride level, 219 mg/dL; creatinine clearance (calculated), 69 mL/min; urine, 45 mg albumin/g creatinine.

MR’s self-measured fasting plasma glucose (FPG) has ranged from 121 to 143 mg/dL over the past month; isolated postprandial glucose (PPG) measurements show 194 to 258 mg/dL.

MR works as a vocational teacher at the local high school, and he teaches driver education after school. Review of his pharmacy records suggests his adherence over the past year has been: metformin (88%), hydrochlorothiazide (72%), and lisinopril (72%).

Assessment:

  • A1C level of 7.8% indicates an estimated average glucose (eAG) of 177 mg/dL13
    • –Mildly elevated FPG and PPG
    • –Evidence of microvascular disease (retinopathy, nephropathy)
    • –Creatinine clearance 69 mL/min and microalbuminuria indicate stage 2 chronic kidney disease14

In addition to referring MR for physical rehabilitation of his knee, you discuss with MR the need and options for intensifying his diabetes therapy.

Does the fact that MR is male affect your management plan?

 

 

In people diagnosed with T2DM, there are differences between men and women with respect to risk for cardiovascular and other comorbid diseases, as well as in their psychosocial well-being and coping strategies.

Risk for Cardiovascular Disease in Type 2 Diabetes Mellitus

 

A systematic literature review shows that men with T2DM generally fare better than women with T2DM regarding their risk for CVD. Men with T2DM have a 2- to 3-fold increase in the risk of developing coronary heart disease (CHD) compared with men without T2DM, whereas women with T2DM have a 4- to 6-fold increase in risk compared with women without T2DM.15 Compared with women with T2DM, men with T2DM also have a better prognosis after myocardial infarction (MI) and a lower risk of death overall from CVD. Possible reasons for these differences include a lower risk of hypertension, a less severe form of dyslipidemia, and a lower prevalence of obesity in men with T2DM compared with women with T2DM.15 These same reasons for observed differences between men and women were seen in a meta-analysis of 29 studies, where the RR of fatal MI in men with T2DM compared with women with T2DM was 0.68.16 Similar findings were seen in the Skaraborg Project, which involved 1116 Swedish patients with hypertension and/or T2DM.17 Compared with a healthy population, the age-adjusted HR for fatal MI was 1.9 for men with T2DM and 5.0 for women with T2DM over 8.1 years of follow-up (RR, 0.38 for men vs women). Analysis of the data indicated that these results were not explained by the more favorable survival rate in women without T2DM than in men without T2DM.17

Somewhat different results have been reported by the Italian Diabetes and Informatics Study Group in a slightly different T2DM population. This investigation involved men and women with T2DM (N = 11,644) who could have microvascular but not macrovascular disease.18 After 4 years of follow-up, the age-adjusted incident rates for first CHD event (composite of acute MI, coronary artery bypass grafting, percutaneous transluminal coronary angioplasty) were 28.8 per 1000 person-years in men and 23.3 per 1000 person-years in women. Incident rates (per 1000 person-years) of acute MI (10.3 vs 4.7), major CHD events (13.1 vs 5.8), and fatal CHD (2.6 vs 0.6) were all significantly more frequent in men than in women, respectively. Multivariate analysis showed that hypertension and A1C were additional risk factors for CHD in men; for each 20% increment above the A1C upper limit of normal, there was a 14% risk increase for CHD. The presence of microvascular complications increased risk by 20% in men and 35% in women. In this analysis, glycemic control and hypertension were found to be the predominant risk factors in men, while high triglyceride levels, low HDL-C levels, and microangiopathy were predominant in women.

Additional multivariate analyses provide greater insight into specific factors that affect the risk of CVD and outcomes in men with T2DM. One investigation compared men and women with T2DM who were normotensive without evidence of CVD but with microalbuminuria. After 4.7 years of follow-up, men were found to be at lower risk (RR, 0.12) for a composite of death, acute MI, unstable angina, coronary interventions, heart failure, cerebral ischemic stroke or transient ischemic attack, and peripheral artery disease.19 Other investigators have reported a lower risk of stroke, including fatal stroke, in men with T2DM compared with women with T2DM.20,21 For example, analysis of the General Practice Research Database identified 22,178 men and 19,621 women with T2DM between the ages of 35 and 89 years.20 The stroke rate per 1000 person-years across all ages was 10.82 (95% confidence interval (CI), 10.17-11.51) in men and 13.16 (95% CI, 12.40-13.97) in women. In men, the rate per 1000 person-years rose from 1.81 in the 35 to 44 year age group to 28.35 in men 85 years of age or older. Although the rate of stroke per 1000 person-years was lower in women than men in the 35 to 44 year age group (1.53 vs 1.81), the rate in women exceeded that of men in the 85 years of age or older group (32.20 vs 28.35).

Other Chronic Complications

Kidney disease is affected by blood lipids, specifically HDL-C, in men with T2DM. An investigation in men and women with T2DM with normoalbuminuria or microalbuminuria at baseline showed that a low HDL-C level was an independent predictor of progression to a more advanced stage of albuminuria over 4.3 years of follow-up (HR, 0.391 for men with normal HDL-C compared with men with low HDL-C). In women, no lipid parameters were associated with progression of albuminuria.22

 

 

While these investigations do not provide a clear picture of the differences regarding cardiovascular risk between men and women with T2DM, they suggest that men with T2DM have a lower risk of nonfatal and fatal CVD and stroke than do women with T2DM. However, the lower risk seen in men may be affected by the cardiovascular endpoints measured and the presence of microvascular disease. Possible independent risk factors for CVD in men with T2DM include hypertension, poor glycemic control, and low HDL-C.

 

Risk factors that place MR at greater risk for CVD compared with a woman with T2DM and therefore serve as key treatment targets include:

  • Hypertension—although controlled (126/78 mm Hg) with hydrochlorothiazide and lisinopril
  • Poor glycemic control—A1C, 7.8% (eAG, 177mg/dL)
    • –Increase physical activity—refer for knee rehabilitation
    • –Intensify glucose-lowering therapy by adding an additional glucose-lowering agent (eg, dipeptidyl peptidase-4 inhibitor, glucagon-like peptide-1 receptor agonist, thiazolidinedione, α-glucosidase inhibitor, sulfonylurea, glinide, or basal insulin)
  • Microalbuminuria (45 mg urinary albumin/g creatinine)—encourage better adherence to lisinopril; monitor renal function
  • Hypertriglyceridemia—initiate omega-3 fatty acid or extended-release niacin

Psychosocial Well-Being, Benefit of Self-Care, and Coping Strategies

Type 2 diabetes mellitus is a chronic disease with glycemic control largely determined by patient self-management, and the attitudes and beliefs of patients with T2DM are important factors to consider from diagnosis onward.23 There are important differences between men and women with T2DM regarding attitudes and beliefs. Published investigations provide some, although not entirely consistent, insight into these psychosocial differences between men and women with T2DM. These differences are summarized in TABLE 2 .24-32 Taking these differences into account when planning treatment and when communicating with and educating the patient is essential for improved patient self-management.

TABLE 2

Psychosocial and Coping Characteristics of Men with Type 2 Diabetes Mellitus (T2DM)24-32

 

Compared with women with T2DM, generally, men with T2DM:
  • Experience less diabetes-related distress and greater well-being
  • Are less likely to experience symptoms of depression
  • Experience a slower deterioration in physical function
  • Exercise more
  • Perceive less support from their healthcare team
  • Have lower expectations regarding the benefits of self-management
  • Are less informed about T2DM, particularly pharmacologic and nonpharmacologic treatment options
  • Exert less effort and employ fewer strategies to cope with T2DM
  • Have less adaptive attitudes toward T2DM
  • Are influenced more by symptoms of hypoglycemia and hyperglycemia
  • Believe they have more family and social support and are more influenced by such support
  • Fear losing control of their disease and resist being “policed” by their social support system

 

 

Key interventions for MR:

  • Maintain a dialogue and enhance collaboration with MR
  • Establish shared goals that are customized to incorporate MR’s personal goals
  • Problem solve with MR to identify ways he can better integrate the diabetes self-care objectives of dietary changes and blood glucose self-monitoring into his daily life
  • Emphasize that enhanced or greater disease control can be achieved by good self-management, including better adherence to the management plan
  • Remind MR that T2DM is a progressive disease that requires intermittent medication adjustments to keep pace with its progression
  • Build upon the belief that T2DM can be controlled by reminding MR that the disease was well controlled before his knee injury
    • –Focus on the importance of rehabilitating his knee
    • –Develop a rehabilitation plan
  • Provide informational support regarding options for intensifying diabetes therapy (eg, dipeptidyl peptidase-4 inhibitor, thiazolidinedione, glucagon-like peptide-1 receptor agonist, sulfonylurea, or insulin)
    • –Discuss MR’s needs and concerns, as well as barriers for each treatment option, particularly hypoglycemia and weight gain
    • –Provide instruction or educational materials regarding injection devices
    • –Involve the healthcare team, as appropriate
  • Keep the treatment regimen as simple as possible; consider pill combinations where appropriate

Summary

The growing epidemic of T2DM requires intervention to assist patients who have been diagnosed to better manage the disease, to reduce the risk of developing the disease in those who have not yet been diagnosed, and to manage the associated complications. In addition to individualizing interventions based on a patient’s needs, concerns, and capabilities, taking gender into account is necessary. In otherwise healthy people, several independent factors appear to pose a higher risk of T2DM in men relative to women, including systolic hypertension, regular smoking, and alcohol intake ≥ 40 g/d. At the same time, men achieve greater risk reduction from moderate daily alcohol intake and a diet high in fish and seafood, low-fat dairy products, whole grains, and magnesium.

Once diagnosed with T2DM, men generally fare better than women regarding the risk for CVD; they also have a better prognosis after MI and a lower risk of death overall from CVD. Possible independent risk factors for CVD in men with T2DM that are especially important may include hypertension, poor glycemic control, and low HDL-C levels. Psychosocial complications, such as depression, are less likely in men with T2DM. However, men expend less effort coping, are less likely to utilize healthcare services, and are less informed about treatment options. Although men have a lower expectation of the benefit of self-management, they find support from family and friends more helpful than do women, but they are fearful of losing control of their disease.

Taking these gender differences into account should prove helpful as family care physicians work with men to reduce their risk of developing T2DM and in helping men diagnosed with T2DM to better self-manage their disease.

References

1. Centers for Disease Control and Prevention. National diabetes fact sheet: national estimates and general information on diabetes and prediabetes in the United States, 2011. http://www.cdc.gov/diabetes/pubs/pdf/ndfs_2011.pdf. Published 2011. Accessed May 2, 2011.

2. Meisinger C, Thorand B, Schneider A, Stieber J, Doring A, Lowel H. Sex differences in risk factors for incident type 2 diabetes mellitus: the MONICA Augsburg cohort study. Arch Intern Med. 2002;162(1):82-89.

3. Meisinger C, Kandler U, Ladwig KH. Living alone is associated with an increased risk of type 2 diabetes mellitus in men but not women from the general population: the MONICA/KORA Augsburg Cohort Study. Psychosom Med. 2009;71(7):784-788.

4. Baliunas DO, Taylor BJ, Irving H, et al. Alcohol as a risk factor for type 2 diabetes: a systematic review and meta-analysis. Diabetes Care. 2009;32(11):2123-2132.

5. Mozaffarian D, Kamineni A, Carnethon M, Djoussé L, Mukamal KJ, Siscovick D. Lifestyle risk factors and new-onset diabetes mellitus in older adults: the cardiovascular health study. Arch Intern Med. 2009;169(8):798-807.

6. Joosten MM, Grobbee DE, van der A DL, Verschuren WM, Hendriks HF, Beulens JW. Combined effect of alcohol consumption and lifestyle behaviors on risk of type 2 diabetes. Am J Clin Nutr. 2010;91(6):1777-1783.

7. Gigleux I, Gagnon J, St-Pierre A, et al. Moderate alcohol consumption is more cardioprotective in men with the metabolic syndrome. J Nutr. 2006;136(12):3027-3032.

8. Joosten MM, Chiuve SE, Mukamal KJ, Hu FB, Hendriks HF, Rimm EB. Changes in alcohol consumption and subsequent risk of type 2 diabetes in men. Diabetes. 2011;60(1):74-79.

9. Nanri A, Mizoue T, Noda M, et al. Fish intake and type 2 diabetes in Japanese men and women: the Japan Public Health Center-based Prospective Study. Am J Clin Nutr. 2011;94(3):884-891.

10. Choi HK, Willett WC, Stampfer MJ, Rimm E, Hu FB. Dairy consumption and risk of type 2 diabetes mellitus in men: a prospective study. Arch Intern Med. 2005;165(9):997-1003.

11. Fung TT, Hu FB, Pereira MA, et al. Whole-grain intake and the risk of type 2 diabetes: a prospective study in men. Am J Clin Nutr. 2002;76(3):535-540.

12. Lopez-Ridaura R, Willett WC, Rimm EB, et al. Magnesium intake and risk of type 2 diabetes in men and women. Diabetes Care. 2004;27(1):134-140.

13. Nathan DM, Kuenen J, Borg R, et al. Translating the A1C assay into estimated average glucose values. Diabetes Care. 2008;31(8):1473-1478.

14. Levey AS, Coresh J, Balk E, et al. National Kidney Foundation practice guidelines for chronic kidney disease: evaluation, classification, and stratification. Ann Intern Med. 2003;139(2):137-147.

15. Legato MJ, Gelzer A, Goland R, et al. Gender-specific care of the patient with diabetes: review and recommendations. Gend Med. 2006;3(2):131-158.

16. Huxley R, Barzi F, Woodward M. Excess risk of fatal coronary heart disease associated with diabetes in men and women: meta-analysis of 37 prospective cohort studies. BMJ. 2006;332(7533):73-78.

17. Larsson CA, Gullberg B, Merlo J, Rastam L, Lindblad U. Female advantage in AMI mortality is reversed in patients with type 2 diabetes in the Skaraborg Project. Diabetes Care. 2005;28(9):2246-2248.

18. Avogaro A, Giorda C, Maggini M, et al. Incidence of coronary heart disease in type 2 diabetic men and women: impact of microvascular complications, treatment, and geographic location. Diabetes Care. 2007;30(5):1241-1247.

19. Zandbergen AA, Sijbrands EJ, Lamberts SW, Bootsma AH. Normotensive women with type 2 diabetes and microalbuminuria are at high risk for macrovascular disease. Diabetes Care. 2006;29(8):1851-1855.

20. Mulnier HE, Seaman HE, Raleigh VS, et al. Risk of stroke in people with type 2 diabetes in the UK: a study using the General Practice Research Database. Diabetologia. 2006;49(12):2859-2865.

21. Tuomilehto J, Rastenyte D, Jousilahti P, Sarti C, Vartiainen E. Diabetes mellitus as a risk factor for death from stroke. Prospective study of the middle-aged Finnish population. Stroke. 1996;27(2):210-215.

22. Hanai K, Babazono T, Yoshida N, et al. Gender differences in the association between HDL cholesterol and the progression of diabetic kidney disease in type 2 diabetic patients. Nephrol Dial Transplant. 2012;27(3):1070-1075.

23. Tuerk PW, Mueller M, Egede LE. Estimating physician effects on glycemic control in the treatment of diabetes: methods, effects sizes, and implications for treatment policy. Diabetes Care. 2008;31(5):869-873.

24. Rubin RR, Peyrot M, Siminerio LM. Health care and patient-reported outcomes: results of the cross-national Diabetes Attitudes, Wishes and Needs (DAWN) study. Diabetes Care. 2006;29(6):1249-1255.

25. McCollum M, Hansen LB, Ghushchyan V, Sullivan PW. Inconsistent health perceptions for US women and men with diabetes. J Womens Health (Larchmt). 2007;16(10):1421-1428.

26. Gucciardi E, Wang SC, DeMelo M, Amaral L, Stewart DE. Characteristics of men and women with diabetes: observations during patients’ initial visit to a diabetes education centre. Can Fam Physician. 2008;54(2):219-227.

27. Chiu CJ, Wray LA. Physical disability trajectories in older Americans with and without diabetes: the role of age, gender, race or ethnicity, and education. Gerontologist. 2011;51(1):51-63.

28. Nielsen AB, de Fine Olivarius N, Gannik D, Hindsberger C, Hollnagel H. Structured personal diabetes care in primary health care affects only women’s HbA1c. Diabetes Care. 2006;29(5):963-969.

29. Shalev V, Chodick G, Heymann AD, Kokia E. Gender differences in healthcare utilization and medical indicators among patients with diabetes. Public Health. 2005;119(1):45-49.

30. Kacerovsky-Bielesz G, Lienhardt S, Hagenhofer M, et al. Sex-related psychological effects on metabolic control in type 2 diabetes mellitus. Diabetologia. 2009;52(5):781-788.

31. Brown SA, Harrist RB, Villagomez ET, Segura M, Barton SA, Hanis CL. Gender and treatment differences in knowledge, health beliefs, and metabolic control in Mexican Americans with type 2 diabetes. Diabetes Educ. 2000;26(3):425-438.

32. Liburd LC, Namageyo-Funa A, Jack L, Jr. Understanding “masculinity” and the challenges of managing type-2 diabetes among African-American men. J Natl Med Assoc. 2007;99(5):550-552, 554–558.

Author and Disclosure Information

Richard Aguilar, MD
Medical Director, Diabetes Nation, LLC, Sisters, OR, Director, Seville Medical Center, Downey, CA

Issue
The Journal of Family Practice - 61(6)
Publications
Topics
Page Number
S16-S21
Sections
Author and Disclosure Information

Richard Aguilar, MD
Medical Director, Diabetes Nation, LLC, Sisters, OR, Director, Seville Medical Center, Downey, CA

Author and Disclosure Information

Richard Aguilar, MD
Medical Director, Diabetes Nation, LLC, Sisters, OR, Director, Seville Medical Center, Downey, CA

 

DISCLOSURES

Dr. Aguilar has disclosed that he has ongoing relationships with the following companies: Amylin Pharmaceuticals; Eli Lilly; Janssen Pharmaceuticals, Inc; Novo Nordisk, Inc; and Takeda Pharmaceuticals USA, Inc.

SUPPORT

This program is sponsored by the PCEC and is supported by funding from Novo Nordisk, Inc.

 

The prevalence of type 2 diabetes mellitus (T2DM) is similar in men and women (11.8% vs 10.8%, respectively), however there are gender differences that should be considered when developing a treatment plan (eg, cardiovascular risk, psychosocial factors, coping strategies, and the perception of benefit from self-care) when managing those diagnosed with this disease and those at risk for developing it.1 This article describes these differences in the context of two patients—one at risk for T2DM being seen by his health care provider for a routine physical examination, and one who has been treated for several years for T2DM and is being seen for a follow-up office visit. For each patient, the implications for treatment are discussed.

Men at Risk for Type 2 Diabetes Mellitus

 

JW is a 48-year-old white male being seen for a routine physical examination; he last saw a physician 6 years ago, also for a routine physical. He has no complaints and is taking no medications. Having divorced 7 years ago, he lives alone in an apartment and eats many of his meals at fast food restaurants. JW drinks 2 to 3 beers a night several times a week and more when he socializes with his friends 2 to 3 evenings per week. He smokes socially. His father has a 12-year history of T2DM. His mother has a 4-year history of essential hypertension and a 9-year history of chronic obstructive pulmonary disease.

Physical examination shows that JW is 5’11” tall, weighs 207 pounds (body mass index (BMI), 29 kg/m2), and has a 41” waist circumference; his blood pressure (BP) is 138/86 mm Hg and respiratory rate is 17 breaths/min. The remainder of his physical examination, including eye and neurologic exams, is normal. Laboratory results, including a screening glycated homoglobin (A1C), are pending.

