Neuropathic pain treatment provides unexpected benefit

Article Type
Changed
Fri, 01/18/2019 - 08:47
Display Headline
Neuropathic pain treatment provides unexpected benefit
 

THE CASE

A 57-year-old African American woman was being treated at our clinic for neurogenic urinary incontinence (UI). The UI, which occurred day and night, began 2 years earlier following a laminectomy of vertebrae C3 to C6 with spinal fusion of C3 to C7 for cervical spinal stenosis. The UI persisted despite physical therapy and trials of oxybutynin and imipramine. Since the surgery, the patient had also been experiencing chronic (debilitating) neuropathic pain in both legs, and the sensation of incomplete bladder emptying. She denied bowel incontinence or saddle anesthesia. Her prescription medications included hydrocodone-acetaminophen 7.5/325 mg every 6 hours as needed for pain and lisinopril 20 mg/d for essential hypertension. The patient’s body mass index (BMI) was 23.3.

A urine culture initially grew Klebsiella pneumoniae, which we successfully treated with ciprofloxacin. A urinalysis was unremarkable, and blood urea nitrogen and creatinine levels were within normal limits.

We started the patient on oral duloxetine 30 mg/d for her neuropathic pain. The patient hadn’t undergone a urologic evaluation before starting duloxetine, so no urodynamic studies or measurements had been conducted. At that point, we sent the patient to a urologist for an evaluation.

At a follow-up visit with one of our clinic providers <3 months later, the patient reported that the duloxetine was providing her with some pain relief and that she was “waking up dry” in the mornings and having fewer UI symptoms throughout the day, as well as at night. The patient denied any adverse effects such as nausea, gastrointestinal upset, weight changes, xerostomia, fatigue, insomnia, headaches, or dizziness. Duloxetine was titrated up to 60 mg/d for better control of her neuropathic pain. At the next follow-up visit at our clinic 3 months later, her UI was 80% to 90% improved and she was able to stop her opioid pain medications.

DISCUSSION

UI is a significant problem in the United States and around the world. For women, the prevalence of UI ranges from 15% to 69%; among men, the prevalence is 5% to 24%.1-3 The economic burden of UI includes both medical and nonmedical (eg, pads, diapers, laundry, and dry cleaning) care. The total national cost was estimated at $66 billion in 2007: $49 billion for direct medical costs, $2 billion for direct nonmedical costs, and $15 billion for indirect costs.4 And those costs are expected to increase 25% by 2020, mainly because of the aging population.

Risk factors for UI other than gender include advancing age, obesity, non-Hispanic white race, depression, hypertension, type 2 diabetes mellitus, neurologic disease, and functional limitations/general poor health.5-7 Comorbid depression and BMI >30, as well as the presence and duration of diabetes, increase the odds for developing UI.7,8

Duloxetine has been shown to be effective for the treatment of stress and mixed urinary incontinence. This case suggests it may be useful for neurogenic urinary incontinence, as well.

Risk factors for women include hysterectomy,7 increasing parity, and delivery of at least one infant >9.5 pounds; the risk is the same for both vaginal and cesarean-section delivery.6 Specific risk factors for men include prostate cancer, prostate surgery, and prostate radiation.5

Significant, chronic comorbidities of UI include depression and chronic pain. While quality of life is negatively affected by UI alone, the coexistence of depression and UI produces an additive negative effect on quality of life.9

Types and treatment of UI

There are 5 types of UI: urge, stress, overflow, functional, and mixed.10

  • Urge incontinence is the leakage of urine following a sensation of sudden urgency to void.
  • Stress incontinence is urine leakage associated with increased intra-abdominal pressure such as with coughing or sneezing and is typically associated with weakened pelvic floor musculature.
  • Overflow incontinence is more common in men, and is typically caused by prostatic disease. The urethral outlet is obstructed leading to increased pressure within the bladder and subsequent leakage of urine.
  • Functional incontinence is caused by physical or cognitive impairment leading to a decreased ability to get to a bathroom quickly enough to void.
  • Mixed incontinence is when symptoms of stress and urgency incontinence are present.

There are 3 broad categories of treatment methods for urinary incontinence: behavioral, pharmacologic, and surgical. Behavioral interventions are subdivided into caregiver-dependent (prompted voiding, habit retraining, and timed voiding) and patient-directed (bladder training, pelvic floor muscle training, strategies for bladder control, education, and self-monitoring) techniques. Pharmacologic treatment typically consists of antimuscarinics (eg, oxybutynin, tolterodine, solifenacin) and tricyclic antidepressants (eg, imipramine).11 Injections of onabotulinumtoxinA into the detrusor muscle have also been shown to reduce the symptoms of urinary incontinence.12 Surgical options for treatment of UI include retro-pubic suspension, slings, and, in some instances, artificial urethral sphincters.13

 

 

 

A novel treatment for neurogenic UI?

Despite the many treatments available for UI, none comprehensively addresses UI and its common comorbidities.

The role of duloxetine. Normal micturition is regulated by the somatic nervous system and an autonomic reflex arc; the neurotransmitters serotonin and norepinephrine play an important role in the neural regulation of micturition and urinary continence. Duloxetine, alone or as an adjunctive treatment, is a potential novel therapy that treats 2 common comorbidities of UI—chronic pain and depression.

As a selective serotonin norepinephrine reuptake inhibitor (SNRI), duloxetine acts at the molecular level to block the reuptake of serotonin and norepinephrine from synaptic clefts. Specifically, the medication blocks the 5-hydroxytryptamine (5-HT) reuptake transporters, as well as the norepinephrine transporters, of pre-synaptic neurons.14 Thus, the concentrations of 5-HT and norepinephrine increase in the synaptic cleft.

Functionally, the accumulation of norepinephrine inhibits micturition by relaxing the detrusor muscle and constricting the urethral smooth muscle. In addition, a higher concentration of 5-HT at the neuromuscular junction leads to constriction of the external urethral sphincter.

Duloxetine has been shown to be effective in the treatment of other types of UI, such as stress UI15 and mixed UI.16 Additionally, it was found to be effective when compared with placebo in women with overactive bladder syndrome17 and in women with multiple sclerosis and depression.18 However, we are not aware of any cases using duloxetine for the treatment of neurogenic UI.

THE TAKEAWAY

Duloxetine is a potential novel drug choice for the treatment of neurogenic UI. Its effects on serotonin and norepinephrine at the synaptic cleft and neuromuscular junction could provide relief for those who have not found relief from other therapies. Further research—particularly a prospective, randomized controlled trial—is needed to determine if duloxetine is, in fact, more than just a theoretical candidate to treat UI and, if so, the most effective dosing.

Offering duloxetine for the treatment of neurogenic urinary incontinence would potentially address coexisting conditions, such as pain or depression.

Offering duloxetine for the treatment of neurogenic UI would potentially address coexisting conditions—such as pain or depression—thus improving patient compliance and reducing health care spending. Before beginning therapy, urodynamic studies to identify the type of UI should be completed, or, at a minimum, post-void residual volume should be measured.

ACKNOWLEDGEMENTS
The authors would like to thank Julie Hughbanks, MLS, Library Manager, Parkview Health Resource Library, for her assistance with the library searches used for this case report.

References

1. Markland AD, Richter HE, Fwu CW, et al. Prevalence and trends of urinary incontinence in adults in the United States, 2001 to 2008. J Urol. 2011;186:589-593.

2. Buckley BS, Lapitan MC; Epidemiology Committee of the Fourth International Consultation on Incontinence, Paris, 2008. Prevalence of urinary incontinence in men, women, and children—current evidence: findings of the Fourth International Consultation on Incontinence. Urology. 2010;76:265-270.

3. Gorina Y, Schappert S, Bercovitz A, et al. Prevalence of incontinence among older Americans. Vital Health Stat 3. 2014;1-33.

4. Coyne KS, Wein A, Nicholson S, et al. Economic burden of urgency urinary incontinence in the United States: a systematic review. J Manag Care Pharm. 2014;20:130-140.

5. Shamliyan TA, Wyman JF, Ping R, et al. Male urinary incontinence: prevalence, risk factors, and preventive interventions. Rev Urol. 2009;11:145-165.

6. Matthews CA, Whitehead WE, Townsend MK, et al. Risk factors for urinary, fecal, or dual incontinence in the Nurses’ Health Study. Obstet Gynecol. 2013;122:539-545.

7. Danforth KN, Townsend MK, Lifford K, et al. Risk factors for urinary incontinence among middle-aged women. Am J Obstet Gynecol. 2006;194:339-345.

8. Lifford KL, Curhan GC, Hu FB, et al. Type 2 diabetes mellitus and risk of developing urinary incontinence. J Am Geriatr Soc. 2005;53:1851-1857.

9. Avery JC, Stocks NP, Duggan P, et al. Identifying the quality of life effects of urinary incontinence with depression in an Australian population. BMC Urol. 2013;13:11.

10. National Kidney and Urologic Diseases Information Clearinghouse. Urinary incontinence in women. Available at: http://kidney.niddk.nih.gov/KUDISEASES/pubs/uiwomen/UI-Women_508.pdf. Accessed January 2, 2015.

11. Ontario Medical Advisory Secretariat. Behavioural interventions for urinary incontinence in community-dwelling seniors: an evidence-based analysis. Ontario Health Technology Assessment Series. 2008:8. Available at: http://www.hqontario.ca/Portals/0/Documents/evidence/reports/rev_aic_ui_20081002.pdf. Accessed November 30, 2015.

12. Cox L, Cameron A. OnabotulinumtoxinA for the treatment of overactive bladder. Res Rep Urol. 2014;6:79-89.

13. Dmochowski RR, Blaivas JM, Gormley EA, et al. Update of AUA guideline on the surgical management of female stress urinary incontinence. J Urol. 2010;183:1906-1914.

14. Duloxetine. US National Library of Medicine: National Center for Biotechnology Information. 2015. Available at: http://pubchem.ncbi.nlm.nih.gov/compound/duloxetine. Accessed October 20, 2015.

15. Li J, Yang L, Pu C, et al. The role of duloxetine in stress urinary incontinence: a systematic review and meta-analysis. Int Urol Nephrol. 2013;45:679-686.

16. Bent AE, Gousse AE, Hendrix SL, et al. Duloxetine compared with placebo for the treatment of women with mixed urinary incontinence. Neurourol Urodyn. 2008;27:212-221.

17. Steers WD, Herschorn S, Kreder KJ, et al; Duloxetine OAB Study Group. Duloxetine compared with placebo for treating women with symptoms of overactive bladder. BJU Int. 2007;100:337-345.

18. Di Rezze S, Frasca V, Inghilleri M, et al. Duloxetine for the treatment of overactive bladder syndrome in multiple sclerosis: a pilot study. Clin Neuropharmacol. 2012;35:231-234.

Article PDF
Author and Disclosure Information

Ark City Clinic, Arkansas City, Kans. (Dr. Keesling); University of Saint Francis, Fort Wayne, Ind. (Dr. Wilson); Fort Wayne Medical Education Program, Ind. (Dr. Wilkins)
[email protected]

The authors reported no potential conflict of interest relevant to this article.

Adapted from a poster presentation, Indiana Academy of Family Physicians 2015 Research Day, May 7, 2015, Indianapolis, Ind

Issue
The Journal of Family Practice - 66(6)
Publications
Topics
Page Number
388-390
Sections
Author and Disclosure Information

Ark City Clinic, Arkansas City, Kans. (Dr. Keesling); University of Saint Francis, Fort Wayne, Ind. (Dr. Wilson); Fort Wayne Medical Education Program, Ind. (Dr. Wilkins)
[email protected]

The authors reported no potential conflict of interest relevant to this article.

Adapted from a poster presentation, Indiana Academy of Family Physicians 2015 Research Day, May 7, 2015, Indianapolis, Ind

Author and Disclosure Information

Ark City Clinic, Arkansas City, Kans. (Dr. Keesling); University of Saint Francis, Fort Wayne, Ind. (Dr. Wilson); Fort Wayne Medical Education Program, Ind. (Dr. Wilkins)
[email protected]

The authors reported no potential conflict of interest relevant to this article.

Adapted from a poster presentation, Indiana Academy of Family Physicians 2015 Research Day, May 7, 2015, Indianapolis, Ind

Article PDF
Article PDF
 

THE CASE

A 57-year-old African American woman was being treated at our clinic for neurogenic urinary incontinence (UI). The UI, which occurred day and night, began 2 years earlier following a laminectomy of vertebrae C3 to C6 with spinal fusion of C3 to C7 for cervical spinal stenosis. The UI persisted despite physical therapy and trials of oxybutynin and imipramine. Since the surgery, the patient had also been experiencing chronic (debilitating) neuropathic pain in both legs, and the sensation of incomplete bladder emptying. She denied bowel incontinence or saddle anesthesia. Her prescription medications included hydrocodone-acetaminophen 7.5/325 mg every 6 hours as needed for pain and lisinopril 20 mg/d for essential hypertension. The patient’s body mass index (BMI) was 23.3.

A urine culture initially grew Klebsiella pneumoniae, which we successfully treated with ciprofloxacin. A urinalysis was unremarkable, and blood urea nitrogen and creatinine levels were within normal limits.

We started the patient on oral duloxetine 30 mg/d for her neuropathic pain. The patient hadn’t undergone a urologic evaluation before starting duloxetine, so no urodynamic studies or measurements had been conducted. At that point, we sent the patient to a urologist for an evaluation.

At a follow-up visit with one of our clinic providers <3 months later, the patient reported that the duloxetine was providing her with some pain relief and that she was “waking up dry” in the mornings and having fewer UI symptoms throughout the day, as well as at night. The patient denied any adverse effects such as nausea, gastrointestinal upset, weight changes, xerostomia, fatigue, insomnia, headaches, or dizziness. Duloxetine was titrated up to 60 mg/d for better control of her neuropathic pain. At the next follow-up visit at our clinic 3 months later, her UI was 80% to 90% improved and she was able to stop her opioid pain medications.

DISCUSSION

UI is a significant problem in the United States and around the world. For women, the prevalence of UI ranges from 15% to 69%; among men, the prevalence is 5% to 24%.1-3 The economic burden of UI includes both medical and nonmedical (eg, pads, diapers, laundry, and dry cleaning) care. The total national cost was estimated at $66 billion in 2007: $49 billion for direct medical costs, $2 billion for direct nonmedical costs, and $15 billion for indirect costs.4 And those costs are expected to increase 25% by 2020, mainly because of the aging population.

Risk factors for UI other than gender include advancing age, obesity, non-Hispanic white race, depression, hypertension, type 2 diabetes mellitus, neurologic disease, and functional limitations/general poor health.5-7 Comorbid depression and BMI >30, as well as the presence and duration of diabetes, increase the odds for developing UI.7,8

Duloxetine has been shown to be effective for the treatment of stress and mixed urinary incontinence. This case suggests it may be useful for neurogenic urinary incontinence, as well.

Risk factors for women include hysterectomy,7 increasing parity, and delivery of at least one infant >9.5 pounds; the risk is the same for both vaginal and cesarean-section delivery.6 Specific risk factors for men include prostate cancer, prostate surgery, and prostate radiation.5

Significant, chronic comorbidities of UI include depression and chronic pain. While quality of life is negatively affected by UI alone, the coexistence of depression and UI produces an additive negative effect on quality of life.9

Types and treatment of UI

There are 5 types of UI: urge, stress, overflow, functional, and mixed.10

  • Urge incontinence is the leakage of urine following a sensation of sudden urgency to void.
  • Stress incontinence is urine leakage associated with increased intra-abdominal pressure such as with coughing or sneezing and is typically associated with weakened pelvic floor musculature.
  • Overflow incontinence is more common in men, and is typically caused by prostatic disease. The urethral outlet is obstructed leading to increased pressure within the bladder and subsequent leakage of urine.
  • Functional incontinence is caused by physical or cognitive impairment leading to a decreased ability to get to a bathroom quickly enough to void.
  • Mixed incontinence is when symptoms of stress and urgency incontinence are present.

There are 3 broad categories of treatment methods for urinary incontinence: behavioral, pharmacologic, and surgical. Behavioral interventions are subdivided into caregiver-dependent (prompted voiding, habit retraining, and timed voiding) and patient-directed (bladder training, pelvic floor muscle training, strategies for bladder control, education, and self-monitoring) techniques. Pharmacologic treatment typically consists of antimuscarinics (eg, oxybutynin, tolterodine, solifenacin) and tricyclic antidepressants (eg, imipramine).11 Injections of onabotulinumtoxinA into the detrusor muscle have also been shown to reduce the symptoms of urinary incontinence.12 Surgical options for treatment of UI include retro-pubic suspension, slings, and, in some instances, artificial urethral sphincters.13

 

 

 

A novel treatment for neurogenic UI?

Despite the many treatments available for UI, none comprehensively addresses UI and its common comorbidities.

The role of duloxetine. Normal micturition is regulated by the somatic nervous system and an autonomic reflex arc; the neurotransmitters serotonin and norepinephrine play an important role in the neural regulation of micturition and urinary continence. Duloxetine, alone or as an adjunctive treatment, is a potential novel therapy that treats 2 common comorbidities of UI—chronic pain and depression.

As a selective serotonin norepinephrine reuptake inhibitor (SNRI), duloxetine acts at the molecular level to block the reuptake of serotonin and norepinephrine from synaptic clefts. Specifically, the medication blocks the 5-hydroxytryptamine (5-HT) reuptake transporters, as well as the norepinephrine transporters, of pre-synaptic neurons.14 Thus, the concentrations of 5-HT and norepinephrine increase in the synaptic cleft.

Functionally, the accumulation of norepinephrine inhibits micturition by relaxing the detrusor muscle and constricting the urethral smooth muscle. In addition, a higher concentration of 5-HT at the neuromuscular junction leads to constriction of the external urethral sphincter.

Duloxetine has been shown to be effective in the treatment of other types of UI, such as stress UI15 and mixed UI.16 Additionally, it was found to be effective when compared with placebo in women with overactive bladder syndrome17 and in women with multiple sclerosis and depression.18 However, we are not aware of any cases using duloxetine for the treatment of neurogenic UI.

THE TAKEAWAY

Duloxetine is a potential novel drug choice for the treatment of neurogenic UI. Its effects on serotonin and norepinephrine at the synaptic cleft and neuromuscular junction could provide relief for those who have not found relief from other therapies. Further research—particularly a prospective, randomized controlled trial—is needed to determine if duloxetine is, in fact, more than just a theoretical candidate to treat UI and, if so, the most effective dosing.

Offering duloxetine for the treatment of neurogenic urinary incontinence would potentially address coexisting conditions, such as pain or depression.

Offering duloxetine for the treatment of neurogenic UI would potentially address coexisting conditions—such as pain or depression—thus improving patient compliance and reducing health care spending. Before beginning therapy, urodynamic studies to identify the type of UI should be completed, or, at a minimum, post-void residual volume should be measured.

ACKNOWLEDGEMENTS
The authors would like to thank Julie Hughbanks, MLS, Library Manager, Parkview Health Resource Library, for her assistance with the library searches used for this case report.

 

THE CASE

A 57-year-old African American woman was being treated at our clinic for neurogenic urinary incontinence (UI). The UI, which occurred day and night, began 2 years earlier following a laminectomy of vertebrae C3 to C6 with spinal fusion of C3 to C7 for cervical spinal stenosis. The UI persisted despite physical therapy and trials of oxybutynin and imipramine. Since the surgery, the patient had also been experiencing chronic (debilitating) neuropathic pain in both legs, and the sensation of incomplete bladder emptying. She denied bowel incontinence or saddle anesthesia. Her prescription medications included hydrocodone-acetaminophen 7.5/325 mg every 6 hours as needed for pain and lisinopril 20 mg/d for essential hypertension. The patient’s body mass index (BMI) was 23.3.

A urine culture initially grew Klebsiella pneumoniae, which we successfully treated with ciprofloxacin. A urinalysis was unremarkable, and blood urea nitrogen and creatinine levels were within normal limits.

We started the patient on oral duloxetine 30 mg/d for her neuropathic pain. The patient hadn’t undergone a urologic evaluation before starting duloxetine, so no urodynamic studies or measurements had been conducted. At that point, we sent the patient to a urologist for an evaluation.

At a follow-up visit with one of our clinic providers <3 months later, the patient reported that the duloxetine was providing her with some pain relief and that she was “waking up dry” in the mornings and having fewer UI symptoms throughout the day, as well as at night. The patient denied any adverse effects such as nausea, gastrointestinal upset, weight changes, xerostomia, fatigue, insomnia, headaches, or dizziness. Duloxetine was titrated up to 60 mg/d for better control of her neuropathic pain. At the next follow-up visit at our clinic 3 months later, her UI was 80% to 90% improved and she was able to stop her opioid pain medications.

DISCUSSION

UI is a significant problem in the United States and around the world. For women, the prevalence of UI ranges from 15% to 69%; among men, the prevalence is 5% to 24%.1-3 The economic burden of UI includes both medical and nonmedical (eg, pads, diapers, laundry, and dry cleaning) care. The total national cost was estimated at $66 billion in 2007: $49 billion for direct medical costs, $2 billion for direct nonmedical costs, and $15 billion for indirect costs.4 And those costs are expected to increase 25% by 2020, mainly because of the aging population.

Risk factors for UI other than gender include advancing age, obesity, non-Hispanic white race, depression, hypertension, type 2 diabetes mellitus, neurologic disease, and functional limitations/general poor health.5-7 Comorbid depression and BMI >30, as well as the presence and duration of diabetes, increase the odds for developing UI.7,8

Duloxetine has been shown to be effective for the treatment of stress and mixed urinary incontinence. This case suggests it may be useful for neurogenic urinary incontinence, as well.

Risk factors for women include hysterectomy,7 increasing parity, and delivery of at least one infant >9.5 pounds; the risk is the same for both vaginal and cesarean-section delivery.6 Specific risk factors for men include prostate cancer, prostate surgery, and prostate radiation.5

Significant, chronic comorbidities of UI include depression and chronic pain. While quality of life is negatively affected by UI alone, the coexistence of depression and UI produces an additive negative effect on quality of life.9

Types and treatment of UI

There are 5 types of UI: urge, stress, overflow, functional, and mixed.10

  • Urge incontinence is the leakage of urine following a sensation of sudden urgency to void.
  • Stress incontinence is urine leakage associated with increased intra-abdominal pressure such as with coughing or sneezing and is typically associated with weakened pelvic floor musculature.
  • Overflow incontinence is more common in men, and is typically caused by prostatic disease. The urethral outlet is obstructed leading to increased pressure within the bladder and subsequent leakage of urine.
  • Functional incontinence is caused by physical or cognitive impairment leading to a decreased ability to get to a bathroom quickly enough to void.
  • Mixed incontinence is when symptoms of stress and urgency incontinence are present.

There are 3 broad categories of treatment methods for urinary incontinence: behavioral, pharmacologic, and surgical. Behavioral interventions are subdivided into caregiver-dependent (prompted voiding, habit retraining, and timed voiding) and patient-directed (bladder training, pelvic floor muscle training, strategies for bladder control, education, and self-monitoring) techniques. Pharmacologic treatment typically consists of antimuscarinics (eg, oxybutynin, tolterodine, solifenacin) and tricyclic antidepressants (eg, imipramine).11 Injections of onabotulinumtoxinA into the detrusor muscle have also been shown to reduce the symptoms of urinary incontinence.12 Surgical options for treatment of UI include retro-pubic suspension, slings, and, in some instances, artificial urethral sphincters.13

 

 

 

A novel treatment for neurogenic UI?

Despite the many treatments available for UI, none comprehensively addresses UI and its common comorbidities.

The role of duloxetine. Normal micturition is regulated by the somatic nervous system and an autonomic reflex arc; the neurotransmitters serotonin and norepinephrine play an important role in the neural regulation of micturition and urinary continence. Duloxetine, alone or as an adjunctive treatment, is a potential novel therapy that treats 2 common comorbidities of UI—chronic pain and depression.

As a selective serotonin norepinephrine reuptake inhibitor (SNRI), duloxetine acts at the molecular level to block the reuptake of serotonin and norepinephrine from synaptic clefts. Specifically, the medication blocks the 5-hydroxytryptamine (5-HT) reuptake transporters, as well as the norepinephrine transporters, of pre-synaptic neurons.14 Thus, the concentrations of 5-HT and norepinephrine increase in the synaptic cleft.

Functionally, the accumulation of norepinephrine inhibits micturition by relaxing the detrusor muscle and constricting the urethral smooth muscle. In addition, a higher concentration of 5-HT at the neuromuscular junction leads to constriction of the external urethral sphincter.

Duloxetine has been shown to be effective in the treatment of other types of UI, such as stress UI15 and mixed UI.16 Additionally, it was found to be effective when compared with placebo in women with overactive bladder syndrome17 and in women with multiple sclerosis and depression.18 However, we are not aware of any cases using duloxetine for the treatment of neurogenic UI.

THE TAKEAWAY

Duloxetine is a potential novel drug choice for the treatment of neurogenic UI. Its effects on serotonin and norepinephrine at the synaptic cleft and neuromuscular junction could provide relief for those who have not found relief from other therapies. Further research—particularly a prospective, randomized controlled trial—is needed to determine if duloxetine is, in fact, more than just a theoretical candidate to treat UI and, if so, the most effective dosing.

Offering duloxetine for the treatment of neurogenic urinary incontinence would potentially address coexisting conditions, such as pain or depression.

Offering duloxetine for the treatment of neurogenic UI would potentially address coexisting conditions—such as pain or depression—thus improving patient compliance and reducing health care spending. Before beginning therapy, urodynamic studies to identify the type of UI should be completed, or, at a minimum, post-void residual volume should be measured.

ACKNOWLEDGEMENTS
The authors would like to thank Julie Hughbanks, MLS, Library Manager, Parkview Health Resource Library, for her assistance with the library searches used for this case report.

References

1. Markland AD, Richter HE, Fwu CW, et al. Prevalence and trends of urinary incontinence in adults in the United States, 2001 to 2008. J Urol. 2011;186:589-593.

2. Buckley BS, Lapitan MC; Epidemiology Committee of the Fourth International Consultation on Incontinence, Paris, 2008. Prevalence of urinary incontinence in men, women, and children—current evidence: findings of the Fourth International Consultation on Incontinence. Urology. 2010;76:265-270.

3. Gorina Y, Schappert S, Bercovitz A, et al. Prevalence of incontinence among older Americans. Vital Health Stat 3. 2014;1-33.

4. Coyne KS, Wein A, Nicholson S, et al. Economic burden of urgency urinary incontinence in the United States: a systematic review. J Manag Care Pharm. 2014;20:130-140.

5. Shamliyan TA, Wyman JF, Ping R, et al. Male urinary incontinence: prevalence, risk factors, and preventive interventions. Rev Urol. 2009;11:145-165.

6. Matthews CA, Whitehead WE, Townsend MK, et al. Risk factors for urinary, fecal, or dual incontinence in the Nurses’ Health Study. Obstet Gynecol. 2013;122:539-545.

7. Danforth KN, Townsend MK, Lifford K, et al. Risk factors for urinary incontinence among middle-aged women. Am J Obstet Gynecol. 2006;194:339-345.

8. Lifford KL, Curhan GC, Hu FB, et al. Type 2 diabetes mellitus and risk of developing urinary incontinence. J Am Geriatr Soc. 2005;53:1851-1857.

9. Avery JC, Stocks NP, Duggan P, et al. Identifying the quality of life effects of urinary incontinence with depression in an Australian population. BMC Urol. 2013;13:11.

10. National Kidney and Urologic Diseases Information Clearinghouse. Urinary incontinence in women. Available at: http://kidney.niddk.nih.gov/KUDISEASES/pubs/uiwomen/UI-Women_508.pdf. Accessed January 2, 2015.

11. Ontario Medical Advisory Secretariat. Behavioural interventions for urinary incontinence in community-dwelling seniors: an evidence-based analysis. Ontario Health Technology Assessment Series. 2008:8. Available at: http://www.hqontario.ca/Portals/0/Documents/evidence/reports/rev_aic_ui_20081002.pdf. Accessed November 30, 2015.

12. Cox L, Cameron A. OnabotulinumtoxinA for the treatment of overactive bladder. Res Rep Urol. 2014;6:79-89.

13. Dmochowski RR, Blaivas JM, Gormley EA, et al. Update of AUA guideline on the surgical management of female stress urinary incontinence. J Urol. 2010;183:1906-1914.

14. Duloxetine. US National Library of Medicine: National Center for Biotechnology Information. 2015. Available at: http://pubchem.ncbi.nlm.nih.gov/compound/duloxetine. Accessed October 20, 2015.

15. Li J, Yang L, Pu C, et al. The role of duloxetine in stress urinary incontinence: a systematic review and meta-analysis. Int Urol Nephrol. 2013;45:679-686.

16. Bent AE, Gousse AE, Hendrix SL, et al. Duloxetine compared with placebo for the treatment of women with mixed urinary incontinence. Neurourol Urodyn. 2008;27:212-221.

17. Steers WD, Herschorn S, Kreder KJ, et al; Duloxetine OAB Study Group. Duloxetine compared with placebo for treating women with symptoms of overactive bladder. BJU Int. 2007;100:337-345.

18. Di Rezze S, Frasca V, Inghilleri M, et al. Duloxetine for the treatment of overactive bladder syndrome in multiple sclerosis: a pilot study. Clin Neuropharmacol. 2012;35:231-234.

References

1. Markland AD, Richter HE, Fwu CW, et al. Prevalence and trends of urinary incontinence in adults in the United States, 2001 to 2008. J Urol. 2011;186:589-593.

2. Buckley BS, Lapitan MC; Epidemiology Committee of the Fourth International Consultation on Incontinence, Paris, 2008. Prevalence of urinary incontinence in men, women, and children—current evidence: findings of the Fourth International Consultation on Incontinence. Urology. 2010;76:265-270.

3. Gorina Y, Schappert S, Bercovitz A, et al. Prevalence of incontinence among older Americans. Vital Health Stat 3. 2014;1-33.

4. Coyne KS, Wein A, Nicholson S, et al. Economic burden of urgency urinary incontinence in the United States: a systematic review. J Manag Care Pharm. 2014;20:130-140.

5. Shamliyan TA, Wyman JF, Ping R, et al. Male urinary incontinence: prevalence, risk factors, and preventive interventions. Rev Urol. 2009;11:145-165.

6. Matthews CA, Whitehead WE, Townsend MK, et al. Risk factors for urinary, fecal, or dual incontinence in the Nurses’ Health Study. Obstet Gynecol. 2013;122:539-545.

7. Danforth KN, Townsend MK, Lifford K, et al. Risk factors for urinary incontinence among middle-aged women. Am J Obstet Gynecol. 2006;194:339-345.

8. Lifford KL, Curhan GC, Hu FB, et al. Type 2 diabetes mellitus and risk of developing urinary incontinence. J Am Geriatr Soc. 2005;53:1851-1857.

9. Avery JC, Stocks NP, Duggan P, et al. Identifying the quality of life effects of urinary incontinence with depression in an Australian population. BMC Urol. 2013;13:11.

10. National Kidney and Urologic Diseases Information Clearinghouse. Urinary incontinence in women. Available at: http://kidney.niddk.nih.gov/KUDISEASES/pubs/uiwomen/UI-Women_508.pdf. Accessed January 2, 2015.

11. Ontario Medical Advisory Secretariat. Behavioural interventions for urinary incontinence in community-dwelling seniors: an evidence-based analysis. Ontario Health Technology Assessment Series. 2008:8. Available at: http://www.hqontario.ca/Portals/0/Documents/evidence/reports/rev_aic_ui_20081002.pdf. Accessed November 30, 2015.

12. Cox L, Cameron A. OnabotulinumtoxinA for the treatment of overactive bladder. Res Rep Urol. 2014;6:79-89.

13. Dmochowski RR, Blaivas JM, Gormley EA, et al. Update of AUA guideline on the surgical management of female stress urinary incontinence. J Urol. 2010;183:1906-1914.

14. Duloxetine. US National Library of Medicine: National Center for Biotechnology Information. 2015. Available at: http://pubchem.ncbi.nlm.nih.gov/compound/duloxetine. Accessed October 20, 2015.

15. Li J, Yang L, Pu C, et al. The role of duloxetine in stress urinary incontinence: a systematic review and meta-analysis. Int Urol Nephrol. 2013;45:679-686.

16. Bent AE, Gousse AE, Hendrix SL, et al. Duloxetine compared with placebo for the treatment of women with mixed urinary incontinence. Neurourol Urodyn. 2008;27:212-221.

17. Steers WD, Herschorn S, Kreder KJ, et al; Duloxetine OAB Study Group. Duloxetine compared with placebo for treating women with symptoms of overactive bladder. BJU Int. 2007;100:337-345.

18. Di Rezze S, Frasca V, Inghilleri M, et al. Duloxetine for the treatment of overactive bladder syndrome in multiple sclerosis: a pilot study. Clin Neuropharmacol. 2012;35:231-234.

Issue
The Journal of Family Practice - 66(6)
Issue
The Journal of Family Practice - 66(6)
Page Number
388-390
Page Number
388-390
Publications
Publications
Topics
Article Type
Display Headline
Neuropathic pain treatment provides unexpected benefit
Display Headline
Neuropathic pain treatment provides unexpected benefit
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
PubMed ID
28574523
Article PDF Media

The one thing that’s missing from the health care debate

Article Type
Changed
Wed, 03/27/2019 - 11:48
Display Headline
The one thing that’s missing from the health care debate
 

The Affordable Care Act (aka Obamacare) may soon be out and the American Health Care Act (AHCA) may soon be in. Despite all of the rhetoric about making health care affordable by reducing insurance premiums, one thing has been conspicuously absent from the debate: how we are going to reduce the actual cost of health care. Yes, the AHCA may help reduce premiums, but what is most likely to result is not less expensive health care, but rather people paying less money on premiums and more out of their pockets for medicines and treatments. Especially troublesome is that older and sicker patients will be hit the hardest.

The American conundrum. Why do Americans pay twice what citizens of most other developed nations pay and get health care outcomes that are worse?1,2 Two reasons are that those who provide health care charge more in this country for services and medications, and physicians do a lot more testing and treatment here than their counterparts abroad.

If we control the cost of providing care, insurance premiums will follow suit.

One expert estimated that up to $700 billion could be saved by eliminating testing and treatments that provide marginal or no value to patients.3 For example, knee arthroscopy for moderate knee osteoarthritis produces no better outcomes than medical management.4 And many medications are much more expensive in the United States than in other countries. It seems that pharmaceutical companies are permitted greater profits here than elsewhere in the world, and these profits are at the expense of sick people and taxpayers.

