AGING: Is your patient taking too many pills?

Article Type
Changed
Mon, 01/14/2019 - 11:35
Display Headline
AGING: Is your patient taking too many pills?

PRACTICE RECOMMENDATIONS

Consider the possibility that an adverse drug effect—rather than a new condition—is at play when a patient taking multiple medications develops a new symptom. C

Use an online interaction checker, which can be accessed via a smart phone or tablet, to check for potential drug-drug interactions in patients on multiple medications. C

Cross-check patients’ medications with a list of their medical problems, with the goal of discontinuing any drug that duplicates the action of another or is age-inappropriate, ineffective, or not indicated for the condition for which it was prescribed. 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

Older adults are taking more medications than ever before. Nearly 9 out of 10 US residents who are 60 years of age or older take at least one prescription drug, more than a third take 5 to 9 medications, and 12% take 10 or more.1

The increase is largely driven by newer medications to effectively treat a variety of medical conditions, and by practice guidelines that often recommend multidrug regimens.2

As a result, the term “polypharmacy,” which once referred to a specific number of medications, is now used more broadly to mean “a large number” of drugs.

From a safety standpoint, the number of medications a patient takes matters. The risk of adverse drug effects and dangerous drug-drug interactions increases significantly when an individual takes ≥5 medications.3

More than 4.5 million adverse drug effects occur each year in the United States, and nearly three quarters of them are initially evaluated in outpatient settings.4 Research suggests that about 80% of the time, these adverse effects are not recognized as such by the patient’s physician. So instead of discontinuing the offending medication, physicians treat the drug-related symptoms by adding yet another medication—a phenomenon known as “the prescribing cascade.”5

This review can help you safeguard older patients taking multiple medications by recognizing and responding to drug-related problems, identifying drugs that can be safely eliminated (or, in some cases, drugs that should be added), and checking regularly to ensure that the medication regimen is appropriate and up to date.

CASE Mrs. R, a 79-year-old woman who recently moved to town, is brought to your office by her daughter and son-in-law. The patient has a hard time reporting her medical history, but her daughter tells you her mother has chronic obstructive pulmonary disease (COPD), heart failure, type 2 diabetes, and mild urinary incontinence, and was recently diagnosed with early dementia.

Mrs. R’s daughter has brought in a bagful of medications, but she’s not sure which ones her mother takes regularly. The medications are an albuterol inhaler, alprazolam, digoxin, diphenhydramine, donepezil, furosemide, glargine insulin, guaifenesin, levothyroxine, metformin, extended-release metoprolol, naproxen, omeprazole, simvastatin, tolterodine, and zolpidem—a total of 16 different drugs.

If Mrs. R were your patient, how would you manage her multidrug regimen?

Start with a medication review

The first step in evaluating a patient’s medication regimen is to find out whether the drugs in the patient’s possession and/or in the medical record are the ones he or she is actually taking. Ask older patients who haven’t brought in their medications, or the caregiver of a confused patient, to bring them to the next visit.

The next step: Determine whether the medication regimen is right for the patient.

Polypharmacy may be indicated
Despite the risks associated with polypharmacy, do not assume that it is inappropriate. For some conditions, multiple medications are routinely recommended. Patients with heart failure, for example, have been shown to have better outcomes when they take 3 to 5 medications, including beta-blockers, angiotensin-converting enzyme (ACE) inhibitors, and diuretics.2

Some treatment guidelines also call for multiple medications. Achieving the more stringent blood pressure goals recommended in the Seventh Report of the Joint National Committee on Prevention, for instance, often requires 2 or more antihypertensive agents.6 In many cases, however, patients end up taking more drugs than necessary.

Is the patient taking the right drugs?
Medication reconciliation (determining whether the treatment regimen is appropriate for the patient’s diagnoses) is the way to find out.

The most widely recommended approach to medication reconciliation is to create a table and do a systematic review.7 List all the patient’s medical conditions in the first column and all current medications in the second column. Use the third column to note whether each medication is one the patient should be on, based not only on his or her medical conditions and other drugs being taken but also on current renal and hepatic function and body size, and contraindications.

 

 

A medication may be inappropriate if it duplicates, cancels out the action of, or otherwise interacts with another drug the patient is taking; is contraindicated in older patients; or is ineffective for the condition for which it was prescribed. In one key study of nearly 200 patients 65 years and older who took 5 or more medications, more than half had been prescribed at least one drug that was ineffective for the patient’s condition or that duplicated the action of another medication.8

In addition to finding drugs that the patient should not be taking, medication reconciliation may also reveal that the patient is not receiving optimal therapy and that one or more drugs should be added to his or her treatment regimen.

Check meds after transitions. A move from home to hospital, from emergency department to home, or any other transition relating to patient care should prompt a medication reconciliation. Medications are often added or inadvertently discontinued at such times,9,10 and instructions relating to medication are often misunderstood.11 In one study of 384 frail elderly patients being discharged from a hospital, for example, 44% were found to have been given at least one unnecessary prescription—most commonly for a medication that was neither indicated nor effective for any of the patient’s medical problems.12 It was also common for patients to be given drugs that duplicated the action of others they were already taking.

Even in the absence of such transitions, medication reconciliation should occur at regular intervals. Many physicians do a medication reconciliation at every visit to ensure that the medical record is accurate and the patient’s medication regimen is optimal.

Managing polypharmacy: These resources can help

Numerous tools are available to help you evaluate and monitor patients’ medication regimens, including some that were developed specifically for older patients.

START (Screening Tool to Alert doctors to Right Treatment) identifies drugs and drug classes that are underused with older patients.13 START criteria (TABLE 1)13-17 focus on medications that should be used yet are often omitted in older patients who have the appropriate indications.

TABLE 1
START criteria: Drug therapy that should be given to older patients
13-17

Cardiovascular
  • Anticoagulation or antiplatelet therapy for atrial fibrillation
  • Antiplatelet therapy for patients with known coronary, cerebral, or peripheral vascular disease
  • Antihypertensive therapy for systolic BP >160 mm hg
  • Statins for secondary prevention in patients with coronary, cerebral, or peripheral vascular disease (with life expectancy >5 years)
  • ACE inhibitor for heart failure or after MI
  • Beta-blocker for chronic stable angina
Endocrine
  • Metformin for type 2 diabetes
  • ACE inhibitor for patients with diabetes and nephropathy
  • Antiplatelet and statin therapy for patients with diabetes and CVD risk factors
Gastrointestinal
  • PPI for severe gi reflux or esophageal stricture
  • Fiber supplement for chronic symptomatic diverticular disease
Musculoskeletal
  • Antirheumatic drugs for moderate-to-severe chronic rheumatoid disease
  • Bisphosphonates for patients taking chronic oral steroids
  • Calcium and vitamin D for osteoporosis
Nervous system
  • Levodopa for Parkinson’s disease with functional impairment
  • Antidepressant for moderate-to-severe depression lasting >3 months
Respiratory
  • Daily inhaled beta-agonist or anticholinergic agent for asthma or COPD
  • Daily inhaled steroid for asthma or COPD with FEV1 <50% of predicted value
  • Continuous home oxygen for chronic hypoxemic respiratory failure
ACE, angiotensin-converting enzyme; BP, blood pressure; COPD, chronic obstructive pulmonary disease; CVD, cardiovascular disease; FEV1, forced expiratory volume in 1 second; GI, gastrointestinal; MI, myocardial infarction; PPI, proton-pump inhibitor; START, Screening Tool to alert doctors to right Treatment.

In using START or any other drug-related tool, it is important to keep in mind that therapy should be individualized. Not all the medications in the START criteria are appropriate for every patient, and a medication that is indicated for a given medical condition may or may not provide real benefit for a particular patient. That would depend on the individual’s overall health and life expectancy, the goals of treatment, and how long it would take for the patient to realize any benefit from the drug in question.18 A vigorous 79-year-old might benefit from statin therapy for prevention of cardiovascular events, for instance, while a patient like Mrs. R, who is also 79 but has dementia and multiple other medical problems, would be unlikely to live long enough to realize such a benefit.

”Age” assessment tool. One criterion in deciding whether medication(s) are appropriate for an older patient is his or her “physiologic age”—calculated on the basis of the individual’s chronological age and self-reported health status (TABLE 2).19

TABLE 2
Calculating your patient’s “real” age
19

Actual age (y)Physiologic age (y)
Self-reported health
ExcellentGoodFairPoor
MaleFemaleMaleFemaleMaleFemaleMaleFemale
655860646468667372
706265696973717877
756770747478768382
8072757979838185+85+

Flagging drugs that may be inappropriate
Several tools have been developed to aid clinicians in identifying medications that are potentially inappropriate for older adults, although here, too, decisions about their use must be individualized. Two of the most widely used tools are the Beers criteria and STOPP (Screening Tool of Older Persons’ potentially inappropriate Prescriptions).

 

 

Beers criteria were developed by Mark Beers et al in 199120 and have been updated at regular intervals, most recently by the American Geriatrics Society in 2012.21 The drugs and drug classes included in the Beers criteria should not be prescribed for older patients in most cases, either because the risk of using them outweighs the benefit or because safer alternatives are available. Key components are listed in TABLE 3.21

TABLE 3
Beers criteria:* Drug classes that may be inappropriate for older adults
21

Drug classConcern
Alpha-blockers with peripheral activityOrthostatic hypotension
AnticholinergicsCognitive impairment, urinary retention
AntipsychoticsIncreased death rate when used for behavior control in patients with dementia
NSAIDsRenal dysfunction, GI bleeding, fluid retention, exacerbation of heart failure
Sedative hypnoticsCognitive impairment, delirium
Tricyclic antidepressantsCognitive impairment, delirium, urinary retention
GI, gastrointestinal; NSAIDs, nonsteroidal anti-inflammatory drugs.
*The full Beers criteria contains 53 drugs and drug classes that are generally inappropriate for older adults. The full list is available from the American Geriatrics Society at: www.americangeriatrics.org/files/documents/beers/2012BeersCriteria_JAGS.pdf.

One limitation of the Beers criteria has been its all-or-nothing approach, with many of the medications on the list deemed inappropriate for all older adults regardless of their circumstances. The 2012 update does a better job of individualizing recommendations: Medications are now categorized as those that should be avoided in older patients regardless of their diseases or conditions, those that should be avoided only in patients with certain diseases or conditions, and those that may be used for this patient population but require caution.21

STOPP is similar to the Beers criteria, but uses a different approach: Most medications on this list are considered in the context of specific medical problems.22 While the Beers criteria classify digoxin >0.125 mg/d as generally inappropriate for older adults, for example, STOPP criteria state that long-term dosing at that level is inappropriate only for those with impaired renal function.22 A list of medications identified by STOPP as contributing to hospitalization due to adverse drug effects is available at http://ageing.oxfordjournals.org/content/37/6/673.

Both tools address this drug category. Cumulative anticholinergic burden is a concept applied to the use of anticholinergic medications, which are included in both the Beers and STOPP criteria. Although isolated short-term exposure to a drug with anticholinergic properties may be tolerated by a healthy and cognitively intact older patient, repetitive exposure to such drugs, even if separated in time, has negative effects. One study evaluated more than 500 community-dwelling older adults and found that the more exposure an individual had to anticholinergic medications over the course of a year, the greater the impairment in short-term memory and activities of daily living.23 Another study, this one involving more than 13,000 community-dwelling and institutionalized patients, showed that the longer an older patient takes an anticholinergic medication, the more likely there is to be a measurable decline in performance on the Mini-Mental State Examination.24

Programs that flag potential interactions
Drug-drug interactions are a key concern of polypharmacy, and electronic medical records and prescribing systems that flag potential drug-drug interactions when a new medication is ordered are designed to help physicians avoid them. Unfortunately, clinicians only react to 3% to 9% of such notifications, overriding them because computerized systems often fail to distinguish between important and unimportant interactions.25-27 Thus, clinicians often must decide whether to react to or override warnings, an often difficult decision with patient safety and medicolegal implications. The best advice we can offer is to carefully evaluate drug interaction warnings using common sense, and seek consultation with a clinical pharmacist when uncertainty exists. This approach should prevent prescribing medications that have potentially harmful interactions with drugs the patient is already taking.

For physicians who do not have access to an electronic prescribing system that provides such notification, several online resources are available, some by subscription (eg, Lexicomp, www.lexi.com; Micromedex, www.micromedex.com/index.html; and Pepid, www.pepid.com) and others with free access (eg, AARP, healthtools.aarp.org/drug-interactions; Drugs.com (www.drugs.com/drug_interactions.php; and HealthLine, www.healthline.com/druginteractions).

CASE After doing a medication reconciliation for Mrs. R, you find that she is taking tolterodine, an anticholinergic medication for urge urinary incontinence, and donepezil, a procholinergic medication for dementia. This type of drug-drug interaction, in which the action of one drug effectively cancels out the effect of another, should not be ignored.

Overall, you identify 8 of her medications that could be discontinued: The list includes guaifenesin (a nonessential medication of questionable efficacy); naproxen (inappropriate per Beers criteria; inappropriate in patients with heart failure, according to STOPP); alprazolam, zolpidem, and diphenhydramine (duplicate medications that are all on the Beers criteria as inappropriate for chronic use and ill-advised in patients with cognitive impairment); and omeprazole and levothyroxine (for which nothing in the patient’s history suggests a need), as well as tolterodine. Depending on dose, digoxin is yet another candidate for discontinuation.

 

 

Discontinuing medications: Proceed carefully

Physicians are often reluctant to discontinue chronic medications in older patients—even in those with advanced disease who are not likely to benefit from treatment. Focus groups have identified a number of reasons for their hesitation, including:

  • the assumption that patients have no problem taking large numbers of drugs
  • the fear that patients may misinterpret a plan to discontinue medications as evidence that the physician is giving up on them
  • the belief that physicians must comply with practice guidelines that recommend multiple drug treatments
  • concern that proposing discontinuation of medications often leads to a discussion of life expectancy and end-of-life care.28

Physicians may also fear that discontinuation of certain drugs will increase the risk of adverse outcomes. More than 30 studies have evaluated discontinuation of chronic medications in older adults, however, and found that drugs as diverse as antihypertensives, antipsychotics, benzodiazepines, and selective serotonin reuptake inhibitors (SSRIs) can often be discontinued without adverse outcomes. In many cases, improvement in patient function results.29 Medications that present the most difficulty are those that patients often become physically or psychologically dependent on, such as benzodiazepines, guaifenesin, proton-pump inhibitors, nonsteroidal anti-inflammatory drugs, and SSRIs. Some (eg, benzodiazepines, SSRIs) require a gradual reduction; for others, no taper is required
(TABLE 4).30-37

TABLE 4
Recommendations for discontinuing hard-to-stop drugs

Medication or drug classDiscontinuation regimenComments
Benzodiazepines30Taper dose by 25% q 2 wkNo withdrawal symptoms reported with this taper regimen. Subtle cognitive improvement noted over a period of months
Guaifenesin31Can be discontinued without tapering if not combined with opioids or other medications. Elimination half-life is approximately 1 hourGuaifenesin is often marketed as a combination product with opioids; such combination products require tapering
PPIs32-34Decrease dose by 50% q 2 wk; supplement with H2 blocker if needed, but tapering of H2 blocker may be requiredAbrupt discontinuation after long-term use causes rebound gastric acid hypersecretion and lowers rate of success. Higher success rates with taper regimen and in patients who do not have documented GERD
NSAIDs35No taper requiredShort-term use (<3 mo) acceptable for patients with no contraindications
SSRIs36,37Gradual reduction in dose over 6-8 wkHighest rate of success in patients without a clear diagnosis of depression
GERD, gastroesophageal reflux disease; NSAIDs, nonsteroidal anti-inflammatory drugs; PPIs, proton-pump inhibitors; SSRIs, selective serotonin reuptake inhibitors.

CASE You trim down Mrs. R’s regimen by discontinuing each of the 8 drugs, one at a time, and carefully monitor the patient during the withdrawal period. Because she had been taking alprazolam daily, the dose is tapered slowly to avoid withdrawal. Omeprazole also requires a gradual taper to avoid rebound hyperacidity.3

After confirming that Mrs. R has heart failure and COPD, you identify 2 medications that should be added to her drug regimen—an ACE inhibitor for heart failure and an inhaled anticholinergic for COPD.

Going from 16 medications to 10 saves money, decreases the likelihood of adverse events and drug-drug interactions, and helps with adherence. Mrs. R’s new drug regimen is expected to lead to improvements in memory and overall quality of life, as well.

CORRESPONDENCE 
Barry D. Weiss, MD, Department of Family and Community Medicine, University of Arizona College of Medicine, Tucson, AZ 85724; [email protected]

References

1. Gu Q, Dillon CF, Burt V. Prescription drug use continued to increase: US prescription drug data for 2007-2008. CDC/NCHS Data Brief. 2010;42:1-2.

2. Jessup K, Abraham WT, Casey DE, et al. 2009 focused update: ACCF/AHA guidelines for the diagnosis and management of heart failure in adults. A report of the American College of Cardiology Foundation/American Heart Association Task Force on Practice Guidelines. Circulation. 2009;119:1977-2016.

3. Johnell K, Klarin I. The relationship between number of drugs and potential drug-drug interactions in the elderly: a study of over 600,000 elderly patients from the Swedish Prescribed Drug Register. Drug Saf. 2007;30:911-918.

4. Sarkar U, Lopez A, Maselli JH, et al. Adverse drug events in US adult ambulatory medical care. Health Services Res. 2011;46:1517-1533.

5. Rollason V, Vogt N. Reduction of polypharmacy in the elderly. A systematic review of the role of the pharmacist. Drugs Aging. 2003;20:817-832.

6. National Heart, Lung, and Blood Institute. Seventh Report of the Joint National Committee on Prevention, Detection, Evaluation, and Treatment of High Blood Pressure. Available at: www.nhlbi.nih.gov/guidelines/hypertension/jnc7full.pdf. Accessed October 11, 2012.

7. Steinman MA, Hanlon JT. Managing medications in clinically complex elders. JAMA. 2010;304:1592-1601.

8. Steinman MA, Landefeld CS, Rosenthal GE, et al. Polypharmacy and prescribing quality in older people. J Am Geriatr Soc. 2006;54:1516-23.

9. Bell CM, Brener SS, Gunraj N, et al. Association of ICU or hospital admission with unintentional discontinuations of medications for chronic disease. JAMA. 2011;306:840-847.

10. Moore C, Wisnivesky J, Williams S, et al. Medical errors related to discontinuity of care from an inpatient to an outpatient setting. J Gen Intern Med. 2003;18:646-651.

11. Ziaeian B, Arauho KL, Van Ness PH, et al. Medication reconciliation accuracy and patient understanding of intended medication changes on hospital discharge. J Gen Intern Med. 2012 July 12. ePub ahead of print.

12. Hajjar ER, Hanlon JT, Sloane RJ, et al. Unnecessary drug use in frail older people at hospital discharge. J Am Geriatr Soc. 2005;53:1518-1523.

13. O’Mahony D, Gallagher P, Ryan C, et al. STOPP & START criteria: a new approach to detecting potentially inappropriate prescribing in old age. Eur Geriatr Med. 2010;1:45-51.

14. Denneboom W, Dautzenberg KGH, Grol R, et al. Analysis of polypharmacy in older patients in primary acre using a multidisciplinary expert panel. Br J Gen Pract. 2006;56:504-510.

15. Ko DT, Mamdani M, Alter DA. Lipid-lowering therapy with statins in high-risk elderly patients. JAMA. 2004;291:1864-70.

16. Wright RM, Sloane R, Pieper CF, et al. Underuse of indicated medications among physically frail older US veterans at the time of hospital discharge: results of a cross-sectional analysis of data from the Geriatric Evaluation and Management Drug Study. Am J Geriatr Pharmacother. 2009;7:271-280.

17. Garwood CL. Use of anticoagulation in elderly patients with atrial fibrillation who are risk for falls. Ann Pharmacother. 2008;42:523-532.

18. Holmes HM, Hayley DC, Alexander GC, et al. Reconsidering medication appropriateness for patients late in life. Arch Intern Med. 2006;166:605-609.

19. Simplified Methods for Estimating Life Expectancy. Available at: http://painconsortium.nih.gov/symptomresearch/chapter_14/Part_3/sec4/chspt3s4pg1.htm. Accessed October 9, 2012.

20. Beers MH, Ouslander JG, Rollingher I, et al. Explicit criteria for determining inappropriate medication use in nursing home residents. Arch Intern Med. 1991;151:1825-1832.

21. The American Geriatrics Society 2012 Beers Criteria Update Expert Panel. American Geriatrics Society Update Beers Criteria for potentially inappropriate medication use in older adults. J Am Geriatr Soc. 2012;60:616-631.

22. Gallagher P, O’Mahony D. STOPP (Screening Tool of Older Persons’ potentially inappropriate Prescriptions): application to acutely ill elderly patients and comparison with Beers’ criteria. Age Aging. 2008;37:673-379.

23. Han L, Agostini JV, Allore HG. Cumulative anticholinergic exposure is associated with poor memory and executive function in older men. J Am Geriatr Soc. 2008;56:2203-2210.

24. Fox C, Richardson K, Maidment ID, et al. Anticholinergic medication use and cognitive impairment in the older population: the medical research council cognitive function and ageing study. J Am Geriatr Soc. 2011;59:1477-1483.

25. Knight A, Falade O, Maygers J, et al. Factors associated with medication warning acceptance [abstract]. J Hosp Med. 2012;7(suppl 2):515.-

26. Isaac T, Weissman JS, Davis RB, et al. Overrides of medication alerts in ambulatory care. Arch Intern Med. 2009;169:305-311.

27. Van Der Sijs H, Aarts J, Vulto A, et al. Overriding of drug safety alerts in computerized physician order entry. J Am Med Inform Assoc. 2006;12:138-147.

28. Schuling J, Gebben H, Veehof LJG, et al. Deprescribing medication in very elderly patients with multimorbidity: the view of Dutch GPs. A qualitative study. BMC Family Practice. 2012;13:56. http://www.biomedcentral.com/1471-2296/13/56.

29. Iyer S, Naganathan V, McLachlan AJ, et al. Medication withdrawal trials in people aged 65 years and older. A systematic review. Drugs Aging. 2008;25:1021-1031.

30. Curran HV, Collins R, Fletcher S, et al. Older adults and withdrawal from benzodiazepine hypnotics in general practice: effects on cognitive function, sleep, mood and quality of life. Psychol Med. 2003;33:1223-1237.

31. Krinsky DL, Berardi RR, Ferreris SP, et al. Handbook of Nonprescription Drugs: An Interactive Approach to Self-Care. Washington, DC: American Pharmacists Association; 2012:209.

32. Bjornsson E, Abrahamsson H, Simren M, et al. Discontinuation of proton pump inhibitors in patients on long-term therapy: a double-blind, placebo-controlled trial. Aliment Pharmacol Ther. 2006;24:945-954.

33. Inadomi JM, Jamai R, Murata GH, et al. Step-down management of gastroesophageal reflux disease. Gastroenterology. 2001;131:1095-1100.

34. Hester SA. Proton pump inhibitors and rebound acid hypersecretion. Pharm Lett. 2009;25:250920.-

35. Taylor R, Jr, Lemtouni S, Weiss K, et al. Pain management in the elderly: an FDA safe use initiative expert panel’s view on preventable harm associated with NSAID therapy. Curr Gerontol Geriatr Res. 2012;196159.-

36. Ulfvarson J, Adami J, Wredling R, et al. Controlled withdrawal of selective serotonin reuptake inhibitor drugs in elderly patients in nursing homes with no indication of depression. Eur J Clin Pharmacol. 2003;59:735-740.

37. Lindstrom K, Ekedahl A, Carlsten A, et al. Can selective serotonin inhibitor drugs in elderly patients in nursing homes be reduced? Scand J Prim Health Care. 2007;25:3-8.

Article PDF
Author and Disclosure Information

Barry D. Weiss, MD
Arizona Center on Aging, Department of Family and Community Medicine, University of Arizona College of Medicine, Tucson
[email protected]

Jeannie K. Lee, PharmD, BCPS
Arizona Center on Aging, Department of Pharmacy Practice and Science, University of Arizona College of Pharmacy, Tucson

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

Issue
The Journal of Family Practice - 61(11)
Publications
Topics
Page Number
652-661
Legacy Keywords
Barry D. Weiss;MD; Jeannie K. Lee;PharmD;BCPS; polypharmacy; adverse drug effect; age-inappropriate; multiple medications; medication review; medication reconcilliation
Sections
Author and Disclosure Information

Barry D. Weiss, MD
Arizona Center on Aging, Department of Family and Community Medicine, University of Arizona College of Medicine, Tucson
[email protected]

Jeannie K. Lee, PharmD, BCPS
Arizona Center on Aging, Department of Pharmacy Practice and Science, University of Arizona College of Pharmacy, Tucson

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

Author and Disclosure Information

Barry D. Weiss, MD
Arizona Center on Aging, Department of Family and Community Medicine, University of Arizona College of Medicine, Tucson
[email protected]

Jeannie K. Lee, PharmD, BCPS
Arizona Center on Aging, Department of Pharmacy Practice and Science, University of Arizona College of Pharmacy, Tucson

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

Article PDF
Article PDF

PRACTICE RECOMMENDATIONS

Consider the possibility that an adverse drug effect—rather than a new condition—is at play when a patient taking multiple medications develops a new symptom. C

Use an online interaction checker, which can be accessed via a smart phone or tablet, to check for potential drug-drug interactions in patients on multiple medications. C

Cross-check patients’ medications with a list of their medical problems, with the goal of discontinuing any drug that duplicates the action of another or is age-inappropriate, ineffective, or not indicated for the condition for which it was prescribed. 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

Older adults are taking more medications than ever before. Nearly 9 out of 10 US residents who are 60 years of age or older take at least one prescription drug, more than a third take 5 to 9 medications, and 12% take 10 or more.1

The increase is largely driven by newer medications to effectively treat a variety of medical conditions, and by practice guidelines that often recommend multidrug regimens.2

As a result, the term “polypharmacy,” which once referred to a specific number of medications, is now used more broadly to mean “a large number” of drugs.

From a safety standpoint, the number of medications a patient takes matters. The risk of adverse drug effects and dangerous drug-drug interactions increases significantly when an individual takes ≥5 medications.3

More than 4.5 million adverse drug effects occur each year in the United States, and nearly three quarters of them are initially evaluated in outpatient settings.4 Research suggests that about 80% of the time, these adverse effects are not recognized as such by the patient’s physician. So instead of discontinuing the offending medication, physicians treat the drug-related symptoms by adding yet another medication—a phenomenon known as “the prescribing cascade.”5

This review can help you safeguard older patients taking multiple medications by recognizing and responding to drug-related problems, identifying drugs that can be safely eliminated (or, in some cases, drugs that should be added), and checking regularly to ensure that the medication regimen is appropriate and up to date.

CASE Mrs. R, a 79-year-old woman who recently moved to town, is brought to your office by her daughter and son-in-law. The patient has a hard time reporting her medical history, but her daughter tells you her mother has chronic obstructive pulmonary disease (COPD), heart failure, type 2 diabetes, and mild urinary incontinence, and was recently diagnosed with early dementia.

Mrs. R’s daughter has brought in a bagful of medications, but she’s not sure which ones her mother takes regularly. The medications are an albuterol inhaler, alprazolam, digoxin, diphenhydramine, donepezil, furosemide, glargine insulin, guaifenesin, levothyroxine, metformin, extended-release metoprolol, naproxen, omeprazole, simvastatin, tolterodine, and zolpidem—a total of 16 different drugs.

If Mrs. R were your patient, how would you manage her multidrug regimen?

Start with a medication review

The first step in evaluating a patient’s medication regimen is to find out whether the drugs in the patient’s possession and/or in the medical record are the ones he or she is actually taking. Ask older patients who haven’t brought in their medications, or the caregiver of a confused patient, to bring them to the next visit.

The next step: Determine whether the medication regimen is right for the patient.

Polypharmacy may be indicated
Despite the risks associated with polypharmacy, do not assume that it is inappropriate. For some conditions, multiple medications are routinely recommended. Patients with heart failure, for example, have been shown to have better outcomes when they take 3 to 5 medications, including beta-blockers, angiotensin-converting enzyme (ACE) inhibitors, and diuretics.2

Some treatment guidelines also call for multiple medications. Achieving the more stringent blood pressure goals recommended in the Seventh Report of the Joint National Committee on Prevention, for instance, often requires 2 or more antihypertensive agents.6 In many cases, however, patients end up taking more drugs than necessary.

Is the patient taking the right drugs?
Medication reconciliation (determining whether the treatment regimen is appropriate for the patient’s diagnoses) is the way to find out.

The most widely recommended approach to medication reconciliation is to create a table and do a systematic review.7 List all the patient’s medical conditions in the first column and all current medications in the second column. Use the third column to note whether each medication is one the patient should be on, based not only on his or her medical conditions and other drugs being taken but also on current renal and hepatic function and body size, and contraindications.

 

 

A medication may be inappropriate if it duplicates, cancels out the action of, or otherwise interacts with another drug the patient is taking; is contraindicated in older patients; or is ineffective for the condition for which it was prescribed. In one key study of nearly 200 patients 65 years and older who took 5 or more medications, more than half had been prescribed at least one drug that was ineffective for the patient’s condition or that duplicated the action of another medication.8

In addition to finding drugs that the patient should not be taking, medication reconciliation may also reveal that the patient is not receiving optimal therapy and that one or more drugs should be added to his or her treatment regimen.

Check meds after transitions. A move from home to hospital, from emergency department to home, or any other transition relating to patient care should prompt a medication reconciliation. Medications are often added or inadvertently discontinued at such times,9,10 and instructions relating to medication are often misunderstood.11 In one study of 384 frail elderly patients being discharged from a hospital, for example, 44% were found to have been given at least one unnecessary prescription—most commonly for a medication that was neither indicated nor effective for any of the patient’s medical problems.12 It was also common for patients to be given drugs that duplicated the action of others they were already taking.

Even in the absence of such transitions, medication reconciliation should occur at regular intervals. Many physicians do a medication reconciliation at every visit to ensure that the medical record is accurate and the patient’s medication regimen is optimal.

Managing polypharmacy: These resources can help

Numerous tools are available to help you evaluate and monitor patients’ medication regimens, including some that were developed specifically for older patients.

START (Screening Tool to Alert doctors to Right Treatment) identifies drugs and drug classes that are underused with older patients.13 START criteria (TABLE 1)13-17 focus on medications that should be used yet are often omitted in older patients who have the appropriate indications.

TABLE 1
START criteria: Drug therapy that should be given to older patients
13-17

Cardiovascular
  • Anticoagulation or antiplatelet therapy for atrial fibrillation
  • Antiplatelet therapy for patients with known coronary, cerebral, or peripheral vascular disease
  • Antihypertensive therapy for systolic BP >160 mm hg
  • Statins for secondary prevention in patients with coronary, cerebral, or peripheral vascular disease (with life expectancy >5 years)
  • ACE inhibitor for heart failure or after MI
  • Beta-blocker for chronic stable angina
Endocrine
  • Metformin for type 2 diabetes
  • ACE inhibitor for patients with diabetes and nephropathy
  • Antiplatelet and statin therapy for patients with diabetes and CVD risk factors
Gastrointestinal
  • PPI for severe gi reflux or esophageal stricture
  • Fiber supplement for chronic symptomatic diverticular disease
Musculoskeletal
  • Antirheumatic drugs for moderate-to-severe chronic rheumatoid disease
  • Bisphosphonates for patients taking chronic oral steroids
  • Calcium and vitamin D for osteoporosis
Nervous system
  • Levodopa for Parkinson’s disease with functional impairment
  • Antidepressant for moderate-to-severe depression lasting >3 months
Respiratory
  • Daily inhaled beta-agonist or anticholinergic agent for asthma or COPD
  • Daily inhaled steroid for asthma or COPD with FEV1 <50% of predicted value
  • Continuous home oxygen for chronic hypoxemic respiratory failure
ACE, angiotensin-converting enzyme; BP, blood pressure; COPD, chronic obstructive pulmonary disease; CVD, cardiovascular disease; FEV1, forced expiratory volume in 1 second; GI, gastrointestinal; MI, myocardial infarction; PPI, proton-pump inhibitor; START, Screening Tool to alert doctors to right Treatment.

In using START or any other drug-related tool, it is important to keep in mind that therapy should be individualized. Not all the medications in the START criteria are appropriate for every patient, and a medication that is indicated for a given medical condition may or may not provide real benefit for a particular patient. That would depend on the individual’s overall health and life expectancy, the goals of treatment, and how long it would take for the patient to realize any benefit from the drug in question.18 A vigorous 79-year-old might benefit from statin therapy for prevention of cardiovascular events, for instance, while a patient like Mrs. R, who is also 79 but has dementia and multiple other medical problems, would be unlikely to live long enough to realize such a benefit.

”Age” assessment tool. One criterion in deciding whether medication(s) are appropriate for an older patient is his or her “physiologic age”—calculated on the basis of the individual’s chronological age and self-reported health status (TABLE 2).19

TABLE 2
Calculating your patient’s “real” age
19

Actual age (y)Physiologic age (y)
Self-reported health
ExcellentGoodFairPoor
MaleFemaleMaleFemaleMaleFemaleMaleFemale
655860646468667372
706265696973717877
756770747478768382
8072757979838185+85+

Flagging drugs that may be inappropriate
Several tools have been developed to aid clinicians in identifying medications that are potentially inappropriate for older adults, although here, too, decisions about their use must be individualized. Two of the most widely used tools are the Beers criteria and STOPP (Screening Tool of Older Persons’ potentially inappropriate Prescriptions).

 

 

Beers criteria were developed by Mark Beers et al in 199120 and have been updated at regular intervals, most recently by the American Geriatrics Society in 2012.21 The drugs and drug classes included in the Beers criteria should not be prescribed for older patients in most cases, either because the risk of using them outweighs the benefit or because safer alternatives are available. Key components are listed in TABLE 3.21

TABLE 3
Beers criteria:* Drug classes that may be inappropriate for older adults
21

Drug classConcern
Alpha-blockers with peripheral activityOrthostatic hypotension
AnticholinergicsCognitive impairment, urinary retention
AntipsychoticsIncreased death rate when used for behavior control in patients with dementia
NSAIDsRenal dysfunction, GI bleeding, fluid retention, exacerbation of heart failure
Sedative hypnoticsCognitive impairment, delirium
Tricyclic antidepressantsCognitive impairment, delirium, urinary retention
GI, gastrointestinal; NSAIDs, nonsteroidal anti-inflammatory drugs.
*The full Beers criteria contains 53 drugs and drug classes that are generally inappropriate for older adults. The full list is available from the American Geriatrics Society at: www.americangeriatrics.org/files/documents/beers/2012BeersCriteria_JAGS.pdf.

One limitation of the Beers criteria has been its all-or-nothing approach, with many of the medications on the list deemed inappropriate for all older adults regardless of their circumstances. The 2012 update does a better job of individualizing recommendations: Medications are now categorized as those that should be avoided in older patients regardless of their diseases or conditions, those that should be avoided only in patients with certain diseases or conditions, and those that may be used for this patient population but require caution.21

STOPP is similar to the Beers criteria, but uses a different approach: Most medications on this list are considered in the context of specific medical problems.22 While the Beers criteria classify digoxin >0.125 mg/d as generally inappropriate for older adults, for example, STOPP criteria state that long-term dosing at that level is inappropriate only for those with impaired renal function.22 A list of medications identified by STOPP as contributing to hospitalization due to adverse drug effects is available at http://ageing.oxfordjournals.org/content/37/6/673.

Both tools address this drug category. Cumulative anticholinergic burden is a concept applied to the use of anticholinergic medications, which are included in both the Beers and STOPP criteria. Although isolated short-term exposure to a drug with anticholinergic properties may be tolerated by a healthy and cognitively intact older patient, repetitive exposure to such drugs, even if separated in time, has negative effects. One study evaluated more than 500 community-dwelling older adults and found that the more exposure an individual had to anticholinergic medications over the course of a year, the greater the impairment in short-term memory and activities of daily living.23 Another study, this one involving more than 13,000 community-dwelling and institutionalized patients, showed that the longer an older patient takes an anticholinergic medication, the more likely there is to be a measurable decline in performance on the Mini-Mental State Examination.24

Programs that flag potential interactions
Drug-drug interactions are a key concern of polypharmacy, and electronic medical records and prescribing systems that flag potential drug-drug interactions when a new medication is ordered are designed to help physicians avoid them. Unfortunately, clinicians only react to 3% to 9% of such notifications, overriding them because computerized systems often fail to distinguish between important and unimportant interactions.25-27 Thus, clinicians often must decide whether to react to or override warnings, an often difficult decision with patient safety and medicolegal implications. The best advice we can offer is to carefully evaluate drug interaction warnings using common sense, and seek consultation with a clinical pharmacist when uncertainty exists. This approach should prevent prescribing medications that have potentially harmful interactions with drugs the patient is already taking.

For physicians who do not have access to an electronic prescribing system that provides such notification, several online resources are available, some by subscription (eg, Lexicomp, www.lexi.com; Micromedex, www.micromedex.com/index.html; and Pepid, www.pepid.com) and others with free access (eg, AARP, healthtools.aarp.org/drug-interactions; Drugs.com (www.drugs.com/drug_interactions.php; and HealthLine, www.healthline.com/druginteractions).

CASE After doing a medication reconciliation for Mrs. R, you find that she is taking tolterodine, an anticholinergic medication for urge urinary incontinence, and donepezil, a procholinergic medication for dementia. This type of drug-drug interaction, in which the action of one drug effectively cancels out the effect of another, should not be ignored.

Overall, you identify 8 of her medications that could be discontinued: The list includes guaifenesin (a nonessential medication of questionable efficacy); naproxen (inappropriate per Beers criteria; inappropriate in patients with heart failure, according to STOPP); alprazolam, zolpidem, and diphenhydramine (duplicate medications that are all on the Beers criteria as inappropriate for chronic use and ill-advised in patients with cognitive impairment); and omeprazole and levothyroxine (for which nothing in the patient’s history suggests a need), as well as tolterodine. Depending on dose, digoxin is yet another candidate for discontinuation.

 

 

Discontinuing medications: Proceed carefully

Physicians are often reluctant to discontinue chronic medications in older patients—even in those with advanced disease who are not likely to benefit from treatment. Focus groups have identified a number of reasons for their hesitation, including:

  • the assumption that patients have no problem taking large numbers of drugs
  • the fear that patients may misinterpret a plan to discontinue medications as evidence that the physician is giving up on them
  • the belief that physicians must comply with practice guidelines that recommend multiple drug treatments
  • concern that proposing discontinuation of medications often leads to a discussion of life expectancy and end-of-life care.28

Physicians may also fear that discontinuation of certain drugs will increase the risk of adverse outcomes. More than 30 studies have evaluated discontinuation of chronic medications in older adults, however, and found that drugs as diverse as antihypertensives, antipsychotics, benzodiazepines, and selective serotonin reuptake inhibitors (SSRIs) can often be discontinued without adverse outcomes. In many cases, improvement in patient function results.29 Medications that present the most difficulty are those that patients often become physically or psychologically dependent on, such as benzodiazepines, guaifenesin, proton-pump inhibitors, nonsteroidal anti-inflammatory drugs, and SSRIs. Some (eg, benzodiazepines, SSRIs) require a gradual reduction; for others, no taper is required
(TABLE 4).30-37

TABLE 4
Recommendations for discontinuing hard-to-stop drugs

Medication or drug classDiscontinuation regimenComments
Benzodiazepines30Taper dose by 25% q 2 wkNo withdrawal symptoms reported with this taper regimen. Subtle cognitive improvement noted over a period of months
Guaifenesin31Can be discontinued without tapering if not combined with opioids or other medications. Elimination half-life is approximately 1 hourGuaifenesin is often marketed as a combination product with opioids; such combination products require tapering
PPIs32-34Decrease dose by 50% q 2 wk; supplement with H2 blocker if needed, but tapering of H2 blocker may be requiredAbrupt discontinuation after long-term use causes rebound gastric acid hypersecretion and lowers rate of success. Higher success rates with taper regimen and in patients who do not have documented GERD
NSAIDs35No taper requiredShort-term use (<3 mo) acceptable for patients with no contraindications
SSRIs36,37Gradual reduction in dose over 6-8 wkHighest rate of success in patients without a clear diagnosis of depression
GERD, gastroesophageal reflux disease; NSAIDs, nonsteroidal anti-inflammatory drugs; PPIs, proton-pump inhibitors; SSRIs, selective serotonin reuptake inhibitors.

CASE You trim down Mrs. R’s regimen by discontinuing each of the 8 drugs, one at a time, and carefully monitor the patient during the withdrawal period. Because she had been taking alprazolam daily, the dose is tapered slowly to avoid withdrawal. Omeprazole also requires a gradual taper to avoid rebound hyperacidity.3

After confirming that Mrs. R has heart failure and COPD, you identify 2 medications that should be added to her drug regimen—an ACE inhibitor for heart failure and an inhaled anticholinergic for COPD.

Going from 16 medications to 10 saves money, decreases the likelihood of adverse events and drug-drug interactions, and helps with adherence. Mrs. R’s new drug regimen is expected to lead to improvements in memory and overall quality of life, as well.

CORRESPONDENCE 
Barry D. Weiss, MD, Department of Family and Community Medicine, University of Arizona College of Medicine, Tucson, AZ 85724; [email protected]

PRACTICE RECOMMENDATIONS

Consider the possibility that an adverse drug effect—rather than a new condition—is at play when a patient taking multiple medications develops a new symptom. C

Use an online interaction checker, which can be accessed via a smart phone or tablet, to check for potential drug-drug interactions in patients on multiple medications. C

Cross-check patients’ medications with a list of their medical problems, with the goal of discontinuing any drug that duplicates the action of another or is age-inappropriate, ineffective, or not indicated for the condition for which it was prescribed. 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

Older adults are taking more medications than ever before. Nearly 9 out of 10 US residents who are 60 years of age or older take at least one prescription drug, more than a third take 5 to 9 medications, and 12% take 10 or more.1

The increase is largely driven by newer medications to effectively treat a variety of medical conditions, and by practice guidelines that often recommend multidrug regimens.2

As a result, the term “polypharmacy,” which once referred to a specific number of medications, is now used more broadly to mean “a large number” of drugs.

From a safety standpoint, the number of medications a patient takes matters. The risk of adverse drug effects and dangerous drug-drug interactions increases significantly when an individual takes ≥5 medications.3

More than 4.5 million adverse drug effects occur each year in the United States, and nearly three quarters of them are initially evaluated in outpatient settings.4 Research suggests that about 80% of the time, these adverse effects are not recognized as such by the patient’s physician. So instead of discontinuing the offending medication, physicians treat the drug-related symptoms by adding yet another medication—a phenomenon known as “the prescribing cascade.”5

This review can help you safeguard older patients taking multiple medications by recognizing and responding to drug-related problems, identifying drugs that can be safely eliminated (or, in some cases, drugs that should be added), and checking regularly to ensure that the medication regimen is appropriate and up to date.

CASE Mrs. R, a 79-year-old woman who recently moved to town, is brought to your office by her daughter and son-in-law. The patient has a hard time reporting her medical history, but her daughter tells you her mother has chronic obstructive pulmonary disease (COPD), heart failure, type 2 diabetes, and mild urinary incontinence, and was recently diagnosed with early dementia.

Mrs. R’s daughter has brought in a bagful of medications, but she’s not sure which ones her mother takes regularly. The medications are an albuterol inhaler, alprazolam, digoxin, diphenhydramine, donepezil, furosemide, glargine insulin, guaifenesin, levothyroxine, metformin, extended-release metoprolol, naproxen, omeprazole, simvastatin, tolterodine, and zolpidem—a total of 16 different drugs.

If Mrs. R were your patient, how would you manage her multidrug regimen?

Start with a medication review

The first step in evaluating a patient’s medication regimen is to find out whether the drugs in the patient’s possession and/or in the medical record are the ones he or she is actually taking. Ask older patients who haven’t brought in their medications, or the caregiver of a confused patient, to bring them to the next visit.

The next step: Determine whether the medication regimen is right for the patient.

Polypharmacy may be indicated
Despite the risks associated with polypharmacy, do not assume that it is inappropriate. For some conditions, multiple medications are routinely recommended. Patients with heart failure, for example, have been shown to have better outcomes when they take 3 to 5 medications, including beta-blockers, angiotensin-converting enzyme (ACE) inhibitors, and diuretics.2

Some treatment guidelines also call for multiple medications. Achieving the more stringent blood pressure goals recommended in the Seventh Report of the Joint National Committee on Prevention, for instance, often requires 2 or more antihypertensive agents.6 In many cases, however, patients end up taking more drugs than necessary.

Is the patient taking the right drugs?
Medication reconciliation (determining whether the treatment regimen is appropriate for the patient’s diagnoses) is the way to find out.

The most widely recommended approach to medication reconciliation is to create a table and do a systematic review.7 List all the patient’s medical conditions in the first column and all current medications in the second column. Use the third column to note whether each medication is one the patient should be on, based not only on his or her medical conditions and other drugs being taken but also on current renal and hepatic function and body size, and contraindications.

 

 

A medication may be inappropriate if it duplicates, cancels out the action of, or otherwise interacts with another drug the patient is taking; is contraindicated in older patients; or is ineffective for the condition for which it was prescribed. In one key study of nearly 200 patients 65 years and older who took 5 or more medications, more than half had been prescribed at least one drug that was ineffective for the patient’s condition or that duplicated the action of another medication.8

In addition to finding drugs that the patient should not be taking, medication reconciliation may also reveal that the patient is not receiving optimal therapy and that one or more drugs should be added to his or her treatment regimen.