Key Risk Factors for Type 2 Diabetes Mellitus in Men

This case is not an uncommon presentation of a middle-aged male who has several risk factors for diabetes (see Case Study 1 continued ). JW also has key risk factors for T2DM in men. The Monitoring of Trends and Determinants in Cardiovascular Disease (MONICA) Augsburg surveys identified 128 men and 85 women with T2DM.2 Increasing age and BMI, positive parental history of T2DM, and a low high-density lipoprotein cholesterol (HDL-C) level were independent risk factors predicting the development of T2DM in both men and women. However, several other factors posed a higher risk in men relative to women, including systolic BP (hazard ratio [HR], 1.16 per 10-mm Hg increase), regular smoking (HR, 1.75), and alcohol intake ≥ 40 g/d (HR, 1.95). (Note: 1 fluid ounce 80 proof alcohol ≈ 11 g ethanol; 12 fluid ounces beer [~5% alcohol] ≈ 14 g ethanol). After adjusting for these factors, a separate analysis (4424 men, 4380 women) showed that men who lived alone were more likely to develop T2DM than either men or women who did not live alone (HR, 1.69 in men vs 0.85 in women; P = .006).3 While the number of people with T2DM in MONICA was small, the results suggest that measuring BP, particularly systolic BP, and taking a smoking and alcohol history may be especially important in men.

With respect to alcohol intake, epidemiologic and randomized clinical trials have generally demonstrated an inverse relationship between moderate alcohol consumption (20 to 30 g/d) and the long-term risk of T2DM.2,4-7 Differences among studies in how patients were grouped preclude determination of the daily alcohol consumption that confers the greatest risk benefit, although one recent study conducted over 4 years indicates that the greatest benefit in diabetes risk reduction may occur when men who previously consumed 8

Other nutrition and lifestyle patterns also seem to be particularly beneficial in reducing the risk of T2DM in men. Survey data involving 22,921 Japanese men and 29,759 Japanese women followed over 5 years showed that fish and seafood intake was significantly associated with a decreased risk of T2DM in men but not in women.9 The odds ratio of developing T2DM for the highest quartile versus the lowest quartile of fish and seafood intake was 0.73 (P = .04 for trend). Additional analysis did not identify any significant association with the fat content of fish.

 

 

Results of the Health Professionals Follow-up Study provide evidence of benefit in lowering the risk of T2DM in men who consume high amounts of low-fat dairy products, whole grains, and magnesium ( TABLE 1 ). With respect to dairy food consumption, after 12 years of follow-up involving 1243 incident cases of T2DM, the relative risk (RR) of developing T2DM in men in the top quintile of dairy intake was 0.77 compared with those in the lowest quintile (P = .003 for trend).10 Men in the highest quintile consumed 4.1 servings of dairy food per day compared with 0.5 servings per day in the lowest quintile. Each serving-per-day increase in total dairy intake was associated with a 9% lower risk for T2DM, with a lower risk seen with consumption of low-fat vs high-fat dairy food. With respect to whole-grain intake, the RR of developing T2DM was 0.58 in men in the upper vs lower quintiles (3.2 vs 0.4 servings/d), although the effect was attenuated with BMI (P = .0006 for trend).11 Similar observations were made with respect to magnesium consumption; a RR of 0.76 for T2DM was observed in men with a median magnesium consumption of 457 mg/d compared with those who consumed 270 mg/d.12

TABLE 1

Suggestions for Men Who Are at Risk of or Have Been Diagnosed with Type 2 Diabetes Mellitus (T2DM)*

 

For men who are at risk:
  • Key targets
    • -Systolic BP
    • -Smoking cessation
    • -Alcohol consumption (moderate)
  • Promote healthy diet
    • -Fish/seafood
    • -Low-fat dairy products
    • -Whole grains
    • -Magnesium
For men who have been diagnosed:
  • Key targets
    • -BP
    • -Blood glucose
    • -HDL-C
  • Emphasize the importance of self-management
  • Provide ongoing education/information regarding the progressive nature of T2DM and the need to adjust treatment over time, potentially adding both oral and injectable therapies
  • Recommend a diabetes support group
BP, blood pressure; HDL-C, high-density lipoprotein cholesterol.
*These suggestions are in addition to developing and fostering a collaborative, patient-centered approach.

 

JW has the following risk factors for T2DM:

  • Overweight with central adiposity
  • Physical inactivity
  • First-degree relative with T2DM
  • Possible cardiovascular disease (CVD; hypertension, smoking)
  • High daily alcohol intake (10 to 20 g alcohol/beer x 2-3 beers/d = 20 to 60 g alcohol/d)
  • Poor nutrition
  • Lives alone

Plan:

  • Discuss above risk factors with JW
  • Repeat BP measurement at next visit; implement treatment if BP >140/90 mm Hg (130/80 mm Hg if T2DM is diagnosed)
  • Consider evaluation for alcohol/substance abuse
  • Evaluate for smoking cessation program
  • Nutrition referral for lifestyle and dietary management intervention

Working with men to avoid the development of T2DM is an important objective for family physicians. It is essential to identify men who are at increased risk, including those with prediabetes, provide education about the disease and its risk factors, and implement appropriate risk reduction strategies. Risk reduction strategies should focus on modifiable factors, such as body weight, physical activity, BP, blood lipids, blood glucose, and smoking. With JW, his motivation to “get back into shape” will help move the conversation toward achievable goals that can be set and modified over time. Other strategies that may be helpful in reducing the risk of developing T2DM in men include a moderate daily alcohol intake and a diet high in fish and seafood, low-fat dairy products, whole grains, and magnesium ( TABLE 1 ).

Once diagnosed with T2DM, there are risk management strategies that can be particularly helpful in men. These include strategies that target cardiovascular health, as well as those that consider the psychosocial and coping behaviors of men.

Risk of Complications in Men With Type 2 Diabetes Mellitus

 

MR is a 57-year-old African American male diagnosed with hypertension 5 years ago and T2DM 3 years ago (A1C, 8.2%). Treatment with lifestyle modification and metformin 1000 mg twice daily had lowered his A1C to between 6.8% and 7.1%. However, 9 months ago, MR hurt his knee, which prevents him from walking his usual 1 to 1.5 miles several days a week and doing yard work on the weekends.

Physical examination: BP, 126/78 mm Hg; body weight, 183 pounds (a 13 to 17 pound increase since the knee injury); waist circumference, 38” (BMI, 28 kg/m2); grade 1 retinopathy bilaterally; neurologic exam normal.

Laboratory: A1C, 7.8%; lipids normal except triglyceride level, 219 mg/dL; creatinine clearance (calculated), 69 mL/min; urine, 45 mg albumin/g creatinine.

MR’s self-measured fasting plasma glucose (FPG) has ranged from 121 to 143 mg/dL over the past month; isolated postprandial glucose (PPG) measurements show 194 to 258 mg/dL.

MR works as a vocational teacher at the local high school, and he teaches driver education after school. Review of his pharmacy records suggests his adherence over the past year has been: metformin (88%), hydrochlorothiazide (72%), and lisinopril (72%).

Assessment:

  • A1C level of 7.8% indicates an estimated average glucose (eAG) of 177 mg/dL13
    • –Mildly elevated FPG and PPG
    • –Evidence of microvascular disease (retinopathy, nephropathy)
    • –Creatinine clearance 69 mL/min and microalbuminuria indicate stage 2 chronic kidney disease14

In addition to referring MR for physical rehabilitation of his knee, you discuss with MR the need and options for intensifying his diabetes therapy.

Does the fact that MR is male affect your management plan?

 

 

In people diagnosed with T2DM, there are differences between men and women with respect to risk for cardiovascular and other comorbid diseases, as well as in their psychosocial well-being and coping strategies.

Risk for Cardiovascular Disease in Type 2 Diabetes Mellitus

 

A systematic literature review shows that men with T2DM generally fare better than women with T2DM regarding their risk for CVD. Men with T2DM have a 2- to 3-fold increase in the risk of developing coronary heart disease (CHD) compared with men without T2DM, whereas women with T2DM have a 4- to 6-fold increase in risk compared with women without T2DM.15 Compared with women with T2DM, men with T2DM also have a better prognosis after myocardial infarction (MI) and a lower risk of death overall from CVD. Possible reasons for these differences include a lower risk of hypertension, a less severe form of dyslipidemia, and a lower prevalence of obesity in men with T2DM compared with women with T2DM.15 These same reasons for observed differences between men and women were seen in a meta-analysis of 29 studies, where the RR of fatal MI in men with T2DM compared with women with T2DM was 0.68.16 Similar findings were seen in the Skaraborg Project, which involved 1116 Swedish patients with hypertension and/or T2DM.17 Compared with a healthy population, the age-adjusted HR for fatal MI was 1.9 for men with T2DM and 5.0 for women with T2DM over 8.1 years of follow-up (RR, 0.38 for men vs women). Analysis of the data indicated that these results were not explained by the more favorable survival rate in women without T2DM than in men without T2DM.17

Somewhat different results have been reported by the Italian Diabetes and Informatics Study Group in a slightly different T2DM population. This investigation involved men and women with T2DM (N = 11,644) who could have microvascular but not macrovascular disease.18 After 4 years of follow-up, the age-adjusted incident rates for first CHD event (composite of acute MI, coronary artery bypass grafting, percutaneous transluminal coronary angioplasty) were 28.8 per 1000 person-years in men and 23.3 per 1000 person-years in women. Incident rates (per 1000 person-years) of acute MI (10.3 vs 4.7), major CHD events (13.1 vs 5.8), and fatal CHD (2.6 vs 0.6) were all significantly more frequent in men than in women, respectively. Multivariate analysis showed that hypertension and A1C were additional risk factors for CHD in men; for each 20% increment above the A1C upper limit of normal, there was a 14% risk increase for CHD. The presence of microvascular complications increased risk by 20% in men and 35% in women. In this analysis, glycemic control and hypertension were found to be the predominant risk factors in men, while high triglyceride levels, low HDL-C levels, and microangiopathy were predominant in women.

Additional multivariate analyses provide greater insight into specific factors that affect the risk of CVD and outcomes in men with T2DM. One investigation compared men and women with T2DM who were normotensive without evidence of CVD but with microalbuminuria. After 4.7 years of follow-up, men were found to be at lower risk (RR, 0.12) for a composite of death, acute MI, unstable angina, coronary interventions, heart failure, cerebral ischemic stroke or transient ischemic attack, and peripheral artery disease.19 Other investigators have reported a lower risk of stroke, including fatal stroke, in men with T2DM compared with women with T2DM.20,21 For example, analysis of the General Practice Research Database identified 22,178 men and 19,621 women with T2DM between the ages of 35 and 89 years.20 The stroke rate per 1000 person-years across all ages was 10.82 (95% confidence interval (CI), 10.17-11.51) in men and 13.16 (95% CI, 12.40-13.97) in women. In men, the rate per 1000 person-years rose from 1.81 in the 35 to 44 year age group to 28.35 in men 85 years of age or older. Although the rate of stroke per 1000 person-years was lower in women than men in the 35 to 44 year age group (1.53 vs 1.81), the rate in women exceeded that of men in the 85 years of age or older group (32.20 vs 28.35).

Other Chronic Complications

Kidney disease is affected by blood lipids, specifically HDL-C, in men with T2DM. An investigation in men and women with T2DM with normoalbuminuria or microalbuminuria at baseline showed that a low HDL-C level was an independent predictor of progression to a more advanced stage of albuminuria over 4.3 years of follow-up (HR, 0.391 for men with normal HDL-C compared with men with low HDL-C). In women, no lipid parameters were associated with progression of albuminuria.22

 

 

While these investigations do not provide a clear picture of the differences regarding cardiovascular risk between men and women with T2DM, they suggest that men with T2DM have a lower risk of nonfatal and fatal CVD and stroke than do women with T2DM. However, the lower risk seen in men may be affected by the cardiovascular endpoints measured and the presence of microvascular disease. Possible independent risk factors for CVD in men with T2DM include hypertension, poor glycemic control, and low HDL-C.

 

Risk factors that place MR at greater risk for CVD compared with a woman with T2DM and therefore serve as key treatment targets include:

  • Hypertension—although controlled (126/78 mm Hg) with hydrochlorothiazide and lisinopril
  • Poor glycemic control—A1C, 7.8% (eAG, 177mg/dL)
    • –Increase physical activity—refer for knee rehabilitation
    • –Intensify glucose-lowering therapy by adding an additional glucose-lowering agent (eg, dipeptidyl peptidase-4 inhibitor, glucagon-like peptide-1 receptor agonist, thiazolidinedione, α-glucosidase inhibitor, sulfonylurea, glinide, or basal insulin)
  • Microalbuminuria (45 mg urinary albumin/g creatinine)—encourage better adherence to lisinopril; monitor renal function
  • Hypertriglyceridemia—initiate omega-3 fatty acid or extended-release niacin

Psychosocial Well-Being, Benefit of Self-Care, and Coping Strategies

Type 2 diabetes mellitus is a chronic disease with glycemic control largely determined by patient self-management, and the attitudes and beliefs of patients with T2DM are important factors to consider from diagnosis onward.23 There are important differences between men and women with T2DM regarding attitudes and beliefs. Published investigations provide some, although not entirely consistent, insight into these psychosocial differences between men and women with T2DM. These differences are summarized in TABLE 2 .24-32 Taking these differences into account when planning treatment and when communicating with and educating the patient is essential for improved patient self-management.

TABLE 2

Psychosocial and Coping Characteristics of Men with Type 2 Diabetes Mellitus (T2DM)24-32

 

Compared with women with T2DM, generally, men with T2DM:
  • Experience less diabetes-related distress and greater well-being
  • Are less likely to experience symptoms of depression
  • Experience a slower deterioration in physical function
  • Exercise more
  • Perceive less support from their healthcare team
  • Have lower expectations regarding the benefits of self-management
  • Are less informed about T2DM, particularly pharmacologic and nonpharmacologic treatment options
  • Exert less effort and employ fewer strategies to cope with T2DM
  • Have less adaptive attitudes toward T2DM
  • Are influenced more by symptoms of hypoglycemia and hyperglycemia
  • Believe they have more family and social support and are more influenced by such support
  • Fear losing control of their disease and resist being “policed” by their social support system

 

 

Key interventions for MR:

  • Maintain a dialogue and enhance collaboration with MR
  • Establish shared goals that are customized to incorporate MR’s personal goals
  • Problem solve with MR to identify ways he can better integrate the diabetes self-care objectives of dietary changes and blood glucose self-monitoring into his daily life
  • Emphasize that enhanced or greater disease control can be achieved by good self-management, including better adherence to the management plan
  • Remind MR that T2DM is a progressive disease that requires intermittent medication adjustments to keep pace with its progression
  • Build upon the belief that T2DM can be controlled by reminding MR that the disease was well controlled before his knee injury
    • –Focus on the importance of rehabilitating his knee
    • –Develop a rehabilitation plan
  • Provide informational support regarding options for intensifying diabetes therapy (eg, dipeptidyl peptidase-4 inhibitor, thiazolidinedione, glucagon-like peptide-1 receptor agonist, sulfonylurea, or insulin)
    • –Discuss MR’s needs and concerns, as well as barriers for each treatment option, particularly hypoglycemia and weight gain
    • –Provide instruction or educational materials regarding injection devices
    • –Involve the healthcare team, as appropriate
  • Keep the treatment regimen as simple as possible; consider pill combinations where appropriate

Summary

The growing epidemic of T2DM requires intervention to assist patients who have been diagnosed to better manage the disease, to reduce the risk of developing the disease in those who have not yet been diagnosed, and to manage the associated complications. In addition to individualizing interventions based on a patient’s needs, concerns, and capabilities, taking gender into account is necessary. In otherwise healthy people, several independent factors appear to pose a higher risk of T2DM in men relative to women, including systolic hypertension, regular smoking, and alcohol intake ≥ 40 g/d. At the same time, men achieve greater risk reduction from moderate daily alcohol intake and a diet high in fish and seafood, low-fat dairy products, whole grains, and magnesium.

Once diagnosed with T2DM, men generally fare better than women regarding the risk for CVD; they also have a better prognosis after MI and a lower risk of death overall from CVD. Possible independent risk factors for CVD in men with T2DM that are especially important may include hypertension, poor glycemic control, and low HDL-C levels. Psychosocial complications, such as depression, are less likely in men with T2DM. However, men expend less effort coping, are less likely to utilize healthcare services, and are less informed about treatment options. Although men have a lower expectation of the benefit of self-management, they find support from family and friends more helpful than do women, but they are fearful of losing control of their disease.

Taking these gender differences into account should prove helpful as family care physicians work with men to reduce their risk of developing T2DM and in helping men diagnosed with T2DM to better self-manage their disease.

 

DISCLOSURES

Dr. Aguilar has disclosed that he has ongoing relationships with the following companies: Amylin Pharmaceuticals; Eli Lilly; Janssen Pharmaceuticals, Inc; Novo Nordisk, Inc; and Takeda Pharmaceuticals USA, Inc.

SUPPORT

This program is sponsored by the PCEC and is supported by funding from Novo Nordisk, Inc.

 

The prevalence of type 2 diabetes mellitus (T2DM) is similar in men and women (11.8% vs 10.8%, respectively), however there are gender differences that should be considered when developing a treatment plan (eg, cardiovascular risk, psychosocial factors, coping strategies, and the perception of benefit from self-care) when managing those diagnosed with this disease and those at risk for developing it.1 This article describes these differences in the context of two patients—one at risk for T2DM being seen by his health care provider for a routine physical examination, and one who has been treated for several years for T2DM and is being seen for a follow-up office visit. For each patient, the implications for treatment are discussed.

Men at Risk for Type 2 Diabetes Mellitus

 

JW is a 48-year-old white male being seen for a routine physical examination; he last saw a physician 6 years ago, also for a routine physical. He has no complaints and is taking no medications. Having divorced 7 years ago, he lives alone in an apartment and eats many of his meals at fast food restaurants. JW drinks 2 to 3 beers a night several times a week and more when he socializes with his friends 2 to 3 evenings per week. He smokes socially. His father has a 12-year history of T2DM. His mother has a 4-year history of essential hypertension and a 9-year history of chronic obstructive pulmonary disease.

Physical examination shows that JW is 5’11” tall, weighs 207 pounds (body mass index (BMI), 29 kg/m2), and has a 41” waist circumference; his blood pressure (BP) is 138/86 mm Hg and respiratory rate is 17 breaths/min. The remainder of his physical examination, including eye and neurologic exams, is normal. Laboratory results, including a screening glycated homoglobin (A1C), are pending.

Key Risk Factors for Type 2 Diabetes Mellitus in Men

This case is not an uncommon presentation of a middle-aged male who has several risk factors for diabetes (see Case Study 1 continued ). JW also has key risk factors for T2DM in men. The Monitoring of Trends and Determinants in Cardiovascular Disease (MONICA) Augsburg surveys identified 128 men and 85 women with T2DM.2 Increasing age and BMI, positive parental history of T2DM, and a low high-density lipoprotein cholesterol (HDL-C) level were independent risk factors predicting the development of T2DM in both men and women. However, several other factors posed a higher risk in men relative to women, including systolic BP (hazard ratio [HR], 1.16 per 10-mm Hg increase), regular smoking (HR, 1.75), and alcohol intake ≥ 40 g/d (HR, 1.95). (Note: 1 fluid ounce 80 proof alcohol ≈ 11 g ethanol; 12 fluid ounces beer [~5% alcohol] ≈ 14 g ethanol). After adjusting for these factors, a separate analysis (4424 men, 4380 women) showed that men who lived alone were more likely to develop T2DM than either men or women who did not live alone (HR, 1.69 in men vs 0.85 in women; P = .006).3 While the number of people with T2DM in MONICA was small, the results suggest that measuring BP, particularly systolic BP, and taking a smoking and alcohol history may be especially important in men.

With respect to alcohol intake, epidemiologic and randomized clinical trials have generally demonstrated an inverse relationship between moderate alcohol consumption (20 to 30 g/d) and the long-term risk of T2DM.2,4-7 Differences among studies in how patients were grouped preclude determination of the daily alcohol consumption that confers the greatest risk benefit, although one recent study conducted over 4 years indicates that the greatest benefit in diabetes risk reduction may occur when men who previously consumed 8

Other nutrition and lifestyle patterns also seem to be particularly beneficial in reducing the risk of T2DM in men. Survey data involving 22,921 Japanese men and 29,759 Japanese women followed over 5 years showed that fish and seafood intake was significantly associated with a decreased risk of T2DM in men but not in women.9 The odds ratio of developing T2DM for the highest quartile versus the lowest quartile of fish and seafood intake was 0.73 (P = .04 for trend). Additional analysis did not identify any significant association with the fat content of fish.