 

 

 

How do we bend the cost curve downward? This is a tough question with no single correct answer, but we can all help. Some health care organizations have already reduced costs significantly without sacrificing quality by using team-based primary care as their foundation. Two examples are Nuka Health and Iora Health.5,6

As primary care physicians, we are in an ideal position to constrain unnecessary testing and treatments by establishing trusting relationships with patients, who will believe us when we tell them they don’t need an antibiotic for their chest cold or an MRI for their back pain.

If we control the cost of providing care, insurance premiums will follow suit.

References

1. The Commonwealth Fund. U.S. health care from a global perspective. Available at: http://www.commonwealthfund.org/publications/issue-briefs/2015/oct/us-health-care-from-a-global-perspective. Accessed May 14, 2017.

2. The Commonwealth Fund. US health system ranks last among eleven countries on measures of access, equity, quality, efficiency, and healthy lives. Available at: http://www.commonwealthfund.org/publications/press-releases/2014/jun/us-health-system-ranks-last. Accessed May 14, 2017.

3. Kelley R. Where can $700 billion in waste be cut annually from the U.S. healthcare system? Available at: http://www.hcca-info.org/Portals/0/PDFs/Resources/Conference_Handouts/Compliance_Institute/2010/P8handout6.pdf. Accessed May 14, 2017.

4. Kirkley A, Birmingham TB, Litchfield RB, et al. A randomized trial of arthroscopic surgery for osteoarthritis of the knee. N Engl J Med. 2008;359:1097-1107.

5. Gottlieb K. The Nuka System of Care: improving health through ownership and relationships. Int J Circumpolar Health. 2013;72. doi: 10.3402/ijch.v72i0.21118.

6. Iorahealth. Available at: www.iorahealth.com. Accessed May 14, 2017.

Article PDF
Author and Disclosure Information

Editor-in-Chief

John Hickner, MD, MSc

Issue
The Journal of Family Practice - 66(6)
Publications
Topics
Page Number
344
Sections
Author and Disclosure Information

Editor-in-Chief

John Hickner, MD, MSc

Author and Disclosure Information

Editor-in-Chief

John Hickner, MD, MSc

Article PDF
Article PDF
 

The Affordable Care Act (aka Obamacare) may soon be out and the American Health Care Act (AHCA) may soon be in. Despite all of the rhetoric about making health care affordable by reducing insurance premiums, one thing has been conspicuously absent from the debate: how we are going to reduce the actual cost of health care. Yes, the AHCA may help reduce premiums, but what is most likely to result is not less expensive health care, but rather people paying less money on premiums and more out of their pockets for medicines and treatments. Especially troublesome is that older and sicker patients will be hit the hardest.

The American conundrum. Why do Americans pay twice what citizens of most other developed nations pay and get health care outcomes that are worse?1,2 Two reasons are that those who provide health care charge more in this country for services and medications, and physicians do a lot more testing and treatment here than their counterparts abroad.

If we control the cost of providing care, insurance premiums will follow suit.

One expert estimated that up to $700 billion could be saved by eliminating testing and treatments that provide marginal or no value to patients.3 For example, knee arthroscopy for moderate knee osteoarthritis produces no better outcomes than medical management.4 And many medications are much more expensive in the United States than in other countries. It seems that pharmaceutical companies are permitted greater profits here than elsewhere in the world, and these profits are at the expense of sick people and taxpayers.

 

 

 

How do we bend the cost curve downward? This is a tough question with no single correct answer, but we can all help. Some health care organizations have already reduced costs significantly without sacrificing quality by using team-based primary care as their foundation. Two examples are Nuka Health and Iora Health.5,6

As primary care physicians, we are in an ideal position to constrain unnecessary testing and treatments by establishing trusting relationships with patients, who will believe us when we tell them they don’t need an antibiotic for their chest cold or an MRI for their back pain.

If we control the cost of providing care, insurance premiums will follow suit.

 

The Affordable Care Act (aka Obamacare) may soon be out and the American Health Care Act (AHCA) may soon be in. Despite all of the rhetoric about making health care affordable by reducing insurance premiums, one thing has been conspicuously absent from the debate: how we are going to reduce the actual cost of health care. Yes, the AHCA may help reduce premiums, but what is most likely to result is not less expensive health care, but rather people paying less money on premiums and more out of their pockets for medicines and treatments. Especially troublesome is that older and sicker patients will be hit the hardest.

The American conundrum. Why do Americans pay twice what citizens of most other developed nations pay and get health care outcomes that are worse?1,2 Two reasons are that those who provide health care charge more in this country for services and medications, and physicians do a lot more testing and treatment here than their counterparts abroad.

If we control the cost of providing care, insurance premiums will follow suit.

One expert estimated that up to $700 billion could be saved by eliminating testing and treatments that provide marginal or no value to patients.3 For example, knee arthroscopy for moderate knee osteoarthritis produces no better outcomes than medical management.4 And many medications are much more expensive in the United States than in other countries. It seems that pharmaceutical companies are permitted greater profits here than elsewhere in the world, and these profits are at the expense of sick people and taxpayers.

 

 

 

How do we bend the cost curve downward? This is a tough question with no single correct answer, but we can all help. Some health care organizations have already reduced costs significantly without sacrificing quality by using team-based primary care as their foundation. Two examples are Nuka Health and Iora Health.5,6

As primary care physicians, we are in an ideal position to constrain unnecessary testing and treatments by establishing trusting relationships with patients, who will believe us when we tell them they don’t need an antibiotic for their chest cold or an MRI for their back pain.

If we control the cost of providing care, insurance premiums will follow suit.

References

1. The Commonwealth Fund. U.S. health care from a global perspective. Available at: http://www.commonwealthfund.org/publications/issue-briefs/2015/oct/us-health-care-from-a-global-perspective. Accessed May 14, 2017.

2. The Commonwealth Fund. US health system ranks last among eleven countries on measures of access, equity, quality, efficiency, and healthy lives. Available at: http://www.commonwealthfund.org/publications/press-releases/2014/jun/us-health-system-ranks-last. Accessed May 14, 2017.

3. Kelley R. Where can $700 billion in waste be cut annually from the U.S. healthcare system? Available at: http://www.hcca-info.org/Portals/0/PDFs/Resources/Conference_Handouts/Compliance_Institute/2010/P8handout6.pdf. Accessed May 14, 2017.

4. Kirkley A, Birmingham TB, Litchfield RB, et al. A randomized trial of arthroscopic surgery for osteoarthritis of the knee. N Engl J Med. 2008;359:1097-1107.

5. Gottlieb K. The Nuka System of Care: improving health through ownership and relationships. Int J Circumpolar Health. 2013;72. doi: 10.3402/ijch.v72i0.21118.

6. Iorahealth. Available at: www.iorahealth.com. Accessed May 14, 2017.

References

1. The Commonwealth Fund. U.S. health care from a global perspective. Available at: http://www.commonwealthfund.org/publications/issue-briefs/2015/oct/us-health-care-from-a-global-perspective. Accessed May 14, 2017.

2. The Commonwealth Fund. US health system ranks last among eleven countries on measures of access, equity, quality, efficiency, and healthy lives. Available at: http://www.commonwealthfund.org/publications/press-releases/2014/jun/us-health-system-ranks-last. Accessed May 14, 2017.

3. Kelley R. Where can $700 billion in waste be cut annually from the U.S. healthcare system? Available at: http://www.hcca-info.org/Portals/0/PDFs/Resources/Conference_Handouts/Compliance_Institute/2010/P8handout6.pdf. Accessed May 14, 2017.

4. Kirkley A, Birmingham TB, Litchfield RB, et al. A randomized trial of arthroscopic surgery for osteoarthritis of the knee. N Engl J Med. 2008;359:1097-1107.

5. Gottlieb K. The Nuka System of Care: improving health through ownership and relationships. Int J Circumpolar Health. 2013;72. doi: 10.3402/ijch.v72i0.21118.

6. Iorahealth. Available at: www.iorahealth.com. Accessed May 14, 2017.

Issue
The Journal of Family Practice - 66(6)
Issue
The Journal of Family Practice - 66(6)
Page Number
344
Page Number
344
Publications
Publications
Topics
Article Type
Display Headline
The one thing that’s missing from the health care debate
Display Headline
The one thing that’s missing from the health care debate
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
PubMed ID
28574518
Article PDF Media

Post-bariatric surgery patients: Your role in their long-term care

Article Type
Changed
Fri, 01/18/2019 - 08:47
Display Headline
Post-bariatric surgery patients: Your role in their long-term care
 

More than one-third of American adults and approximately 17% of children and adolescents between the ages of 2 and 19 years are obese.1,2 Poor diet coupled with a sedentary lifestyle is the highest ranked cause of non-communicable disease and a leading cause of preventable death, according to the National Research Council.3

Bariatric surgery (BS) is a viable therapeutic option for obese patients who do not respond to conventional lifestyle interventions for losing weight. There are multiple gastrointestinal (GI) procedures available that are classified as either malabsorptive (Roux-en-Y gastric bypass [RYGB] and biliopancreatic diversion [BPD] with or without duodenal switch) or restrictive (laparoscopic adjustable gastric banding [LAGB] and vertical sleeve gastrectomy [VSG]).

Approximately half of the 196,000 bariatric procedures performed in the United States in 2015 were of the sleeve variety, another 23% were RYGB, and the remaining percentage was divided among the other types.4 Postoperative risks include nutritional deficiencies, decreased bone mineral density (BMD), dumping syndrome (when food rapidly dumps from the stomach to the intestine), and gastroesophageal reflux disease (GERD) with possible ulceration.

Despite these potential complications, a systematic review and meta-analysis found that obese people who underwent BS (gastric banding or gastric bypass) had significantly reduced risks of global, non-cardiovascular (CV), and CV mortality compared with obese controls.5 Helping patients to realize these benefits requires that the entire health care team—especially the family physician—is aware of the special considerations for this population.

Patients undergoing bariatric surgery require routine lifetime screening for nutritional deficiencies.

To that end, this article reviews the details of diagnosing and managing post-surgical complications. It also addresses issues unique to managing certain subpopulations, such as post-BS patients who require revision surgery or who want to pursue body contouring surgery; adolescents who undergo BS surgery; and women who want to get pregnant postoperatively.

Monitor patients for these post-surgery complications

Postoperative BS follow-up varies depending on location, surgeon preference, and availability of multidisciplinary resources. At our institution, patients have a minimum of 3 follow-up visits with their surgeon (during hospitalization and 2 weeks and 2 months postoperatively). This is followed by visits with Endocrinology 6 months after surgery and annually thereafter. Given the variability of follow-up, family physicians should coordinate with specialists where appropriate and be aware of postoperative complications and monitoring since it is likely they will have the most frequent contact with these patients.

Nutritional deficiencies are common and require lifelong screening

Nutritional deficiencies are the most common complications of malabsorptive BS. Guidelines from the Endocrine Society, as well as guidelines from the American Association of Clinical Endocrinologists (AACE), The Obesity Society (TOS), and the American Society for Metabolic and Bariatric Surgery (ASMBS), recommend routine lifetime screening for deficiencies after surgery.6,7 Complete blood cell count, electrolytes, glucose, creatinine, and liver function tests should be obtained at one, 3, 6, 12, 18, and 24 months following surgery and annually thereafter.6

Multiple factors contribute to nutritional and micronutrient deficiencies, including reduced oral intake of food, decreased GI absorption, food intolerance, nausea/vomiting, and nonadherence with dietary supplements.8 Oral supplementation should be in chewable, powder, or liquid form because pill and capsule absorption may be altered.8,9 Over-the-counter multivitamins may not contain the requisite daily doses recommended after BS.9 Patients and physicians should evaluate supplements together to ensure appropriate nutritional and micronutrient supplementation (TABLE 16,8-11).

Bone mineral density can start to decrease soon after surgery

Studies evaluating BMD after BS have produced variable findings. In obese patients, dual-energy x-ray absorptiometry (DEXA) measurements may not be accurate due to adipose tissue artifact and table weight limits. In addition, limited data exist on the incidence of fractures after BS. Of 2 notable studies, only one, a population-based study involving 258 Minnesota residents who underwent a first bariatric surgery between 1985 and 2004, demonstrated a significantly increased incidence of fractures.12,13

In addition, studies show bone turnover markers, including C-terminal telopeptide, increase as early as 3 months after BS.14 Several guidelines recommend routine BMD screening after BS (TABLE 2).6,7 The mechanism of bone demineralization is likely multifactorial—a function of the magnitude of the weight loss and skeletal unloading, calcium and vitamin D deficiencies, and associated secondary hyperparathyroidism.15 Treatment for secondary hyperparathyroidism is adequate supplementation with vitamin D and calcium.

Optimal dosing for vitamin D has not been determined. One recent systematic review suggests routine prophylaxis with at least 2000 international units (IU)/d and found the greatest improvement for known deficiency with doses of 1500-9100 IU/d following malabsorptive surgeries.11 After laparoscopic sleeve gastrectomy, at least 1000 IU/d vitamin D is recommended.11

Overall, high variability exists among patients, and an individualized approach for dosing is recommended.11 Vitamin D levels should be monitored 2 and 4 weeks after initiation of treatment and every 3 months thereafter.11 Normal levels of serum calcium, 25-OH vitamin D, bone-specific alkaline phosphatase, and 24-hour urinary calcium excretion indicate adequate calcium and vitamin D supplementation.6

 

 

 

Dumping syndrome can lead to hypoglycemia

Dumping syndrome is a common complication following BS, with prevalence ranging from 25% to 75%, depending upon the type of procedure performed.16,17 There are 2 types: early and late. Early dumping syndrome occurs within 30 minutes of eating. Symptoms are related to the robust release of gastrointestinal hormones caused by rapid gastric emptying. Symptoms include nausea, abdominal pain, diarrhea, flushing, hypotension, and tachycardia.

Late dumping is characterized as postprandial hypoglycemia occurring one to 3 hours after eating. Late dumping is likely caused by a combination of changes within the pancreatic beta cells and abnormal insulin response to glucose.16-18 Rapid gastric emptying leads to rapid release of glucose in the gut, which, in turn, leads to brisk insulin secretion. Since glucose is absorbed faster than insulin’s half-life, the resulting (relatively) high levels of insulin may cause hypoglycemia.16-18

Sigstad’s scoring system can be used to confirm suspected cases of dumping syndrome (TABLE 316,17,19). A diagnosis can also be made with an oral glucose challenge in which pulse, blood pressure, glucose, and hematocrit are measured after ingestion of 50 g glucose. The test is positive if heart rate increases by 10 beats per minute, hematocrit increases by 3% 30 minutes after ingestion, or glucose falls below 60 mg/dL 2 to 3 hours after ingestion.17

First-line treatment of dumping syndrome consists of dietary modifications. The goal is to slow the rate of gastric emptying by eating smaller, more frequent meals; separating beverages from food; decreasing carbohydrates; and increasing fiber and protein content.

If results are suboptimal after dietary changes, medications can be prescribed including acarbose to prevent postprandial hypoglycemia; anticholinergics such as dicyclomine to slow gastric emptying; and somatostatin to decrease gastric emptying and inhibit GI hormone release.17 Lastly, for resistant and severe postprandial hypoglycemia, a few patients have undergone pancreatectomy, but only about 65% experienced improvement in symptoms and 12% developed diabetes post-surgically.20

Gout attacks may initially increase, but then decrease

BS affects the incidence of gout attacks in patients with a history of gout. One comparative study of approximately 150 patients demonstrated that those with a history of gout had significantly more gout attacks in the first month after BS compared with obese patients with a history of gout undergoing other upper GI surgeries.21 There was no difference between malabsorptive and restrictive procedures. But after the first month, BS patients had significantly fewer gout attacks and lower uric acid levels than their obese counterparts.21

Protein rich diets, catabolism potentiated by aggressive caloric restriction following BS, and dehydration contribute to the initial increase. Therefore, patients who have had at least one gout attack in the year prior to surgery or who are on hypouricemic medication may benefit from at least one month of prophylactic therapy (eg, allopurinol and colchicine) after surgery.

GERD and ulceration: How to respond

Obesity is a known risk factor for GERD, but the effect of BS on GERD is uncertain and seems to vary with the procedure performed. RYGB decreases GERD and is, therefore, used as both a secondary treatment in those not responding to medications and a revision treatment for fundoplication and other types of BSs. Sleeve gastrectomy and adjustable gastric banding have mixed effects on GERD. A systematic review by de Jong et al revealed a decreased prevalence of reflux symptoms and GERD medication use after LAGB; however, during longer follow-up, 15% of previously unaffected patients reported experiencing GERD.22 The 2011 International Sleeve Gastrectomy Expert Panel Consensus Statement retrospectively noted a postoperative incidence of GERD as high as 31%.23

BS patients with GERD should be treated with a proton pump inhibitor. If this fails, refer patients to a gastroenterologist for further evaluation.24

Ulcers after BS may be an indication for revision surgery. Data are mixed regarding increased risk of marginal ulceration from nonsteroidal anti-inflammatory drug (NSAID) use, but NSAIDs have been linked to an increased risk of anastomotic leakage.25-28 Thus, it seems prudent to avoid NSAIDs in people who have undergone BS.

Keeping watch over psychiatric comorbidities

A recent meta-analysis by Dawes et al29 showed that about 23% of patients pursuing BS have a comorbid mood disorder. Specifically, the preoperative prevalence of depression (19%) and binge-eating disorder (17%) were found to be higher than rates in the general population.29 The meta-analysis found improvement in the prevalence of depression with fewer symptoms and less antidepressant medication use in the first 3 years after surgery and a decrease in the rate of binge-eating disorder, although there were fewer supporting data for the latter. These findings were observed with both restrictive and malabsorptive procedures.

 

 

 

The data are mixed regarding rates of alcohol abuse and suicide. Further research is necessary in this field. Patients who have had BS should receive ongoing psychiatric and psychological care from a multidisciplinary team as a matter of course.

Will a second surgery be needed?

Revision surgery. In 2015, about 14% of the almost 200,000 BSs performed were revisions.4 Revision surgery is indicated in BS patients with weight regain, recurrent comorbid diseases (eg, diabetes, hypertension), or complications of primary BS. Restrictive procedures have a higher revision rate than malabsorptive procedures, primarily due to a higher rate of weight regain.6,30

Because revision surgery is associated with more complications and possibly longer hospital stays than primary BS, it should be performed by a bariatric surgeon with extensive experience.30,31 Restrictive revisions are typically converted to malabsorptive procedures. Cost is a limiting factor as many patients’ insurance coverage is limited to one BS per lifetime.

Body contouring. Body contouring surgery (BCS) can improve physical and mental well-being and may be a protective factor for weight regain after bariatric surgery.32 Despite its desirability—particularly to women, adolescents, and those with large decreases in body mass index (BMI)—few patients can afford BCS since it is rarely covered by insurance.

Complications of BCS vary, but are most commonly infection and wound dehiscence. This is, in part, due to poorer wound healing in BS patients compared to those with nonsurgical massive weight loss. The cause of poor wound healing is thought to be secondary to nutritional deficiencies and the catabolic state induced by post-surgical weight loss. Recommendations for BCS include weight stability for more than one year after BS, age >16 years, excess skin causing significant functional impairment, non-smoking status, and presence of good social support.33

Bariatric surgery in adolescents is on the rise

Children in the highest body mass index quartile have more than twice the death rate of those in the lowest BMI quartile.34 Thus, it is not surprising that the rate of BS in adolescents is increasing.7 BS in this age group is successful for weight loss and improvement of comorbid conditions, with relatively low complication rates.35 Options include malabsorptive and restrictive procedures, although gastric banding has not been approved by the US Food and Drug Administration for patients under the age of 18 years.

Monitor vitamin D levels 2 and 4 weeks after initiation of treatment and every 3 months thereafter.After BS, adolescent girls should be counseled regarding the possibility of pregnancy (restoration of fertility) and appropriate contraception. Adolescent patients require nutritional supplementation after BS as indicated in TABLE 1.6,8-11

When determining which adolescents to refer for BS, we recommend the following criteria: 35-38

  • failure of a minimum 6-month trial of a staged treatment approach, as recommended by Barlow et al,36 including diet, exercise, and pharmacologic treatment
  • BMI 35 with type 2 diabetes or severe sleep apnea (apnea hypopnea index [AHI] >15)37
  • BMI 40 with mild sleep apnea (AHI >5), hypertension, or pre-diabetes37
  • Tanner stage IV or V
  • at least 95% skeletal growth (for malabsorptive surgery).37 This can be determined using an estimated adult height from mid-parental height formula and assessing growth plate closure with hand radiographs for bone age
  • appropriate maturity level permitting adherence
  • good psychological support
  • a multidisciplinary team for postoperative and long-term follow-up care.

Planning for the future: Exploring the possibility of pregnancy

Obesity is the primary cause of maternal and fetal morbidity during pregnancy. It is associated with increased rates of early miscarriage, congenital defects, macrosomia, and fetal death. Maternal risks of obesity include: gestational hypertension, gestational diabetes mellitus (GDM), and pre-eclampsia. Obese mothers also have a higher incidence of failed induction, caesarean section, and breastfeeding failure.10,39 Given that half of all BSs are performed in women of reproductive age, this population deserves special consideration.10

A recent meta-analysis by Galazis et al40 concluded that BS performed prior to pregnancy led to decreased rates of preeclampsia, GDM, large neonates, preterm birth, and neonatal intensive care unit admission. Perinatal mortality did not increase after BS. However, BS led to higher rates of maternal anemia. There was no significant difference between groups in incidence of cesarean section.

The post BS female patient should be advised to use a reliable form of contraception for a minimum of 12 to 18 months after surgery.6,10,39 Involve high-risk obstetric specialists during pregnancies. Diet should be supplemented as indicated in TABLE 1.6,8-11

CORRESPONDENCE
Amy Rothberg, MD, PhD, Domino’s Farms, Lobby G, Suite 1500, 24 Frank Lloyd Wright Drive, Ann Arbor, MI 48106; [email protected].

References

1. Centers for Disease Control and Prevention. Overweight and obesity. Adult obesity facts. Available at: https://www.cdc.gov/obesity/data/adult.html. Accessed April 5, 2017.

2. Centers for Disease Control and Prevention. Overweight and obesity. Childhood obesity facts. Available at: https://www.cdc.gov/obesity/data/childhood.html. Accessed April 5, 2017.

3. McGinnis JM. Actual causes of death, 1990-2010. Workshop on Determinants of Premature Mortality, September 18, 2013, National Research Council, Washington, DC.

4. American Society of Metabolic and Bariatric Surgery. Estimate of bariatric surgery numbers, 2011-2015. Available at: https://asmbs.org/resources/estimate-of-bariatric-surgery-numbers. Accessed April 5, 2017.

5. Pontiroli AE, Morabito A. Long-term prevention of mortality in morbid obesity through bariatric surgery. a systematic review and meta-analysis of trials performed with gastric banding and gastric bypass. Ann Surg. 2011;253:484-487.

6. Heber D, Greenway FL, Kaplan LM, et al. Endocrine and nutritional management of the post-bariatric surgery patient: an Endocrine Society clinical practice guideline. J Clin Endocrinol Metab. 2010;95:4823-4843.

7. Mechanick JI, Youdim A, Jones DB, et al. Clinical practice guidelines for the perioperative nutritional, metabolic, and nonsurgical support of the bariatric surgery patient—2013 update: cosponsored by American Association of Clinical Endocrinologists, The Obesity Society, and American Society for Metabolic & Bariatric Surgery. Obesity. 2013;21:S1-S27.

8. Stein J, Stier C, Raab H, et al. Review article: the nutritional and pharmacological consequences of obesity surgery. Aliment Pharmacol Ther. 2014;40:582-609.

9. Boyce SG, Goriparthi R, Clark J, et al. Can composite nutritional supplement based on the current guidelines prevent vitamin and mineral deficiency after weight loss surgery? Obes Surg. 2016;26:966-971.

10. Beard JH, Bell RL, Duffy AJ. Reproductive considerations and pregnancy after bariatric surgery: current evidence and recommendations. Obes Surg. 2008;18:1023-1027.

11. Chakhtoura MT, Nakhoul NN, Shawwa K, et al. Hypovitaminosis D in bariatric surgery: a systematic review of observational studies. Metabolism. 2016;65:574-585.

12. Nakamura KM, Haglind EG, Clowes JA, et al. Fracture risk following bariatric surgery: a population-based study. Osteoporosis Int. 2014;25:151-158.

13. Lalmohamed A, de Vries F, Bazelier MT, et al. Risk of fracture after bariatric surgery in the United Kingdom: population-based, retrospective cohort study. BMJ. 2012;345:e5085.

14. Coates PS, Fernstrom JD, Fernstrom MH, et al. Gastric bypass surgery for morbid obesity leads to an increase in bone turnover and a decrease in bone mass. J Clin Endocrinol Metab. 2004;89:1061-1065.

15. Stein EM, Silverberg SJ. Bone loss after bariatric surgery: causes, consequences and management. Lancet Diabetes Endocrinol. 2014;2:165-174.

16. Tack J, Deloose E. Complications of bariatric surgery: dumping syndrome, reflux and vitamin deficiencies. Best Pract Res Clin Gastroenterol. 2014;28:741-749.

17. Berg P, McCallum R. Dumping syndrome: a review of the current concepts of pathophysiology, diagnosis, and treatment. Dig Dis Sci. 2016;61:11-18.

18. Ritz P, Vaurs C, Barigou M, et al. Hypoglycaemia after gastric bypass: mechanisms and treatment. Diabetes Obes Metab. 2016;18:217-223.

19. Sigstad H. A clinical diagnostic index in the diagnosis of the dumping syndrome. Changes in plasma volume and blood sugar after a test meal. Acta Med Scand. 1970;188:479-486.

20. Mala T. Postprandial hyperinsulinemic hypoglycaemia after gastric bypass surgical treatment. Surg Obes Relat Dis. 2014;10:1220-1225.

21. Romero-Talamás H, Daigle CR, Aminian A. The effect of bariatric surgery on gout: a comparative study. Surg Obes Relat Dis. 2014;10:1161-1165.

22. de Jong JR, Besselink MG, van Ramshorst B, et al. Effects of adjustable gastric banding on gastroesophageal reflux and esophageal motility: a systematic review. Obes Rev. 2010;11:297-305.

23. Rosenthal RJ; International Sleeve Gastrectomy Expert Panel. International Sleeve Gastrectomy Expert Panel Consensus Statement: best practice guidelines based on experience of >12,000 cases. Surg Obes Relat Dis. 2012;8:8-19.

24. Altieri MS, Pryor AD. Gastroesophageal reflux disease after bariatric procedures. Surg Clin North Am. 2015;95:579-591.

25. Hakkarainen TW, Steele SR, Bastaworous A, et al. Nonsteroidal anti-inflammatory drugs and the risk for anastomotic failure: a report from Washington State’s Surgical Care and Outcomes Assessment Program (SCOAP). JAMA Surg. 2015;150:223-228.

26. El-Hayek K, Timratana P, Shimizu H, et al. Marginal ulcer after Roux-en-Y gastric bypass: what have we really learned? Surg Endosc. 2012;26:2789-2796.

27. Sverdén E, Mattsson F, Sondén AM, et al. Risk factors for marginal ulcer after gastric bypass surgery for obesity: a population-based cohort study. Ann Surg. 2016;263:733-737.

28. Azagury DE, Abu Dayyeh BK, Greenwalt IT, et al. Marginal ulceration after Roux-en-Y gastric bypass surgery: characteristics, risk factors, treatment, and outcomes. Endoscopy. 2011;43:950-954.

29. Dawes AJ, Maggard-Gibbons M, Maher AR, et al. Mental health conditions among patients seeking and undergoing bariatric surgery: A meta-analysis. JAMA. 2016;315:150-163.

30. Ferrer-Márquez MP, Belda-Lozano R, Solvas-Salmerón MJ, et al. Revisional surgery after laparoscopic sleeve gastrectomy. Surg Laparosc Endosc Percutan Tech. 2015;25:6-9.

31. Sanchez H, Cabrera A, Cabrera K, et al. Laparoscopic Roux-en-Y gastric bypass as a revision procedure after restrictive bariatric surgery. Obes Surg. 2008;18:1539-1543.

32. van der Beek ES, Te Riele W, Specken TF, et al. The impact of reconstructive procedures following bariatric surgery on patient well-being and quality of life. Obes Surg. 2010;20:36-41.

 

 

33. Ellison JM, Steffen KJ, Sarwer DB. Body contouring after bariatric surgery. Eur Eat Disord Rev. 2015;23:479-487.

34. Franks PW, Hanson RL, Knowler WC, et al. Childhood obesity, other cardiovascular risk factors, and premature death. NEJM. 2010;362:485-489.

35. Gravelle BL, Broyles M. Interventions of weight reduction and prevention in children and adolescents: update. Am J Ther. 2015;22:159-166.

36. Barlow SE. Expert committee recommendations regarding the prevention, assessment, and treatment of child and adolescent overweight and obesity: summary report. Pediatrics. 2007;120:S164-S192.

37. Pratt JS, Lenders CM, Dionne EA, et al. Best practice updates for pediatric/adolescent weight loss surgery. Obesity. 2009;17:901-910.

38. Nogueira I, Hrovat K. Adolescent bariatric surgery: review on nutrition considerations. Nutr Clin Pract. 2014;29:740-746.

39. Nicklas JM, Barbour LA. Optimizing weight for maternal and infant health: tenable, or too late? Expert Rev Endocrinol Metab. 2015;10:227-242.

40. Galazis N, Docheva N, Simillis C, et al. Maternal and neonatal outcomes in women undergoing bariatric surgery: a systematic review and meta-analysis. Eur J Obstet Gynecol Reprod Biol. 2014;181:45-53.

Article PDF
Author and Disclosure Information

University of Michigan, Ann Arbor (Drs. Kreykes and Rothberg); Oregon Health & Science University, Portland (Dr. Choxi)
[email protected]

The authors reported no potential conflict of interest relevant to this article.

Issue
The Journal of Family Practice - 66(6)
Publications
Topics
Page Number
356-363
Sections
Author and Disclosure Information

University of Michigan, Ann Arbor (Drs. Kreykes and Rothberg); Oregon Health & Science University, Portland (Dr. Choxi)
[email protected]

The authors reported no potential conflict of interest relevant to this article.

Author and Disclosure Information

University of Michigan, Ann Arbor (Drs. Kreykes and Rothberg); Oregon Health & Science University, Portland (Dr. Choxi)
[email protected]

The authors reported no potential conflict of interest relevant to this article.

Article PDF
Article PDF
 

More than one-third of American adults and approximately 17% of children and adolescents between the ages of 2 and 19 years are obese.1,2 Poor diet coupled with a sedentary lifestyle is the highest ranked cause of non-communicable disease and a leading cause of preventable death, according to the National Research Council.3

Bariatric surgery (BS) is a viable therapeutic option for obese patients who do not respond to conventional lifestyle interventions for losing weight. There are multiple gastrointestinal (GI) procedures available that are classified as either malabsorptive (Roux-en-Y gastric bypass [RYGB] and biliopancreatic diversion [BPD] with or without duodenal switch) or restrictive (laparoscopic adjustable gastric banding [LAGB] and vertical sleeve gastrectomy [VSG]).

Approximately half of the 196,000 bariatric procedures performed in the United States in 2015 were of the sleeve variety, another 23% were RYGB, and the remaining percentage was divided among the other types.4 Postoperative risks include nutritional deficiencies, decreased bone mineral density (BMD), dumping syndrome (when food rapidly dumps from the stomach to the intestine), and gastroesophageal reflux disease (GERD) with possible ulceration.

Despite these potential complications, a systematic review and meta-analysis found that obese people who underwent BS (gastric banding or gastric bypass) had significantly reduced risks of global, non-cardiovascular (CV), and CV mortality compared with obese controls.5 Helping patients to realize these benefits requires that the entire health care team—especially the family physician—is aware of the special considerations for this population.

Patients undergoing bariatric surgery require routine lifetime screening for nutritional deficiencies.

To that end, this article reviews the details of diagnosing and managing post-surgical complications. It also addresses issues unique to managing certain subpopulations, such as post-BS patients who require revision surgery or who want to pursue body contouring surgery; adolescents who undergo BS surgery; and women who want to get pregnant postoperatively.

Monitor patients for these post-surgery complications

Postoperative BS follow-up varies depending on location, surgeon preference, and availability of multidisciplinary resources. At our institution, patients have a minimum of 3 follow-up visits with their surgeon (during hospitalization and 2 weeks and 2 months postoperatively). This is followed by visits with Endocrinology 6 months after surgery and annually thereafter. Given the variability of follow-up, family physicians should coordinate with specialists where appropriate and be aware of postoperative complications and monitoring since it is likely they will have the most frequent contact with these patients.

Nutritional deficiencies are common and require lifelong screening

Nutritional deficiencies are the most common complications of malabsorptive BS. Guidelines from the Endocrine Society, as well as guidelines from the American Association of Clinical Endocrinologists (AACE), The Obesity Society (TOS), and the American Society for Metabolic and Bariatric Surgery (ASMBS), recommend routine lifetime screening for deficiencies after surgery.6,7 Complete blood cell count, electrolytes, glucose, creatinine, and liver function tests should be obtained at one, 3, 6, 12, 18, and 24 months following surgery and annually thereafter.6

Multiple factors contribute to nutritional and micronutrient deficiencies, including reduced oral intake of food, decreased GI absorption, food intolerance, nausea/vomiting, and nonadherence with dietary supplements.8 Oral supplementation should be in chewable, powder, or liquid form because pill and capsule absorption may be altered.8,9 Over-the-counter multivitamins may not contain the requisite daily doses recommended after BS.9 Patients and physicians should evaluate supplements together to ensure appropriate nutritional and micronutrient supplementation (TABLE 16,8-11).

Bone mineral density can start to decrease soon after surgery

Studies evaluating BMD after BS have produced variable findings. In obese patients, dual-energy x-ray absorptiometry (DEXA) measurements may not be accurate due to adipose tissue artifact and table weight limits. In addition, limited data exist on the incidence of fractures after BS. Of 2 notable studies, only one, a population-based study involving 258 Minnesota residents who underwent a first bariatric surgery between 1985 and 2004, demonstrated a significantly increased incidence of fractures.12,13

In addition, studies show bone turnover markers, including C-terminal telopeptide, increase as early as 3 months after BS.14 Several guidelines recommend routine BMD screening after BS (TABLE 2).6,7 The mechanism of bone demineralization is likely multifactorial—a function of the magnitude of the weight loss and skeletal unloading, calcium and vitamin D deficiencies, and associated secondary hyperparathyroidism.15 Treatment for secondary hyperparathyroidism is adequate supplementation with vitamin D and calcium.