Check meds after transitions. A move from home to hospital, from emergency department to home, or any other transition relating to patient care should prompt a medication reconciliation. Medications are often added or inadvertently discontinued at such times,9,10 and instructions relating to medication are often misunderstood.11 In one study of 384 frail elderly patients being discharged from a hospital, for example, 44% were found to have been given at least one unnecessary prescription—most commonly for a medication that was neither indicated nor effective for any of the patient’s medical problems.12 It was also common for patients to be given drugs that duplicated the action of others they were already taking.

Even in the absence of such transitions, medication reconciliation should occur at regular intervals. Many physicians do a medication reconciliation at every visit to ensure that the medical record is accurate and the patient’s medication regimen is optimal.

Managing polypharmacy: These resources can help

Numerous tools are available to help you evaluate and monitor patients’ medication regimens, including some that were developed specifically for older patients.

START (Screening Tool to Alert doctors to Right Treatment) identifies drugs and drug classes that are underused with older patients.13 START criteria (TABLE 1)13-17 focus on medications that should be used yet are often omitted in older patients who have the appropriate indications.

TABLE 1
START criteria: Drug therapy that should be given to older patients
13-17

Cardiovascular
  • Anticoagulation or antiplatelet therapy for atrial fibrillation
  • Antiplatelet therapy for patients with known coronary, cerebral, or peripheral vascular disease
  • Antihypertensive therapy for systolic BP >160 mm hg
  • Statins for secondary prevention in patients with coronary, cerebral, or peripheral vascular disease (with life expectancy >5 years)
  • ACE inhibitor for heart failure or after MI
  • Beta-blocker for chronic stable angina
Endocrine
  • Metformin for type 2 diabetes
  • ACE inhibitor for patients with diabetes and nephropathy
  • Antiplatelet and statin therapy for patients with diabetes and CVD risk factors
Gastrointestinal
  • PPI for severe gi reflux or esophageal stricture
  • Fiber supplement for chronic symptomatic diverticular disease
Musculoskeletal
  • Antirheumatic drugs for moderate-to-severe chronic rheumatoid disease
  • Bisphosphonates for patients taking chronic oral steroids
  • Calcium and vitamin D for osteoporosis
Nervous system
  • Levodopa for Parkinson’s disease with functional impairment
  • Antidepressant for moderate-to-severe depression lasting >3 months
Respiratory
  • Daily inhaled beta-agonist or anticholinergic agent for asthma or COPD
  • Daily inhaled steroid for asthma or COPD with FEV1 <50% of predicted value
  • Continuous home oxygen for chronic hypoxemic respiratory failure
ACE, angiotensin-converting enzyme; BP, blood pressure; COPD, chronic obstructive pulmonary disease; CVD, cardiovascular disease; FEV1, forced expiratory volume in 1 second; GI, gastrointestinal; MI, myocardial infarction; PPI, proton-pump inhibitor; START, Screening Tool to alert doctors to right Treatment.

In using START or any other drug-related tool, it is important to keep in mind that therapy should be individualized. Not all the medications in the START criteria are appropriate for every patient, and a medication that is indicated for a given medical condition may or may not provide real benefit for a particular patient. That would depend on the individual’s overall health and life expectancy, the goals of treatment, and how long it would take for the patient to realize any benefit from the drug in question.18 A vigorous 79-year-old might benefit from statin therapy for prevention of cardiovascular events, for instance, while a patient like Mrs. R, who is also 79 but has dementia and multiple other medical problems, would be unlikely to live long enough to realize such a benefit.

”Age” assessment tool. One criterion in deciding whether medication(s) are appropriate for an older patient is his or her “physiologic age”—calculated on the basis of the individual’s chronological age and self-reported health status (TABLE 2).19

TABLE 2
Calculating your patient’s “real” age
19

Actual age (y)Physiologic age (y)
Self-reported health
ExcellentGoodFairPoor
MaleFemaleMaleFemaleMaleFemaleMaleFemale
655860646468667372
706265696973717877
756770747478768382
8072757979838185+85+

Flagging drugs that may be inappropriate
Several tools have been developed to aid clinicians in identifying medications that are potentially inappropriate for older adults, although here, too, decisions about their use must be individualized. Two of the most widely used tools are the Beers criteria and STOPP (Screening Tool of Older Persons’ potentially inappropriate Prescriptions).

 

 

Beers criteria were developed by Mark Beers et al in 199120 and have been updated at regular intervals, most recently by the American Geriatrics Society in 2012.21 The drugs and drug classes included in the Beers criteria should not be prescribed for older patients in most cases, either because the risk of using them outweighs the benefit or because safer alternatives are available. Key components are listed in TABLE 3.21

TABLE 3
Beers criteria:* Drug classes that may be inappropriate for older adults
21

Drug classConcern
Alpha-blockers with peripheral activityOrthostatic hypotension
AnticholinergicsCognitive impairment, urinary retention
AntipsychoticsIncreased death rate when used for behavior control in patients with dementia
NSAIDsRenal dysfunction, GI bleeding, fluid retention, exacerbation of heart failure
Sedative hypnoticsCognitive impairment, delirium
Tricyclic antidepressantsCognitive impairment, delirium, urinary retention
GI, gastrointestinal; NSAIDs, nonsteroidal anti-inflammatory drugs.
*The full Beers criteria contains 53 drugs and drug classes that are generally inappropriate for older adults. The full list is available from the American Geriatrics Society at: www.americangeriatrics.org/files/documents/beers/2012BeersCriteria_JAGS.pdf.

One limitation of the Beers criteria has been its all-or-nothing approach, with many of the medications on the list deemed inappropriate for all older adults regardless of their circumstances. The 2012 update does a better job of individualizing recommendations: Medications are now categorized as those that should be avoided in older patients regardless of their diseases or conditions, those that should be avoided only in patients with certain diseases or conditions, and those that may be used for this patient population but require caution.21

STOPP is similar to the Beers criteria, but uses a different approach: Most medications on this list are considered in the context of specific medical problems.22 While the Beers criteria classify digoxin >0.125 mg/d as generally inappropriate for older adults, for example, STOPP criteria state that long-term dosing at that level is inappropriate only for those with impaired renal function.22 A list of medications identified by STOPP as contributing to hospitalization due to adverse drug effects is available at http://ageing.oxfordjournals.org/content/37/6/673.

Both tools address this drug category. Cumulative anticholinergic burden is a concept applied to the use of anticholinergic medications, which are included in both the Beers and STOPP criteria. Although isolated short-term exposure to a drug with anticholinergic properties may be tolerated by a healthy and cognitively intact older patient, repetitive exposure to such drugs, even if separated in time, has negative effects. One study evaluated more than 500 community-dwelling older adults and found that the more exposure an individual had to anticholinergic medications over the course of a year, the greater the impairment in short-term memory and activities of daily living.23 Another study, this one involving more than 13,000 community-dwelling and institutionalized patients, showed that the longer an older patient takes an anticholinergic medication, the more likely there is to be a measurable decline in performance on the Mini-Mental State Examination.24

Programs that flag potential interactions
Drug-drug interactions are a key concern of polypharmacy, and electronic medical records and prescribing systems that flag potential drug-drug interactions when a new medication is ordered are designed to help physicians avoid them. Unfortunately, clinicians only react to 3% to 9% of such notifications, overriding them because computerized systems often fail to distinguish between important and unimportant interactions.25-27 Thus, clinicians often must decide whether to react to or override warnings, an often difficult decision with patient safety and medicolegal implications. The best advice we can offer is to carefully evaluate drug interaction warnings using common sense, and seek consultation with a clinical pharmacist when uncertainty exists. This approach should prevent prescribing medications that have potentially harmful interactions with drugs the patient is already taking.

For physicians who do not have access to an electronic prescribing system that provides such notification, several online resources are available, some by subscription (eg, Lexicomp, www.lexi.com; Micromedex, www.micromedex.com/index.html; and Pepid, www.pepid.com) and others with free access (eg, AARP, healthtools.aarp.org/drug-interactions; Drugs.com (www.drugs.com/drug_interactions.php; and HealthLine, www.healthline.com/druginteractions).

CASE After doing a medication reconciliation for Mrs. R, you find that she is taking tolterodine, an anticholinergic medication for urge urinary incontinence, and donepezil, a procholinergic medication for dementia. This type of drug-drug interaction, in which the action of one drug effectively cancels out the effect of another, should not be ignored.

Overall, you identify 8 of her medications that could be discontinued: The list includes guaifenesin (a nonessential medication of questionable efficacy); naproxen (inappropriate per Beers criteria; inappropriate in patients with heart failure, according to STOPP); alprazolam, zolpidem, and diphenhydramine (duplicate medications that are all on the Beers criteria as inappropriate for chronic use and ill-advised in patients with cognitive impairment); and omeprazole and levothyroxine (for which nothing in the patient’s history suggests a need), as well as tolterodine. Depending on dose, digoxin is yet another candidate for discontinuation.

 

 

Discontinuing medications: Proceed carefully

Physicians are often reluctant to discontinue chronic medications in older patients—even in those with advanced disease who are not likely to benefit from treatment. Focus groups have identified a number of reasons for their hesitation, including:

  • the assumption that patients have no problem taking large numbers of drugs
  • the fear that patients may misinterpret a plan to discontinue medications as evidence that the physician is giving up on them
  • the belief that physicians must comply with practice guidelines that recommend multiple drug treatments
  • concern that proposing discontinuation of medications often leads to a discussion of life expectancy and end-of-life care.28

Physicians may also fear that discontinuation of certain drugs will increase the risk of adverse outcomes. More than 30 studies have evaluated discontinuation of chronic medications in older adults, however, and found that drugs as diverse as antihypertensives, antipsychotics, benzodiazepines, and selective serotonin reuptake inhibitors (SSRIs) can often be discontinued without adverse outcomes. In many cases, improvement in patient function results.29 Medications that present the most difficulty are those that patients often become physically or psychologically dependent on, such as benzodiazepines, guaifenesin, proton-pump inhibitors, nonsteroidal anti-inflammatory drugs, and SSRIs. Some (eg, benzodiazepines, SSRIs) require a gradual reduction; for others, no taper is required
(TABLE 4).30-37

TABLE 4
Recommendations for discontinuing hard-to-stop drugs

Medication or drug classDiscontinuation regimenComments
Benzodiazepines30Taper dose by 25% q 2 wkNo withdrawal symptoms reported with this taper regimen. Subtle cognitive improvement noted over a period of months
Guaifenesin31Can be discontinued without tapering if not combined with opioids or other medications. Elimination half-life is approximately 1 hourGuaifenesin is often marketed as a combination product with opioids; such combination products require tapering
PPIs32-34Decrease dose by 50% q 2 wk; supplement with H2 blocker if needed, but tapering of H2 blocker may be requiredAbrupt discontinuation after long-term use causes rebound gastric acid hypersecretion and lowers rate of success. Higher success rates with taper regimen and in patients who do not have documented GERD
NSAIDs35No taper requiredShort-term use (<3 mo) acceptable for patients with no contraindications
SSRIs36,37Gradual reduction in dose over 6-8 wkHighest rate of success in patients without a clear diagnosis of depression
GERD, gastroesophageal reflux disease; NSAIDs, nonsteroidal anti-inflammatory drugs; PPIs, proton-pump inhibitors; SSRIs, selective serotonin reuptake inhibitors.

CASE You trim down Mrs. R’s regimen by discontinuing each of the 8 drugs, one at a time, and carefully monitor the patient during the withdrawal period. Because she had been taking alprazolam daily, the dose is tapered slowly to avoid withdrawal. Omeprazole also requires a gradual taper to avoid rebound hyperacidity.3

After confirming that Mrs. R has heart failure and COPD, you identify 2 medications that should be added to her drug regimen—an ACE inhibitor for heart failure and an inhaled anticholinergic for COPD.

Going from 16 medications to 10 saves money, decreases the likelihood of adverse events and drug-drug interactions, and helps with adherence. Mrs. R’s new drug regimen is expected to lead to improvements in memory and overall quality of life, as well.

CORRESPONDENCE 
Barry D. Weiss, MD, Department of Family and Community Medicine, University of Arizona College of Medicine, Tucson, AZ 85724; [email protected]

References

1. Gu Q, Dillon CF, Burt V. Prescription drug use continued to increase: US prescription drug data for 2007-2008. CDC/NCHS Data Brief. 2010;42:1-2.

2. Jessup K, Abraham WT, Casey DE, et al. 2009 focused update: ACCF/AHA guidelines for the diagnosis and management of heart failure in adults. A report of the American College of Cardiology Foundation/American Heart Association Task Force on Practice Guidelines. Circulation. 2009;119:1977-2016.

3. Johnell K, Klarin I. The relationship between number of drugs and potential drug-drug interactions in the elderly: a study of over 600,000 elderly patients from the Swedish Prescribed Drug Register. Drug Saf. 2007;30:911-918.

4. Sarkar U, Lopez A, Maselli JH, et al. Adverse drug events in US adult ambulatory medical care. Health Services Res. 2011;46:1517-1533.

5. Rollason V, Vogt N. Reduction of polypharmacy in the elderly. A systematic review of the role of the pharmacist. Drugs Aging. 2003;20:817-832.

6. National Heart, Lung, and Blood Institute. Seventh Report of the Joint National Committee on Prevention, Detection, Evaluation, and Treatment of High Blood Pressure. Available at: www.nhlbi.nih.gov/guidelines/hypertension/jnc7full.pdf. Accessed October 11, 2012.

7. Steinman MA, Hanlon JT. Managing medications in clinically complex elders. JAMA. 2010;304:1592-1601.

8. Steinman MA, Landefeld CS, Rosenthal GE, et al. Polypharmacy and prescribing quality in older people. J Am Geriatr Soc. 2006;54:1516-23.

9. Bell CM, Brener SS, Gunraj N, et al. Association of ICU or hospital admission with unintentional discontinuations of medications for chronic disease. JAMA. 2011;306:840-847.

10. Moore C, Wisnivesky J, Williams S, et al. Medical errors related to discontinuity of care from an inpatient to an outpatient setting. J Gen Intern Med. 2003;18:646-651.

11. Ziaeian B, Arauho KL, Van Ness PH, et al. Medication reconciliation accuracy and patient understanding of intended medication changes on hospital discharge. J Gen Intern Med. 2012 July 12. ePub ahead of print.

12. Hajjar ER, Hanlon JT, Sloane RJ, et al. Unnecessary drug use in frail older people at hospital discharge. J Am Geriatr Soc. 2005;53:1518-1523.

13. O’Mahony D, Gallagher P, Ryan C, et al. STOPP & START criteria: a new approach to detecting potentially inappropriate prescribing in old age. Eur Geriatr Med. 2010;1:45-51.

14. Denneboom W, Dautzenberg KGH, Grol R, et al. Analysis of polypharmacy in older patients in primary acre using a multidisciplinary expert panel. Br J Gen Pract. 2006;56:504-510.

15. Ko DT, Mamdani M, Alter DA. Lipid-lowering therapy with statins in high-risk elderly patients. JAMA. 2004;291:1864-70.

16. Wright RM, Sloane R, Pieper CF, et al. Underuse of indicated medications among physically frail older US veterans at the time of hospital discharge: results of a cross-sectional analysis of data from the Geriatric Evaluation and Management Drug Study. Am J Geriatr Pharmacother. 2009;7:271-280.

17. Garwood CL. Use of anticoagulation in elderly patients with atrial fibrillation who are risk for falls. Ann Pharmacother. 2008;42:523-532.

18. Holmes HM, Hayley DC, Alexander GC, et al. Reconsidering medication appropriateness for patients late in life. Arch Intern Med. 2006;166:605-609.

19. Simplified Methods for Estimating Life Expectancy. Available at: http://painconsortium.nih.gov/symptomresearch/chapter_14/Part_3/sec4/chspt3s4pg1.htm. Accessed October 9, 2012.

20. Beers MH, Ouslander JG, Rollingher I, et al. Explicit criteria for determining inappropriate medication use in nursing home residents. Arch Intern Med. 1991;151:1825-1832.

21. The American Geriatrics Society 2012 Beers Criteria Update Expert Panel. American Geriatrics Society Update Beers Criteria for potentially inappropriate medication use in older adults. J Am Geriatr Soc. 2012;60:616-631.

22. Gallagher P, O’Mahony D. STOPP (Screening Tool of Older Persons’ potentially inappropriate Prescriptions): application to acutely ill elderly patients and comparison with Beers’ criteria. Age Aging. 2008;37:673-379.

23. Han L, Agostini JV, Allore HG. Cumulative anticholinergic exposure is associated with poor memory and executive function in older men. J Am Geriatr Soc. 2008;56:2203-2210.

24. Fox C, Richardson K, Maidment ID, et al. Anticholinergic medication use and cognitive impairment in the older population: the medical research council cognitive function and ageing study. J Am Geriatr Soc. 2011;59:1477-1483.

25. Knight A, Falade O, Maygers J, et al. Factors associated with medication warning acceptance [abstract]. J Hosp Med. 2012;7(suppl 2):515.-

26. Isaac T, Weissman JS, Davis RB, et al. Overrides of medication alerts in ambulatory care. Arch Intern Med. 2009;169:305-311.

27. Van Der Sijs H, Aarts J, Vulto A, et al. Overriding of drug safety alerts in computerized physician order entry. J Am Med Inform Assoc. 2006;12:138-147.

28. Schuling J, Gebben H, Veehof LJG, et al. Deprescribing medication in very elderly patients with multimorbidity: the view of Dutch GPs. A qualitative study. BMC Family Practice. 2012;13:56. http://www.biomedcentral.com/1471-2296/13/56.

29. Iyer S, Naganathan V, McLachlan AJ, et al. Medication withdrawal trials in people aged 65 years and older. A systematic review. Drugs Aging. 2008;25:1021-1031.

30. Curran HV, Collins R, Fletcher S, et al. Older adults and withdrawal from benzodiazepine hypnotics in general practice: effects on cognitive function, sleep, mood and quality of life. Psychol Med. 2003;33:1223-1237.

31. Krinsky DL, Berardi RR, Ferreris SP, et al. Handbook of Nonprescription Drugs: An Interactive Approach to Self-Care. Washington, DC: American Pharmacists Association; 2012:209.

32. Bjornsson E, Abrahamsson H, Simren M, et al. Discontinuation of proton pump inhibitors in patients on long-term therapy: a double-blind, placebo-controlled trial. Aliment Pharmacol Ther. 2006;24:945-954.

33. Inadomi JM, Jamai R, Murata GH, et al. Step-down management of gastroesophageal reflux disease. Gastroenterology. 2001;131:1095-1100.

34. Hester SA. Proton pump inhibitors and rebound acid hypersecretion. Pharm Lett. 2009;25:250920.-

35. Taylor R, Jr, Lemtouni S, Weiss K, et al. Pain management in the elderly: an FDA safe use initiative expert panel’s view on preventable harm associated with NSAID therapy. Curr Gerontol Geriatr Res. 2012;196159.-

36. Ulfvarson J, Adami J, Wredling R, et al. Controlled withdrawal of selective serotonin reuptake inhibitor drugs in elderly patients in nursing homes with no indication of depression. Eur J Clin Pharmacol. 2003;59:735-740.

37. Lindstrom K, Ekedahl A, Carlsten A, et al. Can selective serotonin inhibitor drugs in elderly patients in nursing homes be reduced? Scand J Prim Health Care. 2007;25:3-8.

References

1. Gu Q, Dillon CF, Burt V. Prescription drug use continued to increase: US prescription drug data for 2007-2008. CDC/NCHS Data Brief. 2010;42:1-2.

2. Jessup K, Abraham WT, Casey DE, et al. 2009 focused update: ACCF/AHA guidelines for the diagnosis and management of heart failure in adults. A report of the American College of Cardiology Foundation/American Heart Association Task Force on Practice Guidelines. Circulation. 2009;119:1977-2016.

3. Johnell K, Klarin I. The relationship between number of drugs and potential drug-drug interactions in the elderly: a study of over 600,000 elderly patients from the Swedish Prescribed Drug Register. Drug Saf. 2007;30:911-918.

4. Sarkar U, Lopez A, Maselli JH, et al. Adverse drug events in US adult ambulatory medical care. Health Services Res. 2011;46:1517-1533.

5. Rollason V, Vogt N. Reduction of polypharmacy in the elderly. A systematic review of the role of the pharmacist. Drugs Aging. 2003;20:817-832.

6. National Heart, Lung, and Blood Institute. Seventh Report of the Joint National Committee on Prevention, Detection, Evaluation, and Treatment of High Blood Pressure. Available at: www.nhlbi.nih.gov/guidelines/hypertension/jnc7full.pdf. Accessed October 11, 2012.

7. Steinman MA, Hanlon JT. Managing medications in clinically complex elders. JAMA. 2010;304:1592-1601.

8. Steinman MA, Landefeld CS, Rosenthal GE, et al. Polypharmacy and prescribing quality in older people. J Am Geriatr Soc. 2006;54:1516-23.

9. Bell CM, Brener SS, Gunraj N, et al. Association of ICU or hospital admission with unintentional discontinuations of medications for chronic disease. JAMA. 2011;306:840-847.

10. Moore C, Wisnivesky J, Williams S, et al. Medical errors related to discontinuity of care from an inpatient to an outpatient setting. J Gen Intern Med. 2003;18:646-651.

11. Ziaeian B, Arauho KL, Van Ness PH, et al. Medication reconciliation accuracy and patient understanding of intended medication changes on hospital discharge. J Gen Intern Med. 2012 July 12. ePub ahead of print.

12. Hajjar ER, Hanlon JT, Sloane RJ, et al. Unnecessary drug use in frail older people at hospital discharge. J Am Geriatr Soc. 2005;53:1518-1523.

13. O’Mahony D, Gallagher P, Ryan C, et al. STOPP & START criteria: a new approach to detecting potentially inappropriate prescribing in old age. Eur Geriatr Med. 2010;1:45-51.

14. Denneboom W, Dautzenberg KGH, Grol R, et al. Analysis of polypharmacy in older patients in primary acre using a multidisciplinary expert panel. Br J Gen Pract. 2006;56:504-510.

15. Ko DT, Mamdani M, Alter DA. Lipid-lowering therapy with statins in high-risk elderly patients. JAMA. 2004;291:1864-70.

16. Wright RM, Sloane R, Pieper CF, et al. Underuse of indicated medications among physically frail older US veterans at the time of hospital discharge: results of a cross-sectional analysis of data from the Geriatric Evaluation and Management Drug Study. Am J Geriatr Pharmacother. 2009;7:271-280.

17. Garwood CL. Use of anticoagulation in elderly patients with atrial fibrillation who are risk for falls. Ann Pharmacother. 2008;42:523-532.

18. Holmes HM, Hayley DC, Alexander GC, et al. Reconsidering medication appropriateness for patients late in life. Arch Intern Med. 2006;166:605-609.

19. Simplified Methods for Estimating Life Expectancy. Available at: http://painconsortium.nih.gov/symptomresearch/chapter_14/Part_3/sec4/chspt3s4pg1.htm. Accessed October 9, 2012.

20. Beers MH, Ouslander JG, Rollingher I, et al. Explicit criteria for determining inappropriate medication use in nursing home residents. Arch Intern Med. 1991;151:1825-1832.

21. The American Geriatrics Society 2012 Beers Criteria Update Expert Panel. American Geriatrics Society Update Beers Criteria for potentially inappropriate medication use in older adults. J Am Geriatr Soc. 2012;60:616-631.

22. Gallagher P, O’Mahony D. STOPP (Screening Tool of Older Persons’ potentially inappropriate Prescriptions): application to acutely ill elderly patients and comparison with Beers’ criteria. Age Aging. 2008;37:673-379.

23. Han L, Agostini JV, Allore HG. Cumulative anticholinergic exposure is associated with poor memory and executive function in older men. J Am Geriatr Soc. 2008;56:2203-2210.

24. Fox C, Richardson K, Maidment ID, et al. Anticholinergic medication use and cognitive impairment in the older population: the medical research council cognitive function and ageing study. J Am Geriatr Soc. 2011;59:1477-1483.

25. Knight A, Falade O, Maygers J, et al. Factors associated with medication warning acceptance [abstract]. J Hosp Med. 2012;7(suppl 2):515.-

26. Isaac T, Weissman JS, Davis RB, et al. Overrides of medication alerts in ambulatory care. Arch Intern Med. 2009;169:305-311.

27. Van Der Sijs H, Aarts J, Vulto A, et al. Overriding of drug safety alerts in computerized physician order entry. J Am Med Inform Assoc. 2006;12:138-147.

28. Schuling J, Gebben H, Veehof LJG, et al. Deprescribing medication in very elderly patients with multimorbidity: the view of Dutch GPs. A qualitative study. BMC Family Practice. 2012;13:56. http://www.biomedcentral.com/1471-2296/13/56.

29. Iyer S, Naganathan V, McLachlan AJ, et al. Medication withdrawal trials in people aged 65 years and older. A systematic review. Drugs Aging. 2008;25:1021-1031.

30. Curran HV, Collins R, Fletcher S, et al. Older adults and withdrawal from benzodiazepine hypnotics in general practice: effects on cognitive function, sleep, mood and quality of life. Psychol Med. 2003;33:1223-1237.

31. Krinsky DL, Berardi RR, Ferreris SP, et al. Handbook of Nonprescription Drugs: An Interactive Approach to Self-Care. Washington, DC: American Pharmacists Association; 2012:209.

32. Bjornsson E, Abrahamsson H, Simren M, et al. Discontinuation of proton pump inhibitors in patients on long-term therapy: a double-blind, placebo-controlled trial. Aliment Pharmacol Ther. 2006;24:945-954.

33. Inadomi JM, Jamai R, Murata GH, et al. Step-down management of gastroesophageal reflux disease. Gastroenterology. 2001;131:1095-1100.

34. Hester SA. Proton pump inhibitors and rebound acid hypersecretion. Pharm Lett. 2009;25:250920.-

35. Taylor R, Jr, Lemtouni S, Weiss K, et al. Pain management in the elderly: an FDA safe use initiative expert panel’s view on preventable harm associated with NSAID therapy. Curr Gerontol Geriatr Res. 2012;196159.-

36. Ulfvarson J, Adami J, Wredling R, et al. Controlled withdrawal of selective serotonin reuptake inhibitor drugs in elderly patients in nursing homes with no indication of depression. Eur J Clin Pharmacol. 2003;59:735-740.

37. Lindstrom K, Ekedahl A, Carlsten A, et al. Can selective serotonin inhibitor drugs in elderly patients in nursing homes be reduced? Scand J Prim Health Care. 2007;25:3-8.

Issue
The Journal of Family Practice - 61(11)
Issue
The Journal of Family Practice - 61(11)
Page Number
652-661
Page Number
652-661
Publications
Publications
Topics
Article Type
Display Headline
AGING: Is your patient taking too many pills?
Display Headline
AGING: Is your patient taking too many pills?
Legacy Keywords
Barry D. Weiss;MD; Jeannie K. Lee;PharmD;BCPS; polypharmacy; adverse drug effect; age-inappropriate; multiple medications; medication review; medication reconcilliation
Legacy Keywords
Barry D. Weiss;MD; Jeannie K. Lee;PharmD;BCPS; polypharmacy; adverse drug effect; age-inappropriate; multiple medications; medication review; medication reconcilliation
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media

Infant’s brain damage blamed on delayed delivery … and more

Article Type
Changed
Thu, 03/28/2019 - 16:12
Display Headline
Infant’s brain damage blamed on delayed delivery … and more

Infant’s brain damage blamed on delayed delivery

DURING DELIVERY, THE MOTHER’S PERINATOLOGIST recognized a severe shoulder dystocia. The perinatologist abandoned vaginal delivery and ordered an emergency cesarean delivery. The mother was transferred to an operating room (OR) with the baby’s head out between her legs. In the OR, the perinatologist pushed the baby’s head back into the uterus and performed a cesarean extraction. Nineteen minutes elapsed from when the vaginal delivery was abandoned and the baby was delivered.

The child was unresponsive at birth with no spontaneous movement or respiration. She was intubated and transferred to the NICU, where she was resuscitated. MRI confirmed that the child had hypoxic ischemia and severe, permanent brain damage from acute birth asphyxia. The child is blind, deaf, hypertensive, and has diffuse spasticity. She has a tracheostomy, a gastrostomy tube, and requires 24-hour care.

PARENTS’ CLAIM The perinatologist was negligent for abandoning vaginal delivery when delivery was progressing appropriately and there was no fetal distress. If the perinatologist had rotated the baby’s shoulder to the oblique position and/or used suprapubic pressure, the shoulder would have become disimpacted and the baby would have been safely delivered within seconds. Delay in delivery allowed for 19 minutes of umbilical cord compression, resulting in brain damage.

PHYSICIAN’S DEFENSE Cesarean delivery was appropriate; the baby did not suffer cord compression. Injury to the brain occurred days before delivery, based on prenatal ultrasonography.

VERDICT A $5.5 million California settlement was reached.

Failure to diagnose breast cancer: death

A 38-YEAR-OLD WOMAN went to her primary care physician (PCP) 3 years after giving birth. She reported breast pain, nipple discharge, and a dime-sized lump. The woman was still breastfeeding. An exam by the nurse practitioner (NP) was limited because the patient had breast implants. The NP suspected a galactocele and advised the patient to stop breastfeeding and apply ice packs. When the patient returned in 2 weeks, only the lump remained. The PCP determined that she had mastitis.

Five months later, she returned with additional lumps in both breasts, and was referred to a gynecologist. Ultrasonography (US) was ordered, but the patient never followed up. A year later, the patient was found to have metastatic breast cancer and died after 3 years of treatment.

ESTATE’S CLAIM The PCP and NP were negligent for not referring her for a breast biopsy when a lump was first detected.

DEFENDANTS’ DEFENSE Proper care was given. An earlier diagnosis would not have changed the outcome.

VERDICT A $750,000 Massachusetts settlement was reached.

What caused this child’s autism?

AFTER 33 HOURS OF LABOR, a baby was delivered vaginally by an ObGyn, nurse, and midwife. The child was diagnosed with autism several years later. His development is delayed, and he suffers cognitive impairment.

PARENTS’ CLAIM The child’s autism is due to a prolonged hypoxic event during labor. Fetal heart-rate monitoring demonstrated fetal distress, with a bradycardia. A cesarean delivery should have been performed.

PHYSICIAN’S DEFENSE The child has genetic autism unrelated to the birth process.

VERDICT A $1.35 million New York settlement was reached.

Was oxytocin the culprit?

DURING AN EXTENDED LABOR, the ObGyn continued to give the mother oxytocin, although there were signs of fetal distress. The child was born with brain damage, cannot walk, talk, or see, and requires 24-hour care.

PATIENT’S CLAIM The use of oxytocin was inappropriate given the signs of fetal distress. Oxytocin caused a lack of oxygen to the child, resulting in brain damage. A cesarean delivery should have been performed when fetal distress was identified.

DEFENDANTS’ DEFENSE The case was settled before trial.

VERDICT A $12 million Illinois settlement was reached: $11 million from the hospital and $1 million from the ObGyn.

Bowel perforation, sepsis after ovary removal

DURING LEFT OOPHORECTOMY, the ObGyn encountered adhesions. Five days later, the 41-year-old patient reported severe pain. A second procedure revealed sepsis and perforation of the large bowel. A colostomy was performed. The patient underwent additional corrective operations.

PATIENT’S CLAIM The ObGyn was negligent for causing tissue damage to the colon that perforated and escalated into sepsis. A surgeon should have been consulted when the ObGyn found the adhesions, so the bowel could be properly inspected before the abdomen was closed. The physician was also negligent for not recognizing symptoms of sepsis earlier.

PHYSICIAN’S DEFENSE Bowel injury is a known complication of oophorectomy. The patient appeared to be making a fairly good recovery until infection became evident; she was immediately treated.

VERDICT A $6.3 million New Jersey verdict was returned, including $300,000 for the husband’s loss of consortium.

Traumatic delivery causes seizures

 

 

DURING CESAREAN DELIVERY, the ObGyn rotated the baby from a transverse to a cephalic lie, and used a vacuum extractor to deliver the head through the hysterotomy incision.

When the child was 25 hours old, he suffered a seizure that lasted 6 minutes. Focal seizure activity involving the left side of his body and a skull fracture were identified. He was transferred to another hospital, where radiologic studies indicated a middle right cerebral artery infarct. The child developed an ongoing seizure disorder, speech and language delays, and mild, left-sided weakness.

PARENTS’ CLAIM The baby’s head was not properly delivered through the cesarean incision nor should the ObGyn have used vacuum extraction. The combination of the rotation and use of vacuum caused trauma to the infant’s head. In addition, the baby was placed in the well-baby nursery, which was inappropriate because he was born through thick meconium, resuscitated by a neonatal nurse, and had a depressed skull fracture.

PHYSICIAN’S DEFENSE Delivery was not traumatic; all treatment was appropriate.

VERDICT A $4.6 million New York settlement was reached with the hospital and ObGyn’s insurer.

Mother gets severe headache during birth

A 35-YEAR-OLD WOMAN began having a severe headache during delivery that continued after birth. She was discharged from the hospital and collapsed at home a day later. She was returned to the ED, where she was left in a hallway for 6 hours. She lost consciousness while in the hallway. Imaging and neurologic evaluation determined that she suffered a hypoxic brain injury from intracranial bleeding. She has slow response time, difficulty with all aspects of everyday life, and requires full-time attendant care.

PATIENT’S CLAIM Although she complained of a headache, no testing was done prior to her hospital discharge. Treatment was extremely delayed in the ED; an earlier diagnosis could have prevented brain damage.

PHYSICIAN’S DEFENSE Nothing could have prevented the brain damage.

VERDICT A $3.5 million California settlement was mediated.

Shoulder dystocia; brachial plexus injury

WHEN SHOULDER DYSTOCIA was encountered during delivery, the ObGyn applied gentle pressure to deliver the head. He was assisted by an ObGyn resident. The child was born with a brachial plexus injury, causing left-arm paralysis. She underwent surgery that increased her range of motion, but she will need years of physical therapy.

PATIENT’S CLAIM The ObGyn applied excessive traction and the resident improperly applied fundal pressure.

DEFENDANTS’ DEFENSE Only gentle traction was used. The resident did not apply fundal pressure.

VERDICT A New York jury found the ObGyn at fault and awarded the patient $3.5 million. The resident was vindicated.

Was premature baby viable?

AN EXPECTANT MOTHER MISCARRIED AT HOME at 6 months’ gestation, and an ambulance was called. After the EMTs helped the mother to the ambulance, they retrieved the fetus. When the baby was seen moving its head, the EMTs requested assistance from the advanced life support (ALS) team. ALS personnel visually assessed the fetus, determined it was nonviable, and placed the baby in a small container. The mother and baby arrived at the hospital 17 minutes after the ambulance was called.

At the hospital, a nurse noticed that the fetus was warm and had a heartbeat. The baby was taken to a special-care nursery for resuscitation and then transferred to another hospital’s NICU. The baby died after 46 days from severe brain damage due to lack of oxygen.

PARENTS’ CLAIM The EMTs and ALS team should have provided better evaluation and treatment for the infant; they were not trained to determine an infant’s viability. Placing the infant inside a plastic bag inside a box with a lid further deprived the baby of oxygen.

DEFENDANTS’ DEFENSE The case was settled before trial.

VERDICT A $1 million Massachusetts settlement was reached.

Were records altered because of a delayed diagnosis?

A WOMAN FOUND A LUMP in her left breast. A gynecologist ordered mammography. In January 2006, the radiologist requested ultrasonography (US), and reported that it conclusively indicated that the mass was a cyst. The gynecologist told the patient the tests were normal; further action was unnecessary. The patient saw the gynecologist four more times before being referred to a breast surgeon. In June 2006, she underwent surgical resection and chemotherapy for a malignant breast tumor.

PATIENT’S CLAIM The gynecologist was negligent for not referring the patient to a surgeon earlier. The gynecologist altered records: excerpts from the mammogram and US reports had been scanned in with a notation that the gynecologist had told the patient to follow up with a surgeon. When the gynecologist faxed the same reports to the surgeon, the annotations were absent. The gynecologist also changed the December 2005 chart, which referred to an US she never ordered.

 

 

PHYSICIAN’S DEFENSE The gynecologist stated that she regularly “merged” two reports into one document in her practice.

VERDICT A $700,000 Pennsylvania verdict was returned.

Excessive force or standard of care?

SHOULDER DYSTOCIA occurred during labor. The child sustained left brachial plexus palsy. At age 6, his left arm is paralyzed and smaller than the right arm. He has trouble performing normal daily tasks.

PATIENT’S CLAIM The ObGyn used excessive force by pulling on the baby’s head to complete the delivery. Standard of care required the ObGyn to take a more gentle approach to achieve delivery.

PHYSICIAN’S DEFENSE Delivery was performed appropriately, and did not deviate from standard of care.

VERDICT A $20.881 million Maryland verdict was returned, including $20 million for pain and suffering. The total award was reduced to $1,531,082 when the pain and suffering award was cut to $650,000 under the state’s statutory cap.

Preterm birth from an asymptomatic UTI?

A BABY WAS BORN AT 31 WEEKS’ gestation. The child has cerebral palsy, spastic quadriplegia, and requires assistance in all aspects of life.

PARENTS’ CLAIM Chorioamnionitis from a urinary tract infection (UTI) caused preterm birth. Urinalysis performed 7 weeks earlier indicated an infection, but the second-year resident caring for the mother failed to treat the UTI. The resident should have obtained a confirming urine culture, prescribed antibiotics, and monitored the mother more closely. The resident was poorly supervised.

DEFENDANTS’ DEFENSE Chorioamnionitis developed just before birth and could not be detected or prevented. A UTI cannot remain asymptomatic for 7 weeks and still cause premature birth. The mother was at increased risk of premature delivery because she had given birth to an anencephalic infant a year earlier. She began prenatal care in the middle of her pregnancy and ignored a referral to a high-risk maternal fetal specialist.

VERDICT A New York defense verdict was returned.

References

These cases were selected by the editors of OBG Management from Medical Malpractice Verdicts, Settlements & Experts, with permission of the editor, Lewis Laska (www.verdictslaska.com). The information available to the editors about the cases presented here is sometimes incomplete. Moreover, the cases may or may not have merit. Nevertheless, these cases represent the types of clinical situations that typically result in litigation and are meant to illustrate nationwide variation in jury verdicts and awards.

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

Article PDF
Author and Disclosure Information

Issue
OBG Management - 24(11)
Publications
Topics
Page Number
46-49
Legacy Keywords
Medical malpractice;medical verdicts;negligence;defense;settlement;Lewis Laska;Medical Malpractice Verdicts Settlements and Experts;delayed delivery;brain damage;perinatologist;vaginal delivery;cesarean;NICU;breast cancer;galactocele;breast biopsy;autism;fetal distress;oxytocin;bowel perforation;sepsis;traumatic delivery;focal seizure;vacuum extraction;headache;hypoxic brain injury;shoulder dystocia;brachial plexus injury;miscarriage;ALS team;delayed diagnosis;chorioamnionitis;UTI
Sections
Author and Disclosure Information

Author and Disclosure Information

Article PDF
Article PDF

Infant’s brain damage blamed on delayed delivery

DURING DELIVERY, THE MOTHER’S PERINATOLOGIST recognized a severe shoulder dystocia. The perinatologist abandoned vaginal delivery and ordered an emergency cesarean delivery. The mother was transferred to an operating room (OR) with the baby’s head out between her legs. In the OR, the perinatologist pushed the baby’s head back into the uterus and performed a cesarean extraction. Nineteen minutes elapsed from when the vaginal delivery was abandoned and the baby was delivered.

The child was unresponsive at birth with no spontaneous movement or respiration. She was intubated and transferred to the NICU, where she was resuscitated. MRI confirmed that the child had hypoxic ischemia and severe, permanent brain damage from acute birth asphyxia. The child is blind, deaf, hypertensive, and has diffuse spasticity. She has a tracheostomy, a gastrostomy tube, and requires 24-hour care.

PARENTS’ CLAIM The perinatologist was negligent for abandoning vaginal delivery when delivery was progressing appropriately and there was no fetal distress. If the perinatologist had rotated the baby’s shoulder to the oblique position and/or used suprapubic pressure, the shoulder would have become disimpacted and the baby would have been safely delivered within seconds. Delay in delivery allowed for 19 minutes of umbilical cord compression, resulting in brain damage.

PHYSICIAN’S DEFENSE Cesarean delivery was appropriate; the baby did not suffer cord compression. Injury to the brain occurred days before delivery, based on prenatal ultrasonography.

VERDICT A $5.5 million California settlement was reached.

Failure to diagnose breast cancer: death

A 38-YEAR-OLD WOMAN went to her primary care physician (PCP) 3 years after giving birth. She reported breast pain, nipple discharge, and a dime-sized lump. The woman was still breastfeeding. An exam by the nurse practitioner (NP) was limited because the patient had breast implants. The NP suspected a galactocele and advised the patient to stop breastfeeding and apply ice packs. When the patient returned in 2 weeks, only the lump remained. The PCP determined that she had mastitis.

Five months later, she returned with additional lumps in both breasts, and was referred to a gynecologist. Ultrasonography (US) was ordered, but the patient never followed up. A year later, the patient was found to have metastatic breast cancer and died after 3 years of treatment.

ESTATE’S CLAIM The PCP and NP were negligent for not referring her for a breast biopsy when a lump was first detected.

DEFENDANTS’ DEFENSE Proper care was given. An earlier diagnosis would not have changed the outcome.

VERDICT A $750,000 Massachusetts settlement was reached.

What caused this child’s autism?

AFTER 33 HOURS OF LABOR, a baby was delivered vaginally by an ObGyn, nurse, and midwife. The child was diagnosed with autism several years later. His development is delayed, and he suffers cognitive impairment.

PARENTS’ CLAIM The child’s autism is due to a prolonged hypoxic event during labor. Fetal heart-rate monitoring demonstrated fetal distress, with a bradycardia. A cesarean delivery should have been performed.

PHYSICIAN’S DEFENSE The child has genetic autism unrelated to the birth process.

VERDICT A $1.35 million New York settlement was reached.

Was oxytocin the culprit?

DURING AN EXTENDED LABOR, the ObGyn continued to give the mother oxytocin, although there were signs of fetal distress. The child was born with brain damage, cannot walk, talk, or see, and requires 24-hour care.

PATIENT’S CLAIM The use of oxytocin was inappropriate given the signs of fetal distress. Oxytocin caused a lack of oxygen to the child, resulting in brain damage. A cesarean delivery should have been performed when fetal distress was identified.

DEFENDANTS’ DEFENSE The case was settled before trial.

VERDICT A $12 million Illinois settlement was reached: $11 million from the hospital and $1 million from the ObGyn.

Bowel perforation, sepsis after ovary removal

DURING LEFT OOPHORECTOMY, the ObGyn encountered adhesions. Five days later, the 41-year-old patient reported severe pain. A second procedure revealed sepsis and perforation of the large bowel. A colostomy was performed. The patient underwent additional corrective operations.

PATIENT’S CLAIM The ObGyn was negligent for causing tissue damage to the colon that perforated and escalated into sepsis. A surgeon should have been consulted when the ObGyn found the adhesions, so the bowel could be properly inspected before the abdomen was closed. The physician was also negligent for not recognizing symptoms of sepsis earlier.

PHYSICIAN’S DEFENSE Bowel injury is a known complication of oophorectomy. The patient appeared to be making a fairly good recovery until infection became evident; she was immediately treated.

VERDICT A $6.3 million New Jersey verdict was returned, including $300,000 for the husband’s loss of consortium.

Traumatic delivery causes seizures

 

 

DURING CESAREAN DELIVERY, the ObGyn rotated the baby from a transverse to a cephalic lie, and used a vacuum extractor to deliver the head through the hysterotomy incision.

When the child was 25 hours old, he suffered a seizure that lasted 6 minutes. Focal seizure activity involving the left side of his body and a skull fracture were identified. He was transferred to another hospital, where radiologic studies indicated a middle right cerebral artery infarct. The child developed an ongoing seizure disorder, speech and language delays, and mild, left-sided weakness.

PARENTS’ CLAIM The baby’s head was not properly delivered through the cesarean incision nor should the ObGyn have used vacuum extraction. The combination of the rotation and use of vacuum caused trauma to the infant’s head. In addition, the baby was placed in the well-baby nursery, which was inappropriate because he was born through thick meconium, resuscitated by a neonatal nurse, and had a depressed skull fracture.

PHYSICIAN’S DEFENSE Delivery was not traumatic; all treatment was appropriate.

VERDICT A $4.6 million New York settlement was reached with the hospital and ObGyn’s insurer.

Mother gets severe headache during birth

A 35-YEAR-OLD WOMAN began having a severe headache during delivery that continued after birth. She was discharged from the hospital and collapsed at home a day later. She was returned to the ED, where she was left in a hallway for 6 hours. She lost consciousness while in the hallway. Imaging and neurologic evaluation determined that she suffered a hypoxic brain injury from intracranial bleeding. She has slow response time, difficulty with all aspects of everyday life, and requires full-time attendant care.

PATIENT’S CLAIM Although she complained of a headache, no testing was done prior to her hospital discharge. Treatment was extremely delayed in the ED; an earlier diagnosis could have prevented brain damage.

PHYSICIAN’S DEFENSE Nothing could have prevented the brain damage.

VERDICT A $3.5 million California settlement was mediated.