 

 

Results of the Health Professionals Follow-up Study provide evidence of benefit in lowering the risk of T2DM in men who consume high amounts of low-fat dairy products, whole grains, and magnesium ( TABLE 1 ). With respect to dairy food consumption, after 12 years of follow-up involving 1243 incident cases of T2DM, the relative risk (RR) of developing T2DM in men in the top quintile of dairy intake was 0.77 compared with those in the lowest quintile (P = .003 for trend).10 Men in the highest quintile consumed 4.1 servings of dairy food per day compared with 0.5 servings per day in the lowest quintile. Each serving-per-day increase in total dairy intake was associated with a 9% lower risk for T2DM, with a lower risk seen with consumption of low-fat vs high-fat dairy food. With respect to whole-grain intake, the RR of developing T2DM was 0.58 in men in the upper vs lower quintiles (3.2 vs 0.4 servings/d), although the effect was attenuated with BMI (P = .0006 for trend).11 Similar observations were made with respect to magnesium consumption; a RR of 0.76 for T2DM was observed in men with a median magnesium consumption of 457 mg/d compared with those who consumed 270 mg/d.12

TABLE 1

Suggestions for Men Who Are at Risk of or Have Been Diagnosed with Type 2 Diabetes Mellitus (T2DM)*

 

For men who are at risk:
  • Key targets
    • -Systolic BP
    • -Smoking cessation
    • -Alcohol consumption (moderate)
  • Promote healthy diet
    • -Fish/seafood
    • -Low-fat dairy products
    • -Whole grains
    • -Magnesium
For men who have been diagnosed:
  • Key targets
    • -BP
    • -Blood glucose
    • -HDL-C
  • Emphasize the importance of self-management
  • Provide ongoing education/information regarding the progressive nature of T2DM and the need to adjust treatment over time, potentially adding both oral and injectable therapies
  • Recommend a diabetes support group
BP, blood pressure; HDL-C, high-density lipoprotein cholesterol.
*These suggestions are in addition to developing and fostering a collaborative, patient-centered approach.

 

JW has the following risk factors for T2DM:

  • Overweight with central adiposity
  • Physical inactivity
  • First-degree relative with T2DM
  • Possible cardiovascular disease (CVD; hypertension, smoking)
  • High daily alcohol intake (10 to 20 g alcohol/beer x 2-3 beers/d = 20 to 60 g alcohol/d)
  • Poor nutrition
  • Lives alone

Plan:

  • Discuss above risk factors with JW
  • Repeat BP measurement at next visit; implement treatment if BP >140/90 mm Hg (130/80 mm Hg if T2DM is diagnosed)
  • Consider evaluation for alcohol/substance abuse
  • Evaluate for smoking cessation program
  • Nutrition referral for lifestyle and dietary management intervention

Working with men to avoid the development of T2DM is an important objective for family physicians. It is essential to identify men who are at increased risk, including those with prediabetes, provide education about the disease and its risk factors, and implement appropriate risk reduction strategies. Risk reduction strategies should focus on modifiable factors, such as body weight, physical activity, BP, blood lipids, blood glucose, and smoking. With JW, his motivation to “get back into shape” will help move the conversation toward achievable goals that can be set and modified over time. Other strategies that may be helpful in reducing the risk of developing T2DM in men include a moderate daily alcohol intake and a diet high in fish and seafood, low-fat dairy products, whole grains, and magnesium ( TABLE 1 ).

Once diagnosed with T2DM, there are risk management strategies that can be particularly helpful in men. These include strategies that target cardiovascular health, as well as those that consider the psychosocial and coping behaviors of men.

Risk of Complications in Men With Type 2 Diabetes Mellitus

 

MR is a 57-year-old African American male diagnosed with hypertension 5 years ago and T2DM 3 years ago (A1C, 8.2%). Treatment with lifestyle modification and metformin 1000 mg twice daily had lowered his A1C to between 6.8% and 7.1%. However, 9 months ago, MR hurt his knee, which prevents him from walking his usual 1 to 1.5 miles several days a week and doing yard work on the weekends.

Physical examination: BP, 126/78 mm Hg; body weight, 183 pounds (a 13 to 17 pound increase since the knee injury); waist circumference, 38” (BMI, 28 kg/m2); grade 1 retinopathy bilaterally; neurologic exam normal.

Laboratory: A1C, 7.8%; lipids normal except triglyceride level, 219 mg/dL; creatinine clearance (calculated), 69 mL/min; urine, 45 mg albumin/g creatinine.

MR’s self-measured fasting plasma glucose (FPG) has ranged from 121 to 143 mg/dL over the past month; isolated postprandial glucose (PPG) measurements show 194 to 258 mg/dL.

MR works as a vocational teacher at the local high school, and he teaches driver education after school. Review of his pharmacy records suggests his adherence over the past year has been: metformin (88%), hydrochlorothiazide (72%), and lisinopril (72%).

Assessment:

  • A1C level of 7.8% indicates an estimated average glucose (eAG) of 177 mg/dL13
    • –Mildly elevated FPG and PPG
    • –Evidence of microvascular disease (retinopathy, nephropathy)
    • –Creatinine clearance 69 mL/min and microalbuminuria indicate stage 2 chronic kidney disease14

In addition to referring MR for physical rehabilitation of his knee, you discuss with MR the need and options for intensifying his diabetes therapy.

Does the fact that MR is male affect your management plan?

 

 

In people diagnosed with T2DM, there are differences between men and women with respect to risk for cardiovascular and other comorbid diseases, as well as in their psychosocial well-being and coping strategies.

Risk for Cardiovascular Disease in Type 2 Diabetes Mellitus

 

A systematic literature review shows that men with T2DM generally fare better than women with T2DM regarding their risk for CVD. Men with T2DM have a 2- to 3-fold increase in the risk of developing coronary heart disease (CHD) compared with men without T2DM, whereas women with T2DM have a 4- to 6-fold increase in risk compared with women without T2DM.15 Compared with women with T2DM, men with T2DM also have a better prognosis after myocardial infarction (MI) and a lower risk of death overall from CVD. Possible reasons for these differences include a lower risk of hypertension, a less severe form of dyslipidemia, and a lower prevalence of obesity in men with T2DM compared with women with T2DM.15 These same reasons for observed differences between men and women were seen in a meta-analysis of 29 studies, where the RR of fatal MI in men with T2DM compared with women with T2DM was 0.68.16 Similar findings were seen in the Skaraborg Project, which involved 1116 Swedish patients with hypertension and/or T2DM.17 Compared with a healthy population, the age-adjusted HR for fatal MI was 1.9 for men with T2DM and 5.0 for women with T2DM over 8.1 years of follow-up (RR, 0.38 for men vs women). Analysis of the data indicated that these results were not explained by the more favorable survival rate in women without T2DM than in men without T2DM.17

Somewhat different results have been reported by the Italian Diabetes and Informatics Study Group in a slightly different T2DM population. This investigation involved men and women with T2DM (N = 11,644) who could have microvascular but not macrovascular disease.18 After 4 years of follow-up, the age-adjusted incident rates for first CHD event (composite of acute MI, coronary artery bypass grafting, percutaneous transluminal coronary angioplasty) were 28.8 per 1000 person-years in men and 23.3 per 1000 person-years in women. Incident rates (per 1000 person-years) of acute MI (10.3 vs 4.7), major CHD events (13.1 vs 5.8), and fatal CHD (2.6 vs 0.6) were all significantly more frequent in men than in women, respectively. Multivariate analysis showed that hypertension and A1C were additional risk factors for CHD in men; for each 20% increment above the A1C upper limit of normal, there was a 14% risk increase for CHD. The presence of microvascular complications increased risk by 20% in men and 35% in women. In this analysis, glycemic control and hypertension were found to be the predominant risk factors in men, while high triglyceride levels, low HDL-C levels, and microangiopathy were predominant in women.

Additional multivariate analyses provide greater insight into specific factors that affect the risk of CVD and outcomes in men with T2DM. One investigation compared men and women with T2DM who were normotensive without evidence of CVD but with microalbuminuria. After 4.7 years of follow-up, men were found to be at lower risk (RR, 0.12) for a composite of death, acute MI, unstable angina, coronary interventions, heart failure, cerebral ischemic stroke or transient ischemic attack, and peripheral artery disease.19 Other investigators have reported a lower risk of stroke, including fatal stroke, in men with T2DM compared with women with T2DM.20,21 For example, analysis of the General Practice Research Database identified 22,178 men and 19,621 women with T2DM between the ages of 35 and 89 years.20 The stroke rate per 1000 person-years across all ages was 10.82 (95% confidence interval (CI), 10.17-11.51) in men and 13.16 (95% CI, 12.40-13.97) in women. In men, the rate per 1000 person-years rose from 1.81 in the 35 to 44 year age group to 28.35 in men 85 years of age or older. Although the rate of stroke per 1000 person-years was lower in women than men in the 35 to 44 year age group (1.53 vs 1.81), the rate in women exceeded that of men in the 85 years of age or older group (32.20 vs 28.35).

Other Chronic Complications

Kidney disease is affected by blood lipids, specifically HDL-C, in men with T2DM. An investigation in men and women with T2DM with normoalbuminuria or microalbuminuria at baseline showed that a low HDL-C level was an independent predictor of progression to a more advanced stage of albuminuria over 4.3 years of follow-up (HR, 0.391 for men with normal HDL-C compared with men with low HDL-C). In women, no lipid parameters were associated with progression of albuminuria.22

 

 

While these investigations do not provide a clear picture of the differences regarding cardiovascular risk between men and women with T2DM, they suggest that men with T2DM have a lower risk of nonfatal and fatal CVD and stroke than do women with T2DM. However, the lower risk seen in men may be affected by the cardiovascular endpoints measured and the presence of microvascular disease. Possible independent risk factors for CVD in men with T2DM include hypertension, poor glycemic control, and low HDL-C.

 

Risk factors that place MR at greater risk for CVD compared with a woman with T2DM and therefore serve as key treatment targets include:

  • Hypertension—although controlled (126/78 mm Hg) with hydrochlorothiazide and lisinopril
  • Poor glycemic control—A1C, 7.8% (eAG, 177mg/dL)
    • –Increase physical activity—refer for knee rehabilitation
    • –Intensify glucose-lowering therapy by adding an additional glucose-lowering agent (eg, dipeptidyl peptidase-4 inhibitor, glucagon-like peptide-1 receptor agonist, thiazolidinedione, α-glucosidase inhibitor, sulfonylurea, glinide, or basal insulin)
  • Microalbuminuria (45 mg urinary albumin/g creatinine)—encourage better adherence to lisinopril; monitor renal function
  • Hypertriglyceridemia—initiate omega-3 fatty acid or extended-release niacin

Psychosocial Well-Being, Benefit of Self-Care, and Coping Strategies

Type 2 diabetes mellitus is a chronic disease with glycemic control largely determined by patient self-management, and the attitudes and beliefs of patients with T2DM are important factors to consider from diagnosis onward.23 There are important differences between men and women with T2DM regarding attitudes and beliefs. Published investigations provide some, although not entirely consistent, insight into these psychosocial differences between men and women with T2DM. These differences are summarized in TABLE 2 .24-32 Taking these differences into account when planning treatment and when communicating with and educating the patient is essential for improved patient self-management.

TABLE 2

Psychosocial and Coping Characteristics of Men with Type 2 Diabetes Mellitus (T2DM)24-32

 

Compared with women with T2DM, generally, men with T2DM:
  • Experience less diabetes-related distress and greater well-being
  • Are less likely to experience symptoms of depression
  • Experience a slower deterioration in physical function
  • Exercise more
  • Perceive less support from their healthcare team
  • Have lower expectations regarding the benefits of self-management
  • Are less informed about T2DM, particularly pharmacologic and nonpharmacologic treatment options
  • Exert less effort and employ fewer strategies to cope with T2DM
  • Have less adaptive attitudes toward T2DM
  • Are influenced more by symptoms of hypoglycemia and hyperglycemia
  • Believe they have more family and social support and are more influenced by such support
  • Fear losing control of their disease and resist being “policed” by their social support system

 

 

Key interventions for MR:

  • Maintain a dialogue and enhance collaboration with MR
  • Establish shared goals that are customized to incorporate MR’s personal goals
  • Problem solve with MR to identify ways he can better integrate the diabetes self-care objectives of dietary changes and blood glucose self-monitoring into his daily life
  • Emphasize that enhanced or greater disease control can be achieved by good self-management, including better adherence to the management plan
  • Remind MR that T2DM is a progressive disease that requires intermittent medication adjustments to keep pace with its progression
  • Build upon the belief that T2DM can be controlled by reminding MR that the disease was well controlled before his knee injury
    • –Focus on the importance of rehabilitating his knee
    • –Develop a rehabilitation plan
  • Provide informational support regarding options for intensifying diabetes therapy (eg, dipeptidyl peptidase-4 inhibitor, thiazolidinedione, glucagon-like peptide-1 receptor agonist, sulfonylurea, or insulin)
    • –Discuss MR’s needs and concerns, as well as barriers for each treatment option, particularly hypoglycemia and weight gain
    • –Provide instruction or educational materials regarding injection devices
    • –Involve the healthcare team, as appropriate
  • Keep the treatment regimen as simple as possible; consider pill combinations where appropriate

Summary

The growing epidemic of T2DM requires intervention to assist patients who have been diagnosed to better manage the disease, to reduce the risk of developing the disease in those who have not yet been diagnosed, and to manage the associated complications. In addition to individualizing interventions based on a patient’s needs, concerns, and capabilities, taking gender into account is necessary. In otherwise healthy people, several independent factors appear to pose a higher risk of T2DM in men relative to women, including systolic hypertension, regular smoking, and alcohol intake ≥ 40 g/d. At the same time, men achieve greater risk reduction from moderate daily alcohol intake and a diet high in fish and seafood, low-fat dairy products, whole grains, and magnesium.

Once diagnosed with T2DM, men generally fare better than women regarding the risk for CVD; they also have a better prognosis after MI and a lower risk of death overall from CVD. Possible independent risk factors for CVD in men with T2DM that are especially important may include hypertension, poor glycemic control, and low HDL-C levels. Psychosocial complications, such as depression, are less likely in men with T2DM. However, men expend less effort coping, are less likely to utilize healthcare services, and are less informed about treatment options. Although men have a lower expectation of the benefit of self-management, they find support from family and friends more helpful than do women, but they are fearful of losing control of their disease.

Taking these gender differences into account should prove helpful as family care physicians work with men to reduce their risk of developing T2DM and in helping men diagnosed with T2DM to better self-manage their disease.

References

1. Centers for Disease Control and Prevention. National diabetes fact sheet: national estimates and general information on diabetes and prediabetes in the United States, 2011. http://www.cdc.gov/diabetes/pubs/pdf/ndfs_2011.pdf. Published 2011. Accessed May 2, 2011.

2. Meisinger C, Thorand B, Schneider A, Stieber J, Doring A, Lowel H. Sex differences in risk factors for incident type 2 diabetes mellitus: the MONICA Augsburg cohort study. Arch Intern Med. 2002;162(1):82-89.

3. Meisinger C, Kandler U, Ladwig KH. Living alone is associated with an increased risk of type 2 diabetes mellitus in men but not women from the general population: the MONICA/KORA Augsburg Cohort Study. Psychosom Med. 2009;71(7):784-788.

4. Baliunas DO, Taylor BJ, Irving H, et al. Alcohol as a risk factor for type 2 diabetes: a systematic review and meta-analysis. Diabetes Care. 2009;32(11):2123-2132.

5. Mozaffarian D, Kamineni A, Carnethon M, Djoussé L, Mukamal KJ, Siscovick D. Lifestyle risk factors and new-onset diabetes mellitus in older adults: the cardiovascular health study. Arch Intern Med. 2009;169(8):798-807.

6. Joosten MM, Grobbee DE, van der A DL, Verschuren WM, Hendriks HF, Beulens JW. Combined effect of alcohol consumption and lifestyle behaviors on risk of type 2 diabetes. Am J Clin Nutr. 2010;91(6):1777-1783.

7. Gigleux I, Gagnon J, St-Pierre A, et al. Moderate alcohol consumption is more cardioprotective in men with the metabolic syndrome. J Nutr. 2006;136(12):3027-3032.

8. Joosten MM, Chiuve SE, Mukamal KJ, Hu FB, Hendriks HF, Rimm EB. Changes in alcohol consumption and subsequent risk of type 2 diabetes in men. Diabetes. 2011;60(1):74-79.

9. Nanri A, Mizoue T, Noda M, et al. Fish intake and type 2 diabetes in Japanese men and women: the Japan Public Health Center-based Prospective Study. Am J Clin Nutr. 2011;94(3):884-891.

10. Choi HK, Willett WC, Stampfer MJ, Rimm E, Hu FB. Dairy consumption and risk of type 2 diabetes mellitus in men: a prospective study. Arch Intern Med. 2005;165(9):997-1003.

11. Fung TT, Hu FB, Pereira MA, et al. Whole-grain intake and the risk of type 2 diabetes: a prospective study in men. Am J Clin Nutr. 2002;76(3):535-540.

12. Lopez-Ridaura R, Willett WC, Rimm EB, et al. Magnesium intake and risk of type 2 diabetes in men and women. Diabetes Care. 2004;27(1):134-140.

13. Nathan DM, Kuenen J, Borg R, et al. Translating the A1C assay into estimated average glucose values. Diabetes Care. 2008;31(8):1473-1478.

14. Levey AS, Coresh J, Balk E, et al. National Kidney Foundation practice guidelines for chronic kidney disease: evaluation, classification, and stratification. Ann Intern Med. 2003;139(2):137-147.

15. Legato MJ, Gelzer A, Goland R, et al. Gender-specific care of the patient with diabetes: review and recommendations. Gend Med. 2006;3(2):131-158.

16. Huxley R, Barzi F, Woodward M. Excess risk of fatal coronary heart disease associated with diabetes in men and women: meta-analysis of 37 prospective cohort studies. BMJ. 2006;332(7533):73-78.

17. Larsson CA, Gullberg B, Merlo J, Rastam L, Lindblad U. Female advantage in AMI mortality is reversed in patients with type 2 diabetes in the Skaraborg Project. Diabetes Care. 2005;28(9):2246-2248.

18. Avogaro A, Giorda C, Maggini M, et al. Incidence of coronary heart disease in type 2 diabetic men and women: impact of microvascular complications, treatment, and geographic location. Diabetes Care. 2007;30(5):1241-1247.

19. Zandbergen AA, Sijbrands EJ, Lamberts SW, Bootsma AH. Normotensive women with type 2 diabetes and microalbuminuria are at high risk for macrovascular disease. Diabetes Care. 2006;29(8):1851-1855.

20. Mulnier HE, Seaman HE, Raleigh VS, et al. Risk of stroke in people with type 2 diabetes in the UK: a study using the General Practice Research Database. Diabetologia. 2006;49(12):2859-2865.

21. Tuomilehto J, Rastenyte D, Jousilahti P, Sarti C, Vartiainen E. Diabetes mellitus as a risk factor for death from stroke. Prospective study of the middle-aged Finnish population. Stroke. 1996;27(2):210-215.

22. Hanai K, Babazono T, Yoshida N, et al. Gender differences in the association between HDL cholesterol and the progression of diabetic kidney disease in type 2 diabetic patients. Nephrol Dial Transplant. 2012;27(3):1070-1075.

23. Tuerk PW, Mueller M, Egede LE. Estimating physician effects on glycemic control in the treatment of diabetes: methods, effects sizes, and implications for treatment policy. Diabetes Care. 2008;31(5):869-873.

24. Rubin RR, Peyrot M, Siminerio LM. Health care and patient-reported outcomes: results of the cross-national Diabetes Attitudes, Wishes and Needs (DAWN) study. Diabetes Care. 2006;29(6):1249-1255.

25. McCollum M, Hansen LB, Ghushchyan V, Sullivan PW. Inconsistent health perceptions for US women and men with diabetes. J Womens Health (Larchmt). 2007;16(10):1421-1428.