Optimal dosing for vitamin D has not been determined. One recent systematic review suggests routine prophylaxis with at least 2000 international units (IU)/d and found the greatest improvement for known deficiency with doses of 1500-9100 IU/d following malabsorptive surgeries.11 After laparoscopic sleeve gastrectomy, at least 1000 IU/d vitamin D is recommended.11

Overall, high variability exists among patients, and an individualized approach for dosing is recommended.11 Vitamin D levels should be monitored 2 and 4 weeks after initiation of treatment and every 3 months thereafter.11 Normal levels of serum calcium, 25-OH vitamin D, bone-specific alkaline phosphatase, and 24-hour urinary calcium excretion indicate adequate calcium and vitamin D supplementation.6

 

 

 

Dumping syndrome can lead to hypoglycemia

Dumping syndrome is a common complication following BS, with prevalence ranging from 25% to 75%, depending upon the type of procedure performed.16,17 There are 2 types: early and late. Early dumping syndrome occurs within 30 minutes of eating. Symptoms are related to the robust release of gastrointestinal hormones caused by rapid gastric emptying. Symptoms include nausea, abdominal pain, diarrhea, flushing, hypotension, and tachycardia.

Late dumping is characterized as postprandial hypoglycemia occurring one to 3 hours after eating. Late dumping is likely caused by a combination of changes within the pancreatic beta cells and abnormal insulin response to glucose.16-18 Rapid gastric emptying leads to rapid release of glucose in the gut, which, in turn, leads to brisk insulin secretion. Since glucose is absorbed faster than insulin’s half-life, the resulting (relatively) high levels of insulin may cause hypoglycemia.16-18

Sigstad’s scoring system can be used to confirm suspected cases of dumping syndrome (TABLE 316,17,19). A diagnosis can also be made with an oral glucose challenge in which pulse, blood pressure, glucose, and hematocrit are measured after ingestion of 50 g glucose. The test is positive if heart rate increases by 10 beats per minute, hematocrit increases by 3% 30 minutes after ingestion, or glucose falls below 60 mg/dL 2 to 3 hours after ingestion.17

First-line treatment of dumping syndrome consists of dietary modifications. The goal is to slow the rate of gastric emptying by eating smaller, more frequent meals; separating beverages from food; decreasing carbohydrates; and increasing fiber and protein content.

If results are suboptimal after dietary changes, medications can be prescribed including acarbose to prevent postprandial hypoglycemia; anticholinergics such as dicyclomine to slow gastric emptying; and somatostatin to decrease gastric emptying and inhibit GI hormone release.17 Lastly, for resistant and severe postprandial hypoglycemia, a few patients have undergone pancreatectomy, but only about 65% experienced improvement in symptoms and 12% developed diabetes post-surgically.20

Gout attacks may initially increase, but then decrease

BS affects the incidence of gout attacks in patients with a history of gout. One comparative study of approximately 150 patients demonstrated that those with a history of gout had significantly more gout attacks in the first month after BS compared with obese patients with a history of gout undergoing other upper GI surgeries.21 There was no difference between malabsorptive and restrictive procedures. But after the first month, BS patients had significantly fewer gout attacks and lower uric acid levels than their obese counterparts.21

Protein rich diets, catabolism potentiated by aggressive caloric restriction following BS, and dehydration contribute to the initial increase. Therefore, patients who have had at least one gout attack in the year prior to surgery or who are on hypouricemic medication may benefit from at least one month of prophylactic therapy (eg, allopurinol and colchicine) after surgery.

GERD and ulceration: How to respond

Obesity is a known risk factor for GERD, but the effect of BS on GERD is uncertain and seems to vary with the procedure performed. RYGB decreases GERD and is, therefore, used as both a secondary treatment in those not responding to medications and a revision treatment for fundoplication and other types of BSs. Sleeve gastrectomy and adjustable gastric banding have mixed effects on GERD. A systematic review by de Jong et al revealed a decreased prevalence of reflux symptoms and GERD medication use after LAGB; however, during longer follow-up, 15% of previously unaffected patients reported experiencing GERD.22 The 2011 International Sleeve Gastrectomy Expert Panel Consensus Statement retrospectively noted a postoperative incidence of GERD as high as 31%.23

BS patients with GERD should be treated with a proton pump inhibitor. If this fails, refer patients to a gastroenterologist for further evaluation.24

Ulcers after BS may be an indication for revision surgery. Data are mixed regarding increased risk of marginal ulceration from nonsteroidal anti-inflammatory drug (NSAID) use, but NSAIDs have been linked to an increased risk of anastomotic leakage.25-28 Thus, it seems prudent to avoid NSAIDs in people who have undergone BS.

Keeping watch over psychiatric comorbidities

A recent meta-analysis by Dawes et al29 showed that about 23% of patients pursuing BS have a comorbid mood disorder. Specifically, the preoperative prevalence of depression (19%) and binge-eating disorder (17%) were found to be higher than rates in the general population.29 The meta-analysis found improvement in the prevalence of depression with fewer symptoms and less antidepressant medication use in the first 3 years after surgery and a decrease in the rate of binge-eating disorder, although there were fewer supporting data for the latter. These findings were observed with both restrictive and malabsorptive procedures.

 

 

 

The data are mixed regarding rates of alcohol abuse and suicide. Further research is necessary in this field. Patients who have had BS should receive ongoing psychiatric and psychological care from a multidisciplinary team as a matter of course.

Will a second surgery be needed?

Revision surgery. In 2015, about 14% of the almost 200,000 BSs performed were revisions.4 Revision surgery is indicated in BS patients with weight regain, recurrent comorbid diseases (eg, diabetes, hypertension), or complications of primary BS. Restrictive procedures have a higher revision rate than malabsorptive procedures, primarily due to a higher rate of weight regain.6,30

Because revision surgery is associated with more complications and possibly longer hospital stays than primary BS, it should be performed by a bariatric surgeon with extensive experience.30,31 Restrictive revisions are typically converted to malabsorptive procedures. Cost is a limiting factor as many patients’ insurance coverage is limited to one BS per lifetime.

Body contouring. Body contouring surgery (BCS) can improve physical and mental well-being and may be a protective factor for weight regain after bariatric surgery.32 Despite its desirability—particularly to women, adolescents, and those with large decreases in body mass index (BMI)—few patients can afford BCS since it is rarely covered by insurance.

Complications of BCS vary, but are most commonly infection and wound dehiscence. This is, in part, due to poorer wound healing in BS patients compared to those with nonsurgical massive weight loss. The cause of poor wound healing is thought to be secondary to nutritional deficiencies and the catabolic state induced by post-surgical weight loss. Recommendations for BCS include weight stability for more than one year after BS, age >16 years, excess skin causing significant functional impairment, non-smoking status, and presence of good social support.33

Bariatric surgery in adolescents is on the rise

Children in the highest body mass index quartile have more than twice the death rate of those in the lowest BMI quartile.34 Thus, it is not surprising that the rate of BS in adolescents is increasing.7 BS in this age group is successful for weight loss and improvement of comorbid conditions, with relatively low complication rates.35 Options include malabsorptive and restrictive procedures, although gastric banding has not been approved by the US Food and Drug Administration for patients under the age of 18 years.

Monitor vitamin D levels 2 and 4 weeks after initiation of treatment and every 3 months thereafter.After BS, adolescent girls should be counseled regarding the possibility of pregnancy (restoration of fertility) and appropriate contraception. Adolescent patients require nutritional supplementation after BS as indicated in TABLE 1.6,8-11

When determining which adolescents to refer for BS, we recommend the following criteria: 35-38

  • failure of a minimum 6-month trial of a staged treatment approach, as recommended by Barlow et al,36 including diet, exercise, and pharmacologic treatment
  • BMI 35 with type 2 diabetes or severe sleep apnea (apnea hypopnea index [AHI] >15)37
  • BMI 40 with mild sleep apnea (AHI >5), hypertension, or pre-diabetes37
  • Tanner stage IV or V
  • at least 95% skeletal growth (for malabsorptive surgery).37 This can be determined using an estimated adult height from mid-parental height formula and assessing growth plate closure with hand radiographs for bone age
  • appropriate maturity level permitting adherence
  • good psychological support
  • a multidisciplinary team for postoperative and long-term follow-up care.

Planning for the future: Exploring the possibility of pregnancy

Obesity is the primary cause of maternal and fetal morbidity during pregnancy. It is associated with increased rates of early miscarriage, congenital defects, macrosomia, and fetal death. Maternal risks of obesity include: gestational hypertension, gestational diabetes mellitus (GDM), and pre-eclampsia. Obese mothers also have a higher incidence of failed induction, caesarean section, and breastfeeding failure.10,39 Given that half of all BSs are performed in women of reproductive age, this population deserves special consideration.10

A recent meta-analysis by Galazis et al40 concluded that BS performed prior to pregnancy led to decreased rates of preeclampsia, GDM, large neonates, preterm birth, and neonatal intensive care unit admission. Perinatal mortality did not increase after BS. However, BS led to higher rates of maternal anemia. There was no significant difference between groups in incidence of cesarean section.

The post BS female patient should be advised to use a reliable form of contraception for a minimum of 12 to 18 months after surgery.6,10,39 Involve high-risk obstetric specialists during pregnancies. Diet should be supplemented as indicated in TABLE 1.6,8-11

CORRESPONDENCE
Amy Rothberg, MD, PhD, Domino’s Farms, Lobby G, Suite 1500, 24 Frank Lloyd Wright Drive, Ann Arbor, MI 48106; [email protected].

 

More than one-third of American adults and approximately 17% of children and adolescents between the ages of 2 and 19 years are obese.1,2 Poor diet coupled with a sedentary lifestyle is the highest ranked cause of non-communicable disease and a leading cause of preventable death, according to the National Research Council.3

Bariatric surgery (BS) is a viable therapeutic option for obese patients who do not respond to conventional lifestyle interventions for losing weight. There are multiple gastrointestinal (GI) procedures available that are classified as either malabsorptive (Roux-en-Y gastric bypass [RYGB] and biliopancreatic diversion [BPD] with or without duodenal switch) or restrictive (laparoscopic adjustable gastric banding [LAGB] and vertical sleeve gastrectomy [VSG]).

Approximately half of the 196,000 bariatric procedures performed in the United States in 2015 were of the sleeve variety, another 23% were RYGB, and the remaining percentage was divided among the other types.4 Postoperative risks include nutritional deficiencies, decreased bone mineral density (BMD), dumping syndrome (when food rapidly dumps from the stomach to the intestine), and gastroesophageal reflux disease (GERD) with possible ulceration.

Despite these potential complications, a systematic review and meta-analysis found that obese people who underwent BS (gastric banding or gastric bypass) had significantly reduced risks of global, non-cardiovascular (CV), and CV mortality compared with obese controls.5 Helping patients to realize these benefits requires that the entire health care team—especially the family physician—is aware of the special considerations for this population.

Patients undergoing bariatric surgery require routine lifetime screening for nutritional deficiencies.

To that end, this article reviews the details of diagnosing and managing post-surgical complications. It also addresses issues unique to managing certain subpopulations, such as post-BS patients who require revision surgery or who want to pursue body contouring surgery; adolescents who undergo BS surgery; and women who want to get pregnant postoperatively.

Monitor patients for these post-surgery complications

Postoperative BS follow-up varies depending on location, surgeon preference, and availability of multidisciplinary resources. At our institution, patients have a minimum of 3 follow-up visits with their surgeon (during hospitalization and 2 weeks and 2 months postoperatively). This is followed by visits with Endocrinology 6 months after surgery and annually thereafter. Given the variability of follow-up, family physicians should coordinate with specialists where appropriate and be aware of postoperative complications and monitoring since it is likely they will have the most frequent contact with these patients.

Nutritional deficiencies are common and require lifelong screening

Nutritional deficiencies are the most common complications of malabsorptive BS. Guidelines from the Endocrine Society, as well as guidelines from the American Association of Clinical Endocrinologists (AACE), The Obesity Society (TOS), and the American Society for Metabolic and Bariatric Surgery (ASMBS), recommend routine lifetime screening for deficiencies after surgery.6,7 Complete blood cell count, electrolytes, glucose, creatinine, and liver function tests should be obtained at one, 3, 6, 12, 18, and 24 months following surgery and annually thereafter.6

Multiple factors contribute to nutritional and micronutrient deficiencies, including reduced oral intake of food, decreased GI absorption, food intolerance, nausea/vomiting, and nonadherence with dietary supplements.8 Oral supplementation should be in chewable, powder, or liquid form because pill and capsule absorption may be altered.8,9 Over-the-counter multivitamins may not contain the requisite daily doses recommended after BS.9 Patients and physicians should evaluate supplements together to ensure appropriate nutritional and micronutrient supplementation (TABLE 16,8-11).

Bone mineral density can start to decrease soon after surgery

Studies evaluating BMD after BS have produced variable findings. In obese patients, dual-energy x-ray absorptiometry (DEXA) measurements may not be accurate due to adipose tissue artifact and table weight limits. In addition, limited data exist on the incidence of fractures after BS. Of 2 notable studies, only one, a population-based study involving 258 Minnesota residents who underwent a first bariatric surgery between 1985 and 2004, demonstrated a significantly increased incidence of fractures.12,13

In addition, studies show bone turnover markers, including C-terminal telopeptide, increase as early as 3 months after BS.14 Several guidelines recommend routine BMD screening after BS (TABLE 2).6,7 The mechanism of bone demineralization is likely multifactorial—a function of the magnitude of the weight loss and skeletal unloading, calcium and vitamin D deficiencies, and associated secondary hyperparathyroidism.15 Treatment for secondary hyperparathyroidism is adequate supplementation with vitamin D and calcium.

Optimal dosing for vitamin D has not been determined. One recent systematic review suggests routine prophylaxis with at least 2000 international units (IU)/d and found the greatest improvement for known deficiency with doses of 1500-9100 IU/d following malabsorptive surgeries.11 After laparoscopic sleeve gastrectomy, at least 1000 IU/d vitamin D is recommended.11

Overall, high variability exists among patients, and an individualized approach for dosing is recommended.11 Vitamin D levels should be monitored 2 and 4 weeks after initiation of treatment and every 3 months thereafter.11 Normal levels of serum calcium, 25-OH vitamin D, bone-specific alkaline phosphatase, and 24-hour urinary calcium excretion indicate adequate calcium and vitamin D supplementation.6

 

 

 

Dumping syndrome can lead to hypoglycemia

Dumping syndrome is a common complication following BS, with prevalence ranging from 25% to 75%, depending upon the type of procedure performed.16,17 There are 2 types: early and late. Early dumping syndrome occurs within 30 minutes of eating. Symptoms are related to the robust release of gastrointestinal hormones caused by rapid gastric emptying. Symptoms include nausea, abdominal pain, diarrhea, flushing, hypotension, and tachycardia.

Late dumping is characterized as postprandial hypoglycemia occurring one to 3 hours after eating. Late dumping is likely caused by a combination of changes within the pancreatic beta cells and abnormal insulin response to glucose.16-18 Rapid gastric emptying leads to rapid release of glucose in the gut, which, in turn, leads to brisk insulin secretion. Since glucose is absorbed faster than insulin’s half-life, the resulting (relatively) high levels of insulin may cause hypoglycemia.16-18

Sigstad’s scoring system can be used to confirm suspected cases of dumping syndrome (TABLE 316,17,19). A diagnosis can also be made with an oral glucose challenge in which pulse, blood pressure, glucose, and hematocrit are measured after ingestion of 50 g glucose. The test is positive if heart rate increases by 10 beats per minute, hematocrit increases by 3% 30 minutes after ingestion, or glucose falls below 60 mg/dL 2 to 3 hours after ingestion.17

First-line treatment of dumping syndrome consists of dietary modifications. The goal is to slow the rate of gastric emptying by eating smaller, more frequent meals; separating beverages from food; decreasing carbohydrates; and increasing fiber and protein content.

If results are suboptimal after dietary changes, medications can be prescribed including acarbose to prevent postprandial hypoglycemia; anticholinergics such as dicyclomine to slow gastric emptying; and somatostatin to decrease gastric emptying and inhibit GI hormone release.17 Lastly, for resistant and severe postprandial hypoglycemia, a few patients have undergone pancreatectomy, but only about 65% experienced improvement in symptoms and 12% developed diabetes post-surgically.20

Gout attacks may initially increase, but then decrease

BS affects the incidence of gout attacks in patients with a history of gout. One comparative study of approximately 150 patients demonstrated that those with a history of gout had significantly more gout attacks in the first month after BS compared with obese patients with a history of gout undergoing other upper GI surgeries.21 There was no difference between malabsorptive and restrictive procedures. But after the first month, BS patients had significantly fewer gout attacks and lower uric acid levels than their obese counterparts.21

Protein rich diets, catabolism potentiated by aggressive caloric restriction following BS, and dehydration contribute to the initial increase. Therefore, patients who have had at least one gout attack in the year prior to surgery or who are on hypouricemic medication may benefit from at least one month of prophylactic therapy (eg, allopurinol and colchicine) after surgery.

GERD and ulceration: How to respond

Obesity is a known risk factor for GERD, but the effect of BS on GERD is uncertain and seems to vary with the procedure performed. RYGB decreases GERD and is, therefore, used as both a secondary treatment in those not responding to medications and a revision treatment for fundoplication and other types of BSs. Sleeve gastrectomy and adjustable gastric banding have mixed effects on GERD. A systematic review by de Jong et al revealed a decreased prevalence of reflux symptoms and GERD medication use after LAGB; however, during longer follow-up, 15% of previously unaffected patients reported experiencing GERD.22 The 2011 International Sleeve Gastrectomy Expert Panel Consensus Statement retrospectively noted a postoperative incidence of GERD as high as 31%.23

BS patients with GERD should be treated with a proton pump inhibitor. If this fails, refer patients to a gastroenterologist for further evaluation.24

Ulcers after BS may be an indication for revision surgery. Data are mixed regarding increased risk of marginal ulceration from nonsteroidal anti-inflammatory drug (NSAID) use, but NSAIDs have been linked to an increased risk of anastomotic leakage.25-28 Thus, it seems prudent to avoid NSAIDs in people who have undergone BS.

Keeping watch over psychiatric comorbidities

A recent meta-analysis by Dawes et al29 showed that about 23% of patients pursuing BS have a comorbid mood disorder. Specifically, the preoperative prevalence of depression (19%) and binge-eating disorder (17%) were found to be higher than rates in the general population.29 The meta-analysis found improvement in the prevalence of depression with fewer symptoms and less antidepressant medication use in the first 3 years after surgery and a decrease in the rate of binge-eating disorder, although there were fewer supporting data for the latter. These findings were observed with both restrictive and malabsorptive procedures.

 

 

 

The data are mixed regarding rates of alcohol abuse and suicide. Further research is necessary in this field. Patients who have had BS should receive ongoing psychiatric and psychological care from a multidisciplinary team as a matter of course.

Will a second surgery be needed?

Revision surgery. In 2015, about 14% of the almost 200,000 BSs performed were revisions.4 Revision surgery is indicated in BS patients with weight regain, recurrent comorbid diseases (eg, diabetes, hypertension), or complications of primary BS. Restrictive procedures have a higher revision rate than malabsorptive procedures, primarily due to a higher rate of weight regain.6,30

Because revision surgery is associated with more complications and possibly longer hospital stays than primary BS, it should be performed by a bariatric surgeon with extensive experience.30,31 Restrictive revisions are typically converted to malabsorptive procedures. Cost is a limiting factor as many patients’ insurance coverage is limited to one BS per lifetime.

Body contouring. Body contouring surgery (BCS) can improve physical and mental well-being and may be a protective factor for weight regain after bariatric surgery.32 Despite its desirability—particularly to women, adolescents, and those with large decreases in body mass index (BMI)—few patients can afford BCS since it is rarely covered by insurance.

Complications of BCS vary, but are most commonly infection and wound dehiscence. This is, in part, due to poorer wound healing in BS patients compared to those with nonsurgical massive weight loss. The cause of poor wound healing is thought to be secondary to nutritional deficiencies and the catabolic state induced by post-surgical weight loss. Recommendations for BCS include weight stability for more than one year after BS, age >16 years, excess skin causing significant functional impairment, non-smoking status, and presence of good social support.33

Bariatric surgery in adolescents is on the rise

Children in the highest body mass index quartile have more than twice the death rate of those in the lowest BMI quartile.34 Thus, it is not surprising that the rate of BS in adolescents is increasing.7 BS in this age group is successful for weight loss and improvement of comorbid conditions, with relatively low complication rates.35 Options include malabsorptive and restrictive procedures, although gastric banding has not been approved by the US Food and Drug Administration for patients under the age of 18 years.

Monitor vitamin D levels 2 and 4 weeks after initiation of treatment and every 3 months thereafter.After BS, adolescent girls should be counseled regarding the possibility of pregnancy (restoration of fertility) and appropriate contraception. Adolescent patients require nutritional supplementation after BS as indicated in TABLE 1.6,8-11

When determining which adolescents to refer for BS, we recommend the following criteria: 35-38

  • failure of a minimum 6-month trial of a staged treatment approach, as recommended by Barlow et al,36 including diet, exercise, and pharmacologic treatment
  • BMI 35 with type 2 diabetes or severe sleep apnea (apnea hypopnea index [AHI] >15)37
  • BMI 40 with mild sleep apnea (AHI >5), hypertension, or pre-diabetes37
  • Tanner stage IV or V
  • at least 95% skeletal growth (for malabsorptive surgery).37 This can be determined using an estimated adult height from mid-parental height formula and assessing growth plate closure with hand radiographs for bone age
  • appropriate maturity level permitting adherence
  • good psychological support
  • a multidisciplinary team for postoperative and long-term follow-up care.

Planning for the future: Exploring the possibility of pregnancy

Obesity is the primary cause of maternal and fetal morbidity during pregnancy. It is associated with increased rates of early miscarriage, congenital defects, macrosomia, and fetal death. Maternal risks of obesity include: gestational hypertension, gestational diabetes mellitus (GDM), and pre-eclampsia. Obese mothers also have a higher incidence of failed induction, caesarean section, and breastfeeding failure.10,39 Given that half of all BSs are performed in women of reproductive age, this population deserves special consideration.10

A recent meta-analysis by Galazis et al40 concluded that BS performed prior to pregnancy led to decreased rates of preeclampsia, GDM, large neonates, preterm birth, and neonatal intensive care unit admission. Perinatal mortality did not increase after BS. However, BS led to higher rates of maternal anemia. There was no significant difference between groups in incidence of cesarean section.

The post BS female patient should be advised to use a reliable form of contraception for a minimum of 12 to 18 months after surgery.6,10,39 Involve high-risk obstetric specialists during pregnancies. Diet should be supplemented as indicated in TABLE 1.6,8-11

CORRESPONDENCE
Amy Rothberg, MD, PhD, Domino’s Farms, Lobby G, Suite 1500, 24 Frank Lloyd Wright Drive, Ann Arbor, MI 48106; [email protected].

References

1. Centers for Disease Control and Prevention. Overweight and obesity. Adult obesity facts. Available at: https://www.cdc.gov/obesity/data/adult.html. Accessed April 5, 2017.

2. Centers for Disease Control and Prevention. Overweight and obesity. Childhood obesity facts. Available at: https://www.cdc.gov/obesity/data/childhood.html. Accessed April 5, 2017.

3. McGinnis JM. Actual causes of death, 1990-2010. Workshop on Determinants of Premature Mortality, September 18, 2013, National Research Council, Washington, DC.

4. American Society of Metabolic and Bariatric Surgery. Estimate of bariatric surgery numbers, 2011-2015. Available at: https://asmbs.org/resources/estimate-of-bariatric-surgery-numbers. Accessed April 5, 2017.

5. Pontiroli AE, Morabito A. Long-term prevention of mortality in morbid obesity through bariatric surgery. a systematic review and meta-analysis of trials performed with gastric banding and gastric bypass. Ann Surg. 2011;253:484-487.

6. Heber D, Greenway FL, Kaplan LM, et al. Endocrine and nutritional management of the post-bariatric surgery patient: an Endocrine Society clinical practice guideline. J Clin Endocrinol Metab. 2010;95:4823-4843.

7. Mechanick JI, Youdim A, Jones DB, et al. Clinical practice guidelines for the perioperative nutritional, metabolic, and nonsurgical support of the bariatric surgery patient—2013 update: cosponsored by American Association of Clinical Endocrinologists, The Obesity Society, and American Society for Metabolic & Bariatric Surgery. Obesity. 2013;21:S1-S27.

8. Stein J, Stier C, Raab H, et al. Review article: the nutritional and pharmacological consequences of obesity surgery. Aliment Pharmacol Ther. 2014;40:582-609.

9. Boyce SG, Goriparthi R, Clark J, et al. Can composite nutritional supplement based on the current guidelines prevent vitamin and mineral deficiency after weight loss surgery? Obes Surg. 2016;26:966-971.

10. Beard JH, Bell RL, Duffy AJ. Reproductive considerations and pregnancy after bariatric surgery: current evidence and recommendations. Obes Surg. 2008;18:1023-1027.

11. Chakhtoura MT, Nakhoul NN, Shawwa K, et al. Hypovitaminosis D in bariatric surgery: a systematic review of observational studies. Metabolism. 2016;65:574-585.

12. Nakamura KM, Haglind EG, Clowes JA, et al. Fracture risk following bariatric surgery: a population-based study. Osteoporosis Int. 2014;25:151-158.

13. Lalmohamed A, de Vries F, Bazelier MT, et al. Risk of fracture after bariatric surgery in the United Kingdom: population-based, retrospective cohort study. BMJ. 2012;345:e5085.

14. Coates PS, Fernstrom JD, Fernstrom MH, et al. Gastric bypass surgery for morbid obesity leads to an increase in bone turnover and a decrease in bone mass. J Clin Endocrinol Metab. 2004;89:1061-1065.

15. Stein EM, Silverberg SJ. Bone loss after bariatric surgery: causes, consequences and management. Lancet Diabetes Endocrinol. 2014;2:165-174.

16. Tack J, Deloose E. Complications of bariatric surgery: dumping syndrome, reflux and vitamin deficiencies. Best Pract Res Clin Gastroenterol. 2014;28:741-749.

17. Berg P, McCallum R. Dumping syndrome: a review of the current concepts of pathophysiology, diagnosis, and treatment. Dig Dis Sci. 2016;61:11-18.

18. Ritz P, Vaurs C, Barigou M, et al. Hypoglycaemia after gastric bypass: mechanisms and treatment. Diabetes Obes Metab. 2016;18:217-223.

19. Sigstad H. A clinical diagnostic index in the diagnosis of the dumping syndrome. Changes in plasma volume and blood sugar after a test meal. Acta Med Scand. 1970;188:479-486.

20. Mala T. Postprandial hyperinsulinemic hypoglycaemia after gastric bypass surgical treatment. Surg Obes Relat Dis. 2014;10:1220-1225.

21. Romero-Talamás H, Daigle CR, Aminian A. The effect of bariatric surgery on gout: a comparative study. Surg Obes Relat Dis. 2014;10:1161-1165.

22. de Jong JR, Besselink MG, van Ramshorst B, et al. Effects of adjustable gastric banding on gastroesophageal reflux and esophageal motility: a systematic review. Obes Rev. 2010;11:297-305.

23. Rosenthal RJ; International Sleeve Gastrectomy Expert Panel. International Sleeve Gastrectomy Expert Panel Consensus Statement: best practice guidelines based on experience of >12,000 cases. Surg Obes Relat Dis. 2012;8:8-19.

24. Altieri MS, Pryor AD. Gastroesophageal reflux disease after bariatric procedures. Surg Clin North Am. 2015;95:579-591.

25. Hakkarainen TW, Steele SR, Bastaworous A, et al. Nonsteroidal anti-inflammatory drugs and the risk for anastomotic failure: a report from Washington State’s Surgical Care and Outcomes Assessment Program (SCOAP). JAMA Surg. 2015;150:223-228.

26. El-Hayek K, Timratana P, Shimizu H, et al. Marginal ulcer after Roux-en-Y gastric bypass: what have we really learned? Surg Endosc. 2012;26:2789-2796.

27. Sverdén E, Mattsson F, Sondén AM, et al. Risk factors for marginal ulcer after gastric bypass surgery for obesity: a population-based cohort study. Ann Surg. 2016;263:733-737.

28. Azagury DE, Abu Dayyeh BK, Greenwalt IT, et al. Marginal ulceration after Roux-en-Y gastric bypass surgery: characteristics, risk factors, treatment, and outcomes. Endoscopy. 2011;43:950-954.

29. Dawes AJ, Maggard-Gibbons M, Maher AR, et al. Mental health conditions among patients seeking and undergoing bariatric surgery: A meta-analysis. JAMA. 2016;315:150-163.

30. Ferrer-Márquez MP, Belda-Lozano R, Solvas-Salmerón MJ, et al. Revisional surgery after laparoscopic sleeve gastrectomy. Surg Laparosc Endosc Percutan Tech. 2015;25:6-9.

31. Sanchez H, Cabrera A, Cabrera K, et al. Laparoscopic Roux-en-Y gastric bypass as a revision procedure after restrictive bariatric surgery. Obes Surg. 2008;18:1539-1543.

32. van der Beek ES, Te Riele W, Specken TF, et al. The impact of reconstructive procedures following bariatric surgery on patient well-being and quality of life. Obes Surg. 2010;20:36-41.

 

 

33. Ellison JM, Steffen KJ, Sarwer DB. Body contouring after bariatric surgery. Eur Eat Disord Rev. 2015;23:479-487.

34. Franks PW, Hanson RL, Knowler WC, et al. Childhood obesity, other cardiovascular risk factors, and premature death. NEJM. 2010;362:485-489.

35. Gravelle BL, Broyles M. Interventions of weight reduction and prevention in children and adolescents: update. Am J Ther. 2015;22:159-166.

36. Barlow SE. Expert committee recommendations regarding the prevention, assessment, and treatment of child and adolescent overweight and obesity: summary report. Pediatrics. 2007;120:S164-S192.

37. Pratt JS, Lenders CM, Dionne EA, et al. Best practice updates for pediatric/adolescent weight loss surgery. Obesity. 2009;17:901-910.

38. Nogueira I, Hrovat K. Adolescent bariatric surgery: review on nutrition considerations. Nutr Clin Pract. 2014;29:740-746.

39. Nicklas JM, Barbour LA. Optimizing weight for maternal and infant health: tenable, or too late? Expert Rev Endocrinol Metab. 2015;10:227-242.

40. Galazis N, Docheva N, Simillis C, et al. Maternal and neonatal outcomes in women undergoing bariatric surgery: a systematic review and meta-analysis. Eur J Obstet Gynecol Reprod Biol. 2014;181:45-53.

References

1. Centers for Disease Control and Prevention. Overweight and obesity. Adult obesity facts. Available at: https://www.cdc.gov/obesity/data/adult.html. Accessed April 5, 2017.

2. Centers for Disease Control and Prevention. Overweight and obesity. Childhood obesity facts. Available at: https://www.cdc.gov/obesity/data/childhood.html. Accessed April 5, 2017.

3. McGinnis JM. Actual causes of death, 1990-2010. Workshop on Determinants of Premature Mortality, September 18, 2013, National Research Council, Washington, DC.

4. American Society of Metabolic and Bariatric Surgery. Estimate of bariatric surgery numbers, 2011-2015. Available at: https://asmbs.org/resources/estimate-of-bariatric-surgery-numbers. Accessed April 5, 2017.

5. Pontiroli AE, Morabito A. Long-term prevention of mortality in morbid obesity through bariatric surgery. a systematic review and meta-analysis of trials performed with gastric banding and gastric bypass. Ann Surg. 2011;253:484-487.

6. Heber D, Greenway FL, Kaplan LM, et al. Endocrine and nutritional management of the post-bariatric surgery patient: an Endocrine Society clinical practice guideline. J Clin Endocrinol Metab. 2010;95:4823-4843.

7. Mechanick JI, Youdim A, Jones DB, et al. Clinical practice guidelines for the perioperative nutritional, metabolic, and nonsurgical support of the bariatric surgery patient—2013 update: cosponsored by American Association of Clinical Endocrinologists, The Obesity Society, and American Society for Metabolic & Bariatric Surgery. Obesity. 2013;21:S1-S27.

8. Stein J, Stier C, Raab H, et al. Review article: the nutritional and pharmacological consequences of obesity surgery. Aliment Pharmacol Ther. 2014;40:582-609.

9. Boyce SG, Goriparthi R, Clark J, et al. Can composite nutritional supplement based on the current guidelines prevent vitamin and mineral deficiency after weight loss surgery? Obes Surg. 2016;26:966-971.

10. Beard JH, Bell RL, Duffy AJ. Reproductive considerations and pregnancy after bariatric surgery: current evidence and recommendations. Obes Surg. 2008;18:1023-1027.

11. Chakhtoura MT, Nakhoul NN, Shawwa K, et al. Hypovitaminosis D in bariatric surgery: a systematic review of observational studies. Metabolism. 2016;65:574-585.

12. Nakamura KM, Haglind EG, Clowes JA, et al. Fracture risk following bariatric surgery: a population-based study. Osteoporosis Int. 2014;25:151-158.

13. Lalmohamed A, de Vries F, Bazelier MT, et al. Risk of fracture after bariatric surgery in the United Kingdom: population-based, retrospective cohort study. BMJ. 2012;345:e5085.

14. Coates PS, Fernstrom JD, Fernstrom MH, et al. Gastric bypass surgery for morbid obesity leads to an increase in bone turnover and a decrease in bone mass. J Clin Endocrinol Metab. 2004;89:1061-1065.

15. Stein EM, Silverberg SJ. Bone loss after bariatric surgery: causes, consequences and management. Lancet Diabetes Endocrinol. 2014;2:165-174.

16. Tack J, Deloose E. Complications of bariatric surgery: dumping syndrome, reflux and vitamin deficiencies. Best Pract Res Clin Gastroenterol. 2014;28:741-749.

17. Berg P, McCallum R. Dumping syndrome: a review of the current concepts of pathophysiology, diagnosis, and treatment. Dig Dis Sci. 2016;61:11-18.

18. Ritz P, Vaurs C, Barigou M, et al. Hypoglycaemia after gastric bypass: mechanisms and treatment. Diabetes Obes Metab. 2016;18:217-223.

19. Sigstad H. A clinical diagnostic index in the diagnosis of the dumping syndrome. Changes in plasma volume and blood sugar after a test meal. Acta Med Scand. 1970;188:479-486.

20. Mala T. Postprandial hyperinsulinemic hypoglycaemia after gastric bypass surgical treatment. Surg Obes Relat Dis. 2014;10:1220-1225.

21. Romero-Talamás H, Daigle CR, Aminian A. The effect of bariatric surgery on gout: a comparative study. Surg Obes Relat Dis. 2014;10:1161-1165.

22. de Jong JR, Besselink MG, van Ramshorst B, et al. Effects of adjustable gastric banding on gastroesophageal reflux and esophageal motility: a systematic review. Obes Rev. 2010;11:297-305.

