Clinical Update on Nursing Home Medicine: 2011

Clinical Update on Nursing Home Medicine: 2011

UPDATES FROM THE AMDA MEETING Clinical Update on Nursing Home Medicine: 2011 Barbara J. Messinger-Rapport, MS, PhD, FACP, CMD, John E. Morley, MB, BC...

326KB Sizes 5 Downloads 211 Views

UPDATES FROM THE AMDA MEETING

Clinical Update on Nursing Home Medicine: 2011 Barbara J. Messinger-Rapport, MS, PhD, FACP, CMD, John E. Morley, MB, BCh, David R. Thomas, MD, FACP, AGSF, FGSA, CMD, and Julie K. Gammack, MD

This represents the fifth article in the series on yearly updates of hot topics in long term care (J Am Med Dir Assoc 2011; 12: 615–626)

Keywords: Osteoporosis; Diabetes mellitus; Weight loss; Anticoagulants; Proton pump inhibitors; Delirium; Polypharmacy

PHARMACOLOGICAL OSTEOPOROSIS MANAGEMENT

cer patients. The latter risk of typical femur fracture rises after 5 years of use, but is rare, with an incidence of 2.3 fractures per 10,000 patient-years.13 Although some advocate a ‘‘drug holiday’’ after 5 years of use, evidence for optimal duration of therapy and use of a drug holiday is not available, and the benefit of ‘‘typical’’ femur fracture risk reduction continues past 5 years.14 Denosumab (Prolia) is a recently approved osteoporosis drug which has been shown to reduce the risk of hip fractures in osteoporotic women by 40%15 and vertebral fractures in men with androgen deprivation by 62%.16 A monoclonal antibody, it is a member of the TNF superfamily and binds to the receptor activator of nuclear factor B ligand (RANKL). Cleared by nonrenal mechanisms, it can be administered to persons with CKD, though there is inadequate data on end stage renal failure. It is given subcutaneously twice yearly. Given the potential to suppress immune function, its role in routine osteoporosis treatment in frail elders who may be vulnerable to infection is limited at this time until more postmarketing studies are available. Vitamin D levels are low in the majority of nursing home residents.17–25 Daily Vitamin D, regardless of calcium intake, has been shown to reduce the rate of falls and fracture in older adults26 but the optimal dose and frequency are not known. Very high doses separated by long intervals (for example, 500,000 units yearly) was associated with an increase in falls and fracture in 1 study.27 The value of calcium supplementation above and beyond dietary intake when there is adequate vitamin D supplementation is likely low.26 Additionally, calcium supplements are associated with nephrolithiasis,28 advanced and fatal prostate cancer,29 and can prevent absorption of medications often used in older adults, including floroquinolones, ketoconazole, phenytoin, levo-thyroxine, iron supplements, gabapentin, and others. Recent studies30–32 found an increased cardiovascular risk to supplemental calcium at doses typically used in older adults (600–2000 mg/d). Estrogens and selective estrogen receptor modulator increase bone density and are associated with vertebral fracture

Osteoporosis and hip fractures are extremely common in nursing home residents and are associated with increased disability and mortality.1–10 Despite the plethora of drugs for preventing and treating osteoporosis, there are only 2 drugs which demonstrate an outcome of lowered risk of hip fracture: bisphosphonates, and denosumab. Bisphosphonates reduce the risk of hip fracture by 25–40%, with a NNT of 20–200 depending upon the study and the risk of fracture.11 Bisphosphonates can be administered orally on a daily, weekly, monthly, or quarterly basis, or IV either quarterly (ibandronate) or yearly (zolendronate). Conditions seen in older patients, such as esophageal varices, severe kyphosis, dysphagia, and dementia (impaired oropharyngeal coordination, lack of compliance with upright restrictions, or both) limit the efficacy and safety of the oral versions. The high prevalence of chronic kidney disease (CKD) in the older population limits this drug as well, since it is not approved for use in CKD 4 or 5. Off-label use in those with advanced CKD in the absence of biochemical markers for CKD-associated bone mineral disorders has emerged but long-term and prospective data are absent.12 Potential and emerging adverse effects include osteonecrosis of the jaw and atypical femur fracture. The former risk is seen mainly with IV pamidronate or zolendronic acid in canCleveland Clinic, Section of Geriatric Medicine, Cleveland, OH (B.J.M.-R); Division of Geriatric Medicine, Saint Louis University School of Medicine, St. Louis, MO (J.E.M., D.R.T., J.K.G). This represents the fifth article in the series on yearly updates of hot topics in long-term care. Address correspondence to John E. Morley, MB, BCh, Division of Geriatric Medicine, Saint Louis University School of Medicine, 1402 S. Grand Boulevard, M238, St. Louis, MO 63104. E-mail: [email protected]

Published by Elsevier Inc. on behalf of the American Medical Directors Association, Inc. DOI:10.1016/j.jamda.2011.08.007 UPDATES FROM THE AMDA MEETING