26. Gucciardi E, Wang SC, DeMelo M, Amaral L, Stewart DE. Characteristics of men and women with diabetes: observations during patients’ initial visit to a diabetes education centre. Can Fam Physician. 2008;54(2):219-227.

27. Chiu CJ, Wray LA. Physical disability trajectories in older Americans with and without diabetes: the role of age, gender, race or ethnicity, and education. Gerontologist. 2011;51(1):51-63.

28. Nielsen AB, de Fine Olivarius N, Gannik D, Hindsberger C, Hollnagel H. Structured personal diabetes care in primary health care affects only women’s HbA1c. Diabetes Care. 2006;29(5):963-969.

29. Shalev V, Chodick G, Heymann AD, Kokia E. Gender differences in healthcare utilization and medical indicators among patients with diabetes. Public Health. 2005;119(1):45-49.

30. Kacerovsky-Bielesz G, Lienhardt S, Hagenhofer M, et al. Sex-related psychological effects on metabolic control in type 2 diabetes mellitus. Diabetologia. 2009;52(5):781-788.

31. Brown SA, Harrist RB, Villagomez ET, Segura M, Barton SA, Hanis CL. Gender and treatment differences in knowledge, health beliefs, and metabolic control in Mexican Americans with type 2 diabetes. Diabetes Educ. 2000;26(3):425-438.

32. Liburd LC, Namageyo-Funa A, Jack L, Jr. Understanding “masculinity” and the challenges of managing type-2 diabetes among African-American men. J Natl Med Assoc. 2007;99(5):550-552, 554–558.

References

1. Centers for Disease Control and Prevention. National diabetes fact sheet: national estimates and general information on diabetes and prediabetes in the United States, 2011. http://www.cdc.gov/diabetes/pubs/pdf/ndfs_2011.pdf. Published 2011. Accessed May 2, 2011.

2. Meisinger C, Thorand B, Schneider A, Stieber J, Doring A, Lowel H. Sex differences in risk factors for incident type 2 diabetes mellitus: the MONICA Augsburg cohort study. Arch Intern Med. 2002;162(1):82-89.

3. Meisinger C, Kandler U, Ladwig KH. Living alone is associated with an increased risk of type 2 diabetes mellitus in men but not women from the general population: the MONICA/KORA Augsburg Cohort Study. Psychosom Med. 2009;71(7):784-788.

4. Baliunas DO, Taylor BJ, Irving H, et al. Alcohol as a risk factor for type 2 diabetes: a systematic review and meta-analysis. Diabetes Care. 2009;32(11):2123-2132.

5. Mozaffarian D, Kamineni A, Carnethon M, Djoussé L, Mukamal KJ, Siscovick D. Lifestyle risk factors and new-onset diabetes mellitus in older adults: the cardiovascular health study. Arch Intern Med. 2009;169(8):798-807.

6. Joosten MM, Grobbee DE, van der A DL, Verschuren WM, Hendriks HF, Beulens JW. Combined effect of alcohol consumption and lifestyle behaviors on risk of type 2 diabetes. Am J Clin Nutr. 2010;91(6):1777-1783.

7. Gigleux I, Gagnon J, St-Pierre A, et al. Moderate alcohol consumption is more cardioprotective in men with the metabolic syndrome. J Nutr. 2006;136(12):3027-3032.

8. Joosten MM, Chiuve SE, Mukamal KJ, Hu FB, Hendriks HF, Rimm EB. Changes in alcohol consumption and subsequent risk of type 2 diabetes in men. Diabetes. 2011;60(1):74-79.

9. Nanri A, Mizoue T, Noda M, et al. Fish intake and type 2 diabetes in Japanese men and women: the Japan Public Health Center-based Prospective Study. Am J Clin Nutr. 2011;94(3):884-891.

10. Choi HK, Willett WC, Stampfer MJ, Rimm E, Hu FB. Dairy consumption and risk of type 2 diabetes mellitus in men: a prospective study. Arch Intern Med. 2005;165(9):997-1003.

11. Fung TT, Hu FB, Pereira MA, et al. Whole-grain intake and the risk of type 2 diabetes: a prospective study in men. Am J Clin Nutr. 2002;76(3):535-540.

12. Lopez-Ridaura R, Willett WC, Rimm EB, et al. Magnesium intake and risk of type 2 diabetes in men and women. Diabetes Care. 2004;27(1):134-140.

13. Nathan DM, Kuenen J, Borg R, et al. Translating the A1C assay into estimated average glucose values. Diabetes Care. 2008;31(8):1473-1478.

14. Levey AS, Coresh J, Balk E, et al. National Kidney Foundation practice guidelines for chronic kidney disease: evaluation, classification, and stratification. Ann Intern Med. 2003;139(2):137-147.

15. Legato MJ, Gelzer A, Goland R, et al. Gender-specific care of the patient with diabetes: review and recommendations. Gend Med. 2006;3(2):131-158.

16. Huxley R, Barzi F, Woodward M. Excess risk of fatal coronary heart disease associated with diabetes in men and women: meta-analysis of 37 prospective cohort studies. BMJ. 2006;332(7533):73-78.

17. Larsson CA, Gullberg B, Merlo J, Rastam L, Lindblad U. Female advantage in AMI mortality is reversed in patients with type 2 diabetes in the Skaraborg Project. Diabetes Care. 2005;28(9):2246-2248.

18. Avogaro A, Giorda C, Maggini M, et al. Incidence of coronary heart disease in type 2 diabetic men and women: impact of microvascular complications, treatment, and geographic location. Diabetes Care. 2007;30(5):1241-1247.

19. Zandbergen AA, Sijbrands EJ, Lamberts SW, Bootsma AH. Normotensive women with type 2 diabetes and microalbuminuria are at high risk for macrovascular disease. Diabetes Care. 2006;29(8):1851-1855.

20. Mulnier HE, Seaman HE, Raleigh VS, et al. Risk of stroke in people with type 2 diabetes in the UK: a study using the General Practice Research Database. Diabetologia. 2006;49(12):2859-2865.

21. Tuomilehto J, Rastenyte D, Jousilahti P, Sarti C, Vartiainen E. Diabetes mellitus as a risk factor for death from stroke. Prospective study of the middle-aged Finnish population. Stroke. 1996;27(2):210-215.

22. Hanai K, Babazono T, Yoshida N, et al. Gender differences in the association between HDL cholesterol and the progression of diabetic kidney disease in type 2 diabetic patients. Nephrol Dial Transplant. 2012;27(3):1070-1075.

23. Tuerk PW, Mueller M, Egede LE. Estimating physician effects on glycemic control in the treatment of diabetes: methods, effects sizes, and implications for treatment policy. Diabetes Care. 2008;31(5):869-873.

24. Rubin RR, Peyrot M, Siminerio LM. Health care and patient-reported outcomes: results of the cross-national Diabetes Attitudes, Wishes and Needs (DAWN) study. Diabetes Care. 2006;29(6):1249-1255.

25. McCollum M, Hansen LB, Ghushchyan V, Sullivan PW. Inconsistent health perceptions for US women and men with diabetes. J Womens Health (Larchmt). 2007;16(10):1421-1428.

26. Gucciardi E, Wang SC, DeMelo M, Amaral L, Stewart DE. Characteristics of men and women with diabetes: observations during patients’ initial visit to a diabetes education centre. Can Fam Physician. 2008;54(2):219-227.

27. Chiu CJ, Wray LA. Physical disability trajectories in older Americans with and without diabetes: the role of age, gender, race or ethnicity, and education. Gerontologist. 2011;51(1):51-63.

28. Nielsen AB, de Fine Olivarius N, Gannik D, Hindsberger C, Hollnagel H. Structured personal diabetes care in primary health care affects only women’s HbA1c. Diabetes Care. 2006;29(5):963-969.

29. Shalev V, Chodick G, Heymann AD, Kokia E. Gender differences in healthcare utilization and medical indicators among patients with diabetes. Public Health. 2005;119(1):45-49.

30. Kacerovsky-Bielesz G, Lienhardt S, Hagenhofer M, et al. Sex-related psychological effects on metabolic control in type 2 diabetes mellitus. Diabetologia. 2009;52(5):781-788.

31. Brown SA, Harrist RB, Villagomez ET, Segura M, Barton SA, Hanis CL. Gender and treatment differences in knowledge, health beliefs, and metabolic control in Mexican Americans with type 2 diabetes. Diabetes Educ. 2000;26(3):425-438.

32. Liburd LC, Namageyo-Funa A, Jack L, Jr. Understanding “masculinity” and the challenges of managing type-2 diabetes among African-American men. J Natl Med Assoc. 2007;99(5):550-552, 554–558.

Issue
The Journal of Family Practice - 61(6)
Issue
The Journal of Family Practice - 61(6)
Page Number
S16-S21
Page Number
S16-S21
Publications
Publications
Topics
Article Type
Display Headline
Managing Type 2 Diabetes in Men
Display Headline
Managing Type 2 Diabetes in Men
Sections
Citation Override
June 2012 · Vol. 61, No. 06 Suppl: S16-S21
Disallow All Ads
Alternative CME
Use ProPublica

Introduction

Article Type
Changed
Thu, 01/24/2019 - 15:00
Display Headline
Introduction

 

DISCLOSURES

Dr. Brunton has disclosed that he is on the advisory boards and speakers’ bureaus for Boehringer Ingelheim, Eli Lilly, Kowa, Novo Nordisk, Inc, and Teva Pharmaceuticals, and is on the advisory boards for Abbott and Sunovion.

 

A decade ago, the World Health Organization suggested that “increasing the effectiveness of adherence interventions may have a far greater impact on the health of the population than any improvement in specific medical treatments.”1 A recent survey found that medication adherence rates over the course of 1 year were 24% for patients with depression, 36% with diabetes, 54% with epilepsy, 32% with dyslipidemia, and 42% with hypertension.2 Poor adherence rates such as these contribute to the low rates of disease control in patients with diabetes, dyslipidemia, hypertension, and other chronic diseases.3,4 Since chronic diseases are largely self-managed, effective patient self-management is critical to good health-related outcomes. To help patients self-manage their diseases, the family physician must work collaboratively with each patient to select, initiate, and modify therapy based upon the patient’s needs, interests, and capabilities. Just as there are important differences between children and adults, men and women often manifest diseases differently. In addition, men and women often deal with and manage their diseases in different ways. While “Men’s Health” is often considered to be a focus on the urogenital tract, we have sought to also focus on diseases that have a high prevalence in men, or where treatment in men may be different compared with women.

The first 2 articles in this supplement on men’s health concern 2 diseases increasingly encountered by men as they age. Dr. Martin Miner provides his thoughts about screening for and diagnosing benign prostatic hyperplasia, including strategies to promote patient report of symptoms and the role of the prostate specific antigen test. A case study is utilized to illustrate key considerations when selecting therapy and promoting patient self-management of benign prostatic hyperplasia. Dr. Gary Ruoff follows a patient from initial diagnosis of gout through selection of treatment for the acute flare and chronic treatment with urate-lowering therapy. A treatment plan is presented at each management step. In the next article, Dr. Richard Aguilar takes a case study approach to describe key risk factors for type 2 diabetes mellitus in men. He also discusses how men self-manage type 2 diabetes differently than women and provides insight as to how to address common psychosocial issues in men. Drs. Louis Kuritzky and José Díez review clinical experience with the two newest antiplatelet agents, prasugrel and ticagrelor. Answers are also provided to common questions and problems encountered with the use of antiplatelet agents in primary care. The next 2 articles focus on major modifiable risk factors contributing to cardiovascular disease. In the first, Dr. Michael Cobble focuses on patient assessment and treatment strategies to help men modify abnormal lipid levels and blood pressure for primary prevention of coronary heart disease. Finally, a more in-depth discussion of dyslipidemia is provided by Dr. Peter Toth, who begins by providing a brief overview of the current evidence regarding the long-term benefits of statin therapy, as well as his clinical perspective on the newest statin, pitavastatin. Dr. Toth also provides answers to many problems frequently encountered in the primary care management of patients with dyslipidemia using statin therapy.

It is my hope that the insights provided by these authors will be helpful to family physicians in managing their male patients with these common chronic diseases.

References

1. World Health Organization. Adherence to long-term therapies: evidence for action. http://www.who.int/chp/knowledge/publications/adherence_full_report.pdf. Published 2003. Accessed May 7, 2012.

2. Khanna R, Pace PF, Mahabaleshwarkar R, Basak RS, Datar M, Banahan BF. Medication adherence among recipients with chronic diseases enrolled in a state medicaid program [published online ahead of print March 8, 2012]. Popul Health Manag. doi:10.1089/pop.2011.0069.

3. Ford ES. Trends in the control of risk factors for cardiovascular disease among adults with diagnosed diabetes: findings from the National Health and Nutrition Examination Survey 1999-2008. J Diabetes. 2011;3(4):337-347.

4. Roger VL, Go AS, Lloyd-Jones DM, et al. American Heart Association Statistics Committee and Stroke Statistics Subcommittee. Heart disease and stroke statistics—2012 update: a report from the American Heart Association. Circulation. 2012;125(1):e2-e220.

Author and Disclosure Information

Stephen A. Brunton, MD, FAAFP
Adjunct Clinical Professor, Department of Family Medicine, University of North Carolina, Chapel Hill, NC, Executive Vice President for Education, Primary Care Education Consortium, Charlotte, NC

Issue
The Journal of Family Practice - 61(6)
Publications
Topics
Page Number
S4-S4
Sections
Author and Disclosure Information

Stephen A. Brunton, MD, FAAFP
Adjunct Clinical Professor, Department of Family Medicine, University of North Carolina, Chapel Hill, NC, Executive Vice President for Education, Primary Care Education Consortium, Charlotte, NC

Author and Disclosure Information

Stephen A. Brunton, MD, FAAFP
Adjunct Clinical Professor, Department of Family Medicine, University of North Carolina, Chapel Hill, NC, Executive Vice President for Education, Primary Care Education Consortium, Charlotte, NC

 

DISCLOSURES

Dr. Brunton has disclosed that he is on the advisory boards and speakers’ bureaus for Boehringer Ingelheim, Eli Lilly, Kowa, Novo Nordisk, Inc, and Teva Pharmaceuticals, and is on the advisory boards for Abbott and Sunovion.

 

A decade ago, the World Health Organization suggested that “increasing the effectiveness of adherence interventions may have a far greater impact on the health of the population than any improvement in specific medical treatments.”1 A recent survey found that medication adherence rates over the course of 1 year were 24% for patients with depression, 36% with diabetes, 54% with epilepsy, 32% with dyslipidemia, and 42% with hypertension.2 Poor adherence rates such as these contribute to the low rates of disease control in patients with diabetes, dyslipidemia, hypertension, and other chronic diseases.3,4 Since chronic diseases are largely self-managed, effective patient self-management is critical to good health-related outcomes. To help patients self-manage their diseases, the family physician must work collaboratively with each patient to select, initiate, and modify therapy based upon the patient’s needs, interests, and capabilities. Just as there are important differences between children and adults, men and women often manifest diseases differently. In addition, men and women often deal with and manage their diseases in different ways. While “Men’s Health” is often considered to be a focus on the urogenital tract, we have sought to also focus on diseases that have a high prevalence in men, or where treatment in men may be different compared with women.

The first 2 articles in this supplement on men’s health concern 2 diseases increasingly encountered by men as they age. Dr. Martin Miner provides his thoughts about screening for and diagnosing benign prostatic hyperplasia, including strategies to promote patient report of symptoms and the role of the prostate specific antigen test. A case study is utilized to illustrate key considerations when selecting therapy and promoting patient self-management of benign prostatic hyperplasia. Dr. Gary Ruoff follows a patient from initial diagnosis of gout through selection of treatment for the acute flare and chronic treatment with urate-lowering therapy. A treatment plan is presented at each management step. In the next article, Dr. Richard Aguilar takes a case study approach to describe key risk factors for type 2 diabetes mellitus in men. He also discusses how men self-manage type 2 diabetes differently than women and provides insight as to how to address common psychosocial issues in men. Drs. Louis Kuritzky and José Díez review clinical experience with the two newest antiplatelet agents, prasugrel and ticagrelor. Answers are also provided to common questions and problems encountered with the use of antiplatelet agents in primary care. The next 2 articles focus on major modifiable risk factors contributing to cardiovascular disease. In the first, Dr. Michael Cobble focuses on patient assessment and treatment strategies to help men modify abnormal lipid levels and blood pressure for primary prevention of coronary heart disease. Finally, a more in-depth discussion of dyslipidemia is provided by Dr. Peter Toth, who begins by providing a brief overview of the current evidence regarding the long-term benefits of statin therapy, as well as his clinical perspective on the newest statin, pitavastatin. Dr. Toth also provides answers to many problems frequently encountered in the primary care management of patients with dyslipidemia using statin therapy.

It is my hope that the insights provided by these authors will be helpful to family physicians in managing their male patients with these common chronic diseases.

 

DISCLOSURES

Dr. Brunton has disclosed that he is on the advisory boards and speakers’ bureaus for Boehringer Ingelheim, Eli Lilly, Kowa, Novo Nordisk, Inc, and Teva Pharmaceuticals, and is on the advisory boards for Abbott and Sunovion.

 

A decade ago, the World Health Organization suggested that “increasing the effectiveness of adherence interventions may have a far greater impact on the health of the population than any improvement in specific medical treatments.”1 A recent survey found that medication adherence rates over the course of 1 year were 24% for patients with depression, 36% with diabetes, 54% with epilepsy, 32% with dyslipidemia, and 42% with hypertension.2 Poor adherence rates such as these contribute to the low rates of disease control in patients with diabetes, dyslipidemia, hypertension, and other chronic diseases.3,4 Since chronic diseases are largely self-managed, effective patient self-management is critical to good health-related outcomes. To help patients self-manage their diseases, the family physician must work collaboratively with each patient to select, initiate, and modify therapy based upon the patient’s needs, interests, and capabilities. Just as there are important differences between children and adults, men and women often manifest diseases differently. In addition, men and women often deal with and manage their diseases in different ways. While “Men’s Health” is often considered to be a focus on the urogenital tract, we have sought to also focus on diseases that have a high prevalence in men, or where treatment in men may be different compared with women.

The first 2 articles in this supplement on men’s health concern 2 diseases increasingly encountered by men as they age. Dr. Martin Miner provides his thoughts about screening for and diagnosing benign prostatic hyperplasia, including strategies to promote patient report of symptoms and the role of the prostate specific antigen test. A case study is utilized to illustrate key considerations when selecting therapy and promoting patient self-management of benign prostatic hyperplasia. Dr. Gary Ruoff follows a patient from initial diagnosis of gout through selection of treatment for the acute flare and chronic treatment with urate-lowering therapy. A treatment plan is presented at each management step. In the next article, Dr. Richard Aguilar takes a case study approach to describe key risk factors for type 2 diabetes mellitus in men. He also discusses how men self-manage type 2 diabetes differently than women and provides insight as to how to address common psychosocial issues in men. Drs. Louis Kuritzky and José Díez review clinical experience with the two newest antiplatelet agents, prasugrel and ticagrelor. Answers are also provided to common questions and problems encountered with the use of antiplatelet agents in primary care. The next 2 articles focus on major modifiable risk factors contributing to cardiovascular disease. In the first, Dr. Michael Cobble focuses on patient assessment and treatment strategies to help men modify abnormal lipid levels and blood pressure for primary prevention of coronary heart disease. Finally, a more in-depth discussion of dyslipidemia is provided by Dr. Peter Toth, who begins by providing a brief overview of the current evidence regarding the long-term benefits of statin therapy, as well as his clinical perspective on the newest statin, pitavastatin. Dr. Toth also provides answers to many problems frequently encountered in the primary care management of patients with dyslipidemia using statin therapy.

It is my hope that the insights provided by these authors will be helpful to family physicians in managing their male patients with these common chronic diseases.

References

1. World Health Organization. Adherence to long-term therapies: evidence for action. http://www.who.int/chp/knowledge/publications/adherence_full_report.pdf. Published 2003. Accessed May 7, 2012.

2. Khanna R, Pace PF, Mahabaleshwarkar R, Basak RS, Datar M, Banahan BF. Medication adherence among recipients with chronic diseases enrolled in a state medicaid program [published online ahead of print March 8, 2012]. Popul Health Manag. doi:10.1089/pop.2011.0069.