23. Rosenthal RJ; International Sleeve Gastrectomy Expert Panel. International Sleeve Gastrectomy Expert Panel Consensus Statement: best practice guidelines based on experience of >12,000 cases. Surg Obes Relat Dis. 2012;8:8-19.

24. Altieri MS, Pryor AD. Gastroesophageal reflux disease after bariatric procedures. Surg Clin North Am. 2015;95:579-591.

25. Hakkarainen TW, Steele SR, Bastaworous A, et al. Nonsteroidal anti-inflammatory drugs and the risk for anastomotic failure: a report from Washington State’s Surgical Care and Outcomes Assessment Program (SCOAP). JAMA Surg. 2015;150:223-228.

26. El-Hayek K, Timratana P, Shimizu H, et al. Marginal ulcer after Roux-en-Y gastric bypass: what have we really learned? Surg Endosc. 2012;26:2789-2796.

27. Sverdén E, Mattsson F, Sondén AM, et al. Risk factors for marginal ulcer after gastric bypass surgery for obesity: a population-based cohort study. Ann Surg. 2016;263:733-737.

28. Azagury DE, Abu Dayyeh BK, Greenwalt IT, et al. Marginal ulceration after Roux-en-Y gastric bypass surgery: characteristics, risk factors, treatment, and outcomes. Endoscopy. 2011;43:950-954.

29. Dawes AJ, Maggard-Gibbons M, Maher AR, et al. Mental health conditions among patients seeking and undergoing bariatric surgery: A meta-analysis. JAMA. 2016;315:150-163.

30. Ferrer-Márquez MP, Belda-Lozano R, Solvas-Salmerón MJ, et al. Revisional surgery after laparoscopic sleeve gastrectomy. Surg Laparosc Endosc Percutan Tech. 2015;25:6-9.

31. Sanchez H, Cabrera A, Cabrera K, et al. Laparoscopic Roux-en-Y gastric bypass as a revision procedure after restrictive bariatric surgery. Obes Surg. 2008;18:1539-1543.

32. van der Beek ES, Te Riele W, Specken TF, et al. The impact of reconstructive procedures following bariatric surgery on patient well-being and quality of life. Obes Surg. 2010;20:36-41.

 

 

33. Ellison JM, Steffen KJ, Sarwer DB. Body contouring after bariatric surgery. Eur Eat Disord Rev. 2015;23:479-487.

34. Franks PW, Hanson RL, Knowler WC, et al. Childhood obesity, other cardiovascular risk factors, and premature death. NEJM. 2010;362:485-489.

35. Gravelle BL, Broyles M. Interventions of weight reduction and prevention in children and adolescents: update. Am J Ther. 2015;22:159-166.

36. Barlow SE. Expert committee recommendations regarding the prevention, assessment, and treatment of child and adolescent overweight and obesity: summary report. Pediatrics. 2007;120:S164-S192.

37. Pratt JS, Lenders CM, Dionne EA, et al. Best practice updates for pediatric/adolescent weight loss surgery. Obesity. 2009;17:901-910.

38. Nogueira I, Hrovat K. Adolescent bariatric surgery: review on nutrition considerations. Nutr Clin Pract. 2014;29:740-746.

39. Nicklas JM, Barbour LA. Optimizing weight for maternal and infant health: tenable, or too late? Expert Rev Endocrinol Metab. 2015;10:227-242.

40. Galazis N, Docheva N, Simillis C, et al. Maternal and neonatal outcomes in women undergoing bariatric surgery: a systematic review and meta-analysis. Eur J Obstet Gynecol Reprod Biol. 2014;181:45-53.

Issue
The Journal of Family Practice - 66(6)
Issue
The Journal of Family Practice - 66(6)
Page Number
356-363
Page Number
356-363
Publications
Publications
Topics
Article Type
Display Headline
Post-bariatric surgery patients: Your role in their long-term care
Display Headline
Post-bariatric surgery patients: Your role in their long-term care
Sections
Article Source

From The Journal of Family Practice | 2017;66(6):356-363.

Inside the Article

PRACTICE RECOMMENDATIONS

› Routinely screen bariatric surgery patients for nutritional deficiencies throughout their life. A

› Avoid the use of nonsteroidal anti-inflammatory medications in patients who have had bariatric surgery becauseof the risk of anastomotic ulceration and leakage. A

› Consider revision surgery for bariatric surgery patients with weight regain, recurrent comorbid diseases, or surgical complications. B

› Counsel obese women who want to become pregnant that bariatric surgery decreases rates of future pregnancy complications. B

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

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
PubMed ID
28574521
Article PDF Media

Oral agent offers relief from generalized hyperhidrosis

Article Type
Changed
Tue, 06/23/2020 - 12:37
Display Headline
Oral agent offers relief from generalized hyperhidrosis
 

ILLUSTRATIVE CASE

A 34-year-old woman presents to your office for unbearable sweating. She notes that the sweating occurs nearly daily on her hands, face, and in her axillary regions, causing social embarrassment. She has tried multiple antiperspirants to no avail. Is there anything she can take to reduce the sweating?

Hyperhidrosis is a common, self-limiting problem affecting 2% to 3% of the population in the United States.2 Patients may complain of localized sweating of the hands, feet, face, or underarms or more systemic, generalized sweating in multiple locations. Either way, patients always note a significant impact on their quality of life.

Treatment of hyperhidrosis has traditionally focused on topical therapies to the affected areas. Research has shown that localized treatment with antiperspirants containing aluminum salt is effective by both subjective report and objective measurements at reducing sweating—particularly in the axilla, hands, and feet.3,4 Additionally, a systematic review of observational and experimental studies found topical glycopyrrolate to be efficacious for craniofacial hyperhidrosis with minimal adverse effects.5 The availability of low-cost prescription and over-the-counter aluminum-based antiperspirant agents makes topicals the first-line choice.

More invasive treatments are available for hyperhidrosis that is refractory to topicals. In a double-blind, randomized controlled trial, researchers injected either botulinum toxin type A (BTX-A) 50 U or placebo in patients with bilateral primary axillary hyperhidrosis.6 Of the 207 patients who received treatment injections, 96.1% had at least a 50% reduction of axillary sweating at 4 weeks after one injection, as measured by gravimetric assessment. The BTX-A injections also produced a prolonged effect; mean duration between injections was 30.6 weeks.

Other invasive treatments include iontophoresis, surgery, and laser therapy; however, these methods are not suitable for body-wide application and are, thus, not appropriate for patients with generalized hyperhidrosis.

Oxybutynin is the first oral agent to emerge as a treatment option for hyperhidrosis. This cholinergic antagonist had historically been used to treat overactive bladder. As a cholinergic antagonist, oxybutynin not only reduces urinary frequency, but also decreases secretions in various locations and, thus, can cause dry mouth and reduce perspiration.

In one prospective placebo-controlled trial, 50 patients with generalized hyperhidrosis were randomized to receive either oxybutynin titrated from 2.5 mg orally once daily to 5 mg orally twice daily or placebo for 6 weeks.7 Seventeen (73.9%) patients receiving oxybutynin for palmar or axillary hyperhidrosis reported moderate to “great” resolution of their symptoms compared with 6 (27.3%) patients in the placebo group. Dry mouth was reported in 34.8% of patients receiving oxybutynin vs 9.1% of those who received placebo (P=.038); however, no patients dropped out of the study due to this adverse effect.7

STUDY SUMMARY

This multicenter, randomized controlled trial compared oxybutynin to placebo in 62 adults with localized or generalized hyperhidrosis from 12 outpatient dermatology practices in France. It is the first study to include patients with a localized, as well as a generalized form of the condition.

Patients were included if they were >18 years of age, enrolled in the National Health Insurance system in France, and reported a Hyperhidrosis Disease Severity Scale (HDSS) score ≥2. The HDSS is a validated, one-question tool (“How would you rate the severity of your sweating?”). Patients provide a score of 1 (no perceptible sweating and no interference with everyday life) to 4 (intolerable sweating with constant interference with everyday life).8 Patients were excluded if they had any contraindications to the use of an anticholinergic medication.

This trial used a relatively low dose of oxybutynin, which produced significant benefit while minimizing anticholinergic adverse effects.

Patients randomized to oxybutynin took 2.5 mg/d orally initially and increased gradually over 8 days until reaching an effective dose that was not more than 7.5 mg/d. They then continued at that dose for 6 weeks. The primary outcome was improvement on the HDSS by one or more points measured at the beginning of the trial and at 6 weeks. Secondary outcomes included change in quality of life, as measured by the Dermatology Life Quality Index (DLQI) and reported adverse effects. The DLQI is a dermatology-specific quality-of-life measure consisting of 10 questions. Scores range from 0 (where their disease has no impact on their quality of life) to 30 (maximum impact of their disease on their quality of life).9

Improved HDSS and DLQI scores. Most patients (83%) in the study had generalized hyperhidrosis. Patients were in their mid-thirties. Sixty percent of the patients in the oxybutynin group had an improvement of one point or more on the 4-point HDSS compared to 27% in the placebo group (P<.01). DLQI scores improved by 6.9 points in the oxybutynin group and 2.3 points in the placebo group (P<.01).

The most common adverse effect was dry mouth, which occurred in 13 patients (43%) in the oxybutynin group and in 3 patients (11%) in the placebo group (P<.01); it did not cause any patients to drop out of the study. The second most common adverse effect was blurred vision, which only occurred in the oxybutynin group (4 patients; 13%).

 

 

 

WHAT'S NEW

This is the first randomized controlled trial to demonstrate the efficacy of an oral agent for generalized primary hyperhidrosis. This trial used a relatively low dose of oxybutynin, which produced significant benefit while minimizing anticholinergic adverse effects.

CAVEATS

There are many situations for which anticholinergic medications are inappropriate, including use by geriatric patients and those with gastrointestinal disorders, urinary retention, or glaucoma.

CHALLENGES TO IMPLEMENTATION

Few if any challenges exist to the utilization of oxybutynin; inexpensive generic versions are widely available.

ACKNOWLEDGEMENT

The PURLs Surveillance System was supported in part by Grant Number UL1RR024999 from the National Center For Research Resources, a Clinical Translational Science Award to the University of Chicago. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Center For Research Resources or the National Institutes of Health.

Files
References

1. Schollhammer M, Brenaut E, Menard-Andivot N, et al. Oxybutynin as a treatment for generalized hyperhidrosis: a randomized, placebo-controlled trial. Br J Dermatol. 2015;173:1163-1168.

2. Grabell DA, Hebert AA. Current and emerging medical therapies for primary hyperhidrosis. Dermatol Ther (Heidelb). 2017;7:25-36.

3. Innocenzi D, Lupi F, Bruni F, et al. Efficacy of a new aluminium salt thermophobic foam in the treatment of axillary and palmar primary hyperhidrosis: a pilot exploratory trial. Curr Med Res Opin. 2005;21:1949-1953.

4. Goh CL. Aluminum chloride hexahydrate versus palmar hyperhidrosis. Evaporimeter assessment. Int J Dermatol. 1990;29:368-370.

5. Nicholas R, Quddus A, Baker DM. Treatment of primary craniofacial hyperhidrosis: a systematic review. Am J Clin Dermatol. 2015;16:361-370.

6. Naumann M, Lowe NJ, Kumar CR, et al. Botulinum toxin type a is a safe and effective treatment for axillary hyperhidrosis over 16 months: a prospective study. Arch Dermatol. 2003;139:731-736.

7. Wolosker N, de Campos JR, Kauffman P, et al. A randomized placebo-controlled trial of oxybutynin for the initial treatment of palmar and axillary hyperhidrosis. J Vasc Surg. 2012;55:1696-1700.

8. Varella AY, Fukuda JM, Teivelis MP, et al. Translation and validation of Hyperhidrosis Disease Severity Scale. Rev Assoc Med Bras. 2016;62:843-847.

9. Finlay AY, Khan GK. Dermatology Life Quality Index (DLQI)—a simple practical measure for routine clinical use. Clin Exp Dermatol. 1994;19:210-216.

Article PDF
Author and Disclosure Information

University of Illinois at Chicago (Dr. Jarrett); Nellis Air Force Base Family Medicine Residency, Las Vegas, Nev (Dr. Moss)

DEPUTY EDITOR
Anne Mounsey, MD

Department of Family Medicine, University of North Carolina, Chapel Hill

Issue
The Journal of Family Practice - 66(6)
Publications
Topics
Page Number
392-394
Sections
Files
Files
Author and Disclosure Information

University of Illinois at Chicago (Dr. Jarrett); Nellis Air Force Base Family Medicine Residency, Las Vegas, Nev (Dr. Moss)

DEPUTY EDITOR
Anne Mounsey, MD

Department of Family Medicine, University of North Carolina, Chapel Hill

Author and Disclosure Information

University of Illinois at Chicago (Dr. Jarrett); Nellis Air Force Base Family Medicine Residency, Las Vegas, Nev (Dr. Moss)

DEPUTY EDITOR
Anne Mounsey, MD

Department of Family Medicine, University of North Carolina, Chapel Hill

Article PDF
Article PDF
 

ILLUSTRATIVE CASE

A 34-year-old woman presents to your office for unbearable sweating. She notes that the sweating occurs nearly daily on her hands, face, and in her axillary regions, causing social embarrassment. She has tried multiple antiperspirants to no avail. Is there anything she can take to reduce the sweating?

Hyperhidrosis is a common, self-limiting problem affecting 2% to 3% of the population in the United States.2 Patients may complain of localized sweating of the hands, feet, face, or underarms or more systemic, generalized sweating in multiple locations. Either way, patients always note a significant impact on their quality of life.

Treatment of hyperhidrosis has traditionally focused on topical therapies to the affected areas. Research has shown that localized treatment with antiperspirants containing aluminum salt is effective by both subjective report and objective measurements at reducing sweating—particularly in the axilla, hands, and feet.3,4 Additionally, a systematic review of observational and experimental studies found topical glycopyrrolate to be efficacious for craniofacial hyperhidrosis with minimal adverse effects.5 The availability of low-cost prescription and over-the-counter aluminum-based antiperspirant agents makes topicals the first-line choice.

More invasive treatments are available for hyperhidrosis that is refractory to topicals. In a double-blind, randomized controlled trial, researchers injected either botulinum toxin type A (BTX-A) 50 U or placebo in patients with bilateral primary axillary hyperhidrosis.6 Of the 207 patients who received treatment injections, 96.1% had at least a 50% reduction of axillary sweating at 4 weeks after one injection, as measured by gravimetric assessment. The BTX-A injections also produced a prolonged effect; mean duration between injections was 30.6 weeks.

Other invasive treatments include iontophoresis, surgery, and laser therapy; however, these methods are not suitable for body-wide application and are, thus, not appropriate for patients with generalized hyperhidrosis.

Oxybutynin is the first oral agent to emerge as a treatment option for hyperhidrosis. This cholinergic antagonist had historically been used to treat overactive bladder. As a cholinergic antagonist, oxybutynin not only reduces urinary frequency, but also decreases secretions in various locations and, thus, can cause dry mouth and reduce perspiration.

In one prospective placebo-controlled trial, 50 patients with generalized hyperhidrosis were randomized to receive either oxybutynin titrated from 2.5 mg orally once daily to 5 mg orally twice daily or placebo for 6 weeks.7 Seventeen (73.9%) patients receiving oxybutynin for palmar or axillary hyperhidrosis reported moderate to “great” resolution of their symptoms compared with 6 (27.3%) patients in the placebo group. Dry mouth was reported in 34.8% of patients receiving oxybutynin vs 9.1% of those who received placebo (P=.038); however, no patients dropped out of the study due to this adverse effect.7

STUDY SUMMARY

This multicenter, randomized controlled trial compared oxybutynin to placebo in 62 adults with localized or generalized hyperhidrosis from 12 outpatient dermatology practices in France. It is the first study to include patients with a localized, as well as a generalized form of the condition.

Patients were included if they were >18 years of age, enrolled in the National Health Insurance system in France, and reported a Hyperhidrosis Disease Severity Scale (HDSS) score ≥2. The HDSS is a validated, one-question tool (“How would you rate the severity of your sweating?”). Patients provide a score of 1 (no perceptible sweating and no interference with everyday life) to 4 (intolerable sweating with constant interference with everyday life).8 Patients were excluded if they had any contraindications to the use of an anticholinergic medication.

This trial used a relatively low dose of oxybutynin, which produced significant benefit while minimizing anticholinergic adverse effects.

Patients randomized to oxybutynin took 2.5 mg/d orally initially and increased gradually over 8 days until reaching an effective dose that was not more than 7.5 mg/d. They then continued at that dose for 6 weeks. The primary outcome was improvement on the HDSS by one or more points measured at the beginning of the trial and at 6 weeks. Secondary outcomes included change in quality of life, as measured by the Dermatology Life Quality Index (DLQI) and reported adverse effects. The DLQI is a dermatology-specific quality-of-life measure consisting of 10 questions. Scores range from 0 (where their disease has no impact on their quality of life) to 30 (maximum impact of their disease on their quality of life).9

Improved HDSS and DLQI scores. Most patients (83%) in the study had generalized hyperhidrosis. Patients were in their mid-thirties. Sixty percent of the patients in the oxybutynin group had an improvement of one point or more on the 4-point HDSS compared to 27% in the placebo group (P<.01). DLQI scores improved by 6.9 points in the oxybutynin group and 2.3 points in the placebo group (P<.01).

The most common adverse effect was dry mouth, which occurred in 13 patients (43%) in the oxybutynin group and in 3 patients (11%) in the placebo group (P<.01); it did not cause any patients to drop out of the study. The second most common adverse effect was blurred vision, which only occurred in the oxybutynin group (4 patients; 13%).

 

 

 

WHAT'S NEW

This is the first randomized controlled trial to demonstrate the efficacy of an oral agent for generalized primary hyperhidrosis. This trial used a relatively low dose of oxybutynin, which produced significant benefit while minimizing anticholinergic adverse effects.

CAVEATS

There are many situations for which anticholinergic medications are inappropriate, including use by geriatric patients and those with gastrointestinal disorders, urinary retention, or glaucoma.

CHALLENGES TO IMPLEMENTATION

Few if any challenges exist to the utilization of oxybutynin; inexpensive generic versions are widely available.

ACKNOWLEDGEMENT

The PURLs Surveillance System was supported in part by Grant Number UL1RR024999 from the National Center For Research Resources, a Clinical Translational Science Award to the University of Chicago. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Center For Research Resources or the National Institutes of Health.

 

ILLUSTRATIVE CASE

A 34-year-old woman presents to your office for unbearable sweating. She notes that the sweating occurs nearly daily on her hands, face, and in her axillary regions, causing social embarrassment. She has tried multiple antiperspirants to no avail. Is there anything she can take to reduce the sweating?

Hyperhidrosis is a common, self-limiting problem affecting 2% to 3% of the population in the United States.2 Patients may complain of localized sweating of the hands, feet, face, or underarms or more systemic, generalized sweating in multiple locations. Either way, patients always note a significant impact on their quality of life.

Treatment of hyperhidrosis has traditionally focused on topical therapies to the affected areas. Research has shown that localized treatment with antiperspirants containing aluminum salt is effective by both subjective report and objective measurements at reducing sweating—particularly in the axilla, hands, and feet.3,4 Additionally, a systematic review of observational and experimental studies found topical glycopyrrolate to be efficacious for craniofacial hyperhidrosis with minimal adverse effects.5 The availability of low-cost prescription and over-the-counter aluminum-based antiperspirant agents makes topicals the first-line choice.

More invasive treatments are available for hyperhidrosis that is refractory to topicals. In a double-blind, randomized controlled trial, researchers injected either botulinum toxin type A (BTX-A) 50 U or placebo in patients with bilateral primary axillary hyperhidrosis.6 Of the 207 patients who received treatment injections, 96.1% had at least a 50% reduction of axillary sweating at 4 weeks after one injection, as measured by gravimetric assessment. The BTX-A injections also produced a prolonged effect; mean duration between injections was 30.6 weeks.

Other invasive treatments include iontophoresis, surgery, and laser therapy; however, these methods are not suitable for body-wide application and are, thus, not appropriate for patients with generalized hyperhidrosis.

Oxybutynin is the first oral agent to emerge as a treatment option for hyperhidrosis. This cholinergic antagonist had historically been used to treat overactive bladder. As a cholinergic antagonist, oxybutynin not only reduces urinary frequency, but also decreases secretions in various locations and, thus, can cause dry mouth and reduce perspiration.

In one prospective placebo-controlled trial, 50 patients with generalized hyperhidrosis were randomized to receive either oxybutynin titrated from 2.5 mg orally once daily to 5 mg orally twice daily or placebo for 6 weeks.7 Seventeen (73.9%) patients receiving oxybutynin for palmar or axillary hyperhidrosis reported moderate to “great” resolution of their symptoms compared with 6 (27.3%) patients in the placebo group. Dry mouth was reported in 34.8% of patients receiving oxybutynin vs 9.1% of those who received placebo (P=.038); however, no patients dropped out of the study due to this adverse effect.7

STUDY SUMMARY

This multicenter, randomized controlled trial compared oxybutynin to placebo in 62 adults with localized or generalized hyperhidrosis from 12 outpatient dermatology practices in France. It is the first study to include patients with a localized, as well as a generalized form of the condition.

Patients were included if they were >18 years of age, enrolled in the National Health Insurance system in France, and reported a Hyperhidrosis Disease Severity Scale (HDSS) score ≥2. The HDSS is a validated, one-question tool (“How would you rate the severity of your sweating?”). Patients provide a score of 1 (no perceptible sweating and no interference with everyday life) to 4 (intolerable sweating with constant interference with everyday life).8 Patients were excluded if they had any contraindications to the use of an anticholinergic medication.

This trial used a relatively low dose of oxybutynin, which produced significant benefit while minimizing anticholinergic adverse effects.

Patients randomized to oxybutynin took 2.5 mg/d orally initially and increased gradually over 8 days until reaching an effective dose that was not more than 7.5 mg/d. They then continued at that dose for 6 weeks. The primary outcome was improvement on the HDSS by one or more points measured at the beginning of the trial and at 6 weeks. Secondary outcomes included change in quality of life, as measured by the Dermatology Life Quality Index (DLQI) and reported adverse effects. The DLQI is a dermatology-specific quality-of-life measure consisting of 10 questions. Scores range from 0 (where their disease has no impact on their quality of life) to 30 (maximum impact of their disease on their quality of life).9

Improved HDSS and DLQI scores. Most patients (83%) in the study had generalized hyperhidrosis. Patients were in their mid-thirties. Sixty percent of the patients in the oxybutynin group had an improvement of one point or more on the 4-point HDSS compared to 27% in the placebo group (P<.01). DLQI scores improved by 6.9 points in the oxybutynin group and 2.3 points in the placebo group (P<.01).

The most common adverse effect was dry mouth, which occurred in 13 patients (43%) in the oxybutynin group and in 3 patients (11%) in the placebo group (P<.01); it did not cause any patients to drop out of the study. The second most common adverse effect was blurred vision, which only occurred in the oxybutynin group (4 patients; 13%).

 

 

 

WHAT'S NEW

This is the first randomized controlled trial to demonstrate the efficacy of an oral agent for generalized primary hyperhidrosis. This trial used a relatively low dose of oxybutynin, which produced significant benefit while minimizing anticholinergic adverse effects.

CAVEATS

There are many situations for which anticholinergic medications are inappropriate, including use by geriatric patients and those with gastrointestinal disorders, urinary retention, or glaucoma.

CHALLENGES TO IMPLEMENTATION

Few if any challenges exist to the utilization of oxybutynin; inexpensive generic versions are widely available.

ACKNOWLEDGEMENT

The PURLs Surveillance System was supported in part by Grant Number UL1RR024999 from the National Center For Research Resources, a Clinical Translational Science Award to the University of Chicago. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Center For Research Resources or the National Institutes of Health.

References

1. Schollhammer M, Brenaut E, Menard-Andivot N, et al. Oxybutynin as a treatment for generalized hyperhidrosis: a randomized, placebo-controlled trial. Br J Dermatol. 2015;173:1163-1168.

2. Grabell DA, Hebert AA. Current and emerging medical therapies for primary hyperhidrosis. Dermatol Ther (Heidelb). 2017;7:25-36.

3. Innocenzi D, Lupi F, Bruni F, et al. Efficacy of a new aluminium salt thermophobic foam in the treatment of axillary and palmar primary hyperhidrosis: a pilot exploratory trial. Curr Med Res Opin. 2005;21:1949-1953.

4. Goh CL. Aluminum chloride hexahydrate versus palmar hyperhidrosis. Evaporimeter assessment. Int J Dermatol. 1990;29:368-370.

5. Nicholas R, Quddus A, Baker DM. Treatment of primary craniofacial hyperhidrosis: a systematic review. Am J Clin Dermatol. 2015;16:361-370.

6. Naumann M, Lowe NJ, Kumar CR, et al. Botulinum toxin type a is a safe and effective treatment for axillary hyperhidrosis over 16 months: a prospective study. Arch Dermatol. 2003;139:731-736.

7. Wolosker N, de Campos JR, Kauffman P, et al. A randomized placebo-controlled trial of oxybutynin for the initial treatment of palmar and axillary hyperhidrosis. J Vasc Surg. 2012;55:1696-1700.

8. Varella AY, Fukuda JM, Teivelis MP, et al. Translation and validation of Hyperhidrosis Disease Severity Scale. Rev Assoc Med Bras. 2016;62:843-847.

9. Finlay AY, Khan GK. Dermatology Life Quality Index (DLQI)—a simple practical measure for routine clinical use. Clin Exp Dermatol. 1994;19:210-216.

References

1. Schollhammer M, Brenaut E, Menard-Andivot N, et al. Oxybutynin as a treatment for generalized hyperhidrosis: a randomized, placebo-controlled trial. Br J Dermatol. 2015;173:1163-1168.

2. Grabell DA, Hebert AA. Current and emerging medical therapies for primary hyperhidrosis. Dermatol Ther (Heidelb). 2017;7:25-36.

3. Innocenzi D, Lupi F, Bruni F, et al. Efficacy of a new aluminium salt thermophobic foam in the treatment of axillary and palmar primary hyperhidrosis: a pilot exploratory trial. Curr Med Res Opin. 2005;21:1949-1953.

4. Goh CL. Aluminum chloride hexahydrate versus palmar hyperhidrosis. Evaporimeter assessment. Int J Dermatol. 1990;29:368-370.

5. Nicholas R, Quddus A, Baker DM. Treatment of primary craniofacial hyperhidrosis: a systematic review. Am J Clin Dermatol. 2015;16:361-370.

6. Naumann M, Lowe NJ, Kumar CR, et al. Botulinum toxin type a is a safe and effective treatment for axillary hyperhidrosis over 16 months: a prospective study. Arch Dermatol. 2003;139:731-736.

7. Wolosker N, de Campos JR, Kauffman P, et al. A randomized placebo-controlled trial of oxybutynin for the initial treatment of palmar and axillary hyperhidrosis. J Vasc Surg. 2012;55:1696-1700.

8. Varella AY, Fukuda JM, Teivelis MP, et al. Translation and validation of Hyperhidrosis Disease Severity Scale. Rev Assoc Med Bras. 2016;62:843-847.

9. Finlay AY, Khan GK. Dermatology Life Quality Index (DLQI)—a simple practical measure for routine clinical use. Clin Exp Dermatol. 1994;19:210-216.

Issue
The Journal of Family Practice - 66(6)
Issue
The Journal of Family Practice - 66(6)
Page Number
392-394
Page Number
392-394
Publications
Publications
Topics
Article Type
Display Headline
Oral agent offers relief from generalized hyperhidrosis
Display Headline
Oral agent offers relief from generalized hyperhidrosis
Sections
PURLs Copyright

Copyright © 2017. The Family Physicians Inquiries Network. All rights reserved.

Inside the Article

PRACTICE CHANGER

Use low-dose oxybutynin as a first-line treatment option for patients with primary hyperhidrosis to improve symptoms and quality of life.1

STRENGTH OF RECOMMENDATION

B: Based on a single, good quality, randomized controlled trial.

Schollhammer M, Brenaut E, Menard-Andivot N, et al. Oxybutynin as a treatment for generalized hyperhidrosis: a randomized, placebo-controlled trial. Br J Dermatol. 2015;173:1163-1168.

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
PubMed ID
28574524
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Article PDF Media
Media Files

Severe right hip pain

Article Type
Changed
Fri, 01/18/2019 - 08:47
Display Headline
Severe right hip pain
 

A 63-year-old woman with a 3-year history of osteoporosis presented to our clinic with a 2-week history of severe right hip pain. She had been taking a bisphosphonate—oral ibandronate sodium, 150 mg, once monthlyfor about 6 years. The postmenopausal patient had a history of degenerative disc disease and lumbar back pain, but no known history of recent trauma or falls.

A clinical exam revealed full passive and active range of motion; however, she had pain with weight bearing. A full metabolic panel revealed no significant abnormalities. A leg length discrepancy was noted, so a bone length study was ordered. Anteroposterior x-rays of the bilateral lower extremities demonstrated a focal convexity along the lateral cortical junction of the proximal right femur (FIGURE).

WHAT IS YOUR DIAGNOSIS?
HOW WOULD YOU TREAT THIS PATIENT?

 

 

 

Dx: Bisphosphonate-associated proximal insufficiency fracture

Based on the patient’s clinical history and x-ray findings, we determined that the patient had sustained a bisphosphonate-associated proximal femoral insufficiency fracture. Insufficiency fractures arise from normal physi­­ologic stress on abnormal bone. They commonly occur in conditions that impair normal bone physiology and remodeling, such as osteoporosis, renal insufficiency, rheumatoid arthritis, and diabetes.1

Could a bisphosphonate be to blame? Bisphosphonate therapy has been associated with significant benefits, including increased bone mineral density (BMD), decreased incidence of fracture, and improved mortality.2-4 But it’s been postulated that the global suppression of bone turnover caused by these drugs may also impair the bone remodeling process.5 Some case reports have suggested an association between chronic bisphosphonate use and atypical insufficiency fractures. These atypical femur fractures are characterized by their location (along the diaphysis in the region distal to the lesser trochanter), the patient’s history (there may be minimal to no trauma), and the potential for “beaking” (localized periosteal or endosteal thickening of the lateral cortex).6,7

Bisphosphonate therapy has been associated with significant benefits, but it’s been suggested that these drugs may impair the bone remodeling process.

Several large, population-based, case-control studies have found a temporal relationship between bisphosphonate therapy and a statistically significant increased risk of subtrochanteric fractures.8-10 These studies do note, however, that the absolute risk of insufficiency fracture is very low, and that the benefits of bisphosphonate therapy greatly outweigh the risks. A 2013 meta-analysis came to the same conclusion.11

Treatment options include PT, surgical intervention

When an insufficiency fracture is identified in a patient taking a bisphosphonate, the medication should be discontinued and a consultation with Endocrinology should be arranged. Nonsurgical management ranges from physical therapy to alternative medication regimens, such as teriparatide—a recombinant human parathyroid hormone used to restore bone quality. A variety of surgical stabilization options are also available.6

In contrast to typical subtrochanteric fractures, about half of patients with atypical insufficiency fractures demonstrate poor fracture healing that requires surgical intervention.12 Complete fractures almost always require surgery, while incomplete subtrochanteric femur fractures can usually be managed conservatively by altering pharmacologic prophylaxis (interval dosing or discontinuation of the bisphosphonate and initiation of an alternative therapy like teriparatide) in conjunction with routine radiologic surveillance. Internal fixation may be considered for cases of persistent pain or those that progress to an unstable fracture.13

Our patient declined surgical intervention. We switched her monthly ibandronate dosage to a periodic dosing schedule (6 months on, followed by 6 months off) and advised her to rest and take nonsteroidal anti-inflammatory drugs when needed. While consensus guidelines exist for the management of osteoporosis (see Osteoporosis: Assessing your patient’s risk), there is still debate over the optimal length of bisphosphonate therapy and the impact of drug holidays; a recent review in The BMJ discusses bisphosphonate use in detail.5

SIDEBAR
Osteoporosis: Assessing your patient's risk
Approximately 9.9 million Americans have osteoporosis, and while the disease is more common in Caucasian females, patients with osteoporosis have the same elevated fracture risk regardless of their race.14 The US Preventive Services Task Force recommends bone mineral density (BMD) testing for all women ages 65 years and older (earlier if risk factor profile warrants).15

According to the World Health Organization (WHO), patients with BMD T-scores at the hip or lumbar spine that are ≤2.5 standard deviations below the mean BMD of a young-adult reference population are at highest risk for osteoporotic fractures.16 There are also free online risk assessment tools, like the WHO’s FRAX calculator (available at: http://www.shef.ac.uk/FRAX/tool.jsp?locationValue=9), which integrate clinical data to generate an evidence-based assessment of fracture risk.17

Follow-up x-rays 14 months later revealed that the insufficiency fracture had healed with a bony callus.

CORRESPONDENCE
Joseph S. McMonagle, MD, Eastern Virginia Medical School, Department of Radiology, P.O. Box 1980, Norfolk, VA; [email protected].

References

1. Sheehan SE, Shyu JY, Weaver MJ, et al. Proximal femoral fractures: what the orthopedic surgeon wants to know. Radiographics. 2015;35:1563-1584.

2. Wells G, Cranney A, Peterson J, et al. Risedronate for the primary and secondary prevention of osteoporotic fractures in postmenopausal women. Cochrane Database Syst Rev. 2008;(1):CD004523.

3. Wells GA, Cranney A, Peterson J, et al. Alendronate for the primary and secondary prevention of osteoporotic fractures in postmenopausal women. Cochrane Database Syst Rev. 2008;(1):CD001155.

4. Wells GA, Cranney A, Peterson J, et al. Etidronate for the primary and secondary prevention of osteoporotic fractures in postmenopausal women. Cochrane Database Syst Rev. 2008;(1):CD003376.

5. Maraka S, Kennel KA. Bisphosphonates for the prevention and treatment of osteoporosis. BMJ. 2015;351:h3783.

6. Balach T, Baldwin PC, Intravia J. Atypical femur fractures associated with diphosphonate use. J Am Acad Orthop Surg. 2015;23:550-557.

7. Porrino JA Jr, Kohl CA, Taljanovic M, et al. Diagnosis of proximal femoral insufficiency fractures in patients receiving bisphosphonate therapy. AJR Am J Roentgenol. 2010;194:1061-1064.

8. Edwards BJ, Bunta AD, Lane J, et al. Bisphosphonates and nonhealing femoral fractures: analysis of the FDA Adverse Event Reporting System (FAERS) and international safety efforts: a systematic review from the Research on Adverse Drug Events And Reports (RADAR) project. J Bone Joint Surg Am. 2013;95:297-307.