Shoulder dystocia; brachial plexus injury

WHEN SHOULDER DYSTOCIA was encountered during delivery, the ObGyn applied gentle pressure to deliver the head. He was assisted by an ObGyn resident. The child was born with a brachial plexus injury, causing left-arm paralysis. She underwent surgery that increased her range of motion, but she will need years of physical therapy.

PATIENT’S CLAIM The ObGyn applied excessive traction and the resident improperly applied fundal pressure.

DEFENDANTS’ DEFENSE Only gentle traction was used. The resident did not apply fundal pressure.

VERDICT A New York jury found the ObGyn at fault and awarded the patient $3.5 million. The resident was vindicated.

Was premature baby viable?

AN EXPECTANT MOTHER MISCARRIED AT HOME at 6 months’ gestation, and an ambulance was called. After the EMTs helped the mother to the ambulance, they retrieved the fetus. When the baby was seen moving its head, the EMTs requested assistance from the advanced life support (ALS) team. ALS personnel visually assessed the fetus, determined it was nonviable, and placed the baby in a small container. The mother and baby arrived at the hospital 17 minutes after the ambulance was called.

At the hospital, a nurse noticed that the fetus was warm and had a heartbeat. The baby was taken to a special-care nursery for resuscitation and then transferred to another hospital’s NICU. The baby died after 46 days from severe brain damage due to lack of oxygen.

PARENTS’ CLAIM The EMTs and ALS team should have provided better evaluation and treatment for the infant; they were not trained to determine an infant’s viability. Placing the infant inside a plastic bag inside a box with a lid further deprived the baby of oxygen.

DEFENDANTS’ DEFENSE The case was settled before trial.

VERDICT A $1 million Massachusetts settlement was reached.

Were records altered because of a delayed diagnosis?

A WOMAN FOUND A LUMP in her left breast. A gynecologist ordered mammography. In January 2006, the radiologist requested ultrasonography (US), and reported that it conclusively indicated that the mass was a cyst. The gynecologist told the patient the tests were normal; further action was unnecessary. The patient saw the gynecologist four more times before being referred to a breast surgeon. In June 2006, she underwent surgical resection and chemotherapy for a malignant breast tumor.

PATIENT’S CLAIM The gynecologist was negligent for not referring the patient to a surgeon earlier. The gynecologist altered records: excerpts from the mammogram and US reports had been scanned in with a notation that the gynecologist had told the patient to follow up with a surgeon. When the gynecologist faxed the same reports to the surgeon, the annotations were absent. The gynecologist also changed the December 2005 chart, which referred to an US she never ordered.

 

 

PHYSICIAN’S DEFENSE The gynecologist stated that she regularly “merged” two reports into one document in her practice.

VERDICT A $700,000 Pennsylvania verdict was returned.

Excessive force or standard of care?

SHOULDER DYSTOCIA occurred during labor. The child sustained left brachial plexus palsy. At age 6, his left arm is paralyzed and smaller than the right arm. He has trouble performing normal daily tasks.

PATIENT’S CLAIM The ObGyn used excessive force by pulling on the baby’s head to complete the delivery. Standard of care required the ObGyn to take a more gentle approach to achieve delivery.

PHYSICIAN’S DEFENSE Delivery was performed appropriately, and did not deviate from standard of care.

VERDICT A $20.881 million Maryland verdict was returned, including $20 million for pain and suffering. The total award was reduced to $1,531,082 when the pain and suffering award was cut to $650,000 under the state’s statutory cap.

Preterm birth from an asymptomatic UTI?

A BABY WAS BORN AT 31 WEEKS’ gestation. The child has cerebral palsy, spastic quadriplegia, and requires assistance in all aspects of life.

PARENTS’ CLAIM Chorioamnionitis from a urinary tract infection (UTI) caused preterm birth. Urinalysis performed 7 weeks earlier indicated an infection, but the second-year resident caring for the mother failed to treat the UTI. The resident should have obtained a confirming urine culture, prescribed antibiotics, and monitored the mother more closely. The resident was poorly supervised.

DEFENDANTS’ DEFENSE Chorioamnionitis developed just before birth and could not be detected or prevented. A UTI cannot remain asymptomatic for 7 weeks and still cause premature birth. The mother was at increased risk of premature delivery because she had given birth to an anencephalic infant a year earlier. She began prenatal care in the middle of her pregnancy and ignored a referral to a high-risk maternal fetal specialist.

VERDICT A New York defense verdict was returned.

Infant’s brain damage blamed on delayed delivery

DURING DELIVERY, THE MOTHER’S PERINATOLOGIST recognized a severe shoulder dystocia. The perinatologist abandoned vaginal delivery and ordered an emergency cesarean delivery. The mother was transferred to an operating room (OR) with the baby’s head out between her legs. In the OR, the perinatologist pushed the baby’s head back into the uterus and performed a cesarean extraction. Nineteen minutes elapsed from when the vaginal delivery was abandoned and the baby was delivered.

The child was unresponsive at birth with no spontaneous movement or respiration. She was intubated and transferred to the NICU, where she was resuscitated. MRI confirmed that the child had hypoxic ischemia and severe, permanent brain damage from acute birth asphyxia. The child is blind, deaf, hypertensive, and has diffuse spasticity. She has a tracheostomy, a gastrostomy tube, and requires 24-hour care.

PARENTS’ CLAIM The perinatologist was negligent for abandoning vaginal delivery when delivery was progressing appropriately and there was no fetal distress. If the perinatologist had rotated the baby’s shoulder to the oblique position and/or used suprapubic pressure, the shoulder would have become disimpacted and the baby would have been safely delivered within seconds. Delay in delivery allowed for 19 minutes of umbilical cord compression, resulting in brain damage.

PHYSICIAN’S DEFENSE Cesarean delivery was appropriate; the baby did not suffer cord compression. Injury to the brain occurred days before delivery, based on prenatal ultrasonography.

VERDICT A $5.5 million California settlement was reached.

Failure to diagnose breast cancer: death

A 38-YEAR-OLD WOMAN went to her primary care physician (PCP) 3 years after giving birth. She reported breast pain, nipple discharge, and a dime-sized lump. The woman was still breastfeeding. An exam by the nurse practitioner (NP) was limited because the patient had breast implants. The NP suspected a galactocele and advised the patient to stop breastfeeding and apply ice packs. When the patient returned in 2 weeks, only the lump remained. The PCP determined that she had mastitis.

Five months later, she returned with additional lumps in both breasts, and was referred to a gynecologist. Ultrasonography (US) was ordered, but the patient never followed up. A year later, the patient was found to have metastatic breast cancer and died after 3 years of treatment.

ESTATE’S CLAIM The PCP and NP were negligent for not referring her for a breast biopsy when a lump was first detected.

DEFENDANTS’ DEFENSE Proper care was given. An earlier diagnosis would not have changed the outcome.

VERDICT A $750,000 Massachusetts settlement was reached.

What caused this child’s autism?

AFTER 33 HOURS OF LABOR, a baby was delivered vaginally by an ObGyn, nurse, and midwife. The child was diagnosed with autism several years later. His development is delayed, and he suffers cognitive impairment.

PARENTS’ CLAIM The child’s autism is due to a prolonged hypoxic event during labor. Fetal heart-rate monitoring demonstrated fetal distress, with a bradycardia. A cesarean delivery should have been performed.

PHYSICIAN’S DEFENSE The child has genetic autism unrelated to the birth process.

VERDICT A $1.35 million New York settlement was reached.

Was oxytocin the culprit?

DURING AN EXTENDED LABOR, the ObGyn continued to give the mother oxytocin, although there were signs of fetal distress. The child was born with brain damage, cannot walk, talk, or see, and requires 24-hour care.

PATIENT’S CLAIM The use of oxytocin was inappropriate given the signs of fetal distress. Oxytocin caused a lack of oxygen to the child, resulting in brain damage. A cesarean delivery should have been performed when fetal distress was identified.

DEFENDANTS’ DEFENSE The case was settled before trial.

VERDICT A $12 million Illinois settlement was reached: $11 million from the hospital and $1 million from the ObGyn.

Bowel perforation, sepsis after ovary removal

DURING LEFT OOPHORECTOMY, the ObGyn encountered adhesions. Five days later, the 41-year-old patient reported severe pain. A second procedure revealed sepsis and perforation of the large bowel. A colostomy was performed. The patient underwent additional corrective operations.

PATIENT’S CLAIM The ObGyn was negligent for causing tissue damage to the colon that perforated and escalated into sepsis. A surgeon should have been consulted when the ObGyn found the adhesions, so the bowel could be properly inspected before the abdomen was closed. The physician was also negligent for not recognizing symptoms of sepsis earlier.

PHYSICIAN’S DEFENSE Bowel injury is a known complication of oophorectomy. The patient appeared to be making a fairly good recovery until infection became evident; she was immediately treated.

VERDICT A $6.3 million New Jersey verdict was returned, including $300,000 for the husband’s loss of consortium.

Traumatic delivery causes seizures

 

 

DURING CESAREAN DELIVERY, the ObGyn rotated the baby from a transverse to a cephalic lie, and used a vacuum extractor to deliver the head through the hysterotomy incision.

When the child was 25 hours old, he suffered a seizure that lasted 6 minutes. Focal seizure activity involving the left side of his body and a skull fracture were identified. He was transferred to another hospital, where radiologic studies indicated a middle right cerebral artery infarct. The child developed an ongoing seizure disorder, speech and language delays, and mild, left-sided weakness.

PARENTS’ CLAIM The baby’s head was not properly delivered through the cesarean incision nor should the ObGyn have used vacuum extraction. The combination of the rotation and use of vacuum caused trauma to the infant’s head. In addition, the baby was placed in the well-baby nursery, which was inappropriate because he was born through thick meconium, resuscitated by a neonatal nurse, and had a depressed skull fracture.

PHYSICIAN’S DEFENSE Delivery was not traumatic; all treatment was appropriate.

VERDICT A $4.6 million New York settlement was reached with the hospital and ObGyn’s insurer.

Mother gets severe headache during birth

A 35-YEAR-OLD WOMAN began having a severe headache during delivery that continued after birth. She was discharged from the hospital and collapsed at home a day later. She was returned to the ED, where she was left in a hallway for 6 hours. She lost consciousness while in the hallway. Imaging and neurologic evaluation determined that she suffered a hypoxic brain injury from intracranial bleeding. She has slow response time, difficulty with all aspects of everyday life, and requires full-time attendant care.

PATIENT’S CLAIM Although she complained of a headache, no testing was done prior to her hospital discharge. Treatment was extremely delayed in the ED; an earlier diagnosis could have prevented brain damage.

PHYSICIAN’S DEFENSE Nothing could have prevented the brain damage.

VERDICT A $3.5 million California settlement was mediated.

Shoulder dystocia; brachial plexus injury

WHEN SHOULDER DYSTOCIA was encountered during delivery, the ObGyn applied gentle pressure to deliver the head. He was assisted by an ObGyn resident. The child was born with a brachial plexus injury, causing left-arm paralysis. She underwent surgery that increased her range of motion, but she will need years of physical therapy.

PATIENT’S CLAIM The ObGyn applied excessive traction and the resident improperly applied fundal pressure.

DEFENDANTS’ DEFENSE Only gentle traction was used. The resident did not apply fundal pressure.

VERDICT A New York jury found the ObGyn at fault and awarded the patient $3.5 million. The resident was vindicated.

Was premature baby viable?

AN EXPECTANT MOTHER MISCARRIED AT HOME at 6 months’ gestation, and an ambulance was called. After the EMTs helped the mother to the ambulance, they retrieved the fetus. When the baby was seen moving its head, the EMTs requested assistance from the advanced life support (ALS) team. ALS personnel visually assessed the fetus, determined it was nonviable, and placed the baby in a small container. The mother and baby arrived at the hospital 17 minutes after the ambulance was called.

At the hospital, a nurse noticed that the fetus was warm and had a heartbeat. The baby was taken to a special-care nursery for resuscitation and then transferred to another hospital’s NICU. The baby died after 46 days from severe brain damage due to lack of oxygen.

PARENTS’ CLAIM The EMTs and ALS team should have provided better evaluation and treatment for the infant; they were not trained to determine an infant’s viability. Placing the infant inside a plastic bag inside a box with a lid further deprived the baby of oxygen.

DEFENDANTS’ DEFENSE The case was settled before trial.

VERDICT A $1 million Massachusetts settlement was reached.

Were records altered because of a delayed diagnosis?

A WOMAN FOUND A LUMP in her left breast. A gynecologist ordered mammography. In January 2006, the radiologist requested ultrasonography (US), and reported that it conclusively indicated that the mass was a cyst. The gynecologist told the patient the tests were normal; further action was unnecessary. The patient saw the gynecologist four more times before being referred to a breast surgeon. In June 2006, she underwent surgical resection and chemotherapy for a malignant breast tumor.

PATIENT’S CLAIM The gynecologist was negligent for not referring the patient to a surgeon earlier. The gynecologist altered records: excerpts from the mammogram and US reports had been scanned in with a notation that the gynecologist had told the patient to follow up with a surgeon. When the gynecologist faxed the same reports to the surgeon, the annotations were absent. The gynecologist also changed the December 2005 chart, which referred to an US she never ordered.

 

 

PHYSICIAN’S DEFENSE The gynecologist stated that she regularly “merged” two reports into one document in her practice.

VERDICT A $700,000 Pennsylvania verdict was returned.

Excessive force or standard of care?

SHOULDER DYSTOCIA occurred during labor. The child sustained left brachial plexus palsy. At age 6, his left arm is paralyzed and smaller than the right arm. He has trouble performing normal daily tasks.

PATIENT’S CLAIM The ObGyn used excessive force by pulling on the baby’s head to complete the delivery. Standard of care required the ObGyn to take a more gentle approach to achieve delivery.

PHYSICIAN’S DEFENSE Delivery was performed appropriately, and did not deviate from standard of care.

VERDICT A $20.881 million Maryland verdict was returned, including $20 million for pain and suffering. The total award was reduced to $1,531,082 when the pain and suffering award was cut to $650,000 under the state’s statutory cap.

Preterm birth from an asymptomatic UTI?

A BABY WAS BORN AT 31 WEEKS’ gestation. The child has cerebral palsy, spastic quadriplegia, and requires assistance in all aspects of life.

PARENTS’ CLAIM Chorioamnionitis from a urinary tract infection (UTI) caused preterm birth. Urinalysis performed 7 weeks earlier indicated an infection, but the second-year resident caring for the mother failed to treat the UTI. The resident should have obtained a confirming urine culture, prescribed antibiotics, and monitored the mother more closely. The resident was poorly supervised.

DEFENDANTS’ DEFENSE Chorioamnionitis developed just before birth and could not be detected or prevented. A UTI cannot remain asymptomatic for 7 weeks and still cause premature birth. The mother was at increased risk of premature delivery because she had given birth to an anencephalic infant a year earlier. She began prenatal care in the middle of her pregnancy and ignored a referral to a high-risk maternal fetal specialist.

VERDICT A New York defense verdict was returned.

References

These cases were selected by the editors of OBG Management from Medical Malpractice Verdicts, Settlements & Experts, with permission of the editor, Lewis Laska (www.verdictslaska.com). The information available to the editors about the cases presented here is sometimes incomplete. Moreover, the cases may or may not have merit. Nevertheless, these cases represent the types of clinical situations that typically result in litigation and are meant to illustrate nationwide variation in jury verdicts and awards.

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

References

These cases were selected by the editors of OBG Management from Medical Malpractice Verdicts, Settlements & Experts, with permission of the editor, Lewis Laska (www.verdictslaska.com). The information available to the editors about the cases presented here is sometimes incomplete. Moreover, the cases may or may not have merit. Nevertheless, these cases represent the types of clinical situations that typically result in litigation and are meant to illustrate nationwide variation in jury verdicts and awards.

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

Issue
OBG Management - 24(11)
Issue
OBG Management - 24(11)
Page Number
46-49
Page Number
46-49
Publications
Publications
Topics
Article Type
Display Headline
Infant’s brain damage blamed on delayed delivery … and more
Display Headline
Infant’s brain damage blamed on delayed delivery … and more
Legacy Keywords
Medical malpractice;medical verdicts;negligence;defense;settlement;Lewis Laska;Medical Malpractice Verdicts Settlements and Experts;delayed delivery;brain damage;perinatologist;vaginal delivery;cesarean;NICU;breast cancer;galactocele;breast biopsy;autism;fetal distress;oxytocin;bowel perforation;sepsis;traumatic delivery;focal seizure;vacuum extraction;headache;hypoxic brain injury;shoulder dystocia;brachial plexus injury;miscarriage;ALS team;delayed diagnosis;chorioamnionitis;UTI
Legacy Keywords
Medical malpractice;medical verdicts;negligence;defense;settlement;Lewis Laska;Medical Malpractice Verdicts Settlements and Experts;delayed delivery;brain damage;perinatologist;vaginal delivery;cesarean;NICU;breast cancer;galactocele;breast biopsy;autism;fetal distress;oxytocin;bowel perforation;sepsis;traumatic delivery;focal seizure;vacuum extraction;headache;hypoxic brain injury;shoulder dystocia;brachial plexus injury;miscarriage;ALS team;delayed diagnosis;chorioamnionitis;UTI
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media

When and how to place an autologous rectus fascia pubovaginal sling

Article Type
Changed
Tue, 08/28/2018 - 11:00
Display Headline
When and how to place an autologous rectus fascia pubovaginal sling


Watch 2 intraoperative videos

These videos were selected by Mickey Karram, MD, and presented courtesy of
International Academy of Pelvic Surgery

Developed in Partnership with International Academy of Pelvic Surgery

CASE 1: Recurrent SUI and mesh erosion

A 50-year-old woman reports urinary incontinence that is associated with activity and exertion—stress urinary incontinence (SUI)—and says it has worsened over the past year. She mentions that she underwent vaginal hysterectomy, with placement of a tension-free vaginal tape (TVT), about 2 years earlier.

During physical examination, the patient becomes incontinent when abdominal pressure is increased, with some urethral mobility (cotton-swab deflection to 25° from the horizontal). She is also noted to have erosion of the TVT tape into the vaginal lumen.

Urodynamic testing reveals easily demonstrable SUI at a volume of 150 mL when she is in the sitting position, with a Valsalva leak-point pressure of 55 cm H2O. Her bladder remains stable to a capacity of 520 mL. Cystoscopy yields unremarkable findings.

When she is offered surgical correction of her SUI, the patient expresses a preference for the use of her own tissues and says she does not want to have synthetic mesh placed.

Is this patient a candidate for a rectus fascia pubovaginal sling?

As more patients express reservations about the placement of synthetic mesh during sling procedures, the use of autologous rectus fascia pubovaginal slings has risen. The concept of using a patient’s own tissue as a sling to support the urethra dates to the early 20th century, but it was not until late in that century that the procedure gained widespread appreciation and evolved into its current form. Initially, the procedure entailed mobilizing a strip of abdominal muscle (either rectus or pyramidalis), freeing one end of the strip from its attachment, passing that end under the bladder neck, and reaffixing it to the abdominal muscle wall, forming a “U”-shaped sling around the bladder outlet. Subsequently, overlying abdominal fascia was included in the sling, eventually replacing the muscle altogether. The final innovation: An isolated strip of fascia was suspended by free sutures that were tied to the abdominal wall or attached on top of the abdominal rectus sheath.

The autologous pubovaginal sling supports the proximal urethra and bladder neck to achieve continence by providing a direct compressive force on the urethra and bladder outlet, or by reestablishing a reinforcing platform or hammock against which the urethra is compressed during the transmission of increased abdominal pressure.

The sling is suspended on each end by free sutures that are attached directly to the abdominal wall musculature or, more commonly, tied to each other on the anterior surface of the abdominal wall.

Long-term success depends on healing and fibrotic processes, which occur primarily where the sling passes through the endopelvic fascia.

Who is a candidate?

Although the pubovaginal sling procedure was pioneered as a surgical option for intrinsic sphincter deficiency (ISD), its indications have broadened to encompass all types of SUI. Its reliable results and durable outcomes make it one of the main standards of treatment, and the pubovaginal sling has been used extensively as primary therapy for:

  • SUI related to ISD or urethral hypermobility
  • as a salvage procedure for recurrent SUI
  • as an adjunct to urethral and bladder reconstruction
  • as a way to functionally close the urethra to abandon urethral access to the bladder.

In our opinion, the autologous pubovaginal sling is appropriate for patients with SUI who decline to have synthetic material implanted because of concerns related to long-term placement of synthetic mesh. Other good candidates are women who experience recurrent incontinence after placement of a synthetic sling or who develop a complication, such as vaginal erosion (VIDEO 1, Rectus fascia pubovaginal sling after an unsuccessful TVT), after placement of a synthetic sling. We also prefer to use an autologous sling in patients who have been radiated or who have sustained urethral injuries, as well as in patients who are undergoing simultaneous repair of urethrovaginal fistula or diverticulum—or those who have already undergone such repair.

What is the optimal sling material?

Rectus abdominis fascia versus fascia lata. The two most commonly used autologous tissues are rectus abdominus fascia and fascia lata. Both of these materials have been studied extensively and proven to be effective and reliable. Most surgeons prefer rectus fascia because it is easier and quicker to harvest.

Allogenic and xenogenic tissues. Allogenic (cadaveric) fascia lata and cadaveric dermis provide reasonable efficacy, but durability remains an issue, as high failure rates have been reported. Bovine and porcine dermis, as well as porcine small-intestine submucosa, are also effective for SUI, although durability remains a concern.

 

 

Synthetic materials. Synthetic graft materials of various designs and substances also have been used as sling material. Monofilament, large-pore weave grafts (Type 1 mesh) are recommended for implantation in the vagina. Although good efficacy can be achieved with synthetic mesh, the material also may increase the risk of serious complications, such as infection, vaginal extrusion, and genitourinary erosion, and is not recommended for use beneath the proximal urethra or bladder neck.


The autologous pubovaginal sling supports the proximal urethra and bladder neck to achieve continence by providing a direct compressive force on the urethra and bladder outlet, or by reestablishing a reinforcing platform or hammock against which the urethra is compressed during increased abdominal pressure.

How to harvest rectus fascia and create a sling

1. Choose anesthesia and perioperative antibiotics

Pubovaginal sling procedures are generally carried out under general anesthesia, but spinal or epidural anesthesia also is possible. Full-patient paralysis is not warranted but may facilitate closure of the rectus fascia after fascial harvesting.

Perioperative antibiotics usually are given to ensure appropriate coverage against skin and vaginal flora (for example, a cephalosporin or fluoroquinolone). In fact, perioperative antibiotics have become a mandated quality of care measure in the United States.

2. Position the patient for optimal access

Place the patient in the low lithotomy position with her legs in stirrups. The abdomen and perineum should be sterilely prepared and draped to provide access to the vagina and lower abdomen.

After the bladder is drained with a Foley catheter, place a weighted vaginal speculum and use either lateral labial retraction sutures or a self-retaining retractor system to facilitate vaginal exposure.

3. Make an abdominal incision

Make an 8- to 10-cm Pfannenstiel incision approximately 3 to 5 cm above the pubic bone, carry the dissection down to the level of the rectus fascia using a combination of electrocautery and blunt dissection, and sweep the fat and subcutaneous tissue clear of the rectus tissue (FIGURE 1).



FIGURE 1 Skin incision

Before initiating the operation, delineate the location of the transverse skin incision, which should measure 8 to 10 cm and be situated about 4 cm above the symphysis pubis. A vertical incision is also feasible, although it usually is less aesthetic.

4. Harvest the fascia

The rectus abdominis fascia can be harvested in a transverse or vertical orientation. A fascial segment at least 8 cm in length and 1.5 to 2 cm in width is recommended.

Delineate the fascial segment to be resected using a surgical marking pen or electrocautery, then incise the tissue sharply with a scalpel, scissors, or electrocautery along the drawn lines.

Virgin fascia is preferred, but the presence of fibrotic rectus fascia does not prohibit its use. If you are resecting the fascia close and parallel to the symphysis pubis, leave at least 0.5 to 1.0 cm attached to facilitate closure of the defect created in the fascia. Small Army/Navy retractors permit aggressive retraction of skin edges, making it possible to use a smaller skin incision (FIGURE 2).



FIGURE 2 Resect the fascial strip

After choosing the optimal location for excision, mark the area using electrocautery or a surgical marking pen. Then resect the strip using a scalpel or electrocautery. The strip should measure 8 to 10 cm in length and 1 to 2 cm in width. If the skin incision is small, Army/Navy retractors may enhance exposure.

5. Close the fascial defect

Use heavy-gauge (#1 or #0) delayed, absorbable suture in a running fashion. It may be necessary to mobilize the rectus abdominis fascial edges to ensure appropriate tension-free approximation. It is important that anesthesia be sufficient to ensure muscular relaxation and paralysis during closure.

6. Prepare the fascial sling

Affix a single #1 permanent (for example, polypropylene or polyester) suture to each end of the fascial segment by passing the needle through the undersurface of the sling and then back through the top of the sling. If necessary, defat the sling (FIGURE 3).


FIGURE 3 Attach suspensory sutures
A. Mark the midline of the fascial sling with a pen and gently grasp it using a hemostat. B. Attach a polyester suture to each end of the fascial sling after stripping it of any adipose tissue. Ensure that the initial entry and exit points of the polyester sutures are on the same side of the strip that originally abutted the rectus muscles.

7. Dissect the vagina

Use injectable-grade saline or a local analgesic, such as 1% lidocaine, to hydrodissect the subepithelial tissues of the distal portion of the anterior vaginal wall. Make a midline or inverted “U” incision into the vagina (FIGURE 4).

 

 

Create vaginal flaps that have sufficient mobility to ensure tension-free closure over the sling. Carry out dissection laterally and anteriorly until you encounter the endopelvic fascia, then incise the endopelvic fascia and dissect it from the posterior surface of the pubis to enter the retropubic space.

Although blunt dissection sometimes can be performed, sharp dissection with Mayo scissors is often required, especially in cases that involve recurrent stress incontinence (FIGURE 4).


FIGURE 4 Dissect the vagina
A.
Use an inverted “U” or vertical incision on the vaginal mucosa overlying the midurethra and bladder. B. Carefully dissect the tissue to the pubic rami bilaterally until the urogenital diaphragm is identified, then sharply penetrate it using Mayo scissors. C. Enlarge the opening by repeating the procedure on the opposite side.

8. Pass retropubic needles

Pass Stamey needles or long clamps through the retropubic space from the open abdominal wound immediately posterior to the pubic bone, approximately 4 cm apart. You can maintain distal control of the needles by direct finger guidance through the vaginal incision. Be careful to advance the tip of the needle adjacent to the posterior surface of the pubic bone to avoid inadvertent bladder injury (FIGURE 5). Proper bladder drainage also helps to minimize injury to the bladder, which may be closely adherent to the pubis, especially if a prior retropubic procedure has been performed, as in Case 1.


FIGURE 5 Place the sling
A. Insert the Stamey needle through the rectus fascia and guide it into the vagina with the index finger placed against the tip of the needle. B. Thread both ends of the polyester suture into the eye of the Stamey needle and then retract the needle carefully until the suture ends are delivered abdominally at the level of the fascia.

9. Rule out bladder injury

Careful cystoscopic examination of the bladder is mandatory after passing the needles to rule out inadvertent injury. Injuries to the bladder typically occur at the 1 o’clock and 11 o’clock positions, so use a 70° lens, and fill the bladder completely to expand any mucosal redundancy. Wiggle the needles or clamps to help localize their position relative to the bladder wall.

10. Deploy the sling

Thread the free ends of the sutures affixed to the sling into the ends of the Stamey needles—or grasp them with clamps—and pull each suture up to the anterior abdominal wall through the retropubic space (FIGURE 5). Keep the sling centered and flat at the area of the bladder neck.

Some surgeons fix the sling in the midline to the underlying periurethral tissue using numerous delayed absorbable sutures. We prefer to leave the sling unattached to the underlying urethra and bladder neck.

11. Tension the sling

Various techniques are applicable. To ensure adequate “looseness,” we tie the sutures across the midline while holding a right-angle clamp between the sling material and the posterior urethral surface. The goal is for the sling to prevent the descent of the proximal urethra during increases in abdominal pressure without creating any outlet obstruction to the normal flow of urine (FIGURE 6).


FIGURE 6 Tension the sling
A. Tie the suspensory sutures abdominally above the fascial closure line. Tie the sutures across the assistant’s index finger to avoid excessive tension. B. Assess the tension using a right-angle clamp placed between the pubovaginal sling and the vagina.

12. Close the incisions

Close the abdominal skin incision using 3-0 and 4-0 absorbable sutures. Use 3-0 absorbable sutures to close the vaginal mucosa. We prefer to close the vagina after completion of the tensioning procedure, but some surgeons complete this step prior to tensioning.

13. Place a catheter, packing material

Place a bladder catheter and vaginal gauze packing. Both the catheter and gauze may be removed after 24 hours. If the patient is unable to void at that time, teach her intermittent self-catheterization, or place an indwelling Foley catheter for 1 week.

Outcomes show good efficacy

Pubovaginal slings are highly effective, with success rates between 50% and 75% after follow-up as long as 10 years.1 In 2011, Blaivas and Chaikin reported 4-year follow-up data, with improvement or cure in 100% of patients with uncomplicated SUI and in as many as 93% of patients in more complicated cases.2 Most failures were due to urge incontinence and occurred within the first 6 postoperative months; 3% of these urge patients were thought to have developed de novo urge incontinence.

Other studies have found de novo urgency and storage symptoms in as many as 23% of patients, with 11% of patients reporting voiding dysfunction and as many as 7.8% requiring long-term self-catheterization.1

 

 

Flawed methodology in the few randomized, controlled trials that have compared the pubovaginal sling with the tension-free vaginal tape (TVT) has cast doubt on their findings.3 Basok and colleagues found an increased rate of de novo urgency in the women treated with a pubovaginal sling, compared with those who underwent intravaginal slingplasty,4 whereas Sharifiaghdas and Mortazavi found equal efficacy between pubovaginal and retropubic midurethral synthetic slings.5 The most scientifically valid randomized, controlled trial found equal subjective cure rates and complication rates when a biologic pubovaginal sling was compared with the TVT.6 In that study, the pubovaginal sling was of porcine origin.

In a comparison of autologous and autograft slings, Flynn found equal control of SUI over 2 years, with reduced postoperative discomfort in the allograft group.7

When autologous pubovaginal slings were compared with Burch colposuspension in a randomized, controlled trial, fascial slings were better at controlling incontinence despite an increased morbidity profile.8

A meta-analysis found equal subjective cure rates and overall efficacy between pubovaginal and midurethral synthetic slings.9

Voiding dysfunction is the most common complication

Transient urinary retention may occur in as many as 20% of patients and requires intermittent self-catheterization until resolution (typically 2–4 weeks). Prolonged postoperative voiding dysfunction (lasting more than 4–6 weeks), including de novo urgency, urgency incontinence, and obstructive symptoms, may occur to some degree in as many as 25% of patients. However, fewer than 3% of women require subsequent urethrolysis for treatment of prolonged retention or obstructive voiding symptoms.

DID YOU READ THE GUEST EDITORIAL?

Synthetic full-length midurethral slings remain the standard of care
for SUI

Charles W. Nager, MD (November 2012)

A few technical suggestions

Harvest the fascia first. Because substantial bleeding can occur during vaginal dissection, it is advisable to harvest the autologous fascia and prepare the sling by affixing sutures to it before dissecting the vagina. This facilitates timely insertion of the sling and minimal blood loss. Retropubic bleeding from high in the space that occurs during dissection almost always resolves upon placement of the sling. We recommend against prolonged attempts at hemostasis.

In urethral reconstruction, tension the sling after reconstruction. When placing an autologous pubovaginal sling in the setting of urethral reconstruction or as tissue interposition, harvest the fascia and prepare and deploy the sling (with passage of the retropubic sutures) before reconstructing the urethra—but refrain from tensioning until after the reconstruction is completed. Then affix the sling in the appropriate location and tension it. When the sling is placed after reconstruction, it can damage the reconstruction through traction or direct injury.

Don’t worry about surface orientation. During placement of the autologous sling material, surface orientation does not matter. Conventionally, however, the “body-side” or underside of the graft is placed on the body-side of the patient.

Tensioning varies between patients. For most women, sling tensioning can be accomplished by tying the sutures over one or two fingers placed across the fascia. In patients who have undergone multiple procedures and who have a nonmobile urethra, however, tension should be tighter and must be individualized, based on the patient’s anatomy, lower urinary tract function, and willingness to perform intermittent self-catheterization for a prolonged period of time.

CASE 1: Resolved

After you advise the patient of the risks and benefits of the rectus fascia pubovaginal sling, in comparison with a repeat synthetic midurethral sling, she continues to insist on the use of autologous tissue. She undergoes the pubovaginal sling operation with excision of eroded mesh without complication.

CASE 2: Loss of a cadaveric sling

A 35-year-old woman reports continuous urinary leakage that is not associated with movement. She was previously told that she had an ectopic ureter implanted into a congenitally short urethra, and she underwent repair of the problem, including reimplantation of the ureter and placement of a cadaveric fascia lata sling. A congenital remnant—observed as a blind pouch via cystoscopy—was left attached to the urethra. Two years have passed since that operation.

Physical findings: A pelvic examination reveals complete loss of the posterior urethra. One possible explanation: The remnant became infected and caused a breakdown of the posterior urethra, with complete disappearance of the cadaveric fascia lata.

Recommended management: Complete urethral reconstruction, with transposition of a martius fat pad and repeat placement of a cadaveric fascia pubovaginal sling.

Technique: See Video 2, Urethral reconstruction.

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

Article PDF
Author and Disclosure Information


Mickey Karram, MD
Dr. Karram is Director of the Fellowship Program in Female Pelvic Medicine and Reconstructive Pelvic Surgery, University of Cincinnati/The Christ Hospital, Cincinnati, Ohio; Co-Editor in Chief of the International Academy of Pelvic Surgery (IAPS); and Course Director of the Pelvic Anatomy and Gynecologic Surgery Symposium (PAGS) and the Female Urology and Urogynecology Symposium (FUUS), both co-sponsored by OBG Management.


Dani Zoorob, MD
Dr. Zoorob is a Fellow in Urogynecology at the University of Cincinnati/The Christ Hospital in Cincinnati, Ohio.

The authors report no financial relationships relevant to this article.

Issue
OBG Management - 24(11)
Publications
Topics
Page Number
24-33
Legacy Keywords
autologous rectus fascia pubovaginal sling;Mickey Karram MD;Dani Zoorob MD;surgical techniques;rectus fascia;synthetic midurethral sling;stress urinary incontience;International Academy of Pelvic Surgery;IAPS;urethral reconstruction;unsuccessful TVT;SUI;synthetic mesh;intrinsic sphincter deficiency;ISD;bladder reconstruction;allogenic;xenogenic;fascia lata;Burch colposuspension;midurethral sling;transient urinary retention;voiding dysfunction;nonmibile urethra;self-catheterization
Sections
Author and Disclosure Information


Mickey Karram, MD
Dr. Karram is Director of the Fellowship Program in Female Pelvic Medicine and Reconstructive Pelvic Surgery, University of Cincinnati/The Christ Hospital, Cincinnati, Ohio; Co-Editor in Chief of the International Academy of Pelvic Surgery (IAPS); and Course Director of the Pelvic Anatomy and Gynecologic Surgery Symposium (PAGS) and the Female Urology and Urogynecology Symposium (FUUS), both co-sponsored by OBG Management.


Dani Zoorob, MD
Dr. Zoorob is a Fellow in Urogynecology at the University of Cincinnati/The Christ Hospital in Cincinnati, Ohio.

The authors report no financial relationships relevant to this article.

Author and Disclosure Information


Mickey Karram, MD
Dr. Karram is Director of the Fellowship Program in Female Pelvic Medicine and Reconstructive Pelvic Surgery, University of Cincinnati/The Christ Hospital, Cincinnati, Ohio; Co-Editor in Chief of the International Academy of Pelvic Surgery (IAPS); and Course Director of the Pelvic Anatomy and Gynecologic Surgery Symposium (PAGS) and the Female Urology and Urogynecology Symposium (FUUS), both co-sponsored by OBG Management.


Dani Zoorob, MD
Dr. Zoorob is a Fellow in Urogynecology at the University of Cincinnati/The Christ Hospital in Cincinnati, Ohio.

The authors report no financial relationships relevant to this article.

Article PDF
Article PDF


Watch 2 intraoperative videos

These videos were selected by Mickey Karram, MD, and presented courtesy of
International Academy of Pelvic Surgery

Developed in Partnership with International Academy of Pelvic Surgery

CASE 1: Recurrent SUI and mesh erosion

A 50-year-old woman reports urinary incontinence that is associated with activity and exertion—stress urinary incontinence (SUI)—and says it has worsened over the past year. She mentions that she underwent vaginal hysterectomy, with placement of a tension-free vaginal tape (TVT), about 2 years earlier.

During physical examination, the patient becomes incontinent when abdominal pressure is increased, with some urethral mobility (cotton-swab deflection to 25° from the horizontal). She is also noted to have erosion of the TVT tape into the vaginal lumen.

Urodynamic testing reveals easily demonstrable SUI at a volume of 150 mL when she is in the sitting position, with a Valsalva leak-point pressure of 55 cm H2O. Her bladder remains stable to a capacity of 520 mL. Cystoscopy yields unremarkable findings.

When she is offered surgical correction of her SUI, the patient expresses a preference for the use of her own tissues and says she does not want to have synthetic mesh placed.

Is this patient a candidate for a rectus fascia pubovaginal sling?

As more patients express reservations about the placement of synthetic mesh during sling procedures, the use of autologous rectus fascia pubovaginal slings has risen. The concept of using a patient’s own tissue as a sling to support the urethra dates to the early 20th century, but it was not until late in that century that the procedure gained widespread appreciation and evolved into its current form. Initially, the procedure entailed mobilizing a strip of abdominal muscle (either rectus or pyramidalis), freeing one end of the strip from its attachment, passing that end under the bladder neck, and reaffixing it to the abdominal muscle wall, forming a “U”-shaped sling around the bladder outlet. Subsequently, overlying abdominal fascia was included in the sling, eventually replacing the muscle altogether. The final innovation: An isolated strip of fascia was suspended by free sutures that were tied to the abdominal wall or attached on top of the abdominal rectus sheath.

The autologous pubovaginal sling supports the proximal urethra and bladder neck to achieve continence by providing a direct compressive force on the urethra and bladder outlet, or by reestablishing a reinforcing platform or hammock against which the urethra is compressed during the transmission of increased abdominal pressure.

The sling is suspended on each end by free sutures that are attached directly to the abdominal wall musculature or, more commonly, tied to each other on the anterior surface of the abdominal wall.

Long-term success depends on healing and fibrotic processes, which occur primarily where the sling passes through the endopelvic fascia.

Who is a candidate?

Although the pubovaginal sling procedure was pioneered as a surgical option for intrinsic sphincter deficiency (ISD), its indications have broadened to encompass all types of SUI. Its reliable results and durable outcomes make it one of the main standards of treatment, and the pubovaginal sling has been used extensively as primary therapy for:

  • SUI related to ISD or urethral hypermobility
  • as a salvage procedure for recurrent SUI
  • as an adjunct to urethral and bladder reconstruction
  • as a way to functionally close the urethra to abandon urethral access to the bladder.

In our opinion, the autologous pubovaginal sling is appropriate for patients with SUI who decline to have synthetic material implanted because of concerns related to long-term placement of synthetic mesh. Other good candidates are women who experience recurrent incontinence after placement of a synthetic sling or who develop a complication, such as vaginal erosion (VIDEO 1, Rectus fascia pubovaginal sling after an unsuccessful TVT), after placement of a synthetic sling. We also prefer to use an autologous sling in patients who have been radiated or who have sustained urethral injuries, as well as in patients who are undergoing simultaneous repair of urethrovaginal fistula or diverticulum—or those who have already undergone such repair.

What is the optimal sling material?

Rectus abdominis fascia versus fascia lata. The two most commonly used autologous tissues are rectus abdominus fascia and fascia lata. Both of these materials have been studied extensively and proven to be effective and reliable. Most surgeons prefer rectus fascia because it is easier and quicker to harvest.

Allogenic and xenogenic tissues. Allogenic (cadaveric) fascia lata and cadaveric dermis provide reasonable efficacy, but durability remains an issue, as high failure rates have been reported. Bovine and porcine dermis, as well as porcine small-intestine submucosa, are also effective for SUI, although durability remains a concern.

 

 

Synthetic materials. Synthetic graft materials of various designs and substances also have been used as sling material. Monofilament, large-pore weave grafts (Type 1 mesh) are recommended for implantation in the vagina. Although good efficacy can be achieved with synthetic mesh, the material also may increase the risk of serious complications, such as infection, vaginal extrusion, and genitourinary erosion, and is not recommended for use beneath the proximal urethra or bladder neck.


The autologous pubovaginal sling supports the proximal urethra and bladder neck to achieve continence by providing a direct compressive force on the urethra and bladder outlet, or by reestablishing a reinforcing platform or hammock against which the urethra is compressed during increased abdominal pressure.

How to harvest rectus fascia and create a sling

1. Choose anesthesia and perioperative antibiotics

Pubovaginal sling procedures are generally carried out under general anesthesia, but spinal or epidural anesthesia also is possible. Full-patient paralysis is not warranted but may facilitate closure of the rectus fascia after fascial harvesting.

Perioperative antibiotics usually are given to ensure appropriate coverage against skin and vaginal flora (for example, a cephalosporin or fluoroquinolone). In fact, perioperative antibiotics have become a mandated quality of care measure in the United States.

2. Position the patient for optimal access

Place the patient in the low lithotomy position with her legs in stirrups. The abdomen and perineum should be sterilely prepared and draped to provide access to the vagina and lower abdomen.

After the bladder is drained with a Foley catheter, place a weighted vaginal speculum and use either lateral labial retraction sutures or a self-retaining retractor system to facilitate vaginal exposure.

3. Make an abdominal incision

Make an 8- to 10-cm Pfannenstiel incision approximately 3 to 5 cm above the pubic bone, carry the dissection down to the level of the rectus fascia using a combination of electrocautery and blunt dissection, and sweep the fat and subcutaneous tissue clear of the rectus tissue (FIGURE 1).



FIGURE 1 Skin incision

Before initiating the operation, delineate the location of the transverse skin incision, which should measure 8 to 10 cm and be situated about 4 cm above the symphysis pubis. A vertical incision is also feasible, although it usually is less aesthetic.

4. Harvest the fascia

The rectus abdominis fascia can be harvested in a transverse or vertical orientation. A fascial segment at least 8 cm in length and 1.5 to 2 cm in width is recommended.

Delineate the fascial segment to be resected using a surgical marking pen or electrocautery, then incise the tissue sharply with a scalpel, scissors, or electrocautery along the drawn lines.

Virgin fascia is preferred, but the presence of fibrotic rectus fascia does not prohibit its use. If you are resecting the fascia close and parallel to the symphysis pubis, leave at least 0.5 to 1.0 cm attached to facilitate closure of the defect created in the fascia. Small Army/Navy retractors permit aggressive retraction of skin edges, making it possible to use a smaller skin incision (FIGURE 2).



FIGURE 2 Resect the fascial strip

After choosing the optimal location for excision, mark the area using electrocautery or a surgical marking pen. Then resect the strip using a scalpel or electrocautery. The strip should measure 8 to 10 cm in length and 1 to 2 cm in width. If the skin incision is small, Army/Navy retractors may enhance exposure.

5. Close the fascial defect

Use heavy-gauge (#1 or #0) delayed, absorbable suture in a running fashion. It may be necessary to mobilize the rectus abdominis fascial edges to ensure appropriate tension-free approximation. It is important that anesthesia be sufficient to ensure muscular relaxation and paralysis during closure.

6. Prepare the fascial sling

Affix a single #1 permanent (for example, polypropylene or polyester) suture to each end of the fascial segment by passing the needle through the undersurface of the sling and then back through the top of the sling. If necessary, defat the sling (FIGURE 3).


FIGURE 3 Attach suspensory sutures
A. Mark the midline of the fascial sling with a pen and gently grasp it using a hemostat. B. Attach a polyester suture to each end of the fascial sling after stripping it of any adipose tissue. Ensure that the initial entry and exit points of the polyester sutures are on the same side of the strip that originally abutted the rectus muscles.

7. Dissect the vagina

Use injectable-grade saline or a local analgesic, such as 1% lidocaine, to hydrodissect the subepithelial tissues of the distal portion of the anterior vaginal wall. Make a midline or inverted “U” incision into the vagina (FIGURE 4).

 

 

Create vaginal flaps that have sufficient mobility to ensure tension-free closure over the sling. Carry out dissection laterally and anteriorly until you encounter the endopelvic fascia, then incise the endopelvic fascia and dissect it from the posterior surface of the pubis to enter the retropubic space.

Although blunt dissection sometimes can be performed, sharp dissection with Mayo scissors is often required, especially in cases that involve recurrent stress incontinence (FIGURE 4).


FIGURE 4 Dissect the vagina
A.
Use an inverted “U” or vertical incision on the vaginal mucosa overlying the midurethra and bladder. B. Carefully dissect the tissue to the pubic rami bilaterally until the urogenital diaphragm is identified, then sharply penetrate it using Mayo scissors. C. Enlarge the opening by repeating the procedure on the opposite side.

8. Pass retropubic needles

Pass Stamey needles or long clamps through the retropubic space from the open abdominal wound immediately posterior to the pubic bone, approximately 4 cm apart. You can maintain distal control of the needles by direct finger guidance through the vaginal incision. Be careful to advance the tip of the needle adjacent to the posterior surface of the pubic bone to avoid inadvertent bladder injury (FIGURE 5). Proper bladder drainage also helps to minimize injury to the bladder, which may be closely adherent to the pubis, especially if a prior retropubic procedure has been performed, as in Case 1.