3. Ford ES. Trends in the control of risk factors for cardiovascular disease among adults with diagnosed diabetes: findings from the National Health and Nutrition Examination Survey 1999-2008. J Diabetes. 2011;3(4):337-347.

4. Roger VL, Go AS, Lloyd-Jones DM, et al. American Heart Association Statistics Committee and Stroke Statistics Subcommittee. Heart disease and stroke statistics—2012 update: a report from the American Heart Association. Circulation. 2012;125(1):e2-e220.

References

1. World Health Organization. Adherence to long-term therapies: evidence for action. http://www.who.int/chp/knowledge/publications/adherence_full_report.pdf. Published 2003. Accessed May 7, 2012.

2. Khanna R, Pace PF, Mahabaleshwarkar R, Basak RS, Datar M, Banahan BF. Medication adherence among recipients with chronic diseases enrolled in a state medicaid program [published online ahead of print March 8, 2012]. Popul Health Manag. doi:10.1089/pop.2011.0069.

3. Ford ES. Trends in the control of risk factors for cardiovascular disease among adults with diagnosed diabetes: findings from the National Health and Nutrition Examination Survey 1999-2008. J Diabetes. 2011;3(4):337-347.

4. Roger VL, Go AS, Lloyd-Jones DM, et al. American Heart Association Statistics Committee and Stroke Statistics Subcommittee. Heart disease and stroke statistics—2012 update: a report from the American Heart Association. Circulation. 2012;125(1):e2-e220.

Issue
The Journal of Family Practice - 61(6)
Issue
The Journal of Family Practice - 61(6)
Page Number
S4-S4
Page Number
S4-S4
Publications
Publications
Topics
Article Type
Display Headline
Introduction
Display Headline
Introduction
Sections
Citation Override
June 2012 · Vol. 61, No. 06 Suppl: S4
Disallow All Ads
Alternative CME
Use ProPublica

Sports concussion: A return-to-play guide

Article Type
Changed
Mon, 01/14/2019 - 10:47
Display Headline
Sports concussion: A return-to-play guide

PRACTICE RECOMMENDATIONS

Prohibit sports participation as long as a patient exhibits concussive symptoms after a head injury. C

Evaluate a patient’s balance and cognitive function to help gauge the severity of concussion and the likely delay in a return to sports activity. C

Use a stepwise protocol in returning an asymptomatic patient to full sports activity. C

Strength of recommendation (SOR)

A Good-quality patient-oriented evidence
B Inconsistent or limited-quality patient-oriented evidence
C Consensus, usual practice, opinion, disease-oriented evidence, case series

CASE KD is an 18-year-old high school basketball player who was knocked backwards during a game, hitting her head on the floor. She had immediate head and neck pain but no loss of consciousness; she was transported by EMS to the local emergency department (ED) for further evaluation. Results of head and neck CT scans were normal, and she was discharged home. Four days later, KD’s parents brought her to our office because she was experiencing ongoing headache, phonophobia, nausea, light-headedness, poor balance, increased sleepiness, and irritability.

The Centers for Disease Control and Prevention estimate that approximately 300,000 sports concussions occur yearly in the United States,1 and that 135,000 of these cases are treated in EDs.2 These numbers have not gone unnoticed in the consumer press. Over the past 18 months, Sports Illustrated, Newsweek, and Time3-5 have published stories on sports-related concussion, helping to raise public awareness of its risks.

Recommendations for practitioners have changed. In 1997, the American Academy of Neurology6 published one-size-fits-all guidelines on managing concussion, using levels of symptomatology and loss of consciousness to grade the severity of concussion from 1 to 3. These guidelines were similar to the Cantu and Colorado guidelines of the early 1990s.7,8 Since then, however, the diagnostic criteria and expert opinion about treatment and return to physical activity have changed. Indeed, several medical organizations9-12 now recommend a more individualized approach to evaluation and management, which we describe here.

It begins with a definition

While there is no single agreed-upon characterization of “concussion,” the 3rd International Conference on Concussion in Sport (ICCS)12 provides this definition:

Concussion is defined as a complex pathophysiological process affecting the brain, induced by traumatic biomechanical forces. Several common features that incorporate clinical, pathologic, and biomechanical injury constructs that may be utilized in defining the nature of a concussive head injury include:

  1. Concussion may be caused either by a direct blow to the head, face, or neck or a blow elsewhere on the body with an ‘‘impulsive’’ force transmitted to the head.
  2. Concussion typically results in the rapid onset of short-lived impairment of neurologic function that resolves spontaneously.
  3. Concussion may result in neuropathological changes but the acute clinical symptoms largely reflect a functional disturbance rather than a structural injury.
  4. Concussion results in a graded set of clinical symptoms that may or may not involve loss of consciousness. Resolution of the clinical and cognitive symptoms typically follows a sequential course.… In a small percentage of cases, however, postconcussive symptoms may be prolonged.
  5. No abnormality on standard structural neuroimaging studies is seen in concussion.

Office evaluation
Obtain a thorough history and conduct a neurologic evaluation and musculoskeletal examination of the head and neck.

Clues to expected length of recovery

A patient with a concussion may lose consciousness after the impact, or have a brief convulsion that is not a seizure.13 In the periodimmediately after the injury, the patient may exhibit a constellation of such signs and symptoms as headache, confusion, a dazed look, dilated pupils, amnesia, poor balance, nausea, or vomiting. These features typically resolve over time, but may persist for weeks or months. Anterograde or retrograde amnesia may also occur. TABLE 1 details a more complete list of concussion symptoms. If the patient is a child or young adult, it is useful to have a parent present at the office visit to describe the patient’s mood, sleep, appetite, and overall health after the injury.

Factors that may portend a longer recovery include a previous concussion, retrograde or anterograde amnesia, younger age, and female sex.14

Dire problems beyond concussion. Complaints or historical elements inconsistent with concussion that should be considered red flags include any focal neurologic complaints, vomiting or headache that worsens after a period of improvement, or obtundation or disorientation that has worsened since the injury. With such findings, consider more serious head injuries and arrange for a more complete immediate neurologic work-up.

 

 

CASE Our neurologic examination yielded normal results. However, our patient was unable to balance correctly on one leg. The cognitive exam revealed a deficit in short-term memory. We diagnosed a concussion, advised her to refrain from sports, and prescribed cognitive rest. A return to school for half days would be considered once her symptoms began to resolve.

TABLE 1
Signs and symptoms commonly associated with concussion

Headache
“Pressure in head”
Neck pain
Nausea or vomiting
Dizziness
Blurred vision
Balance problems
Sensitivity to light
Sensitivity to noise
Feeling slowed down
Feeling like “in a fog”
“Don’t feel right”
Difficulty concentrating
Difficulty remembering
Fatigue or low energy
Confusion
Drowsiness
Trouble falling asleep
Irritability
Sadness
Nervousness or anxiety
Adapted from SCAT2 in Appendix 1 of: McCrory P, Meeuwisse W, Johnston K, et al. Br J Sports Med. 2009;43(suppl 1):i76-i90.12

Options for the neurologic exam
With a simple concussion, expect a normal neurologic examination, with the possible exception of the ability to balance. Head imaging is not necessary in the setting of suspected concussion, because results of computed tomography (CT) and magnetic resonance imaging (MRI) will likely be normal.12

Balance testing can assist in the diagnosis of concussion and the monitoring of recovery from injury.15-17 The Balance Error Scoring System (BESS)15 is a validated and simple test that can be done in the office. The test involves 3 consecutive stances: (a) normal stance with feet comfortably apart and hands on hips, (b) with feet aligned heel to toe with the dominant leg in front, and (c) standing on the nondominant leg with the dominant leg flexed 30 degrees at the hip. Have the patient repeat each version of the test for 20 seconds with eyes closed, on a stable and then unstable surface (eg, foam mat).

It’s recommended that another staff member be present to spot the patient in case of a fall. A link to a complete description of the test and scoring details is provided in the Web resources box.

Assess cognitive function. One tool for assessing cognitive function is the Sports Concussion Assessment Tool 2 (SCAT2).12 SCAT2 includes newer, as yet unvalidated sections and several sections that have been independently studied and proven useful in diagnosing concussion. Validated sections are the Maddocks questions, used only at the time and place of injury18 ; the modified BESS15 ; and the Standardized Assessment of Concussion (SAC).19 The SCAT2 and the SAC (which may be used separately) include questions that assist in evaluating short-term memory and attention, and are useful in the physician’s office.

Do computer-based tools help? Another option for cognitive assessment is computer-based neuropsychologic testing developed specifically for use with suspected concussion. Any of these programs can be used in the office by a trained practitioner. Schools may also use the programs under the supervision of an athletic trainer or team physician. Available programs are ImPACT, developed by the University of Pittsburgh (http://impacttest.com); the Cognitive Stability Index (CSI), by HeadMinder (http://www.headminder.com/site/csi/home.html); and the Computerized Cognitive Assessment Tool (CCAT), by CogState/Axon Sports (http://www.axonsports.com). Multiple studies have shown such programs to be useful in diagnosing and monitoring recovery from sports concussion.20-23

However, among sports medicine practitioners, there seems to be a consensus that computer-based neuropsychologic testing is most useful when a baseline score exists. Baseline testing is usually done preseason on athletes in a healthy state. If a baseline score is not available, a patient’s postinjury score is compared with normative data produced by the developer of the individual test.

Few, if any, outcome studies have been conducted to determine whether computer-basedneuropsychologic testing provides any meaningful improvement in the care of athletes who have suffered concussions. There is also concern that few studies by independent sources have replicated the data disseminated by developers of the tests.24,25 The most recent guidelines by the 3rd ICCS recommend using neuropsychologic testing only as an aid to an overall medical evaluation, not as the sole determinant of recovery from concussion.12 Numerous studies now underway may help clarify the role of neuropsychologic testing in concussion.

CASE By the time of our follow-up exam 7 days later (11 days from injury), KD had returned to school for half days, but her phonophobia and headaches worsened at school and she had difficulty focusing on academic tasks. Neurologic, balance, and cognitive exams were all normal. We advised her to gradually return to school full time while abstaining from sporting activity.

 

 

At 16 days’ follow-up (20 days from injury), KD had returned to school full time and said she felt more like herself, although she continued to have daily headaches and phonophobia. All exam results were normal. Sports were still off limits, and we told her to expect at least 7 more days of respite before any return to exercise would be allowed.

At 23 days’ follow-up (27 days from injury), KD’s symptoms had completely resolved, and all exam results were normal. We prescribed a stepwise return to athletic activity over the next 10 days and discussed this plan with the school’s athletic trainer, who would supervise her return to play.

Web resources

American Academy of Neurology (AAN). Position Statement on Sports Concussion. http://www.aan.com/globals/axon/assets/7913.pdf

American Academy of Pediatrics (AAP). Sports-Related Concussion in Children and Adolescents. http://pediatrics.aappublications.org/cgi/content/abstract/126/3/597

The Balance Error Scoring System (BESS). http://www.sportsconcussion.com/pdf/management/BESSProtocolNATA09.pdf

Centers for Disease Control and Prevention. Concussion and Mild TBI. http://www.cdc.gov/concussion/index.html

Sport Concussion Assessment Tool 2 (SCAT2). http://www.athletictherapy.org/en/pdf/SCAT2.pdf

3rd International Conference on Concussion in Sport. http://bjsm.bmj.com/content/43/Suppl_1/i76.full

Individualize management

The one-size-fits-all approach previously recommended6 is no longer the standard of care. In your initial encounter with the patient (and parents, as appropriate), explain the nature of the injury, expected course of recovery, and requirements for a return to play. Also discuss the possibility of postconcussive syndrome and the risk of rare sequelae such as second impact syndrome.

If the patient is symptomatic or exhibits examination findings consistent with concussion, recommend immediate cessation of sports activity.9-12 With a school-aged athlete, if symptoms reported by the patient or parents are significant, consider prescribing cognitive rest, which can be provided through quiet accommodations at school or perhaps even time off from school or exams.12,24 In the early period of recovery, increased cognitive or physical activity can cause symptoms to worsen. With improvement, the patient may return to school half time to lessen the chance of a significant return of symptoms. If half days are tolerated, the patient may transition to full days. Make sure the diagnosis and expectations for recovery are communicated to the appropriate school officials so that necessary accommodationscan be made. If symptoms after the initial office visit are mild, a one-week return to school is appropriate to evaluate the patient’s recovery.

Allowing a return to sports. Once the patient is asymptomatic, and physical and cognitive test results are normal, discuss a return-to-play protocol with the patient (and with parents and athletic trainer or coach, as appropriate). Multiple sources10,11,26 now recommend a stepwise return to play, as detailed by the 3rd ICCS ( TABLE 2 ).12 Increase or decrease the length of the protocol depending on the patient and the specifics of the case.

There is little science to guide the treatment of children with concussion. However, given that their brains are still developing, it’s prudent to be more conservative than with older adolescents or adults. Multiple sources apart from the 3rd ICCS agree with this recommendation. Several authors suggest more cognitive rest and a longer return-to-play protocol in all cases.10,27 In fact, the ICCS committee additionally recommends observing a symptom-free waiting period for pediatric athletes before even starting a return-to-play protocol.

McCrory et al26 suggest that children under age 15 be treated more conservatively than those 15 and older. They suggest treating those 15 and older with the protocol for older adolescents. Specifying an age at which one should always make a decision for or against conservative care can be problematic. However, based on the recommendations above, it would seem reasonable to provide conservative treatment for children younger than high school age and perhaps even those in the early years of high school.

Consider legal implications. Become familiar with state laws that require certain steps in managing sports concussion. The Web site http://www.sportsconcussions.org/laws.html28 lists states with sports concussion statutes, as well as states with bills working their way through the legislative system. Currently, 29 states are listed with laws; 14 more and the District of Columbia have pending legislation.

TABLE 2
Stepwise protocol for return to play

If symptoms recur at any step, have patient return to prior level
1. Light aerobic activityWalking, swimming, exercise bike; keeping exertion <70% of maximum heart rate
2. Sport-specific exercisesExertional drills in sport, eg, running drills in football/soccer, skating drills in hockey
3. Noncontact training drillsProgression to more complex noncontact drills, eg, passing/catching drills in football, shooting/passing in basketball, hitting drills in volleyball
4. Full-contact practiceReturn to full practice if no recurrence of symptoms through first 3 steps and cleared by physician
5. Game activityReturn to full sport participation if no recurrence of symptoms with above steps
Adapted from: McCrory P, Meeuwisse W, Johnston K, et al. Br J Sports Med. 2009;43(suppl 1):i76-i90.12
 

 

Anticipate complications
Most patients with concussions who are managed appropriately do well. However, complications can occur. The most serious complication is second impact syndrome, which usually occurs when concussion is unrecognized or not well managed. While not well understood, this condition is thought to result from a sudden increase in intracranial pressure after a second head injury in an athlete already suffering from concussion symptoms. The injury typically results in serious long-term neurologic deficits, or even fatality.29 Second impact syndrome has been documented as occurring in the same game after an initial injury, as well as in subsequent games.29

A more common, but less serious, complication is postconcussion syndrome.30 This is an ill-defined condition in which the patient suffers from concussive symptoms for an extended period of time, generally for more than 3 months.30 As with acute concussion, the constellation of symptoms ranges from headache to cognitive impairment. In cases of postconcussion syndrome, it is appropriate to consult with neuropsychologists, psychiatrists, or neurologists for assistance with symptoms and associated mood disorders. Similar to acute concussion management, it is generally recommended that athletes not be cleared to resume play while struggling with the symptoms of postconcussion syndrome.30

There have also been recent reports of late-life sequelae in those who have sustained multiple concussions. Depression and dementia have been suggested in surveys of retired NFL players.31,32 There have also been studies both suggesting14 and questioning33,34 whether multiple concussions result in long-term cognitive deficits. While the evidence available at this time is not firm, there seems to be an increasing belief that multiple concussions can affect long-term cognitive abilities. For these reasons, use caution in making return-to-play decisions for patients with multiple concussions or concussions with long-lasting symptoms.

CORRESPONDENCE Aaron M. Lear, MD, 224 West Exchange Street, Suite 440, Akron, OH 44302; [email protected]

References

1. CDC. Sports-related recurrent brain injuries—United States. MMWR Morb Mortal Wkly Rep. 1997;46:224-227.

2. CDC. Brain injury awareness month—March 2010. MMWR Morb Mortal Wkly Rep. 2010;59:235.-

3. Epstein D. The damage done. Sports Illustrated. November 1, 2010:42. Available at: http://sportsillustrated.cnn.com/vault/article/magazine/MAG1176377/index.htm. Accessed May 16, 2012.

4. Kliff S. Heading off sports injuries. Newsweek. February 4, 2010. Available at: http://www.newsweek.com/2010/02/04/heading-off-sports-injuries.html. Accessed February 9, 2011.

5. Kluger J. Headbanger nation. Health special: kids and concussions. Time. February 3, 2011. Available at: http://www.time.com/time/specials/packages/article/0,28804,2043395_2043506_2043494,00.html. Accessed February 9, 2011.

6. American Academy of Neurology. Practice parameter: the management of concussion in sports (summary statement). Report of the quality standards subcommittee. Neurology. 1997;48:581-585.

7. Cantu R. Cerebral concussion in sport. Management and prevention. Sports Med. 1992;14:64-74.

8. Kelly J, Nichols J, Filley C, et al. Concussion in sports. Guidelines for the prevention of catastrophic outcome. JAMA. 1991;266:2867-2869.

9. American Academy of Neurology. Position statement on sports concussion. October 2010. AAN policy 2010-36. Available at: http://www.aan.com/globals/axon/assets/7913.pdf. Accessed February 23, 2011.

10. Halstead M, Walter K. Council on Sports Medicine and Fitness. American Academy of Pediatrics. Clinical report—sport-related concussion in children and adolescents. Pediatrics. 2010;126:597-615.

11. Herring SA, Cantu RC, Guskiewicz KM, et al. Concussion (mild traumatic brain injury) and the team physician: a consensus statement—2011 update. Med Sci Sports Exerc. 2011;43:2412-2422.Available at: http://journals.lww.com/acsm-msse/Fulltext/2011/12000/Concussion__Mild_Traumatic_Brain_Injury__and_the.24.aspx. Accessed February 23, 2011.

12. McCrory P, Meeuwisse W, Johnston K, et al. Consensus statement on concussion in sport: the 3rd International Conference on Concussion in Sport held in Zurich, November 2008. Br J Sports Med. 2009;43(suppl 1):i76-i90.

13. Ropper A, Gorson K. Clinical practice. Concussion. N Engl J Med. 2007;356:166-172.

14. Reddy C, Collins MW. Sports concussion: management and predictors of outcome. Curr Sports Med Rep. 2009;8:10-15.

15. Guskiewicz KM. Assessment of postural stability following sport-related concussion. Curr Sports Med Rep. 2003;2:24-30.

16. Broglio S, Sosnoff J, Ferrara M. The relationship of athlete-reported concussion symptoms and objective measures of neurocognitive function and postural control. Clin J Sport Med. 2009;19:377-382.

17. Reimann B, Guskiewicz K. Effects of mild head injury on postural stability as measured through clinical balance testing. J Athl Train. 2000;35:19-25.

18. Maddocks D, Dicker G, Saling M. The assessment of orientation following concussion in athletes. Clin J Sport Med. 1995;5:32-35.

19. McCrea M. Standardized mental status assessment of sports concussion. Clin J Sport Med. 2001;11:176-181.

20. Collie A, Maruff P, Makdissi M, et al. CogSport: reliability and correlation with conventional cognitive tests used in postconcussion medical evaluations. Clin J Sport Med. 2003;13:28-32.

21. Erlanger D, Saliba E, Barth J, et al. Monitoring resolution of postconcussion symptoms in athletes: preliminary results of a web-based neuropsychological test protocol. J Athl Train. 2001;36:280-287.

22. Schatz P, Pardini J, Lovell M, et al. Sensitivity and specificity of the ImPACT Test battery for concussion in athletes. Arch Clin Neuropsychol. 2006;21:91-99.