9. Park-Wyllie LY, Mamdani MM, Juurlink DN, et al. Bisphosphonate use and the risk of subtrochanteric or femoral shaft fractures in older women. JAMA. 2011;305:783-789.

10. Schilcher J, Michaëlsson K, Aspenberg P. Bisphosphonate use and atypical fractures of the femoral shaft. N Engl J Med. 2011;364:1728-1737.

11. Gedmintas L, Solomon DH, Kim SC. Bisphosphonates and risk of subtrochanteric, femoral shaft, and atypical femur fracture: a systematic review and meta-analysis. J Bone Miner Res. 2013;28:1729-1737.

12. Weil YA, Rivkin G, Safran O, et al. The outcome of surgically treated femur fractures associated with long-term bisphosphonate use. J Trauma. 2011;71:186-190.

13. Khan AA, Leslie WD, Lentle B, et al. Atypical femoral fractures: a teaching perspective. Can Assoc Radiol J. 2015;66:102-107.

14. Cosman F, de Beur SJ, LeBoff MS, et al; National Osteoporosis Foundation. Clinician’s Guide to Prevention and Treatment of Osteoporosis. Osteoporos Int. 2014;25:2359-2381.

15. US Preventive Services Task Force. Screening for osteoporosis: U.S. preventive services task force recommendation statement. Ann Intern Med. 2011;154:356-364.

16. World Health Organization (WHO). 1994. Assessment of fracture risk and its application to screening for postmenopausal osteoporosis: report of a WHO study group. WHO Technical Report Series, Report No. 843. WHO, Geneva, Switzerland.

17. Kanis JA, McCloskey EV, Johansson H, et al. Development and use of FRAX in osteoporosis. Osteoporos Int. 2010;21:S407-S413.

Article PDF
Author and Disclosure Information

Department of Radiology, Eastern Virginia Medical School, Norfolk (Drs. Trace, Hoang, and McMonagle); Hampton Roads Radiology, Norfolk, Va (Dr. McMonagle)
[email protected]

DEPARTMENT EDITOR
Richard P. Usatine, MD

University of Texas Health Science Center at San Antonio

The authors reported no potential conflict of interest relevant to this article.

This case was presented, in part, at the Radiological Society of North America Annual Meeting in 2014 by Dr. Hoang.

Issue
The Journal of Family Practice - 66(6)
Publications
Topics
Page Number
395-397
Sections
Author and Disclosure Information

Department of Radiology, Eastern Virginia Medical School, Norfolk (Drs. Trace, Hoang, and McMonagle); Hampton Roads Radiology, Norfolk, Va (Dr. McMonagle)
[email protected]

DEPARTMENT EDITOR
Richard P. Usatine, MD

University of Texas Health Science Center at San Antonio

The authors reported no potential conflict of interest relevant to this article.

This case was presented, in part, at the Radiological Society of North America Annual Meeting in 2014 by Dr. Hoang.

Author and Disclosure Information

Department of Radiology, Eastern Virginia Medical School, Norfolk (Drs. Trace, Hoang, and McMonagle); Hampton Roads Radiology, Norfolk, Va (Dr. McMonagle)
[email protected]

DEPARTMENT EDITOR
Richard P. Usatine, MD

University of Texas Health Science Center at San Antonio

The authors reported no potential conflict of interest relevant to this article.

This case was presented, in part, at the Radiological Society of North America Annual Meeting in 2014 by Dr. Hoang.

Article PDF
Article PDF
 

A 63-year-old woman with a 3-year history of osteoporosis presented to our clinic with a 2-week history of severe right hip pain. She had been taking a bisphosphonate—oral ibandronate sodium, 150 mg, once monthlyfor about 6 years. The postmenopausal patient had a history of degenerative disc disease and lumbar back pain, but no known history of recent trauma or falls.

A clinical exam revealed full passive and active range of motion; however, she had pain with weight bearing. A full metabolic panel revealed no significant abnormalities. A leg length discrepancy was noted, so a bone length study was ordered. Anteroposterior x-rays of the bilateral lower extremities demonstrated a focal convexity along the lateral cortical junction of the proximal right femur (FIGURE).

WHAT IS YOUR DIAGNOSIS?
HOW WOULD YOU TREAT THIS PATIENT?

 

 

 

Dx: Bisphosphonate-associated proximal insufficiency fracture

Based on the patient’s clinical history and x-ray findings, we determined that the patient had sustained a bisphosphonate-associated proximal femoral insufficiency fracture. Insufficiency fractures arise from normal physi­­ologic stress on abnormal bone. They commonly occur in conditions that impair normal bone physiology and remodeling, such as osteoporosis, renal insufficiency, rheumatoid arthritis, and diabetes.1

Could a bisphosphonate be to blame? Bisphosphonate therapy has been associated with significant benefits, including increased bone mineral density (BMD), decreased incidence of fracture, and improved mortality.2-4 But it’s been postulated that the global suppression of bone turnover caused by these drugs may also impair the bone remodeling process.5 Some case reports have suggested an association between chronic bisphosphonate use and atypical insufficiency fractures. These atypical femur fractures are characterized by their location (along the diaphysis in the region distal to the lesser trochanter), the patient’s history (there may be minimal to no trauma), and the potential for “beaking” (localized periosteal or endosteal thickening of the lateral cortex).6,7

Bisphosphonate therapy has been associated with significant benefits, but it’s been suggested that these drugs may impair the bone remodeling process.

Several large, population-based, case-control studies have found a temporal relationship between bisphosphonate therapy and a statistically significant increased risk of subtrochanteric fractures.8-10 These studies do note, however, that the absolute risk of insufficiency fracture is very low, and that the benefits of bisphosphonate therapy greatly outweigh the risks. A 2013 meta-analysis came to the same conclusion.11

Treatment options include PT, surgical intervention

When an insufficiency fracture is identified in a patient taking a bisphosphonate, the medication should be discontinued and a consultation with Endocrinology should be arranged. Nonsurgical management ranges from physical therapy to alternative medication regimens, such as teriparatide—a recombinant human parathyroid hormone used to restore bone quality. A variety of surgical stabilization options are also available.6

In contrast to typical subtrochanteric fractures, about half of patients with atypical insufficiency fractures demonstrate poor fracture healing that requires surgical intervention.12 Complete fractures almost always require surgery, while incomplete subtrochanteric femur fractures can usually be managed conservatively by altering pharmacologic prophylaxis (interval dosing or discontinuation of the bisphosphonate and initiation of an alternative therapy like teriparatide) in conjunction with routine radiologic surveillance. Internal fixation may be considered for cases of persistent pain or those that progress to an unstable fracture.13

Our patient declined surgical intervention. We switched her monthly ibandronate dosage to a periodic dosing schedule (6 months on, followed by 6 months off) and advised her to rest and take nonsteroidal anti-inflammatory drugs when needed. While consensus guidelines exist for the management of osteoporosis (see Osteoporosis: Assessing your patient’s risk), there is still debate over the optimal length of bisphosphonate therapy and the impact of drug holidays; a recent review in The BMJ discusses bisphosphonate use in detail.5

SIDEBAR
Osteoporosis: Assessing your patient's risk
Approximately 9.9 million Americans have osteoporosis, and while the disease is more common in Caucasian females, patients with osteoporosis have the same elevated fracture risk regardless of their race.14 The US Preventive Services Task Force recommends bone mineral density (BMD) testing for all women ages 65 years and older (earlier if risk factor profile warrants).15

According to the World Health Organization (WHO), patients with BMD T-scores at the hip or lumbar spine that are ≤2.5 standard deviations below the mean BMD of a young-adult reference population are at highest risk for osteoporotic fractures.16 There are also free online risk assessment tools, like the WHO’s FRAX calculator (available at: http://www.shef.ac.uk/FRAX/tool.jsp?locationValue=9), which integrate clinical data to generate an evidence-based assessment of fracture risk.17

Follow-up x-rays 14 months later revealed that the insufficiency fracture had healed with a bony callus.

CORRESPONDENCE
Joseph S. McMonagle, MD, Eastern Virginia Medical School, Department of Radiology, P.O. Box 1980, Norfolk, VA; [email protected].

 

A 63-year-old woman with a 3-year history of osteoporosis presented to our clinic with a 2-week history of severe right hip pain. She had been taking a bisphosphonate—oral ibandronate sodium, 150 mg, once monthlyfor about 6 years. The postmenopausal patient had a history of degenerative disc disease and lumbar back pain, but no known history of recent trauma or falls.

A clinical exam revealed full passive and active range of motion; however, she had pain with weight bearing. A full metabolic panel revealed no significant abnormalities. A leg length discrepancy was noted, so a bone length study was ordered. Anteroposterior x-rays of the bilateral lower extremities demonstrated a focal convexity along the lateral cortical junction of the proximal right femur (FIGURE).

WHAT IS YOUR DIAGNOSIS?
HOW WOULD YOU TREAT THIS PATIENT?

 

 

 

Dx: Bisphosphonate-associated proximal insufficiency fracture

Based on the patient’s clinical history and x-ray findings, we determined that the patient had sustained a bisphosphonate-associated proximal femoral insufficiency fracture. Insufficiency fractures arise from normal physi­­ologic stress on abnormal bone. They commonly occur in conditions that impair normal bone physiology and remodeling, such as osteoporosis, renal insufficiency, rheumatoid arthritis, and diabetes.1

Could a bisphosphonate be to blame? Bisphosphonate therapy has been associated with significant benefits, including increased bone mineral density (BMD), decreased incidence of fracture, and improved mortality.2-4 But it’s been postulated that the global suppression of bone turnover caused by these drugs may also impair the bone remodeling process.5 Some case reports have suggested an association between chronic bisphosphonate use and atypical insufficiency fractures. These atypical femur fractures are characterized by their location (along the diaphysis in the region distal to the lesser trochanter), the patient’s history (there may be minimal to no trauma), and the potential for “beaking” (localized periosteal or endosteal thickening of the lateral cortex).6,7

Bisphosphonate therapy has been associated with significant benefits, but it’s been suggested that these drugs may impair the bone remodeling process.

Several large, population-based, case-control studies have found a temporal relationship between bisphosphonate therapy and a statistically significant increased risk of subtrochanteric fractures.8-10 These studies do note, however, that the absolute risk of insufficiency fracture is very low, and that the benefits of bisphosphonate therapy greatly outweigh the risks. A 2013 meta-analysis came to the same conclusion.11

Treatment options include PT, surgical intervention

When an insufficiency fracture is identified in a patient taking a bisphosphonate, the medication should be discontinued and a consultation with Endocrinology should be arranged. Nonsurgical management ranges from physical therapy to alternative medication regimens, such as teriparatide—a recombinant human parathyroid hormone used to restore bone quality. A variety of surgical stabilization options are also available.6

In contrast to typical subtrochanteric fractures, about half of patients with atypical insufficiency fractures demonstrate poor fracture healing that requires surgical intervention.12 Complete fractures almost always require surgery, while incomplete subtrochanteric femur fractures can usually be managed conservatively by altering pharmacologic prophylaxis (interval dosing or discontinuation of the bisphosphonate and initiation of an alternative therapy like teriparatide) in conjunction with routine radiologic surveillance. Internal fixation may be considered for cases of persistent pain or those that progress to an unstable fracture.13

Our patient declined surgical intervention. We switched her monthly ibandronate dosage to a periodic dosing schedule (6 months on, followed by 6 months off) and advised her to rest and take nonsteroidal anti-inflammatory drugs when needed. While consensus guidelines exist for the management of osteoporosis (see Osteoporosis: Assessing your patient’s risk), there is still debate over the optimal length of bisphosphonate therapy and the impact of drug holidays; a recent review in The BMJ discusses bisphosphonate use in detail.5

SIDEBAR
Osteoporosis: Assessing your patient's risk
Approximately 9.9 million Americans have osteoporosis, and while the disease is more common in Caucasian females, patients with osteoporosis have the same elevated fracture risk regardless of their race.14 The US Preventive Services Task Force recommends bone mineral density (BMD) testing for all women ages 65 years and older (earlier if risk factor profile warrants).15

According to the World Health Organization (WHO), patients with BMD T-scores at the hip or lumbar spine that are ≤2.5 standard deviations below the mean BMD of a young-adult reference population are at highest risk for osteoporotic fractures.16 There are also free online risk assessment tools, like the WHO’s FRAX calculator (available at: http://www.shef.ac.uk/FRAX/tool.jsp?locationValue=9), which integrate clinical data to generate an evidence-based assessment of fracture risk.17

Follow-up x-rays 14 months later revealed that the insufficiency fracture had healed with a bony callus.

CORRESPONDENCE
Joseph S. McMonagle, MD, Eastern Virginia Medical School, Department of Radiology, P.O. Box 1980, Norfolk, VA; [email protected].

References

1. Sheehan SE, Shyu JY, Weaver MJ, et al. Proximal femoral fractures: what the orthopedic surgeon wants to know. Radiographics. 2015;35:1563-1584.

2. Wells G, Cranney A, Peterson J, et al. Risedronate for the primary and secondary prevention of osteoporotic fractures in postmenopausal women. Cochrane Database Syst Rev. 2008;(1):CD004523.

3. Wells GA, Cranney A, Peterson J, et al. Alendronate for the primary and secondary prevention of osteoporotic fractures in postmenopausal women. Cochrane Database Syst Rev. 2008;(1):CD001155.

4. Wells GA, Cranney A, Peterson J, et al. Etidronate for the primary and secondary prevention of osteoporotic fractures in postmenopausal women. Cochrane Database Syst Rev. 2008;(1):CD003376.

5. Maraka S, Kennel KA. Bisphosphonates for the prevention and treatment of osteoporosis. BMJ. 2015;351:h3783.

6. Balach T, Baldwin PC, Intravia J. Atypical femur fractures associated with diphosphonate use. J Am Acad Orthop Surg. 2015;23:550-557.

7. Porrino JA Jr, Kohl CA, Taljanovic M, et al. Diagnosis of proximal femoral insufficiency fractures in patients receiving bisphosphonate therapy. AJR Am J Roentgenol. 2010;194:1061-1064.

8. Edwards BJ, Bunta AD, Lane J, et al. Bisphosphonates and nonhealing femoral fractures: analysis of the FDA Adverse Event Reporting System (FAERS) and international safety efforts: a systematic review from the Research on Adverse Drug Events And Reports (RADAR) project. J Bone Joint Surg Am. 2013;95:297-307.

9. Park-Wyllie LY, Mamdani MM, Juurlink DN, et al. Bisphosphonate use and the risk of subtrochanteric or femoral shaft fractures in older women. JAMA. 2011;305:783-789.

10. Schilcher J, Michaëlsson K, Aspenberg P. Bisphosphonate use and atypical fractures of the femoral shaft. N Engl J Med. 2011;364:1728-1737.

11. Gedmintas L, Solomon DH, Kim SC. Bisphosphonates and risk of subtrochanteric, femoral shaft, and atypical femur fracture: a systematic review and meta-analysis. J Bone Miner Res. 2013;28:1729-1737.

12. Weil YA, Rivkin G, Safran O, et al. The outcome of surgically treated femur fractures associated with long-term bisphosphonate use. J Trauma. 2011;71:186-190.

13. Khan AA, Leslie WD, Lentle B, et al. Atypical femoral fractures: a teaching perspective. Can Assoc Radiol J. 2015;66:102-107.

14. Cosman F, de Beur SJ, LeBoff MS, et al; National Osteoporosis Foundation. Clinician’s Guide to Prevention and Treatment of Osteoporosis. Osteoporos Int. 2014;25:2359-2381.

15. US Preventive Services Task Force. Screening for osteoporosis: U.S. preventive services task force recommendation statement. Ann Intern Med. 2011;154:356-364.

16. World Health Organization (WHO). 1994. Assessment of fracture risk and its application to screening for postmenopausal osteoporosis: report of a WHO study group. WHO Technical Report Series, Report No. 843. WHO, Geneva, Switzerland.

17. Kanis JA, McCloskey EV, Johansson H, et al. Development and use of FRAX in osteoporosis. Osteoporos Int. 2010;21:S407-S413.

References

1. Sheehan SE, Shyu JY, Weaver MJ, et al. Proximal femoral fractures: what the orthopedic surgeon wants to know. Radiographics. 2015;35:1563-1584.

2. Wells G, Cranney A, Peterson J, et al. Risedronate for the primary and secondary prevention of osteoporotic fractures in postmenopausal women. Cochrane Database Syst Rev. 2008;(1):CD004523.

3. Wells GA, Cranney A, Peterson J, et al. Alendronate for the primary and secondary prevention of osteoporotic fractures in postmenopausal women. Cochrane Database Syst Rev. 2008;(1):CD001155.

4. Wells GA, Cranney A, Peterson J, et al. Etidronate for the primary and secondary prevention of osteoporotic fractures in postmenopausal women. Cochrane Database Syst Rev. 2008;(1):CD003376.

5. Maraka S, Kennel KA. Bisphosphonates for the prevention and treatment of osteoporosis. BMJ. 2015;351:h3783.

6. Balach T, Baldwin PC, Intravia J. Atypical femur fractures associated with diphosphonate use. J Am Acad Orthop Surg. 2015;23:550-557.

7. Porrino JA Jr, Kohl CA, Taljanovic M, et al. Diagnosis of proximal femoral insufficiency fractures in patients receiving bisphosphonate therapy. AJR Am J Roentgenol. 2010;194:1061-1064.

8. Edwards BJ, Bunta AD, Lane J, et al. Bisphosphonates and nonhealing femoral fractures: analysis of the FDA Adverse Event Reporting System (FAERS) and international safety efforts: a systematic review from the Research on Adverse Drug Events And Reports (RADAR) project. J Bone Joint Surg Am. 2013;95:297-307.

9. Park-Wyllie LY, Mamdani MM, Juurlink DN, et al. Bisphosphonate use and the risk of subtrochanteric or femoral shaft fractures in older women. JAMA. 2011;305:783-789.

10. Schilcher J, Michaëlsson K, Aspenberg P. Bisphosphonate use and atypical fractures of the femoral shaft. N Engl J Med. 2011;364:1728-1737.

11. Gedmintas L, Solomon DH, Kim SC. Bisphosphonates and risk of subtrochanteric, femoral shaft, and atypical femur fracture: a systematic review and meta-analysis. J Bone Miner Res. 2013;28:1729-1737.

12. Weil YA, Rivkin G, Safran O, et al. The outcome of surgically treated femur fractures associated with long-term bisphosphonate use. J Trauma. 2011;71:186-190.

13. Khan AA, Leslie WD, Lentle B, et al. Atypical femoral fractures: a teaching perspective. Can Assoc Radiol J. 2015;66:102-107.

14. Cosman F, de Beur SJ, LeBoff MS, et al; National Osteoporosis Foundation. Clinician’s Guide to Prevention and Treatment of Osteoporosis. Osteoporos Int. 2014;25:2359-2381.

15. US Preventive Services Task Force. Screening for osteoporosis: U.S. preventive services task force recommendation statement. Ann Intern Med. 2011;154:356-364.

16. World Health Organization (WHO). 1994. Assessment of fracture risk and its application to screening for postmenopausal osteoporosis: report of a WHO study group. WHO Technical Report Series, Report No. 843. WHO, Geneva, Switzerland.

17. Kanis JA, McCloskey EV, Johansson H, et al. Development and use of FRAX in osteoporosis. Osteoporos Int. 2010;21:S407-S413.

Issue
The Journal of Family Practice - 66(6)
Issue
The Journal of Family Practice - 66(6)
Page Number
395-397
Page Number
395-397
Publications
Publications
Topics
Article Type
Display Headline
Severe right hip pain
Display Headline
Severe right hip pain
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
PubMed ID
28574525
Article PDF Media

Bipolar disorder: Making the Dx, selecting the right Rx

Article Type
Changed
Wed, 06/12/2019 - 11:21
Display Headline
Bipolar disorder: Making the Dx, selecting the right Rx
 

THE CASE

A 23-year-old woman seeks medical attention at the request of her boyfriend because she’s been “miserable” for 3 weeks. In the examination room, she slouches in the chair and says her mood is low, her grades have dropped, and she no longer enjoys social gatherings or her other usual activities. She has no thoughts of suicide, no weight loss, and no somatic symptoms.

She says she is generally healthy, does not take any regular medications, and has never been pregnant. When asked about previous similar episodes, she admits to feeling this way about 3 times a year for one to 2 months at a time. She has tried different antidepressants, which haven’t helped much and have made her irritable and interfered with sleep.

When asked about mania or hypomania, she says there are short periods, roughly a couple of weeks 2 or 3 times a year, when she will get a lot of work done and can get by with little sleep. She has never gone on “spending sprees,” though, or indulged in any other unusual or dangerous behavior. And she has never been hospitalized for symptoms.

HOW WOULD YOU PROCEED WITH THIS PATIENT?

 

 

 

Bipolar disorders, over time, typically cause fluctuations in mood, activity, and energy level. If disorders go untreated, a patient’s behavior may cause considerable damage to relationships, finances, and reputations. And for some patients, the disorder can take the ultimate toll, resulting in death by suicide or accident.

Subtypes of bipolar disorder differ in the timing and severity of manic (or hypomanic) and depressive symptoms or episodes. Type I is the classic manic-depressive illness; type II is characterized by chronic treatment-resistant depression punctuated by hypomanic episodes; and cyclothymia leads to chronic fluctuations in mood. The diagnostic category “bipolar disorder not otherwise specified” applies to patients who meet some, but not all, of the criteria for other bipolar disorder subtypes.1

Prevalence. As with other mood symptoms or disorders, patients with bipolar disorder are often seen first in primary care due, in part, to barriers to obtaining psychiatric care or to avoidance of the perceived stigma in seeking such care.2 In a systematic review of patients who were interviewed randomly in primary care settings, 0.5% to 4.3% met criteria for bipolar disorder.3 The average age of onset for bipolar disorder is 15 to 19 years.4 In the United States, the prevalence of bipolar disorder type I is 1%; type II is 1.1%.3

The cause of bipolar disorder is unknown, but familial predisposition, biopsychosocial factors, and environment all seem to play a role. Children of parents with bipolar disorder have a 4% to 15% chance of receiving the same diagnosis, compared with children of parents without bipolar disorder, whose risk is only as high as 2%.5,6

Clinical presentation varies

When patients with bipolar disorder are first seen in the office, their state may be depression, mania, hypomania, or even euthymia. Keep in mind that the first 3 aberrations may indicate other disorders, either psychiatric (TABLE 1)1,4 or medical (eg, hyperthyroidism, delirium).

Verify a true depressive episode

Symptoms must last for 2 weeks and include anhedonia or depressed mood, as well as some combination of changes in sleep, increased feelings of guilt, poor concentration, changes in appetite, loss of energy, psychomotor agitation or retardation, or suicidal thoughts.1

Know the criteria for mania

True mania is a distinct period of abnormally and persistently elevated, expansive, or irritable mood, accompanied by abnormally and persistently increased activity or energy, and lasting at least one week for most of the day, nearly every day (or any duration if hospitalization is necessary).

During that time, the patient must also exhibit at least 3 or more of the following symptoms (not counting irritability, if present): 1

  • distractibility,
  • insomnia,
  • grandiosity,
  • flights of ideas,
  • increased goal-directed activity or agitation,
  • rapid/pressured speech, or
  • reckless behaviors.

How hypomania differs from mania. The symptoms of hypomania are less severe than those of mania—eg, social functioning is less impaired or is even normal, and there is no need for hospitalization. Patients may feel they have been much more productive than usual or have needed less sleep to engage in daily activities. Hypomania may be present but not reported by patients who perceive nothing wrong.1,4

 

 

 

Rule out alternate diagnoses and apply DSM-5 criteria

There are no objective tests to confirm a diagnosis of bipolar disorder. If you suspect bipolar disorder, focus your clinical evaluation on ruling out competing mental health or medical diagnoses, and on determining whether the patient’s history meets criteria for a bipolar disorder as described in the Diagnostic and Statistical Manual of Mental Disorders (DSM-5).1

Explore the patient’s psychiatric history (including hospitalizations, medications, and electroconvulsive therapy), general medical history, family history of psychiatric disorders (including suicide), and social history (including substance use and abuse). And carefully observe mental status. Confirming a diagnosis of bipolar disorder may take multiple visits, but strongly suggestive symptoms could warrant empirical treatment.

Helpful scales. The Patient Health Questionnaire (PHQ-9; https://www.uspreventiveservicestaskforce.org/Home/GetFileByID/218) and the Beck Depression Inventory (http://www.hr.ucdavis.edu/asap/pdf_files/Beck_Depression_Inventory.pdf) are useful for ruling out depressive disorders. Other scales are available, but they cannot confirm bipolar disorder. Laboratory testing selected according to patient symptoms (TABLE 24) can help rule out alternative diagnoses, but are also useful for establishing a baseline for medications.

Pharmacologic treatment: Match agents to symptoms

When treating bipolar disorder, choose a drug that targets a patient’s specific symptoms (TABLE 3).7-10 In primary care, the most commonly-used treatments for bipolar disorder type II are lamotrigine, valproic acid, and lithium.11

When to refer

Many cases of bipolar disorder type II can be managed successfully in a primary care practice, as can some cases of stable bipolar disorder type I. Psychiatric consultation may be most beneficial if the patient has recently attempted suicide or has suicidal ideation, has symptoms refractory to treatment, has poor medication adherence, or is misusing medications.

In primary care, up to 4% of patients randomly interviewed met criteria for bipolar disorder.

Even when patients are co-managed with psychiatric consultation, family physicians often ensure patients’ medication adherence, help patients understand their illness, manage overall health-related behaviors (including getting sufficient sleep), and make sure patients follow up as needed with their psychiatrist. Often, once patients have achieved equilibrium on mood-stabilizing (or other) medications, you can manage them and monitor medications with further consultation only as needed for clinical deterioration or other issues. Cognitive behavioral therapy may be useful as adjunctive treatment, particularly when patients are in active treatment.12

CASE

This case is typical for many patients with depressed mood. A few key features in the patient’s history suggest bipolar disorder type II:

  • depression that has been refractory to treatment
  • multiple failed drug treatments, with mood-related adverse effects
  • hypomania perceived as a “productive time,” and not as a problem
  • absence of overt manic symptoms.

The patient was given a diagnosis of bipolar disorder type II with current depressed mood and no evidence of acute mania. She was started on valproic acid 250 mg po tid. She reported an initial improvement in mood but stopped the medication after one month because it caused intolerable drowsiness. She was then prescribed lamotrigine progressing gradually in 2-week intervals from 25 mg to 100 mg daily. She tolerated the medication well, and after 3 months of treatment, her mood symptoms improved and she had no further episodes of depressed mood.

CORRESPONDENCE
Michael Jason Wells, MD, Department of Family and Geriatric Medicine, University of Louisville School of Medicine, 201 Abraham Flexner Way, Suite 690, Louisville, KY 40202; [email protected].

References

1. American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders, 5th ed (DSM-5). Arlington, VA: American Psychiatric Association; 2013.

2. Kilbourne AM, Goodrich DE, O’Donnell AN, et al. Integrated bipolar disorder management in primary care. Curr Psychiatry Rep. 2012;14:687-695.

3. Cerimele JM, Chwastiak LA, Dodson S, et al. The prevalence of bipolar disorder in primary care samples: a systematic review. Gen Hosp Psychiatry. 2014;36:19-25.

4. Malhi GS, Adams D, Lampe L, et al. Clinical practice recommendations for bipolar disorder. Acta Pscyhiatr Scand. 2009:119(Suppl 439):27-46.

5. Abell S, Ey J. Bipolar Disorder. Clin Pediatr. 2009;48:693-694.

6. Birmaher B, Axelson D, Monk K, et al. Lifetime psychiatric disorders of school-aged offspring of parents with bipolar disorder: the Pittsburgh Bipolar Offspring Study. Arch Gen Psychiatry. 2009;66:287-296.

7. Cipriani A, Hawton K, Stockton S, et al. Lithium in the prevention of suicide in mood disorders: updated systematic review and meta-analysis. BMJ. 2013;346:f3646.

8. De Fruyt J, Deschepper E, Audenaert K, et al. Second generation antipsychotics in the treatment of bipolar depression: a systematic review and meta-analysis. J Psychopharmacol. 2012;26:603-617.

9. Gitlin M, Frye MA. Maintenance therapies in bipolar disorders. Bipolar Disord. 2012:14(Suppl 2):51-65.

10. Labbate LA, Fava M, Rosenbaum JF, et al. Handbook of Psychiatric Drug Therapy. 6th ed. Philadelphia, Pa: Lippincott Williams & Wilkins; 2010.

11. Ostacher M, Tandon R, Suppes T. Florida Best Practice Psychotherapeutic Medication Guidelines for Adults with Bipolar Disorder: a novel, practical, patient-centered guide for clinicians. J Clin Psychiatry. 2016;77:920-926.

12. Szentagotai A, David D. The efficacy of cognitive behavioral therapy in bipolar disorder: a quantitative meta-analysis. J Clin Psychiatry. 2010;71:66-72.

Article PDF
Author and Disclosure Information

Department of Family and Geriatric Medicine, University of Louisville School of Medicine, Ky
[email protected]

The authors reported no potential conflict of interest relevant to this article.

Issue
The Journal of Family Practice - 66(6)
Publications
Topics
Page Number
375-379
Sections
Author and Disclosure Information

Department of Family and Geriatric Medicine, University of Louisville School of Medicine, Ky
[email protected]

The authors reported no potential conflict of interest relevant to this article.

Author and Disclosure Information

Department of Family and Geriatric Medicine, University of Louisville School of Medicine, Ky
[email protected]

The authors reported no potential conflict of interest relevant to this article.

Article PDF
Article PDF
 

THE CASE

A 23-year-old woman seeks medical attention at the request of her boyfriend because she’s been “miserable” for 3 weeks. In the examination room, she slouches in the chair and says her mood is low, her grades have dropped, and she no longer enjoys social gatherings or her other usual activities. She has no thoughts of suicide, no weight loss, and no somatic symptoms.

She says she is generally healthy, does not take any regular medications, and has never been pregnant. When asked about previous similar episodes, she admits to feeling this way about 3 times a year for one to 2 months at a time. She has tried different antidepressants, which haven’t helped much and have made her irritable and interfered with sleep.

When asked about mania or hypomania, she says there are short periods, roughly a couple of weeks 2 or 3 times a year, when she will get a lot of work done and can get by with little sleep. She has never gone on “spending sprees,” though, or indulged in any other unusual or dangerous behavior. And she has never been hospitalized for symptoms.

HOW WOULD YOU PROCEED WITH THIS PATIENT?

 

 

 

Bipolar disorders, over time, typically cause fluctuations in mood, activity, and energy level. If disorders go untreated, a patient’s behavior may cause considerable damage to relationships, finances, and reputations. And for some patients, the disorder can take the ultimate toll, resulting in death by suicide or accident.

Subtypes of bipolar disorder differ in the timing and severity of manic (or hypomanic) and depressive symptoms or episodes. Type I is the classic manic-depressive illness; type II is characterized by chronic treatment-resistant depression punctuated by hypomanic episodes; and cyclothymia leads to chronic fluctuations in mood. The diagnostic category “bipolar disorder not otherwise specified” applies to patients who meet some, but not all, of the criteria for other bipolar disorder subtypes.1

Prevalence. As with other mood symptoms or disorders, patients with bipolar disorder are often seen first in primary care due, in part, to barriers to obtaining psychiatric care or to avoidance of the perceived stigma in seeking such care.2 In a systematic review of patients who were interviewed randomly in primary care settings, 0.5% to 4.3% met criteria for bipolar disorder.3 The average age of onset for bipolar disorder is 15 to 19 years.4 In the United States, the prevalence of bipolar disorder type I is 1%; type II is 1.1%.3

The cause of bipolar disorder is unknown, but familial predisposition, biopsychosocial factors, and environment all seem to play a role. Children of parents with bipolar disorder have a 4% to 15% chance of receiving the same diagnosis, compared with children of parents without bipolar disorder, whose risk is only as high as 2%.5,6

Clinical presentation varies

When patients with bipolar disorder are first seen in the office, their state may be depression, mania, hypomania, or even euthymia. Keep in mind that the first 3 aberrations may indicate other disorders, either psychiatric (TABLE 1)1,4 or medical (eg, hyperthyroidism, delirium).

Verify a true depressive episode

Symptoms must last for 2 weeks and include anhedonia or depressed mood, as well as some combination of changes in sleep, increased feelings of guilt, poor concentration, changes in appetite, loss of energy, psychomotor agitation or retardation, or suicidal thoughts.1

Know the criteria for mania

True mania is a distinct period of abnormally and persistently elevated, expansive, or irritable mood, accompanied by abnormally and persistently increased activity or energy, and lasting at least one week for most of the day, nearly every day (or any duration if hospitalization is necessary).

During that time, the patient must also exhibit at least 3 or more of the following symptoms (not counting irritability, if present): 1

  • distractibility,
  • insomnia,
  • grandiosity,
  • flights of ideas,
  • increased goal-directed activity or agitation,
  • rapid/pressured speech, or
  • reckless behaviors.

How hypomania differs from mania. The symptoms of hypomania are less severe than those of mania—eg, social functioning is less impaired or is even normal, and there is no need for hospitalization. Patients may feel they have been much more productive than usual or have needed less sleep to engage in daily activities. Hypomania may be present but not reported by patients who perceive nothing wrong.1,4

 

 

 

Rule out alternate diagnoses and apply DSM-5 criteria

There are no objective tests to confirm a diagnosis of bipolar disorder. If you suspect bipolar disorder, focus your clinical evaluation on ruling out competing mental health or medical diagnoses, and on determining whether the patient’s history meets criteria for a bipolar disorder as described in the Diagnostic and Statistical Manual of Mental Disorders (DSM-5).1

Explore the patient’s psychiatric history (including hospitalizations, medications, and electroconvulsive therapy), general medical history, family history of psychiatric disorders (including suicide), and social history (including substance use and abuse). And carefully observe mental status. Confirming a diagnosis of bipolar disorder may take multiple visits, but strongly suggestive symptoms could warrant empirical treatment.

Helpful scales. The Patient Health Questionnaire (PHQ-9; https://www.uspreventiveservicestaskforce.org/Home/GetFileByID/218) and the Beck Depression Inventory (http://www.hr.ucdavis.edu/asap/pdf_files/Beck_Depression_Inventory.pdf) are useful for ruling out depressive disorders. Other scales are available, but they cannot confirm bipolar disorder. Laboratory testing selected according to patient symptoms (TABLE 24) can help rule out alternative diagnoses, but are also useful for establishing a baseline for medications.

Pharmacologic treatment: Match agents to symptoms

When treating bipolar disorder, choose a drug that targets a patient’s specific symptoms (TABLE 3).7-10 In primary care, the most commonly-used treatments for bipolar disorder type II are lamotrigine, valproic acid, and lithium.11

When to refer

Many cases of bipolar disorder type II can be managed successfully in a primary care practice, as can some cases of stable bipolar disorder type I. Psychiatric consultation may be most beneficial if the patient has recently attempted suicide or has suicidal ideation, has symptoms refractory to treatment, has poor medication adherence, or is misusing medications.