Messinger-Rapport et al 615

risk reduction as primary outcomes.11 However, estrogen therapy is associated with an excess risk of stroke despite reduction of hip fracture risk.33 Oral estrogen/progesterone combination treatment is associated with an increased risk of coronary heart disease, breast cancer34 and dementia.35 Fracture risk is based primarily on bone density (the World Health Organization utilizes a cut-off of T #2.5 standard deviations). When the bone density does not reach this low level but there is concern, 1 risk tool to consider is the FRAX.36 A FRAX score of at least 3% for hip and 20% for major osteoporotic fractures suggests benefit to pharmacological intervention. This score incorporates bone density, gender, race, age (up to 90), use of alcohol and tobacco, body mass index, glucocorticoids, personal and family history of fracture, secondary causes of osteoporosis. It expands treatment to persons who might not reach the WHO cutoff, and reassures clinicians that some with osteopenia may not need treatment at the current time. The problem with the FRAX score and other standardized risk models such as the Fracture Risk Calculator (FRC) is that many conditions and medications common to geriatric patients increase the risk of falls, thus increasing the risk of fracture. Examples of conditions that increase the risk of falls include visual impairment, orthostatic hypotension, diabetes (with vision impairment and/or neuropathy and/or recurrent hypoglycemic episodes), Parkinson’s disease, and dementia. These conditions are not included in current risk calculators. Older diabetics may have neuropathy, which increases the risk of falls, or may have overt or subclinical hypoglycemia due to medications, increasing the risk of confusion and falls. Movement disorders of all types, particularly Parkinson’s disease and parkinsonian conditions, increase the risk of fall and fracture. The reduced attention span and judgment found in dementia lead to falls. The relatively sedentary lifestyles of these patients may increase the rate of decline of bone density faster than the risk calculators account for as well. In summary, fracture prevention in frail elders, particularly those with dementia and those in long-term care, is based primarily on adequate calcium intake (up to 1000 mg/d), adequate vitamin D intake (800–1000 units daily), maintenance of mobility and balance, appropriate treatment of diseases that predispose to falls, and medication parsimony to reduce falls and limit the decline in bone density. Oral bisphosphonates in frail elders requires judicious monitoring if the patient has severe kyphosis, dysphagia, or is unreliable in taking it in the upright position. Yearly IV infusion of zolendronate may be helpful in mobile but frail elders who may be at risk with the oral route of bisphosphonates. Denosumab is too new to be considered for routine use in mobile, frail elders who may be vulnerable to an as yet unclear immunosuppressive effect.

fecal incontinence.38–44 Diabetics also have a higher rate of falls, fractures, renal failure, depression and mobility impairment.45–48 For all of these reasons there is a need to focus on appropriate diabetic care.49,50 Diabetics tend to have better outcomes when systolic blood pressure is reduced below 160 mm Hg compared to nondiabetics.51 Most available data suggests that controlling the blood pressure around 145 mm Hg is optimal with no evidence to support going below 130 mm Hg.52 Orthostatic hypertension and postprandial hypotension need to be assessed in all persons with diabetes mellitus.53,54 Persons with postprandial hypotension can be treated with alpha-lglucosidase inhibitors such as miglitol or acarbose.55 Acarbose also decreases the occurrence of hypoglycemia.56 The 25(OH) vitamin D levels are often low in diabetics.57 Replacement of vitamin D may improve diabetic control, reduce hypertension, decrease falls and fractures and increase muscle strength.58 Depression is common is persons with diabetes and needs to be vigorously treated.46 Both alphalipoic acid and topiramate have been shown to be efficacious in treating painful peripheral neuropathy.59,60 Studies in general have found that in the United States persons with diabetes have HbA1C levels \7 in 67–89% of residents.61–63 This becomes disturbing when it is recognized that the 3 recent major diabetes trials (ACCORD, ADVANCE and the Veterans Administration Diabetes Trial) found no effect on cardiovascular disease or total mortality when HbA1C was controlled to below 7.0.64–66 Currie et al,67 using a United Kingdom database in a general population, found that in persons on 2 antidiabetic medicines, optimal survival occurred at a HbA1C of 7.5 and in persons on insulin at an HbA1C of 7.9. Falls in older diabetics increase markedly when the HbA1C falls below 7.68 Thus, it would seem that in the nursing home diabetic control should be aimed at maintaining the HbA1C at levels between 7 and 8. Weight loss has been shown to be associated with increased mortality in older persons with diabetes.69 Therapeutic diets have failed to be shown to be of benefit in diabetics in nursing homes.70,71 For this reason, the American Diabetic Association diets in long-term care have been retired in favor of liberalized diets which may be individualized.72 Numerous studies support the concept of both aerobic and resistance exercise to improve diabetic control.73,74 In addition, exercise programs in nursing homes have been shown to improve function, decrease injurious falls, decrease depression, improve cognition and decrease agitated behavior.75–77 Postprandial walking has been found to have better effects on glucose control than walking before meals.78 In older diabetics, every 1-MET increase in exercise activity resulted in a 16% lower mortality.79

MANAGEMENT OF DIABETES MELLITUS

Oral drugs have been the mainstay of modern treatment for type 2 diabetes mellitus.80 Of these only metformin has been shown to decrease mortality and myocardial infarction.81 It needs to be recognized that its effects on atherosclerotic disease may be independent from its glucose lowering effects and related to its ability to increase metric oxide synthase and

Based on national surveys the prevalence of diabetes mellitus in nursing homes has increased to 26.4% in men and 22.2% in females.37 Diabetics in nursing homes tend to have a greater disease burden, are more likely to be frail and have cognitive decline, and have an increase in aspiration pneumonia and 616 Messinger-Rapport et al