23. Schatz P, Putz B. Cross-validation of measures used for computer-based assessment of concussion. Appl Neuropsychol. 2006;13:151-159.

24. Kirkwood M, Randolph C, Yeates K. Returning pediatric athletes to play after concussion: the evidence (or lack thereof) behind baseline neuropsychological testing. Acta Pædiatr. 2009;98:1409-1411.

25. Randolph C. Baseline neuropsychological testing in managing sport-related concussion: does it modify risk? Curr Sports Med Rep. 2011;10:21-26.

26. McCrory P, Collie A, Anderson V, et al. Can we manage sport related concussion in children the same as in adults? Br J Sports Med. 2004;38:516-519.

27. d’Hemecourt P. Subacute symptoms of sports-related concussion outpatient management and return to play. Clin Sports Med. 2011;30:63-72.

28. Concussion laws. Available at: http://www.sportsconcussions.org/laws.html. Accessed July 5, 2011.

29. Wetjen N, Pichelmann M, Atkinson J. Second impact syndrome: concussion and second injury brain complications. J Am Coll Surg. 2010;211:553-557.

30. Jotwani V, Harmon KG. Postconcussion syndrome in athletes. Curr Sports Med Rep. 2010;9:21-26.

31. Guskiewicz K, Marshall S, Bailes J, et al. Association between recurrent concussion and late-life cognitive impairment in retired professional football players. Neurosurgery. 2005;57:719-726.

32. Guskiewicz K, Marshall S, Bailes J, et al. Recurrent concussion and risk of depression in retired professional football players. Med Sci Sports Exerc. 2007;39:903-909.

33. Belanger H, Spiegel E, Vanderploeg R. Neuropsychological performance following a history of multiple self-reported concussions: a meta-analysis. J Int Neuropsychol Soc. 2010;16:262-267.

34. Burce J, Echemendia R. History of multiple self-reported concussions is not associated with reduced cognitive abilities. Neurosurgery. 2009;64:100-106.

Article PDF
Author and Disclosure Information

Aaron M. Lear, MD, CAQ
Akron General Sports Medicine, Akron General Center for Family Medicine, Akron, Ohio
[email protected]

Minh-Ha Hoang, DO
Akron General Center for Family Medicine, Akron, Ohio

Issue
The Journal of Family Practice - 61(6)
Publications
Topics
Page Number
323-328
Legacy Keywords
Aaron M. Lear ;MD;CAQ; Minh-Ha Hoang ;DO;sports concussion; evaluative methods; return-to-play; concussive symptoms; balance; sports participation; complex pathophysiological process; direct blow; impulsive force
Sections
Author and Disclosure Information

Aaron M. Lear, MD, CAQ
Akron General Sports Medicine, Akron General Center for Family Medicine, Akron, Ohio
[email protected]

Minh-Ha Hoang, DO
Akron General Center for Family Medicine, Akron, Ohio

Author and Disclosure Information

Aaron M. Lear, MD, CAQ
Akron General Sports Medicine, Akron General Center for Family Medicine, Akron, Ohio
[email protected]

Minh-Ha Hoang, DO
Akron General Center for Family Medicine, Akron, Ohio

Article PDF
Article PDF

PRACTICE RECOMMENDATIONS

Prohibit sports participation as long as a patient exhibits concussive symptoms after a head injury. C

Evaluate a patient’s balance and cognitive function to help gauge the severity of concussion and the likely delay in a return to sports activity. C

Use a stepwise protocol in returning an asymptomatic patient to full sports activity. C

Strength of recommendation (SOR)

A Good-quality patient-oriented evidence
B Inconsistent or limited-quality patient-oriented evidence
C Consensus, usual practice, opinion, disease-oriented evidence, case series

CASE KD is an 18-year-old high school basketball player who was knocked backwards during a game, hitting her head on the floor. She had immediate head and neck pain but no loss of consciousness; she was transported by EMS to the local emergency department (ED) for further evaluation. Results of head and neck CT scans were normal, and she was discharged home. Four days later, KD’s parents brought her to our office because she was experiencing ongoing headache, phonophobia, nausea, light-headedness, poor balance, increased sleepiness, and irritability.

The Centers for Disease Control and Prevention estimate that approximately 300,000 sports concussions occur yearly in the United States,1 and that 135,000 of these cases are treated in EDs.2 These numbers have not gone unnoticed in the consumer press. Over the past 18 months, Sports Illustrated, Newsweek, and Time3-5 have published stories on sports-related concussion, helping to raise public awareness of its risks.

Recommendations for practitioners have changed. In 1997, the American Academy of Neurology6 published one-size-fits-all guidelines on managing concussion, using levels of symptomatology and loss of consciousness to grade the severity of concussion from 1 to 3. These guidelines were similar to the Cantu and Colorado guidelines of the early 1990s.7,8 Since then, however, the diagnostic criteria and expert opinion about treatment and return to physical activity have changed. Indeed, several medical organizations9-12 now recommend a more individualized approach to evaluation and management, which we describe here.

It begins with a definition

While there is no single agreed-upon characterization of “concussion,” the 3rd International Conference on Concussion in Sport (ICCS)12 provides this definition:

Concussion is defined as a complex pathophysiological process affecting the brain, induced by traumatic biomechanical forces. Several common features that incorporate clinical, pathologic, and biomechanical injury constructs that may be utilized in defining the nature of a concussive head injury include:

  1. Concussion may be caused either by a direct blow to the head, face, or neck or a blow elsewhere on the body with an ‘‘impulsive’’ force transmitted to the head.
  2. Concussion typically results in the rapid onset of short-lived impairment of neurologic function that resolves spontaneously.
  3. Concussion may result in neuropathological changes but the acute clinical symptoms largely reflect a functional disturbance rather than a structural injury.
  4. Concussion results in a graded set of clinical symptoms that may or may not involve loss of consciousness. Resolution of the clinical and cognitive symptoms typically follows a sequential course.… In a small percentage of cases, however, postconcussive symptoms may be prolonged.
  5. No abnormality on standard structural neuroimaging studies is seen in concussion.

Office evaluation
Obtain a thorough history and conduct a neurologic evaluation and musculoskeletal examination of the head and neck.

Clues to expected length of recovery

A patient with a concussion may lose consciousness after the impact, or have a brief convulsion that is not a seizure.13 In the periodimmediately after the injury, the patient may exhibit a constellation of such signs and symptoms as headache, confusion, a dazed look, dilated pupils, amnesia, poor balance, nausea, or vomiting. These features typically resolve over time, but may persist for weeks or months. Anterograde or retrograde amnesia may also occur. TABLE 1 details a more complete list of concussion symptoms. If the patient is a child or young adult, it is useful to have a parent present at the office visit to describe the patient’s mood, sleep, appetite, and overall health after the injury.

Factors that may portend a longer recovery include a previous concussion, retrograde or anterograde amnesia, younger age, and female sex.14

Dire problems beyond concussion. Complaints or historical elements inconsistent with concussion that should be considered red flags include any focal neurologic complaints, vomiting or headache that worsens after a period of improvement, or obtundation or disorientation that has worsened since the injury. With such findings, consider more serious head injuries and arrange for a more complete immediate neurologic work-up.

 

 

CASE Our neurologic examination yielded normal results. However, our patient was unable to balance correctly on one leg. The cognitive exam revealed a deficit in short-term memory. We diagnosed a concussion, advised her to refrain from sports, and prescribed cognitive rest. A return to school for half days would be considered once her symptoms began to resolve.

TABLE 1
Signs and symptoms commonly associated with concussion

Headache
“Pressure in head”
Neck pain
Nausea or vomiting
Dizziness
Blurred vision
Balance problems
Sensitivity to light
Sensitivity to noise
Feeling slowed down
Feeling like “in a fog”
“Don’t feel right”
Difficulty concentrating
Difficulty remembering
Fatigue or low energy
Confusion
Drowsiness
Trouble falling asleep
Irritability
Sadness
Nervousness or anxiety
Adapted from SCAT2 in Appendix 1 of: McCrory P, Meeuwisse W, Johnston K, et al. Br J Sports Med. 2009;43(suppl 1):i76-i90.12

Options for the neurologic exam
With a simple concussion, expect a normal neurologic examination, with the possible exception of the ability to balance. Head imaging is not necessary in the setting of suspected concussion, because results of computed tomography (CT) and magnetic resonance imaging (MRI) will likely be normal.12

Balance testing can assist in the diagnosis of concussion and the monitoring of recovery from injury.15-17 The Balance Error Scoring System (BESS)15 is a validated and simple test that can be done in the office. The test involves 3 consecutive stances: (a) normal stance with feet comfortably apart and hands on hips, (b) with feet aligned heel to toe with the dominant leg in front, and (c) standing on the nondominant leg with the dominant leg flexed 30 degrees at the hip. Have the patient repeat each version of the test for 20 seconds with eyes closed, on a stable and then unstable surface (eg, foam mat).

It’s recommended that another staff member be present to spot the patient in case of a fall. A link to a complete description of the test and scoring details is provided in the Web resources box.

Assess cognitive function. One tool for assessing cognitive function is the Sports Concussion Assessment Tool 2 (SCAT2).12 SCAT2 includes newer, as yet unvalidated sections and several sections that have been independently studied and proven useful in diagnosing concussion. Validated sections are the Maddocks questions, used only at the time and place of injury18 ; the modified BESS15 ; and the Standardized Assessment of Concussion (SAC).19 The SCAT2 and the SAC (which may be used separately) include questions that assist in evaluating short-term memory and attention, and are useful in the physician’s office.

Do computer-based tools help? Another option for cognitive assessment is computer-based neuropsychologic testing developed specifically for use with suspected concussion. Any of these programs can be used in the office by a trained practitioner. Schools may also use the programs under the supervision of an athletic trainer or team physician. Available programs are ImPACT, developed by the University of Pittsburgh (http://impacttest.com); the Cognitive Stability Index (CSI), by HeadMinder (http://www.headminder.com/site/csi/home.html); and the Computerized Cognitive Assessment Tool (CCAT), by CogState/Axon Sports (http://www.axonsports.com). Multiple studies have shown such programs to be useful in diagnosing and monitoring recovery from sports concussion.20-23

However, among sports medicine practitioners, there seems to be a consensus that computer-based neuropsychologic testing is most useful when a baseline score exists. Baseline testing is usually done preseason on athletes in a healthy state. If a baseline score is not available, a patient’s postinjury score is compared with normative data produced by the developer of the individual test.

Few, if any, outcome studies have been conducted to determine whether computer-basedneuropsychologic testing provides any meaningful improvement in the care of athletes who have suffered concussions. There is also concern that few studies by independent sources have replicated the data disseminated by developers of the tests.24,25 The most recent guidelines by the 3rd ICCS recommend using neuropsychologic testing only as an aid to an overall medical evaluation, not as the sole determinant of recovery from concussion.12 Numerous studies now underway may help clarify the role of neuropsychologic testing in concussion.

CASE By the time of our follow-up exam 7 days later (11 days from injury), KD had returned to school for half days, but her phonophobia and headaches worsened at school and she had difficulty focusing on academic tasks. Neurologic, balance, and cognitive exams were all normal. We advised her to gradually return to school full time while abstaining from sporting activity.

 

 

At 16 days’ follow-up (20 days from injury), KD had returned to school full time and said she felt more like herself, although she continued to have daily headaches and phonophobia. All exam results were normal. Sports were still off limits, and we told her to expect at least 7 more days of respite before any return to exercise would be allowed.

At 23 days’ follow-up (27 days from injury), KD’s symptoms had completely resolved, and all exam results were normal. We prescribed a stepwise return to athletic activity over the next 10 days and discussed this plan with the school’s athletic trainer, who would supervise her return to play.

Web resources

American Academy of Neurology (AAN). Position Statement on Sports Concussion. http://www.aan.com/globals/axon/assets/7913.pdf

American Academy of Pediatrics (AAP). Sports-Related Concussion in Children and Adolescents. http://pediatrics.aappublications.org/cgi/content/abstract/126/3/597

The Balance Error Scoring System (BESS). http://www.sportsconcussion.com/pdf/management/BESSProtocolNATA09.pdf

Centers for Disease Control and Prevention. Concussion and Mild TBI. http://www.cdc.gov/concussion/index.html

Sport Concussion Assessment Tool 2 (SCAT2). http://www.athletictherapy.org/en/pdf/SCAT2.pdf

3rd International Conference on Concussion in Sport. http://bjsm.bmj.com/content/43/Suppl_1/i76.full

Individualize management

The one-size-fits-all approach previously recommended6 is no longer the standard of care. In your initial encounter with the patient (and parents, as appropriate), explain the nature of the injury, expected course of recovery, and requirements for a return to play. Also discuss the possibility of postconcussive syndrome and the risk of rare sequelae such as second impact syndrome.

If the patient is symptomatic or exhibits examination findings consistent with concussion, recommend immediate cessation of sports activity.9-12 With a school-aged athlete, if symptoms reported by the patient or parents are significant, consider prescribing cognitive rest, which can be provided through quiet accommodations at school or perhaps even time off from school or exams.12,24 In the early period of recovery, increased cognitive or physical activity can cause symptoms to worsen. With improvement, the patient may return to school half time to lessen the chance of a significant return of symptoms. If half days are tolerated, the patient may transition to full days. Make sure the diagnosis and expectations for recovery are communicated to the appropriate school officials so that necessary accommodationscan be made. If symptoms after the initial office visit are mild, a one-week return to school is appropriate to evaluate the patient’s recovery.

Allowing a return to sports. Once the patient is asymptomatic, and physical and cognitive test results are normal, discuss a return-to-play protocol with the patient (and with parents and athletic trainer or coach, as appropriate). Multiple sources10,11,26 now recommend a stepwise return to play, as detailed by the 3rd ICCS ( TABLE 2 ).12 Increase or decrease the length of the protocol depending on the patient and the specifics of the case.

There is little science to guide the treatment of children with concussion. However, given that their brains are still developing, it’s prudent to be more conservative than with older adolescents or adults. Multiple sources apart from the 3rd ICCS agree with this recommendation. Several authors suggest more cognitive rest and a longer return-to-play protocol in all cases.10,27 In fact, the ICCS committee additionally recommends observing a symptom-free waiting period for pediatric athletes before even starting a return-to-play protocol.

McCrory et al26 suggest that children under age 15 be treated more conservatively than those 15 and older. They suggest treating those 15 and older with the protocol for older adolescents. Specifying an age at which one should always make a decision for or against conservative care can be problematic. However, based on the recommendations above, it would seem reasonable to provide conservative treatment for children younger than high school age and perhaps even those in the early years of high school.

Consider legal implications. Become familiar with state laws that require certain steps in managing sports concussion. The Web site http://www.sportsconcussions.org/laws.html28 lists states with sports concussion statutes, as well as states with bills working their way through the legislative system. Currently, 29 states are listed with laws; 14 more and the District of Columbia have pending legislation.

TABLE 2
Stepwise protocol for return to play

If symptoms recur at any step, have patient return to prior level
1. Light aerobic activityWalking, swimming, exercise bike; keeping exertion <70% of maximum heart rate
2. Sport-specific exercisesExertional drills in sport, eg, running drills in football/soccer, skating drills in hockey
3. Noncontact training drillsProgression to more complex noncontact drills, eg, passing/catching drills in football, shooting/passing in basketball, hitting drills in volleyball
4. Full-contact practiceReturn to full practice if no recurrence of symptoms through first 3 steps and cleared by physician
5. Game activityReturn to full sport participation if no recurrence of symptoms with above steps
Adapted from: McCrory P, Meeuwisse W, Johnston K, et al. Br J Sports Med. 2009;43(suppl 1):i76-i90.12
 

 

Anticipate complications
Most patients with concussions who are managed appropriately do well. However, complications can occur. The most serious complication is second impact syndrome, which usually occurs when concussion is unrecognized or not well managed. While not well understood, this condition is thought to result from a sudden increase in intracranial pressure after a second head injury in an athlete already suffering from concussion symptoms. The injury typically results in serious long-term neurologic deficits, or even fatality.29 Second impact syndrome has been documented as occurring in the same game after an initial injury, as well as in subsequent games.29

A more common, but less serious, complication is postconcussion syndrome.30 This is an ill-defined condition in which the patient suffers from concussive symptoms for an extended period of time, generally for more than 3 months.30 As with acute concussion, the constellation of symptoms ranges from headache to cognitive impairment. In cases of postconcussion syndrome, it is appropriate to consult with neuropsychologists, psychiatrists, or neurologists for assistance with symptoms and associated mood disorders. Similar to acute concussion management, it is generally recommended that athletes not be cleared to resume play while struggling with the symptoms of postconcussion syndrome.30

There have also been recent reports of late-life sequelae in those who have sustained multiple concussions. Depression and dementia have been suggested in surveys of retired NFL players.31,32 There have also been studies both suggesting14 and questioning33,34 whether multiple concussions result in long-term cognitive deficits. While the evidence available at this time is not firm, there seems to be an increasing belief that multiple concussions can affect long-term cognitive abilities. For these reasons, use caution in making return-to-play decisions for patients with multiple concussions or concussions with long-lasting symptoms.

CORRESPONDENCE Aaron M. Lear, MD, 224 West Exchange Street, Suite 440, Akron, OH 44302; [email protected]

PRACTICE RECOMMENDATIONS

Prohibit sports participation as long as a patient exhibits concussive symptoms after a head injury. C

Evaluate a patient’s balance and cognitive function to help gauge the severity of concussion and the likely delay in a return to sports activity. C

Use a stepwise protocol in returning an asymptomatic patient to full sports activity. C

Strength of recommendation (SOR)

A Good-quality patient-oriented evidence
B Inconsistent or limited-quality patient-oriented evidence
C Consensus, usual practice, opinion, disease-oriented evidence, case series

CASE KD is an 18-year-old high school basketball player who was knocked backwards during a game, hitting her head on the floor. She had immediate head and neck pain but no loss of consciousness; she was transported by EMS to the local emergency department (ED) for further evaluation. Results of head and neck CT scans were normal, and she was discharged home. Four days later, KD’s parents brought her to our office because she was experiencing ongoing headache, phonophobia, nausea, light-headedness, poor balance, increased sleepiness, and irritability.

The Centers for Disease Control and Prevention estimate that approximately 300,000 sports concussions occur yearly in the United States,1 and that 135,000 of these cases are treated in EDs.2 These numbers have not gone unnoticed in the consumer press. Over the past 18 months, Sports Illustrated, Newsweek, and Time3-5 have published stories on sports-related concussion, helping to raise public awareness of its risks.

Recommendations for practitioners have changed. In 1997, the American Academy of Neurology6 published one-size-fits-all guidelines on managing concussion, using levels of symptomatology and loss of consciousness to grade the severity of concussion from 1 to 3. These guidelines were similar to the Cantu and Colorado guidelines of the early 1990s.7,8 Since then, however, the diagnostic criteria and expert opinion about treatment and return to physical activity have changed. Indeed, several medical organizations9-12 now recommend a more individualized approach to evaluation and management, which we describe here.

It begins with a definition

While there is no single agreed-upon characterization of “concussion,” the 3rd International Conference on Concussion in Sport (ICCS)12 provides this definition:

Concussion is defined as a complex pathophysiological process affecting the brain, induced by traumatic biomechanical forces. Several common features that incorporate clinical, pathologic, and biomechanical injury constructs that may be utilized in defining the nature of a concussive head injury include:

  1. Concussion may be caused either by a direct blow to the head, face, or neck or a blow elsewhere on the body with an ‘‘impulsive’’ force transmitted to the head.
  2. Concussion typically results in the rapid onset of short-lived impairment of neurologic function that resolves spontaneously.
  3. Concussion may result in neuropathological changes but the acute clinical symptoms largely reflect a functional disturbance rather than a structural injury.
  4. Concussion results in a graded set of clinical symptoms that may or may not involve loss of consciousness. Resolution of the clinical and cognitive symptoms typically follows a sequential course.… In a small percentage of cases, however, postconcussive symptoms may be prolonged.
  5. No abnormality on standard structural neuroimaging studies is seen in concussion.

Office evaluation
Obtain a thorough history and conduct a neurologic evaluation and musculoskeletal examination of the head and neck.