In primary care, up to 4% of patients randomly interviewed met criteria for bipolar disorder.

Even when patients are co-managed with psychiatric consultation, family physicians often ensure patients’ medication adherence, help patients understand their illness, manage overall health-related behaviors (including getting sufficient sleep), and make sure patients follow up as needed with their psychiatrist. Often, once patients have achieved equilibrium on mood-stabilizing (or other) medications, you can manage them and monitor medications with further consultation only as needed for clinical deterioration or other issues. Cognitive behavioral therapy may be useful as adjunctive treatment, particularly when patients are in active treatment.12

CASE

This case is typical for many patients with depressed mood. A few key features in the patient’s history suggest bipolar disorder type II:

  • depression that has been refractory to treatment
  • multiple failed drug treatments, with mood-related adverse effects
  • hypomania perceived as a “productive time,” and not as a problem
  • absence of overt manic symptoms.

The patient was given a diagnosis of bipolar disorder type II with current depressed mood and no evidence of acute mania. She was started on valproic acid 250 mg po tid. She reported an initial improvement in mood but stopped the medication after one month because it caused intolerable drowsiness. She was then prescribed lamotrigine progressing gradually in 2-week intervals from 25 mg to 100 mg daily. She tolerated the medication well, and after 3 months of treatment, her mood symptoms improved and she had no further episodes of depressed mood.

CORRESPONDENCE
Michael Jason Wells, MD, Department of Family and Geriatric Medicine, University of Louisville School of Medicine, 201 Abraham Flexner Way, Suite 690, Louisville, KY 40202; [email protected].

 

THE CASE

A 23-year-old woman seeks medical attention at the request of her boyfriend because she’s been “miserable” for 3 weeks. In the examination room, she slouches in the chair and says her mood is low, her grades have dropped, and she no longer enjoys social gatherings or her other usual activities. She has no thoughts of suicide, no weight loss, and no somatic symptoms.

She says she is generally healthy, does not take any regular medications, and has never been pregnant. When asked about previous similar episodes, she admits to feeling this way about 3 times a year for one to 2 months at a time. She has tried different antidepressants, which haven’t helped much and have made her irritable and interfered with sleep.

When asked about mania or hypomania, she says there are short periods, roughly a couple of weeks 2 or 3 times a year, when she will get a lot of work done and can get by with little sleep. She has never gone on “spending sprees,” though, or indulged in any other unusual or dangerous behavior. And she has never been hospitalized for symptoms.

HOW WOULD YOU PROCEED WITH THIS PATIENT?

 

 

 

Bipolar disorders, over time, typically cause fluctuations in mood, activity, and energy level. If disorders go untreated, a patient’s behavior may cause considerable damage to relationships, finances, and reputations. And for some patients, the disorder can take the ultimate toll, resulting in death by suicide or accident.

Subtypes of bipolar disorder differ in the timing and severity of manic (or hypomanic) and depressive symptoms or episodes. Type I is the classic manic-depressive illness; type II is characterized by chronic treatment-resistant depression punctuated by hypomanic episodes; and cyclothymia leads to chronic fluctuations in mood. The diagnostic category “bipolar disorder not otherwise specified” applies to patients who meet some, but not all, of the criteria for other bipolar disorder subtypes.1

Prevalence. As with other mood symptoms or disorders, patients with bipolar disorder are often seen first in primary care due, in part, to barriers to obtaining psychiatric care or to avoidance of the perceived stigma in seeking such care.2 In a systematic review of patients who were interviewed randomly in primary care settings, 0.5% to 4.3% met criteria for bipolar disorder.3 The average age of onset for bipolar disorder is 15 to 19 years.4 In the United States, the prevalence of bipolar disorder type I is 1%; type II is 1.1%.3

The cause of bipolar disorder is unknown, but familial predisposition, biopsychosocial factors, and environment all seem to play a role. Children of parents with bipolar disorder have a 4% to 15% chance of receiving the same diagnosis, compared with children of parents without bipolar disorder, whose risk is only as high as 2%.5,6

Clinical presentation varies

When patients with bipolar disorder are first seen in the office, their state may be depression, mania, hypomania, or even euthymia. Keep in mind that the first 3 aberrations may indicate other disorders, either psychiatric (TABLE 1)1,4 or medical (eg, hyperthyroidism, delirium).

Verify a true depressive episode

Symptoms must last for 2 weeks and include anhedonia or depressed mood, as well as some combination of changes in sleep, increased feelings of guilt, poor concentration, changes in appetite, loss of energy, psychomotor agitation or retardation, or suicidal thoughts.1

Know the criteria for mania

True mania is a distinct period of abnormally and persistently elevated, expansive, or irritable mood, accompanied by abnormally and persistently increased activity or energy, and lasting at least one week for most of the day, nearly every day (or any duration if hospitalization is necessary).

During that time, the patient must also exhibit at least 3 or more of the following symptoms (not counting irritability, if present): 1

  • distractibility,
  • insomnia,
  • grandiosity,
  • flights of ideas,
  • increased goal-directed activity or agitation,
  • rapid/pressured speech, or
  • reckless behaviors.

How hypomania differs from mania. The symptoms of hypomania are less severe than those of mania—eg, social functioning is less impaired or is even normal, and there is no need for hospitalization. Patients may feel they have been much more productive than usual or have needed less sleep to engage in daily activities. Hypomania may be present but not reported by patients who perceive nothing wrong.1,4

 

 

 

Rule out alternate diagnoses and apply DSM-5 criteria

There are no objective tests to confirm a diagnosis of bipolar disorder. If you suspect bipolar disorder, focus your clinical evaluation on ruling out competing mental health or medical diagnoses, and on determining whether the patient’s history meets criteria for a bipolar disorder as described in the Diagnostic and Statistical Manual of Mental Disorders (DSM-5).1

Explore the patient’s psychiatric history (including hospitalizations, medications, and electroconvulsive therapy), general medical history, family history of psychiatric disorders (including suicide), and social history (including substance use and abuse). And carefully observe mental status. Confirming a diagnosis of bipolar disorder may take multiple visits, but strongly suggestive symptoms could warrant empirical treatment.

Helpful scales. The Patient Health Questionnaire (PHQ-9; https://www.uspreventiveservicestaskforce.org/Home/GetFileByID/218) and the Beck Depression Inventory (http://www.hr.ucdavis.edu/asap/pdf_files/Beck_Depression_Inventory.pdf) are useful for ruling out depressive disorders. Other scales are available, but they cannot confirm bipolar disorder. Laboratory testing selected according to patient symptoms (TABLE 24) can help rule out alternative diagnoses, but are also useful for establishing a baseline for medications.

Pharmacologic treatment: Match agents to symptoms

When treating bipolar disorder, choose a drug that targets a patient’s specific symptoms (TABLE 3).7-10 In primary care, the most commonly-used treatments for bipolar disorder type II are lamotrigine, valproic acid, and lithium.11

When to refer

Many cases of bipolar disorder type II can be managed successfully in a primary care practice, as can some cases of stable bipolar disorder type I. Psychiatric consultation may be most beneficial if the patient has recently attempted suicide or has suicidal ideation, has symptoms refractory to treatment, has poor medication adherence, or is misusing medications.

In primary care, up to 4% of patients randomly interviewed met criteria for bipolar disorder.

Even when patients are co-managed with psychiatric consultation, family physicians often ensure patients’ medication adherence, help patients understand their illness, manage overall health-related behaviors (including getting sufficient sleep), and make sure patients follow up as needed with their psychiatrist. Often, once patients have achieved equilibrium on mood-stabilizing (or other) medications, you can manage them and monitor medications with further consultation only as needed for clinical deterioration or other issues. Cognitive behavioral therapy may be useful as adjunctive treatment, particularly when patients are in active treatment.12

CASE

This case is typical for many patients with depressed mood. A few key features in the patient’s history suggest bipolar disorder type II:

  • depression that has been refractory to treatment
  • multiple failed drug treatments, with mood-related adverse effects
  • hypomania perceived as a “productive time,” and not as a problem
  • absence of overt manic symptoms.

The patient was given a diagnosis of bipolar disorder type II with current depressed mood and no evidence of acute mania. She was started on valproic acid 250 mg po tid. She reported an initial improvement in mood but stopped the medication after one month because it caused intolerable drowsiness. She was then prescribed lamotrigine progressing gradually in 2-week intervals from 25 mg to 100 mg daily. She tolerated the medication well, and after 3 months of treatment, her mood symptoms improved and she had no further episodes of depressed mood.

CORRESPONDENCE
Michael Jason Wells, MD, Department of Family and Geriatric Medicine, University of Louisville School of Medicine, 201 Abraham Flexner Way, Suite 690, Louisville, KY 40202; [email protected].

References

1. American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders, 5th ed (DSM-5). Arlington, VA: American Psychiatric Association; 2013.

2. Kilbourne AM, Goodrich DE, O’Donnell AN, et al. Integrated bipolar disorder management in primary care. Curr Psychiatry Rep. 2012;14:687-695.

3. Cerimele JM, Chwastiak LA, Dodson S, et al. The prevalence of bipolar disorder in primary care samples: a systematic review. Gen Hosp Psychiatry. 2014;36:19-25.

4. Malhi GS, Adams D, Lampe L, et al. Clinical practice recommendations for bipolar disorder. Acta Pscyhiatr Scand. 2009:119(Suppl 439):27-46.

5. Abell S, Ey J. Bipolar Disorder. Clin Pediatr. 2009;48:693-694.

6. Birmaher B, Axelson D, Monk K, et al. Lifetime psychiatric disorders of school-aged offspring of parents with bipolar disorder: the Pittsburgh Bipolar Offspring Study. Arch Gen Psychiatry. 2009;66:287-296.

7. Cipriani A, Hawton K, Stockton S, et al. Lithium in the prevention of suicide in mood disorders: updated systematic review and meta-analysis. BMJ. 2013;346:f3646.

8. De Fruyt J, Deschepper E, Audenaert K, et al. Second generation antipsychotics in the treatment of bipolar depression: a systematic review and meta-analysis. J Psychopharmacol. 2012;26:603-617.

9. Gitlin M, Frye MA. Maintenance therapies in bipolar disorders. Bipolar Disord. 2012:14(Suppl 2):51-65.

10. Labbate LA, Fava M, Rosenbaum JF, et al. Handbook of Psychiatric Drug Therapy. 6th ed. Philadelphia, Pa: Lippincott Williams & Wilkins; 2010.

11. Ostacher M, Tandon R, Suppes T. Florida Best Practice Psychotherapeutic Medication Guidelines for Adults with Bipolar Disorder: a novel, practical, patient-centered guide for clinicians. J Clin Psychiatry. 2016;77:920-926.

12. Szentagotai A, David D. The efficacy of cognitive behavioral therapy in bipolar disorder: a quantitative meta-analysis. J Clin Psychiatry. 2010;71:66-72.

References

1. American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders, 5th ed (DSM-5). Arlington, VA: American Psychiatric Association; 2013.

2. Kilbourne AM, Goodrich DE, O’Donnell AN, et al. Integrated bipolar disorder management in primary care. Curr Psychiatry Rep. 2012;14:687-695.

3. Cerimele JM, Chwastiak LA, Dodson S, et al. The prevalence of bipolar disorder in primary care samples: a systematic review. Gen Hosp Psychiatry. 2014;36:19-25.

4. Malhi GS, Adams D, Lampe L, et al. Clinical practice recommendations for bipolar disorder. Acta Pscyhiatr Scand. 2009:119(Suppl 439):27-46.

5. Abell S, Ey J. Bipolar Disorder. Clin Pediatr. 2009;48:693-694.

6. Birmaher B, Axelson D, Monk K, et al. Lifetime psychiatric disorders of school-aged offspring of parents with bipolar disorder: the Pittsburgh Bipolar Offspring Study. Arch Gen Psychiatry. 2009;66:287-296.

7. Cipriani A, Hawton K, Stockton S, et al. Lithium in the prevention of suicide in mood disorders: updated systematic review and meta-analysis. BMJ. 2013;346:f3646.

8. De Fruyt J, Deschepper E, Audenaert K, et al. Second generation antipsychotics in the treatment of bipolar depression: a systematic review and meta-analysis. J Psychopharmacol. 2012;26:603-617.

9. Gitlin M, Frye MA. Maintenance therapies in bipolar disorders. Bipolar Disord. 2012:14(Suppl 2):51-65.

10. Labbate LA, Fava M, Rosenbaum JF, et al. Handbook of Psychiatric Drug Therapy. 6th ed. Philadelphia, Pa: Lippincott Williams & Wilkins; 2010.

11. Ostacher M, Tandon R, Suppes T. Florida Best Practice Psychotherapeutic Medication Guidelines for Adults with Bipolar Disorder: a novel, practical, patient-centered guide for clinicians. J Clin Psychiatry. 2016;77:920-926.

12. Szentagotai A, David D. The efficacy of cognitive behavioral therapy in bipolar disorder: a quantitative meta-analysis. J Clin Psychiatry. 2010;71:66-72.

Issue
The Journal of Family Practice - 66(6)
Issue
The Journal of Family Practice - 66(6)
Page Number
375-379
Page Number
375-379
Publications
Publications
Topics
Article Type
Display Headline
Bipolar disorder: Making the Dx, selecting the right Rx
Display Headline
Bipolar disorder: Making the Dx, selecting the right Rx
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
PubMed ID
28574522
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Article PDF Media

These 3 tools can help you streamline management of IBS

Article Type
Changed
Fri, 01/18/2019 - 08:47
Display Headline
These 3 tools can help you streamline management of IBS
 

CASE Amber S,* a 33-year-old woman who works on the production line at a bread factory, sought care at my health center with a several month history of non-bloody diarrhea that was increasing in frequency and urgency and was accompanied by painful abdominal bloating and cramping. She said that these symptoms were negatively impacting her interpersonal relationships, as well as her productivity at work. She reported that “almost everything” she ate upset her stomach and “goes right through her,” including fruits, vegetables, and meat, as well as greasy fast food. She had researched her symptoms on the Internet and was worried that she might have something serious like inflammatory bowel disease or cancer.

Irritable bowel syndrome (IBS) is a common functional gastrointestinal disorder (FGID) that negatively impacts the quality of life (QOL) of millions of people worldwide.1 In fact, one study of 179 people with IBS found that 76% of survey respondents reported some degree of IBS-related impairment in at least 5 domains of daily life: daily activities, comorbid psychiatric diagnoses, symptom severity, QOL, and symptom-specific cognitive affective factors related to IBS.2

Estimating prevalence and incidence is a formidable challenge given various diagnostic criteria, the influence of population selection, inclusion or exclusion of non-GI comorbidities, and various cultural influences.3 That said, it’s estimated that IBS impacts approximately 11% of the world’s population, and approximately 30% of these individuals seek treatment.1,4 While there are no significant differences in GI symptoms between those who consult physicians and those who do not, those who do seek treatment report higher pain scores, greater levels of anxiety, and a greater reduction in QOL.5

All ages affected. IBS has been reported in patients of all ages, including children and the elderly, with no definable difference reported in the frequency of subtypes (diarrhea- or constipation-predominant).

This article reviews the latest explanations, diagnostic criteria, and treatment guidelines for this challenging condition so that you can offer your patients confident care without needless testing or referral.

[polldaddy:9755564]

A lack of consensus among practicing physicians

Historically, IBS has been regarded by many primary care physicians (PCPs) as a diagnosis of exclusion. Lab tests would be ordered, nothing significant would be found, and the patient would be referred to the gastroenterologist for a definitive diagnosis.

Perceptions and misconceptions about IBS continue to abound to this day. Many are neither completely right nor wrong partly because so many triggers for IBS exist and partly because of the heretofore lack of simple, standardized criteria to diagnose the condition. Other factors contributing to the confusion are that the diagnosis of IBS is purely symptom-based and that proposals of its pathophysiology have traditionally been complex.

It is presumed that IBS shares common pathophysiologic mechanisms—including visceral hypersensitivity—with syndromes like functional dyspepsia.For example, a 2006 survey-based study of PCPs and gastroenterologists found that PCPs were less likely than gastroenterologists to believe that IBS was related to prior physical or sexual abuse, previous infection, or learned behavior, but were more likely to associate dietary factors or a linkable genetic etiology with IBS.6 Both sets of beliefs, however, may be considered correct.

Similarly, a 2009 qualitative study conducted in the Netherlands found that general practitioners (GPs) considered smoking, caffeine, diet, “hasty lifestyle,” and lack of exercise as potential triggers for IBS symptoms, while PCPs in the United Kingdom considered diet, infection, and travel to be possible triggers.7 Again, all play a role.

While GPs reported that patients should take responsibility for managing their IBS and for minimizing its impact on their daily lives, they admitted limited awareness of the extent to which IBS affected their patients’ daily living.7

A 2013 survey-based study in England determined that GPs understand the relationship between IBS and psychological symptoms including anxiety and stress, and posited that the majority of patients could be managed within primary care without referral for psychological interventions.8 Moreover, they reported that a dedicated risk assessment tool for patients with IBS would be helpful to stratify severity of disease. The study concluded that the reluctance of GPs to refer patients for evidence-based psychological treatments may prevent them from obtaining appropriate services and care.

Newer explanatory model shines light on IBS

A newer explanation that is based on 3 main hypotheses is elucidating the true nature of IBS and providing a pragmatic model for the clinical setting (FIGURE 1).9 According to the model, IBS entails the following 3 elements, which combined lead to the symptoms of IBS:

  • Altered or abnormal peripheral regulation of gut function (including sensory and secretory mechanisms)
  • Altered brain-gut signaling (including visceral hypersensitivity)
  • Psychological distress.

 

 

 

It is reasonable to consider that epigenetic changes may underlie the etiology and pathophysiology of IBS and could increase one’s susceptibility to developing the disorder. Additionally, it is presumed that IBS shares common pathophysiologic mechanisms, including visceral hypersensitivity, with other associated functional syndromes, such as functional dyspepsia.

New criteria make diagnosis on symptoms alone easier

In addition to a new explanatory model, clear criteria for diagnosing the disorder now exist, which should make it easier for PCPs to make the diagnosis without additional testing or referral. The 2016 Rome IV criteria3 provide guidelines for diagnosing the various subtypes of IBS including IBS-D (diarrhea predominant), IBS-C (constipation predominant), and IBS-M (mixed subtypes). A laboratory evaluation is really only needed for patients who fall outside the criteria or who have alarm symptoms, which include:

  • age >50 years at onset of symptoms,
  • new onset of constipation in the elderly,
  • rectal bleeding,
  • unexplained weight loss or anemia,
  • family history of organic GI disease, and
  • a palpable abdominal or rectal mass.

These symptoms should prompt referral to a gastroenterologist. Once alarm symptoms have been excluded, the diagnosis of IBS is based upon the presence of characteristic symptoms and changes in stool habits (FIGURE 23,10).

Patterns of migration. Over time, patients may migrate between subtypes, most commonly from IBS-C or IBS-D to IBS-M; switching between IBS-C and IBS-D occurs less commonly.11 Patients who meet criteria for IBS but whose bowel habits and symptoms cannot be grouped into any of these 3 categories are considered to have IBS unclassified. The Bristol Stool Form Scale (available at: https://www.niddk.nih.gov/health-information/health-communication-programs/bowel-control-awareness-campaign/Documents/Bristol_Stool_Form_Scale_508.pdf) should be used to gauge and track stool consistency.

A novel diagnostic test for IBS has been validated for differentiating patients with IBS-D from those with inflammatory bowel disease (IBD).12 The test focused on the beliefs that cytolethal distending toxin B (CdtB) is produced by bacteria that cause acute viral gastroenteritis (eg, norovirus, rotavirus), and that host antibodies to CdtB cross-react with the protein vinculin in the host gut, producing an “IBS-like phenotype.”

Additional treatment options for diarrhea-predominant IBS include antidepressants (tricyclics or SSRIs) and antispasmodics, such as dicyclomine and hyoscyamine.In a 2015 large-scale multicenter trial, both anti-CdtB and anti-vinculin antibodies were found to be significantly elevated in subjects with IBS-D compared to non-IBS subjects,12 providing evidence to support the long-held belief that viral gastroenteritis is often at the root of IBS.

Treatment aims to decrease symptoms and improve QOL

Treatment of IBS is directed at decreasing symptoms of abdominal pain and discomfort, bloating, diarrhea, and constipation while improving QOL. Therapeutic options for treatment of each symptom are listed in FIGURE 3,13,14 including several that are commonly used and have moderate efficacy, but are not currently approved by the US Food and Drug Administration for this purpose.

Current evidence-based pharmacologic guidelines from the American Gastroenterological Association (AGA) can be found at: https://www.guideline.gov/summaries/summary/49122?osrc=12. Figure 313,14 provides a few additional options not included in the AGA guidelines and presents the information in a simple schematic.

Pharmacologic therapies for IBS-D

Eluxadoline is a novel mixed mu opioid receptor agonist and delta opioid receptor antagonist developed for the treatment of IBS-D. It normalizes GI transit and defecation under conditions of environmental stress or post-inflammatory altered GI function.15 A 2016 study involving almost 2500 patients found that eluxadoline was significantly better than placebo at decreasing abdominal pain and improving stool consistency on the same day for at least half of a 26-week period.13 The most common adverse effects were nausea, constipation, and abdominal pain. Pancreatitis occurred rarely.

Rifaximin. Because GI flora play a central role in the pathophysiology of IBS, researchers have found that rifaximin, a minimally absorbed antibiotic, is a potentially important player in treatment. Two double-blind, placebo-controlled trials (TARGET 1 and TARGET 2) found that after 4 weeks of treatment, patients experienced significant improvement in global IBS symptoms including bloating, abdominal pain, and stool consistency on rifaximin vs placebo (40.7% vs 31.7%; P<.001 in the 2 studies combined).16 The incidence of adverse effects (headache, upper respiratory infection, nausea, abdominal pain, diarrhea, and urinary tract infection) was comparable to that with placebo.

Alosetron. Research has shown this selective 5-HT3 receptor antagonist to improve all IBS QOL measures, restriction of daily activities, and patient satisfaction significantly more than placebo in women.17 While initial use of alosetron in 2000 was widespread, the rare serious adverse event of ischemic colitis led to its withdrawal from the US market within a few months.18 Alosetron returned to the market in 2002 with restricted marketing (to treat only women with severe diarrhea-predominant IBS). (See Lotronex [alosetron hydrochloride] full prescribing information available at: https://lotronex.com/hcp/index.html.) Data from a 9-year risk management program subsequently found a cumulative incidence rate for ischemic colitis of 1.03 cases per 1000 patient/years.19

Current evidence suggests that targeted carbohydrate and gluten exclusion plays a role in the treatment and symptomatic improvement of patients with IBS.Other possible options include various antidepressants (tricyclics such as amitriptyline, imipramine, and nortriptyline; or selective serotonin reuptake inhibitors [SSRIs] such as citalopram, fluoxetine, and paroxetine) and antispasmodics such as dicyclomine and hyoscyamine.

 

 

 

Pharmacologic therapies for IBS-C

Linaclotide is a guanylate cyclase-C agonist with an indication for treatment of IBS-C. A double-blind, parallel-group, placebo-controlled trial found that the percentage of patients who experienced a decrease in abdominal pain was nearly 25%, with statistically significant improvements in bloating, straining, and stool consistency over a 26-week period.20 In a report on 2 phase 3 trials, researchers found that linaclotide improved global symptom scores and significantly decreased abdominal bloating and fullness, pain, cramping, and discomfort vs placebo. Diarrhea was the most commonly reported adverse event in patients with severe abdominal symptoms (18.8%-21%).21

Lubiprostone is a prostaglandin E1 analogue that activates type-2-chloride channels on the apical membrane of epithelial cells in the intestine. In a combined analysis of 2 phase 3 randomized trials, lubiprostone was administered twice daily for 12 weeks vs placebo and patients were asked to describe how they felt after the trial period. Survey responders reported significant improvements in global IBS-C symptoms (17.9% vs 10.1%; P=.001).22 A meta-analysis of studies on lubiprostone found that diarrhea, nausea, and abdominal pain were the most common adverse effects, but their occurrence was not that much greater than with placebo.23

Diet and probiotics can play a significant role

The role of dietary components in the treatment of IBS is gaining increasing attention. Such components can have a direct effect on gastric and intestinal motility, visceral sensation, immune activation, brain-gut interactions, and the microbiome. Current evidence suggests that targeted carbohydrate and gluten exclusion plays a favorable role in the treatment and symptomatic improvement of patients with IBS.24

A 2014 study conducted in Australia showed that a diet low in FODMAPs (fermentable oligosaccharides, disaccharides, monosaccharides, and polyols), which is characterized by avoiding foods containing gluten and those that are high in fructose, reduced overall GI symptom scores (including scores involving abdominal bloating, pain, and flatus) in patients with IBS compared to those consuming a normal Australian diet.25 The International Foundation for Functional Gastrointestinal Disorders’ Web site provides a detailed guide to low FODMAP foods and can be found at: http://www.aboutibs.org/low-fodmap-diet.html.

Probiotics are now commonly used in the symptomatic treatment of many upper and lower GI disorders. While much anecdotal evidence exists to support their benefit, there is a paucity of large-scale and rigorous research to provide substantial outcomes-based evidence. The theory for their use is that they support regulation of the gut microbiome, which in turn improves the imbalance between the intestinal microbiome and a dysfunctional intestinal barrier.

Probiotics are now used in the symptomatic treatment of many upper and lower GI disorders.A 2014 randomized, double-blind, placebo-controlled trial involving multispecies probiotics (a mixture of Bifidobacterium longum, B.bifidum, B.lactis, Lactobacillus acidophilus, L.rhamnosus, and Streptococcus thermophilus) found that patients who received probiotics had significantly reduced symptoms of IBS after 4 weeks compared with placebo, and modest improvement in abdominal pain and discomfort as well as bloating.26 One study involving 122 patients from 2011 found that B. bifidum MIMBb75 reduced the global assessment of IBS symptoms by -88 points (95% CI, -1.07 to -0.69) when compared with only -0.16 (95% CI, -.32 to 0.00) points in the placebo group (P<.0001).27 MIMBb75 also significantly improved the IBS symptoms of pain/discomfort, distension/bloating, urgency, and digestive disorder. And one randomized, double-blind, placebo-controlled study involving 67 patients found that QOL scores improved two-fold when patients took Saccharomyces boulardii (15.4% vs 7.0%; P<.05).28

Dried plums or prunes have been used successfully for decades for the symptomatic treatment of constipation. A single-blinded, randomized, cross-over study compared prunes 50 g/d to psyllium fiber 11 g/d and found that prunes were more efficacious (P<.05) with spontaneous bowel movements and stool consistency scores.29

Peppermint oil has been studied as an alternative therapy for symptoms of IBS, but efficacy and tolerability are concerns. A meta-analysis of randomized controlled trials with a minimum duration of 2 weeks found that compared with placebo, peppermint oil provided improvement in abdominal pain, bloating, and global symptoms, but some patients reported transient heartburn.30 A 4-week, randomized, double-blind, placebo-controlled clinical trial sponsored by IM HealthScience found a novel oral formulation of triple-enteric-coated sustained-release peppermint oil microspheres caused less heartburn than was reported in the previous study, but still significantly improved abdominal symptoms and lessened pain on defecation and fecal urgency.31

CASE Suspecting IBS-D, the FP ordered a complete blood count, tissue transglutaminase antibodies, and a stool culture, all of which were unremarkable. Ms. S has been trying to follow a low FODMAP diet and has been taking some over-the-counter probiotics with only minimal relief of abdominal bloating and cramping and no improvement in stool consistency. Her FP started her on eluxadoline 100 mg twice daily with food. After 12 weeks of therapy, she reports significant improvement in global IBS symptoms and nearly complete resolution of her diarrhea.

*Amber S is a real patient in my practice. Her name has been changed to protect her identity.

CORRESPONDENCE
Joel J. Heidelbaugh, MD, FAAFP, FACG, Ypsilanti Health Center, 200 Arnet Suite 200, Ypsilanti, MI 48198; [email protected].

References

1. Lovell RM, Ford AC. Global prevalence of and risk factors for irritable bowel syndrome: a meta-analysis. Clin Gastroenterol Hepatol. 2012;10:712-721.

2. Ballou S, Keefer L. The impact of irritable bowel syndrome on daily functioning: characterizing and understanding daily consequences of IBS. Neurogastroenterol Motil. 2017;29. Epub 2016 Oct 25.

3. Heidelbaugh J, Hungin P, eds. ROME IV: Functional Gastrointestinal Disorders for Primary Care and Non-GI Clinicians. 1st ed. Raleigh, NC: Rome Foundation, Inc.; 2016.

4. Canavan C, West J, Card T. The epidemiology of irritable bowel syndrome. Clin Epidemiol. 2014;6:71-80.

5. Lee V, Guthrie E, Robinson A, et al. Functional bowel disorders in primary care: factors associated with health-related quality of life and doctor consultation. J Psychosom Res. 2008;64:129-138.

6. Lacy BE, Rosemore J, Robertson D, et al. Physicians’ attitudes and practices in the evaluation and treatment of irritable bowel syndrome. Scand J Gastroenterol. 2006;41:892-902.

7. Casiday RE, Hungin AP, Cornford CS, et al. GPs’ explanatory models for irritable bowel syndrome: a mismatch with patient models? J Fam Pract. 2009;26:34-39.

8. Harkness EF, Harrington V, Hinder S, et al. GP perspectives of irritable bowel syndrome—an accepted illness, but management deviates from guidelines: a qualitative study. BMC Fam Pract. 2013;14:92.

9. Hungin AP, Becher A, Cayley B, et al. Irritable bowel syndrome: an integrated explanatory model for clinical practice. Neurogastroenterol Motil. 2015;27:750-753.

10. Lacy BE, Mearin F, Chang L, et al. Bowel Disorders. Gastroenterol. 2016;150:1393-1407.

11. Engsbro AL, Simren M, Bytzer P. Short-term stability of subtypes in the irritable bowel syndrome: prospective evaluation using the Rome III classification. Aliment Pharmacol Ther. 2012;35:350-359.

12. Pimentel M, Morales W, Rezaie A, et al. Development and validation of a biomarker for diarrhea-predominant irritable bowel syndrome in human subjects. PLoS One. 2015;10:e0126438.

13. Lembo AJ, Lacy BE, Zuckerman MJ, et al. Eluxadoline for irritable bowel syndrome with diarrhea. N Engl J Med. 2016;374:242-253.

14. Ford AC, Quigley EM, Lacy BE, et al. Efficacy of prebiotics, probiotics, and synbiotics in irritable bowel syndrome and chronic idiopathic constipation: systematic review and meta-analysis. Am J Gastroenterol. 2014;109:1547-1561.

15. Fujita W, Gomes I, Dove LS, et al. Molecular characterization of eluxadoline as a potential ligand targeting mu-delta opioid receptor heteromers. Biochem Pharmacol. 2014;92:448-456.

16. Pimentel M, Lembo A, Chey WD, et al, for the TARGET Study Group. Rifaximin therapy for patients with irritable bowel syndrome without constipation. N Engl J Med. 2011;364:22-32.

17. Cremonini F, Nicandro JP, Atkinson V, et al. Randomised clinical trial: alosetron improves quality of life and reduces restriction of daily activities in women with severe diarrhoea-predominant IBS. Aliment Pharmacol Ther. 2012;36:437-448.

18. Lewis JH. Alosetron for severe diarrhea-predominant irritable bowel syndrome: safety and efficacy in perspective. Expert Rev Gastroenterol Hepatol. 2010;4:13-29.

19. Tong K, Nicandro JP, Shringarpure R, et al. A 9-year evaluation of temporal trends in alosetron postmarketing safety under the risk management program. Therap Adv Gastroenterol. 2013;6:344-357.

20. Chey WD, Lembo AJ, Lavins BJ, et al. Linaclotide for irritable bowel syndrome with constipation: a 26-week, randomized, double-blind, placebo-controlled trial to evaluate efficacy and safety. Am J Gastroenterol. 2012;107:1702-1712.

21. Rao SS, Quigley EM, Shiff SJ, et al. Effect of linaclotide on severe abdominal symptoms in patients with irritable bowel syndrome with constipation. Clin Gastroenterol Hepatol. 2014;12:616-623.

22. Drossman DA, Chey WD, Johanson JF, et al. Clinical trial: lubiprostone in patients with constipation-associated irritable bowel syndrome—results of two randomized, placebo-controlled studies. Aliment Pharmacol Ther. 2009;29:329-341.

23. Lacy BE, Chey WD. Lubiprostone: chronic constipation and irritable bowel syndrome with constipation. Expert Opin Pharmacother. 2009;10:143-152.

24. Spencer M, Chey WD, Eswaran S. Dietary Renaissance in IBS: has food replaced medications as a primary treatment strategy? Curr Treat Options Gastroenterol. 2014;12:424-440.

25. Halmos EP, Power VA, Shepherd SJ, et al. A diet low in FODMAPs reduces symptoms of irritable bowel syndrome. Gastroenterology. 2014;146:67-75.

26. Yoon JS, Sohn W, Lee OY, et al. Effect of multispecies probiotics on irritable bowel syndrome: a randomized, double-blind, placebo-controlled trial. J Gastroenterol Hepatol. 2014;29:52-59.

27. Guglielmetti S, Mora D, Gschwender M, et al. Randomised clinical trial: Bifidobacterium bifidum MIMBb75 significantly alleviates irritable bowel syndrome and improves quality of life—a double-blind, placebo-controlled study. Aliment Pharmacol Ther. 2011;33:1123-1132.

28. Choi CH, Jo SY, Park HJ, et al. A randomized, double-blind, placebo-controlled multicenter trial of saccharomyces boulardii in irritable bowel syndrome: effect on quality of life. J Clin Gastroenterol. 2011;45:679-683.

29. Attaluri A, Donahoe R, Valestin J, et al. Randomised clinical trial: dried plums (prunes) vs. psyllium for constipation. Aliment Pharmacol Ther. 2011;33:822-828.

30. Khanna R, MacDonald JK, Levesque BG. Peppermint oil for the treatment of irritable bowel syndrome: a systematic review and meta-analysis. J Clin Gastroenterol. 2014;48:505-512.

31. Cash BD, Epstein MS, Shah SM. A novel delivery system of peppermint oil is an effective therapy for irritable bowel syndrome symptoms. Dig Dis Sci. 2016;61:560-571.

Article PDF
Author and Disclosure Information

Departments of Family Medicine and Urology, University of Michigan Medical School, Ann Arbor
[email protected]

The author is the co-editor and co-author of ROME IV: Functional Gastrointestinal Disorders for Primary Care and Non-GI Clinicians, which is described in this review.