Choice of Hypoglycemic Agent in Older Adults

JAMDA – November 2011

thus alter blood flow.82 Metformin produces anorexia and weight loss and when severe, may need to be discontinued.83 While there is a fear of lactic acidosis, metformin can be used in persons over 80 years old provided they do not have chronic kidney disease. Thiazolidinediones appeared to be potentially excellent drugs to manage diabetes in older persons. Unfortunately, it is possible the rosiglitazone has a negative effect on cardiovascular disease.84 In addition, thiazolidinediones decrease osteoblast function, increase bone loss and increase differentiation of mesenchymal stem cells towards adipocytes rather than osteoblasts through increased PPARg activation.85,86 In the ADOPT trial thiozolidendiones increased fracture risk in women.87 Rosiglitazone decreases bone mineral density and markers of bone formation in humans.1 This effect of thiazolidinediones on increasing osteoporosis makes these drugs inappropriate for use in older nursing home residents. Glucagon-like peptide I (GLP-I) is an incretin that stimulates insulin and decreases glucagon, It also inhibits hepatic glucose production and produces anorexia and delayed gastric emptying. Glucagon-like peptide 7 levels are decreased in type 2 diabetics. Analogs of GLP-I, originally derived from the gila monster, have been developed to treat obese type 2 diabetics. As they cause major weight loss they should be avoided in the nursing home. Interestingly alpha-1glucosidase inhibitors (i.e., acarbose and miglitol) both increase GLP-I.88,89 More recently dipeptydyl IV inhibitors have been developed.90 These drugs increase GLP-I levels by blocking their breakdown. Both sitagleptin and vildagleptin lower HbA1C by 0.65–0.8%. Side effects include tremor and rarely a severe skin rash. Unlike the alpha-1glucosidase inhibitors, they do not reduce weight. Saxagliptin is metabolized by the CYP system and has a high risk of drug– drug interaction.91 Sulfonylureas are the oldest of the antidiabetic drugs. They all produce hypoglycemia.91 They act by increasing insulin. The evidence suggests that in the long-term they may have negative effects on the cardiovascular system. As such, they are no longer first line drugs for older persons. In the nursing home insulin should be substituted for oral agents when the HbA1C cannot be maintained below 8.92 The choice of insulin depends more upon affordability and adverse effect than efficacy. Studies which compare NPH with the 2 newer, more expensive, but more truly ‘‘basal’’ insulins, detemir and glargine, suggest that all 3 are similarly efficacious in lowering HbA1C.93–95 However, there is less hypoglycemia, particularly nocturnal hypoglycemia, with the newer agents, detemir and glargine. In frail elders predisposed to cognitive impairment and falls, preventing hypoglycemia is essential. Some frail elders on NPH with or without Regular humulin may be better served on detemir or glargine. Additionally, transitioning persons with NPH plus a ‘‘sliding scale’’ of regular humulin to a basal regimen may reduce hypoglycemia as well as facilitate a simpler but as efficacious diabetic treatment regimen. Glargine may be the easiest basal insulin to utilize in a long-term care setting due to its pharmacodynamics. Caution should be taken UPDATES FROM THE AMDA MEETING

with frail elders, however, since if a person becomes hypoglycemic, the episode can be prolonged.96 The approach to residents in the nursing home should be to use generic metformin, when it is safe to use. The second choice would be between a generic sulfonylurea (glipizide, glyburide, or glimipride) or the much more expensive DPPIV inhibitors. If the resident has postprandial hypotension, acarbose or miglitol should be used. When the resident is on 2 oral agents and the HbA1C is greater than 8, insulin glargine should be started and the oral antidiabetic drugs stopped. Utilizing more than 2 antidiabetic drugs leads to polypharmacy and multiple studies have demonstrated the dangers of polypharmacy in the nursing home.97–100 A number of new drugs are under development to treat diabetes. Among those closest to clinical use is the antianginal sodium channel modulator101 and Depaglifozin, a SGLT-2 antagonist, that inhibits glucose reabsorption in the kidney tubules.102 Finally, all nursing homes should have a policy for glucose monitoring and regularly undertake a quality improvement program on diabetes.103 Identifying and Reaching the Glycemic Target in a Facility The spectrum of diabetes in LTC includes newly diagnosed disease in robust individuals undergoing rehabilitation as well as advanced disease in frail elders. The target intensity of glycemic control varies depending upon the comorbidities, medical goals, and overall personal goals. Additionally, the ability to monitor and deliver the care needed to accomplish the desired glycemic control requires accommodating the facility, and engaging the team of clinicians, pharmacy consultants, emergency contingency kit, nursing aides, nurses, patients, families, and information technology consultants, where appropriate. The glycemic goal of 6.5% or less in type 2 Diabetes is not supported by interventional trials in older adults.66,104,105 except, possibly, in the subset of those with shorter duration of disease and less total burden of calcium.106 Older adults with early disease and active lifestyles might be able to maintain this target safely. For most elders, a HbA1C goal of 7– 7.5% is appropriate. Higher HbA1C targets (less than 8%, or even \ 9%) would be appropriate for those persons with a shorter life expectancy, where the goals of care are to reduce symptoms from geriatric syndromes and limit hospitalization. When the HbA1C is approximately 6%–9%, postprandial glucose is a better measure of control than fasting glucose,107 and can be incorporated into periodic monitoring. Fasting blood glucose contributes a greater predictive value as glycemic control worsens. Early type 2 diabetics managed with ‘‘diet’’ or sensitizers (glitazones or metformin) do not need any scheduled fasting capillary blood glucose (CBG) testing; it is very unlikely that they will become iatrogenically hypoglycemic. A postprandial CBG periodically (weekly is reasonable) reflects HbA1C when the patient has reasonable glycemic control.89 The postprandial CBG is typically 30–50 mg/dL higher than the preprandial value, but will vary depending upon the meal and the timing of the postprandial test. Combined with Messinger-Rapport et al 617

a premeal blood glucose, the difference offers insight into both mealtime carbohydrate intake and overall glycemic control. Once a pattern of postprandial CBG is established, it can be used as a therapeutic target to reduce or liberalize the HbA1C. Longstanding type 2 diabetes—glycemic control is more difficult than in early diabetes, requiring 1 or 2 oral medications or a basal insulin injection plus or minus a sensitizer daily. These persons typically have been diagnosed with diabetes for several years (their actual disease duration may be unknown). A regimen for a fairly stable patient on 1 or 2 hypoglycemic agents might consist of a fasting CBG Monday, Wednesday, and Friday to identify a pattern of iatrogenic hypoglycemia, and a postprandial CBG Tuesday and Thursday to monitor efficacy and diet. Patient/staff can check additional blood glucose as symptoms indicate. Additionally, the older regimen of a sliding scale of regular humulin should be discouraged, since it treats elevated blood glucose reactively, rather than proactively.92,108 Because of the relatively long half-life of regular humulin, the person may become hypoglycemic prior to the next meal. The patient and nursing staff may need guidance that an elevated postprandial CBG does not, in general, require action to immediately lower the level. Notification of the clinician is performed to review the recent pattern of CBG and to adjust the basal dose, which will lower the CBG over the next 1–2 weeks. Type 1 diabetes is frequently seen in those receiving rehabilitation at a facility. They usually need 1–2 daily doses of a basal insulin and plus prandial doses of rapid-acting insulin in order to maintain glycemic control. Adjusting preprandial or postprandial rapid-acting insulin dosing to intake and preprandial glucose measurements makes the management more complicated but may be required for this individual. Episodic postprandial CBG can be helpful to verify mealtime carbohydrate intake and overall glycemic control. Advanced or ‘‘end-stage’’ type 2 diabetes with blood glucose that is difficult to control may needed to be treated as if they were a type 1 diabetic, with 1–2 daily doses of a basal insulin plus prandial doses of rapid-acting insulin according to consumption and possibly adjusted to CBG. These persons have had diabetes, diagnosed or not, for many years. Many have gastroparesis, resulting in wild glucose swings. Addition of metoclopramide or, outside of the United States, domperidone, may be helpful. End of life: A patient with longstanding type 2 diabetes close to the end of life may still require hypoglycemic drugs and monitoring in order to stay comfortable. Uncontrolled diabetes may impair comfort if it leads to thirst, dehydration, infections, urinary incontinence, and falls. A sample palliative treatment plan might include 1 hypoglycemic agent daily (whether oral or injectable insulin) with a capillary blood glucose (CBG) a few times weekly, perhaps alternating a fasting measurement (to verify lack of iatrogenically induced hypoglycemia) with a measurement 2 hours after the largest meal (to verify efficacy). Verification of control with a serum HbA1C would not be necessary if the goal is symptom management. Hypoglycemic agents could be adjusted to keep 618 Messinger-Rapport et al