Clues to expected length of recovery

A patient with a concussion may lose consciousness after the impact, or have a brief convulsion that is not a seizure.13 In the periodimmediately after the injury, the patient may exhibit a constellation of such signs and symptoms as headache, confusion, a dazed look, dilated pupils, amnesia, poor balance, nausea, or vomiting. These features typically resolve over time, but may persist for weeks or months. Anterograde or retrograde amnesia may also occur. TABLE 1 details a more complete list of concussion symptoms. If the patient is a child or young adult, it is useful to have a parent present at the office visit to describe the patient’s mood, sleep, appetite, and overall health after the injury.

Factors that may portend a longer recovery include a previous concussion, retrograde or anterograde amnesia, younger age, and female sex.14

Dire problems beyond concussion. Complaints or historical elements inconsistent with concussion that should be considered red flags include any focal neurologic complaints, vomiting or headache that worsens after a period of improvement, or obtundation or disorientation that has worsened since the injury. With such findings, consider more serious head injuries and arrange for a more complete immediate neurologic work-up.

 

 

CASE Our neurologic examination yielded normal results. However, our patient was unable to balance correctly on one leg. The cognitive exam revealed a deficit in short-term memory. We diagnosed a concussion, advised her to refrain from sports, and prescribed cognitive rest. A return to school for half days would be considered once her symptoms began to resolve.

TABLE 1
Signs and symptoms commonly associated with concussion

Headache
“Pressure in head”
Neck pain
Nausea or vomiting
Dizziness
Blurred vision
Balance problems
Sensitivity to light
Sensitivity to noise
Feeling slowed down
Feeling like “in a fog”
“Don’t feel right”
Difficulty concentrating
Difficulty remembering
Fatigue or low energy
Confusion
Drowsiness
Trouble falling asleep
Irritability
Sadness
Nervousness or anxiety
Adapted from SCAT2 in Appendix 1 of: McCrory P, Meeuwisse W, Johnston K, et al. Br J Sports Med. 2009;43(suppl 1):i76-i90.12

Options for the neurologic exam
With a simple concussion, expect a normal neurologic examination, with the possible exception of the ability to balance. Head imaging is not necessary in the setting of suspected concussion, because results of computed tomography (CT) and magnetic resonance imaging (MRI) will likely be normal.12

Balance testing can assist in the diagnosis of concussion and the monitoring of recovery from injury.15-17 The Balance Error Scoring System (BESS)15 is a validated and simple test that can be done in the office. The test involves 3 consecutive stances: (a) normal stance with feet comfortably apart and hands on hips, (b) with feet aligned heel to toe with the dominant leg in front, and (c) standing on the nondominant leg with the dominant leg flexed 30 degrees at the hip. Have the patient repeat each version of the test for 20 seconds with eyes closed, on a stable and then unstable surface (eg, foam mat).

It’s recommended that another staff member be present to spot the patient in case of a fall. A link to a complete description of the test and scoring details is provided in the Web resources box.

Assess cognitive function. One tool for assessing cognitive function is the Sports Concussion Assessment Tool 2 (SCAT2).12 SCAT2 includes newer, as yet unvalidated sections and several sections that have been independently studied and proven useful in diagnosing concussion. Validated sections are the Maddocks questions, used only at the time and place of injury18 ; the modified BESS15 ; and the Standardized Assessment of Concussion (SAC).19 The SCAT2 and the SAC (which may be used separately) include questions that assist in evaluating short-term memory and attention, and are useful in the physician’s office.

Do computer-based tools help? Another option for cognitive assessment is computer-based neuropsychologic testing developed specifically for use with suspected concussion. Any of these programs can be used in the office by a trained practitioner. Schools may also use the programs under the supervision of an athletic trainer or team physician. Available programs are ImPACT, developed by the University of Pittsburgh (http://impacttest.com); the Cognitive Stability Index (CSI), by HeadMinder (http://www.headminder.com/site/csi/home.html); and the Computerized Cognitive Assessment Tool (CCAT), by CogState/Axon Sports (http://www.axonsports.com). Multiple studies have shown such programs to be useful in diagnosing and monitoring recovery from sports concussion.20-23

However, among sports medicine practitioners, there seems to be a consensus that computer-based neuropsychologic testing is most useful when a baseline score exists. Baseline testing is usually done preseason on athletes in a healthy state. If a baseline score is not available, a patient’s postinjury score is compared with normative data produced by the developer of the individual test.

Few, if any, outcome studies have been conducted to determine whether computer-basedneuropsychologic testing provides any meaningful improvement in the care of athletes who have suffered concussions. There is also concern that few studies by independent sources have replicated the data disseminated by developers of the tests.24,25 The most recent guidelines by the 3rd ICCS recommend using neuropsychologic testing only as an aid to an overall medical evaluation, not as the sole determinant of recovery from concussion.12 Numerous studies now underway may help clarify the role of neuropsychologic testing in concussion.

CASE By the time of our follow-up exam 7 days later (11 days from injury), KD had returned to school for half days, but her phonophobia and headaches worsened at school and she had difficulty focusing on academic tasks. Neurologic, balance, and cognitive exams were all normal. We advised her to gradually return to school full time while abstaining from sporting activity.

 

 

At 16 days’ follow-up (20 days from injury), KD had returned to school full time and said she felt more like herself, although she continued to have daily headaches and phonophobia. All exam results were normal. Sports were still off limits, and we told her to expect at least 7 more days of respite before any return to exercise would be allowed.

At 23 days’ follow-up (27 days from injury), KD’s symptoms had completely resolved, and all exam results were normal. We prescribed a stepwise return to athletic activity over the next 10 days and discussed this plan with the school’s athletic trainer, who would supervise her return to play.

Web resources

American Academy of Neurology (AAN). Position Statement on Sports Concussion. http://www.aan.com/globals/axon/assets/7913.pdf

American Academy of Pediatrics (AAP). Sports-Related Concussion in Children and Adolescents. http://pediatrics.aappublications.org/cgi/content/abstract/126/3/597

The Balance Error Scoring System (BESS). http://www.sportsconcussion.com/pdf/management/BESSProtocolNATA09.pdf

Centers for Disease Control and Prevention. Concussion and Mild TBI. http://www.cdc.gov/concussion/index.html

Sport Concussion Assessment Tool 2 (SCAT2). http://www.athletictherapy.org/en/pdf/SCAT2.pdf

3rd International Conference on Concussion in Sport. http://bjsm.bmj.com/content/43/Suppl_1/i76.full

Individualize management

The one-size-fits-all approach previously recommended6 is no longer the standard of care. In your initial encounter with the patient (and parents, as appropriate), explain the nature of the injury, expected course of recovery, and requirements for a return to play. Also discuss the possibility of postconcussive syndrome and the risk of rare sequelae such as second impact syndrome.

If the patient is symptomatic or exhibits examination findings consistent with concussion, recommend immediate cessation of sports activity.9-12 With a school-aged athlete, if symptoms reported by the patient or parents are significant, consider prescribing cognitive rest, which can be provided through quiet accommodations at school or perhaps even time off from school or exams.12,24 In the early period of recovery, increased cognitive or physical activity can cause symptoms to worsen. With improvement, the patient may return to school half time to lessen the chance of a significant return of symptoms. If half days are tolerated, the patient may transition to full days. Make sure the diagnosis and expectations for recovery are communicated to the appropriate school officials so that necessary accommodationscan be made. If symptoms after the initial office visit are mild, a one-week return to school is appropriate to evaluate the patient’s recovery.

Allowing a return to sports. Once the patient is asymptomatic, and physical and cognitive test results are normal, discuss a return-to-play protocol with the patient (and with parents and athletic trainer or coach, as appropriate). Multiple sources10,11,26 now recommend a stepwise return to play, as detailed by the 3rd ICCS ( TABLE 2 ).12 Increase or decrease the length of the protocol depending on the patient and the specifics of the case.

There is little science to guide the treatment of children with concussion. However, given that their brains are still developing, it’s prudent to be more conservative than with older adolescents or adults. Multiple sources apart from the 3rd ICCS agree with this recommendation. Several authors suggest more cognitive rest and a longer return-to-play protocol in all cases.10,27 In fact, the ICCS committee additionally recommends observing a symptom-free waiting period for pediatric athletes before even starting a return-to-play protocol.

McCrory et al26 suggest that children under age 15 be treated more conservatively than those 15 and older. They suggest treating those 15 and older with the protocol for older adolescents. Specifying an age at which one should always make a decision for or against conservative care can be problematic. However, based on the recommendations above, it would seem reasonable to provide conservative treatment for children younger than high school age and perhaps even those in the early years of high school.

Consider legal implications. Become familiar with state laws that require certain steps in managing sports concussion. The Web site http://www.sportsconcussions.org/laws.html28 lists states with sports concussion statutes, as well as states with bills working their way through the legislative system. Currently, 29 states are listed with laws; 14 more and the District of Columbia have pending legislation.

TABLE 2
Stepwise protocol for return to play

If symptoms recur at any step, have patient return to prior level
1. Light aerobic activityWalking, swimming, exercise bike; keeping exertion <70% of maximum heart rate
2. Sport-specific exercisesExertional drills in sport, eg, running drills in football/soccer, skating drills in hockey
3. Noncontact training drillsProgression to more complex noncontact drills, eg, passing/catching drills in football, shooting/passing in basketball, hitting drills in volleyball
4. Full-contact practiceReturn to full practice if no recurrence of symptoms through first 3 steps and cleared by physician
5. Game activityReturn to full sport participation if no recurrence of symptoms with above steps
Adapted from: McCrory P, Meeuwisse W, Johnston K, et al. Br J Sports Med. 2009;43(suppl 1):i76-i90.12
 

 

Anticipate complications
Most patients with concussions who are managed appropriately do well. However, complications can occur. The most serious complication is second impact syndrome, which usually occurs when concussion is unrecognized or not well managed. While not well understood, this condition is thought to result from a sudden increase in intracranial pressure after a second head injury in an athlete already suffering from concussion symptoms. The injury typically results in serious long-term neurologic deficits, or even fatality.29 Second impact syndrome has been documented as occurring in the same game after an initial injury, as well as in subsequent games.29

A more common, but less serious, complication is postconcussion syndrome.30 This is an ill-defined condition in which the patient suffers from concussive symptoms for an extended period of time, generally for more than 3 months.30 As with acute concussion, the constellation of symptoms ranges from headache to cognitive impairment. In cases of postconcussion syndrome, it is appropriate to consult with neuropsychologists, psychiatrists, or neurologists for assistance with symptoms and associated mood disorders. Similar to acute concussion management, it is generally recommended that athletes not be cleared to resume play while struggling with the symptoms of postconcussion syndrome.30

There have also been recent reports of late-life sequelae in those who have sustained multiple concussions. Depression and dementia have been suggested in surveys of retired NFL players.31,32 There have also been studies both suggesting14 and questioning33,34 whether multiple concussions result in long-term cognitive deficits. While the evidence available at this time is not firm, there seems to be an increasing belief that multiple concussions can affect long-term cognitive abilities. For these reasons, use caution in making return-to-play decisions for patients with multiple concussions or concussions with long-lasting symptoms.

CORRESPONDENCE Aaron M. Lear, MD, 224 West Exchange Street, Suite 440, Akron, OH 44302; [email protected]

References

1. CDC. Sports-related recurrent brain injuries—United States. MMWR Morb Mortal Wkly Rep. 1997;46:224-227.

2. CDC. Brain injury awareness month—March 2010. MMWR Morb Mortal Wkly Rep. 2010;59:235.-

3. Epstein D. The damage done. Sports Illustrated. November 1, 2010:42. Available at: http://sportsillustrated.cnn.com/vault/article/magazine/MAG1176377/index.htm. Accessed May 16, 2012.

4. Kliff S. Heading off sports injuries. Newsweek. February 4, 2010. Available at: http://www.newsweek.com/2010/02/04/heading-off-sports-injuries.html. Accessed February 9, 2011.

5. Kluger J. Headbanger nation. Health special: kids and concussions. Time. February 3, 2011. Available at: http://www.time.com/time/specials/packages/article/0,28804,2043395_2043506_2043494,00.html. Accessed February 9, 2011.

6. American Academy of Neurology. Practice parameter: the management of concussion in sports (summary statement). Report of the quality standards subcommittee. Neurology. 1997;48:581-585.

7. Cantu R. Cerebral concussion in sport. Management and prevention. Sports Med. 1992;14:64-74.

8. Kelly J, Nichols J, Filley C, et al. Concussion in sports. Guidelines for the prevention of catastrophic outcome. JAMA. 1991;266:2867-2869.

9. American Academy of Neurology. Position statement on sports concussion. October 2010. AAN policy 2010-36. Available at: http://www.aan.com/globals/axon/assets/7913.pdf. Accessed February 23, 2011.

10. Halstead M, Walter K. Council on Sports Medicine and Fitness. American Academy of Pediatrics. Clinical report—sport-related concussion in children and adolescents. Pediatrics. 2010;126:597-615.

11. Herring SA, Cantu RC, Guskiewicz KM, et al. Concussion (mild traumatic brain injury) and the team physician: a consensus statement—2011 update. Med Sci Sports Exerc. 2011;43:2412-2422.Available at: http://journals.lww.com/acsm-msse/Fulltext/2011/12000/Concussion__Mild_Traumatic_Brain_Injury__and_the.24.aspx. Accessed February 23, 2011.

12. McCrory P, Meeuwisse W, Johnston K, et al. Consensus statement on concussion in sport: the 3rd International Conference on Concussion in Sport held in Zurich, November 2008. Br J Sports Med. 2009;43(suppl 1):i76-i90.

13. Ropper A, Gorson K. Clinical practice. Concussion. N Engl J Med. 2007;356:166-172.

14. Reddy C, Collins MW. Sports concussion: management and predictors of outcome. Curr Sports Med Rep. 2009;8:10-15.

15. Guskiewicz KM. Assessment of postural stability following sport-related concussion. Curr Sports Med Rep. 2003;2:24-30.

16. Broglio S, Sosnoff J, Ferrara M. The relationship of athlete-reported concussion symptoms and objective measures of neurocognitive function and postural control. Clin J Sport Med. 2009;19:377-382.

17. Reimann B, Guskiewicz K. Effects of mild head injury on postural stability as measured through clinical balance testing. J Athl Train. 2000;35:19-25.

18. Maddocks D, Dicker G, Saling M. The assessment of orientation following concussion in athletes. Clin J Sport Med. 1995;5:32-35.

19. McCrea M. Standardized mental status assessment of sports concussion. Clin J Sport Med. 2001;11:176-181.

20. Collie A, Maruff P, Makdissi M, et al. CogSport: reliability and correlation with conventional cognitive tests used in postconcussion medical evaluations. Clin J Sport Med. 2003;13:28-32.

21. Erlanger D, Saliba E, Barth J, et al. Monitoring resolution of postconcussion symptoms in athletes: preliminary results of a web-based neuropsychological test protocol. J Athl Train. 2001;36:280-287.

22. Schatz P, Pardini J, Lovell M, et al. Sensitivity and specificity of the ImPACT Test battery for concussion in athletes. Arch Clin Neuropsychol. 2006;21:91-99.

23. Schatz P, Putz B. Cross-validation of measures used for computer-based assessment of concussion. Appl Neuropsychol. 2006;13:151-159.

24. Kirkwood M, Randolph C, Yeates K. Returning pediatric athletes to play after concussion: the evidence (or lack thereof) behind baseline neuropsychological testing. Acta Pædiatr. 2009;98:1409-1411.

25. Randolph C. Baseline neuropsychological testing in managing sport-related concussion: does it modify risk? Curr Sports Med Rep. 2011;10:21-26.

26. McCrory P, Collie A, Anderson V, et al. Can we manage sport related concussion in children the same as in adults? Br J Sports Med. 2004;38:516-519.

27. d’Hemecourt P. Subacute symptoms of sports-related concussion outpatient management and return to play. Clin Sports Med. 2011;30:63-72.

28. Concussion laws. Available at: http://www.sportsconcussions.org/laws.html. Accessed July 5, 2011.

29. Wetjen N, Pichelmann M, Atkinson J. Second impact syndrome: concussion and second injury brain complications. J Am Coll Surg. 2010;211:553-557.

30. Jotwani V, Harmon KG. Postconcussion syndrome in athletes. Curr Sports Med Rep. 2010;9:21-26.

31. Guskiewicz K, Marshall S, Bailes J, et al. Association between recurrent concussion and late-life cognitive impairment in retired professional football players. Neurosurgery. 2005;57:719-726.

32. Guskiewicz K, Marshall S, Bailes J, et al. Recurrent concussion and risk of depression in retired professional football players. Med Sci Sports Exerc. 2007;39:903-909.

33. Belanger H, Spiegel E, Vanderploeg R. Neuropsychological performance following a history of multiple self-reported concussions: a meta-analysis. J Int Neuropsychol Soc. 2010;16:262-267.

34. Burce J, Echemendia R. History of multiple self-reported concussions is not associated with reduced cognitive abilities. Neurosurgery. 2009;64:100-106.

References

1. CDC. Sports-related recurrent brain injuries—United States. MMWR Morb Mortal Wkly Rep. 1997;46:224-227.

2. CDC. Brain injury awareness month—March 2010. MMWR Morb Mortal Wkly Rep. 2010;59:235.-

3. Epstein D. The damage done. Sports Illustrated. November 1, 2010:42. Available at: http://sportsillustrated.cnn.com/vault/article/magazine/MAG1176377/index.htm. Accessed May 16, 2012.

4. Kliff S. Heading off sports injuries. Newsweek. February 4, 2010. Available at: http://www.newsweek.com/2010/02/04/heading-off-sports-injuries.html. Accessed February 9, 2011.

5. Kluger J. Headbanger nation. Health special: kids and concussions. Time. February 3, 2011. Available at: http://www.time.com/time/specials/packages/article/0,28804,2043395_2043506_2043494,00.html. Accessed February 9, 2011.

6. American Academy of Neurology. Practice parameter: the management of concussion in sports (summary statement). Report of the quality standards subcommittee. Neurology. 1997;48:581-585.

7. Cantu R. Cerebral concussion in sport. Management and prevention. Sports Med. 1992;14:64-74.

8. Kelly J, Nichols J, Filley C, et al. Concussion in sports. Guidelines for the prevention of catastrophic outcome. JAMA. 1991;266:2867-2869.

9. American Academy of Neurology. Position statement on sports concussion. October 2010. AAN policy 2010-36. Available at: http://www.aan.com/globals/axon/assets/7913.pdf. Accessed February 23, 2011.

10. Halstead M, Walter K. Council on Sports Medicine and Fitness. American Academy of Pediatrics. Clinical report—sport-related concussion in children and adolescents. Pediatrics. 2010;126:597-615.

11. Herring SA, Cantu RC, Guskiewicz KM, et al. Concussion (mild traumatic brain injury) and the team physician: a consensus statement—2011 update. Med Sci Sports Exerc. 2011;43:2412-2422.Available at: http://journals.lww.com/acsm-msse/Fulltext/2011/12000/Concussion__Mild_Traumatic_Brain_Injury__and_the.24.aspx. Accessed February 23, 2011.

12. McCrory P, Meeuwisse W, Johnston K, et al. Consensus statement on concussion in sport: the 3rd International Conference on Concussion in Sport held in Zurich, November 2008. Br J Sports Med. 2009;43(suppl 1):i76-i90.

13. Ropper A, Gorson K. Clinical practice. Concussion. N Engl J Med. 2007;356:166-172.

14. Reddy C, Collins MW. Sports concussion: management and predictors of outcome. Curr Sports Med Rep. 2009;8:10-15.

15. Guskiewicz KM. Assessment of postural stability following sport-related concussion. Curr Sports Med Rep. 2003;2:24-30.

16. Broglio S, Sosnoff J, Ferrara M. The relationship of athlete-reported concussion symptoms and objective measures of neurocognitive function and postural control. Clin J Sport Med. 2009;19:377-382.

17. Reimann B, Guskiewicz K. Effects of mild head injury on postural stability as measured through clinical balance testing. J Athl Train. 2000;35:19-25.

18. Maddocks D, Dicker G, Saling M. The assessment of orientation following concussion in athletes. Clin J Sport Med. 1995;5:32-35.