FIGURE 5 Place the sling
A. Insert the Stamey needle through the rectus fascia and guide it into the vagina with the index finger placed against the tip of the needle. B. Thread both ends of the polyester suture into the eye of the Stamey needle and then retract the needle carefully until the suture ends are delivered abdominally at the level of the fascia.

9. Rule out bladder injury

Careful cystoscopic examination of the bladder is mandatory after passing the needles to rule out inadvertent injury. Injuries to the bladder typically occur at the 1 o’clock and 11 o’clock positions, so use a 70° lens, and fill the bladder completely to expand any mucosal redundancy. Wiggle the needles or clamps to help localize their position relative to the bladder wall.

10. Deploy the sling

Thread the free ends of the sutures affixed to the sling into the ends of the Stamey needles—or grasp them with clamps—and pull each suture up to the anterior abdominal wall through the retropubic space (FIGURE 5). Keep the sling centered and flat at the area of the bladder neck.

Some surgeons fix the sling in the midline to the underlying periurethral tissue using numerous delayed absorbable sutures. We prefer to leave the sling unattached to the underlying urethra and bladder neck.

11. Tension the sling

Various techniques are applicable. To ensure adequate “looseness,” we tie the sutures across the midline while holding a right-angle clamp between the sling material and the posterior urethral surface. The goal is for the sling to prevent the descent of the proximal urethra during increases in abdominal pressure without creating any outlet obstruction to the normal flow of urine (FIGURE 6).


FIGURE 6 Tension the sling
A. Tie the suspensory sutures abdominally above the fascial closure line. Tie the sutures across the assistant’s index finger to avoid excessive tension. B. Assess the tension using a right-angle clamp placed between the pubovaginal sling and the vagina.

12. Close the incisions

Close the abdominal skin incision using 3-0 and 4-0 absorbable sutures. Use 3-0 absorbable sutures to close the vaginal mucosa. We prefer to close the vagina after completion of the tensioning procedure, but some surgeons complete this step prior to tensioning.

13. Place a catheter, packing material

Place a bladder catheter and vaginal gauze packing. Both the catheter and gauze may be removed after 24 hours. If the patient is unable to void at that time, teach her intermittent self-catheterization, or place an indwelling Foley catheter for 1 week.

Outcomes show good efficacy

Pubovaginal slings are highly effective, with success rates between 50% and 75% after follow-up as long as 10 years.1 In 2011, Blaivas and Chaikin reported 4-year follow-up data, with improvement or cure in 100% of patients with uncomplicated SUI and in as many as 93% of patients in more complicated cases.2 Most failures were due to urge incontinence and occurred within the first 6 postoperative months; 3% of these urge patients were thought to have developed de novo urge incontinence.

Other studies have found de novo urgency and storage symptoms in as many as 23% of patients, with 11% of patients reporting voiding dysfunction and as many as 7.8% requiring long-term self-catheterization.1

 

 

Flawed methodology in the few randomized, controlled trials that have compared the pubovaginal sling with the tension-free vaginal tape (TVT) has cast doubt on their findings.3 Basok and colleagues found an increased rate of de novo urgency in the women treated with a pubovaginal sling, compared with those who underwent intravaginal slingplasty,4 whereas Sharifiaghdas and Mortazavi found equal efficacy between pubovaginal and retropubic midurethral synthetic slings.5 The most scientifically valid randomized, controlled trial found equal subjective cure rates and complication rates when a biologic pubovaginal sling was compared with the TVT.6 In that study, the pubovaginal sling was of porcine origin.

In a comparison of autologous and autograft slings, Flynn found equal control of SUI over 2 years, with reduced postoperative discomfort in the allograft group.7

When autologous pubovaginal slings were compared with Burch colposuspension in a randomized, controlled trial, fascial slings were better at controlling incontinence despite an increased morbidity profile.8

A meta-analysis found equal subjective cure rates and overall efficacy between pubovaginal and midurethral synthetic slings.9

Voiding dysfunction is the most common complication

Transient urinary retention may occur in as many as 20% of patients and requires intermittent self-catheterization until resolution (typically 2–4 weeks). Prolonged postoperative voiding dysfunction (lasting more than 4–6 weeks), including de novo urgency, urgency incontinence, and obstructive symptoms, may occur to some degree in as many as 25% of patients. However, fewer than 3% of women require subsequent urethrolysis for treatment of prolonged retention or obstructive voiding symptoms.

DID YOU READ THE GUEST EDITORIAL?

Synthetic full-length midurethral slings remain the standard of care
for SUI

Charles W. Nager, MD (November 2012)

A few technical suggestions

Harvest the fascia first. Because substantial bleeding can occur during vaginal dissection, it is advisable to harvest the autologous fascia and prepare the sling by affixing sutures to it before dissecting the vagina. This facilitates timely insertion of the sling and minimal blood loss. Retropubic bleeding from high in the space that occurs during dissection almost always resolves upon placement of the sling. We recommend against prolonged attempts at hemostasis.

In urethral reconstruction, tension the sling after reconstruction. When placing an autologous pubovaginal sling in the setting of urethral reconstruction or as tissue interposition, harvest the fascia and prepare and deploy the sling (with passage of the retropubic sutures) before reconstructing the urethra—but refrain from tensioning until after the reconstruction is completed. Then affix the sling in the appropriate location and tension it. When the sling is placed after reconstruction, it can damage the reconstruction through traction or direct injury.

Don’t worry about surface orientation. During placement of the autologous sling material, surface orientation does not matter. Conventionally, however, the “body-side” or underside of the graft is placed on the body-side of the patient.

Tensioning varies between patients. For most women, sling tensioning can be accomplished by tying the sutures over one or two fingers placed across the fascia. In patients who have undergone multiple procedures and who have a nonmobile urethra, however, tension should be tighter and must be individualized, based on the patient’s anatomy, lower urinary tract function, and willingness to perform intermittent self-catheterization for a prolonged period of time.

CASE 1: Resolved

After you advise the patient of the risks and benefits of the rectus fascia pubovaginal sling, in comparison with a repeat synthetic midurethral sling, she continues to insist on the use of autologous tissue. She undergoes the pubovaginal sling operation with excision of eroded mesh without complication.

CASE 2: Loss of a cadaveric sling

A 35-year-old woman reports continuous urinary leakage that is not associated with movement. She was previously told that she had an ectopic ureter implanted into a congenitally short urethra, and she underwent repair of the problem, including reimplantation of the ureter and placement of a cadaveric fascia lata sling. A congenital remnant—observed as a blind pouch via cystoscopy—was left attached to the urethra. Two years have passed since that operation.

Physical findings: A pelvic examination reveals complete loss of the posterior urethra. One possible explanation: The remnant became infected and caused a breakdown of the posterior urethra, with complete disappearance of the cadaveric fascia lata.

Recommended management: Complete urethral reconstruction, with transposition of a martius fat pad and repeat placement of a cadaveric fascia pubovaginal sling.

Technique: See Video 2, Urethral reconstruction.

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


Watch 2 intraoperative videos

These videos were selected by Mickey Karram, MD, and presented courtesy of
International Academy of Pelvic Surgery

Developed in Partnership with International Academy of Pelvic Surgery

CASE 1: Recurrent SUI and mesh erosion

A 50-year-old woman reports urinary incontinence that is associated with activity and exertion—stress urinary incontinence (SUI)—and says it has worsened over the past year. She mentions that she underwent vaginal hysterectomy, with placement of a tension-free vaginal tape (TVT), about 2 years earlier.

During physical examination, the patient becomes incontinent when abdominal pressure is increased, with some urethral mobility (cotton-swab deflection to 25° from the horizontal). She is also noted to have erosion of the TVT tape into the vaginal lumen.

Urodynamic testing reveals easily demonstrable SUI at a volume of 150 mL when she is in the sitting position, with a Valsalva leak-point pressure of 55 cm H2O. Her bladder remains stable to a capacity of 520 mL. Cystoscopy yields unremarkable findings.

When she is offered surgical correction of her SUI, the patient expresses a preference for the use of her own tissues and says she does not want to have synthetic mesh placed.

Is this patient a candidate for a rectus fascia pubovaginal sling?

As more patients express reservations about the placement of synthetic mesh during sling procedures, the use of autologous rectus fascia pubovaginal slings has risen. The concept of using a patient’s own tissue as a sling to support the urethra dates to the early 20th century, but it was not until late in that century that the procedure gained widespread appreciation and evolved into its current form. Initially, the procedure entailed mobilizing a strip of abdominal muscle (either rectus or pyramidalis), freeing one end of the strip from its attachment, passing that end under the bladder neck, and reaffixing it to the abdominal muscle wall, forming a “U”-shaped sling around the bladder outlet. Subsequently, overlying abdominal fascia was included in the sling, eventually replacing the muscle altogether. The final innovation: An isolated strip of fascia was suspended by free sutures that were tied to the abdominal wall or attached on top of the abdominal rectus sheath.

The autologous pubovaginal sling supports the proximal urethra and bladder neck to achieve continence by providing a direct compressive force on the urethra and bladder outlet, or by reestablishing a reinforcing platform or hammock against which the urethra is compressed during the transmission of increased abdominal pressure.

The sling is suspended on each end by free sutures that are attached directly to the abdominal wall musculature or, more commonly, tied to each other on the anterior surface of the abdominal wall.

Long-term success depends on healing and fibrotic processes, which occur primarily where the sling passes through the endopelvic fascia.

Who is a candidate?

Although the pubovaginal sling procedure was pioneered as a surgical option for intrinsic sphincter deficiency (ISD), its indications have broadened to encompass all types of SUI. Its reliable results and durable outcomes make it one of the main standards of treatment, and the pubovaginal sling has been used extensively as primary therapy for:

  • SUI related to ISD or urethral hypermobility
  • as a salvage procedure for recurrent SUI
  • as an adjunct to urethral and bladder reconstruction
  • as a way to functionally close the urethra to abandon urethral access to the bladder.

In our opinion, the autologous pubovaginal sling is appropriate for patients with SUI who decline to have synthetic material implanted because of concerns related to long-term placement of synthetic mesh. Other good candidates are women who experience recurrent incontinence after placement of a synthetic sling or who develop a complication, such as vaginal erosion (VIDEO 1, Rectus fascia pubovaginal sling after an unsuccessful TVT), after placement of a synthetic sling. We also prefer to use an autologous sling in patients who have been radiated or who have sustained urethral injuries, as well as in patients who are undergoing simultaneous repair of urethrovaginal fistula or diverticulum—or those who have already undergone such repair.

What is the optimal sling material?

Rectus abdominis fascia versus fascia lata. The two most commonly used autologous tissues are rectus abdominus fascia and fascia lata. Both of these materials have been studied extensively and proven to be effective and reliable. Most surgeons prefer rectus fascia because it is easier and quicker to harvest.

Allogenic and xenogenic tissues. Allogenic (cadaveric) fascia lata and cadaveric dermis provide reasonable efficacy, but durability remains an issue, as high failure rates have been reported. Bovine and porcine dermis, as well as porcine small-intestine submucosa, are also effective for SUI, although durability remains a concern.

 

 

Synthetic materials. Synthetic graft materials of various designs and substances also have been used as sling material. Monofilament, large-pore weave grafts (Type 1 mesh) are recommended for implantation in the vagina. Although good efficacy can be achieved with synthetic mesh, the material also may increase the risk of serious complications, such as infection, vaginal extrusion, and genitourinary erosion, and is not recommended for use beneath the proximal urethra or bladder neck.


The autologous pubovaginal sling supports the proximal urethra and bladder neck to achieve continence by providing a direct compressive force on the urethra and bladder outlet, or by reestablishing a reinforcing platform or hammock against which the urethra is compressed during increased abdominal pressure.

How to harvest rectus fascia and create a sling

1. Choose anesthesia and perioperative antibiotics

Pubovaginal sling procedures are generally carried out under general anesthesia, but spinal or epidural anesthesia also is possible. Full-patient paralysis is not warranted but may facilitate closure of the rectus fascia after fascial harvesting.

Perioperative antibiotics usually are given to ensure appropriate coverage against skin and vaginal flora (for example, a cephalosporin or fluoroquinolone). In fact, perioperative antibiotics have become a mandated quality of care measure in the United States.

2. Position the patient for optimal access

Place the patient in the low lithotomy position with her legs in stirrups. The abdomen and perineum should be sterilely prepared and draped to provide access to the vagina and lower abdomen.

After the bladder is drained with a Foley catheter, place a weighted vaginal speculum and use either lateral labial retraction sutures or a self-retaining retractor system to facilitate vaginal exposure.

3. Make an abdominal incision

Make an 8- to 10-cm Pfannenstiel incision approximately 3 to 5 cm above the pubic bone, carry the dissection down to the level of the rectus fascia using a combination of electrocautery and blunt dissection, and sweep the fat and subcutaneous tissue clear of the rectus tissue (FIGURE 1).



FIGURE 1 Skin incision

Before initiating the operation, delineate the location of the transverse skin incision, which should measure 8 to 10 cm and be situated about 4 cm above the symphysis pubis. A vertical incision is also feasible, although it usually is less aesthetic.

4. Harvest the fascia

The rectus abdominis fascia can be harvested in a transverse or vertical orientation. A fascial segment at least 8 cm in length and 1.5 to 2 cm in width is recommended.

Delineate the fascial segment to be resected using a surgical marking pen or electrocautery, then incise the tissue sharply with a scalpel, scissors, or electrocautery along the drawn lines.

Virgin fascia is preferred, but the presence of fibrotic rectus fascia does not prohibit its use. If you are resecting the fascia close and parallel to the symphysis pubis, leave at least 0.5 to 1.0 cm attached to facilitate closure of the defect created in the fascia. Small Army/Navy retractors permit aggressive retraction of skin edges, making it possible to use a smaller skin incision (FIGURE 2).



FIGURE 2 Resect the fascial strip

After choosing the optimal location for excision, mark the area using electrocautery or a surgical marking pen. Then resect the strip using a scalpel or electrocautery. The strip should measure 8 to 10 cm in length and 1 to 2 cm in width. If the skin incision is small, Army/Navy retractors may enhance exposure.

5. Close the fascial defect

Use heavy-gauge (#1 or #0) delayed, absorbable suture in a running fashion. It may be necessary to mobilize the rectus abdominis fascial edges to ensure appropriate tension-free approximation. It is important that anesthesia be sufficient to ensure muscular relaxation and paralysis during closure.

6. Prepare the fascial sling

Affix a single #1 permanent (for example, polypropylene or polyester) suture to each end of the fascial segment by passing the needle through the undersurface of the sling and then back through the top of the sling. If necessary, defat the sling (FIGURE 3).


FIGURE 3 Attach suspensory sutures
A. Mark the midline of the fascial sling with a pen and gently grasp it using a hemostat. B. Attach a polyester suture to each end of the fascial sling after stripping it of any adipose tissue. Ensure that the initial entry and exit points of the polyester sutures are on the same side of the strip that originally abutted the rectus muscles.

7. Dissect the vagina

Use injectable-grade saline or a local analgesic, such as 1% lidocaine, to hydrodissect the subepithelial tissues of the distal portion of the anterior vaginal wall. Make a midline or inverted “U” incision into the vagina (FIGURE 4).

 

 

Create vaginal flaps that have sufficient mobility to ensure tension-free closure over the sling. Carry out dissection laterally and anteriorly until you encounter the endopelvic fascia, then incise the endopelvic fascia and dissect it from the posterior surface of the pubis to enter the retropubic space.

Although blunt dissection sometimes can be performed, sharp dissection with Mayo scissors is often required, especially in cases that involve recurrent stress incontinence (FIGURE 4).


FIGURE 4 Dissect the vagina
A.
Use an inverted “U” or vertical incision on the vaginal mucosa overlying the midurethra and bladder. B. Carefully dissect the tissue to the pubic rami bilaterally until the urogenital diaphragm is identified, then sharply penetrate it using Mayo scissors. C. Enlarge the opening by repeating the procedure on the opposite side.

8. Pass retropubic needles

Pass Stamey needles or long clamps through the retropubic space from the open abdominal wound immediately posterior to the pubic bone, approximately 4 cm apart. You can maintain distal control of the needles by direct finger guidance through the vaginal incision. Be careful to advance the tip of the needle adjacent to the posterior surface of the pubic bone to avoid inadvertent bladder injury (FIGURE 5). Proper bladder drainage also helps to minimize injury to the bladder, which may be closely adherent to the pubis, especially if a prior retropubic procedure has been performed, as in Case 1.


FIGURE 5 Place the sling
A. Insert the Stamey needle through the rectus fascia and guide it into the vagina with the index finger placed against the tip of the needle. B. Thread both ends of the polyester suture into the eye of the Stamey needle and then retract the needle carefully until the suture ends are delivered abdominally at the level of the fascia.

9. Rule out bladder injury

Careful cystoscopic examination of the bladder is mandatory after passing the needles to rule out inadvertent injury. Injuries to the bladder typically occur at the 1 o’clock and 11 o’clock positions, so use a 70° lens, and fill the bladder completely to expand any mucosal redundancy. Wiggle the needles or clamps to help localize their position relative to the bladder wall.

10. Deploy the sling

Thread the free ends of the sutures affixed to the sling into the ends of the Stamey needles—or grasp them with clamps—and pull each suture up to the anterior abdominal wall through the retropubic space (FIGURE 5). Keep the sling centered and flat at the area of the bladder neck.

Some surgeons fix the sling in the midline to the underlying periurethral tissue using numerous delayed absorbable sutures. We prefer to leave the sling unattached to the underlying urethra and bladder neck.

11. Tension the sling

Various techniques are applicable. To ensure adequate “looseness,” we tie the sutures across the midline while holding a right-angle clamp between the sling material and the posterior urethral surface. The goal is for the sling to prevent the descent of the proximal urethra during increases in abdominal pressure without creating any outlet obstruction to the normal flow of urine (FIGURE 6).


FIGURE 6 Tension the sling
A. Tie the suspensory sutures abdominally above the fascial closure line. Tie the sutures across the assistant’s index finger to avoid excessive tension. B. Assess the tension using a right-angle clamp placed between the pubovaginal sling and the vagina.

12. Close the incisions

Close the abdominal skin incision using 3-0 and 4-0 absorbable sutures. Use 3-0 absorbable sutures to close the vaginal mucosa. We prefer to close the vagina after completion of the tensioning procedure, but some surgeons complete this step prior to tensioning.

13. Place a catheter, packing material

Place a bladder catheter and vaginal gauze packing. Both the catheter and gauze may be removed after 24 hours. If the patient is unable to void at that time, teach her intermittent self-catheterization, or place an indwelling Foley catheter for 1 week.

Outcomes show good efficacy

Pubovaginal slings are highly effective, with success rates between 50% and 75% after follow-up as long as 10 years.1 In 2011, Blaivas and Chaikin reported 4-year follow-up data, with improvement or cure in 100% of patients with uncomplicated SUI and in as many as 93% of patients in more complicated cases.2 Most failures were due to urge incontinence and occurred within the first 6 postoperative months; 3% of these urge patients were thought to have developed de novo urge incontinence.

Other studies have found de novo urgency and storage symptoms in as many as 23% of patients, with 11% of patients reporting voiding dysfunction and as many as 7.8% requiring long-term self-catheterization.1

 

 

Flawed methodology in the few randomized, controlled trials that have compared the pubovaginal sling with the tension-free vaginal tape (TVT) has cast doubt on their findings.3 Basok and colleagues found an increased rate of de novo urgency in the women treated with a pubovaginal sling, compared with those who underwent intravaginal slingplasty,4 whereas Sharifiaghdas and Mortazavi found equal efficacy between pubovaginal and retropubic midurethral synthetic slings.5 The most scientifically valid randomized, controlled trial found equal subjective cure rates and complication rates when a biologic pubovaginal sling was compared with the TVT.6 In that study, the pubovaginal sling was of porcine origin.

In a comparison of autologous and autograft slings, Flynn found equal control of SUI over 2 years, with reduced postoperative discomfort in the allograft group.7

When autologous pubovaginal slings were compared with Burch colposuspension in a randomized, controlled trial, fascial slings were better at controlling incontinence despite an increased morbidity profile.8

A meta-analysis found equal subjective cure rates and overall efficacy between pubovaginal and midurethral synthetic slings.9

Voiding dysfunction is the most common complication

Transient urinary retention may occur in as many as 20% of patients and requires intermittent self-catheterization until resolution (typically 2–4 weeks). Prolonged postoperative voiding dysfunction (lasting more than 4–6 weeks), including de novo urgency, urgency incontinence, and obstructive symptoms, may occur to some degree in as many as 25% of patients. However, fewer than 3% of women require subsequent urethrolysis for treatment of prolonged retention or obstructive voiding symptoms.

DID YOU READ THE GUEST EDITORIAL?

Synthetic full-length midurethral slings remain the standard of care
for SUI

Charles W. Nager, MD (November 2012)

A few technical suggestions

Harvest the fascia first. Because substantial bleeding can occur during vaginal dissection, it is advisable to harvest the autologous fascia and prepare the sling by affixing sutures to it before dissecting the vagina. This facilitates timely insertion of the sling and minimal blood loss. Retropubic bleeding from high in the space that occurs during dissection almost always resolves upon placement of the sling. We recommend against prolonged attempts at hemostasis.

In urethral reconstruction, tension the sling after reconstruction. When placing an autologous pubovaginal sling in the setting of urethral reconstruction or as tissue interposition, harvest the fascia and prepare and deploy the sling (with passage of the retropubic sutures) before reconstructing the urethra—but refrain from tensioning until after the reconstruction is completed. Then affix the sling in the appropriate location and tension it. When the sling is placed after reconstruction, it can damage the reconstruction through traction or direct injury.

Don’t worry about surface orientation. During placement of the autologous sling material, surface orientation does not matter. Conventionally, however, the “body-side” or underside of the graft is placed on the body-side of the patient.

Tensioning varies between patients. For most women, sling tensioning can be accomplished by tying the sutures over one or two fingers placed across the fascia. In patients who have undergone multiple procedures and who have a nonmobile urethra, however, tension should be tighter and must be individualized, based on the patient’s anatomy, lower urinary tract function, and willingness to perform intermittent self-catheterization for a prolonged period of time.

CASE 1: Resolved

After you advise the patient of the risks and benefits of the rectus fascia pubovaginal sling, in comparison with a repeat synthetic midurethral sling, she continues to insist on the use of autologous tissue. She undergoes the pubovaginal sling operation with excision of eroded mesh without complication.

CASE 2: Loss of a cadaveric sling

A 35-year-old woman reports continuous urinary leakage that is not associated with movement. She was previously told that she had an ectopic ureter implanted into a congenitally short urethra, and she underwent repair of the problem, including reimplantation of the ureter and placement of a cadaveric fascia lata sling. A congenital remnant—observed as a blind pouch via cystoscopy—was left attached to the urethra. Two years have passed since that operation.

Physical findings: A pelvic examination reveals complete loss of the posterior urethra. One possible explanation: The remnant became infected and caused a breakdown of the posterior urethra, with complete disappearance of the cadaveric fascia lata.

Recommended management: Complete urethral reconstruction, with transposition of a martius fat pad and repeat placement of a cadaveric fascia pubovaginal sling.

Technique: See Video 2, Urethral reconstruction.

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

Issue
OBG Management - 24(11)
Issue
OBG Management - 24(11)
Page Number
24-33
Page Number
24-33
Publications
Publications
Topics
Article Type
Display Headline
When and how to place an autologous rectus fascia pubovaginal sling
Display Headline
When and how to place an autologous rectus fascia pubovaginal sling
Legacy Keywords
autologous rectus fascia pubovaginal sling;Mickey Karram MD;Dani Zoorob MD;surgical techniques;rectus fascia;synthetic midurethral sling;stress urinary incontience;International Academy of Pelvic Surgery;IAPS;urethral reconstruction;unsuccessful TVT;SUI;synthetic mesh;intrinsic sphincter deficiency;ISD;bladder reconstruction;allogenic;xenogenic;fascia lata;Burch colposuspension;midurethral sling;transient urinary retention;voiding dysfunction;nonmibile urethra;self-catheterization
Legacy Keywords
autologous rectus fascia pubovaginal sling;Mickey Karram MD;Dani Zoorob MD;surgical techniques;rectus fascia;synthetic midurethral sling;stress urinary incontience;International Academy of Pelvic Surgery;IAPS;urethral reconstruction;unsuccessful TVT;SUI;synthetic mesh;intrinsic sphincter deficiency;ISD;bladder reconstruction;allogenic;xenogenic;fascia lata;Burch colposuspension;midurethral sling;transient urinary retention;voiding dysfunction;nonmibile urethra;self-catheterization
Sections
Article Source

PURLs Copyright

Inside the Article
Article PDF Media

Vitamin D deficiency in older adults

Article Type
Changed
Tue, 12/11/2018 - 15:19
Display Headline
Vitamin D deficiency in older adults

Low vitamin D levels can impact cognitive functioning in older adults.1 As vitamin D levels decrease, cognitive impairment increases.

Vitamin D deficiency can occur because few foods contain this nutrient2 and patients have limited exposure to sunlight—vitamin D is produced when sunlight strikes the skin.2 In addition to rickets, low levels of vitamin D have been linked to slower information processing in middle age and older men, cognitive decline, mood disorders, and altered brain development and function resulting in neurodegenerative diseases and other medical disorders.3

One study suggested that one-half of adults age >60 do not get sufficient vitamin D, with an even higher rate among women with Alzheimer’s disease.4 Patients in dementia units typically are not tested for vitamin D levels. These patients rarely leave the unit, which leaves them deprived of the vitamin D provided by sunlight. Even patients exposed to sunlight may receive minimal vitamin D because they use sunscreen.

The following protocol can help patients who may benefit from vitamin D supplementation and increased sun exposure.

Obtain and assess vitamin D levels. Evaluate your patient’s level in the context of physical or cognitive symptoms and other lab values:

  • deficient: <12 ng/mL
  • inadequate: 12 to 20 ng/mL
  • adequate: ≥20 ng/mL.2

Order dietary assessment to identify foods that may increase vitamin D levels. The best sources are fish—salmon, tuna, and mackerel—fish oils, beef, liver, cheese, and egg yolks.2 Several food products, including milk and orange juice, are fortified with vitamin D.

Suggest a daily vitamin D supplement ranging from 400 IU/d to 1,000 IU/d. The Institute of Medicine suggests 600 IU/d for patients age 60 to 70 and 800 IU/d for those age ≥71. For vitamin D deficient patients, recommend >1,000 IU/d.1

Recommend 15 minutes per day in the sun without sunscreen from spring to autumn; late summer to fall is ideal because vitamin D’s half-life is 30 days. Midday is the best time to produce vitamin D.5

Recheck the patient’s Mini-Mental State Examination score every 4 months. Vitamin D supplementation is correlated with cognitive functioning.6

Disclosure

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

References

1. Mayo Clinic. Vitamin D. http://www.mayoclinic.com/health/vitamin-d/NS_patient-vitamind/DSECTION=dosing. Updated October 1 2011. Accessed September 26, 2012.

2. National Institutes of Health. Office of Dietary Supplements. Dietary supplement fact sheet: vitamin D. http://ods.od.nih.gov/factsheets/VitaminD-HealthProfessional. Accessed September 26, 2012.

3. Lee DM, Tajar A, Ulubaev A, et al. Association between 25-hydroxyvitamin D levels and cognitive performance in middle-aged and older European men. J Neurol Neurosurg Psychiatry. 2009;80(7):722-729.

4. Wilkins CH, Sheline YI, Roe CM, et al. Vitamin D deficiency is associated with low mood and worse cognitive performance in older adults. Am J Geriatr Psychiatry. 2006;14(12):1032-1040.

5. Webb AR, Engelsen O. Calculated ultraviolet exposure levels for a healthy vitamin D status. Photochem Photobiol. 2006;82(6):1697-1703.

6. Przybelski RJ, Binkley NC. Is vitamin D important for preserving cognition? A positive correlation of serum 25-hydroxyvitamin D concentration with cognitive function. Arch Biochem Biophys. 2007;460(2):202-205.

Article PDF
Author and Disclosure Information

Michael C. LaFerney, PhD, RN, PMHCNS-BC
Psychiatric, Clinical Nurse Specialist, Arbour, SeniorCare, Haverhill, MA

Issue
Current Psychiatry - 11(11)
Publications
Topics
Page Number
63-63
Legacy Keywords
vitamin D; vitamin D deficiency; older adults; cognitive decline; rickets
Sections
Author and Disclosure Information

Michael C. LaFerney, PhD, RN, PMHCNS-BC
Psychiatric, Clinical Nurse Specialist, Arbour, SeniorCare, Haverhill, MA

Author and Disclosure Information

Michael C. LaFerney, PhD, RN, PMHCNS-BC
Psychiatric, Clinical Nurse Specialist, Arbour, SeniorCare, Haverhill, MA

Article PDF
Article PDF

Low vitamin D levels can impact cognitive functioning in older adults.1 As vitamin D levels decrease, cognitive impairment increases.

Vitamin D deficiency can occur because few foods contain this nutrient2 and patients have limited exposure to sunlight—vitamin D is produced when sunlight strikes the skin.2 In addition to rickets, low levels of vitamin D have been linked to slower information processing in middle age and older men, cognitive decline, mood disorders, and altered brain development and function resulting in neurodegenerative diseases and other medical disorders.3

One study suggested that one-half of adults age >60 do not get sufficient vitamin D, with an even higher rate among women with Alzheimer’s disease.4 Patients in dementia units typically are not tested for vitamin D levels. These patients rarely leave the unit, which leaves them deprived of the vitamin D provided by sunlight. Even patients exposed to sunlight may receive minimal vitamin D because they use sunscreen.

The following protocol can help patients who may benefit from vitamin D supplementation and increased sun exposure.

Obtain and assess vitamin D levels. Evaluate your patient’s level in the context of physical or cognitive symptoms and other lab values:

  • deficient: <12 ng/mL
  • inadequate: 12 to 20 ng/mL
  • adequate: ≥20 ng/mL.2

Order dietary assessment to identify foods that may increase vitamin D levels. The best sources are fish—salmon, tuna, and mackerel—fish oils, beef, liver, cheese, and egg yolks.2 Several food products, including milk and orange juice, are fortified with vitamin D.

Suggest a daily vitamin D supplement ranging from 400 IU/d to 1,000 IU/d. The Institute of Medicine suggests 600 IU/d for patients age 60 to 70 and 800 IU/d for those age ≥71. For vitamin D deficient patients, recommend >1,000 IU/d.1

Recommend 15 minutes per day in the sun without sunscreen from spring to autumn; late summer to fall is ideal because vitamin D’s half-life is 30 days. Midday is the best time to produce vitamin D.5

Recheck the patient’s Mini-Mental State Examination score every 4 months. Vitamin D supplementation is correlated with cognitive functioning.6

Disclosure

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

Low vitamin D levels can impact cognitive functioning in older adults.1 As vitamin D levels decrease, cognitive impairment increases.

Vitamin D deficiency can occur because few foods contain this nutrient2 and patients have limited exposure to sunlight—vitamin D is produced when sunlight strikes the skin.2 In addition to rickets, low levels of vitamin D have been linked to slower information processing in middle age and older men, cognitive decline, mood disorders, and altered brain development and function resulting in neurodegenerative diseases and other medical disorders.3

One study suggested that one-half of adults age >60 do not get sufficient vitamin D, with an even higher rate among women with Alzheimer’s disease.4 Patients in dementia units typically are not tested for vitamin D levels. These patients rarely leave the unit, which leaves them deprived of the vitamin D provided by sunlight. Even patients exposed to sunlight may receive minimal vitamin D because they use sunscreen.

The following protocol can help patients who may benefit from vitamin D supplementation and increased sun exposure.

Obtain and assess vitamin D levels. Evaluate your patient’s level in the context of physical or cognitive symptoms and other lab values:

  • deficient: <12 ng/mL
  • inadequate: 12 to 20 ng/mL
  • adequate: ≥20 ng/mL.2

Order dietary assessment to identify foods that may increase vitamin D levels. The best sources are fish—salmon, tuna, and mackerel—fish oils, beef, liver, cheese, and egg yolks.2 Several food products, including milk and orange juice, are fortified with vitamin D.

Suggest a daily vitamin D supplement ranging from 400 IU/d to 1,000 IU/d. The Institute of Medicine suggests 600 IU/d for patients age 60 to 70 and 800 IU/d for those age ≥71. For vitamin D deficient patients, recommend >1,000 IU/d.1

Recommend 15 minutes per day in the sun without sunscreen from spring to autumn; late summer to fall is ideal because vitamin D’s half-life is 30 days. Midday is the best time to produce vitamin D.5

Recheck the patient’s Mini-Mental State Examination score every 4 months. Vitamin D supplementation is correlated with cognitive functioning.6

Disclosure

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

References

1. Mayo Clinic. Vitamin D. http://www.mayoclinic.com/health/vitamin-d/NS_patient-vitamind/DSECTION=dosing. Updated October 1 2011. Accessed September 26, 2012.

2. National Institutes of Health. Office of Dietary Supplements. Dietary supplement fact sheet: vitamin D. http://ods.od.nih.gov/factsheets/VitaminD-HealthProfessional. Accessed September 26, 2012.

3. Lee DM, Tajar A, Ulubaev A, et al. Association between 25-hydroxyvitamin D levels and cognitive performance in middle-aged and older European men. J Neurol Neurosurg Psychiatry. 2009;80(7):722-729.

4. Wilkins CH, Sheline YI, Roe CM, et al. Vitamin D deficiency is associated with low mood and worse cognitive performance in older adults. Am J Geriatr Psychiatry. 2006;14(12):1032-1040.

5. Webb AR, Engelsen O. Calculated ultraviolet exposure levels for a healthy vitamin D status. Photochem Photobiol. 2006;82(6):1697-1703.

6. Przybelski RJ, Binkley NC. Is vitamin D important for preserving cognition? A positive correlation of serum 25-hydroxyvitamin D concentration with cognitive function. Arch Biochem Biophys. 2007;460(2):202-205.

References

1. Mayo Clinic. Vitamin D. http://www.mayoclinic.com/health/vitamin-d/NS_patient-vitamind/DSECTION=dosing. Updated October 1 2011. Accessed September 26, 2012.

2. National Institutes of Health. Office of Dietary Supplements. Dietary supplement fact sheet: vitamin D. http://ods.od.nih.gov/factsheets/VitaminD-HealthProfessional. Accessed September 26, 2012.

3. Lee DM, Tajar A, Ulubaev A, et al. Association between 25-hydroxyvitamin D levels and cognitive performance in middle-aged and older European men. J Neurol Neurosurg Psychiatry. 2009;80(7):722-729.

4. Wilkins CH, Sheline YI, Roe CM, et al. Vitamin D deficiency is associated with low mood and worse cognitive performance in older adults. Am J Geriatr Psychiatry. 2006;14(12):1032-1040.

5. Webb AR, Engelsen O. Calculated ultraviolet exposure levels for a healthy vitamin D status. Photochem Photobiol. 2006;82(6):1697-1703.

6. Przybelski RJ, Binkley NC. Is vitamin D important for preserving cognition? A positive correlation of serum 25-hydroxyvitamin D concentration with cognitive function. Arch Biochem Biophys. 2007;460(2):202-205.

Issue
Current Psychiatry - 11(11)
Issue
Current Psychiatry - 11(11)
Page Number
63-63
Page Number
63-63
Publications
Publications
Topics
Article Type
Display Headline
Vitamin D deficiency in older adults
Display Headline
Vitamin D deficiency in older adults
Legacy Keywords
vitamin D; vitamin D deficiency; older adults; cognitive decline; rickets
Legacy Keywords
vitamin D; vitamin D deficiency; older adults; cognitive decline; rickets
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media

An open-label trial of escitalopram for PPD: Considerations for research

Article Type
Changed
Tue, 12/11/2018 - 15:03
Display Headline
An open-label trial of escitalopram for PPD: Considerations for research

Challenges in recruiting women to postpartum depression (PPD) antidepressant treatment trials, which we encountered when conducting a trial of escitalopram, contribute to the limited body of knowledge about PPD treatment. Here we discuss results from a preliminary trial of escitalopram for PPD, and challenges of research in this area.

Escitalopram, the S-enantiomer of citalopram, is a selective serotonin reuptake inhibitor with high selectivity and potency that is FDA-approved for treating major depressive disorder (MDD) and generalized anxiety disorder. An agent with antidepressant and anxiolytic effects is particularly desirable for PPD because anxiety is more common in postpartum major depressive episodes than non-postpartum MDD.1 Anxiety and depressive disorders commonly are comorbid in postpartum women.2

We conducted an open-label trial of escitalopram for women with PPD and anxiety. We initially attempted to recruit 20 women.

Methods

Patients received 8 weeks of treatment with escitalopram, 10 to 20 mg/d (flexible dose). After completing the initial phone screen, patients had 5 follow-up visits, once every 2 weeks for 8 weeks. The institutional review board at Massachusetts General Hospital approved this study and we obtained written informed consent from all patients at the first visit. Twelve patients completed the phone screen and 7 eligible patients were enrolled in the study over 32 months. Reasons for ineligibility included having a history of psychosis, onset of symptoms >3 months postpartum, or presenting >6 months after onset. Others declined to participate because of concern about the time commitment or because they pursued nonpharmacologic treatments after the evaluation visit. One patient was lost to follow-up. Three patients completed the study. The study was halted because of the slow pace of recruitment.

Patient selection. Patients were screened for a major depressive episode with postpartum onset within 3 months of childbirth; depressive symptoms may have developed during pregnancy and worsened postpartum to meet criteria for MDD. Women were eligible for the study if they:

  • were age 18 to 45
  • experienced a major depressive episode with symptoms developing within 3 months of childbirth
  • presented within 6 months of childbirth
  • had a Montgomery-Åsberg Depression Rating Scale (MADRS) score >15
  • had a Beck Anxiety Inventory (BAI) score >10.

Patients who were pregnant or breast-feeding were excluded from the study per an agreement with the sponsor. In addition, women were excluded if they had taken any psychotropic medication within 2 weeks of enrollment; had active suicidal ideation, homicidal ideation, or presence of psychotic symptoms; had chronic depression or dysthymia; had chronic or treatment-resistant anxiety disorders; had a history of mania or hypomania; or had active alcohol or substance abuse within the past year.

Treatment. Patients received escitalopram, 10 mg/d, after the baseline visit. At the investigator’s discretion, the dose could be increased to 20 mg/d or lowered to 5 mg/d if side effects occurred.

Measures. At the first visit, patients were assessed with the Mini-International Neuropsychiatric Interview to verify MDD and exclude diagnoses that would determine ineligibility. MADRS and Edinburgh Postnatal Depression Scale (EPDS) were used at each visit to measure depressive symptoms.3,4 The BAI was completed at each visit to measure anxiety symptoms. Obsessions and compulsions were measured with the Yale-Brown Obsessive Compulsive Scale (Y-BOCS)5 at baseline, and at all following visits if the patient scored >8 at baseline. The Clinical Global Impression Scales for severity and improvement were completed at each visit.6

Results

Of 7 patients enrolled, 3 completed the study, 2 were ineligible after the baseline visit, and 2 did not participate after the baseline visit (1 selected to pursue psychotherapy, and 1 was lost to follow-up).

Two of 3 patients responded to escitalopram (≥50% decrease on MADRS), and both were remitters (MADRS score <7). All 3 patients were responders on EPDS and BAI. One patient had Y-BOCS >8 at baseline (Total Y-BOCS score of 9, and final Y-BOCS score of 8) (Table).

Table

Symptom rating scale scores at baseline and study end

 Baseline (Visit 1)Final (Visit 5)
PatientMADRSBAIEPDSMADRSBAIEPDS
Ms. A2118221200
Ms. B282819452
Ms. C37619620
BAI: Beck Anxiety Inventory; EPDS: Edinburgh Postnatal Depression Scale; MADRS: Montgomery-Åsberg Depression Rating Scale

Discussion

Patients who stayed in treatment improved during the course of this study. Recruitment was difficult; we were able to recruit only 7 patients out of a projected 20 for the screening visit. We solicited feedback from local obstetrics health care providers and social workers on recruitment and attractiveness of the study as part of our routine collaboration with obstetrical services that screen for PPD. Primary reasons patients were not referred were that they were breast-feeding or they stated they would prefer to receive treatment from their primary care doctor. Recruitment difficulty in this study was in stark contrast to other recent studies completed at our center. For example, we have successfully recruited for menopausal depression and premenstrual dysphoric disorder treatment studies, and have completed large naturalistic studies of women with unipolar depression and bipolar disorder across pregnancy and postpartum. We suspect that many patients who were eligible for the study preferred to seek care from an obstetrician or primary care doctor with whom they already had a therapeutic alliance, and we also suspect that many women with PPD do not seek treatment at all, which is consistent with findings from other research groups.

 

 

Lessons learned from PPD research include:

  • Including women who are breast-feeding is important because many women choose to breast-feed and suffer from PPD. Because antidepressant use during breast-feeding has been closely studied, it is appropriate to include potential research participants who are breast-feeding as long as they receive adequate information and are able to provide informed consent.
  • Participants in PPD studies may require accommodations that take into account their role as a new mother, such as on-site childcare, home visits, or other strategies.
  • Because of recruitment challenges in postpartum patients, multisite trials may be required to include adequate numbers of participants.

Related Resource

  • Freeman MP, Joffe H, Cohen LS. Postpartum depression: Help patients find the right treatment. Current Psychiatry. 2012;11(11):14-21.

Drug Brand Names

  • Citalopram • Celexa
  • Escitalopram • Lexapro

Disclosures

Dr. Freeman has received grant or research support from Eli Lilly and Company, Forest Laboratories, and GlaxoSmithKline, is on the advisory boards of Otsuka and Takeda/Lundbeck, and is a consultant for PamLab LLC.

Dr. Joffe has received grant or research support from Cephalon/Teva, and is a consultant to Noven and Sunovion.

Dr. Cohen has received research support from AstraZeneca, Bayer HealthCare Pharmaceuticals, Bristol-Myers Squibb, Forest Laboratories, GlaxoSmithKline, National Institute of Mental Health, National Institute on Aging, National Institutes of Health, Ortho-McNeil Janssen, and Pfizer and has served on an advisory board for PamLab LLC.

This study was funded as an investigator-initiated trial by Forest Pharmaceuticals.

References

1. Bernstein IH, Rush AJ, Yonkers K, et al. Symptom features of postpartum depression: are they distinct? Depress Anxiety. 2008;25(1):20-26.

2. Wenzel A, Haugen EN, Jackson LC, et al. Anxiety symptoms and disorders at eight weeks postpartum. J Anxiety Disord. 2005;19(3):295-311.

3. Cox JL, Holden JM, Sagovsky R. Detection of postnatal depression. Development of the 10-item Edinburgh Postnatal Depression Scale. Br J Psychiatry. 1987;150:782-786.

4. Montgomery SA, Åsberg M. A new depression scale designed to be sensitive to change. Br J Psychiatry. 1979;134:382-389.

5. Goodman WK, Price LH, Rasmussen SA, et al. The Yale-Brown Obsessive Compulsive Scale. I. Development, use, and reliability. Arch Gen Psychiatry. 1989;46(11):1006-1011.

6. Guy W. ECDEU assessment manual for psychopharmacology. Rockville MD: US Department of Health and Human Services; 1976. Department of Health, Education, and Welfare Publication (ADM) 76–338.

Article PDF
Author and Disclosure Information

Marlene P. Freeman, MD
Associate Professor of Psychiatry, Harvard Medical School, Director of Clinical Services, Perinatal and Reproductive Psychiatry Program, Massachusetts General Hospital, Boston, MA
Hadine Joffe, MD, MSc
Associate Professor, Harvard Medical School, Director of Research, Center for Women’s Mental Health, Massachusetts General Hospital, Boston, MA
Lee S. Cohen, MD
Carpenter Chair in Psychiatry and Women’s Mental Health, Harvard Medical School, Director, Center for Women’s Mental Health, Massachusetts General Hospital, Boston, MA

Issue
Current Psychiatry - 11(11)
Publications
Topics
Page Number
E3-E5
Legacy Keywords
escitalopram; postpartum depression; pregnancy; depression
Sections
Author and Disclosure Information

Marlene P. Freeman, MD
Associate Professor of Psychiatry, Harvard Medical School, Director of Clinical Services, Perinatal and Reproductive Psychiatry Program, Massachusetts General Hospital, Boston, MA
Hadine Joffe, MD, MSc
Associate Professor, Harvard Medical School, Director of Research, Center for Women’s Mental Health, Massachusetts General Hospital, Boston, MA
Lee S. Cohen, MD
Carpenter Chair in Psychiatry and Women’s Mental Health, Harvard Medical School, Director, Center for Women’s Mental Health, Massachusetts General Hospital, Boston, MA

Author and Disclosure Information

Marlene P. Freeman, MD
Associate Professor of Psychiatry, Harvard Medical School, Director of Clinical Services, Perinatal and Reproductive Psychiatry Program, Massachusetts General Hospital, Boston, MA
Hadine Joffe, MD, MSc
Associate Professor, Harvard Medical School, Director of Research, Center for Women’s Mental Health, Massachusetts General Hospital, Boston, MA
Lee S. Cohen, MD
Carpenter Chair in Psychiatry and Women’s Mental Health, Harvard Medical School, Director, Center for Women’s Mental Health, Massachusetts General Hospital, Boston, MA

Article PDF
Article PDF

Challenges in recruiting women to postpartum depression (PPD) antidepressant treatment trials, which we encountered when conducting a trial of escitalopram, contribute to the limited body of knowledge about PPD treatment. Here we discuss results from a preliminary trial of escitalopram for PPD, and challenges of research in this area.