the fasting BG greater than 100 and the postprandial BG less than 350. Many patients, nurses, and physicians are accustomed to a ‘‘sliding scale’’ prior to mealtime and bedtime for the diabetic patient. In fact, facilities may have very few persons requiring complicated administration and glycemic monitoring regimens once the target HbA1C and postprandial CBG are explicitly identified. Those needing an intense level of monitoring are typically the type 1 diabetics (particularly in the postacute unit receiving skilled services until they return home) and the ‘‘end stage’’ type 2 diabetics with wildly (and often dangerously) fluctuating blood glucose. Most elders with diabetes have type 2 diabetes in an early or middle stage. These persons can be managed with diet, or a sensitizer, or with 1–2 oral agents, or 1 daily dose of basal insulin, with monitoring needed 1–7 times per week. Understanding facility limitations will help staff design safe environments for glycemic control, particularly for those few persons judged to require a complex administration and monitoring regimen. The inability to administer injectibles in the dining room may limit the ability to provide rapidacting insulin safely. A patient receiving rapid-acting insulin at an interval greater than the package insert lists for proximity to a meal may be at risk for harm or, ineffectiveness. Additionally, the facility may risk citation. The accuracy of the aide in assessing meal consumption is limited and is a barrier to fine-tuning rapid-acting insulin administration. Aides vary by ability and estimated meal consumption is typically higher than actual.109 On the other hand, registered nurses are taught carbohydrate counting (as grams or servings) as part of their training and facilities can incorporate carbohydrate counting when appropriate for adjusting rapid-acting insulin dosing for type 1 or brittle type 2 diabetics. The facility dietician plays a key role in educating aides and nurses in appropriate estimates of consumption. In summary, intensity of glycemic control is established by the attending physician, with input from patients, family, and facility staff. Target of the HbA1C and complexity of CBG management follow from the goals of care. Most older diabetics can be managed with a liberal diet and an oral hypoglycemic or simple basal regimen. The few patients in a facility requiring more complicated management need a team approach to their diabetic care. Non– physician providers can educate the patient, nurses, and families regarding goals and management and the engaged team works together to implement a comprehensive and individualized glycemic administration and monitoring regimen. WEIGHT LOSS Weight loss is extremely common in persons following hospitalization and in the nursing home for rehabilitation.110 It is now generally recognized that weight loss in old frail individuals is strongly related with increased mortality.111–113 Weight loss also leads to increased institutionalization114 hip fracture,115 pressure ulcers, infections, poor cognition, JAMDA – November 2011

anemia, muscle weakness, fatigue and edema.116 Weight loss was recognized by the IAGG as a major area for further research in the nursing home.117 Older persons develop a physiological anorexia of aging.118 This has multiple causes including loss of taste and smell,119 alterations in fundal compliance,120,121 increased cholecystokinin levels and activity122 and in males low testosterone leading to elevated leptin levels.123 These appear to interact within the central nervous system with a decline in the opioid feeding system.124,125 Anorexia has been shown to independently predict mortality.126 There are major causes of weight loss viz., anorexia, cachexia, malabsorption, hypermetabolism, dehydration and sarcopenia.127 Anorexia can be detected early by using the Simplified Nutrition Assessment Questionnaire (SNAQ).128 This has been validated in a nursing population. The MiniNutritional Assessment is an excellent screening test for malnutrition.129–132 It also tends to recognize persons with frailty.133,134 Serum proteins such as albumin, prealbumin and transthyretin are poor indicators of nutritional status as these levels are markedly decreased in the presence of cytokine excess.135 In the nursing home depression is the most common cause of anorexia and weight loss.136–138 Almost a third of nursing home residents who lose weight are depressed. A simple list of the causes of reversible weight loss is the MEALS-ONWHEELS mnemonic (Table 1). Cachexia is generally due to proinflammatory cytokine excess and results in a loss of both muscle and adipose tissue.139–143 The increase in cytokines is usually related to underlying disease processes such as congestive heart failure, chronic obstructive pulmonary disease, kidney disease or cancer.144,145 Sarcopenia is muscle loss associated with aging.146,147 Newer definitions have stressed that for it to be considered a geriatric syndrome it should be associated with loss of strength, power and/or mobility.148–150 Sarcopenia is not always associated with weight loss as muscle loss may be replaced by fat – the obese sarcopenic or ‘‘fat frail’’ syndrome.151,152 Dehydration is a not uncommon cause of weight loss that is often forgotten.153,154 An algorhythm for the management of weight loss in the nursing home has been published.155 A key to preventing weight loss in nursing homes is having an attractive dining ambience.156 Ice cream parlors and snack carts can also help to maintain resident weight.157 A meta-analysis has shown that protein and energy supplementation in elderly malnourished persons decreases mortality and length of hospitalization.158 Oral liquid supplements need to be given between meals to be optimally effective.159 The Intercom Trial showed that an early nutritional intervention in COPD continued for 24 months improved weight, fat free mass, muscle strength and 6 minute walking distance and decreased hospitalization.160 Neelemaat et al161 showed that a 3-month nutritional intervention, including an energy and protein enriched diet, resulted in improved function 3 months after discharge from hospital. A leucine enriched essential amino acid UPDATES FROM THE AMDA MEETING