19. McCrea M. Standardized mental status assessment of sports concussion. Clin J Sport Med. 2001;11:176-181.

20. Collie A, Maruff P, Makdissi M, et al. CogSport: reliability and correlation with conventional cognitive tests used in postconcussion medical evaluations. Clin J Sport Med. 2003;13:28-32.

21. Erlanger D, Saliba E, Barth J, et al. Monitoring resolution of postconcussion symptoms in athletes: preliminary results of a web-based neuropsychological test protocol. J Athl Train. 2001;36:280-287.

22. Schatz P, Pardini J, Lovell M, et al. Sensitivity and specificity of the ImPACT Test battery for concussion in athletes. Arch Clin Neuropsychol. 2006;21:91-99.

23. Schatz P, Putz B. Cross-validation of measures used for computer-based assessment of concussion. Appl Neuropsychol. 2006;13:151-159.

24. Kirkwood M, Randolph C, Yeates K. Returning pediatric athletes to play after concussion: the evidence (or lack thereof) behind baseline neuropsychological testing. Acta Pædiatr. 2009;98:1409-1411.

25. Randolph C. Baseline neuropsychological testing in managing sport-related concussion: does it modify risk? Curr Sports Med Rep. 2011;10:21-26.

26. McCrory P, Collie A, Anderson V, et al. Can we manage sport related concussion in children the same as in adults? Br J Sports Med. 2004;38:516-519.

27. d’Hemecourt P. Subacute symptoms of sports-related concussion outpatient management and return to play. Clin Sports Med. 2011;30:63-72.

28. Concussion laws. Available at: http://www.sportsconcussions.org/laws.html. Accessed July 5, 2011.

29. Wetjen N, Pichelmann M, Atkinson J. Second impact syndrome: concussion and second injury brain complications. J Am Coll Surg. 2010;211:553-557.

30. Jotwani V, Harmon KG. Postconcussion syndrome in athletes. Curr Sports Med Rep. 2010;9:21-26.

31. Guskiewicz K, Marshall S, Bailes J, et al. Association between recurrent concussion and late-life cognitive impairment in retired professional football players. Neurosurgery. 2005;57:719-726.

32. Guskiewicz K, Marshall S, Bailes J, et al. Recurrent concussion and risk of depression in retired professional football players. Med Sci Sports Exerc. 2007;39:903-909.

33. Belanger H, Spiegel E, Vanderploeg R. Neuropsychological performance following a history of multiple self-reported concussions: a meta-analysis. J Int Neuropsychol Soc. 2010;16:262-267.

34. Burce J, Echemendia R. History of multiple self-reported concussions is not associated with reduced cognitive abilities. Neurosurgery. 2009;64:100-106.

Issue
The Journal of Family Practice - 61(6)
Issue
The Journal of Family Practice - 61(6)
Page Number
323-328
Page Number
323-328
Publications
Publications
Topics
Article Type
Display Headline
Sports concussion: A return-to-play guide
Display Headline
Sports concussion: A return-to-play guide
Legacy Keywords
Aaron M. Lear ;MD;CAQ; Minh-Ha Hoang ;DO;sports concussion; evaluative methods; return-to-play; concussive symptoms; balance; sports participation; complex pathophysiological process; direct blow; impulsive force
Legacy Keywords
Aaron M. Lear ;MD;CAQ; Minh-Ha Hoang ;DO;sports concussion; evaluative methods; return-to-play; concussive symptoms; balance; sports participation; complex pathophysiological process; direct blow; impulsive force
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media

Community Oncology Podcast - Erwinia asparaginase for acute lymphoblastic leukemia

Article Type
Changed
Fri, 01/04/2019 - 11:05
Display Headline
Community Oncology Podcast - Erwinia asparaginase for acute lymphoblastic leukemia

Join Community Oncology Editor-in Chief Dr David H. Henry for an audio tour of the May issue, featuring "Erwinia asparaginase for acute lymphoblastic leukemia in children with hypersensitivity to E. coli-derived asparaginase" and "Creating a community-based, patient-centered cancer survivorship program."

Author and Disclosure Information

 

 

Audio / Podcast
Publications
Topics
Legacy Keywords
Dr David H. Henry, "Erwinia asparaginase, acute lymphoblastic leukemia, hypersensitivity to E. coli-derived asparaginase, patient-centered cancer survivorship program
Audio / Podcast
Audio / Podcast
Author and Disclosure Information

 

 

Author and Disclosure Information

 

 

Join Community Oncology Editor-in Chief Dr David H. Henry for an audio tour of the May issue, featuring "Erwinia asparaginase for acute lymphoblastic leukemia in children with hypersensitivity to E. coli-derived asparaginase" and "Creating a community-based, patient-centered cancer survivorship program."

Join Community Oncology Editor-in Chief Dr David H. Henry for an audio tour of the May issue, featuring "Erwinia asparaginase for acute lymphoblastic leukemia in children with hypersensitivity to E. coli-derived asparaginase" and "Creating a community-based, patient-centered cancer survivorship program."

Publications
Publications
Topics
Article Type
Display Headline
Community Oncology Podcast - Erwinia asparaginase for acute lymphoblastic leukemia
Display Headline
Community Oncology Podcast - Erwinia asparaginase for acute lymphoblastic leukemia
Legacy Keywords
Dr David H. Henry, "Erwinia asparaginase, acute lymphoblastic leukemia, hypersensitivity to E. coli-derived asparaginase, patient-centered cancer survivorship program
Legacy Keywords
Dr David H. Henry, "Erwinia asparaginase, acute lymphoblastic leukemia, hypersensitivity to E. coli-derived asparaginase, patient-centered cancer survivorship program
Disallow All Ads
Alternative CME

Statins Appear Safe, Even Beneficial, in Cirrhosis

Article Type
Changed
Fri, 01/18/2019 - 11:59
Display Headline
Statins Appear Safe, Even Beneficial, in Cirrhosis

SAN DIEGO – Statin therapy is not only safe for patients with cirrhosis, it may slow the progression of their liver disease to hepatic decompensation and help them live longer, a study has shown.

The findings, reported at the annual Digestive Disease Week, should help allay fears that decreased hepatic clearance of statins could lead to complications in patients with advanced liver disease, as was previously hypothesized, said lead investigator Dr. Sonal Kumar.

"In fact, it seems the opposite may be true," said Dr. Kumar of Brigham and Women’s Hospital, Boston, referring to the results of her retrospective study in which statin therapy in cirrhosis patients was associated with a decreased risk of hepatic decompensation, a delay in the time to decompensation, and reduced all-cause mortality, compared with cirrhosis patients not taking statins.

The investigators were aware that some clinicians either do not initiate statin therapy or discontinue statins in patients with advanced liver disease because of perceived safety concerns. Dr. Kumar and her colleagues sought to determine the actual effect of statin therapy on the risk of hepatic decomposition in cirrhosis.

They identified all patients with biopsy-proven cirrhosis who had taken statins for a minimum of 3 months for treatment of dyslipidemia. A control population of cirrhosis patients not on statins was matched 2:1 for age, gender, and Child-Pugh class from the Partners HealthCare System Research Patient Data Registry, which includes demographic and diagnostic information on patients treated at Massachusetts General Hospital and Brigham and Women’s Hospital.

The primary outcomes of the study were hepatic decompensation, defined as the development of ascites, jaundice (bilirubin greater than 2.5 mg/dL), hepatic encephalopathy, or variceal hemorrhage, and time to decompensation. Mortality was a secondary outcome, Dr. Kumar explained.

The investigators created a Cox proportional hazards model for decompensation to control for age, Child-Pugh class, diabetes, coronary artery disease, and hepatocellular carcinoma, and they used conditional logistic regression to assess mortality, she said.

Of 243 cirrhosis patients included in the analysis, 81 were statin users and 162 were matched controls. "In each group, approximately 70% of patients were Child-Pugh A and 30% were Child-Pugh B/C, and the MELD [Model for End-Stage Liver Disease] score, albumin, presence of varices, and beta-blocker use were similar between groups," Dr. Kumar noted. In the statin group, which was followed for a mean of 1,756 days, decompensation was reported in 31 patients (38.2%), compared with 80 patients (50.62%) in the control group.

The control patients were followed for a mean of 1,503 days, and on Cox analysis, "statin therapy was the only factor significantly associated with lower decompensation risk, with a hazard ratio of 0.46." Additionally, Kaplan-Meier curves showed a significantly longer time to decompensation in patients receiving statin therapy, she said. In subgroup analyses, significantly longer time to decompensation was observed in Child-Pugh A and Child-Pugh B/C patients.

Overall mortality was significantly lower in the statin group, at 37.0%, than in the control group, at 50.6%, said Dr. Kumar. Statin use remained significantly associated with decreased mortality in multivariate analysis, with an odds ratio of 0.36, while coronary artery disease and hepatocellular carcinoma were associated with increased mortality, with respective odds ratios of 3.6 and 4.9.

There were no statistically significant differences in cause of death between the two groups, "however it is important to note that cause of death was not documented for approximately one-third of the study patients, so we cannot make definitive statements about whether patients on statins were less likely to die of liver-related or cardiovascular causes than patients in the control group," said Dr. Kumar.

The apparent hepatoprotective effect of statin therapy in cirrhosis patients may be a function of previously observed hemodynamic and molecular effects of statins, Dr. Kumar hypothesized. Sinusoidal endothelial dysfunction with decreased nitric oxide production contributes to increased hepatic resistance in cirrhosis, she explained. Just as statins improve endothelial dysfunction in the peripheral vasculature, they may also improve the vascular disturbances that contribute to portal hypertension in cirrhosis by selectively increasing nitric oxide availability in the liver, thus reducing pressures, she said.

The retrospective design of the study limits the conclusions that can be taken from it. Specifically, "we can’t say that all patients with liver disease should be prescribed statins," Dr. Kumar said. "What we can say is that statin therapy is safe in this population, it may be beneficial for its effects on the liver as well as the cardiovascular system, and clinicians should not hesitate to prescribe it for appropriate cardiovascular indications in cirrhosis patients." The findings also indicate that prospective studies are warranted to clarify the role of statins in advanced liver disease, she stressed.

 

 

Dr. Kumar disclosed no relevant financial conflicts of interest.

Meeting/Event
Author and Disclosure Information

Publications
Topics
Legacy Keywords
statin therapy, statins cirrhosis, statins liver disease, hepatic decompensation, Digestive Disease Week 2012, Dr. Sonal Kumar
Sections
Author and Disclosure Information

Author and Disclosure Information

Meeting/Event
Meeting/Event

SAN DIEGO – Statin therapy is not only safe for patients with cirrhosis, it may slow the progression of their liver disease to hepatic decompensation and help them live longer, a study has shown.

The findings, reported at the annual Digestive Disease Week, should help allay fears that decreased hepatic clearance of statins could lead to complications in patients with advanced liver disease, as was previously hypothesized, said lead investigator Dr. Sonal Kumar.

"In fact, it seems the opposite may be true," said Dr. Kumar of Brigham and Women’s Hospital, Boston, referring to the results of her retrospective study in which statin therapy in cirrhosis patients was associated with a decreased risk of hepatic decompensation, a delay in the time to decompensation, and reduced all-cause mortality, compared with cirrhosis patients not taking statins.

The investigators were aware that some clinicians either do not initiate statin therapy or discontinue statins in patients with advanced liver disease because of perceived safety concerns. Dr. Kumar and her colleagues sought to determine the actual effect of statin therapy on the risk of hepatic decomposition in cirrhosis.

They identified all patients with biopsy-proven cirrhosis who had taken statins for a minimum of 3 months for treatment of dyslipidemia. A control population of cirrhosis patients not on statins was matched 2:1 for age, gender, and Child-Pugh class from the Partners HealthCare System Research Patient Data Registry, which includes demographic and diagnostic information on patients treated at Massachusetts General Hospital and Brigham and Women’s Hospital.

The primary outcomes of the study were hepatic decompensation, defined as the development of ascites, jaundice (bilirubin greater than 2.5 mg/dL), hepatic encephalopathy, or variceal hemorrhage, and time to decompensation. Mortality was a secondary outcome, Dr. Kumar explained.

The investigators created a Cox proportional hazards model for decompensation to control for age, Child-Pugh class, diabetes, coronary artery disease, and hepatocellular carcinoma, and they used conditional logistic regression to assess mortality, she said.

Of 243 cirrhosis patients included in the analysis, 81 were statin users and 162 were matched controls. "In each group, approximately 70% of patients were Child-Pugh A and 30% were Child-Pugh B/C, and the MELD [Model for End-Stage Liver Disease] score, albumin, presence of varices, and beta-blocker use were similar between groups," Dr. Kumar noted. In the statin group, which was followed for a mean of 1,756 days, decompensation was reported in 31 patients (38.2%), compared with 80 patients (50.62%) in the control group.

The control patients were followed for a mean of 1,503 days, and on Cox analysis, "statin therapy was the only factor significantly associated with lower decompensation risk, with a hazard ratio of 0.46." Additionally, Kaplan-Meier curves showed a significantly longer time to decompensation in patients receiving statin therapy, she said. In subgroup analyses, significantly longer time to decompensation was observed in Child-Pugh A and Child-Pugh B/C patients.

Overall mortality was significantly lower in the statin group, at 37.0%, than in the control group, at 50.6%, said Dr. Kumar. Statin use remained significantly associated with decreased mortality in multivariate analysis, with an odds ratio of 0.36, while coronary artery disease and hepatocellular carcinoma were associated with increased mortality, with respective odds ratios of 3.6 and 4.9.

There were no statistically significant differences in cause of death between the two groups, "however it is important to note that cause of death was not documented for approximately one-third of the study patients, so we cannot make definitive statements about whether patients on statins were less likely to die of liver-related or cardiovascular causes than patients in the control group," said Dr. Kumar.

The apparent hepatoprotective effect of statin therapy in cirrhosis patients may be a function of previously observed hemodynamic and molecular effects of statins, Dr. Kumar hypothesized. Sinusoidal endothelial dysfunction with decreased nitric oxide production contributes to increased hepatic resistance in cirrhosis, she explained. Just as statins improve endothelial dysfunction in the peripheral vasculature, they may also improve the vascular disturbances that contribute to portal hypertension in cirrhosis by selectively increasing nitric oxide availability in the liver, thus reducing pressures, she said.

The retrospective design of the study limits the conclusions that can be taken from it. Specifically, "we can’t say that all patients with liver disease should be prescribed statins," Dr. Kumar said. "What we can say is that statin therapy is safe in this population, it may be beneficial for its effects on the liver as well as the cardiovascular system, and clinicians should not hesitate to prescribe it for appropriate cardiovascular indications in cirrhosis patients." The findings also indicate that prospective studies are warranted to clarify the role of statins in advanced liver disease, she stressed.

 

 

Dr. Kumar disclosed no relevant financial conflicts of interest.

SAN DIEGO – Statin therapy is not only safe for patients with cirrhosis, it may slow the progression of their liver disease to hepatic decompensation and help them live longer, a study has shown.

The findings, reported at the annual Digestive Disease Week, should help allay fears that decreased hepatic clearance of statins could lead to complications in patients with advanced liver disease, as was previously hypothesized, said lead investigator Dr. Sonal Kumar.

"In fact, it seems the opposite may be true," said Dr. Kumar of Brigham and Women’s Hospital, Boston, referring to the results of her retrospective study in which statin therapy in cirrhosis patients was associated with a decreased risk of hepatic decompensation, a delay in the time to decompensation, and reduced all-cause mortality, compared with cirrhosis patients not taking statins.

The investigators were aware that some clinicians either do not initiate statin therapy or discontinue statins in patients with advanced liver disease because of perceived safety concerns. Dr. Kumar and her colleagues sought to determine the actual effect of statin therapy on the risk of hepatic decomposition in cirrhosis.

They identified all patients with biopsy-proven cirrhosis who had taken statins for a minimum of 3 months for treatment of dyslipidemia. A control population of cirrhosis patients not on statins was matched 2:1 for age, gender, and Child-Pugh class from the Partners HealthCare System Research Patient Data Registry, which includes demographic and diagnostic information on patients treated at Massachusetts General Hospital and Brigham and Women’s Hospital.

The primary outcomes of the study were hepatic decompensation, defined as the development of ascites, jaundice (bilirubin greater than 2.5 mg/dL), hepatic encephalopathy, or variceal hemorrhage, and time to decompensation. Mortality was a secondary outcome, Dr. Kumar explained.

The investigators created a Cox proportional hazards model for decompensation to control for age, Child-Pugh class, diabetes, coronary artery disease, and hepatocellular carcinoma, and they used conditional logistic regression to assess mortality, she said.

Of 243 cirrhosis patients included in the analysis, 81 were statin users and 162 were matched controls. "In each group, approximately 70% of patients were Child-Pugh A and 30% were Child-Pugh B/C, and the MELD [Model for End-Stage Liver Disease] score, albumin, presence of varices, and beta-blocker use were similar between groups," Dr. Kumar noted. In the statin group, which was followed for a mean of 1,756 days, decompensation was reported in 31 patients (38.2%), compared with 80 patients (50.62%) in the control group.

The control patients were followed for a mean of 1,503 days, and on Cox analysis, "statin therapy was the only factor significantly associated with lower decompensation risk, with a hazard ratio of 0.46." Additionally, Kaplan-Meier curves showed a significantly longer time to decompensation in patients receiving statin therapy, she said. In subgroup analyses, significantly longer time to decompensation was observed in Child-Pugh A and Child-Pugh B/C patients.

Overall mortality was significantly lower in the statin group, at 37.0%, than in the control group, at 50.6%, said Dr. Kumar. Statin use remained significantly associated with decreased mortality in multivariate analysis, with an odds ratio of 0.36, while coronary artery disease and hepatocellular carcinoma were associated with increased mortality, with respective odds ratios of 3.6 and 4.9.

There were no statistically significant differences in cause of death between the two groups, "however it is important to note that cause of death was not documented for approximately one-third of the study patients, so we cannot make definitive statements about whether patients on statins were less likely to die of liver-related or cardiovascular causes than patients in the control group," said Dr. Kumar.

The apparent hepatoprotective effect of statin therapy in cirrhosis patients may be a function of previously observed hemodynamic and molecular effects of statins, Dr. Kumar hypothesized. Sinusoidal endothelial dysfunction with decreased nitric oxide production contributes to increased hepatic resistance in cirrhosis, she explained. Just as statins improve endothelial dysfunction in the peripheral vasculature, they may also improve the vascular disturbances that contribute to portal hypertension in cirrhosis by selectively increasing nitric oxide availability in the liver, thus reducing pressures, she said.

The retrospective design of the study limits the conclusions that can be taken from it. Specifically, "we can’t say that all patients with liver disease should be prescribed statins," Dr. Kumar said. "What we can say is that statin therapy is safe in this population, it may be beneficial for its effects on the liver as well as the cardiovascular system, and clinicians should not hesitate to prescribe it for appropriate cardiovascular indications in cirrhosis patients." The findings also indicate that prospective studies are warranted to clarify the role of statins in advanced liver disease, she stressed.

 

 

Dr. Kumar disclosed no relevant financial conflicts of interest.

Publications
Publications
Topics
Article Type
Display Headline
Statins Appear Safe, Even Beneficial, in Cirrhosis
Display Headline
Statins Appear Safe, Even Beneficial, in Cirrhosis
Legacy Keywords
statin therapy, statins cirrhosis, statins liver disease, hepatic decompensation, Digestive Disease Week 2012, Dr. Sonal Kumar
Legacy Keywords
statin therapy, statins cirrhosis, statins liver disease, hepatic decompensation, Digestive Disease Week 2012, Dr. Sonal Kumar
Sections
Article Source

FROM THE ANNUAL DIGESTIVE DISEASE WEEK

PURLs Copyright

Inside the Article

Vitals

Major Finding: Among patients with advanced liver disease, the hepatic decompensation rate was 38.2% in patients on statin therapy and 50.6% in those not using statins.

Data Source: This was a retrospective analysis of medical record data for 243 patients with biopsy-proven cirrhosis: 81 taking statins for dyslipidemia and 162 controls.

Disclosures: Dr. Kumar disclosed no relevant financial conflicts of interest.