Issue
The Journal of Family Practice - 66(6)
Publications
Topics
Page Number
346-348,350-353
Sections
Author and Disclosure Information

Departments of Family Medicine and Urology, University of Michigan Medical School, Ann Arbor
[email protected]

The author is the co-editor and co-author of ROME IV: Functional Gastrointestinal Disorders for Primary Care and Non-GI Clinicians, which is described in this review.

Author and Disclosure Information

Departments of Family Medicine and Urology, University of Michigan Medical School, Ann Arbor
[email protected]

The author is the co-editor and co-author of ROME IV: Functional Gastrointestinal Disorders for Primary Care and Non-GI Clinicians, which is described in this review.

Article PDF
Article PDF
 

CASE Amber S,* a 33-year-old woman who works on the production line at a bread factory, sought care at my health center with a several month history of non-bloody diarrhea that was increasing in frequency and urgency and was accompanied by painful abdominal bloating and cramping. She said that these symptoms were negatively impacting her interpersonal relationships, as well as her productivity at work. She reported that “almost everything” she ate upset her stomach and “goes right through her,” including fruits, vegetables, and meat, as well as greasy fast food. She had researched her symptoms on the Internet and was worried that she might have something serious like inflammatory bowel disease or cancer.

Irritable bowel syndrome (IBS) is a common functional gastrointestinal disorder (FGID) that negatively impacts the quality of life (QOL) of millions of people worldwide.1 In fact, one study of 179 people with IBS found that 76% of survey respondents reported some degree of IBS-related impairment in at least 5 domains of daily life: daily activities, comorbid psychiatric diagnoses, symptom severity, QOL, and symptom-specific cognitive affective factors related to IBS.2

Estimating prevalence and incidence is a formidable challenge given various diagnostic criteria, the influence of population selection, inclusion or exclusion of non-GI comorbidities, and various cultural influences.3 That said, it’s estimated that IBS impacts approximately 11% of the world’s population, and approximately 30% of these individuals seek treatment.1,4 While there are no significant differences in GI symptoms between those who consult physicians and those who do not, those who do seek treatment report higher pain scores, greater levels of anxiety, and a greater reduction in QOL.5

All ages affected. IBS has been reported in patients of all ages, including children and the elderly, with no definable difference reported in the frequency of subtypes (diarrhea- or constipation-predominant).

This article reviews the latest explanations, diagnostic criteria, and treatment guidelines for this challenging condition so that you can offer your patients confident care without needless testing or referral.

[polldaddy:9755564]

A lack of consensus among practicing physicians

Historically, IBS has been regarded by many primary care physicians (PCPs) as a diagnosis of exclusion. Lab tests would be ordered, nothing significant would be found, and the patient would be referred to the gastroenterologist for a definitive diagnosis.

Perceptions and misconceptions about IBS continue to abound to this day. Many are neither completely right nor wrong partly because so many triggers for IBS exist and partly because of the heretofore lack of simple, standardized criteria to diagnose the condition. Other factors contributing to the confusion are that the diagnosis of IBS is purely symptom-based and that proposals of its pathophysiology have traditionally been complex.

It is presumed that IBS shares common pathophysiologic mechanisms—including visceral hypersensitivity—with syndromes like functional dyspepsia.For example, a 2006 survey-based study of PCPs and gastroenterologists found that PCPs were less likely than gastroenterologists to believe that IBS was related to prior physical or sexual abuse, previous infection, or learned behavior, but were more likely to associate dietary factors or a linkable genetic etiology with IBS.6 Both sets of beliefs, however, may be considered correct.

Similarly, a 2009 qualitative study conducted in the Netherlands found that general practitioners (GPs) considered smoking, caffeine, diet, “hasty lifestyle,” and lack of exercise as potential triggers for IBS symptoms, while PCPs in the United Kingdom considered diet, infection, and travel to be possible triggers.7 Again, all play a role.

While GPs reported that patients should take responsibility for managing their IBS and for minimizing its impact on their daily lives, they admitted limited awareness of the extent to which IBS affected their patients’ daily living.7

A 2013 survey-based study in England determined that GPs understand the relationship between IBS and psychological symptoms including anxiety and stress, and posited that the majority of patients could be managed within primary care without referral for psychological interventions.8 Moreover, they reported that a dedicated risk assessment tool for patients with IBS would be helpful to stratify severity of disease. The study concluded that the reluctance of GPs to refer patients for evidence-based psychological treatments may prevent them from obtaining appropriate services and care.

Newer explanatory model shines light on IBS

A newer explanation that is based on 3 main hypotheses is elucidating the true nature of IBS and providing a pragmatic model for the clinical setting (FIGURE 1).9 According to the model, IBS entails the following 3 elements, which combined lead to the symptoms of IBS:

  • Altered or abnormal peripheral regulation of gut function (including sensory and secretory mechanisms)
  • Altered brain-gut signaling (including visceral hypersensitivity)
  • Psychological distress.

 

 

 

It is reasonable to consider that epigenetic changes may underlie the etiology and pathophysiology of IBS and could increase one’s susceptibility to developing the disorder. Additionally, it is presumed that IBS shares common pathophysiologic mechanisms, including visceral hypersensitivity, with other associated functional syndromes, such as functional dyspepsia.

New criteria make diagnosis on symptoms alone easier

In addition to a new explanatory model, clear criteria for diagnosing the disorder now exist, which should make it easier for PCPs to make the diagnosis without additional testing or referral. The 2016 Rome IV criteria3 provide guidelines for diagnosing the various subtypes of IBS including IBS-D (diarrhea predominant), IBS-C (constipation predominant), and IBS-M (mixed subtypes). A laboratory evaluation is really only needed for patients who fall outside the criteria or who have alarm symptoms, which include:

  • age >50 years at onset of symptoms,
  • new onset of constipation in the elderly,
  • rectal bleeding,
  • unexplained weight loss or anemia,
  • family history of organic GI disease, and
  • a palpable abdominal or rectal mass.

These symptoms should prompt referral to a gastroenterologist. Once alarm symptoms have been excluded, the diagnosis of IBS is based upon the presence of characteristic symptoms and changes in stool habits (FIGURE 23,10).

Patterns of migration. Over time, patients may migrate between subtypes, most commonly from IBS-C or IBS-D to IBS-M; switching between IBS-C and IBS-D occurs less commonly.11 Patients who meet criteria for IBS but whose bowel habits and symptoms cannot be grouped into any of these 3 categories are considered to have IBS unclassified. The Bristol Stool Form Scale (available at: https://www.niddk.nih.gov/health-information/health-communication-programs/bowel-control-awareness-campaign/Documents/Bristol_Stool_Form_Scale_508.pdf) should be used to gauge and track stool consistency.

A novel diagnostic test for IBS has been validated for differentiating patients with IBS-D from those with inflammatory bowel disease (IBD).12 The test focused on the beliefs that cytolethal distending toxin B (CdtB) is produced by bacteria that cause acute viral gastroenteritis (eg, norovirus, rotavirus), and that host antibodies to CdtB cross-react with the protein vinculin in the host gut, producing an “IBS-like phenotype.”

Additional treatment options for diarrhea-predominant IBS include antidepressants (tricyclics or SSRIs) and antispasmodics, such as dicyclomine and hyoscyamine.In a 2015 large-scale multicenter trial, both anti-CdtB and anti-vinculin antibodies were found to be significantly elevated in subjects with IBS-D compared to non-IBS subjects,12 providing evidence to support the long-held belief that viral gastroenteritis is often at the root of IBS.

Treatment aims to decrease symptoms and improve QOL

Treatment of IBS is directed at decreasing symptoms of abdominal pain and discomfort, bloating, diarrhea, and constipation while improving QOL. Therapeutic options for treatment of each symptom are listed in FIGURE 3,13,14 including several that are commonly used and have moderate efficacy, but are not currently approved by the US Food and Drug Administration for this purpose.

Current evidence-based pharmacologic guidelines from the American Gastroenterological Association (AGA) can be found at: https://www.guideline.gov/summaries/summary/49122?osrc=12. Figure 313,14 provides a few additional options not included in the AGA guidelines and presents the information in a simple schematic.

Pharmacologic therapies for IBS-D

Eluxadoline is a novel mixed mu opioid receptor agonist and delta opioid receptor antagonist developed for the treatment of IBS-D. It normalizes GI transit and defecation under conditions of environmental stress or post-inflammatory altered GI function.15 A 2016 study involving almost 2500 patients found that eluxadoline was significantly better than placebo at decreasing abdominal pain and improving stool consistency on the same day for at least half of a 26-week period.13 The most common adverse effects were nausea, constipation, and abdominal pain. Pancreatitis occurred rarely.

Rifaximin. Because GI flora play a central role in the pathophysiology of IBS, researchers have found that rifaximin, a minimally absorbed antibiotic, is a potentially important player in treatment. Two double-blind, placebo-controlled trials (TARGET 1 and TARGET 2) found that after 4 weeks of treatment, patients experienced significant improvement in global IBS symptoms including bloating, abdominal pain, and stool consistency on rifaximin vs placebo (40.7% vs 31.7%; P<.001 in the 2 studies combined).16 The incidence of adverse effects (headache, upper respiratory infection, nausea, abdominal pain, diarrhea, and urinary tract infection) was comparable to that with placebo.

Alosetron. Research has shown this selective 5-HT3 receptor antagonist to improve all IBS QOL measures, restriction of daily activities, and patient satisfaction significantly more than placebo in women.17 While initial use of alosetron in 2000 was widespread, the rare serious adverse event of ischemic colitis led to its withdrawal from the US market within a few months.18 Alosetron returned to the market in 2002 with restricted marketing (to treat only women with severe diarrhea-predominant IBS). (See Lotronex [alosetron hydrochloride] full prescribing information available at: https://lotronex.com/hcp/index.html.) Data from a 9-year risk management program subsequently found a cumulative incidence rate for ischemic colitis of 1.03 cases per 1000 patient/years.19

Current evidence suggests that targeted carbohydrate and gluten exclusion plays a role in the treatment and symptomatic improvement of patients with IBS.Other possible options include various antidepressants (tricyclics such as amitriptyline, imipramine, and nortriptyline; or selective serotonin reuptake inhibitors [SSRIs] such as citalopram, fluoxetine, and paroxetine) and antispasmodics such as dicyclomine and hyoscyamine.

 

 

 

Pharmacologic therapies for IBS-C

Linaclotide is a guanylate cyclase-C agonist with an indication for treatment of IBS-C. A double-blind, parallel-group, placebo-controlled trial found that the percentage of patients who experienced a decrease in abdominal pain was nearly 25%, with statistically significant improvements in bloating, straining, and stool consistency over a 26-week period.20 In a report on 2 phase 3 trials, researchers found that linaclotide improved global symptom scores and significantly decreased abdominal bloating and fullness, pain, cramping, and discomfort vs placebo. Diarrhea was the most commonly reported adverse event in patients with severe abdominal symptoms (18.8%-21%).21

Lubiprostone is a prostaglandin E1 analogue that activates type-2-chloride channels on the apical membrane of epithelial cells in the intestine. In a combined analysis of 2 phase 3 randomized trials, lubiprostone was administered twice daily for 12 weeks vs placebo and patients were asked to describe how they felt after the trial period. Survey responders reported significant improvements in global IBS-C symptoms (17.9% vs 10.1%; P=.001).22 A meta-analysis of studies on lubiprostone found that diarrhea, nausea, and abdominal pain were the most common adverse effects, but their occurrence was not that much greater than with placebo.23

Diet and probiotics can play a significant role

The role of dietary components in the treatment of IBS is gaining increasing attention. Such components can have a direct effect on gastric and intestinal motility, visceral sensation, immune activation, brain-gut interactions, and the microbiome. Current evidence suggests that targeted carbohydrate and gluten exclusion plays a favorable role in the treatment and symptomatic improvement of patients with IBS.24

A 2014 study conducted in Australia showed that a diet low in FODMAPs (fermentable oligosaccharides, disaccharides, monosaccharides, and polyols), which is characterized by avoiding foods containing gluten and those that are high in fructose, reduced overall GI symptom scores (including scores involving abdominal bloating, pain, and flatus) in patients with IBS compared to those consuming a normal Australian diet.25 The International Foundation for Functional Gastrointestinal Disorders’ Web site provides a detailed guide to low FODMAP foods and can be found at: http://www.aboutibs.org/low-fodmap-diet.html.

Probiotics are now commonly used in the symptomatic treatment of many upper and lower GI disorders. While much anecdotal evidence exists to support their benefit, there is a paucity of large-scale and rigorous research to provide substantial outcomes-based evidence. The theory for their use is that they support regulation of the gut microbiome, which in turn improves the imbalance between the intestinal microbiome and a dysfunctional intestinal barrier.

Probiotics are now used in the symptomatic treatment of many upper and lower GI disorders.A 2014 randomized, double-blind, placebo-controlled trial involving multispecies probiotics (a mixture of Bifidobacterium longum, B.bifidum, B.lactis, Lactobacillus acidophilus, L.rhamnosus, and Streptococcus thermophilus) found that patients who received probiotics had significantly reduced symptoms of IBS after 4 weeks compared with placebo, and modest improvement in abdominal pain and discomfort as well as bloating.26 One study involving 122 patients from 2011 found that B. bifidum MIMBb75 reduced the global assessment of IBS symptoms by -88 points (95% CI, -1.07 to -0.69) when compared with only -0.16 (95% CI, -.32 to 0.00) points in the placebo group (P<.0001).27 MIMBb75 also significantly improved the IBS symptoms of pain/discomfort, distension/bloating, urgency, and digestive disorder. And one randomized, double-blind, placebo-controlled study involving 67 patients found that QOL scores improved two-fold when patients took Saccharomyces boulardii (15.4% vs 7.0%; P<.05).28

Dried plums or prunes have been used successfully for decades for the symptomatic treatment of constipation. A single-blinded, randomized, cross-over study compared prunes 50 g/d to psyllium fiber 11 g/d and found that prunes were more efficacious (P<.05) with spontaneous bowel movements and stool consistency scores.29

Peppermint oil has been studied as an alternative therapy for symptoms of IBS, but efficacy and tolerability are concerns. A meta-analysis of randomized controlled trials with a minimum duration of 2 weeks found that compared with placebo, peppermint oil provided improvement in abdominal pain, bloating, and global symptoms, but some patients reported transient heartburn.30 A 4-week, randomized, double-blind, placebo-controlled clinical trial sponsored by IM HealthScience found a novel oral formulation of triple-enteric-coated sustained-release peppermint oil microspheres caused less heartburn than was reported in the previous study, but still significantly improved abdominal symptoms and lessened pain on defecation and fecal urgency.31

CASE Suspecting IBS-D, the FP ordered a complete blood count, tissue transglutaminase antibodies, and a stool culture, all of which were unremarkable. Ms. S has been trying to follow a low FODMAP diet and has been taking some over-the-counter probiotics with only minimal relief of abdominal bloating and cramping and no improvement in stool consistency. Her FP started her on eluxadoline 100 mg twice daily with food. After 12 weeks of therapy, she reports significant improvement in global IBS symptoms and nearly complete resolution of her diarrhea.

*Amber S is a real patient in my practice. Her name has been changed to protect her identity.

CORRESPONDENCE
Joel J. Heidelbaugh, MD, FAAFP, FACG, Ypsilanti Health Center, 200 Arnet Suite 200, Ypsilanti, MI 48198; [email protected].

 

CASE Amber S,* a 33-year-old woman who works on the production line at a bread factory, sought care at my health center with a several month history of non-bloody diarrhea that was increasing in frequency and urgency and was accompanied by painful abdominal bloating and cramping. She said that these symptoms were negatively impacting her interpersonal relationships, as well as her productivity at work. She reported that “almost everything” she ate upset her stomach and “goes right through her,” including fruits, vegetables, and meat, as well as greasy fast food. She had researched her symptoms on the Internet and was worried that she might have something serious like inflammatory bowel disease or cancer.

Irritable bowel syndrome (IBS) is a common functional gastrointestinal disorder (FGID) that negatively impacts the quality of life (QOL) of millions of people worldwide.1 In fact, one study of 179 people with IBS found that 76% of survey respondents reported some degree of IBS-related impairment in at least 5 domains of daily life: daily activities, comorbid psychiatric diagnoses, symptom severity, QOL, and symptom-specific cognitive affective factors related to IBS.2

Estimating prevalence and incidence is a formidable challenge given various diagnostic criteria, the influence of population selection, inclusion or exclusion of non-GI comorbidities, and various cultural influences.3 That said, it’s estimated that IBS impacts approximately 11% of the world’s population, and approximately 30% of these individuals seek treatment.1,4 While there are no significant differences in GI symptoms between those who consult physicians and those who do not, those who do seek treatment report higher pain scores, greater levels of anxiety, and a greater reduction in QOL.5

All ages affected. IBS has been reported in patients of all ages, including children and the elderly, with no definable difference reported in the frequency of subtypes (diarrhea- or constipation-predominant).

This article reviews the latest explanations, diagnostic criteria, and treatment guidelines for this challenging condition so that you can offer your patients confident care without needless testing or referral.

[polldaddy:9755564]

A lack of consensus among practicing physicians

Historically, IBS has been regarded by many primary care physicians (PCPs) as a diagnosis of exclusion. Lab tests would be ordered, nothing significant would be found, and the patient would be referred to the gastroenterologist for a definitive diagnosis.

Perceptions and misconceptions about IBS continue to abound to this day. Many are neither completely right nor wrong partly because so many triggers for IBS exist and partly because of the heretofore lack of simple, standardized criteria to diagnose the condition. Other factors contributing to the confusion are that the diagnosis of IBS is purely symptom-based and that proposals of its pathophysiology have traditionally been complex.

It is presumed that IBS shares common pathophysiologic mechanisms—including visceral hypersensitivity—with syndromes like functional dyspepsia.For example, a 2006 survey-based study of PCPs and gastroenterologists found that PCPs were less likely than gastroenterologists to believe that IBS was related to prior physical or sexual abuse, previous infection, or learned behavior, but were more likely to associate dietary factors or a linkable genetic etiology with IBS.6 Both sets of beliefs, however, may be considered correct.

Similarly, a 2009 qualitative study conducted in the Netherlands found that general practitioners (GPs) considered smoking, caffeine, diet, “hasty lifestyle,” and lack of exercise as potential triggers for IBS symptoms, while PCPs in the United Kingdom considered diet, infection, and travel to be possible triggers.7 Again, all play a role.

While GPs reported that patients should take responsibility for managing their IBS and for minimizing its impact on their daily lives, they admitted limited awareness of the extent to which IBS affected their patients’ daily living.7

A 2013 survey-based study in England determined that GPs understand the relationship between IBS and psychological symptoms including anxiety and stress, and posited that the majority of patients could be managed within primary care without referral for psychological interventions.8 Moreover, they reported that a dedicated risk assessment tool for patients with IBS would be helpful to stratify severity of disease. The study concluded that the reluctance of GPs to refer patients for evidence-based psychological treatments may prevent them from obtaining appropriate services and care.

Newer explanatory model shines light on IBS

A newer explanation that is based on 3 main hypotheses is elucidating the true nature of IBS and providing a pragmatic model for the clinical setting (FIGURE 1).9 According to the model, IBS entails the following 3 elements, which combined lead to the symptoms of IBS:

  • Altered or abnormal peripheral regulation of gut function (including sensory and secretory mechanisms)
  • Altered brain-gut signaling (including visceral hypersensitivity)
  • Psychological distress.

 

 

 

It is reasonable to consider that epigenetic changes may underlie the etiology and pathophysiology of IBS and could increase one’s susceptibility to developing the disorder. Additionally, it is presumed that IBS shares common pathophysiologic mechanisms, including visceral hypersensitivity, with other associated functional syndromes, such as functional dyspepsia.

New criteria make diagnosis on symptoms alone easier

In addition to a new explanatory model, clear criteria for diagnosing the disorder now exist, which should make it easier for PCPs to make the diagnosis without additional testing or referral. The 2016 Rome IV criteria3 provide guidelines for diagnosing the various subtypes of IBS including IBS-D (diarrhea predominant), IBS-C (constipation predominant), and IBS-M (mixed subtypes). A laboratory evaluation is really only needed for patients who fall outside the criteria or who have alarm symptoms, which include:

  • age >50 years at onset of symptoms,
  • new onset of constipation in the elderly,
  • rectal bleeding,
  • unexplained weight loss or anemia,
  • family history of organic GI disease, and
  • a palpable abdominal or rectal mass.

These symptoms should prompt referral to a gastroenterologist. Once alarm symptoms have been excluded, the diagnosis of IBS is based upon the presence of characteristic symptoms and changes in stool habits (FIGURE 23,10).

Patterns of migration. Over time, patients may migrate between subtypes, most commonly from IBS-C or IBS-D to IBS-M; switching between IBS-C and IBS-D occurs less commonly.11 Patients who meet criteria for IBS but whose bowel habits and symptoms cannot be grouped into any of these 3 categories are considered to have IBS unclassified. The Bristol Stool Form Scale (available at: https://www.niddk.nih.gov/health-information/health-communication-programs/bowel-control-awareness-campaign/Documents/Bristol_Stool_Form_Scale_508.pdf) should be used to gauge and track stool consistency.

A novel diagnostic test for IBS has been validated for differentiating patients with IBS-D from those with inflammatory bowel disease (IBD).12 The test focused on the beliefs that cytolethal distending toxin B (CdtB) is produced by bacteria that cause acute viral gastroenteritis (eg, norovirus, rotavirus), and that host antibodies to CdtB cross-react with the protein vinculin in the host gut, producing an “IBS-like phenotype.”

Additional treatment options for diarrhea-predominant IBS include antidepressants (tricyclics or SSRIs) and antispasmodics, such as dicyclomine and hyoscyamine.In a 2015 large-scale multicenter trial, both anti-CdtB and anti-vinculin antibodies were found to be significantly elevated in subjects with IBS-D compared to non-IBS subjects,12 providing evidence to support the long-held belief that viral gastroenteritis is often at the root of IBS.

Treatment aims to decrease symptoms and improve QOL

Treatment of IBS is directed at decreasing symptoms of abdominal pain and discomfort, bloating, diarrhea, and constipation while improving QOL. Therapeutic options for treatment of each symptom are listed in FIGURE 3,13,14 including several that are commonly used and have moderate efficacy, but are not currently approved by the US Food and Drug Administration for this purpose.

Current evidence-based pharmacologic guidelines from the American Gastroenterological Association (AGA) can be found at: https://www.guideline.gov/summaries/summary/49122?osrc=12. Figure 313,14 provides a few additional options not included in the AGA guidelines and presents the information in a simple schematic.

Pharmacologic therapies for IBS-D

Eluxadoline is a novel mixed mu opioid receptor agonist and delta opioid receptor antagonist developed for the treatment of IBS-D. It normalizes GI transit and defecation under conditions of environmental stress or post-inflammatory altered GI function.15 A 2016 study involving almost 2500 patients found that eluxadoline was significantly better than placebo at decreasing abdominal pain and improving stool consistency on the same day for at least half of a 26-week period.13 The most common adverse effects were nausea, constipation, and abdominal pain. Pancreatitis occurred rarely.

Rifaximin. Because GI flora play a central role in the pathophysiology of IBS, researchers have found that rifaximin, a minimally absorbed antibiotic, is a potentially important player in treatment. Two double-blind, placebo-controlled trials (TARGET 1 and TARGET 2) found that after 4 weeks of treatment, patients experienced significant improvement in global IBS symptoms including bloating, abdominal pain, and stool consistency on rifaximin vs placebo (40.7% vs 31.7%; P<.001 in the 2 studies combined).16 The incidence of adverse effects (headache, upper respiratory infection, nausea, abdominal pain, diarrhea, and urinary tract infection) was comparable to that with placebo.

Alosetron. Research has shown this selective 5-HT3 receptor antagonist to improve all IBS QOL measures, restriction of daily activities, and patient satisfaction significantly more than placebo in women.17 While initial use of alosetron in 2000 was widespread, the rare serious adverse event of ischemic colitis led to its withdrawal from the US market within a few months.18 Alosetron returned to the market in 2002 with restricted marketing (to treat only women with severe diarrhea-predominant IBS). (See Lotronex [alosetron hydrochloride] full prescribing information available at: https://lotronex.com/hcp/index.html.) Data from a 9-year risk management program subsequently found a cumulative incidence rate for ischemic colitis of 1.03 cases per 1000 patient/years.19

Current evidence suggests that targeted carbohydrate and gluten exclusion plays a role in the treatment and symptomatic improvement of patients with IBS.Other possible options include various antidepressants (tricyclics such as amitriptyline, imipramine, and nortriptyline; or selective serotonin reuptake inhibitors [SSRIs] such as citalopram, fluoxetine, and paroxetine) and antispasmodics such as dicyclomine and hyoscyamine.

 

 

 

Pharmacologic therapies for IBS-C

Linaclotide is a guanylate cyclase-C agonist with an indication for treatment of IBS-C. A double-blind, parallel-group, placebo-controlled trial found that the percentage of patients who experienced a decrease in abdominal pain was nearly 25%, with statistically significant improvements in bloating, straining, and stool consistency over a 26-week period.20 In a report on 2 phase 3 trials, researchers found that linaclotide improved global symptom scores and significantly decreased abdominal bloating and fullness, pain, cramping, and discomfort vs placebo. Diarrhea was the most commonly reported adverse event in patients with severe abdominal symptoms (18.8%-21%).21

Lubiprostone is a prostaglandin E1 analogue that activates type-2-chloride channels on the apical membrane of epithelial cells in the intestine. In a combined analysis of 2 phase 3 randomized trials, lubiprostone was administered twice daily for 12 weeks vs placebo and patients were asked to describe how they felt after the trial period. Survey responders reported significant improvements in global IBS-C symptoms (17.9% vs 10.1%; P=.001).22 A meta-analysis of studies on lubiprostone found that diarrhea, nausea, and abdominal pain were the most common adverse effects, but their occurrence was not that much greater than with placebo.23

Diet and probiotics can play a significant role

The role of dietary components in the treatment of IBS is gaining increasing attention. Such components can have a direct effect on gastric and intestinal motility, visceral sensation, immune activation, brain-gut interactions, and the microbiome. Current evidence suggests that targeted carbohydrate and gluten exclusion plays a favorable role in the treatment and symptomatic improvement of patients with IBS.24

A 2014 study conducted in Australia showed that a diet low in FODMAPs (fermentable oligosaccharides, disaccharides, monosaccharides, and polyols), which is characterized by avoiding foods containing gluten and those that are high in fructose, reduced overall GI symptom scores (including scores involving abdominal bloating, pain, and flatus) in patients with IBS compared to those consuming a normal Australian diet.25 The International Foundation for Functional Gastrointestinal Disorders’ Web site provides a detailed guide to low FODMAP foods and can be found at: http://www.aboutibs.org/low-fodmap-diet.html.

Probiotics are now commonly used in the symptomatic treatment of many upper and lower GI disorders. While much anecdotal evidence exists to support their benefit, there is a paucity of large-scale and rigorous research to provide substantial outcomes-based evidence. The theory for their use is that they support regulation of the gut microbiome, which in turn improves the imbalance between the intestinal microbiome and a dysfunctional intestinal barrier.

Probiotics are now used in the symptomatic treatment of many upper and lower GI disorders.A 2014 randomized, double-blind, placebo-controlled trial involving multispecies probiotics (a mixture of Bifidobacterium longum, B.bifidum, B.lactis, Lactobacillus acidophilus, L.rhamnosus, and Streptococcus thermophilus) found that patients who received probiotics had significantly reduced symptoms of IBS after 4 weeks compared with placebo, and modest improvement in abdominal pain and discomfort as well as bloating.26 One study involving 122 patients from 2011 found that B. bifidum MIMBb75 reduced the global assessment of IBS symptoms by -88 points (95% CI, -1.07 to -0.69) when compared with only -0.16 (95% CI, -.32 to 0.00) points in the placebo group (P<.0001).27 MIMBb75 also significantly improved the IBS symptoms of pain/discomfort, distension/bloating, urgency, and digestive disorder. And one randomized, double-blind, placebo-controlled study involving 67 patients found that QOL scores improved two-fold when patients took Saccharomyces boulardii (15.4% vs 7.0%; P<.05).28

Dried plums or prunes have been used successfully for decades for the symptomatic treatment of constipation. A single-blinded, randomized, cross-over study compared prunes 50 g/d to psyllium fiber 11 g/d and found that prunes were more efficacious (P<.05) with spontaneous bowel movements and stool consistency scores.29

Peppermint oil has been studied as an alternative therapy for symptoms of IBS, but efficacy and tolerability are concerns. A meta-analysis of randomized controlled trials with a minimum duration of 2 weeks found that compared with placebo, peppermint oil provided improvement in abdominal pain, bloating, and global symptoms, but some patients reported transient heartburn.30 A 4-week, randomized, double-blind, placebo-controlled clinical trial sponsored by IM HealthScience found a novel oral formulation of triple-enteric-coated sustained-release peppermint oil microspheres caused less heartburn than was reported in the previous study, but still significantly improved abdominal symptoms and lessened pain on defecation and fecal urgency.31

CASE Suspecting IBS-D, the FP ordered a complete blood count, tissue transglutaminase antibodies, and a stool culture, all of which were unremarkable. Ms. S has been trying to follow a low FODMAP diet and has been taking some over-the-counter probiotics with only minimal relief of abdominal bloating and cramping and no improvement in stool consistency. Her FP started her on eluxadoline 100 mg twice daily with food. After 12 weeks of therapy, she reports significant improvement in global IBS symptoms and nearly complete resolution of her diarrhea.

*Amber S is a real patient in my practice. Her name has been changed to protect her identity.

CORRESPONDENCE
Joel J. Heidelbaugh, MD, FAAFP, FACG, Ypsilanti Health Center, 200 Arnet Suite 200, Ypsilanti, MI 48198; [email protected].

References

1. Lovell RM, Ford AC. Global prevalence of and risk factors for irritable bowel syndrome: a meta-analysis. Clin Gastroenterol Hepatol. 2012;10:712-721.

2. Ballou S, Keefer L. The impact of irritable bowel syndrome on daily functioning: characterizing and understanding daily consequences of IBS. Neurogastroenterol Motil. 2017;29. Epub 2016 Oct 25.

3. Heidelbaugh J, Hungin P, eds. ROME IV: Functional Gastrointestinal Disorders for Primary Care and Non-GI Clinicians. 1st ed. Raleigh, NC: Rome Foundation, Inc.; 2016.

4. Canavan C, West J, Card T. The epidemiology of irritable bowel syndrome. Clin Epidemiol. 2014;6:71-80.

5. Lee V, Guthrie E, Robinson A, et al. Functional bowel disorders in primary care: factors associated with health-related quality of life and doctor consultation. J Psychosom Res. 2008;64:129-138.

6. Lacy BE, Rosemore J, Robertson D, et al. Physicians’ attitudes and practices in the evaluation and treatment of irritable bowel syndrome. Scand J Gastroenterol. 2006;41:892-902.

7. Casiday RE, Hungin AP, Cornford CS, et al. GPs’ explanatory models for irritable bowel syndrome: a mismatch with patient models? J Fam Pract. 2009;26:34-39.

8. Harkness EF, Harrington V, Hinder S, et al. GP perspectives of irritable bowel syndrome—an accepted illness, but management deviates from guidelines: a qualitative study. BMC Fam Pract. 2013;14:92.

9. Hungin AP, Becher A, Cayley B, et al. Irritable bowel syndrome: an integrated explanatory model for clinical practice. Neurogastroenterol Motil. 2015;27:750-753.

10. Lacy BE, Mearin F, Chang L, et al. Bowel Disorders. Gastroenterol. 2016;150:1393-1407.

11. Engsbro AL, Simren M, Bytzer P. Short-term stability of subtypes in the irritable bowel syndrome: prospective evaluation using the Rome III classification. Aliment Pharmacol Ther. 2012;35:350-359.

12. Pimentel M, Morales W, Rezaie A, et al. Development and validation of a biomarker for diarrhea-predominant irritable bowel syndrome in human subjects. PLoS One. 2015;10:e0126438.

13. Lembo AJ, Lacy BE, Zuckerman MJ, et al. Eluxadoline for irritable bowel syndrome with diarrhea. N Engl J Med. 2016;374:242-253.

14. Ford AC, Quigley EM, Lacy BE, et al. Efficacy of prebiotics, probiotics, and synbiotics in irritable bowel syndrome and chronic idiopathic constipation: systematic review and meta-analysis. Am J Gastroenterol. 2014;109:1547-1561.

15. Fujita W, Gomes I, Dove LS, et al. Molecular characterization of eluxadoline as a potential ligand targeting mu-delta opioid receptor heteromers. Biochem Pharmacol. 2014;92:448-456.

16. Pimentel M, Lembo A, Chey WD, et al, for the TARGET Study Group. Rifaximin therapy for patients with irritable bowel syndrome without constipation. N Engl J Med. 2011;364:22-32.

17. Cremonini F, Nicandro JP, Atkinson V, et al. Randomised clinical trial: alosetron improves quality of life and reduces restriction of daily activities in women with severe diarrhoea-predominant IBS. Aliment Pharmacol Ther. 2012;36:437-448.

18. Lewis JH. Alosetron for severe diarrhea-predominant irritable bowel syndrome: safety and efficacy in perspective. Expert Rev Gastroenterol Hepatol. 2010;4:13-29.

19. Tong K, Nicandro JP, Shringarpure R, et al. A 9-year evaluation of temporal trends in alosetron postmarketing safety under the risk management program. Therap Adv Gastroenterol. 2013;6:344-357.

20. Chey WD, Lembo AJ, Lavins BJ, et al. Linaclotide for irritable bowel syndrome with constipation: a 26-week, randomized, double-blind, placebo-controlled trial to evaluate efficacy and safety. Am J Gastroenterol. 2012;107:1702-1712.

21. Rao SS, Quigley EM, Shiff SJ, et al. Effect of linaclotide on severe abdominal symptoms in patients with irritable bowel syndrome with constipation. Clin Gastroenterol Hepatol. 2014;12:616-623.

22. Drossman DA, Chey WD, Johanson JF, et al. Clinical trial: lubiprostone in patients with constipation-associated irritable bowel syndrome—results of two randomized, placebo-controlled studies. Aliment Pharmacol Ther. 2009;29:329-341.

23. Lacy BE, Chey WD. Lubiprostone: chronic constipation and irritable bowel syndrome with constipation. Expert Opin Pharmacother. 2009;10:143-152.

24. Spencer M, Chey WD, Eswaran S. Dietary Renaissance in IBS: has food replaced medications as a primary treatment strategy? Curr Treat Options Gastroenterol. 2014;12:424-440.