Table 1. MEALS-ON-WHEELS Mnemonic for Treatable Causes of Weight Loss in Older Persons Medications (e.g., digoxin, theophylline, cimetidine) Emotional (e.g., depression) Alcoholism, elder abuse, anorexia tardive Late life paranoia Swallowing problems Oral factors Nosocomial infections (e.g., tuberculosis) Wandering and other dementia related factors Hyperthyroidism, Hypercalcemia, Hypoadrenalism Enteral problems (e.g., gluten enteropathy) Eating problems Low salt, low cholesterol and other therapeutic diets Stones (cholecystitis)

supplementation is considered the supplement most likely to enhance muscle strength.162 Megestrol acetate is the drug most effective at producing weight gain.113,163 It also decreases cytokine activity.164 It tends to increase predominantly adipose tissue. There is no evidence that it improves function or mortality. As megestrol acetate is poorly absorbed in the fasting state, a megestrol acetate nanocrystal disperson oral suspension has been developed and has much improved absorption in the fasting state.165 Thus, in persons with low food intake, the megace ES form should be used for the first month. Dronabinol is a tetrahydrocannabinoid that increases food intake.166,167 It produces minimal weight gain. It is an excellent drug for palliative care. Testosterone levels decline markedly with age.168 Recently studies have found that testosterone replacement may increase muscle mass and walking speed and decrease hospitalizations in persons with frailty and heart failure.169–173 The major side effect is increased edema. A number of new drugs are under development to enhance food intake and improve weight gain. These include selective androgen receptor molecules, ghrelin agonists and a novel peptide–nucleic acid compound.174–176 It would appear that tube feeding, especially G-tubes, are grossly overused in demented elderly persons.177,178 Ethical thought processes fail to support the use of G-tubes in most demented older persons. CLINICAL USE OF ANTICOAGULANTS The use of anticoagulants is a major problem in nursing homes.179–181 In part, this is related to drug interactions, many of which, such as the acetaminophen–warfarin interaction, are underappreciated.182 New data and new drugs drive a reevaluation of use of anticoagulation in clinical practice. An overview of several frequent clinical questions follows. Is Warfarin Superior to Antiplatelet Agents in Patients With Atrial Fibrillation? Most guidelines recommend warfarin therapy for nonvalvular atrial fibrillation in persons at high risk for stroke, and antiplatelet agents (chiefly aspirin) for persons considered Messinger-Rapport et al 619

to either be at low risk of stroke or for those persons who cannot safely receive warfarin therapy. From the standpoint of long-term care physicians, most older persons are judged to be a high risk, but also judged to be more likely not to be able to safely take adjusted dose warfarin therapy. There are several risk stratification schemes to predict future stroke risk, but the cut points and specificity are somewhat controversial. A meta-analysis of 8 trials comparing adjusted dose warfarin to antiplatelet therapy in persons with nonvalvular atrial fibrillation and no prior history of stroke or transient ischemic attack demonstrates that warfarin is superior to antiplatelet agents for the prevention of all stroke, ischemic stroke, and systemic emboli, but is not different for fatal stroke, myocardial infarction, vascular death, or all cause death. The incidence of an intracranial bleed is higher with warfarin compared to aspirin.183 In relative terms warfarin reduces the risk of stroke by about 30% and aspirin reduces the risk by about 20%. However, the incidence of stroke in the studied populations is relatively small. Thus, the absolute risk reduction is estimated to be about 1%; that is, 100 persons would be treated for 1 year to prevent 1 additional stroke. In persons who have a history of prior stroke or transient ischemic attack, warfarin is clearly superior to antiplatelet agents alone for the secondary prevention of recurrent stroke.184 A useful risk assessment tool for secondary prevention is the CHADS2 score.185 With a history of prior stroke or transient ischemic attack and age over 75 years, the CHADS2 score would be 3 (without any additional risk factors), and the relative risk for stroke would be 6% per 100 patient-years. The addition of congestive heart failure, hypertension, and diabetes further increases the risk to about 18 per 100 patient-years. A new direct thrombin inhibitor is available for treatment. In direct comparisons, noninferiority has been demonstrated for the prevention of stroke or systemic embolization compared to warfarin (1.1% versus 1.5%), and no difference in total mortality was observed. Hemorrhagic stroke occurred less often in the dabigitran group (0.1% versus 0.4%), and fewer bleeding complications were observed in the dabigitran group compared to warfarin (2.7% versus 3.4%).186 Dabigitran offers the advantage of not requiring venous blood sampling or dose adjustment, but also has the disadvantage of being highly dependent on renal function and does not have a rapid antidote in the presence of bleeding. One of the chief reasons cited for avoiding anticoagulation in older persons is the fear of intra cranial bleeding or other bleeding injury from falling. Elderly persons who fall have a mean of 1.8 falls per year. At this rate, decision analysis techniques suggest that the risk of subdural hematoma must be 535-fold or greater for the risks of warfarin therapy to outweigh the benefits. Persons taking warfarin must fall about 295 times in 1 year for warfarin to not be the optimal therapy.187 In terms of quality-adjusted life-years (QUALYs) per patient in persons with average risks of stroke and falling, warfarin therapy was associated with 12.90 QALYs, aspirin therapy with 11.17 QUALYs, and no antithrombotic therapy with 10.15 QUALYs. 620 Messinger-Rapport et al