Escitalopram, the S-enantiomer of citalopram, is a selective serotonin reuptake inhibitor with high selectivity and potency that is FDA-approved for treating major depressive disorder (MDD) and generalized anxiety disorder. An agent with antidepressant and anxiolytic effects is particularly desirable for PPD because anxiety is more common in postpartum major depressive episodes than non-postpartum MDD.1 Anxiety and depressive disorders commonly are comorbid in postpartum women.2

We conducted an open-label trial of escitalopram for women with PPD and anxiety. We initially attempted to recruit 20 women.

Methods

Patients received 8 weeks of treatment with escitalopram, 10 to 20 mg/d (flexible dose). After completing the initial phone screen, patients had 5 follow-up visits, once every 2 weeks for 8 weeks. The institutional review board at Massachusetts General Hospital approved this study and we obtained written informed consent from all patients at the first visit. Twelve patients completed the phone screen and 7 eligible patients were enrolled in the study over 32 months. Reasons for ineligibility included having a history of psychosis, onset of symptoms >3 months postpartum, or presenting >6 months after onset. Others declined to participate because of concern about the time commitment or because they pursued nonpharmacologic treatments after the evaluation visit. One patient was lost to follow-up. Three patients completed the study. The study was halted because of the slow pace of recruitment.

Patient selection. Patients were screened for a major depressive episode with postpartum onset within 3 months of childbirth; depressive symptoms may have developed during pregnancy and worsened postpartum to meet criteria for MDD. Women were eligible for the study if they:

  • were age 18 to 45
  • experienced a major depressive episode with symptoms developing within 3 months of childbirth
  • presented within 6 months of childbirth
  • had a Montgomery-Åsberg Depression Rating Scale (MADRS) score >15
  • had a Beck Anxiety Inventory (BAI) score >10.

Patients who were pregnant or breast-feeding were excluded from the study per an agreement with the sponsor. In addition, women were excluded if they had taken any psychotropic medication within 2 weeks of enrollment; had active suicidal ideation, homicidal ideation, or presence of psychotic symptoms; had chronic depression or dysthymia; had chronic or treatment-resistant anxiety disorders; had a history of mania or hypomania; or had active alcohol or substance abuse within the past year.

Treatment. Patients received escitalopram, 10 mg/d, after the baseline visit. At the investigator’s discretion, the dose could be increased to 20 mg/d or lowered to 5 mg/d if side effects occurred.

Measures. At the first visit, patients were assessed with the Mini-International Neuropsychiatric Interview to verify MDD and exclude diagnoses that would determine ineligibility. MADRS and Edinburgh Postnatal Depression Scale (EPDS) were used at each visit to measure depressive symptoms.3,4 The BAI was completed at each visit to measure anxiety symptoms. Obsessions and compulsions were measured with the Yale-Brown Obsessive Compulsive Scale (Y-BOCS)5 at baseline, and at all following visits if the patient scored >8 at baseline. The Clinical Global Impression Scales for severity and improvement were completed at each visit.6

Results

Of 7 patients enrolled, 3 completed the study, 2 were ineligible after the baseline visit, and 2 did not participate after the baseline visit (1 selected to pursue psychotherapy, and 1 was lost to follow-up).

Two of 3 patients responded to escitalopram (≥50% decrease on MADRS), and both were remitters (MADRS score <7). All 3 patients were responders on EPDS and BAI. One patient had Y-BOCS >8 at baseline (Total Y-BOCS score of 9, and final Y-BOCS score of 8) (Table).

Table

Symptom rating scale scores at baseline and study end

 Baseline (Visit 1)Final (Visit 5)
PatientMADRSBAIEPDSMADRSBAIEPDS
Ms. A2118221200
Ms. B282819452
Ms. C37619620
BAI: Beck Anxiety Inventory; EPDS: Edinburgh Postnatal Depression Scale; MADRS: Montgomery-Åsberg Depression Rating Scale

Discussion

Patients who stayed in treatment improved during the course of this study. Recruitment was difficult; we were able to recruit only 7 patients out of a projected 20 for the screening visit. We solicited feedback from local obstetrics health care providers and social workers on recruitment and attractiveness of the study as part of our routine collaboration with obstetrical services that screen for PPD. Primary reasons patients were not referred were that they were breast-feeding or they stated they would prefer to receive treatment from their primary care doctor. Recruitment difficulty in this study was in stark contrast to other recent studies completed at our center. For example, we have successfully recruited for menopausal depression and premenstrual dysphoric disorder treatment studies, and have completed large naturalistic studies of women with unipolar depression and bipolar disorder across pregnancy and postpartum. We suspect that many patients who were eligible for the study preferred to seek care from an obstetrician or primary care doctor with whom they already had a therapeutic alliance, and we also suspect that many women with PPD do not seek treatment at all, which is consistent with findings from other research groups.

 

 

Lessons learned from PPD research include:

  • Including women who are breast-feeding is important because many women choose to breast-feed and suffer from PPD. Because antidepressant use during breast-feeding has been closely studied, it is appropriate to include potential research participants who are breast-feeding as long as they receive adequate information and are able to provide informed consent.
  • Participants in PPD studies may require accommodations that take into account their role as a new mother, such as on-site childcare, home visits, or other strategies.
  • Because of recruitment challenges in postpartum patients, multisite trials may be required to include adequate numbers of participants.

Related Resource

  • Freeman MP, Joffe H, Cohen LS. Postpartum depression: Help patients find the right treatment. Current Psychiatry. 2012;11(11):14-21.

Drug Brand Names

  • Citalopram • Celexa
  • Escitalopram • Lexapro

Disclosures

Dr. Freeman has received grant or research support from Eli Lilly and Company, Forest Laboratories, and GlaxoSmithKline, is on the advisory boards of Otsuka and Takeda/Lundbeck, and is a consultant for PamLab LLC.

Dr. Joffe has received grant or research support from Cephalon/Teva, and is a consultant to Noven and Sunovion.

Dr. Cohen has received research support from AstraZeneca, Bayer HealthCare Pharmaceuticals, Bristol-Myers Squibb, Forest Laboratories, GlaxoSmithKline, National Institute of Mental Health, National Institute on Aging, National Institutes of Health, Ortho-McNeil Janssen, and Pfizer and has served on an advisory board for PamLab LLC.

This study was funded as an investigator-initiated trial by Forest Pharmaceuticals.

Challenges in recruiting women to postpartum depression (PPD) antidepressant treatment trials, which we encountered when conducting a trial of escitalopram, contribute to the limited body of knowledge about PPD treatment. Here we discuss results from a preliminary trial of escitalopram for PPD, and challenges of research in this area.

Escitalopram, the S-enantiomer of citalopram, is a selective serotonin reuptake inhibitor with high selectivity and potency that is FDA-approved for treating major depressive disorder (MDD) and generalized anxiety disorder. An agent with antidepressant and anxiolytic effects is particularly desirable for PPD because anxiety is more common in postpartum major depressive episodes than non-postpartum MDD.1 Anxiety and depressive disorders commonly are comorbid in postpartum women.2

We conducted an open-label trial of escitalopram for women with PPD and anxiety. We initially attempted to recruit 20 women.

Methods

Patients received 8 weeks of treatment with escitalopram, 10 to 20 mg/d (flexible dose). After completing the initial phone screen, patients had 5 follow-up visits, once every 2 weeks for 8 weeks. The institutional review board at Massachusetts General Hospital approved this study and we obtained written informed consent from all patients at the first visit. Twelve patients completed the phone screen and 7 eligible patients were enrolled in the study over 32 months. Reasons for ineligibility included having a history of psychosis, onset of symptoms >3 months postpartum, or presenting >6 months after onset. Others declined to participate because of concern about the time commitment or because they pursued nonpharmacologic treatments after the evaluation visit. One patient was lost to follow-up. Three patients completed the study. The study was halted because of the slow pace of recruitment.

Patient selection. Patients were screened for a major depressive episode with postpartum onset within 3 months of childbirth; depressive symptoms may have developed during pregnancy and worsened postpartum to meet criteria for MDD. Women were eligible for the study if they:

  • were age 18 to 45
  • experienced a major depressive episode with symptoms developing within 3 months of childbirth
  • presented within 6 months of childbirth
  • had a Montgomery-Åsberg Depression Rating Scale (MADRS) score >15
  • had a Beck Anxiety Inventory (BAI) score >10.

Patients who were pregnant or breast-feeding were excluded from the study per an agreement with the sponsor. In addition, women were excluded if they had taken any psychotropic medication within 2 weeks of enrollment; had active suicidal ideation, homicidal ideation, or presence of psychotic symptoms; had chronic depression or dysthymia; had chronic or treatment-resistant anxiety disorders; had a history of mania or hypomania; or had active alcohol or substance abuse within the past year.

Treatment. Patients received escitalopram, 10 mg/d, after the baseline visit. At the investigator’s discretion, the dose could be increased to 20 mg/d or lowered to 5 mg/d if side effects occurred.

Measures. At the first visit, patients were assessed with the Mini-International Neuropsychiatric Interview to verify MDD and exclude diagnoses that would determine ineligibility. MADRS and Edinburgh Postnatal Depression Scale (EPDS) were used at each visit to measure depressive symptoms.3,4 The BAI was completed at each visit to measure anxiety symptoms. Obsessions and compulsions were measured with the Yale-Brown Obsessive Compulsive Scale (Y-BOCS)5 at baseline, and at all following visits if the patient scored >8 at baseline. The Clinical Global Impression Scales for severity and improvement were completed at each visit.6

Results

Of 7 patients enrolled, 3 completed the study, 2 were ineligible after the baseline visit, and 2 did not participate after the baseline visit (1 selected to pursue psychotherapy, and 1 was lost to follow-up).

Two of 3 patients responded to escitalopram (≥50% decrease on MADRS), and both were remitters (MADRS score <7). All 3 patients were responders on EPDS and BAI. One patient had Y-BOCS >8 at baseline (Total Y-BOCS score of 9, and final Y-BOCS score of 8) (Table).

Table

Symptom rating scale scores at baseline and study end

 Baseline (Visit 1)Final (Visit 5)
PatientMADRSBAIEPDSMADRSBAIEPDS
Ms. A2118221200
Ms. B282819452
Ms. C37619620
BAI: Beck Anxiety Inventory; EPDS: Edinburgh Postnatal Depression Scale; MADRS: Montgomery-Åsberg Depression Rating Scale

Discussion

Patients who stayed in treatment improved during the course of this study. Recruitment was difficult; we were able to recruit only 7 patients out of a projected 20 for the screening visit. We solicited feedback from local obstetrics health care providers and social workers on recruitment and attractiveness of the study as part of our routine collaboration with obstetrical services that screen for PPD. Primary reasons patients were not referred were that they were breast-feeding or they stated they would prefer to receive treatment from their primary care doctor. Recruitment difficulty in this study was in stark contrast to other recent studies completed at our center. For example, we have successfully recruited for menopausal depression and premenstrual dysphoric disorder treatment studies, and have completed large naturalistic studies of women with unipolar depression and bipolar disorder across pregnancy and postpartum. We suspect that many patients who were eligible for the study preferred to seek care from an obstetrician or primary care doctor with whom they already had a therapeutic alliance, and we also suspect that many women with PPD do not seek treatment at all, which is consistent with findings from other research groups.

 

 

Lessons learned from PPD research include:

  • Including women who are breast-feeding is important because many women choose to breast-feed and suffer from PPD. Because antidepressant use during breast-feeding has been closely studied, it is appropriate to include potential research participants who are breast-feeding as long as they receive adequate information and are able to provide informed consent.
  • Participants in PPD studies may require accommodations that take into account their role as a new mother, such as on-site childcare, home visits, or other strategies.
  • Because of recruitment challenges in postpartum patients, multisite trials may be required to include adequate numbers of participants.

Related Resource

  • Freeman MP, Joffe H, Cohen LS. Postpartum depression: Help patients find the right treatment. Current Psychiatry. 2012;11(11):14-21.

Drug Brand Names

  • Citalopram • Celexa
  • Escitalopram • Lexapro

Disclosures

Dr. Freeman has received grant or research support from Eli Lilly and Company, Forest Laboratories, and GlaxoSmithKline, is on the advisory boards of Otsuka and Takeda/Lundbeck, and is a consultant for PamLab LLC.

Dr. Joffe has received grant or research support from Cephalon/Teva, and is a consultant to Noven and Sunovion.

Dr. Cohen has received research support from AstraZeneca, Bayer HealthCare Pharmaceuticals, Bristol-Myers Squibb, Forest Laboratories, GlaxoSmithKline, National Institute of Mental Health, National Institute on Aging, National Institutes of Health, Ortho-McNeil Janssen, and Pfizer and has served on an advisory board for PamLab LLC.

This study was funded as an investigator-initiated trial by Forest Pharmaceuticals.

References

1. Bernstein IH, Rush AJ, Yonkers K, et al. Symptom features of postpartum depression: are they distinct? Depress Anxiety. 2008;25(1):20-26.

2. Wenzel A, Haugen EN, Jackson LC, et al. Anxiety symptoms and disorders at eight weeks postpartum. J Anxiety Disord. 2005;19(3):295-311.

3. Cox JL, Holden JM, Sagovsky R. Detection of postnatal depression. Development of the 10-item Edinburgh Postnatal Depression Scale. Br J Psychiatry. 1987;150:782-786.

4. Montgomery SA, Åsberg M. A new depression scale designed to be sensitive to change. Br J Psychiatry. 1979;134:382-389.

5. Goodman WK, Price LH, Rasmussen SA, et al. The Yale-Brown Obsessive Compulsive Scale. I. Development, use, and reliability. Arch Gen Psychiatry. 1989;46(11):1006-1011.

6. Guy W. ECDEU assessment manual for psychopharmacology. Rockville MD: US Department of Health and Human Services; 1976. Department of Health, Education, and Welfare Publication (ADM) 76–338.

References

1. Bernstein IH, Rush AJ, Yonkers K, et al. Symptom features of postpartum depression: are they distinct? Depress Anxiety. 2008;25(1):20-26.

2. Wenzel A, Haugen EN, Jackson LC, et al. Anxiety symptoms and disorders at eight weeks postpartum. J Anxiety Disord. 2005;19(3):295-311.

3. Cox JL, Holden JM, Sagovsky R. Detection of postnatal depression. Development of the 10-item Edinburgh Postnatal Depression Scale. Br J Psychiatry. 1987;150:782-786.

4. Montgomery SA, Åsberg M. A new depression scale designed to be sensitive to change. Br J Psychiatry. 1979;134:382-389.

5. Goodman WK, Price LH, Rasmussen SA, et al. The Yale-Brown Obsessive Compulsive Scale. I. Development, use, and reliability. Arch Gen Psychiatry. 1989;46(11):1006-1011.

6. Guy W. ECDEU assessment manual for psychopharmacology. Rockville MD: US Department of Health and Human Services; 1976. Department of Health, Education, and Welfare Publication (ADM) 76–338.

Issue
Current Psychiatry - 11(11)
Issue
Current Psychiatry - 11(11)
Page Number
E3-E5
Page Number
E3-E5
Publications
Publications
Topics
Article Type
Display Headline
An open-label trial of escitalopram for PPD: Considerations for research
Display Headline
An open-label trial of escitalopram for PPD: Considerations for research
Legacy Keywords
escitalopram; postpartum depression; pregnancy; depression
Legacy Keywords
escitalopram; postpartum depression; pregnancy; depression
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media

Panic disorder: Break the fear circuit

Article Type
Changed
Tue, 12/11/2018 - 15:03
Display Headline
Panic disorder: Break the fear circuit


Dr. Dunlop: State-of-the-art treatment for panic disorder


Ms. K, a 24-year-old waitress who lives with her boyfriend, was referred by her primary care physician for evaluation of panic attacks that began “out of nowhere” at work approximately 6 months ago. The unpredictable attacks occur multiple times per week, causing her to leave work and cancel shifts.

Ms. K reports that before the panic attacks began, she felt happy in her relationship, enjoyed hobbies, and was hopeful about the future. However, she has become concerned that a potentially catastrophic illness is causing her panic attacks. She researches her symptoms on the Internet, and is preoccupied with the possibility of sudden death due to an undiagnosed heart condition. Multiple visits to the emergency room have not identified any physical abnormalities. Her primary care doctor prescribed alprazolam, 0.5 mg as needed for panic attacks, which she reports is helpful, “but only in the moment of the attacks.” Ms. K avoids alcohol and illicit substances and limits her caffeine intake. She is not willing to accept that her life “feels so limited.” Her dream of earning a nursing degree and eventually starting a family now seems unattainable.

Panic disorder (PD) occurs in 3% to 5% of adults, with women affected at roughly twice the rate of men.1 Causing a broad range of distress and varying degrees of impairment, PD commonly occurs with other psychiatric disorders. For most patients, treatment is effective, but those who do not respond to initial approaches require a thoughtful, stepped approach to care. Key considerations include establishing an accurate diagnosis, clarifying comorbid illnesses, ascertaining patient beliefs and expectations, and providing appropriately dosed and maintained treatments.

Panic attacks vs PD

Panic attacks consist of rapid onset of intense anxiety, with prominent somatic symptoms, that peaks within 10 minutes (Figure).2 Attacks in which <4 of the listed symptoms occur are considered limited-symptom panic attacks.


Figure: Body locations of panic attack symptoms
Diagnosis of a panic attack requires the sudden development of intense fear or discomfort characterized by ≥4 of the 13 symptoms listed above that peaks in intensity within 10 minutes of onset
Source: Reference 2

Panic attacks can occur with various disorders, including other anxiety disorders, mood disorders, and substance intoxication or withdrawal. Because serious medical conditions can present with panic-like symptoms, the initial occurrence of such symptoms warrants consideration of physiological causes. For a Box2 that describes the differential diagnosis of panic attacks, see this article at CurrentPsychiatry.com.

Box

Differential diagnosis of panic attacks

To meet diagnostic criteria for panic disorder, panic attacks must initially occur “out of the blue,” meaning no specific object or situation induced the attack. The differential diagnosis of panic attacks includes assessing for other psychiatric disorders that may involve panic attacks. Evaluation requires considering the context in which the panic attacks occur, including their start date, pattern of attacks, instigating situations, and associated thoughts.

Social phobia. Attacks occur only during or immediately before a social interaction in which the patient fears embarrassing himself or herself.

Obsessive-compulsive disorder (OCD). Attacks occur when the patient cannot avoid exposure to an obsessional fear or is prevented from performing a ritual that diffuses obsessional anxiety.

Posttraumatic stress disorder (PTSD). Attacks occur when confronted by a trauma-related memory or trigger.

Specific phobia. Attacks occur only when the patient encounters a specifically feared object, place, or situation, unrelated to social phobia, OCD, or PTSD.

Medical conditions. Conditions to consider include—but are not limited to—hyperthyroidism, pulmonary embolism, myocardial infarction, cardiac dysrhythmias, hypoglycemia, asthma, partial complex seizures, and pheochromocytoma.

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

A PD diagnosis requires that repeated panic attacks initially must occur from “out of the blue,” meaning no specific object or situation induced the attack. In addition, the diagnosis requires 1 of 3 types of psychological or behavioral changes as a result of the attacks (Table 1).2 Agoraphobia is diagnosed if 1 of the behavioral changes is avoidance of places or situations from which escape might be embarrassing or difficult should an attack occur. A patient can be diagnosed as having PD with agoraphobia, PD without agoraphobia, or agoraphobia without PD (ie, experiences only limited symptom panic attacks, but avoids situations or stimuli associated with them).

Table 1

Definitions of panic disorder and agoraphobia

Panic disorder
  1. Both 1 and 2:
  2. The panic attacks do not arise from the direct physiological effects of substance intoxication (eg, caffeine, cocaine) or withdrawal (eg, cannabis, gamma-hydroxybutyrate) or from a general medical condition
  3. The panic attacks are not better accounted for by another mental disorder
Agoraphobia
Anxiety about, or avoidance of, being in places or situations from which escape might be difficult or embarrassing, or in which help may not be available in the event of having an unexpected or situationally predisposed panic attack or panic-like symptoms. Agoraphobic fears typically involve characteristic clusters of situations that include being outside the home alone, being in a crowd, standing in a line, being on a bridge, or traveling in a bus, train, or automobile
Source: Reference 2
 

 

Comorbidities are common in patients with PD and predict greater difficulty achieving remission (Box).1,3-6

Box

Comorbidities: How they affect panic disorder treatment

The most common psychiatric conditions that co-occur with panic disorder (PD) are other anxiety disorders, mood disorders, personality disorders, and substance use disorders.1 Carefully assess the severity and degree of impairment or distress arising from each condition to prioritize treatment goals. For example, treating panic attacks would be a lower priority in a patient with untreated bipolar disorder.

Assessing comorbid substance abuse is important in selecting PD treatments. Benzodiazepines should almost always be avoided in patients with a history of drug abuse—illicit or prescribed. Although complete abstinence should not be a prerequisite for beginning PD treatment, detoxification and concomitant substance abuse treatment are essential.3

Comorbid mood disorders also affect the course of PD treatment. Antidepressants are effective for treating depression and PD, whereas benzodiazepines are not effective for depression.4 Antidepressants in patients with bipolar disorder are controversial because these medications might induce mixed or elevated mood states or rapid cycling. In these complicated patients, consider antidepressants lower in the treatment algorithm.5

Other conditions to consider before beginning treatment include pregnancy or the possibility of becoming pregnant in the near future and suicidal ideation. PD is associated with increased risk for suicidal ideation and progression to suicide attempts, particularly in patients with a comorbid mood or psychotic disorder.6 In addition, consider the potential impact of medications on comorbid medical conditions.

Treatment begins with education

The goal of treatment is remission of symptoms, ideally including an absence of panic attacks, agoraphobic avoidance, and anticipatory anxiety.1 The Panic Disorder Severity Scale self-report is a validated measure of panic symptoms that may be useful in clinical practice.7

The first step in treatment is educating patients about panic attacks, framing them as an overreactive fear circuit in the brain that produces physical symptoms that are not dangerous. Using a brain model that shows the location of the amygdala, hippocampus, and prefrontal cortex—which play crucial roles in generating and controlling anxiety and fear—can make this discussion more concrete.8 Although highly simplified, such models allow clinicians to demonstrate that excessive reactivity of limbic regions can be reduced by both top-down (cortico-limbic connections via cognitive-behavioral therapy [CBT]) and bottom-up (pharmacotherapy directly acting on limbic structures) approaches. Such discussions lead to treatment recommendations for CBT, pharmacotherapy, or their combination.

No single treatment has emerged as the definitive “best” for PD, and no reliable predictors can guide specific treatment for an individual.3 Combining CBT with pharmacotherapy produces higher short-term response rates than either treatment alone, but in the long term, combination treatment does not appear to be superior to CBT alone.9 Base the initial treatment selection for PD on patient preference, treatment availability and cost, and comorbid medical and psychiatric conditions. For an Algorithm to guide treatment decisions, see this article at CurrentPsychiatry.com.


Algorithm: Treatment for panic disorder: A suggested algorithm
aPoor response to an SSRI should lead to a switch to venlafaxine extended-release, and vice versa
bBenzodiazepines are relatively contraindicated in geriatric patients and patients with a history of substance abuse or dependence
CBT: cognitive-behavioral therapy; MAOI: monoamine oxidase inhibitor; SSRI: selective serotonin reuptake inhibitor; TCA: tricyclic antidepressant; Ven XR: venlafaxine extended-release

First-line treatments

Psychotherapy. CBT is the most efficacious psychotherapy for PD. Twelve to 15 sessions of CBT has demonstrated efficacy for PD, with additional effects on comorbid anxiety and depressive symptoms.10 No large clinical trials of CBT have used cognitive restructuring alone; all have included at least some component of exposure that requires the patient to confront feared physical sensations. Gains during treatment may be steady and gradual or sudden and uneven, with rapid improvement in some but not all symptoms. CBT and pharmacotherapy have demonstrated similar levels of benefit in short-term trials, but CBT has proven superior in most9 but not all11 trials evaluating long-term outcomes, particularly compared with pharmacotherapy that is discontinued during follow-up. Although less studied, group CBT also may be considered if a patient cannot afford individual CBT.

Pharmacotherapy. Evidence supports selective serotonin reuptake inhibitors (SSRIs), venlafaxine extended-release (XR), benzodiazepines, and tricyclic antidepressants (TCAs) as effective treatments for PD.3 No class of medication has demonstrated superiority over others in short-term treatment.3,12 Because of the medical risks associated with benzodiazepines and TCAs, an SSRI or venlafaxine XR should be the first medication option for most patients. Fluoxetine, paroxetine, sertraline, and venlafaxine XR are FDA-approved for PD. Paroxetine is associated with weight gain and may increase the risk for panic recurrence upon discontinuation more than sertraline, making it a less favorable option for many patients.13 Start doses at half the normal starting dose used for treating major depressive disorder and continue for 4 to 7 days, then increase to the minimal effective dose. For a Table3 that lists dosing recommendations for antidepressants to treat PD, see this article at CurrentPsychiatry.com. If there is no improvement by 4 weeks, increase the dose every 2 to 4 weeks until remission is achieved or side effects prevent further dose increases.

 

 

Table

Recommended doses for antidepressants used to treat panic disorder

MedicationStarting dose (mg/d)Therapeutic range (mg/d)
SSRIs
   Citalopram1020 to 40
   Escitalopram510 to 40
   Fluoxetine5 to 1020 to 80
   Fluvoxamine25100 to 300
   Paroxetine1020 to 80
   Paroxetine CR12.525 to 50
   Sertraline25100 to 200
SNRIs
   Duloxetine20 to 3060 to 120
   Venlafaxine XR37.5150 to 225
TCAs
   Clomipramine10 to 25100 to 300
   Imipramine10100 to 300
MAOI
   Phenelzine1545 to 90
CR: controlled release; MAOI: monoamine oxidase inhibitor; SNRIs: serotonin-norepinephrine reuptake inhibitors; SSRIs: selective serotonin reuptake inhibitors; TCAs: tricyclic antidepressants; XR: extended release
Source: American Psychiatric Association. Practice guideline for the treatment of patients with panic disorder. 2nd ed. Washington, DC: American Psychiatric Association; 2009

Treatment nonresponse. True non-response needs to be distinguished from poor response caused by inadequate treatment delivery, eg, patients not completing homework assignments in CBT or not adhering to pharmacotherapy. Asking patients about adverse effects or personal and family beliefs about treatment may reveal reasons for nonadherence.

Second-line treatments

Little data are available to guide next-step treatment options in patients who don’t achieve remission from their initial treatment. Patients who benefit from an SSRI, venlafaxine XR, or CBT but still have symptoms should be started on combination treatment. For a patient who experiences complete non-response to the initial treatment, discontinue the first treatment and switch to the other modality. In general, completely ineffective treatments should be discontinued when another treatment is added, but when partial improvement (>30%) occurs, continue the original treatment and augment it with another approach.

For patients pursuing pharmacotherapy, poor response to an adequate SSRI trial usually should lead to a switch to venlafaxine XR, and vice versa. Failure to respond to both of these medication classes should prompt a switch to a benzodiazepine or TCA.

Benzodiazepines are a fast-acting, effective treatment for PD, with efficacy similar to SSRIs in acute and long-term treatment.14 Benzodiazepines may be prescribed with antidepressants at the beginning of treatment to improve response speed.15 Clonazepam and alprazolam are FDA-approved for treating PD. A high-potency, long-acting agent, clonazepam is the preferred initial benzodiazepine, dosed 0.5 to 4 mg/d on a fixed schedule. Although substantial data support using alprazolam for PD, it requires more frequent dosing and has a greater risk of rebound anxiety and abuse potential because of its more rapid onset of action. Compared with immediate-release alprazolam, alprazolam XR has a slower absorption rate and longer steady state in the blood, but this formulation does not have lower abuse potential or greater efficacy. Although not FDA-approved for PD, diazepam and lorazepam also have proven efficacy for PD.3

Benzodiazepines should be considered contraindicated in patients with a history of substance abuse, except in select cases.4 Benzodiazepines generally should be avoided in older patients because of increased risk for falls, cognitive impairment, and motor vehicle accidents. Table 2 lists situations in which benzodiazepines may be used to treat PD.

Table 2

Clinical scenarios in which to consider using benzodiazepines

Coadministration for 2 to 4 weeks when initiating treatment with an SSRI or venlafaxine XR to achieve more rapid relief and mitigate potential antidepressant-induced anxiety
For patients who wish to avoid antidepressants because of concern about sexual dysfunction
For patients who need chronic aspirin or an NSAID, which may increase the risk for upper gastrointestinal bleeding when taken in combination with an SSRI
For patients with comorbid bipolar disorder or epilepsy
Next-step monotherapy or augmentation in patients who respond poorly to an SSRI, venlafaxine XR, TCA, or CBT
CBT: cognitive-behavioral therapy; NSAID: nonsteroidal anti-inflammatory drug; SSRI: selective serotonin reuptake inhibitor; TCA: tricyclic antidepressant; XR: extended release

TCAs are effective as monotherapy for PD. Most support comes from studies of imipramine or clomipramine.12 Similar to SSRIs and venlafaxine XR, use a low initial dose and gradually increase until the patient remits or side effects prevent further increases. SSRI and TCA combinations rarely are used unless the TCA is a relatively specific norepinephrine reuptake inhibitor (eg, desipramine, nortriptyline). Because TCAs are metabolized via the cytochrome P450 2D6 system and some SSRIs—particularly fluoxetine and paroxetine—strongly inhibit 2D6, combinations of TCAs with these agents may lead to dangerously high plasma TCA levels, placing patients at risk for cardiac dysrhythmias and other side effects.16

Monoamine oxidase inhibitors (MAOIs)—particularly phenelzine—are underused for PD. They have the strongest efficacy data for any class of medications outside the first- and second-line agents and have a unique mechanism of action. In patients who can comply with the dietary and medication limitations, an MAOI generally should be the next step after nonresponse to other treatments.3

 

 

Alternative treatments

For patients who do not respond to any of the treatments described above, data from uncontrolled studies support mirtazapine, levetiracetam, and the serotonin-norepinephrine reuptake inhibitors duloxetine and milnacipran as monotherapy for PD.17 Pindolol—a beta blocker and 5-HT1A receptor antagonist—proved superior to placebo as an adjunctive agent to SSRIs in treatment-resistant PD in 1 of 2 trials.17 Minimal evidence supports the atypical antipsychotics risperidone and olanzapine in treatment-resistant PD, although a placebo-controlled trial of quetiapine SR coadministered with SSRIs recently was completed (NCT00619892; results pending). Atypical antipsychotics are best reserved for patients with a primary psychotic disorder or bipolar disorder who experience panic attacks.5

Panic-focused psychodynamic psychotherapy, a 12-week (approximately 24 sessions) form of psychotherapy, has demonstrated superiority vs applied relaxation therapy.18 This treatment could be considered for patients who do not respond to standard first-line treatments, but few community therapists are familiar with this method.

For many patients with PD, complementary and alternative medicine (CAM) approaches are appealing. See this article at CurrentPsychiatry.com for a Box that discusses CAM for PD.

Box

Complementary and alternative medicine for panic disorder

Although no complementary and alternative medicine treatments have strong evidence of efficacy as monotherapy for panic disorder (PD), several have data that suggest benefit with little evidence of risk. These include bibliotherapy, yoga, aerobic exercise, and the dietary supplements kava and inositol.a Exercise as a treatment poses a challenge because it can induce symptoms that the patient fears, such as tachycardia and shortness of breath. In addition to any direct physiologic benefit from aerobic exercise, there is also an exposure component that can be harnessed by gradually increasing the exertion level.

Another approach undergoing extensive evaluation is Internet-provided cognitive-behavioral therapy (CBT). Using guided CBT modules with or without therapist support, Internet-provided CBT provides an option for motivated patients unable to complete in-person CBT because of logistical factors.b A helpful resource that reviews Internet self-help and psychotherapy guided programs for PD and other psychiatric conditions is http://beacon.anu.edu.au.

References

a. Antonacci DJ, Davis E, Bloch RM, et al. CAM for your anxious patient: what the evidence says. Current Psychiatry. 2010;9(10):42-52.

b. Johnston L, Titov N, Andrews G, et al. A RCT of a transdiagnostic internet-delivered treatment for three anxiety disorders: examination of support roles and disorder-specific outcomes. PLoS One. 2011;6(11):e28079.

Maintenance treatment

Patients who complete a course of CBT for PD often follow up with several “booster sessions” at monthly or longer intervals that focus on relapse prevention techniques. Few controlled trials have evaluated pharmacotherapy discontinuation in PD. Most guidelines recommend continuing treatment for ≥1 year after achieving remission to minimize the risk of relapse.3 Researchers are focusing on whether medication dosage can be reduced during maintenance without loss of efficacy.

Treatment discontinuation

In the absence of urgent medical need, taper medications for PD gradually over several months. PD patients are highly sensitive to unusual physical sensations, which can occur while discontinuing antidepressants or benzodiazepines. If a benzodiazepine is used in conjunction with an antidepressant, the benzodiazepine should be discontinued first, so that the antidepressant can help ease benzodiazepine-associated discontinuation symptoms. A brief course of CBT during pharmacotherapy discontinuation may increase the likelihood of successful tapering.19

CASE CONTINUED: A successful switch

Ms. K has to discontinue sequential trials of fluoxetine, 40 mg/d, and venlafaxine XR, 225 mg/d because of side effects, and she does not reduce the frequency of her alprazolam use. She agrees to switch from alprazolam to clonazepam, 0.5 mg every morning and 1 mg at bedtime, and to start CBT. Clonazepam reduces her anxiety sufficiently so she can address her symptoms in therapy. Through CBT she becomes motivated to monitor her thoughts and treat them as guesses rather than facts, reviewing the evidence for her thoughts and generating rational responses. She participates in exposure exercises, which she practices between sessions, and grows to tolerate uncomfortable sensations until they no longer signal danger. After 12 CBT sessions, she is panic-free. Despite some trepidation, she agrees to a slow taper off clonazepam, reducing the dose by 0.25 mg every 2 weeks. She continues booster sessions with her therapist to manage any re-emerging anxiety. After an additional 12 weeks, she successfully discontinues clonazepam and remains panic-free.

Related Resources

Drug Brand Names

 

 

  • Alprazolam • Xanax
  • Alprazolam XR • Xanax XR
  • Citalopram • Celexa
  • Clomipramine • Anafranil
  • Clonazepam • Klonopin
  • Desipramine • Norpramin
  • Diazepam • Valium
  • Duloxetine • Cymbalta
  • Escitalopram • Lexapro
  • Fluoxetine • Prozac
  • Fluvoxamine • Luvox
  • Imipramine • Tofranil
  • Levetiracetam • Keppra
  • Lorazepam • Ativan
  • Milnacipran • Savella
  • Mirtazapine • Remeron
  • Nortriptyline • Aventyl, Pamelor
  • Olanzapine • Zyprexa
  • Paroxetine • Paxil
  • Paroxetine CR • Paxil CR
  • Phenelzine • Nardil
  • Pindolol • Visken
  • Quetiapine SR • Seroquel SR
  • Risperidone • Risperdal
  • Sertraline • Zoloft
  • Venlafaxine XR • Effexor XR

Disclosures

Dr. Dunlop receives research support from Bristol-Myers Squibb, GlaxoSmithKline, and the National Institute of Mental Health. He serves as a consultant to MedAvante and Roche.

Ms. Schneider and Dr. Gerardi report no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.

References

1. Roy-Byrne PP, Craske MG, Stein MB. Panic disorder. Lancet. 2006;368(9540):1023-1032.

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

3. American Psychiatric Association. Practice guideline for the treatment of patients with panic disorder. 2nd ed. Washington DC: American Psychiatric Association; 2009.

4. Dunlop BW, Davis PG. Combination treatment with benzodiazepines and SSRIs for comorbid anxiety and depression: a review. Prim Care Companion J Clin Psychiatry. 2008;10(3):222-228.

5. Rakofsky JJ, Dunlop BW. Treating nonspecific anxiety and anxiety disorders in patients with bipolar disorder: a review. J Clin Psychiatry. 2011;72(1):81-90.

6. Sareen J, Cox BJ, Afifi TO, et al. Anxiety disorders and risk for suicidal ideation and suicide attempts: a population-based longitudinal study of adults. Arch Gen Psychiatry. 2005;62(11):1249-1257.

7. Houck PR, Spiegel DA, Shear MK, et al. Reliability of the self-report version of the panic disorder severity scale. Depress Anxiety. 2002;15(4):183-185.

8. Ninan PT, Dunlop BW. Neurobiology and etiology of panic disorder. J Clin Psychiatry. 2005;66(suppl 4):3-7.

9. Furukawa TA, Watanabe N, Churchill R. Psychotherapy plus antidepressant for panic disorder with or without agoraphobia: systematic review. Br J Psychiatry. 2006;188:305-312.

10. Barlow DH, Gorman JM, Shear MK, et al. Cognitive-behavioral therapy, imipramine, or their combination for panic disorder: a randomized controlled trial. JAMA. 2000;283(19):2529-2536.

11. van Apeldoorn FJ, Timmerman ME, Mersch PP, et al. A randomized trial of cognitive-behavioral therapy or selective serotonin reuptake inhibitor or both combined for panic disorder with or without agoraphobia: treatment results through 1-year follow-up. J Clin Psychiatry. 2010;71(5):574-586.

12. Bakker A, van Balkom AJ, Spinhoven P. SSRIs vs. TCAs in the treatment of panic disorder: a meta-analysis. Acta Psychiatr Scand. 2002;106(3):163-167.

13. Bandelow B, Behnke K, Lenoir S, et al. Sertraline versus paroxetine in the treatment of panic disorder: an acute, double-blind noninferiority comparison. J Clin Psychiatry. 2004;65(3):405-413.

14. Nardi AE, Freire RC, Mochcovitch MD, et al. A randomized, naturalistic, parallel-group study for the long-term treatment of panic disorder with clonazepam or paroxetine. J Clin Psychopharmacol. 2012;32(1):120-126.

15. Goddard AW, Brouette T, Almai A, et al. Early coadministration of clonazepam with sertraline for panic disorder. Arch Gen Psychiatry. 2001;58(7):681-686.

16. Preskorn SH, Shah R, Neff M, et al. The potential for clinically significant drug-drug interactions involving the CYP 2D6 system: effects with fluoxetine and paroxetine versus sertraline. J Psychiatr Pract. 2007;13(1):5-12.

17. Perna G, Guerriero G, Caldirola D. Emerging drugs for panic disorder. Expert Opin Emerg Drugs. 2011;16(4):631-645.

18. Milrod B, Leon AC, Busch F, et al. A randomized controlled clinical trial of psychoanalytic psychotherapy for panic disorder. Am J Psychiatry. 2007;164(2):265-272.

19. Otto MW, Pollack MH, Sachs GS, et al. Discontinuation of benzodiazepine treatment: efficacy of cognitive-behavioral therapy for patients with panic disorder. Am J Psychiatry. 1993;150(10):1485-1490.

Article PDF
Author and Disclosure Information

Boadie W. Dunlop, MD, MSCR
Assistant Professor, Director, Mood and Anxiety Disorders Program, Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA
Rebecca Schneider, BA
Research Coordinator, Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA
Maryrose Gerardi, PhD
Assistant Professor, Trauma and Anxiety Recovery Program, Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA

Issue
Current Psychiatry - 11(11)
Publications
Topics
Page Number
36-47
Legacy Keywords
panic disorder; fear; cognitive-behavioral therapy; antidepressants; panic attacks
Sections
Author and Disclosure Information

Boadie W. Dunlop, MD, MSCR
Assistant Professor, Director, Mood and Anxiety Disorders Program, Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA
Rebecca Schneider, BA
Research Coordinator, Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA
Maryrose Gerardi, PhD
Assistant Professor, Trauma and Anxiety Recovery Program, Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA

Author and Disclosure Information

Boadie W. Dunlop, MD, MSCR
Assistant Professor, Director, Mood and Anxiety Disorders Program, Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA
Rebecca Schneider, BA
Research Coordinator, Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA
Maryrose Gerardi, PhD
Assistant Professor, Trauma and Anxiety Recovery Program, Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA

Article PDF
Article PDF


Dr. Dunlop: State-of-the-art treatment for panic disorder


Ms. K, a 24-year-old waitress who lives with her boyfriend, was referred by her primary care physician for evaluation of panic attacks that began “out of nowhere” at work approximately 6 months ago. The unpredictable attacks occur multiple times per week, causing her to leave work and cancel shifts.

Ms. K reports that before the panic attacks began, she felt happy in her relationship, enjoyed hobbies, and was hopeful about the future. However, she has become concerned that a potentially catastrophic illness is causing her panic attacks. She researches her symptoms on the Internet, and is preoccupied with the possibility of sudden death due to an undiagnosed heart condition. Multiple visits to the emergency room have not identified any physical abnormalities. Her primary care doctor prescribed alprazolam, 0.5 mg as needed for panic attacks, which she reports is helpful, “but only in the moment of the attacks.” Ms. K avoids alcohol and illicit substances and limits her caffeine intake. She is not willing to accept that her life “feels so limited.” Her dream of earning a nursing degree and eventually starting a family now seems unattainable.

Panic disorder (PD) occurs in 3% to 5% of adults, with women affected at roughly twice the rate of men.1 Causing a broad range of distress and varying degrees of impairment, PD commonly occurs with other psychiatric disorders. For most patients, treatment is effective, but those who do not respond to initial approaches require a thoughtful, stepped approach to care. Key considerations include establishing an accurate diagnosis, clarifying comorbid illnesses, ascertaining patient beliefs and expectations, and providing appropriately dosed and maintained treatments.

Panic attacks vs PD

Panic attacks consist of rapid onset of intense anxiety, with prominent somatic symptoms, that peaks within 10 minutes (Figure).2 Attacks in which <4 of the listed symptoms occur are considered limited-symptom panic attacks.


Figure: Body locations of panic attack symptoms
Diagnosis of a panic attack requires the sudden development of intense fear or discomfort characterized by ≥4 of the 13 symptoms listed above that peaks in intensity within 10 minutes of onset
Source: Reference 2

Panic attacks can occur with various disorders, including other anxiety disorders, mood disorders, and substance intoxication or withdrawal. Because serious medical conditions can present with panic-like symptoms, the initial occurrence of such symptoms warrants consideration of physiological causes. For a Box2 that describes the differential diagnosis of panic attacks, see this article at CurrentPsychiatry.com.

Box

Differential diagnosis of panic attacks

To meet diagnostic criteria for panic disorder, panic attacks must initially occur “out of the blue,” meaning no specific object or situation induced the attack. The differential diagnosis of panic attacks includes assessing for other psychiatric disorders that may involve panic attacks. Evaluation requires considering the context in which the panic attacks occur, including their start date, pattern of attacks, instigating situations, and associated thoughts.

Social phobia. Attacks occur only during or immediately before a social interaction in which the patient fears embarrassing himself or herself.

Obsessive-compulsive disorder (OCD). Attacks occur when the patient cannot avoid exposure to an obsessional fear or is prevented from performing a ritual that diffuses obsessional anxiety.

Posttraumatic stress disorder (PTSD). Attacks occur when confronted by a trauma-related memory or trigger.

Specific phobia. Attacks occur only when the patient encounters a specifically feared object, place, or situation, unrelated to social phobia, OCD, or PTSD.

Medical conditions. Conditions to consider include—but are not limited to—hyperthyroidism, pulmonary embolism, myocardial infarction, cardiac dysrhythmias, hypoglycemia, asthma, partial complex seizures, and pheochromocytoma.

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

A PD diagnosis requires that repeated panic attacks initially must occur from “out of the blue,” meaning no specific object or situation induced the attack. In addition, the diagnosis requires 1 of 3 types of psychological or behavioral changes as a result of the attacks (Table 1).2 Agoraphobia is diagnosed if 1 of the behavioral changes is avoidance of places or situations from which escape might be embarrassing or difficult should an attack occur. A patient can be diagnosed as having PD with agoraphobia, PD without agoraphobia, or agoraphobia without PD (ie, experiences only limited symptom panic attacks, but avoids situations or stimuli associated with them).

Table 1

Definitions of panic disorder and agoraphobia

Panic disorder
  1. Both 1 and 2:
  2. The panic attacks do not arise from the direct physiological effects of substance intoxication (eg, caffeine, cocaine) or withdrawal (eg, cannabis, gamma-hydroxybutyrate) or from a general medical condition
  3. The panic attacks are not better accounted for by another mental disorder
Agoraphobia
Anxiety about, or avoidance of, being in places or situations from which escape might be difficult or embarrassing, or in which help may not be available in the event of having an unexpected or situationally predisposed panic attack or panic-like symptoms. Agoraphobic fears typically involve characteristic clusters of situations that include being outside the home alone, being in a crowd, standing in a line, being on a bridge, or traveling in a bus, train, or automobile
Source: Reference 2
 

 

Comorbidities are common in patients with PD and predict greater difficulty achieving remission (Box).1,3-6

Box

Comorbidities: How they affect panic disorder treatment

The most common psychiatric conditions that co-occur with panic disorder (PD) are other anxiety disorders, mood disorders, personality disorders, and substance use disorders.1 Carefully assess the severity and degree of impairment or distress arising from each condition to prioritize treatment goals. For example, treating panic attacks would be a lower priority in a patient with untreated bipolar disorder.