25. Halmos EP, Power VA, Shepherd SJ, et al. A diet low in FODMAPs reduces symptoms of irritable bowel syndrome. Gastroenterology. 2014;146:67-75.

26. Yoon JS, Sohn W, Lee OY, et al. Effect of multispecies probiotics on irritable bowel syndrome: a randomized, double-blind, placebo-controlled trial. J Gastroenterol Hepatol. 2014;29:52-59.

27. Guglielmetti S, Mora D, Gschwender M, et al. Randomised clinical trial: Bifidobacterium bifidum MIMBb75 significantly alleviates irritable bowel syndrome and improves quality of life—a double-blind, placebo-controlled study. Aliment Pharmacol Ther. 2011;33:1123-1132.

28. Choi CH, Jo SY, Park HJ, et al. A randomized, double-blind, placebo-controlled multicenter trial of saccharomyces boulardii in irritable bowel syndrome: effect on quality of life. J Clin Gastroenterol. 2011;45:679-683.

29. Attaluri A, Donahoe R, Valestin J, et al. Randomised clinical trial: dried plums (prunes) vs. psyllium for constipation. Aliment Pharmacol Ther. 2011;33:822-828.

30. Khanna R, MacDonald JK, Levesque BG. Peppermint oil for the treatment of irritable bowel syndrome: a systematic review and meta-analysis. J Clin Gastroenterol. 2014;48:505-512.

31. Cash BD, Epstein MS, Shah SM. A novel delivery system of peppermint oil is an effective therapy for irritable bowel syndrome symptoms. Dig Dis Sci. 2016;61:560-571.

References

1. Lovell RM, Ford AC. Global prevalence of and risk factors for irritable bowel syndrome: a meta-analysis. Clin Gastroenterol Hepatol. 2012;10:712-721.

2. Ballou S, Keefer L. The impact of irritable bowel syndrome on daily functioning: characterizing and understanding daily consequences of IBS. Neurogastroenterol Motil. 2017;29. Epub 2016 Oct 25.

3. Heidelbaugh J, Hungin P, eds. ROME IV: Functional Gastrointestinal Disorders for Primary Care and Non-GI Clinicians. 1st ed. Raleigh, NC: Rome Foundation, Inc.; 2016.

4. Canavan C, West J, Card T. The epidemiology of irritable bowel syndrome. Clin Epidemiol. 2014;6:71-80.

5. Lee V, Guthrie E, Robinson A, et al. Functional bowel disorders in primary care: factors associated with health-related quality of life and doctor consultation. J Psychosom Res. 2008;64:129-138.

6. Lacy BE, Rosemore J, Robertson D, et al. Physicians’ attitudes and practices in the evaluation and treatment of irritable bowel syndrome. Scand J Gastroenterol. 2006;41:892-902.

7. Casiday RE, Hungin AP, Cornford CS, et al. GPs’ explanatory models for irritable bowel syndrome: a mismatch with patient models? J Fam Pract. 2009;26:34-39.

8. Harkness EF, Harrington V, Hinder S, et al. GP perspectives of irritable bowel syndrome—an accepted illness, but management deviates from guidelines: a qualitative study. BMC Fam Pract. 2013;14:92.

9. Hungin AP, Becher A, Cayley B, et al. Irritable bowel syndrome: an integrated explanatory model for clinical practice. Neurogastroenterol Motil. 2015;27:750-753.

10. Lacy BE, Mearin F, Chang L, et al. Bowel Disorders. Gastroenterol. 2016;150:1393-1407.

11. Engsbro AL, Simren M, Bytzer P. Short-term stability of subtypes in the irritable bowel syndrome: prospective evaluation using the Rome III classification. Aliment Pharmacol Ther. 2012;35:350-359.

12. Pimentel M, Morales W, Rezaie A, et al. Development and validation of a biomarker for diarrhea-predominant irritable bowel syndrome in human subjects. PLoS One. 2015;10:e0126438.

13. Lembo AJ, Lacy BE, Zuckerman MJ, et al. Eluxadoline for irritable bowel syndrome with diarrhea. N Engl J Med. 2016;374:242-253.

14. Ford AC, Quigley EM, Lacy BE, et al. Efficacy of prebiotics, probiotics, and synbiotics in irritable bowel syndrome and chronic idiopathic constipation: systematic review and meta-analysis. Am J Gastroenterol. 2014;109:1547-1561.

15. Fujita W, Gomes I, Dove LS, et al. Molecular characterization of eluxadoline as a potential ligand targeting mu-delta opioid receptor heteromers. Biochem Pharmacol. 2014;92:448-456.

16. Pimentel M, Lembo A, Chey WD, et al, for the TARGET Study Group. Rifaximin therapy for patients with irritable bowel syndrome without constipation. N Engl J Med. 2011;364:22-32.

17. Cremonini F, Nicandro JP, Atkinson V, et al. Randomised clinical trial: alosetron improves quality of life and reduces restriction of daily activities in women with severe diarrhoea-predominant IBS. Aliment Pharmacol Ther. 2012;36:437-448.

18. Lewis JH. Alosetron for severe diarrhea-predominant irritable bowel syndrome: safety and efficacy in perspective. Expert Rev Gastroenterol Hepatol. 2010;4:13-29.

19. Tong K, Nicandro JP, Shringarpure R, et al. A 9-year evaluation of temporal trends in alosetron postmarketing safety under the risk management program. Therap Adv Gastroenterol. 2013;6:344-357.

20. Chey WD, Lembo AJ, Lavins BJ, et al. Linaclotide for irritable bowel syndrome with constipation: a 26-week, randomized, double-blind, placebo-controlled trial to evaluate efficacy and safety. Am J Gastroenterol. 2012;107:1702-1712.

21. Rao SS, Quigley EM, Shiff SJ, et al. Effect of linaclotide on severe abdominal symptoms in patients with irritable bowel syndrome with constipation. Clin Gastroenterol Hepatol. 2014;12:616-623.

22. Drossman DA, Chey WD, Johanson JF, et al. Clinical trial: lubiprostone in patients with constipation-associated irritable bowel syndrome—results of two randomized, placebo-controlled studies. Aliment Pharmacol Ther. 2009;29:329-341.

23. Lacy BE, Chey WD. Lubiprostone: chronic constipation and irritable bowel syndrome with constipation. Expert Opin Pharmacother. 2009;10:143-152.

24. Spencer M, Chey WD, Eswaran S. Dietary Renaissance in IBS: has food replaced medications as a primary treatment strategy? Curr Treat Options Gastroenterol. 2014;12:424-440.

25. Halmos EP, Power VA, Shepherd SJ, et al. A diet low in FODMAPs reduces symptoms of irritable bowel syndrome. Gastroenterology. 2014;146:67-75.

26. Yoon JS, Sohn W, Lee OY, et al. Effect of multispecies probiotics on irritable bowel syndrome: a randomized, double-blind, placebo-controlled trial. J Gastroenterol Hepatol. 2014;29:52-59.

27. Guglielmetti S, Mora D, Gschwender M, et al. Randomised clinical trial: Bifidobacterium bifidum MIMBb75 significantly alleviates irritable bowel syndrome and improves quality of life—a double-blind, placebo-controlled study. Aliment Pharmacol Ther. 2011;33:1123-1132.

28. Choi CH, Jo SY, Park HJ, et al. A randomized, double-blind, placebo-controlled multicenter trial of saccharomyces boulardii in irritable bowel syndrome: effect on quality of life. J Clin Gastroenterol. 2011;45:679-683.

29. Attaluri A, Donahoe R, Valestin J, et al. Randomised clinical trial: dried plums (prunes) vs. psyllium for constipation. Aliment Pharmacol Ther. 2011;33:822-828.

30. Khanna R, MacDonald JK, Levesque BG. Peppermint oil for the treatment of irritable bowel syndrome: a systematic review and meta-analysis. J Clin Gastroenterol. 2014;48:505-512.

31. Cash BD, Epstein MS, Shah SM. A novel delivery system of peppermint oil is an effective therapy for irritable bowel syndrome symptoms. Dig Dis Sci. 2016;61:560-571.

Issue
The Journal of Family Practice - 66(6)
Issue
The Journal of Family Practice - 66(6)
Page Number
346-348,350-353
Page Number
346-348,350-353
Publications
Publications
Topics
Article Type
Display Headline
These 3 tools can help you streamline management of IBS
Display Headline
These 3 tools can help you streamline management of IBS
Sections
Inside the Article

PRACTICE RECOMMENDATIONS

› Prescribe eluxadoline, rifaximin, or alosetron for diarrhea-predominant IBS because all 3 have proven efficacy with this diagnosis. A

› Prescribe linaclotide or lubiprostone for constipation-predominant IBS, as both have proven efficacy with this condition. A

› Suggest that patients with IBS follow a low FODMAP (fermentable oligosaccharides, disaccharides, monosaccharides, and polyols) diet; probiotics, prunes, and peppermint oil may also offer some improvement of IBS symptoms. 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

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
PubMed ID
28574520
Article PDF Media

How much can we really do to slow age-related cognitive decline?

Article Type
Changed
Fri, 01/18/2019 - 08:46
Display Headline
How much can we really do to slow age-related cognitive decline?

Author and Disclosure Information

Dr. Campos-Outcalt is an assistant editor of The Journal of Family Practice and the medical director for Mercy Care Plan in Phoenix, Ariz.

Issue
The Journal of Family Practice - 66(6)
Publications
Topics
Page Number
audio
Author and Disclosure Information

Dr. Campos-Outcalt is an assistant editor of The Journal of Family Practice and the medical director for Mercy Care Plan in Phoenix, Ariz.

Author and Disclosure Information

Dr. Campos-Outcalt is an assistant editor of The Journal of Family Practice and the medical director for Mercy Care Plan in Phoenix, Ariz.

Issue
The Journal of Family Practice - 66(6)
Issue
The Journal of Family Practice - 66(6)
Page Number
audio
Page Number
audio
Publications
Publications
Topics
Article Type
Display Headline
How much can we really do to slow age-related cognitive decline?
Display Headline
How much can we really do to slow age-related cognitive decline?
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Use ProPublica

Flashback to 2012

Article Type
Changed
Thu, 06/01/2017 - 02:51

 

It’s a whole new biosimilar world. In the April 2012 issue of GI & Hepatology News (GIHN) there was a small article on the issued Food and Drug Administration guidance on how to develop biosimilars. A biosimilar molecule must be structurally similar to the reference or originator product with the expectation that the safety and efficacy will be the same. The European Medicines Agency (EMA) established a legal framework for approving biologics in the European Union in 2003 and guidelines for approval in 2005 to 2006 with the first biosimilar approved in 2006 (somatropin [Omnitrope]).

The first monoclonal antibody biosimilar approved by the EMA was CT-P13 (infliximab-dyyb) in June 2013. There are now over 23 biosimilars approved for use in Europe. In 2012 there were no biosimilars on the market in the United States. This past year (2016) has been the year of the biosimilar with two of the four approved compounds used in inflammatory bowel disease – Inflectra (infliximab-dyyb, Hospira) April 2016 and Amjevita (adalimumab-atto, Amgen) September 2016 appearing.

Dr. Kim L. Isaacs


The launch of these biosimilars raises a whole new series of questions. First and foremost for gastroenterologists – are the biosimilars truly similar in patients with inflammatory bowel disease? Adalimumab-atto was approved on the basis of two phase III studies in psoriasis and in rheumatoid arthritis and infliximab-dyyb was approved on the basis of studies in rheumatoid arthritis and ankylosing spondylitis. Other questions arise: 1. Can a patient who is doing well on the originator be safely switched to the biosimilar? 2. Can we use the same assays for drug monitoring? 3. Will use of biosimilars lead to a lower cost structure for patients and hospitals? 4. What are the regulations and guidelines for interchangeability? (GIHN March 2017) In the United States, development of biosimilars was slow to start but we expect to see an explosion in development of these agents in gastroenterology as the patents expire on the biologics currently in use.
 

Kim L. Isaacs, MD, PhD, is professor of medicine in the division of gastroenterology and hepatology at the University of North Carolina at Chapel Hill. She is codirector of the UNC Center for Inflammatory Bowel Disease. She is an Associate Editor for GI and Hepatology News.

Publications
Topics
Sections

 

It’s a whole new biosimilar world. In the April 2012 issue of GI & Hepatology News (GIHN) there was a small article on the issued Food and Drug Administration guidance on how to develop biosimilars. A biosimilar molecule must be structurally similar to the reference or originator product with the expectation that the safety and efficacy will be the same. The European Medicines Agency (EMA) established a legal framework for approving biologics in the European Union in 2003 and guidelines for approval in 2005 to 2006 with the first biosimilar approved in 2006 (somatropin [Omnitrope]).

The first monoclonal antibody biosimilar approved by the EMA was CT-P13 (infliximab-dyyb) in June 2013. There are now over 23 biosimilars approved for use in Europe. In 2012 there were no biosimilars on the market in the United States. This past year (2016) has been the year of the biosimilar with two of the four approved compounds used in inflammatory bowel disease – Inflectra (infliximab-dyyb, Hospira) April 2016 and Amjevita (adalimumab-atto, Amgen) September 2016 appearing.

Dr. Kim L. Isaacs


The launch of these biosimilars raises a whole new series of questions. First and foremost for gastroenterologists – are the biosimilars truly similar in patients with inflammatory bowel disease? Adalimumab-atto was approved on the basis of two phase III studies in psoriasis and in rheumatoid arthritis and infliximab-dyyb was approved on the basis of studies in rheumatoid arthritis and ankylosing spondylitis. Other questions arise: 1. Can a patient who is doing well on the originator be safely switched to the biosimilar? 2. Can we use the same assays for drug monitoring? 3. Will use of biosimilars lead to a lower cost structure for patients and hospitals? 4. What are the regulations and guidelines for interchangeability? (GIHN March 2017) In the United States, development of biosimilars was slow to start but we expect to see an explosion in development of these agents in gastroenterology as the patents expire on the biologics currently in use.
 

Kim L. Isaacs, MD, PhD, is professor of medicine in the division of gastroenterology and hepatology at the University of North Carolina at Chapel Hill. She is codirector of the UNC Center for Inflammatory Bowel Disease. She is an Associate Editor for GI and Hepatology News.

 

It’s a whole new biosimilar world. In the April 2012 issue of GI & Hepatology News (GIHN) there was a small article on the issued Food and Drug Administration guidance on how to develop biosimilars. A biosimilar molecule must be structurally similar to the reference or originator product with the expectation that the safety and efficacy will be the same. The European Medicines Agency (EMA) established a legal framework for approving biologics in the European Union in 2003 and guidelines for approval in 2005 to 2006 with the first biosimilar approved in 2006 (somatropin [Omnitrope]).

The first monoclonal antibody biosimilar approved by the EMA was CT-P13 (infliximab-dyyb) in June 2013. There are now over 23 biosimilars approved for use in Europe. In 2012 there were no biosimilars on the market in the United States. This past year (2016) has been the year of the biosimilar with two of the four approved compounds used in inflammatory bowel disease – Inflectra (infliximab-dyyb, Hospira) April 2016 and Amjevita (adalimumab-atto, Amgen) September 2016 appearing.

Dr. Kim L. Isaacs


The launch of these biosimilars raises a whole new series of questions. First and foremost for gastroenterologists – are the biosimilars truly similar in patients with inflammatory bowel disease? Adalimumab-atto was approved on the basis of two phase III studies in psoriasis and in rheumatoid arthritis and infliximab-dyyb was approved on the basis of studies in rheumatoid arthritis and ankylosing spondylitis. Other questions arise: 1. Can a patient who is doing well on the originator be safely switched to the biosimilar? 2. Can we use the same assays for drug monitoring? 3. Will use of biosimilars lead to a lower cost structure for patients and hospitals? 4. What are the regulations and guidelines for interchangeability? (GIHN March 2017) In the United States, development of biosimilars was slow to start but we expect to see an explosion in development of these agents in gastroenterology as the patents expire on the biologics currently in use.
 

Kim L. Isaacs, MD, PhD, is professor of medicine in the division of gastroenterology and hepatology at the University of North Carolina at Chapel Hill. She is codirector of the UNC Center for Inflammatory Bowel Disease. She is an Associate Editor for GI and Hepatology News.

Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME

Blazing A Trail in Medical Education

Article Type
Changed
Fri, 01/12/2018 - 13:51

 

What led you to pursue a career in medical education?

Believe it or not, I pursued my path in medical education even prior to attending medical school. I was a high school teacher with a master’s in education, working during the summer of 1979 under the auspices of the Student Conservation Association at Grand Canyon National Park. Sitting on the edge of the canyon at sunset, I made the momentous decision to attend medical school, requiring attendance at a postbaccalaureate program at Columbia University. While considering medical schools, I knew that I wanted to combine my interest in education with medicine and I therefore chose to attend Case Western University School of Medicine. Since the mid-1950s, Case had been committed to innovative educational programs with a systems-based approach to the curriculum.

Dr. Suzanne Rose
Throughout my career I focused on medical education, preparing my senior resident talk on “the resident as teacher” – not yet a hot topic. My path as a GI fellow, including a chief fellow year at the Cleveland Clinic, reconfirmed my interest in education leadership. During my first postfellowship position at the University of Pittsburgh, I was able to lead the GI second-year course, oversee GI electives for students and residents, and work on the GI fellowship curriculum. It was at that time that I began my involvement in AGA with committee work related to education and women’s issues in GI. I also refocused my scholarly work in education, eventually editing a textbook in GI and hepatobiliary pathophysiology, and working on other projects.
 

What do you enjoy most about working in medical education?

There are so many aspects of medical education that make work fun and rewarding. Perhaps the most rewarding is the ability to make a difference that affects the learner as well as the patients and communities that they will serve. I also enjoy the diverse experiences and opportunities in education and the ability to work with others in creative endeavors.

What are your responsibilities in a typical week?

One of the great things about a focus in education is that there never is a typical week. In the 32 years since my graduation from medical school, I have had the great fortune to fill many different roles: course director, electives director, fellowship program director, associate dean for student affairs, associate dean for undergraduate medical education, and associate dean for continuing medical education. For the past 6 years, I have been the senior associate dean for education at the University of Connecticut School of Medicine, overseeing undergraduate medical education, graduate medical education, continuing medical education, and the graduate school.

Over time I have had less interaction with students and residents as my administrative responsibilities have grown, but I know it is critical to maintain a presence with learners and I endeavor to do so in limited ways. Since our current priorities are in implementing a new curriculum and in planning for an accreditation visit, there are many days that are filled with meetings, planning, organizing, and writing. To me, the most precious responsibility is shaping a vision and bringing together a team to operationalize that vision in a collaborative and creative way, with learners, teachers, and administrators working together.
 

What are the different career options available for early-career GIs who are interested in medical education?

There are so many options in medical education for early-career gastroenterologists. For those working in private, group, or community practices, there are opportunities to precept students, residents, and fellows. For those working in an academic setting, opportunities abound. It is often a good idea to start within the division: get involved in teaching fellows in a clinical setting, or creating a new simulation experience or case workshop for fellows. There are opportunities to teach and supervise students. One of my first opportunities was in teaching in the physical diagnosis course. There are options to be involved in curriculum committees, admissions, CME, and to engage in educational initiatives at your institution.

The Association of American Medical Colleges has defined five areas of scholarship in education, and it is possible to get promoted to full professor – and even to attain academic tenure, as I have – if you fulfill the requirements for promotion at your institution. These areas include teaching, curriculum development, assessment, mentorship/advising, and leadership. There are also many ways to get involved in the AGA (http://www.gastro.org/trainees) and other organizations.1,2

 

 

Are there advanced training options available for those interested in medical education?

The AGA Academy of Educators (http://www.gastro.org/about/initiatives/aga-academy-of-educators)3 is a wonderful resource for networking. It has a competitive process for educational project grants as well as faculty development sessions and networking events at DDW®. There are also national leadership academies in medicine that have a focus in medical education. The Harvard Macy Institute is one such opportunity. Many medical schools have their own academies to support educators and teachers. I have been privileged to be one of the co-leaders of the AGA Future Leaders Program (http://www.gastro.org/about/initiatives/aga-future-leaders-program) and those with a niche interest in education can benefit and pursue related projects.4 One group was successful in publishing an educational article after completing the Future Leaders program.5 There are also several master’s programs for further education and training in educational theory. Some of these programs are available online or largely online, with limited requirements for onsite classes.

How do you go about finding a job in medical education?

First of all, you have to do your “day job.” In order to be a credible medical clinician-educator you must have clinical experience in patient care. It is important for the first years of your career to make sure that you have at least 70% clinical roles that can be reduced over time to accommodate advancing educational responsibilities. Get involved in teaching fellows. If you are in a practice, reach out to your local medical school or hospital to see how you might participate in educational programs. If you are in an academic setting, meet with the deans in education to express your interest and look for opportunities to get involved in an area of interest. If you are in academia, you have to make your work “count twice:” being productive in a scholarly way is not only important as a role model for learners, but it is important for you as a faculty member to grow and advance in your professional career.

It is always wise to think about when to say “yes” and when to say “no.” An important point is not to overextend yourself. Your reputation of completing tasks not only well, but on time, and thoroughly, is critical to your success. This includes making sure your learner evaluations are submitted on time, that you complete the administrative work in order to participate in CME programs, and that you honor your commitments by attending committee meetings.
 

What are the resources available to early-career GIs interested in medical education?

It is easy to find resources within your practice, your institution, or externally. The AGA has many resources available with a good start being the AGA Academy of Educators. Opportunities for creativity are numerous and with new advances in team-based learning, simulation, and interprofessional learning, there are new areas for involvement evolving all the time.6,7

Finally, pursuing a career in education is exciting, fun, and fulfilling. Having the opportunity to influence learners, which in turn will impact patient care, is an awesome privilege.
 

Dr. Rose is a professor of medicine and senior associate dean for education at the University of Connecticut School of Medicine.

References

1. Gusic M, et al. MedEdPORTAL; 2013. Available from: http://www.mededportal.org/publication/9313.

2. Gusic ME, et al. Acad Med. 2014;89(7):1006-11.

3. Pfeil SA, et al. Gastroenterology 2015;149(6):1309-14.

4. Cryer B, Rose S. Gastroenterology 2015;149:246-8.

5. Shah BJ, et al. Gastroenterology 2016;151(2):218-21.

6. Shah BJ, Rose S. Gastroenterology 2012;142:684-9.

7. Shah BJ, Rose S. AGA Perspectives 2012;April-May:20-21.
 

Publications
Sections

 

What led you to pursue a career in medical education?

Believe it or not, I pursued my path in medical education even prior to attending medical school. I was a high school teacher with a master’s in education, working during the summer of 1979 under the auspices of the Student Conservation Association at Grand Canyon National Park. Sitting on the edge of the canyon at sunset, I made the momentous decision to attend medical school, requiring attendance at a postbaccalaureate program at Columbia University. While considering medical schools, I knew that I wanted to combine my interest in education with medicine and I therefore chose to attend Case Western University School of Medicine. Since the mid-1950s, Case had been committed to innovative educational programs with a systems-based approach to the curriculum.

Dr. Suzanne Rose
Throughout my career I focused on medical education, preparing my senior resident talk on “the resident as teacher” – not yet a hot topic. My path as a GI fellow, including a chief fellow year at the Cleveland Clinic, reconfirmed my interest in education leadership. During my first postfellowship position at the University of Pittsburgh, I was able to lead the GI second-year course, oversee GI electives for students and residents, and work on the GI fellowship curriculum. It was at that time that I began my involvement in AGA with committee work related to education and women’s issues in GI. I also refocused my scholarly work in education, eventually editing a textbook in GI and hepatobiliary pathophysiology, and working on other projects.
 

What do you enjoy most about working in medical education?

There are so many aspects of medical education that make work fun and rewarding. Perhaps the most rewarding is the ability to make a difference that affects the learner as well as the patients and communities that they will serve. I also enjoy the diverse experiences and opportunities in education and the ability to work with others in creative endeavors.

What are your responsibilities in a typical week?

One of the great things about a focus in education is that there never is a typical week. In the 32 years since my graduation from medical school, I have had the great fortune to fill many different roles: course director, electives director, fellowship program director, associate dean for student affairs, associate dean for undergraduate medical education, and associate dean for continuing medical education. For the past 6 years, I have been the senior associate dean for education at the University of Connecticut School of Medicine, overseeing undergraduate medical education, graduate medical education, continuing medical education, and the graduate school.

Over time I have had less interaction with students and residents as my administrative responsibilities have grown, but I know it is critical to maintain a presence with learners and I endeavor to do so in limited ways. Since our current priorities are in implementing a new curriculum and in planning for an accreditation visit, there are many days that are filled with meetings, planning, organizing, and writing. To me, the most precious responsibility is shaping a vision and bringing together a team to operationalize that vision in a collaborative and creative way, with learners, teachers, and administrators working together.
 

What are the different career options available for early-career GIs who are interested in medical education?

There are so many options in medical education for early-career gastroenterologists. For those working in private, group, or community practices, there are opportunities to precept students, residents, and fellows. For those working in an academic setting, opportunities abound. It is often a good idea to start within the division: get involved in teaching fellows in a clinical setting, or creating a new simulation experience or case workshop for fellows. There are opportunities to teach and supervise students. One of my first opportunities was in teaching in the physical diagnosis course. There are options to be involved in curriculum committees, admissions, CME, and to engage in educational initiatives at your institution.

The Association of American Medical Colleges has defined five areas of scholarship in education, and it is possible to get promoted to full professor – and even to attain academic tenure, as I have – if you fulfill the requirements for promotion at your institution. These areas include teaching, curriculum development, assessment, mentorship/advising, and leadership. There are also many ways to get involved in the AGA (http://www.gastro.org/trainees) and other organizations.1,2

 

 

Are there advanced training options available for those interested in medical education?

The AGA Academy of Educators (http://www.gastro.org/about/initiatives/aga-academy-of-educators)3 is a wonderful resource for networking. It has a competitive process for educational project grants as well as faculty development sessions and networking events at DDW®. There are also national leadership academies in medicine that have a focus in medical education. The Harvard Macy Institute is one such opportunity. Many medical schools have their own academies to support educators and teachers. I have been privileged to be one of the co-leaders of the AGA Future Leaders Program (http://www.gastro.org/about/initiatives/aga-future-leaders-program) and those with a niche interest in education can benefit and pursue related projects.4 One group was successful in publishing an educational article after completing the Future Leaders program.5 There are also several master’s programs for further education and training in educational theory. Some of these programs are available online or largely online, with limited requirements for onsite classes.

How do you go about finding a job in medical education?

First of all, you have to do your “day job.” In order to be a credible medical clinician-educator you must have clinical experience in patient care. It is important for the first years of your career to make sure that you have at least 70% clinical roles that can be reduced over time to accommodate advancing educational responsibilities. Get involved in teaching fellows. If you are in a practice, reach out to your local medical school or hospital to see how you might participate in educational programs. If you are in an academic setting, meet with the deans in education to express your interest and look for opportunities to get involved in an area of interest. If you are in academia, you have to make your work “count twice:” being productive in a scholarly way is not only important as a role model for learners, but it is important for you as a faculty member to grow and advance in your professional career.

It is always wise to think about when to say “yes” and when to say “no.” An important point is not to overextend yourself. Your reputation of completing tasks not only well, but on time, and thoroughly, is critical to your success. This includes making sure your learner evaluations are submitted on time, that you complete the administrative work in order to participate in CME programs, and that you honor your commitments by attending committee meetings.
 

What are the resources available to early-career GIs interested in medical education?

It is easy to find resources within your practice, your institution, or externally. The AGA has many resources available with a good start being the AGA Academy of Educators. Opportunities for creativity are numerous and with new advances in team-based learning, simulation, and interprofessional learning, there are new areas for involvement evolving all the time.6,7

Finally, pursuing a career in education is exciting, fun, and fulfilling. Having the opportunity to influence learners, which in turn will impact patient care, is an awesome privilege.
 

Dr. Rose is a professor of medicine and senior associate dean for education at the University of Connecticut School of Medicine.

References

1. Gusic M, et al. MedEdPORTAL; 2013. Available from: http://www.mededportal.org/publication/9313.

2. Gusic ME, et al. Acad Med. 2014;89(7):1006-11.

3. Pfeil SA, et al. Gastroenterology 2015;149(6):1309-14.

4. Cryer B, Rose S. Gastroenterology 2015;149:246-8.

5. Shah BJ, et al. Gastroenterology 2016;151(2):218-21.

6. Shah BJ, Rose S. Gastroenterology 2012;142:684-9.

7. Shah BJ, Rose S. AGA Perspectives 2012;April-May:20-21.
 

 

What led you to pursue a career in medical education?

Believe it or not, I pursued my path in medical education even prior to attending medical school. I was a high school teacher with a master’s in education, working during the summer of 1979 under the auspices of the Student Conservation Association at Grand Canyon National Park. Sitting on the edge of the canyon at sunset, I made the momentous decision to attend medical school, requiring attendance at a postbaccalaureate program at Columbia University. While considering medical schools, I knew that I wanted to combine my interest in education with medicine and I therefore chose to attend Case Western University School of Medicine. Since the mid-1950s, Case had been committed to innovative educational programs with a systems-based approach to the curriculum.

Dr. Suzanne Rose
Throughout my career I focused on medical education, preparing my senior resident talk on “the resident as teacher” – not yet a hot topic. My path as a GI fellow, including a chief fellow year at the Cleveland Clinic, reconfirmed my interest in education leadership. During my first postfellowship position at the University of Pittsburgh, I was able to lead the GI second-year course, oversee GI electives for students and residents, and work on the GI fellowship curriculum. It was at that time that I began my involvement in AGA with committee work related to education and women’s issues in GI. I also refocused my scholarly work in education, eventually editing a textbook in GI and hepatobiliary pathophysiology, and working on other projects.
 

What do you enjoy most about working in medical education?

There are so many aspects of medical education that make work fun and rewarding. Perhaps the most rewarding is the ability to make a difference that affects the learner as well as the patients and communities that they will serve. I also enjoy the diverse experiences and opportunities in education and the ability to work with others in creative endeavors.

What are your responsibilities in a typical week?

One of the great things about a focus in education is that there never is a typical week. In the 32 years since my graduation from medical school, I have had the great fortune to fill many different roles: course director, electives director, fellowship program director, associate dean for student affairs, associate dean for undergraduate medical education, and associate dean for continuing medical education. For the past 6 years, I have been the senior associate dean for education at the University of Connecticut School of Medicine, overseeing undergraduate medical education, graduate medical education, continuing medical education, and the graduate school.

Over time I have had less interaction with students and residents as my administrative responsibilities have grown, but I know it is critical to maintain a presence with learners and I endeavor to do so in limited ways. Since our current priorities are in implementing a new curriculum and in planning for an accreditation visit, there are many days that are filled with meetings, planning, organizing, and writing. To me, the most precious responsibility is shaping a vision and bringing together a team to operationalize that vision in a collaborative and creative way, with learners, teachers, and administrators working together.
 

What are the different career options available for early-career GIs who are interested in medical education?

There are so many options in medical education for early-career gastroenterologists. For those working in private, group, or community practices, there are opportunities to precept students, residents, and fellows. For those working in an academic setting, opportunities abound. It is often a good idea to start within the division: get involved in teaching fellows in a clinical setting, or creating a new simulation experience or case workshop for fellows. There are opportunities to teach and supervise students. One of my first opportunities was in teaching in the physical diagnosis course. There are options to be involved in curriculum committees, admissions, CME, and to engage in educational initiatives at your institution.

The Association of American Medical Colleges has defined five areas of scholarship in education, and it is possible to get promoted to full professor – and even to attain academic tenure, as I have – if you fulfill the requirements for promotion at your institution. These areas include teaching, curriculum development, assessment, mentorship/advising, and leadership. There are also many ways to get involved in the AGA (http://www.gastro.org/trainees) and other organizations.1,2

 

 

Are there advanced training options available for those interested in medical education?

The AGA Academy of Educators (http://www.gastro.org/about/initiatives/aga-academy-of-educators)3 is a wonderful resource for networking. It has a competitive process for educational project grants as well as faculty development sessions and networking events at DDW®. There are also national leadership academies in medicine that have a focus in medical education. The Harvard Macy Institute is one such opportunity. Many medical schools have their own academies to support educators and teachers. I have been privileged to be one of the co-leaders of the AGA Future Leaders Program (http://www.gastro.org/about/initiatives/aga-future-leaders-program) and those with a niche interest in education can benefit and pursue related projects.4 One group was successful in publishing an educational article after completing the Future Leaders program.5 There are also several master’s programs for further education and training in educational theory. Some of these programs are available online or largely online, with limited requirements for onsite classes.

How do you go about finding a job in medical education?

First of all, you have to do your “day job.” In order to be a credible medical clinician-educator you must have clinical experience in patient care. It is important for the first years of your career to make sure that you have at least 70% clinical roles that can be reduced over time to accommodate advancing educational responsibilities. Get involved in teaching fellows. If you are in a practice, reach out to your local medical school or hospital to see how you might participate in educational programs. If you are in an academic setting, meet with the deans in education to express your interest and look for opportunities to get involved in an area of interest. If you are in academia, you have to make your work “count twice:” being productive in a scholarly way is not only important as a role model for learners, but it is important for you as a faculty member to grow and advance in your professional career.

It is always wise to think about when to say “yes” and when to say “no.” An important point is not to overextend yourself. Your reputation of completing tasks not only well, but on time, and thoroughly, is critical to your success. This includes making sure your learner evaluations are submitted on time, that you complete the administrative work in order to participate in CME programs, and that you honor your commitments by attending committee meetings.
 

What are the resources available to early-career GIs interested in medical education?

It is easy to find resources within your practice, your institution, or externally. The AGA has many resources available with a good start being the AGA Academy of Educators. Opportunities for creativity are numerous and with new advances in team-based learning, simulation, and interprofessional learning, there are new areas for involvement evolving all the time.6,7

Finally, pursuing a career in education is exciting, fun, and fulfilling. Having the opportunity to influence learners, which in turn will impact patient care, is an awesome privilege.
 

Dr. Rose is a professor of medicine and senior associate dean for education at the University of Connecticut School of Medicine.

References

1. Gusic M, et al. MedEdPORTAL; 2013. Available from: http://www.mededportal.org/publication/9313.

2. Gusic ME, et al. Acad Med. 2014;89(7):1006-11.

3. Pfeil SA, et al. Gastroenterology 2015;149(6):1309-14.

4. Cryer B, Rose S. Gastroenterology 2015;149:246-8.

5. Shah BJ, et al. Gastroenterology 2016;151(2):218-21.

6. Shah BJ, Rose S. Gastroenterology 2012;142:684-9.

7. Shah BJ, Rose S. AGA Perspectives 2012;April-May:20-21.
 

Publications
Publications
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default