What About Primary Prevention in Patients With Normal Sinus Rhythm? Primary and secondary prevention in persons with atrial fibrillation is clearly beneficial. Primary prevention in persons without atrial fibrillation (normal sinus rhythm) is much more problematic. Two studies examined the benefit of several prevention strategies with the outcome of death, nonfatal myocardial infarction, and stroke. There were no differences in groups treated with aspirin, warfarin, or no treatment in the first trial,188 or between aspirin, warfarin, or clopidrogel in the second trial.189 In a trial of patients at high risk for a cardiovascular event, clopidrogel plus low-dose aspirin (75–162 mg) was compared to placebo plus low-dose aspirin. At 28 months, the incidence of myocardial infarction, stroke, or cardiovascular death was not different between groups.190 In a subset analysis of this trial, no difference in stroke or stroke and transient ischemic attack was observed between groups.191 This data suggest that clinicians should not add clopidogrel to standard long-term aspirin therapy for preventing cardiovascular or neurological events in people at high risk of cardiovascular disease and in those with established cardiovascular disease.192 How Long Should Antiplatelet Therapy Be Continued in Patients With Stents? Stent thrombosis most commonly occurs in the first month after stent implantation, classified as ‘‘subacute stent thrombosis.’’ However, numerous cases of ‘‘late’’ stent thrombosis, particularly in patients who have been treated with drug eluting stents, have been described as occurring months or even years after stent implantation. In the majority of cases, stent thrombosis is a catastrophic event, resulting in life-threatening complications.193 In a trial which followed subjects with stent placement for 12 months, current use of clopidrogrel at either 6 or 12 months was associated with a reduction in stent thrombois in drug-eluting stents, but demonstrated no difference in persons receiving bare metal stents. Therefore, suggested recommendations for treatment include clopidogrel 75 mg and aspirin 325 mg at a minimum of 1 month after bare-metal stent implantation; 3 months after sirolimus drug eluting stent (DES) implantation; 6 months after paclitaxel DES implantation; and ideally, up to 12 months if the patient is not at high risk for bleeding.193 In patients with acute non–ST elevation myocardial infarction (NSTEMI), there is evidence of benefit outweighing harms caused by major bleeding with aspirin plus clopridrogel. Subjects were randomized to clopidogrel (loading dose, 300 mg orally, followed by 75 mg/day or to placebo plus aspirin (75–325 mg/day). For every 1000 persons treated for 9 months, 23 cardiovascular events were prevented and 10 major bleeds were caused in the clopridogrel group.194 It has been hypothesized that certain groups of patients with high platelet reactivity might be at higher risk for DES stent occlusion, and thus higher loading doses of clopridogrel may be more effective. Subjects with high in vitro platelet reactivity at 24 hours after DES placement were randomized to JAMDA – November 2011

a 600 mg loading dose of clopridogrel followed by 150 mg daily plus aspirin 75–162 mg daily or to placebo loading dose of clopridogrel followed by 75 mg daily plus aspirin 75–162 mg daily. At months of followup, both groups experienced the same rate of cardiovascular death, nonfatal myocardial infarction, or stent thrombosis (2.3%).195 In summary, warfarin is superior to aspirin for the prevention of stroke and systemic emboli in persons with atrial fibrillation. Withholding warfarin for patients who have a falling history is overdone, particularly in persons with a high CHADS2 score. However, therapy can be individualized, since the absolute risk reduction for primary prevention of stroke is small. Darbigitran is equivalent to warfarin for primary prevention in atrial fibrillation. There is no advantage of aspirin or other anticoagulants over no therapy for primary prevention of cardiovascular events in patients who are in normal sinus rhythm. There is no benefit for the addition of clopidogrel to aspirin for the primary prevention of stroke or transient ischemic attack. The addition of clopidogrel to aspirin therapy provides benefit in patients with NSTEMI. The duration of therapy with clopidogrel and aspirin varies with the type of stent, but when possible, should be continued for 12 months after drug-eluting stent placement. DEADLY PROPHYLACTIC PROTON PUMP INHIBITORS? The development of proton pump inhibitors has been a boon to the treatment of gastrointestinal diseases. Proton pump inhibitors (PPIs) are superior to placebo or to histamine-2 receptor antagosists (H2RAs) in healing peptic ulcers. PPIs reduce rebleeding following endoscopic therapy for bleeding ulcers by 54% and reduce the need for surgical intervention by 41%. Symptoms in gastroesophageal reflux disease are improved in 65–75% of patients by 8 weeks, and healing of erosive gastritis has been demonstrated in 85– 95% of patients by 8 weeks. However, symptomatic relief in nonerosive esophagitis is lower, around 37% at 4 weeks. PPIs are superior to double-dose H2RAs or placebo controls in reducing nonsteroidal anti-inflammatory drug induced gastritis, leading to widespread prophylactic use.196 Another widespread area of prophylactic use has been in Table 2.

the prevention of stress-induced ulcers in critically ill patients. Clinically important bleeding occurs in 1.5% of critically ill patients, but differs considerably by patient risk factors. Bleeding occurs in 3.7% of patients with respiratory failure (odds ratio 15.6) and coagulopathy (odds ratio 4.3), but in only 0.1% of patients without these risk factors.197 This low incidence may explain nonsignificant differences in prophylaxsis clinical trials comparing PPIs, H2RAs, and sucalafrate to placebo.198 No difference in the incidence of ventilator acquired pneumonia has been observed comparing patients treated with PPI, H2R, or sucalafrate to control subjects.199 No difference in incidence of pneumonia or mortality has been observed in patients treated for stress ulcer prophylaxsis comparing a PPI versus a H2R.200 The widespread use of PPIs in critical care settings has led some authors to conclude that prophylactic use is frequently inappropriate. Classification of appropriate use is shown in Table 2. Using these definitions, 71% of subjects receiving intravenous PPIs in a teaching hospital did not meet appropriate use criteria. Once started, PPIs are continued, mostly by intravenous route, in about 50% of subjects after being discharged to nonintensive care settings.201 The data suggest that PPIs should only be continued until the patient either begins an oral diet or until discharge from the critical care setting.202 The risk of clinically important bleeding outside of the critical care setting is considered too low to justify continued prophylaxsis.203 Outside of the acute setting PPIs are often used for years at a time. Indeed, estimates suggest that 40% of the US population receives a PPI chronically.204 Recent concerns about interactions of PPI with drugs and with geriatric diseases are relevant to long-term care physicians. Omeprazole and Clopidrogel Interaction? An potential interaction with omeprazole and clopidrogel was suggested by the observation that men taking both drugs after an acute myocardial infarction had a higher recurrence rate 90 days after hospital discharge compared to men not taking omeprazole.205 In another trial, using a unique