Assessing comorbid substance abuse is important in selecting PD treatments. Benzodiazepines should almost always be avoided in patients with a history of drug abuse—illicit or prescribed. Although complete abstinence should not be a prerequisite for beginning PD treatment, detoxification and concomitant substance abuse treatment are essential.3

Comorbid mood disorders also affect the course of PD treatment. Antidepressants are effective for treating depression and PD, whereas benzodiazepines are not effective for depression.4 Antidepressants in patients with bipolar disorder are controversial because these medications might induce mixed or elevated mood states or rapid cycling. In these complicated patients, consider antidepressants lower in the treatment algorithm.5

Other conditions to consider before beginning treatment include pregnancy or the possibility of becoming pregnant in the near future and suicidal ideation. PD is associated with increased risk for suicidal ideation and progression to suicide attempts, particularly in patients with a comorbid mood or psychotic disorder.6 In addition, consider the potential impact of medications on comorbid medical conditions.

Treatment begins with education

The goal of treatment is remission of symptoms, ideally including an absence of panic attacks, agoraphobic avoidance, and anticipatory anxiety.1 The Panic Disorder Severity Scale self-report is a validated measure of panic symptoms that may be useful in clinical practice.7

The first step in treatment is educating patients about panic attacks, framing them as an overreactive fear circuit in the brain that produces physical symptoms that are not dangerous. Using a brain model that shows the location of the amygdala, hippocampus, and prefrontal cortex—which play crucial roles in generating and controlling anxiety and fear—can make this discussion more concrete.8 Although highly simplified, such models allow clinicians to demonstrate that excessive reactivity of limbic regions can be reduced by both top-down (cortico-limbic connections via cognitive-behavioral therapy [CBT]) and bottom-up (pharmacotherapy directly acting on limbic structures) approaches. Such discussions lead to treatment recommendations for CBT, pharmacotherapy, or their combination.

No single treatment has emerged as the definitive “best” for PD, and no reliable predictors can guide specific treatment for an individual.3 Combining CBT with pharmacotherapy produces higher short-term response rates than either treatment alone, but in the long term, combination treatment does not appear to be superior to CBT alone.9 Base the initial treatment selection for PD on patient preference, treatment availability and cost, and comorbid medical and psychiatric conditions. For an Algorithm to guide treatment decisions, see this article at CurrentPsychiatry.com.


Algorithm: Treatment for panic disorder: A suggested algorithm
aPoor response to an SSRI should lead to a switch to venlafaxine extended-release, and vice versa
bBenzodiazepines are relatively contraindicated in geriatric patients and patients with a history of substance abuse or dependence
CBT: cognitive-behavioral therapy; MAOI: monoamine oxidase inhibitor; SSRI: selective serotonin reuptake inhibitor; TCA: tricyclic antidepressant; Ven XR: venlafaxine extended-release

First-line treatments

Psychotherapy. CBT is the most efficacious psychotherapy for PD. Twelve to 15 sessions of CBT has demonstrated efficacy for PD, with additional effects on comorbid anxiety and depressive symptoms.10 No large clinical trials of CBT have used cognitive restructuring alone; all have included at least some component of exposure that requires the patient to confront feared physical sensations. Gains during treatment may be steady and gradual or sudden and uneven, with rapid improvement in some but not all symptoms. CBT and pharmacotherapy have demonstrated similar levels of benefit in short-term trials, but CBT has proven superior in most9 but not all11 trials evaluating long-term outcomes, particularly compared with pharmacotherapy that is discontinued during follow-up. Although less studied, group CBT also may be considered if a patient cannot afford individual CBT.

Pharmacotherapy. Evidence supports selective serotonin reuptake inhibitors (SSRIs), venlafaxine extended-release (XR), benzodiazepines, and tricyclic antidepressants (TCAs) as effective treatments for PD.3 No class of medication has demonstrated superiority over others in short-term treatment.3,12 Because of the medical risks associated with benzodiazepines and TCAs, an SSRI or venlafaxine XR should be the first medication option for most patients. Fluoxetine, paroxetine, sertraline, and venlafaxine XR are FDA-approved for PD. Paroxetine is associated with weight gain and may increase the risk for panic recurrence upon discontinuation more than sertraline, making it a less favorable option for many patients.13 Start doses at half the normal starting dose used for treating major depressive disorder and continue for 4 to 7 days, then increase to the minimal effective dose. For a Table3 that lists dosing recommendations for antidepressants to treat PD, see this article at CurrentPsychiatry.com. If there is no improvement by 4 weeks, increase the dose every 2 to 4 weeks until remission is achieved or side effects prevent further dose increases.

 

 

Table

Recommended doses for antidepressants used to treat panic disorder

MedicationStarting dose (mg/d)Therapeutic range (mg/d)
SSRIs
   Citalopram1020 to 40
   Escitalopram510 to 40
   Fluoxetine5 to 1020 to 80
   Fluvoxamine25100 to 300
   Paroxetine1020 to 80
   Paroxetine CR12.525 to 50
   Sertraline25100 to 200
SNRIs
   Duloxetine20 to 3060 to 120
   Venlafaxine XR37.5150 to 225
TCAs
   Clomipramine10 to 25100 to 300
   Imipramine10100 to 300
MAOI
   Phenelzine1545 to 90
CR: controlled release; MAOI: monoamine oxidase inhibitor; SNRIs: serotonin-norepinephrine reuptake inhibitors; SSRIs: selective serotonin reuptake inhibitors; TCAs: tricyclic antidepressants; XR: extended release
Source: American Psychiatric Association. Practice guideline for the treatment of patients with panic disorder. 2nd ed. Washington, DC: American Psychiatric Association; 2009

Treatment nonresponse. True non-response needs to be distinguished from poor response caused by inadequate treatment delivery, eg, patients not completing homework assignments in CBT or not adhering to pharmacotherapy. Asking patients about adverse effects or personal and family beliefs about treatment may reveal reasons for nonadherence.

Second-line treatments

Little data are available to guide next-step treatment options in patients who don’t achieve remission from their initial treatment. Patients who benefit from an SSRI, venlafaxine XR, or CBT but still have symptoms should be started on combination treatment. For a patient who experiences complete non-response to the initial treatment, discontinue the first treatment and switch to the other modality. In general, completely ineffective treatments should be discontinued when another treatment is added, but when partial improvement (>30%) occurs, continue the original treatment and augment it with another approach.

For patients pursuing pharmacotherapy, poor response to an adequate SSRI trial usually should lead to a switch to venlafaxine XR, and vice versa. Failure to respond to both of these medication classes should prompt a switch to a benzodiazepine or TCA.

Benzodiazepines are a fast-acting, effective treatment for PD, with efficacy similar to SSRIs in acute and long-term treatment.14 Benzodiazepines may be prescribed with antidepressants at the beginning of treatment to improve response speed.15 Clonazepam and alprazolam are FDA-approved for treating PD. A high-potency, long-acting agent, clonazepam is the preferred initial benzodiazepine, dosed 0.5 to 4 mg/d on a fixed schedule. Although substantial data support using alprazolam for PD, it requires more frequent dosing and has a greater risk of rebound anxiety and abuse potential because of its more rapid onset of action. Compared with immediate-release alprazolam, alprazolam XR has a slower absorption rate and longer steady state in the blood, but this formulation does not have lower abuse potential or greater efficacy. Although not FDA-approved for PD, diazepam and lorazepam also have proven efficacy for PD.3

Benzodiazepines should be considered contraindicated in patients with a history of substance abuse, except in select cases.4 Benzodiazepines generally should be avoided in older patients because of increased risk for falls, cognitive impairment, and motor vehicle accidents. Table 2 lists situations in which benzodiazepines may be used to treat PD.

Table 2

Clinical scenarios in which to consider using benzodiazepines

Coadministration for 2 to 4 weeks when initiating treatment with an SSRI or venlafaxine XR to achieve more rapid relief and mitigate potential antidepressant-induced anxiety
For patients who wish to avoid antidepressants because of concern about sexual dysfunction
For patients who need chronic aspirin or an NSAID, which may increase the risk for upper gastrointestinal bleeding when taken in combination with an SSRI
For patients with comorbid bipolar disorder or epilepsy
Next-step monotherapy or augmentation in patients who respond poorly to an SSRI, venlafaxine XR, TCA, or CBT
CBT: cognitive-behavioral therapy; NSAID: nonsteroidal anti-inflammatory drug; SSRI: selective serotonin reuptake inhibitor; TCA: tricyclic antidepressant; XR: extended release

TCAs are effective as monotherapy for PD. Most support comes from studies of imipramine or clomipramine.12 Similar to SSRIs and venlafaxine XR, use a low initial dose and gradually increase until the patient remits or side effects prevent further increases. SSRI and TCA combinations rarely are used unless the TCA is a relatively specific norepinephrine reuptake inhibitor (eg, desipramine, nortriptyline). Because TCAs are metabolized via the cytochrome P450 2D6 system and some SSRIs—particularly fluoxetine and paroxetine—strongly inhibit 2D6, combinations of TCAs with these agents may lead to dangerously high plasma TCA levels, placing patients at risk for cardiac dysrhythmias and other side effects.16

Monoamine oxidase inhibitors (MAOIs)—particularly phenelzine—are underused for PD. They have the strongest efficacy data for any class of medications outside the first- and second-line agents and have a unique mechanism of action. In patients who can comply with the dietary and medication limitations, an MAOI generally should be the next step after nonresponse to other treatments.3

 

 

Alternative treatments

For patients who do not respond to any of the treatments described above, data from uncontrolled studies support mirtazapine, levetiracetam, and the serotonin-norepinephrine reuptake inhibitors duloxetine and milnacipran as monotherapy for PD.17 Pindolol—a beta blocker and 5-HT1A receptor antagonist—proved superior to placebo as an adjunctive agent to SSRIs in treatment-resistant PD in 1 of 2 trials.17 Minimal evidence supports the atypical antipsychotics risperidone and olanzapine in treatment-resistant PD, although a placebo-controlled trial of quetiapine SR coadministered with SSRIs recently was completed (NCT00619892; results pending). Atypical antipsychotics are best reserved for patients with a primary psychotic disorder or bipolar disorder who experience panic attacks.5

Panic-focused psychodynamic psychotherapy, a 12-week (approximately 24 sessions) form of psychotherapy, has demonstrated superiority vs applied relaxation therapy.18 This treatment could be considered for patients who do not respond to standard first-line treatments, but few community therapists are familiar with this method.

For many patients with PD, complementary and alternative medicine (CAM) approaches are appealing. See this article at CurrentPsychiatry.com for a Box that discusses CAM for PD.

Box

Complementary and alternative medicine for panic disorder

Although no complementary and alternative medicine treatments have strong evidence of efficacy as monotherapy for panic disorder (PD), several have data that suggest benefit with little evidence of risk. These include bibliotherapy, yoga, aerobic exercise, and the dietary supplements kava and inositol.a Exercise as a treatment poses a challenge because it can induce symptoms that the patient fears, such as tachycardia and shortness of breath. In addition to any direct physiologic benefit from aerobic exercise, there is also an exposure component that can be harnessed by gradually increasing the exertion level.

Another approach undergoing extensive evaluation is Internet-provided cognitive-behavioral therapy (CBT). Using guided CBT modules with or without therapist support, Internet-provided CBT provides an option for motivated patients unable to complete in-person CBT because of logistical factors.b A helpful resource that reviews Internet self-help and psychotherapy guided programs for PD and other psychiatric conditions is http://beacon.anu.edu.au.

References

a. Antonacci DJ, Davis E, Bloch RM, et al. CAM for your anxious patient: what the evidence says. Current Psychiatry. 2010;9(10):42-52.

b. Johnston L, Titov N, Andrews G, et al. A RCT of a transdiagnostic internet-delivered treatment for three anxiety disorders: examination of support roles and disorder-specific outcomes. PLoS One. 2011;6(11):e28079.

Maintenance treatment

Patients who complete a course of CBT for PD often follow up with several “booster sessions” at monthly or longer intervals that focus on relapse prevention techniques. Few controlled trials have evaluated pharmacotherapy discontinuation in PD. Most guidelines recommend continuing treatment for ≥1 year after achieving remission to minimize the risk of relapse.3 Researchers are focusing on whether medication dosage can be reduced during maintenance without loss of efficacy.

Treatment discontinuation

In the absence of urgent medical need, taper medications for PD gradually over several months. PD patients are highly sensitive to unusual physical sensations, which can occur while discontinuing antidepressants or benzodiazepines. If a benzodiazepine is used in conjunction with an antidepressant, the benzodiazepine should be discontinued first, so that the antidepressant can help ease benzodiazepine-associated discontinuation symptoms. A brief course of CBT during pharmacotherapy discontinuation may increase the likelihood of successful tapering.19

CASE CONTINUED: A successful switch

Ms. K has to discontinue sequential trials of fluoxetine, 40 mg/d, and venlafaxine XR, 225 mg/d because of side effects, and she does not reduce the frequency of her alprazolam use. She agrees to switch from alprazolam to clonazepam, 0.5 mg every morning and 1 mg at bedtime, and to start CBT. Clonazepam reduces her anxiety sufficiently so she can address her symptoms in therapy. Through CBT she becomes motivated to monitor her thoughts and treat them as guesses rather than facts, reviewing the evidence for her thoughts and generating rational responses. She participates in exposure exercises, which she practices between sessions, and grows to tolerate uncomfortable sensations until they no longer signal danger. After 12 CBT sessions, she is panic-free. Despite some trepidation, she agrees to a slow taper off clonazepam, reducing the dose by 0.25 mg every 2 weeks. She continues booster sessions with her therapist to manage any re-emerging anxiety. After an additional 12 weeks, she successfully discontinues clonazepam and remains panic-free.

Related Resources

Drug Brand Names

 

 

  • Alprazolam • Xanax
  • Alprazolam XR • Xanax XR
  • Citalopram • Celexa
  • Clomipramine • Anafranil
  • Clonazepam • Klonopin
  • Desipramine • Norpramin
  • Diazepam • Valium
  • Duloxetine • Cymbalta
  • Escitalopram • Lexapro
  • Fluoxetine • Prozac
  • Fluvoxamine • Luvox
  • Imipramine • Tofranil
  • Levetiracetam • Keppra
  • Lorazepam • Ativan
  • Milnacipran • Savella
  • Mirtazapine • Remeron
  • Nortriptyline • Aventyl, Pamelor
  • Olanzapine • Zyprexa
  • Paroxetine • Paxil
  • Paroxetine CR • Paxil CR
  • Phenelzine • Nardil
  • Pindolol • Visken
  • Quetiapine SR • Seroquel SR
  • Risperidone • Risperdal
  • Sertraline • Zoloft
  • Venlafaxine XR • Effexor XR

Disclosures

Dr. Dunlop receives research support from Bristol-Myers Squibb, GlaxoSmithKline, and the National Institute of Mental Health. He serves as a consultant to MedAvante and Roche.

Ms. Schneider and Dr. Gerardi report no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.


Dr. Dunlop: State-of-the-art treatment for panic disorder


Ms. K, a 24-year-old waitress who lives with her boyfriend, was referred by her primary care physician for evaluation of panic attacks that began “out of nowhere” at work approximately 6 months ago. The unpredictable attacks occur multiple times per week, causing her to leave work and cancel shifts.

Ms. K reports that before the panic attacks began, she felt happy in her relationship, enjoyed hobbies, and was hopeful about the future. However, she has become concerned that a potentially catastrophic illness is causing her panic attacks. She researches her symptoms on the Internet, and is preoccupied with the possibility of sudden death due to an undiagnosed heart condition. Multiple visits to the emergency room have not identified any physical abnormalities. Her primary care doctor prescribed alprazolam, 0.5 mg as needed for panic attacks, which she reports is helpful, “but only in the moment of the attacks.” Ms. K avoids alcohol and illicit substances and limits her caffeine intake. She is not willing to accept that her life “feels so limited.” Her dream of earning a nursing degree and eventually starting a family now seems unattainable.

Panic disorder (PD) occurs in 3% to 5% of adults, with women affected at roughly twice the rate of men.1 Causing a broad range of distress and varying degrees of impairment, PD commonly occurs with other psychiatric disorders. For most patients, treatment is effective, but those who do not respond to initial approaches require a thoughtful, stepped approach to care. Key considerations include establishing an accurate diagnosis, clarifying comorbid illnesses, ascertaining patient beliefs and expectations, and providing appropriately dosed and maintained treatments.

Panic attacks vs PD

Panic attacks consist of rapid onset of intense anxiety, with prominent somatic symptoms, that peaks within 10 minutes (Figure).2 Attacks in which <4 of the listed symptoms occur are considered limited-symptom panic attacks.


Figure: Body locations of panic attack symptoms
Diagnosis of a panic attack requires the sudden development of intense fear or discomfort characterized by ≥4 of the 13 symptoms listed above that peaks in intensity within 10 minutes of onset
Source: Reference 2

Panic attacks can occur with various disorders, including other anxiety disorders, mood disorders, and substance intoxication or withdrawal. Because serious medical conditions can present with panic-like symptoms, the initial occurrence of such symptoms warrants consideration of physiological causes. For a Box2 that describes the differential diagnosis of panic attacks, see this article at CurrentPsychiatry.com.

Box

Differential diagnosis of panic attacks

To meet diagnostic criteria for panic disorder, panic attacks must initially occur “out of the blue,” meaning no specific object or situation induced the attack. The differential diagnosis of panic attacks includes assessing for other psychiatric disorders that may involve panic attacks. Evaluation requires considering the context in which the panic attacks occur, including their start date, pattern of attacks, instigating situations, and associated thoughts.

Social phobia. Attacks occur only during or immediately before a social interaction in which the patient fears embarrassing himself or herself.

Obsessive-compulsive disorder (OCD). Attacks occur when the patient cannot avoid exposure to an obsessional fear or is prevented from performing a ritual that diffuses obsessional anxiety.

Posttraumatic stress disorder (PTSD). Attacks occur when confronted by a trauma-related memory or trigger.

Specific phobia. Attacks occur only when the patient encounters a specifically feared object, place, or situation, unrelated to social phobia, OCD, or PTSD.

Medical conditions. Conditions to consider include—but are not limited to—hyperthyroidism, pulmonary embolism, myocardial infarction, cardiac dysrhythmias, hypoglycemia, asthma, partial complex seizures, and pheochromocytoma.

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

A PD diagnosis requires that repeated panic attacks initially must occur from “out of the blue,” meaning no specific object or situation induced the attack. In addition, the diagnosis requires 1 of 3 types of psychological or behavioral changes as a result of the attacks (Table 1).2 Agoraphobia is diagnosed if 1 of the behavioral changes is avoidance of places or situations from which escape might be embarrassing or difficult should an attack occur. A patient can be diagnosed as having PD with agoraphobia, PD without agoraphobia, or agoraphobia without PD (ie, experiences only limited symptom panic attacks, but avoids situations or stimuli associated with them).

Table 1

Definitions of panic disorder and agoraphobia

Panic disorder
  1. Both 1 and 2:
  2. The panic attacks do not arise from the direct physiological effects of substance intoxication (eg, caffeine, cocaine) or withdrawal (eg, cannabis, gamma-hydroxybutyrate) or from a general medical condition
  3. The panic attacks are not better accounted for by another mental disorder
Agoraphobia
Anxiety about, or avoidance of, being in places or situations from which escape might be difficult or embarrassing, or in which help may not be available in the event of having an unexpected or situationally predisposed panic attack or panic-like symptoms. Agoraphobic fears typically involve characteristic clusters of situations that include being outside the home alone, being in a crowd, standing in a line, being on a bridge, or traveling in a bus, train, or automobile
Source: Reference 2
 

 

Comorbidities are common in patients with PD and predict greater difficulty achieving remission (Box).1,3-6

Box

Comorbidities: How they affect panic disorder treatment

The most common psychiatric conditions that co-occur with panic disorder (PD) are other anxiety disorders, mood disorders, personality disorders, and substance use disorders.1 Carefully assess the severity and degree of impairment or distress arising from each condition to prioritize treatment goals. For example, treating panic attacks would be a lower priority in a patient with untreated bipolar disorder.

Assessing comorbid substance abuse is important in selecting PD treatments. Benzodiazepines should almost always be avoided in patients with a history of drug abuse—illicit or prescribed. Although complete abstinence should not be a prerequisite for beginning PD treatment, detoxification and concomitant substance abuse treatment are essential.3

Comorbid mood disorders also affect the course of PD treatment. Antidepressants are effective for treating depression and PD, whereas benzodiazepines are not effective for depression.4 Antidepressants in patients with bipolar disorder are controversial because these medications might induce mixed or elevated mood states or rapid cycling. In these complicated patients, consider antidepressants lower in the treatment algorithm.5

Other conditions to consider before beginning treatment include pregnancy or the possibility of becoming pregnant in the near future and suicidal ideation. PD is associated with increased risk for suicidal ideation and progression to suicide attempts, particularly in patients with a comorbid mood or psychotic disorder.6 In addition, consider the potential impact of medications on comorbid medical conditions.

Treatment begins with education

The goal of treatment is remission of symptoms, ideally including an absence of panic attacks, agoraphobic avoidance, and anticipatory anxiety.1 The Panic Disorder Severity Scale self-report is a validated measure of panic symptoms that may be useful in clinical practice.7

The first step in treatment is educating patients about panic attacks, framing them as an overreactive fear circuit in the brain that produces physical symptoms that are not dangerous. Using a brain model that shows the location of the amygdala, hippocampus, and prefrontal cortex—which play crucial roles in generating and controlling anxiety and fear—can make this discussion more concrete.8 Although highly simplified, such models allow clinicians to demonstrate that excessive reactivity of limbic regions can be reduced by both top-down (cortico-limbic connections via cognitive-behavioral therapy [CBT]) and bottom-up (pharmacotherapy directly acting on limbic structures) approaches. Such discussions lead to treatment recommendations for CBT, pharmacotherapy, or their combination.

No single treatment has emerged as the definitive “best” for PD, and no reliable predictors can guide specific treatment for an individual.3 Combining CBT with pharmacotherapy produces higher short-term response rates than either treatment alone, but in the long term, combination treatment does not appear to be superior to CBT alone.9 Base the initial treatment selection for PD on patient preference, treatment availability and cost, and comorbid medical and psychiatric conditions. For an Algorithm to guide treatment decisions, see this article at CurrentPsychiatry.com.


Algorithm: Treatment for panic disorder: A suggested algorithm
aPoor response to an SSRI should lead to a switch to venlafaxine extended-release, and vice versa
bBenzodiazepines are relatively contraindicated in geriatric patients and patients with a history of substance abuse or dependence
CBT: cognitive-behavioral therapy; MAOI: monoamine oxidase inhibitor; SSRI: selective serotonin reuptake inhibitor; TCA: tricyclic antidepressant; Ven XR: venlafaxine extended-release

First-line treatments

Psychotherapy. CBT is the most efficacious psychotherapy for PD. Twelve to 15 sessions of CBT has demonstrated efficacy for PD, with additional effects on comorbid anxiety and depressive symptoms.10 No large clinical trials of CBT have used cognitive restructuring alone; all have included at least some component of exposure that requires the patient to confront feared physical sensations. Gains during treatment may be steady and gradual or sudden and uneven, with rapid improvement in some but not all symptoms. CBT and pharmacotherapy have demonstrated similar levels of benefit in short-term trials, but CBT has proven superior in most9 but not all11 trials evaluating long-term outcomes, particularly compared with pharmacotherapy that is discontinued during follow-up. Although less studied, group CBT also may be considered if a patient cannot afford individual CBT.

Pharmacotherapy. Evidence supports selective serotonin reuptake inhibitors (SSRIs), venlafaxine extended-release (XR), benzodiazepines, and tricyclic antidepressants (TCAs) as effective treatments for PD.3 No class of medication has demonstrated superiority over others in short-term treatment.3,12 Because of the medical risks associated with benzodiazepines and TCAs, an SSRI or venlafaxine XR should be the first medication option for most patients. Fluoxetine, paroxetine, sertraline, and venlafaxine XR are FDA-approved for PD. Paroxetine is associated with weight gain and may increase the risk for panic recurrence upon discontinuation more than sertraline, making it a less favorable option for many patients.13 Start doses at half the normal starting dose used for treating major depressive disorder and continue for 4 to 7 days, then increase to the minimal effective dose. For a Table3 that lists dosing recommendations for antidepressants to treat PD, see this article at CurrentPsychiatry.com. If there is no improvement by 4 weeks, increase the dose every 2 to 4 weeks until remission is achieved or side effects prevent further dose increases.

 

 

Table

Recommended doses for antidepressants used to treat panic disorder

MedicationStarting dose (mg/d)Therapeutic range (mg/d)
SSRIs
   Citalopram1020 to 40
   Escitalopram510 to 40
   Fluoxetine5 to 1020 to 80
   Fluvoxamine25100 to 300
   Paroxetine1020 to 80
   Paroxetine CR12.525 to 50
   Sertraline25100 to 200
SNRIs
   Duloxetine20 to 3060 to 120
   Venlafaxine XR37.5150 to 225
TCAs
   Clomipramine10 to 25100 to 300
   Imipramine10100 to 300
MAOI
   Phenelzine1545 to 90
CR: controlled release; MAOI: monoamine oxidase inhibitor; SNRIs: serotonin-norepinephrine reuptake inhibitors; SSRIs: selective serotonin reuptake inhibitors; TCAs: tricyclic antidepressants; XR: extended release
Source: American Psychiatric Association. Practice guideline for the treatment of patients with panic disorder. 2nd ed. Washington, DC: American Psychiatric Association; 2009

Treatment nonresponse. True non-response needs to be distinguished from poor response caused by inadequate treatment delivery, eg, patients not completing homework assignments in CBT or not adhering to pharmacotherapy. Asking patients about adverse effects or personal and family beliefs about treatment may reveal reasons for nonadherence.

Second-line treatments

Little data are available to guide next-step treatment options in patients who don’t achieve remission from their initial treatment. Patients who benefit from an SSRI, venlafaxine XR, or CBT but still have symptoms should be started on combination treatment. For a patient who experiences complete non-response to the initial treatment, discontinue the first treatment and switch to the other modality. In general, completely ineffective treatments should be discontinued when another treatment is added, but when partial improvement (>30%) occurs, continue the original treatment and augment it with another approach.

For patients pursuing pharmacotherapy, poor response to an adequate SSRI trial usually should lead to a switch to venlafaxine XR, and vice versa. Failure to respond to both of these medication classes should prompt a switch to a benzodiazepine or TCA.

Benzodiazepines are a fast-acting, effective treatment for PD, with efficacy similar to SSRIs in acute and long-term treatment.14 Benzodiazepines may be prescribed with antidepressants at the beginning of treatment to improve response speed.15 Clonazepam and alprazolam are FDA-approved for treating PD. A high-potency, long-acting agent, clonazepam is the preferred initial benzodiazepine, dosed 0.5 to 4 mg/d on a fixed schedule. Although substantial data support using alprazolam for PD, it requires more frequent dosing and has a greater risk of rebound anxiety and abuse potential because of its more rapid onset of action. Compared with immediate-release alprazolam, alprazolam XR has a slower absorption rate and longer steady state in the blood, but this formulation does not have lower abuse potential or greater efficacy. Although not FDA-approved for PD, diazepam and lorazepam also have proven efficacy for PD.3

Benzodiazepines should be considered contraindicated in patients with a history of substance abuse, except in select cases.4 Benzodiazepines generally should be avoided in older patients because of increased risk for falls, cognitive impairment, and motor vehicle accidents. Table 2 lists situations in which benzodiazepines may be used to treat PD.

Table 2

Clinical scenarios in which to consider using benzodiazepines

Coadministration for 2 to 4 weeks when initiating treatment with an SSRI or venlafaxine XR to achieve more rapid relief and mitigate potential antidepressant-induced anxiety
For patients who wish to avoid antidepressants because of concern about sexual dysfunction
For patients who need chronic aspirin or an NSAID, which may increase the risk for upper gastrointestinal bleeding when taken in combination with an SSRI
For patients with comorbid bipolar disorder or epilepsy
Next-step monotherapy or augmentation in patients who respond poorly to an SSRI, venlafaxine XR, TCA, or CBT
CBT: cognitive-behavioral therapy; NSAID: nonsteroidal anti-inflammatory drug; SSRI: selective serotonin reuptake inhibitor; TCA: tricyclic antidepressant; XR: extended release

TCAs are effective as monotherapy for PD. Most support comes from studies of imipramine or clomipramine.12 Similar to SSRIs and venlafaxine XR, use a low initial dose and gradually increase until the patient remits or side effects prevent further increases. SSRI and TCA combinations rarely are used unless the TCA is a relatively specific norepinephrine reuptake inhibitor (eg, desipramine, nortriptyline). Because TCAs are metabolized via the cytochrome P450 2D6 system and some SSRIs—particularly fluoxetine and paroxetine—strongly inhibit 2D6, combinations of TCAs with these agents may lead to dangerously high plasma TCA levels, placing patients at risk for cardiac dysrhythmias and other side effects.16

Monoamine oxidase inhibitors (MAOIs)—particularly phenelzine—are underused for PD. They have the strongest efficacy data for any class of medications outside the first- and second-line agents and have a unique mechanism of action. In patients who can comply with the dietary and medication limitations, an MAOI generally should be the next step after nonresponse to other treatments.3

 

 

Alternative treatments

For patients who do not respond to any of the treatments described above, data from uncontrolled studies support mirtazapine, levetiracetam, and the serotonin-norepinephrine reuptake inhibitors duloxetine and milnacipran as monotherapy for PD.17 Pindolol—a beta blocker and 5-HT1A receptor antagonist—proved superior to placebo as an adjunctive agent to SSRIs in treatment-resistant PD in 1 of 2 trials.17 Minimal evidence supports the atypical antipsychotics risperidone and olanzapine in treatment-resistant PD, although a placebo-controlled trial of quetiapine SR coadministered with SSRIs recently was completed (NCT00619892; results pending). Atypical antipsychotics are best reserved for patients with a primary psychotic disorder or bipolar disorder who experience panic attacks.5

Panic-focused psychodynamic psychotherapy, a 12-week (approximately 24 sessions) form of psychotherapy, has demonstrated superiority vs applied relaxation therapy.18 This treatment could be considered for patients who do not respond to standard first-line treatments, but few community therapists are familiar with this method.

For many patients with PD, complementary and alternative medicine (CAM) approaches are appealing. See this article at CurrentPsychiatry.com for a Box that discusses CAM for PD.

Box

Complementary and alternative medicine for panic disorder

Although no complementary and alternative medicine treatments have strong evidence of efficacy as monotherapy for panic disorder (PD), several have data that suggest benefit with little evidence of risk. These include bibliotherapy, yoga, aerobic exercise, and the dietary supplements kava and inositol.a Exercise as a treatment poses a challenge because it can induce symptoms that the patient fears, such as tachycardia and shortness of breath. In addition to any direct physiologic benefit from aerobic exercise, there is also an exposure component that can be harnessed by gradually increasing the exertion level.

Another approach undergoing extensive evaluation is Internet-provided cognitive-behavioral therapy (CBT). Using guided CBT modules with or without therapist support, Internet-provided CBT provides an option for motivated patients unable to complete in-person CBT because of logistical factors.b A helpful resource that reviews Internet self-help and psychotherapy guided programs for PD and other psychiatric conditions is http://beacon.anu.edu.au.

References

a. Antonacci DJ, Davis E, Bloch RM, et al. CAM for your anxious patient: what the evidence says. Current Psychiatry. 2010;9(10):42-52.

b. Johnston L, Titov N, Andrews G, et al. A RCT of a transdiagnostic internet-delivered treatment for three anxiety disorders: examination of support roles and disorder-specific outcomes. PLoS One. 2011;6(11):e28079.

Maintenance treatment

Patients who complete a course of CBT for PD often follow up with several “booster sessions” at monthly or longer intervals that focus on relapse prevention techniques. Few controlled trials have evaluated pharmacotherapy discontinuation in PD. Most guidelines recommend continuing treatment for ≥1 year after achieving remission to minimize the risk of relapse.3 Researchers are focusing on whether medication dosage can be reduced during maintenance without loss of efficacy.

Treatment discontinuation

In the absence of urgent medical need, taper medications for PD gradually over several months. PD patients are highly sensitive to unusual physical sensations, which can occur while discontinuing antidepressants or benzodiazepines. If a benzodiazepine is used in conjunction with an antidepressant, the benzodiazepine should be discontinued first, so that the antidepressant can help ease benzodiazepine-associated discontinuation symptoms. A brief course of CBT during pharmacotherapy discontinuation may increase the likelihood of successful tapering.19

CASE CONTINUED: A successful switch

Ms. K has to discontinue sequential trials of fluoxetine, 40 mg/d, and venlafaxine XR, 225 mg/d because of side effects, and she does not reduce the frequency of her alprazolam use. She agrees to switch from alprazolam to clonazepam, 0.5 mg every morning and 1 mg at bedtime, and to start CBT. Clonazepam reduces her anxiety sufficiently so she can address her symptoms in therapy. Through CBT she becomes motivated to monitor her thoughts and treat them as guesses rather than facts, reviewing the evidence for her thoughts and generating rational responses. She participates in exposure exercises, which she practices between sessions, and grows to tolerate uncomfortable sensations until they no longer signal danger. After 12 CBT sessions, she is panic-free. Despite some trepidation, she agrees to a slow taper off clonazepam, reducing the dose by 0.25 mg every 2 weeks. She continues booster sessions with her therapist to manage any re-emerging anxiety. After an additional 12 weeks, she successfully discontinues clonazepam and remains panic-free.

Related Resources

Drug Brand Names

 

 

  • Alprazolam • Xanax
  • Alprazolam XR • Xanax XR
  • Citalopram • Celexa
  • Clomipramine • Anafranil
  • Clonazepam • Klonopin
  • Desipramine • Norpramin
  • Diazepam • Valium
  • Duloxetine • Cymbalta
  • Escitalopram • Lexapro
  • Fluoxetine • Prozac
  • Fluvoxamine • Luvox
  • Imipramine • Tofranil
  • Levetiracetam • Keppra
  • Lorazepam • Ativan
  • Milnacipran • Savella
  • Mirtazapine • Remeron
  • Nortriptyline • Aventyl, Pamelor
  • Olanzapine • Zyprexa
  • Paroxetine • Paxil
  • Paroxetine CR • Paxil CR
  • Phenelzine • Nardil
  • Pindolol • Visken
  • Quetiapine SR • Seroquel SR
  • Risperidone • Risperdal
  • Sertraline • Zoloft
  • Venlafaxine XR • Effexor XR

Disclosures

Dr. Dunlop receives research support from Bristol-Myers Squibb, GlaxoSmithKline, and the National Institute of Mental Health. He serves as a consultant to MedAvante and Roche.

Ms. Schneider and Dr. Gerardi report no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.

References

1. Roy-Byrne PP, Craske MG, Stein MB. Panic disorder. Lancet. 2006;368(9540):1023-1032.

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

3. American Psychiatric Association. Practice guideline for the treatment of patients with panic disorder. 2nd ed. Washington DC: American Psychiatric Association; 2009.

4. Dunlop BW, Davis PG. Combination treatment with benzodiazepines and SSRIs for comorbid anxiety and depression: a review. Prim Care Companion J Clin Psychiatry. 2008;10(3):222-228.

5. Rakofsky JJ, Dunlop BW. Treating nonspecific anxiety and anxiety disorders in patients with bipolar disorder: a review. J Clin Psychiatry. 2011;72(1):81-90.

6. Sareen J, Cox BJ, Afifi TO, et al. Anxiety disorders and risk for suicidal ideation and suicide attempts: a population-based longitudinal study of adults. Arch Gen Psychiatry. 2005;62(11):1249-1257.

7. Houck PR, Spiegel DA, Shear MK, et al. Reliability of the self-report version of the panic disorder severity scale. Depress Anxiety. 2002;15(4):183-185.

8. Ninan PT, Dunlop BW. Neurobiology and etiology of panic disorder. J Clin Psychiatry. 2005;66(suppl 4):3-7.

9. Furukawa TA, Watanabe N, Churchill R. Psychotherapy plus antidepressant for panic disorder with or without agoraphobia: systematic review. Br J Psychiatry. 2006;188:305-312.

10. Barlow DH, Gorman JM, Shear MK, et al. Cognitive-behavioral therapy, imipramine, or their combination for panic disorder: a randomized controlled trial. JAMA. 2000;283(19):2529-2536.

11. van Apeldoorn FJ, Timmerman ME, Mersch PP, et al. A randomized trial of cognitive-behavioral therapy or selective serotonin reuptake inhibitor or both combined for panic disorder with or without agoraphobia: treatment results through 1-year follow-up. J Clin Psychiatry. 2010;71(5):574-586.

12. Bakker A, van Balkom AJ, Spinhoven P. SSRIs vs. TCAs in the treatment of panic disorder: a meta-analysis. Acta Psychiatr Scand. 2002;106(3):163-167.

13. Bandelow B, Behnke K, Lenoir S, et al. Sertraline versus paroxetine in the treatment of panic disorder: an acute, double-blind noninferiority comparison. J Clin Psychiatry. 2004;65(3):405-413.

14. Nardi AE, Freire RC, Mochcovitch MD, et al. A randomized, naturalistic, parallel-group study for the long-term treatment of panic disorder with clonazepam or paroxetine. J Clin Psychopharmacol. 2012;32(1):120-126.

15. Goddard AW, Brouette T, Almai A, et al. Early coadministration of clonazepam with sertraline for panic disorder. Arch Gen Psychiatry. 2001;58(7):681-686.

16. Preskorn SH, Shah R, Neff M, et al. The potential for clinically significant drug-drug interactions involving the CYP 2D6 system: effects with fluoxetine and paroxetine versus sertraline. J Psychiatr Pract. 2007;13(1):5-12.

17. Perna G, Guerriero G, Caldirola D. Emerging drugs for panic disorder. Expert Opin Emerg Drugs. 2011;16(4):631-645.

18. Milrod B, Leon AC, Busch F, et al. A randomized controlled clinical trial of psychoanalytic psychotherapy for panic disorder. Am J Psychiatry. 2007;164(2):265-272.

19. Otto MW, Pollack MH, Sachs GS, et al. Discontinuation of benzodiazepine treatment: efficacy of cognitive-behavioral therapy for patients with panic disorder. Am J Psychiatry. 1993;150(10):1485-1490.

References

1. Roy-Byrne PP, Craske MG, Stein MB. Panic disorder. Lancet. 2006;368(9540):1023-1032.

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

3. American Psychiatric Association. Practice guideline for the treatment of patients with panic disorder. 2nd ed. Washington DC: American Psychiatric Association; 2009.

4. Dunlop BW, Davis PG. Combination treatment with benzodiazepines and SSRIs for comorbid anxiety and depression: a review. Prim Care Companion J Clin Psychiatry. 2008;10(3):222-228.

5. Rakofsky JJ, Dunlop BW. Treating nonspecific anxiety and anxiety disorders in patients with bipolar disorder: a review. J Clin Psychiatry. 2011;72(1):81-90.

6. Sareen J, Cox BJ, Afifi TO, et al. Anxiety disorders and risk for suicidal ideation and suicide attempts: a population-based longitudinal study of adults. Arch Gen Psychiatry. 2005;62(11):1249-1257.

7. Houck PR, Spiegel DA, Shear MK, et al. Reliability of the self-report version of the panic disorder severity scale. Depress Anxiety. 2002;15(4):183-185.

8. Ninan PT, Dunlop BW. Neurobiology and etiology of panic disorder. J Clin Psychiatry. 2005;66(suppl 4):3-7.

9. Furukawa TA, Watanabe N, Churchill R. Psychotherapy plus antidepressant for panic disorder with or without agoraphobia: systematic review. Br J Psychiatry. 2006;188:305-312.

10. Barlow DH, Gorman JM, Shear MK, et al. Cognitive-behavioral therapy, imipramine, or their combination for panic disorder: a randomized controlled trial. JAMA. 2000;283(19):2529-2536.

11. van Apeldoorn FJ, Timmerman ME, Mersch PP, et al. A randomized trial of cognitive-behavioral therapy or selective serotonin reuptake inhibitor or both combined for panic disorder with or without agoraphobia: treatment results through 1-year follow-up. J Clin Psychiatry. 2010;71(5):574-586.

12. Bakker A, van Balkom AJ, Spinhoven P. SSRIs vs. TCAs in the treatment of panic disorder: a meta-analysis. Acta Psychiatr Scand. 2002;106(3):163-167.

13. Bandelow B, Behnke K, Lenoir S, et al. Sertraline versus paroxetine in the treatment of panic disorder: an acute, double-blind noninferiority comparison. J Clin Psychiatry. 2004;65(3):405-413.

14. Nardi AE, Freire RC, Mochcovitch MD, et al. A randomized, naturalistic, parallel-group study for the long-term treatment of panic disorder with clonazepam or paroxetine. J Clin Psychopharmacol. 2012;32(1):120-126.

15. Goddard AW, Brouette T, Almai A, et al. Early coadministration of clonazepam with sertraline for panic disorder. Arch Gen Psychiatry. 2001;58(7):681-686.

16. Preskorn SH, Shah R, Neff M, et al. The potential for clinically significant drug-drug interactions involving the CYP 2D6 system: effects with fluoxetine and paroxetine versus sertraline. J Psychiatr Pract. 2007;13(1):5-12.

17. Perna G, Guerriero G, Caldirola D. Emerging drugs for panic disorder. Expert Opin Emerg Drugs. 2011;16(4):631-645.

18. Milrod B, Leon AC, Busch F, et al. A randomized controlled clinical trial of psychoanalytic psychotherapy for panic disorder. Am J Psychiatry. 2007;164(2):265-272.

19. Otto MW, Pollack MH, Sachs GS, et al. Discontinuation of benzodiazepine treatment: efficacy of cognitive-behavioral therapy for patients with panic disorder. Am J Psychiatry. 1993;150(10):1485-1490.

Issue
Current Psychiatry - 11(11)
Issue
Current Psychiatry - 11(11)
Page Number
36-47
Page Number
36-47
Publications
Publications
Topics
Article Type
Display Headline
Panic disorder: Break the fear circuit
Display Headline
Panic disorder: Break the fear circuit
Legacy Keywords
panic disorder; fear; cognitive-behavioral therapy; antidepressants; panic attacks
Legacy Keywords
panic disorder; fear; cognitive-behavioral therapy; antidepressants; panic attacks
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media

Differentiating Alzheimer’s disease from dementia with Lewy bodies

Article Type
Changed
Tue, 12/11/2018 - 15:03
Display Headline
Differentiating Alzheimer’s disease from dementia with Lewy bodies

Discuss this article at www.facebook.com/CurrentPsychiatry

Alzheimer’s disease (AD) and dementia with Lewy bodies (DLB) are the first and second most common causes of neurodegenerative dementia, respectively.“New Alzheimer’s disease guidelines: Implications for clinicians,” Current Psychiatry, March 2012, p. 15-20; http://bit.ly/UNYikk.

The 2005 report of the DLB Consortium5 recognizes central, core, suggestive, and supportive features of DLB (Table 1).5,10 These features are considered in the context of other confounding clinical conditions and the timing of cognitive and motor symptoms. The revised DLB criteria5 require a central feature of progressive cognitive decline. “Probable DLB” is when a patient presents with 2 core features or 1 core feature and ≥1 suggestive features. A diagnosis of “possible DLB” requires 1 core feature or 1 suggestive feature in the presence of progressive cognitive decline.

Table 1

Diagnostic criteria for AD and DLB

NIA-AA criteria for AD (2011)10
Possible AD: Clinical and cognitive criteria (DSM-IV-TR) for AD are met and there is an absence of biomarkers to support the diagnosis or there is evidence of a secondary disorder that can cause dementia
Probable AD: Clinical and cognitive criteria for AD are met and there is documented progressive cognitive decline or abnormal biomarker(s) suggestive of AD or evidence of proven AD autosomal dominant genetic mutation (presenilin-1, presenilin-2, amyloid-β precursor protein)
Definite AD: Clinical criteria for probable AD are met and there is histopathologic evidence of the disorder
Revised clinical diagnostic criteria for DLB (2005)5
Core features: Fluctuating cognition, recurrent visual hallucinations, soft motor features of parkinsonism
Suggestive features: REM sleep behavior disorder, severe antipsychotic sensitivity, decreased tracer uptake in striatum on SPECT dopamine transporter imaging or on myocardial scintigraphy with MIBG
Supportive features (common but lacking diagnostic specificity): repeated falls and syncope; transient, unexplained loss of consciousness; systematized delusions; hallucinations other than visual; relative preservation of medial temporal lobe on CT or MRI scan; decreased tracer uptake on SPECT or PET imaging in occipital regions; prominent slow waves on EEG with temporal lobe transient sharp waves
AD: Alzheimer’s disease; DLB: dementia with Lewy bodies; MIBG: metaiodobenzylguanidine; NIA-AA: National Institute on Aging and the Alzheimer’s Association; PET: positron emission tomography; REM: rapid eye movement; SPECT: single photon emission computed tomography

Biomarkers for AD, but not DLB

The 2011 diagnostic criteria for AD incorporate biomarkers that can be measured in vivo and reflect speci?c features of disease-related pathophysiologic processes. Biomarkers for AD are divided into 2 categories:11

  • amyloid-beta (Aβ) accumulation: abnormal tracer retention on amyloid positron emission topography (PET) imaging and low cerebrospinal fluid (CSF) Aβ42
  • neuronal degeneration or injury: elevated CSF tau (total and phosphorylated tau), decreased ?uorodeoxyglucose uptake on PET in temporo-parietal cortices, and atrophy on structural MRI in the hippocampal and temporo-parietal regions.