Appropriate and Nonappropriate use of PPIs

Appropriate Use in UGIB

Appropriate Use in UGIB

Inappropriate Use

After endoscopic hemostasis of gastrointestinal bleeding

Stress ulcer prophylaxes in NPO patients with either mechanical ventilation or coagulopathy Severe dysmotility or illeus

Stress ulcer prophylaxes in patients not meeting criteria

Suspected UGIB 12 hours prior to endoscopy Confirmed endoscopic UGIB for 72 hours High risk for UGIB for 72 hours

Zollinger-Ellison syndrome History of esophagitis in NPO patient

Nausea or vomiting Suspected esophageal varices bleed Probable gastritis Lower gastrointestinal bleed History of peptic ulcer disease Esophagitis, not NPO No indication, not NPO Standard prophylaxes in ICU

UGIB 5 upper gastrointestinal bleeding; NPO 5 nothing by mouth. Adapted from: Hoover JG, et al. Use of intravenous proton-pump inhibitors in a teaching hospital practice. Dig Dis Sci 2009;54:1947–1952.219

UPDATES FROM THE AMDA MEETING

Messinger-Rapport et al 621

preparation of omeprazole, the study was halted before completion. However, no difference in cardiovascular events were observed.206 Although controversial, there is a clear invitro effect of omeprazole on platelet function, and no association has been reported for other PPIs such as pantoprazole. PPIs and Community-Acquired Pneumonia? The second line of the body’s defense against bacteria is high acidity in the stomach, which is active against most organisms. This has led to speculation that low gastric acidity may allow overgrowth of bacteria in stomach contents. Several observational studies seem to bear this out. Newly diagnosed community-acquired pneumonia was associated with current use of proton pump inhibitors (odds ratio1.16, 95% confidence intervals (CI) 1.03–1.31), but not with use of H2RAs (odds ratio 0.98, CI 0.80–1.20).207 Acid-suppressive drug users had a higher incidence rate compared with non acid suppressive drug users (OR 1.73, 95% CI 1.33–2.25). The risk for community acquired pneumonia was also higher among PPI users compared with those who stopped use of a PPI (OR 1.89 (95% CI 1.36–2.62).208,209 PPIs and Skeletal Fractures? The use of PPIs has also been associated with an increase in skeletal fractures. Persons who used a PPI within the last year had an overall increase in risk of fracture (OR 5 1.18, 95% CI 1.12–1.43), an increased risk of hip fracture (OR 5 1.45, 95% CI 1.28–1.65), and an increased risk of spine fracture (OR 5 1.60, 95% CI 1.25–2.04).210 In this same population, the use of an H2RA was associated with a lower risk of fracture. A higher risk of osteoporotic fractures has also been observed among users of PPIs, which is time-dependent. Exposure of 7 or more years was associated with increased risk of all osteoporotic fractures (OR 1.92, 95% CI 1.16–3.18). An increased risk of hip fracture began after 5 or more years of exposure (OR 1.62, 95% CI 1.02–2.58), and increased after 7 or more years of exposure (OR 4.55, 95% CI 1.68–12.29).211 PPIs and Clostridium difficile Infection? Although antibiotic use is associated with risk of Clostridium difficile infection, over half of persons admitted to acute care settings have no antibiotic exposure within the past 45 days. However, the current use of a PPI was associated with a higher risk of infection (relative risk 1.6, 95% CI 1.3–2.0).212 In a hospital outbreak, only the use of a PPI or the use of a cephalosporin was a risk factor for Clostridium difficile infection. Other risk factors, including use of an H2RA, a fluroquinalone, an antipsychotic, or the presence of chronic obstructive pulmonary disease, were not associated with infection.213 A systematic review of twelve studies analyzing the use of PPIs and enteric infection confirmed an increased risk of Clostridium difficile infection (OR 1.94, 95% CI 1.37–2.75).214 The risk was also higher for Salmonella, Campylobacter, and other enteric infections in studies. In a long-term care setting, 60% of persons with a Clostridium difficile infection had used a PPI, compared to 32% of controls.215 622 Messinger-Rapport et al