No clinically applicable genotypic or CSF markers exist to support a DLB diagnosis, but there are many promising candidates, including elevated levels of CSF p-tau 181, CSF levels of alpha- and beta-synuclein,12 and CSF beta-glucocerebrosidase levels.13 PET mapping of brain acetylcholinesterase activity,14 123I-2β-carbomethoxy-3β- (4-iodophenyl)-N-(3-fluoropropyl)nortropane single photon emission computed tomography (SPECT) dopamine transporter (DaT) imaging15 and metaiodobenzylguanidine (MIBG) scintigraphy also are promising methods. DaT scan SPECT is FDA-approved for detecting loss of functional dopaminergic neuron terminals in the striatum and can differentiate between AD and DLB with a sensitivity and specificity of 78% to 88% and 94% to 100%, respectively.16 This test is covered by Medicare for differentiating AD and DLB.

Differences in presentation

Cognitive impairment. Contrary to the early memory impairment that characterizes AD, memory deficits in DLB usually appear later in the disease course.5 Patients with DLB manifest greater attentional, visuospatial, and executive impairments than those with AD, whereas AD causes more profound episodic (declarative) memory impairment than DLB. DLB patients show more preserved consolidation and storage of verbal information than AD patients because of less neuroanatomical and cholinergic compromise in the medial temporal lobe. There is no evidence of significant differences in remote memory, semantic memory, and language (naming and fluency).

Compromised attention in DLB may be the basis for fluctuating cognition, a characteristic of the disease. The greater attentional impairment and reaction time variability in DLB compared with AD is evident during complex tasks for attention and may be a function of the executive and visuospatial demands of the tasks.17

Executive functions critical to adaptive, goal-directed behavior are more impaired in DLB than AD. DLB patients are more susceptible to distraction and have difficulty engaging in a task and shifting from 1 task to another. This, together with a tendency for confabulation and perseveration, are signs of executive dysfunction.

 

 

Neuropsychiatric features. DLB patients are more likely than AD patients to exhibit psychiatric symptoms and have more functional impairment.18 In an analysis of autopsy-confirmed cases, hallucinations and delusions were more frequent with Lewy body pathology (75%) than AD (21%) at initial clinical evaluation.18 By the end stages of both illnesses, the degree of psychotic symptoms is comparable.19 Depression is common in DLB; whether base rates of depressed mood and major depression differ between DLB and AD is uncertain.20

Psychosis in AD can be induced by medication or delirium, or triggered by poor sensory perceptions. Psychotic symptoms occur more frequently during the moderate and advanced stages of AD, when patients present with visual hallucinations, delusions, or delusional misidentifications. As many as 10% to 20% of patients with AD experience hallucinations, typically visual. Delusions occur in 30% to 50% of AD patients, usually in the later stages of the disease. The most common delusional themes are infidelity, theft, and paranoia. Female sex is a risk factor for psychosis in AD. Delusions co-occur with aggression, anxiety, and aberrant motor behavior.

Visual hallucinations—mostly vivid, well-formed, false perceptions of insects, animals, or people—are the defining feature of DLB.21 Many patients recognize that they are experiencing visual hallucinations and can ignore them. DLB patients also may experience visual illusions, such as misperceiving household objects as living beings. Delusions—typically paranoid—are common among DLB patients, as are depression and anxiety.1 Agitation or aggressive behavior tends to occur late in the illness, if at all.

The causes of psychotic symptoms in DLB are not fully understood, but dopamine dysfunction likely is involved in hallucinations, delusions, and agitation, and serotonin dysfunction may be associated with depression and anxiety. Rapid eye movement (REM) sleep/wakefulness dysregulation, in which the dream imagery of REM sleep may occur during wakefulness, also has been proposed as a mechanism for visual hallucinations in DLB.22 In DLB, psychotic symptoms occur early and are a hallmark of this illness, whereas in AD they usually occur in the middle to late stages of the disease.

Motor symptoms. In AD, extrapyramidal symptoms (EPS) are common later in the disease, are strongly correlated with disease severity, and are a strong, independent predictor of depression severity.23 EPS are more common in DLB than in AD24 and DLB patients are at higher risk of developing EPS even with low doses of typical antipsychotics, compared with AD patients.25

Other symptoms. REM sleep behavior disorder (RBD) is characterized by enacting dreams—often violent—during REM sleep. RBD is common in DLB and many patients also have excessive daytime somnolence. Other sleep disorders in DLB include insomnia, obstructive sleep apnea, central sleep apnea, restless legs syndrome, and periodic limb movements during sleep.

In AD patients, common sleep behaviors include confusion in the early evening (“sundowning”) and frequent nighttime awakenings, often accompanied by wandering.26 Orthostatic hypotension, impotence, urinary incontinence, and constipation are common in DLB. Lack of insight concerning personal cognitive, mood, and behavioral state is highly prevalent in AD patients and more common than in DLB.

Diagnostic evaluation

Because there are no definitive clinical markers for DLB, diagnosis is based on a detailed clinical and family history from the patient and a reliable informant, as well as a physical, neurologic, and mental status examination that looks for associated noncognitive symptoms, and neuropsychological evaluation. Reasons DLB may be misdiagnosed include:

  • Some “core” clinical features of DLB may not appear or may overlap with AD.
  • Presence and severity of concurrent AD pathology in DLB may modify the clinical presentation, with decreased rates of hallucinations and parkinsonism and increased neurofibrillary tangles.
  • Failure to reliably identify fluctuations—variations in cognition and arousal, such as periods of unresponsiveness while awake (“zoning out”), excessive daytime somnolence, and disorganized speech.27

Detecting and characterizing cognitive deficits in dementia patients using neuropsychological testing is important in establishing a clinical diagnosis, determining baseline levels of impairment, forming a prognosis, and initiating disease-specific treatments. Differences in neuropsychological findings in AD and DLB are outlined in Table 2.16,28-33 Several studies have suggested using these measures to differentiate patients with DLB from those with AD.20

Table 2

Diagnostic testing for Alzheimer’s disease and dementia with Lewy bodies

Alzheimer’s diseaseDementia with Lewy bodies
Neuropsychological testing findings
Relatively more impairment on verbal memory tasks, delayed recall, delayed recognition, and encoding and storing information.28 Dysfunction of episodic memory functionRelatively more impairment on attention or concentration, verbal fluency, visuoperceptual, visuoconstructive, visual memory tests, and frontal executive functions.28 Relatively preserved confrontation naming and verbal memory
MRI findings
Diffuse cortical atrophy, relatively greater volume loss in hippocampus and medial temporal lobe structures (strong correlation with severity)29Mild generalized cerebral cortical atrophy with minimal hippocampal atrophy and relative preservation of medial temporal lobe structures30
[18F]FDG PET
Widespread metabolic deficits in neocortical association areas, with sparing of the basal ganglia, thalamus, cerebellum, primary sensory motor cortex, and visual cortexWidespread cortical hypometabolism, more marked in primary visual and occipital association areas, and less severe in parietal, frontal, and anterior cingulate cortices.31 Severe cholinergic deafferentation of the neocortex, particularly in posterior cortical regions32
Single photon emission computed tomography
Parietotemporal hypoperfusionOccipital hypoperfusion
123I-FP-CIT SPECT (DaT scan)
No significant loss of DaTLow nigrostriatal terminal density of DaT caused by severe nigrostriatal degeneration16
Myocardial scintigraphy with MIBG
No significant change in MIBG uptakeDecreased MIBG uptake33
123I-FP-CIT: 123I-2β-carbomethoxy-3β-(4-iodophenyl)-N-(3-fluoropropyl)nortropane; DaT: dopamine transporter; FDG PET: [18F]-fluoro-d-glucose positron emission tomography; MIBG: metaiodobenzylguanidine; SPECT: single photon emission computed tomography
 

 

Evidence is insufficient to support using electroencephalographic and polysomnographic studies when initially evaluating patients with dementia. Brain CT or MRI are recommended as part of the initial evaluation of dementia patients to exclude treatable causes of dementia and help clarify the differential diagnosis. Occipital hypometabolism and hypoperfusion demonstrated on PET and SPECT imaging have high sensitivity and specificity for differentiating AD from DLB.

To diagnose DLB more consistently, look for core features of the disease, RBD, antipsychotic hypersensitivity, and decreased striatal binding at presynaptic DaT sites.15 Abnormal (low binding) DaT activity is the most reliable diagnostic marker for DLB.34 Myocardial scintigraphy with MIBG is sensitive to pathologic changes of DLB before clinical expression and could overcome the difficulties of using clinical criteria alone to identify patients with DLB.35 MIBG scintigraphy may be preferred to DaT scan because it is less expensive and its sensitivity and specificity to DLB are independent of the presence of parkinsonism.35

For an overview of pharmacotherapy options for patients with AD or DLB, see Box 2.

Box 2

Treatments for Alzheimer’s disease and dementia with Lewy bodies

Pharmacotherapy options for patients with Alzheimer’s disease (AD) or dementia with Lewy bodies (DLB) include cholinesterase inhibitors, memantine, antipsychotics, and other agents.

Cholinesterase inhibitors. Donepezil, rivastigmine, and galantamine are FDA-approved for treating AD. Their efficacy appears to be similar, so the choice of agent is based largely on cost, patient tolerability, and physician experience.

No medications are FDA-approved for treating DLB. Neocortical cholinergic activity assessed by choline acetyltransferase levels is more severely depleted in DLB than in AD, and this deficit is correlated with the presence of visual hallucinations and global severity of cognitive impairment.a Therefore, drugs that enhance central cholinergic function offer a therapeutic approach for DLB; cognitive and hallucinatory symptoms are the anticipated targets. Multiple anecdotal reports, open-label studies,b,c and 1 randomized, placebo-controlled triald suggest that cholinesterase inhibitors are efficacious in DLB, with reported benefits in cognition, fluctuations, psychotic symptoms, and parkinsonian symptoms. A 20-week randomized, double-blind, placebo-controlled multicenter studyd of patients with DLB found rivastigmine, 6 to 12 mg/d, was superior to placebo. Patients receiving rivastigmine exhibited significantly reduced anxiety, delusions, and hallucinations and significantly better performance on a computerized battery of neuropsychological tests, especially tasks that required sustained attention. Differences between rivastigmine and placebo disappeared after drug discontinuation.

Memantine is a noncompetitive antagonist of N-methyl-d-aspartate receptors that is effective in AD.e The possible involvement of glutamate in DLB has provided a rationale for treating DLB with memantine. Two randomized controlled trials in DLB found that patients treated with memantine for 24 weeks performed better on Clinical Global Impression of Change, but not on most other secondary outcome measures.f,g In both studies, memantine was well tolerated. However, other studies have noted worsening of delusions and hallucinations with memantine in DLB patients.h

Antipsychotics. Agitation is common in moderate and advanced AD. Atypical antipsychotics have been used with variable efficacy to treat agitation, but their use is associated with excess mortality. DLB patients pose a considerable therapeutic challenge because antipsychotics—the mainstay of treatment of psychosis and behavioral problems in most other disorders—can provoke severe, irreversible, and often fatal sensitivity reactions in this type of dementia.i A 2- to 3-fold increased mortality risk associated with antipsychotic sensitivity reactions in DLB is partly mediated via acute blockade of postsynaptic dopamine D2 receptors in the striatum. For severe and disabling psychosis, a trial of a cholinesterase inhibitor and/or lowering the dose of antiparkinsonian medication should be considered first. In urgent situations, small doses of an atypical antipsychotic that is least associated with parkinsonism side effects—such as quetiapine or aripiprazole—should be used.

Other treatments. Treatment of parkinsonian symptoms in DLB patients is similar to that for Parkinson’s disease, but the risk of psychotic symptoms in DLB warrants a conservative approach. Levodopa seems to be more effective than dopamine agonists and produces fewer side effects.j Rapid eye movement sleep behavior disorder often responds to low doses of clonazepam (0.25 to 1.5 mg). Depression and anxiety disorders are common in AD at all stages and their treatment is not fundamentally different than in geriatric patients without dementia. Selective serotonin reuptake inhibitors and electroconvulsive therapy have been used successfully in depressed patients with AD or DLB.k,l

Disease-modifying treatments. Researchers are evaluating an array of antiamyloid and neuroprotective therapeutic approaches for AD based on the hypothesis that amyloid-beta protein plays a pivotal role in disease onset and progression. Interventions that reduce amyloid production, limit aggregation, or increase clearance may block the cascade of events comprising AD pathogenesis. Reducing tau hyperphosphorylation, limiting oxidation and excitotoxicity, and controlling inflammation also may be beneficial strategies. Potentially neuroprotective and restorative treatments such as neurotrophins, neurotrophic factor enhancers, and stem cell-related approaches also are being investigated.

There are no large-scale studies of disease-modifying treatments for DLB. Potential areas of research include the relationship between proteasome function and a-synuclein pathology, the role of beta-synuclein, and the impact of alterations to alpha-synuclein on its propensity to aggregate.

References

a. Ballard C, Ziabreva I, Perry R, et al. Differences in neuropathologic characteristics across the Lewy body dementia spectrum. Neurology. 2006;67(11):1931-1934.

b. Beversdorf DQ, Warner JL, Davis RA, et al. Donepezil in the treatment of dementia with lewy bodies. Am J Geriatr Psychiatry. 2004;12(5):542-544.

c. Edwards K, Royall D, Hershey L, et al. Efficacy and safety of galantamine in patients with dementia with Lewy bodies: a 24-week open-label study. Dement Geriatr Cogn Disord. 2007;23(6):401-405.

d. McKeith I, Del Ser T, Spano P, et al. Efficacy of rivastigmine in dementia with Lewy bodies: a randomised, double-blind, placebo-controlled international study. Lancet. 2000;356(9247):2031-2036.

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

f. Aarsland D, Ballard C, Walker Z, et al. Memantine in patients with Parkinson’s disease dementia or dementia with Lewy bodies: a double-blind, placebo-controlled, multicentre trial. Lancet Neurol. 2009;8(7):613-618.

g. Emre M, Tsolaki M, Bonuccelli U, et al. Memantine for patients with Parkinson’s disease dementia or dementia with Lewy bodies: a randomised, double-blind, placebo-controlled trial. Lancet Neurol. 2010;9(10):969-977.

h. Ridha BH, Josephs KA, Rossor MN. Delusions and hallucinations in dementia with Lewy bodies: worsening with memantine. Neurology. 2005;65(3):481-482.

i. McKeith I, Fairbairn A, Perry R, et al. Neuroleptic sensitivity in patients with senile dementia of Lewy body type. BMJ. 1992;305(6855):673-678.

j. Fernandez HH, Wu CK, Ott BR. Pharmacotherapy of dementia with Lewy bodies. Expert Opin Pharmacother. 2003;4(11):2027-2037.

k. Swartz M, Barak Y, Mirecki I, et al. Treating depression in Alzheimer’s disease: integration of differing guidelines. Int Psychogeriatr. 2000;12(3):353-358.

l. Takahashi S, Mizukami K, Yasuno F, et al. Depression associated with dementia with Lewy bodies (DLB) and the effect of somatotherapy. Psychogeriatrics. 2009;9(2):56-61.

 

 

Related Resources

  • Hanyu H, Sato T, Hirao K, et al. Differences in clinical course between dementia with Lewy bodies and Alzheimer’s disease. Eur J Neurol. 2009;16(2):212-217.
  • Walker Z, McKeith I, Rodda J, et al. Comparison of cognitive decline between dementia with Lewy bodies and Alzheimer’s disease: a cohort study. BMJ Open. 2012;2:e000380.

Drug Brand Names

  • Aripiprazole • Abilify
  • Clonazepam • Klonopin
  • Donepezil • Aricept
  • Galantamine • Razadyne, Reminyl
  • Levodopa • Dopar, Larodopa
  • Memantine • Namenda
  • Quetiapine • Seroquel
  • Rivastigmine • Exelon

Disclosure

Drs. Bishnoi and Manepalli report no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.

Dr. Grossberg serves as a consultant to Forest, Janssen, Novartis, and Pfizer. His department receives research funding from Novartis, Janssen, and Pfizer.

References

1. McKeith IG, Galasko D, Kosaka K, et al. Consensus guidelines for the clinical and pathologic diagnosis of dementia with Lewy bodies (DLB): report of the consortium on DLB international workshop. Neurology. 1996;47(5):1113-1124.

2. Buracchio T, Arvanitakis Z, Gorbien M. Dementia with Lewy bodies: current concepts. Dement Geriatr Cogn Disord. 2005;20(5):306-320.

3. Fujishiro H, Iseki E, Higashi S, et al. Distribution of cerebral amyloid deposition and its relevance to clinical phenotype in Lewy body dementia. Neurosci Lett. 2010;486(1):19-23.

4. Kosaka K. Diffuse Lewy body disease. Neuropathology. 2000;20(suppl):S73-S78.

5. McKeith IG, Dickson DW, Lowe J, et al. Consortium on DLB. Diagnosis and management of dementia with Lewy bodies: third report of the DLB Consortium. Neurology. 2005;65(12):1863-1872.

6. Cummings JL, Cole G. Alzheimer disease. JAMA. 2002;287(18):2335-2338.

7. Zaccai J, McCracken C, Brayne C. A systematic review of prevalence and incidence studies of dementia with Lewy bodies. Age Ageing. 2005;34(6):561-566.

8. Bradshaw J, Saling M, Hopwood M, et al. Fluctuating cognition in dementia with Lewy bodies and Alzheimer’s disease is qualitatively distinct. J Neurol Neurosurg Psychiatry. 2004;75(3):382-387.

9. Singleton AB, Wharton A, O’Brien KK, et al. Clinical and neuropathological correlates of apolipoprotein E genotype in dementia with Lewy bodies. Dement Geriatr Cogn Disord. 2002;14(4):167-175.

10. McKhann GM, Knopman DS, Chertkow H, et al. The diagnosis of dementia due to Alzheimer’s disease: recommendations from the National Institute on Aging-Alzheimer’s Association workgroups on diagnostic guidelines for Alzheimer’s disease. Alzheimers Dement. 2011;7(3):263-269.

11. Jack CR, Jr, Albert MS, Knopman DS, et al. Introduction to the recommendations from the National Institute on Aging-Alzheimer’s Association workgroups on diagnostic guidelines for Alzheimer’s disease. Alzheimers Dement. 2011;7(3):257-262.

12. Mollenhauer B, Cullen V, Kahn I, et al. Direct quantification of CSF alpha-synuclein by ELISA and first cross-sectional study in patients with neurodegeneration. Exp Neurol. 2008;213(2):315-325.

13. Parnetti L, Balducci C, Pierguidi L, et al. Cerebrospinal fluid beta-glucocerebrosidase activity is reduced in dementia with Lewy bodies. Neurobiol Dis. 2009;34(3):484-486.

14. Shimada H, Hirano S, Shinotoh H, et al. Mapping of brain acetylcholinesterase alterations in Lewy body disease by PET. Neurology. 2009;73(4):273-278.

15. McKeith I, O’Brien J, Walker Z, et al. Sensitivity and specificity of dopamine transporter imaging with 123I-FP-CIT SPECT in dementia with Lewy bodies: a phase III, multicentre study. Lancet Neurol. 2007;6(4):305-313.

16. Walker Z, Jaros E, Walker RW, et al. Dementia with Lewy bodies: a comparison of clinical diagnosis, FP-CIT single photon emission computed tomography imaging and autopsy. J Neurol Neurosurg Psychiatry. 2007;78(11):1176-1181.

17. Bradshaw JM, Saling M, Anderson V, et al. Higher cortical deficits influence attentional processing in dementia with Lewy bodies, relative to patients with dementia of the Alzheimer’s type and controls. J Neurol Neurosurg Psychiatry. 2006;77(10):1129-1135.

18. Weiner MF, Hynan LS, Parikh B, et al. Can Alzheimer’s disease and dementias with Lewy bodies be distinguished clinically? J Geriatr Psychiatry Neurol. 2003;16(4):245-250.

19. Stavitsky K, Brickman AM, Scarmeas N, et al. The progression of cognition, psychiatric symptoms, and functional abilities in dementia with Lewy bodies and Alzheimer disease. Arch Neurol. 2006;63(10):1450-1456.

20. Ferman TJ, Smith GE, Boeve BF, et al. Neuropsychological differentiation of dementia with Lewy bodies from normal aging and Alzheimer’s disease. Clin Neuropsychol. 2006;20(4):623-636.

21. McKeith IG, Perry EK, Perry RH. Report of the second dementia with Lewy body international workshop: diagnosis and treatment. Consortium on Dementia with Lewy Bodies. Neurology. 1999;53(5):902-905.

22. Boeve BF, Silber MH, Ferman TJ, et al. Association of REM sleep behavior disorder and neurodegenerative disease may reflect an underlying synucleinopathy. Mov Disord. 2001;16(4):622-630.

23. Portet F, Scarmeas N, Cosentino S, et al. Extrapyramidal signs before and after diagnosis of incident Alzheimer disease in a prospective population study. Arch Neurol. 2009;66(9):1120-1126.

24. McKeith I, Fairbairn A, Perry R, et al. Neuroleptic sensitivity in patients with senile dementia of Lewy body type. BMJ. 1992;305(6855):673-678.

25. Tarawneh R, Galvin JE. Distinguishing Lewy body dementias from Alzheimer’s disease. Expert Rev Neurother. 2007;7(11):1499-1516.

26. Ancoli-Israel S, Klauber MR, Gillin JC, et al. Sleep in non-institutionalized Alzheimer’s disease patients. Aging (Milano). 1994;6(6):451-458.

27. Ferman TJ, Smith GE, Boeve BF, et al. DLB fluctuations: specific features that reliably differentiate DLB from AD and normal aging. Neurology. 2004;62(2):181-187.

28. Salmon DP, Galasko D, Hansen LA, et al. Neuropsychological deficits associated with diffuse Lewy body disease. Brain Cogn. 1996;31(2):148-165.

29. Jack CR, Jr, Petersen RC, Xu Y, et al. Rates of hippocampal atrophy correlate with change in clinical status in aging and AD. Neurology. 2000;55(4):484-489.

30. Burton EJ, Barber R, Mukaetova-Ladinska EB, et al. Medial temporal lobe atrophy on MRI differentiates Alzheimer’s disease from dementia with Lewy bodies and vascular cognitive impairment: a prospective study with pathological verification of diagnosis. Brain. 2009;132(pt 1):195-203.

31. Ishii K, Soma T, Kono AK, et al. Comparison of regional brain volume and glucose metabolism between patients with mild dementia with lewy bodies and those with mild Alzheimer’s disease. J Nucl Med. 2007;48(5):704-711.

32. Klein JC, Eggers C, Kalbe E, et al. Neurotransmitter changes in dementia with Lewy bodies and Parkinson disease dementia in vivo. Neurology. 2010;74(11):885-892.

33. Fujishiro H, Nakamura S, Kitazawa M, et al. Early detection of dementia with Lewy bodies in patients with amnestic mild cognitive impairment using 123I-MIBG cardiac scintigraphy. J Neurol Sci. 2012;315(1-2):115-119.

34. O’Brien JT, McKeith IG, Walker Z, et al. Diagnostic accuracy of 123I-FP-CIT SPECT in possible dementia with Lewy bodies. Br J Psychiatry. 2009;194:34-39.

35. Yoshita M, Taki J, Yokoyama K, et al. Value of 123I-MIBG radioactivity in the differential diagnosis of DLB from AD. Neurology. 2006;66(12):1850-1854.

Article PDF
Author and Disclosure Information

Ram J. Bishnoi, MD
Fellow, Geriatric Psychiatry, Department of Neurology and Psychiatry, St. Louis University School of Medicine, St. Louis, MO
George T. Grossberg, MD
Samuel W. Fordyce Professor, Director of Geriatric Psychiatry, Department of Neurology and Psychiatry, St. Louis University School of Medicine, St. Louis, MO
Jothika Manepalli, MD
Professor, Division of Geriatric Psychiatry, Department of Neurology and Psychiatry, St. Louis University School of Medicine, St. Louis, MO

Issue
Current Psychiatry - 11(11)
Publications
Topics
Page Number
22-30
Legacy Keywords
Alzheimer's disease; dementia; dementia with Lewy bodies; Lewy bodies
Sections
Author and Disclosure Information

Ram J. Bishnoi, MD
Fellow, Geriatric Psychiatry, Department of Neurology and Psychiatry, St. Louis University School of Medicine, St. Louis, MO
George T. Grossberg, MD
Samuel W. Fordyce Professor, Director of Geriatric Psychiatry, Department of Neurology and Psychiatry, St. Louis University School of Medicine, St. Louis, MO
Jothika Manepalli, MD
Professor, Division of Geriatric Psychiatry, Department of Neurology and Psychiatry, St. Louis University School of Medicine, St. Louis, MO

Author and Disclosure Information

Ram J. Bishnoi, MD
Fellow, Geriatric Psychiatry, Department of Neurology and Psychiatry, St. Louis University School of Medicine, St. Louis, MO
George T. Grossberg, MD
Samuel W. Fordyce Professor, Director of Geriatric Psychiatry, Department of Neurology and Psychiatry, St. Louis University School of Medicine, St. Louis, MO
Jothika Manepalli, MD
Professor, Division of Geriatric Psychiatry, Department of Neurology and Psychiatry, St. Louis University School of Medicine, St. Louis, MO

Article PDF
Article PDF

Discuss this article at www.facebook.com/CurrentPsychiatry

Alzheimer’s disease (AD) and dementia with Lewy bodies (DLB) are the first and second most common causes of neurodegenerative dementia, respectively.“New Alzheimer’s disease guidelines: Implications for clinicians,” Current Psychiatry, March 2012, p. 15-20; http://bit.ly/UNYikk.

The 2005 report of the DLB Consortium5 recognizes central, core, suggestive, and supportive features of DLB (Table 1).5,10 These features are considered in the context of other confounding clinical conditions and the timing of cognitive and motor symptoms. The revised DLB criteria5 require a central feature of progressive cognitive decline. “Probable DLB” is when a patient presents with 2 core features or 1 core feature and ≥1 suggestive features. A diagnosis of “possible DLB” requires 1 core feature or 1 suggestive feature in the presence of progressive cognitive decline.

Table 1

Diagnostic criteria for AD and DLB

NIA-AA criteria for AD (2011)10
Possible AD: Clinical and cognitive criteria (DSM-IV-TR) for AD are met and there is an absence of biomarkers to support the diagnosis or there is evidence of a secondary disorder that can cause dementia
Probable AD: Clinical and cognitive criteria for AD are met and there is documented progressive cognitive decline or abnormal biomarker(s) suggestive of AD or evidence of proven AD autosomal dominant genetic mutation (presenilin-1, presenilin-2, amyloid-β precursor protein)
Definite AD: Clinical criteria for probable AD are met and there is histopathologic evidence of the disorder
Revised clinical diagnostic criteria for DLB (2005)5
Core features: Fluctuating cognition, recurrent visual hallucinations, soft motor features of parkinsonism
Suggestive features: REM sleep behavior disorder, severe antipsychotic sensitivity, decreased tracer uptake in striatum on SPECT dopamine transporter imaging or on myocardial scintigraphy with MIBG
Supportive features (common but lacking diagnostic specificity): repeated falls and syncope; transient, unexplained loss of consciousness; systematized delusions; hallucinations other than visual; relative preservation of medial temporal lobe on CT or MRI scan; decreased tracer uptake on SPECT or PET imaging in occipital regions; prominent slow waves on EEG with temporal lobe transient sharp waves
AD: Alzheimer’s disease; DLB: dementia with Lewy bodies; MIBG: metaiodobenzylguanidine; NIA-AA: National Institute on Aging and the Alzheimer’s Association; PET: positron emission tomography; REM: rapid eye movement; SPECT: single photon emission computed tomography

Biomarkers for AD, but not DLB

The 2011 diagnostic criteria for AD incorporate biomarkers that can be measured in vivo and reflect speci?c features of disease-related pathophysiologic processes. Biomarkers for AD are divided into 2 categories:11

  • amyloid-beta (Aβ) accumulation: abnormal tracer retention on amyloid positron emission topography (PET) imaging and low cerebrospinal fluid (CSF) Aβ42
  • neuronal degeneration or injury: elevated CSF tau (total and phosphorylated tau), decreased ?uorodeoxyglucose uptake on PET in temporo-parietal cortices, and atrophy on structural MRI in the hippocampal and temporo-parietal regions.

No clinically applicable genotypic or CSF markers exist to support a DLB diagnosis, but there are many promising candidates, including elevated levels of CSF p-tau 181, CSF levels of alpha- and beta-synuclein,12 and CSF beta-glucocerebrosidase levels.13 PET mapping of brain acetylcholinesterase activity,14 123I-2β-carbomethoxy-3β- (4-iodophenyl)-N-(3-fluoropropyl)nortropane single photon emission computed tomography (SPECT) dopamine transporter (DaT) imaging15 and metaiodobenzylguanidine (MIBG) scintigraphy also are promising methods. DaT scan SPECT is FDA-approved for detecting loss of functional dopaminergic neuron terminals in the striatum and can differentiate between AD and DLB with a sensitivity and specificity of 78% to 88% and 94% to 100%, respectively.16 This test is covered by Medicare for differentiating AD and DLB.

Differences in presentation

Cognitive impairment. Contrary to the early memory impairment that characterizes AD, memory deficits in DLB usually appear later in the disease course.5 Patients with DLB manifest greater attentional, visuospatial, and executive impairments than those with AD, whereas AD causes more profound episodic (declarative) memory impairment than DLB. DLB patients show more preserved consolidation and storage of verbal information than AD patients because of less neuroanatomical and cholinergic compromise in the medial temporal lobe. There is no evidence of significant differences in remote memory, semantic memory, and language (naming and fluency).

Compromised attention in DLB may be the basis for fluctuating cognition, a characteristic of the disease. The greater attentional impairment and reaction time variability in DLB compared with AD is evident during complex tasks for attention and may be a function of the executive and visuospatial demands of the tasks.17

Executive functions critical to adaptive, goal-directed behavior are more impaired in DLB than AD. DLB patients are more susceptible to distraction and have difficulty engaging in a task and shifting from 1 task to another. This, together with a tendency for confabulation and perseveration, are signs of executive dysfunction.

 

 

Neuropsychiatric features. DLB patients are more likely than AD patients to exhibit psychiatric symptoms and have more functional impairment.18 In an analysis of autopsy-confirmed cases, hallucinations and delusions were more frequent with Lewy body pathology (75%) than AD (21%) at initial clinical evaluation.18 By the end stages of both illnesses, the degree of psychotic symptoms is comparable.19 Depression is common in DLB; whether base rates of depressed mood and major depression differ between DLB and AD is uncertain.20

Psychosis in AD can be induced by medication or delirium, or triggered by poor sensory perceptions. Psychotic symptoms occur more frequently during the moderate and advanced stages of AD, when patients present with visual hallucinations, delusions, or delusional misidentifications. As many as 10% to 20% of patients with AD experience hallucinations, typically visual. Delusions occur in 30% to 50% of AD patients, usually in the later stages of the disease. The most common delusional themes are infidelity, theft, and paranoia. Female sex is a risk factor for psychosis in AD. Delusions co-occur with aggression, anxiety, and aberrant motor behavior.

Visual hallucinations—mostly vivid, well-formed, false perceptions of insects, animals, or people—are the defining feature of DLB.21 Many patients recognize that they are experiencing visual hallucinations and can ignore them. DLB patients also may experience visual illusions, such as misperceiving household objects as living beings. Delusions—typically paranoid—are common among DLB patients, as are depression and anxiety.1 Agitation or aggressive behavior tends to occur late in the illness, if at all.

The causes of psychotic symptoms in DLB are not fully understood, but dopamine dysfunction likely is involved in hallucinations, delusions, and agitation, and serotonin dysfunction may be associated with depression and anxiety. Rapid eye movement (REM) sleep/wakefulness dysregulation, in which the dream imagery of REM sleep may occur during wakefulness, also has been proposed as a mechanism for visual hallucinations in DLB.22 In DLB, psychotic symptoms occur early and are a hallmark of this illness, whereas in AD they usually occur in the middle to late stages of the disease.

Motor symptoms. In AD, extrapyramidal symptoms (EPS) are common later in the disease, are strongly correlated with disease severity, and are a strong, independent predictor of depression severity.23 EPS are more common in DLB than in AD24 and DLB patients are at higher risk of developing EPS even with low doses of typical antipsychotics, compared with AD patients.25

Other symptoms. REM sleep behavior disorder (RBD) is characterized by enacting dreams—often violent—during REM sleep. RBD is common in DLB and many patients also have excessive daytime somnolence. Other sleep disorders in DLB include insomnia, obstructive sleep apnea, central sleep apnea, restless legs syndrome, and periodic limb movements during sleep.

In AD patients, common sleep behaviors include confusion in the early evening (“sundowning”) and frequent nighttime awakenings, often accompanied by wandering.26 Orthostatic hypotension, impotence, urinary incontinence, and constipation are common in DLB. Lack of insight concerning personal cognitive, mood, and behavioral state is highly prevalent in AD patients and more common than in DLB.

Diagnostic evaluation

Because there are no definitive clinical markers for DLB, diagnosis is based on a detailed clinical and family history from the patient and a reliable informant, as well as a physical, neurologic, and mental status examination that looks for associated noncognitive symptoms, and neuropsychological evaluation. Reasons DLB may be misdiagnosed include:

  • Some “core” clinical features of DLB may not appear or may overlap with AD.
  • Presence and severity of concurrent AD pathology in DLB may modify the clinical presentation, with decreased rates of hallucinations and parkinsonism and increased neurofibrillary tangles.
  • Failure to reliably identify fluctuations—variations in cognition and arousal, such as periods of unresponsiveness while awake (“zoning out”), excessive daytime somnolence, and disorganized speech.27

Detecting and characterizing cognitive deficits in dementia patients using neuropsychological testing is important in establishing a clinical diagnosis, determining baseline levels of impairment, forming a prognosis, and initiating disease-specific treatments. Differences in neuropsychological findings in AD and DLB are outlined in Table 2.16,28-33 Several studies have suggested using these measures to differentiate patients with DLB from those with AD.20

Table 2

Diagnostic testing for Alzheimer’s disease and dementia with Lewy bodies

Alzheimer’s diseaseDementia with Lewy bodies
Neuropsychological testing findings
Relatively more impairment on verbal memory tasks, delayed recall, delayed recognition, and encoding and storing information.28 Dysfunction of episodic memory functionRelatively more impairment on attention or concentration, verbal fluency, visuoperceptual, visuoconstructive, visual memory tests, and frontal executive functions.28 Relatively preserved confrontation naming and verbal memory
MRI findings
Diffuse cortical atrophy, relatively greater volume loss in hippocampus and medial temporal lobe structures (strong correlation with severity)29Mild generalized cerebral cortical atrophy with minimal hippocampal atrophy and relative preservation of medial temporal lobe structures30
[18F]FDG PET
Widespread metabolic deficits in neocortical association areas, with sparing of the basal ganglia, thalamus, cerebellum, primary sensory motor cortex, and visual cortexWidespread cortical hypometabolism, more marked in primary visual and occipital association areas, and less severe in parietal, frontal, and anterior cingulate cortices.31 Severe cholinergic deafferentation of the neocortex, particularly in posterior cortical regions32
Single photon emission computed tomography
Parietotemporal hypoperfusionOccipital hypoperfusion
123I-FP-CIT SPECT (DaT scan)
No significant loss of DaTLow nigrostriatal terminal density of DaT caused by severe nigrostriatal degeneration16
Myocardial scintigraphy with MIBG
No significant change in MIBG uptakeDecreased MIBG uptake33
123I-FP-CIT: 123I-2β-carbomethoxy-3β-(4-iodophenyl)-N-(3-fluoropropyl)nortropane; DaT: dopamine transporter; FDG PET: [18F]-fluoro-d-glucose positron emission tomography; MIBG: metaiodobenzylguanidine; SPECT: single photon emission computed tomography
 

 

Evidence is insufficient to support using electroencephalographic and polysomnographic studies when initially evaluating patients with dementia. Brain CT or MRI are recommended as part of the initial evaluation of dementia patients to exclude treatable causes of dementia and help clarify the differential diagnosis. Occipital hypometabolism and hypoperfusion demonstrated on PET and SPECT imaging have high sensitivity and specificity for differentiating AD from DLB.

To diagnose DLB more consistently, look for core features of the disease, RBD, antipsychotic hypersensitivity, and decreased striatal binding at presynaptic DaT sites.15 Abnormal (low binding) DaT activity is the most reliable diagnostic marker for DLB.34 Myocardial scintigraphy with MIBG is sensitive to pathologic changes of DLB before clinical expression and could overcome the difficulties of using clinical criteria alone to identify patients with DLB.35 MIBG scintigraphy may be preferred to DaT scan because it is less expensive and its sensitivity and specificity to DLB are independent of the presence of parkinsonism.35

For an overview of pharmacotherapy options for patients with AD or DLB, see Box 2.

Box 2

Treatments for Alzheimer’s disease and dementia with Lewy bodies

Pharmacotherapy options for patients with Alzheimer’s disease (AD) or dementia with Lewy bodies (DLB) include cholinesterase inhibitors, memantine, antipsychotics, and other agents.

Cholinesterase inhibitors. Donepezil, rivastigmine, and galantamine are FDA-approved for treating AD. Their efficacy appears to be similar, so the choice of agent is based largely on cost, patient tolerability, and physician experience.

No medications are FDA-approved for treating DLB. Neocortical cholinergic activity assessed by choline acetyltransferase levels is more severely depleted in DLB than in AD, and this deficit is correlated with the presence of visual hallucinations and global severity of cognitive impairment.a Therefore, drugs that enhance central cholinergic function offer a therapeutic approach for DLB; cognitive and hallucinatory symptoms are the anticipated targets. Multiple anecdotal reports, open-label studies,b,c and 1 randomized, placebo-controlled triald suggest that cholinesterase inhibitors are efficacious in DLB, with reported benefits in cognition, fluctuations, psychotic symptoms, and parkinsonian symptoms. A 20-week randomized, double-blind, placebo-controlled multicenter studyd of patients with DLB found rivastigmine, 6 to 12 mg/d, was superior to placebo. Patients receiving rivastigmine exhibited significantly reduced anxiety, delusions, and hallucinations and significantly better performance on a computerized battery of neuropsychological tests, especially tasks that required sustained attention. Differences between rivastigmine and placebo disappeared after drug discontinuation.

Memantine is a noncompetitive antagonist of N-methyl-d-aspartate receptors that is effective in AD.e The possible involvement of glutamate in DLB has provided a rationale for treating DLB with memantine. Two randomized controlled trials in DLB found that patients treated with memantine for 24 weeks performed better on Clinical Global Impression of Change, but not on most other secondary outcome measures.f,g In both studies, memantine was well tolerated. However, other studies have noted worsening of delusions and hallucinations with memantine in DLB patients.h

Antipsychotics. Agitation is common in moderate and advanced AD. Atypical antipsychotics have been used with variable efficacy to treat agitation, but their use is associated with excess mortality. DLB patients pose a considerable therapeutic challenge because antipsychotics—the mainstay of treatment of psychosis and behavioral problems in most other disorders—can provoke severe, irreversible, and often fatal sensitivity reactions in this type of dementia.i A 2- to 3-fold increased mortality risk associated with antipsychotic sensitivity reactions in DLB is partly mediated via acute blockade of postsynaptic dopamine D2 receptors in the striatum. For severe and disabling psychosis, a trial of a cholinesterase inhibitor and/or lowering the dose of antiparkinsonian medication should be considered first. In urgent situations, small doses of an atypical antipsychotic that is least associated with parkinsonism side effects—such as quetiapine or aripiprazole—should be used.

Other treatments. Treatment of parkinsonian symptoms in DLB patients is similar to that for Parkinson’s disease, but the risk of psychotic symptoms in DLB warrants a conservative approach. Levodopa seems to be more effective than dopamine agonists and produces fewer side effects.j Rapid eye movement sleep behavior disorder often responds to low doses of clonazepam (0.25 to 1.5 mg). Depression and anxiety disorders are common in AD at all stages and their treatment is not fundamentally different than in geriatric patients without dementia. Selective serotonin reuptake inhibitors and electroconvulsive therapy have been used successfully in depressed patients with AD or DLB.k,l

Disease-modifying treatments. Researchers are evaluating an array of antiamyloid and neuroprotective therapeutic approaches for AD based on the hypothesis that amyloid-beta protein plays a pivotal role in disease onset and progression. Interventions that reduce amyloid production, limit aggregation, or increase clearance may block the cascade of events comprising AD pathogenesis. Reducing tau hyperphosphorylation, limiting oxidation and excitotoxicity, and controlling inflammation also may be beneficial strategies. Potentially neuroprotective and restorative treatments such as neurotrophins, neurotrophic factor enhancers, and stem cell-related approaches also are being investigated.

There are no large-scale studies of disease-modifying treatments for DLB. Potential areas of research include the relationship between proteasome function and a-synuclein pathology, the role of beta-synuclein, and the impact of alterations to alpha-synuclein on its propensity to aggregate.

References

a. Ballard C, Ziabreva I, Perry R, et al. Differences in neuropathologic characteristics across the Lewy body dementia spectrum. Neurology. 2006;67(11):1931-1934.

b. Beversdorf DQ, Warner JL, Davis RA, et al. Donepezil in the treatment of dementia with lewy bodies. Am J Geriatr Psychiatry. 2004;12(5):542-544.

c. Edwards K, Royall D, Hershey L, et al. Efficacy and safety of galantamine in patients with dementia with Lewy bodies: a 24-week open-label study. Dement Geriatr Cogn Disord. 2007;23(6):401-405.

d. McKeith I, Del Ser T, Spano P, et al. Efficacy of rivastigmine in dementia with Lewy bodies: a randomised, double-blind, placebo-controlled international study. Lancet. 2000;356(9247):2031-2036.

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

f. Aarsland D, Ballard C, Walker Z, et al. Memantine in patients with Parkinson’s disease dementia or dementia with Lewy bodies: a double-blind, placebo-controlled, multicentre trial. Lancet Neurol. 2009;8(7):613-618.

g. Emre M, Tsolaki M, Bonuccelli U, et al. Memantine for patients with Parkinson’s disease dementia or dementia with Lewy bodies: a randomised, double-blind, placebo-controlled trial. Lancet Neurol. 2010;9(10):969-977.

h. Ridha BH, Josephs KA, Rossor MN. Delusions and hallucinations in dementia with Lewy bodies: worsening with memantine. Neurology. 2005;65(3):481-482.

i. McKeith I, Fairbairn A, Perry R, et al. Neuroleptic sensitivity in patients with senile dementia of Lewy body type. BMJ. 1992;305(6855):673-678.

j. Fernandez HH, Wu CK, Ott BR. Pharmacotherapy of dementia with Lewy bodies. Expert Opin Pharmacother. 2003;4(11):2027-2037.

k. Swartz M, Barak Y, Mirecki I, et al. Treating depression in Alzheimer’s disease: integration of differing guidelines. Int Psychogeriatr. 2000;12(3):353-358.

l. Takahashi S, Mizukami K, Yasuno F, et al. Depression associated with dementia with Lewy bodies (DLB) and the effect of somatotherapy. Psychogeriatrics. 2009;9(2):56-61.

 

 

Related Resources

  • Hanyu H, Sato T, Hirao K, et al. Differences in clinical course between dementia with Lewy bodies and Alzheimer’s disease. Eur J Neurol. 2009;16(2):212-217.
  • Walker Z, McKeith I, Rodda J, et al. Comparison of cognitive decline between dementia with Lewy bodies and Alzheimer’s disease: a cohort study. BMJ Open. 2012;2:e000380.

Drug Brand Names

  • Aripiprazole • Abilify
  • Clonazepam • Klonopin
  • Donepezil • Aricept
  • Galantamine • Razadyne, Reminyl
  • Levodopa • Dopar, Larodopa
  • Memantine • Namenda
  • Quetiapine • Seroquel
  • Rivastigmine • Exelon

Disclosure

Drs. Bishnoi and Manepalli report no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.

Dr. Grossberg serves as a consultant to Forest, Janssen, Novartis, and Pfizer. His department receives research funding from Novartis, Janssen, and Pfizer.

Discuss this article at www.facebook.com/CurrentPsychiatry

Alzheimer’s disease (AD) and dementia with Lewy bodies (DLB) are the first and second most common causes of neurodegenerative dementia, respectively.“New Alzheimer’s disease guidelines: Implications for clinicians,” Current Psychiatry, March 2012, p. 15-20; http://bit.ly/UNYikk.

The 2005 report of the DLB Consortium5 recognizes central, core, suggestive, and supportive features of DLB (Table 1).5,10 These features are considered in the context of other confounding clinical conditions and the timing of cognitive and motor symptoms. The revised DLB criteria5 require a central feature of progressive cognitive decline. “Probable DLB” is when a patient presents with 2 core features or 1 core feature and ≥1 suggestive features. A diagnosis of “possible DLB” requires 1 core feature or 1 suggestive feature in the presence of progressive cognitive decline.

Table 1

Diagnostic criteria for AD and DLB

NIA-AA criteria for AD (2011)10
Possible AD: Clinical and cognitive criteria (DSM-IV-TR) for AD are met and there is an absence of biomarkers to support the diagnosis or there is evidence of a secondary disorder that can cause dementia
Probable AD: Clinical and cognitive criteria for AD are met and there is documented progressive cognitive decline or abnormal biomarker(s) suggestive of AD or evidence of proven AD autosomal dominant genetic mutation (presenilin-1, presenilin-2, amyloid-β precursor protein)
Definite AD: Clinical criteria for probable AD are met and there is histopathologic evidence of the disorder
Revised clinical diagnostic criteria for DLB (2005)5
Core features: Fluctuating cognition, recurrent visual hallucinations, soft motor features of parkinsonism
Suggestive features: REM sleep behavior disorder, severe antipsychotic sensitivity, decreased tracer uptake in striatum on SPECT dopamine transporter imaging or on myocardial scintigraphy with MIBG
Supportive features (common but lacking diagnostic specificity): repeated falls and syncope; transient, unexplained loss of consciousness; systematized delusions; hallucinations other than visual; relative preservation of medial temporal lobe on CT or MRI scan; decreased tracer uptake on SPECT or PET imaging in occipital regions; prominent slow waves on EEG with temporal lobe transient sharp waves
AD: Alzheimer’s disease; DLB: dementia with Lewy bodies; MIBG: metaiodobenzylguanidine; NIA-AA: National Institute on Aging and the Alzheimer’s Association; PET: positron emission tomography; REM: rapid eye movement; SPECT: single photon emission computed tomography

Biomarkers for AD, but not DLB

The 2011 diagnostic criteria for AD incorporate biomarkers that can be measured in vivo and reflect speci?c features of disease-related pathophysiologic processes. Biomarkers for AD are divided into 2 categories:11

  • amyloid-beta (Aβ) accumulation: abnormal tracer retention on amyloid positron emission topography (PET) imaging and low cerebrospinal fluid (CSF) Aβ42
  • neuronal degeneration or injury: elevated CSF tau (total and phosphorylated tau), decreased ?uorodeoxyglucose uptake on PET in temporo-parietal cortices, and atrophy on structural MRI in the hippocampal and temporo-parietal regions.

No clinically applicable genotypic or CSF markers exist to support a DLB diagnosis, but there are many promising candidates, including elevated levels of CSF p-tau 181, CSF levels of alpha- and beta-synuclein,12 and CSF beta-glucocerebrosidase levels.13 PET mapping of brain acetylcholinesterase activity,14 123I-2β-carbomethoxy-3β- (4-iodophenyl)-N-(3-fluoropropyl)nortropane single photon emission computed tomography (SPECT) dopamine transporter (DaT) imaging15 and metaiodobenzylguanidine (MIBG) scintigraphy also are promising methods. DaT scan SPECT is FDA-approved for detecting loss of functional dopaminergic neuron terminals in the striatum and can differentiate between AD and DLB with a sensitivity and specificity of 78% to 88% and 94% to 100%, respectively.16 This test is covered by Medicare for differentiating AD and DLB.

Differences in presentation

Cognitive impairment. Contrary to the early memory impairment that characterizes AD, memory deficits in DLB usually appear later in the disease course.5 Patients with DLB manifest greater attentional, visuospatial, and executive impairments than those with AD, whereas AD causes more profound episodic (declarative) memory impairment than DLB. DLB patients show more preserved consolidation and storage of verbal information than AD patients because of less neuroanatomical and cholinergic compromise in the medial temporal lobe. There is no evidence of significant differences in remote memory, semantic memory, and language (naming and fluency).

Compromised attention in DLB may be the basis for fluctuating cognition, a characteristic of the disease. The greater attentional impairment and reaction time variability in DLB compared with AD is evident during complex tasks for attention and may be a function of the executive and visuospatial demands of the tasks.17

Executive functions critical to adaptive, goal-directed behavior are more impaired in DLB than AD. DLB patients are more susceptible to distraction and have difficulty engaging in a task and shifting from 1 task to another. This, together with a tendency for confabulation and perseveration, are signs of executive dysfunction.

 

 

Neuropsychiatric features. DLB patients are more likely than AD patients to exhibit psychiatric symptoms and have more functional impairment.18 In an analysis of autopsy-confirmed cases, hallucinations and delusions were more frequent with Lewy body pathology (75%) than AD (21%) at initial clinical evaluation.18 By the end stages of both illnesses, the degree of psychotic symptoms is comparable.19 Depression is common in DLB; whether base rates of depressed mood and major depression differ between DLB and AD is uncertain.20

Psychosis in AD can be induced by medication or delirium, or triggered by poor sensory perceptions. Psychotic symptoms occur more frequently during the moderate and advanced stages of AD, when patients present with visual hallucinations, delusions, or delusional misidentifications. As many as 10% to 20% of patients with AD experience hallucinations, typically visual. Delusions occur in 30% to 50% of AD patients, usually in the later stages of the disease. The most common delusional themes are infidelity, theft, and paranoia. Female sex is a risk factor for psychosis in AD. Delusions co-occur with aggression, anxiety, and aberrant motor behavior.

Visual hallucinations—mostly vivid, well-formed, false perceptions of insects, animals, or people—are the defining feature of DLB.21 Many patients recognize that they are experiencing visual hallucinations and can ignore them. DLB patients also may experience visual illusions, such as misperceiving household objects as living beings. Delusions—typically paranoid—are common among DLB patients, as are depression and anxiety.1 Agitation or aggressive behavior tends to occur late in the illness, if at all.

The causes of psychotic symptoms in DLB are not fully understood, but dopamine dysfunction likely is involved in hallucinations, delusions, and agitation, and serotonin dysfunction may be associated with depression and anxiety. Rapid eye movement (REM) sleep/wakefulness dysregulation, in which the dream imagery of REM sleep may occur during wakefulness, also has been proposed as a mechanism for visual hallucinations in DLB.22 In DLB, psychotic symptoms occur early and are a hallmark of this illness, whereas in AD they usually occur in the middle to late stages of the disease.

Motor symptoms. In AD, extrapyramidal symptoms (EPS) are common later in the disease, are strongly correlated with disease severity, and are a strong, independent predictor of depression severity.23 EPS are more common in DLB than in AD24 and DLB patients are at higher risk of developing EPS even with low doses of typical antipsychotics, compared with AD patients.25

Other symptoms. REM sleep behavior disorder (RBD) is characterized by enacting dreams—often violent—during REM sleep. RBD is common in DLB and many patients also have excessive daytime somnolence. Other sleep disorders in DLB include insomnia, obstructive sleep apnea, central sleep apnea, restless legs syndrome, and periodic limb movements during sleep.

In AD patients, common sleep behaviors include confusion in the early evening (“sundowning”) and frequent nighttime awakenings, often accompanied by wandering.26 Orthostatic hypotension, impotence, urinary incontinence, and constipation are common in DLB. Lack of insight concerning personal cognitive, mood, and behavioral state is highly prevalent in AD patients and more common than in DLB.

Diagnostic evaluation

Because there are no definitive clinical markers for DLB, diagnosis is based on a detailed clinical and family history from the patient and a reliable informant, as well as a physical, neurologic, and mental status examination that looks for associated noncognitive symptoms, and neuropsychological evaluation. Reasons DLB may be misdiagnosed include:

  • Some “core” clinical features of DLB may not appear or may overlap with AD.
  • Presence and severity of concurrent AD pathology in DLB may modify the clinical presentation, with decreased rates of hallucinations and parkinsonism and increased neurofibrillary tangles.
  • Failure to reliably identify fluctuations—variations in cognition and arousal, such as periods of unresponsiveness while awake (“zoning out”), excessive daytime somnolence, and disorganized speech.27

Detecting and characterizing cognitive deficits in dementia patients using neuropsychological testing is important in establishing a clinical diagnosis, determining baseline levels of impairment, forming a prognosis, and initiating disease-specific treatments. Differences in neuropsychological findings in AD and DLB are outlined in Table 2.16,28-33 Several studies have suggested using these measures to differentiate patients with DLB from those with AD.20

Table 2

Diagnostic testing for Alzheimer’s disease and dementia with Lewy bodies

Alzheimer’s diseaseDementia with Lewy bodies
Neuropsychological testing findings
Relatively more impairment on verbal memory tasks, delayed recall, delayed recognition, and encoding and storing information.28 Dysfunction of episodic memory functionRelatively more impairment on attention or concentration, verbal fluency, visuoperceptual, visuoconstructive, visual memory tests, and frontal executive functions.28 Relatively preserved confrontation naming and verbal memory
MRI findings
Diffuse cortical atrophy, relatively greater volume loss in hippocampus and medial temporal lobe structures (strong correlation with severity)29Mild generalized cerebral cortical atrophy with minimal hippocampal atrophy and relative preservation of medial temporal lobe structures30
[18F]FDG PET
Widespread metabolic deficits in neocortical association areas, with sparing of the basal ganglia, thalamus, cerebellum, primary sensory motor cortex, and visual cortexWidespread cortical hypometabolism, more marked in primary visual and occipital association areas, and less severe in parietal, frontal, and anterior cingulate cortices.31 Severe cholinergic deafferentation of the neocortex, particularly in posterior cortical regions32
Single photon emission computed tomography
Parietotemporal hypoperfusionOccipital hypoperfusion
123I-FP-CIT SPECT (DaT scan)
No significant loss of DaTLow nigrostriatal terminal density of DaT caused by severe nigrostriatal degeneration16
Myocardial scintigraphy with MIBG
No significant change in MIBG uptakeDecreased MIBG uptake33
123I-FP-CIT: 123I-2β-carbomethoxy-3β-(4-iodophenyl)-N-(3-fluoropropyl)nortropane; DaT: dopamine transporter; FDG PET: [18F]-fluoro-d-glucose positron emission tomography; MIBG: metaiodobenzylguanidine; SPECT: single photon emission computed tomography
 

 

Evidence is insufficient to support using electroencephalographic and polysomnographic studies when initially evaluating patients with dementia. Brain CT or MRI are recommended as part of the initial evaluation of dementia patients to exclude treatable causes of dementia and help clarify the differential diagnosis. Occipital hypometabolism and hypoperfusion demonstrated on PET and SPECT imaging have high sensitivity and specificity for differentiating AD from DLB.

To diagnose DLB more consistently, look for core features of the disease, RBD, antipsychotic hypersensitivity, and decreased striatal binding at presynaptic DaT sites.15 Abnormal (low binding) DaT activity is the most reliable diagnostic marker for DLB.34 Myocardial scintigraphy with MIBG is sensitive to pathologic changes of DLB before clinical expression and could overcome the difficulties of using clinical criteria alone to identify patients with DLB.35 MIBG scintigraphy may be preferred to DaT scan because it is less expensive and its sensitivity and specificity to DLB are independent of the presence of parkinsonism.35

For an overview of pharmacotherapy options for patients with AD or DLB, see Box 2.

Box 2

Treatments for Alzheimer’s disease and dementia with Lewy bodies

Pharmacotherapy options for patients with Alzheimer’s disease (AD) or dementia with Lewy bodies (DLB) include cholinesterase inhibitors, memantine, antipsychotics, and other agents.

Cholinesterase inhibitors. Donepezil, rivastigmine, and galantamine are FDA-approved for treating AD. Their efficacy appears to be similar, so the choice of agent is based largely on cost, patient tolerability, and physician experience.

No medications are FDA-approved for treating DLB. Neocortical cholinergic activity assessed by choline acetyltransferase levels is more severely depleted in DLB than in AD, and this deficit is correlated with the presence of visual hallucinations and global severity of cognitive impairment.a Therefore, drugs that enhance central cholinergic function offer a therapeutic approach for DLB; cognitive and hallucinatory symptoms are the anticipated targets. Multiple anecdotal reports, open-label studies,b,c and 1 randomized, placebo-controlled triald suggest that cholinesterase inhibitors are efficacious in DLB, with reported benefits in cognition, fluctuations, psychotic symptoms, and parkinsonian symptoms. A 20-week randomized, double-blind, placebo-controlled multicenter studyd of patients with DLB found rivastigmine, 6 to 12 mg/d, was superior to placebo. Patients receiving rivastigmine exhibited significantly reduced anxiety, delusions, and hallucinations and significantly better performance on a computerized battery of neuropsychological tests, especially tasks that required sustained attention. Differences between rivastigmine and placebo disappeared after drug discontinuation.

Memantine is a noncompetitive antagonist of N-methyl-d-aspartate receptors that is effective in AD.e The possible involvement of glutamate in DLB has provided a rationale for treating DLB with memantine. Two randomized controlled trials in DLB found that patients treated with memantine for 24 weeks performed better on Clinical Global Impression of Change, but not on most other secondary outcome measures.f,g In both studies, memantine was well tolerated. However, other studies have noted worsening of delusions and hallucinations with memantine in DLB patients.h

Antipsychotics. Agitation is common in moderate and advanced AD. Atypical antipsychotics have been used with variable efficacy to treat agitation, but their use is associated with excess mortality. DLB patients pose a considerable therapeutic challenge because antipsychotics—the mainstay of treatment of psychosis and behavioral problems in most other disorders—can provoke severe, irreversible, and often fatal sensitivity reactions in this type of dementia.i A 2- to 3-fold increased mortality risk associated with antipsychotic sensitivity reactions in DLB is partly mediated via acute blockade of postsynaptic dopamine D2 receptors in the striatum. For severe and disabling psychosis, a trial of a cholinesterase inhibitor and/or lowering the dose of antiparkinsonian medication should be considered first. In urgent situations, small doses of an atypical antipsychotic that is least associated with parkinsonism side effects—such as quetiapine or aripiprazole—should be used.

Other treatments. Treatment of parkinsonian symptoms in DLB patients is similar to that for Parkinson’s disease, but the risk of psychotic symptoms in DLB warrants a conservative approach. Levodopa seems to be more effective than dopamine agonists and produces fewer side effects.j Rapid eye movement sleep behavior disorder often responds to low doses of clonazepam (0.25 to 1.5 mg). Depression and anxiety disorders are common in AD at all stages and their treatment is not fundamentally different than in geriatric patients without dementia. Selective serotonin reuptake inhibitors and electroconvulsive therapy have been used successfully in depressed patients with AD or DLB.k,l

Disease-modifying treatments. Researchers are evaluating an array of antiamyloid and neuroprotective therapeutic approaches for AD based on the hypothesis that amyloid-beta protein plays a pivotal role in disease onset and progression. Interventions that reduce amyloid production, limit aggregation, or increase clearance may block the cascade of events comprising AD pathogenesis. Reducing tau hyperphosphorylation, limiting oxidation and excitotoxicity, and controlling inflammation also may be beneficial strategies. Potentially neuroprotective and restorative treatments such as neurotrophins, neurotrophic factor enhancers, and stem cell-related approaches also are being investigated.

There are no large-scale studies of disease-modifying treatments for DLB. Potential areas of research include the relationship between proteasome function and a-synuclein pathology, the role of beta-synuclein, and the impact of alterations to alpha-synuclein on its propensity to aggregate.

References

a. Ballard C, Ziabreva I, Perry R, et al. Differences in neuropathologic characteristics across the Lewy body dementia spectrum. Neurology. 2006;67(11):1931-1934.

b. Beversdorf DQ, Warner JL, Davis RA, et al. Donepezil in the treatment of dementia with lewy bodies. Am J Geriatr Psychiatry. 2004;12(5):542-544.

c. Edwards K, Royall D, Hershey L, et al. Efficacy and safety of galantamine in patients with dementia with Lewy bodies: a 24-week open-label study. Dement Geriatr Cogn Disord. 2007;23(6):401-405.

d. McKeith I, Del Ser T, Spano P, et al. Efficacy of rivastigmine in dementia with Lewy bodies: a randomised, double-blind, placebo-controlled international study. Lancet. 2000;356(9247):2031-2036.

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

f. Aarsland D, Ballard C, Walker Z, et al. Memantine in patients with Parkinson’s disease dementia or dementia with Lewy bodies: a double-blind, placebo-controlled, multicentre trial. Lancet Neurol. 2009;8(7):613-618.

g. Emre M, Tsolaki M, Bonuccelli U, et al. Memantine for patients with Parkinson’s disease dementia or dementia with Lewy bodies: a randomised, double-blind, placebo-controlled trial. Lancet Neurol. 2010;9(10):969-977.

h. Ridha BH, Josephs KA, Rossor MN. Delusions and hallucinations in dementia with Lewy bodies: worsening with memantine. Neurology. 2005;65(3):481-482.

i. McKeith I, Fairbairn A, Perry R, et al. Neuroleptic sensitivity in patients with senile dementia of Lewy body type. BMJ. 1992;305(6855):673-678.

j. Fernandez HH, Wu CK, Ott BR. Pharmacotherapy of dementia with Lewy bodies. Expert Opin Pharmacother. 2003;4(11):2027-2037.

k. Swartz M, Barak Y, Mirecki I, et al. Treating depression in Alzheimer’s disease: integration of differing guidelines. Int Psychogeriatr. 2000;12(3):353-358.

l. Takahashi S, Mizukami K, Yasuno F, et al. Depression associated with dementia with Lewy bodies (DLB) and the effect of somatotherapy. Psychogeriatrics. 2009;9(2):56-61.

 

 

Related Resources

  • Hanyu H, Sato T, Hirao K, et al. Differences in clinical course between dementia with Lewy bodies and Alzheimer’s disease. Eur J Neurol. 2009;16(2):212-217.
  • Walker Z, McKeith I, Rodda J, et al. Comparison of cognitive decline between dementia with Lewy bodies and Alzheimer’s disease: a cohort study. BMJ Open. 2012;2:e000380.

Drug Brand Names

  • Aripiprazole • Abilify
  • Clonazepam • Klonopin
  • Donepezil • Aricept
  • Galantamine • Razadyne, Reminyl
  • Levodopa • Dopar, Larodopa
  • Memantine • Namenda
  • Quetiapine • Seroquel
  • Rivastigmine • Exelon

Disclosure

Drs. Bishnoi and Manepalli report no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.

Dr. Grossberg serves as a consultant to Forest, Janssen, Novartis, and Pfizer. His department receives research funding from Novartis, Janssen, and Pfizer.

References

1. McKeith IG, Galasko D, Kosaka K, et al. Consensus guidelines for the clinical and pathologic diagnosis of dementia with Lewy bodies (DLB): report of the consortium on DLB international workshop. Neurology. 1996;47(5):1113-1124.

2. Buracchio T, Arvanitakis Z, Gorbien M. Dementia with Lewy bodies: current concepts. Dement Geriatr Cogn Disord. 2005;20(5):306-320.

3. Fujishiro H, Iseki E, Higashi S, et al. Distribution of cerebral amyloid deposition and its relevance to clinical phenotype in Lewy body dementia. Neurosci Lett. 2010;486(1):19-23.

4. Kosaka K. Diffuse Lewy body disease. Neuropathology. 2000;20(suppl):S73-S78.

5. McKeith IG, Dickson DW, Lowe J, et al. Consortium on DLB. Diagnosis and management of dementia with Lewy bodies: third report of the DLB Consortium. Neurology. 2005;65(12):1863-1872.

6. Cummings JL, Cole G. Alzheimer disease. JAMA. 2002;287(18):2335-2338.

7. Zaccai J, McCracken C, Brayne C. A systematic review of prevalence and incidence studies of dementia with Lewy bodies. Age Ageing. 2005;34(6):561-566.

8. Bradshaw J, Saling M, Hopwood M, et al. Fluctuating cognition in dementia with Lewy bodies and Alzheimer’s disease is qualitatively distinct. J Neurol Neurosurg Psychiatry. 2004;75(3):382-387.

9. Singleton AB, Wharton A, O’Brien KK, et al. Clinical and neuropathological correlates of apolipoprotein E genotype in dementia with Lewy bodies. Dement Geriatr Cogn Disord. 2002;14(4):167-175.

10. McKhann GM, Knopman DS, Chertkow H, et al. The diagnosis of dementia due to Alzheimer’s disease: recommendations from the National Institute on Aging-Alzheimer’s Association workgroups on diagnostic guidelines for Alzheimer’s disease. Alzheimers Dement. 2011;7(3):263-269.

11. Jack CR, Jr, Albert MS, Knopman DS, et al. Introduction to the recommendations from the National Institute on Aging-Alzheimer’s Association workgroups on diagnostic guidelines for Alzheimer’s disease. Alzheimers Dement. 2011;7(3):257-262.

12. Mollenhauer B, Cullen V, Kahn I, et al. Direct quantification of CSF alpha-synuclein by ELISA and first cross-sectional study in patients with neurodegeneration. Exp Neurol. 2008;213(2):315-325.

13. Parnetti L, Balducci C, Pierguidi L, et al. Cerebrospinal fluid beta-glucocerebrosidase activity is reduced in dementia with Lewy bodies. Neurobiol Dis. 2009;34(3):484-486.

14. Shimada H, Hirano S, Shinotoh H, et al. Mapping of brain acetylcholinesterase alterations in Lewy body disease by PET. Neurology. 2009;73(4):273-278.

15. McKeith I, O’Brien J, Walker Z, et al. Sensitivity and specificity of dopamine transporter imaging with 123I-FP-CIT SPECT in dementia with Lewy bodies: a phase III, multicentre study. Lancet Neurol. 2007;6(4):305-313.

16. Walker Z, Jaros E, Walker RW, et al. Dementia with Lewy bodies: a comparison of clinical diagnosis, FP-CIT single photon emission computed tomography imaging and autopsy. J Neurol Neurosurg Psychiatry. 2007;78(11):1176-1181.

17. Bradshaw JM, Saling M, Anderson V, et al. Higher cortical deficits influence attentional processing in dementia with Lewy bodies, relative to patients with dementia of the Alzheimer’s type and controls. J Neurol Neurosurg Psychiatry. 2006;77(10):1129-1135.

18. Weiner MF, Hynan LS, Parikh B, et al. Can Alzheimer’s disease and dementias with Lewy bodies be distinguished clinically? J Geriatr Psychiatry Neurol. 2003;16(4):245-250.

19. Stavitsky K, Brickman AM, Scarmeas N, et al. The progression of cognition, psychiatric symptoms, and functional abilities in dementia with Lewy bodies and Alzheimer disease. Arch Neurol. 2006;63(10):1450-1456.

20. Ferman TJ, Smith GE, Boeve BF, et al. Neuropsychological differentiation of dementia with Lewy bodies from normal aging and Alzheimer’s disease. Clin Neuropsychol. 2006;20(4):623-636.

21. McKeith IG, Perry EK, Perry RH. Report of the second dementia with Lewy body international workshop: diagnosis and treatment. Consortium on Dementia with Lewy Bodies. Neurology. 1999;53(5):902-905.

22. Boeve BF, Silber MH, Ferman TJ, et al. Association of REM sleep behavior disorder and neurodegenerative disease may reflect an underlying synucleinopathy. Mov Disord. 2001;16(4):622-630.

23. Portet F, Scarmeas N, Cosentino S, et al. Extrapyramidal signs before and after diagnosis of incident Alzheimer disease in a prospective population study. Arch Neurol. 2009;66(9):1120-1126.

24. McKeith I, Fairbairn A, Perry R, et al. Neuroleptic sensitivity in patients with senile dementia of Lewy body type. BMJ. 1992;305(6855):673-678.

25. Tarawneh R, Galvin JE. Distinguishing Lewy body dementias from Alzheimer’s disease. Expert Rev Neurother. 2007;7(11):1499-1516.

26. Ancoli-Israel S, Klauber MR, Gillin JC, et al. Sleep in non-institutionalized Alzheimer’s disease patients. Aging (Milano). 1994;6(6):451-458.

27. Ferman TJ, Smith GE, Boeve BF, et al. DLB fluctuations: specific features that reliably differentiate DLB from AD and normal aging. Neurology. 2004;62(2):181-187.

28. Salmon DP, Galasko D, Hansen LA, et al. Neuropsychological deficits associated with diffuse Lewy body disease. Brain Cogn. 1996;31(2):148-165.

29. Jack CR, Jr, Petersen RC, Xu Y, et al. Rates of hippocampal atrophy correlate with change in clinical status in aging and AD. Neurology. 2000;55(4):484-489.

30. Burton EJ, Barber R, Mukaetova-Ladinska EB, et al. Medial temporal lobe atrophy on MRI differentiates Alzheimer’s disease from dementia with Lewy bodies and vascular cognitive impairment: a prospective study with pathological verification of diagnosis. Brain. 2009;132(pt 1):195-203.

31. Ishii K, Soma T, Kono AK, et al. Comparison of regional brain volume and glucose metabolism between patients with mild dementia with lewy bodies and those with mild Alzheimer’s disease. J Nucl Med. 2007;48(5):704-711.

32. Klein JC, Eggers C, Kalbe E, et al. Neurotransmitter changes in dementia with Lewy bodies and Parkinson disease dementia in vivo. Neurology. 2010;74(11):885-892.

33. Fujishiro H, Nakamura S, Kitazawa M, et al. Early detection of dementia with Lewy bodies in patients with amnestic mild cognitive impairment using 123I-MIBG cardiac scintigraphy. J Neurol Sci. 2012;315(1-2):115-119.

34. O’Brien JT, McKeith IG, Walker Z, et al. Diagnostic accuracy of 123I-FP-CIT SPECT in possible dementia with Lewy bodies. Br J Psychiatry. 2009;194:34-39.

35. Yoshita M, Taki J, Yokoyama K, et al. Value of 123I-MIBG radioactivity in the differential diagnosis of DLB from AD. Neurology. 2006;66(12):1850-1854.

References

1. McKeith IG, Galasko D, Kosaka K, et al. Consensus guidelines for the clinical and pathologic diagnosis of dementia with Lewy bodies (DLB): report of the consortium on DLB international workshop. Neurology. 1996;47(5):1113-1124.

2. Buracchio T, Arvanitakis Z, Gorbien M. Dementia with Lewy bodies: current concepts. Dement Geriatr Cogn Disord. 2005;20(5):306-320.

3. Fujishiro H, Iseki E, Higashi S, et al. Distribution of cerebral amyloid deposition and its relevance to clinical phenotype in Lewy body dementia. Neurosci Lett. 2010;486(1):19-23.

4. Kosaka K. Diffuse Lewy body disease. Neuropathology. 2000;20(suppl):S73-S78.

5. McKeith IG, Dickson DW, Lowe J, et al. Consortium on DLB. Diagnosis and management of dementia with Lewy bodies: third report of the DLB Consortium. Neurology. 2005;65(12):1863-1872.

6. Cummings JL, Cole G. Alzheimer disease. JAMA. 2002;287(18):2335-2338.

7. Zaccai J, McCracken C, Brayne C. A systematic review of prevalence and incidence studies of dementia with Lewy bodies. Age Ageing. 2005;34(6):561-566.

8. Bradshaw J, Saling M, Hopwood M, et al. Fluctuating cognition in dementia with Lewy bodies and Alzheimer’s disease is qualitatively distinct. J Neurol Neurosurg Psychiatry. 2004;75(3):382-387.

9. Singleton AB, Wharton A, O’Brien KK, et al. Clinical and neuropathological correlates of apolipoprotein E genotype in dementia with Lewy bodies. Dement Geriatr Cogn Disord. 2002;14(4):167-175.

10. McKhann GM, Knopman DS, Chertkow H, et al. The diagnosis of dementia due to Alzheimer’s disease: recommendations from the National Institute on Aging-Alzheimer’s Association workgroups on diagnostic guidelines for Alzheimer’s disease. Alzheimers Dement. 2011;7(3):263-269.

11. Jack CR, Jr, Albert MS, Knopman DS, et al. Introduction to the recommendations from the National Institute on Aging-Alzheimer’s Association workgroups on diagnostic guidelines for Alzheimer’s disease. Alzheimers Dement. 2011;7(3):257-262.

12. Mollenhauer B, Cullen V, Kahn I, et al. Direct quantification of CSF alpha-synuclein by ELISA and first cross-sectional study in patients with neurodegeneration. Exp Neurol. 2008;213(2):315-325.

13. Parnetti L, Balducci C, Pierguidi L, et al. Cerebrospinal fluid beta-glucocerebrosidase activity is reduced in dementia with Lewy bodies. Neurobiol Dis. 2009;34(3):484-486.

14. Shimada H, Hirano S, Shinotoh H, et al. Mapping of brain acetylcholinesterase alterations in Lewy body disease by PET. Neurology. 2009;73(4):273-278.

15. McKeith I, O’Brien J, Walker Z, et al. Sensitivity and specificity of dopamine transporter imaging with 123I-FP-CIT SPECT in dementia with Lewy bodies: a phase III, multicentre study. Lancet Neurol. 2007;6(4):305-313.

16. Walker Z, Jaros E, Walker RW, et al. Dementia with Lewy bodies: a comparison of clinical diagnosis, FP-CIT single photon emission computed tomography imaging and autopsy. J Neurol Neurosurg Psychiatry. 2007;78(11):1176-1181.

17. Bradshaw JM, Saling M, Anderson V, et al. Higher cortical deficits influence attentional processing in dementia with Lewy bodies, relative to patients with dementia of the Alzheimer’s type and controls. J Neurol Neurosurg Psychiatry. 2006;77(10):1129-1135.

18. Weiner MF, Hynan LS, Parikh B, et al. Can Alzheimer’s disease and dementias with Lewy bodies be distinguished clinically? J Geriatr Psychiatry Neurol. 2003;16(4):245-250.

19. Stavitsky K, Brickman AM, Scarmeas N, et al. The progression of cognition, psychiatric symptoms, and functional abilities in dementia with Lewy bodies and Alzheimer disease. Arch Neurol. 2006;63(10):1450-1456.

20. Ferman TJ, Smith GE, Boeve BF, et al. Neuropsychological differentiation of dementia with Lewy bodies from normal aging and Alzheimer’s disease. Clin Neuropsychol. 2006;20(4):623-636.

21. McKeith IG, Perry EK, Perry RH. Report of the second dementia with Lewy body international workshop: diagnosis and treatment. Consortium on Dementia with Lewy Bodies. Neurology. 1999;53(5):902-905.

22. Boeve BF, Silber MH, Ferman TJ, et al. Association of REM sleep behavior disorder and neurodegenerative disease may reflect an underlying synucleinopathy. Mov Disord. 2001;16(4):622-630.

23. Portet F, Scarmeas N, Cosentino S, et al. Extrapyramidal signs before and after diagnosis of incident Alzheimer disease in a prospective population study. Arch Neurol. 2009;66(9):1120-1126.

24. McKeith I, Fairbairn A, Perry R, et al. Neuroleptic sensitivity in patients with senile dementia of Lewy body type. BMJ. 1992;305(6855):673-678.

25. Tarawneh R, Galvin JE. Distinguishing Lewy body dementias from Alzheimer’s disease. Expert Rev Neurother. 2007;7(11):1499-1516.

26. Ancoli-Israel S, Klauber MR, Gillin JC, et al. Sleep in non-institutionalized Alzheimer’s disease patients. Aging (Milano). 1994;6(6):451-458.

27. Ferman TJ, Smith GE, Boeve BF, et al. DLB fluctuations: specific features that reliably differentiate DLB from AD and normal aging. Neurology. 2004;62(2):181-187.

28. Salmon DP, Galasko D, Hansen LA, et al. Neuropsychological deficits associated with diffuse Lewy body disease. Brain Cogn. 1996;31(2):148-165.

29. Jack CR, Jr, Petersen RC, Xu Y, et al. Rates of hippocampal atrophy correlate with change in clinical status in aging and AD. Neurology. 2000;55(4):484-489.

30. Burton EJ, Barber R, Mukaetova-Ladinska EB, et al. Medial temporal lobe atrophy on MRI differentiates Alzheimer’s disease from dementia with Lewy bodies and vascular cognitive impairment: a prospective study with pathological verification of diagnosis. Brain. 2009;132(pt 1):195-203.

31. Ishii K, Soma T, Kono AK, et al. Comparison of regional brain volume and glucose metabolism between patients with mild dementia with lewy bodies and those with mild Alzheimer’s disease. J Nucl Med. 2007;48(5):704-711.

32. Klein JC, Eggers C, Kalbe E, et al. Neurotransmitter changes in dementia with Lewy bodies and Parkinson disease dementia in vivo. Neurology. 2010;74(11):885-892.

33. Fujishiro H, Nakamura S, Kitazawa M, et al. Early detection of dementia with Lewy bodies in patients with amnestic mild cognitive impairment using 123I-MIBG cardiac scintigraphy. J Neurol Sci. 2012;315(1-2):115-119.

34. O’Brien JT, McKeith IG, Walker Z, et al. Diagnostic accuracy of 123I-FP-CIT SPECT in possible dementia with Lewy bodies. Br J Psychiatry. 2009;194:34-39.

35. Yoshita M, Taki J, Yokoyama K, et al. Value of 123I-MIBG radioactivity in the differential diagnosis of DLB from AD. Neurology. 2006;66(12):1850-1854.

Issue
Current Psychiatry - 11(11)
Issue
Current Psychiatry - 11(11)
Page Number
22-30
Page Number
22-30
Publications
Publications
Topics
Article Type
Display Headline
Differentiating Alzheimer’s disease from dementia with Lewy bodies
Display Headline
Differentiating Alzheimer’s disease from dementia with Lewy bodies
Legacy Keywords
Alzheimer's disease; dementia; dementia with Lewy bodies; Lewy bodies
Legacy Keywords
Alzheimer's disease; dementia; dementia with Lewy bodies; Lewy bodies
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media

Why do cancer patients smoke and what can providers do about it?

Article Type
Changed
Fri, 01/04/2019 - 11:06
Display Headline
Why do cancer patients smoke and what can providers do about it?

Despite the widespread dissemination of information about the health risks associated with smoking, many cancer patients continue to smoke, which results in a decreased quality of life, an increased probability of cancer recurrence, and a decreased survival time. Efficacious interventions are available to assist cancer patients to quit smoking, yet smoking cessation interventions are often not implemented. This review describes how clinicians, administrators, insurers, and purchasers can encourage a culture of health care in which tobacco cessation interventions are implemented consistent with evidence-based standards of care. Implementing efficacious tobacco cessation interventions can reduce morbidity and mortality among cancer patients...

*Click on the link to the left of this introduction for a PDF of the full article.  

Article PDF
Author and Disclosure Information

Publications
Topics
Legacy Keywords
lung cancer, smoking, smoking cessation, smoking interventions
Sections
Author and Disclosure Information

Author and Disclosure Information

Article PDF
Article PDF

Despite the widespread dissemination of information about the health risks associated with smoking, many cancer patients continue to smoke, which results in a decreased quality of life, an increased probability of cancer recurrence, and a decreased survival time. Efficacious interventions are available to assist cancer patients to quit smoking, yet smoking cessation interventions are often not implemented. This review describes how clinicians, administrators, insurers, and purchasers can encourage a culture of health care in which tobacco cessation interventions are implemented consistent with evidence-based standards of care. Implementing efficacious tobacco cessation interventions can reduce morbidity and mortality among cancer patients...

*Click on the link to the left of this introduction for a PDF of the full article.  

Despite the widespread dissemination of information about the health risks associated with smoking, many cancer patients continue to smoke, which results in a decreased quality of life, an increased probability of cancer recurrence, and a decreased survival time. Efficacious interventions are available to assist cancer patients to quit smoking, yet smoking cessation interventions are often not implemented. This review describes how clinicians, administrators, insurers, and purchasers can encourage a culture of health care in which tobacco cessation interventions are implemented consistent with evidence-based standards of care. Implementing efficacious tobacco cessation interventions can reduce morbidity and mortality among cancer patients...

*Click on the link to the left of this introduction for a PDF of the full article.  

Publications
Publications
Topics
Article Type
Display Headline
Why do cancer patients smoke and what can providers do about it?
Display Headline
Why do cancer patients smoke and what can providers do about it?
Legacy Keywords
lung cancer, smoking, smoking cessation, smoking interventions
Legacy Keywords
lung cancer, smoking, smoking cessation, smoking interventions
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media

Dasatinib in the first-line treatment of chronic myeloid leukemia

Article Type
Changed
Fri, 01/04/2019 - 11:06
Display Headline
Dasatinib in the first-line treatment of chronic myeloid leukemia

Dasatinib has been approved for first-line treatment of chronic-phase chronic myeloid leukemia by the Food and Drug Administration and is recommended as a first-line treatment option by the National Comprehensive Cancer Network. Based on in vitro data, dasatinib seems to be less susceptible to the resistance mechanisms that affect imatinib. Dasatinib is an effective second-line treatment in patients who are resistant to imatinib. First-line clinical data show that dasatinib provides more rapid and deeper degrees of response than does imatinib, which may correlate with improvements in long-term patient outcome. Grade 1 or 2 cytopenias are the most common adverse events of first-line dasatinib treatment. In a phase 3 comparison with imatinib, several types of nonhematologic adverse events were less frequent in the dasatinib arm; frequencies of grade 3 and 4 events were   2%. Among patients with a minimum follow-up of 24 months, grade 1 or 2 pleural effusion was reported in 14% of dasatinib-treated patients and was manageable in almost all cases; no grade 3 or 4 pleural effusion occurred. Prompt and effective monitoring and management of dasatinib toxicities is essential to minimize intolerance and nonadherence to therapy. Patient education is important to increase the likelihood of prompt management and provide reassurance. Recommendations for patient monitoring, management, and education are provided.

*For a PDF of the full article, click on the link to the left of this introduction.

Article PDF
Author and Disclosure Information

Publications
Topics
Legacy Keywords
dasatinib, chronic myeloid leukemia, toxicities
Sections
Author and Disclosure Information

Author and Disclosure Information

Article PDF
Article PDF

Dasatinib has been approved for first-line treatment of chronic-phase chronic myeloid leukemia by the Food and Drug Administration and is recommended as a first-line treatment option by the National Comprehensive Cancer Network. Based on in vitro data, dasatinib seems to be less susceptible to the resistance mechanisms that affect imatinib. Dasatinib is an effective second-line treatment in patients who are resistant to imatinib. First-line clinical data show that dasatinib provides more rapid and deeper degrees of response than does imatinib, which may correlate with improvements in long-term patient outcome. Grade 1 or 2 cytopenias are the most common adverse events of first-line dasatinib treatment. In a phase 3 comparison with imatinib, several types of nonhematologic adverse events were less frequent in the dasatinib arm; frequencies of grade 3 and 4 events were   2%. Among patients with a minimum follow-up of 24 months, grade 1 or 2 pleural effusion was reported in 14% of dasatinib-treated patients and was manageable in almost all cases; no grade 3 or 4 pleural effusion occurred. Prompt and effective monitoring and management of dasatinib toxicities is essential to minimize intolerance and nonadherence to therapy. Patient education is important to increase the likelihood of prompt management and provide reassurance. Recommendations for patient monitoring, management, and education are provided.

*For a PDF of the full article, click on the link to the left of this introduction.

Dasatinib has been approved for first-line treatment of chronic-phase chronic myeloid leukemia by the Food and Drug Administration and is recommended as a first-line treatment option by the National Comprehensive Cancer Network. Based on in vitro data, dasatinib seems to be less susceptible to the resistance mechanisms that affect imatinib. Dasatinib is an effective second-line treatment in patients who are resistant to imatinib. First-line clinical data show that dasatinib provides more rapid and deeper degrees of response than does imatinib, which may correlate with improvements in long-term patient outcome. Grade 1 or 2 cytopenias are the most common adverse events of first-line dasatinib treatment. In a phase 3 comparison with imatinib, several types of nonhematologic adverse events were less frequent in the dasatinib arm; frequencies of grade 3 and 4 events were   2%. Among patients with a minimum follow-up of 24 months, grade 1 or 2 pleural effusion was reported in 14% of dasatinib-treated patients and was manageable in almost all cases; no grade 3 or 4 pleural effusion occurred. Prompt and effective monitoring and management of dasatinib toxicities is essential to minimize intolerance and nonadherence to therapy. Patient education is important to increase the likelihood of prompt management and provide reassurance. Recommendations for patient monitoring, management, and education are provided.

*For a PDF of the full article, click on the link to the left of this introduction.

Publications
Publications
Topics
Article Type
Display Headline
Dasatinib in the first-line treatment of chronic myeloid leukemia
Display Headline
Dasatinib in the first-line treatment of chronic myeloid leukemia
Legacy Keywords
dasatinib, chronic myeloid leukemia, toxicities
Legacy Keywords
dasatinib, chronic myeloid leukemia, toxicities
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media

Hospitalists Urged to Watch for Fungal Meningitis Cases in Midst of National Outbreak

Article Type
Changed
Fri, 09/14/2018 - 12:21
Display Headline
Hospitalists Urged to Watch for Fungal Meningitis Cases in Midst of National Outbreak

A national outbreak of fungal meningitis tied to contaminated steroids in epidural injections should make hospitalists vigilant with patients who present potential symptoms, says an infectious-disease (ID) specialist.

Earlier this month, health officials linked the outbreak to tainted batches of steroids used in spinal injections, and they say it could be weeks, or even months, before they know whether the incubation period for the disease is over, according to The New York Times.

"The key in my mind is that hospitalists ought to have a high index of suspicion for this right now," says hospitalist and ID expert James Pile, MD, FACP, SFHM, of the Cleveland Clinic. "If you encounter a patient you think may have meningitis, may have a brain stem stroke, may have an epidural abscess or vertebral osteomyelitis...at least think and ask the patient, or their family member, 'Did you receive an epidural steroid injection recently?'"

The answer to that question will help determine the best care delivery for hospitalists, and physicians should not rely on patients to relay the information without being asked for it, Dr. Pile says.

The outbreak has been traced to three contaminated batches of methylprednisolone produced by the New England Compounding Center in Framingham, Mass. The company, which is under criminal investigation, has been linked to at least 25 deaths and more than 317 infected patients. Although 14,000 people might have been injected with the contaminated compound, CDC officials say the likelihood of infection remains relatively low.

Dr. Pile says that while hospitalists might see only a handful of fungal meningitis cases in their careers, they still need to keep the possibility in mind when examining patients. It's a safe approach to take, particularly as the CDC continues to investigate the extent of the outbreak. The CDC has advised against antifungal prophylaxis or presumptive treatment of exposed asymptomatic patients without a diagnosed case of meningitis.

"This is just unfolding so quickly, it's a moving target," Dr. Pile adds. "How big it ends up being and what kinds of new or unusual manifestations present remain to be seen."

 

Visit our website for more information about infectious disease and hospital medicine.


 

 

Issue
The Hospitalist - 2012(10)
Publications
Sections

A national outbreak of fungal meningitis tied to contaminated steroids in epidural injections should make hospitalists vigilant with patients who present potential symptoms, says an infectious-disease (ID) specialist.

Earlier this month, health officials linked the outbreak to tainted batches of steroids used in spinal injections, and they say it could be weeks, or even months, before they know whether the incubation period for the disease is over, according to The New York Times.

"The key in my mind is that hospitalists ought to have a high index of suspicion for this right now," says hospitalist and ID expert James Pile, MD, FACP, SFHM, of the Cleveland Clinic. "If you encounter a patient you think may have meningitis, may have a brain stem stroke, may have an epidural abscess or vertebral osteomyelitis...at least think and ask the patient, or their family member, 'Did you receive an epidural steroid injection recently?'"

The answer to that question will help determine the best care delivery for hospitalists, and physicians should not rely on patients to relay the information without being asked for it, Dr. Pile says.

The outbreak has been traced to three contaminated batches of methylprednisolone produced by the New England Compounding Center in Framingham, Mass. The company, which is under criminal investigation, has been linked to at least 25 deaths and more than 317 infected patients. Although 14,000 people might have been injected with the contaminated compound, CDC officials say the likelihood of infection remains relatively low.

Dr. Pile says that while hospitalists might see only a handful of fungal meningitis cases in their careers, they still need to keep the possibility in mind when examining patients. It's a safe approach to take, particularly as the CDC continues to investigate the extent of the outbreak. The CDC has advised against antifungal prophylaxis or presumptive treatment of exposed asymptomatic patients without a diagnosed case of meningitis.

"This is just unfolding so quickly, it's a moving target," Dr. Pile adds. "How big it ends up being and what kinds of new or unusual manifestations present remain to be seen."

 

Visit our website for more information about infectious disease and hospital medicine.


 

 

A national outbreak of fungal meningitis tied to contaminated steroids in epidural injections should make hospitalists vigilant with patients who present potential symptoms, says an infectious-disease (ID) specialist.

Earlier this month, health officials linked the outbreak to tainted batches of steroids used in spinal injections, and they say it could be weeks, or even months, before they know whether the incubation period for the disease is over, according to The New York Times.

"The key in my mind is that hospitalists ought to have a high index of suspicion for this right now," says hospitalist and ID expert James Pile, MD, FACP, SFHM, of the Cleveland Clinic. "If you encounter a patient you think may have meningitis, may have a brain stem stroke, may have an epidural abscess or vertebral osteomyelitis...at least think and ask the patient, or their family member, 'Did you receive an epidural steroid injection recently?'"

The answer to that question will help determine the best care delivery for hospitalists, and physicians should not rely on patients to relay the information without being asked for it, Dr. Pile says.

The outbreak has been traced to three contaminated batches of methylprednisolone produced by the New England Compounding Center in Framingham, Mass. The company, which is under criminal investigation, has been linked to at least 25 deaths and more than 317 infected patients. Although 14,000 people might have been injected with the contaminated compound, CDC officials say the likelihood of infection remains relatively low.

Dr. Pile says that while hospitalists might see only a handful of fungal meningitis cases in their careers, they still need to keep the possibility in mind when examining patients. It's a safe approach to take, particularly as the CDC continues to investigate the extent of the outbreak. The CDC has advised against antifungal prophylaxis or presumptive treatment of exposed asymptomatic patients without a diagnosed case of meningitis.

"This is just unfolding so quickly, it's a moving target," Dr. Pile adds. "How big it ends up being and what kinds of new or unusual manifestations present remain to be seen."

 

Visit our website for more information about infectious disease and hospital medicine.


 

 

Issue
The Hospitalist - 2012(10)
Issue
The Hospitalist - 2012(10)
Publications
Publications
Article Type
Display Headline
Hospitalists Urged to Watch for Fungal Meningitis Cases in Midst of National Outbreak
Display Headline
Hospitalists Urged to Watch for Fungal Meningitis Cases in Midst of National Outbreak
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)