PPIs and Drug Interactions? Because of the low to absent gastric acidity in persons taking a PPI, the absorption of drugs requiring an acid medium may be impaired. A large number of drugs may be either poorly absorbed (cefuroxime and other antibiotics) or more actively absorbed (digoxin). The frequency of drug interactions require that new drugs added to a person’s regimen be screened for compatibility with a PPI. Other nutrients may be affected by interaction with PPIs. There is some evidence that PPI therapy lowers intragastric concentrations of vitamin C, and may affect serum vitamin C levels.216 Absorption of vitamin B12 may potentially be affected, but the clinical evidence is weak. PPIs reduced the absorption of nonheme iron from a test meal by approximately 50%.217 Thus, in persons with iron deficiency anemia, replacement of iron may be limited by concurrent use of a PPI.218,219 In summary, the availability of PPIs have revolutionized the treatment of hyperacidity gastrointestinal diseases. However, prophylactic use and long durations of therapy have raised concerns about unintended consequences. The large number of potential drug interactions require diligence in careful prescribing. Duration of exposure to PPI should be limited to the shortest possible times consistent with therapeutic goals. For those patients who require long-term maintenance therapy, the use of H2RA has not been associated with the major long-term adverse effects of PPI use. DELIRIUM Delirium is a dangerous diagnosis for older adults in the hospital or long-term care setting. It is associated with a loss of functional ability, increased hospital length of stay, and higher likelihood of long-term care placement. Delirium is an independent risk factor for posthospital institutionalization and rehospitalization.220 Wilcox and colleagues found that delirium carries a 1-year institutionalization rate of 33.4% compared to a baseline rate of 10.7% (odds ratio 2.41; 95% CI 1.77–3.29).221 At 2 years posthospitalization, the mortality rate for individuals having had a diagnosis of delirium was 38% versus 28% in the nondelirious population (HR 1.95; 95% CI 1.51–2.52). Individuals with both dementia and delirium were at greatest risk for institutionalization, with 62.5% being placed in longterm care over 4 years, compared with 8.1% in a nondemented population (OR 12.52; 95% CI 1.86–84.21). In another study, those admitted to postacute rehabilitation with clinical instability or delirium were 15 times more likely to be readmitted to the hospital (30.1% versus 2%, p \ .001) and more than twice as likely to have poor functional recovery (86% versus 38%, p 5 .033).222 Poor functional recovery may be a result of impaired motor functioning in the delirious state. Italian researchers have demonstrated that individuals entering postacute rehabilitation with delirium or dementia have significantly more dysfunction with trunk control and with gait and balance compared with individuals without these diagnoses.223 Those with delirium often demonstrated JAMDA – November 2011

Table 3.

Table 4.

Diagnostic Features of Delirium

Risk Factors for Delirium

1. Inattention AND 2. Disorganized thinking PLUS At least 2 of the following:  Reduced level of consciousness  Perceptual disturbances  Disturbance of sleep-wake cycle  Increased or decreased psychomotor activity  Disorientation  Memory impairment  Abrupt onset and fluctuating course

D E L

a significant decline in motor function before clinical improvement was made. Delirium is a common diagnosis, yet underrecognized by health care providers. Studies consistently demonstrate that delirium is present in up to one third of individuals at the time of hospital admission or during hospitalization.224–227 Delirium is present in 50–75% of postoperative elders. Despite the prevalence of the condition, delirium is missed in one-third to two-thirds of hospitalized patients. A Veterans Affairs committee has suggested that a method for systematically and objectively measuring mental status is needed, and that ‘‘Mental Status’’ should represent the sixth vital sign.228

This cohort study found that those with the allele were more likely to develop delirium (OR 1.7; 95% CI 1.1–2.6) after correcting for age, cognitive status or functional abilities.232 A meta-analysis combining 4 large studies (n51100) of this genetic factor did not find a similar positive relationship.232 Individuals who were homozygous for a known dopamine transporter gene, were found to have a lower likelihood of developing delirium (OR 0.4; 95% CI 0.2–0.6, p\ .001). A second gene for the dopamine receptor provided no protective or beneficial effect for delirium.233

I R I U M S

Diagnostic Criteria for Delirium Delirium has a clear definition based on the Diagnostic and Statistical Manual of Mental Disorders version IV (DSM-IV; see Table 3).229 The DSM-IV diagnostic criteria have been utilized in the development of several delirium screening tools for clinicians. The most widely recognized of these tools is the Confusion Assessment Method (CAM).230 In order to meet screening criteria, the CAM requires the presence of (1) acute change in mental status with fluctuating course, plus (2) inattention and the presence of either, (3) disorganized thinking, or (4) altered level of consciousness. This screening tool has a sensitivity of .94% and specificity of .90%.230 Delirium Risk Factors The risk factors for delirium are many, and include underlying or new medical conditions, medications, and metabolic abnormalities. A mnemonic can assist in recalling potential risk factors and high risk medications (Table 4, and Table 5).231 In addition to these factors, there is growing interest in genetic risks that may predispose individuals to the development of delirium. In 1 study, researchers looked at the presence of the apolipoprotein E-4 allele as a risk for delirium.

Delirium Screening Tools In order to better identify and categorize delirium, several new screening tools have been developed. The Nursing Home CAM (NH-CAM) uses data from the Minimum Data Set (MDS) to retrospectively identify delirium based on presence of diagnostic features recorded through this database. The CAM criteria were identified using the MDS (version 2.0) sections B (question 5a–e) and E (questions 3 and 5). Using this method, a Swiss study of nearly 12,000 nursing home residents identified delirium in 6.5% of the population and subsyndromal delirium (SSD) in 39.7%.234 SSD is defined as meeting 2, but not all, of the delirium criteria and may represent a precursor state to the development of full criterion-based delirium. In another long-term care study, nearly half of residents with dementia with change in mental status were found to meet criteria for SSD.235 The NH-CAM has also been used to measure delirium outcomes of 90-day mortality and rehospitalization. Using a Medicare sample of nearly 38,000 nursing home admissions, patients with delirium had a 57% survival rate at 90 days compared with 83% without delirium. Rehospitalization occurred in 69% without and 62% with delirium. The presence of even 1 or 2 criteria for delirium increased the 90-day risk of death or rehospitalization.236 In a cohort of 457 delirious nursing home patients, mortality was highest in the hypoactive group compared with normal patients (HR 1.6; 94% CI 1.09–2.34)

Table 5.

Drug Categories That Can Cause Acute Change in Mental Status (MS)

A C U T E

anti-Parkinson corticosteroids urinary incontinence theophylline emptying drugs

UPDATES FROM THE AMDA MEETING

C H A N G E

Drugs Eyes-Ears (vision or hearing impairment) Low oxygen states (acute coronary syndrome, stroke, pulmonary embolism) Infection Retention (urine or stool) Ictal state Underhydration / Undernutrition Metabolic causes Subdural hematoma

cardiovascular H2 blockers antibiotics narcotics gero-psychiatry ear-nose-throat

I N

insomnia NSAIDS

M S

muscle relaxants seizure

Messinger-Rapport et al 623

after controlling for age, gender, comorbidity, dementia, and delirium severity.237

Delirium Prevention and Treatment Prevention of delirium is classically based on reduction of risk factors. Recent studies have looked at the use of medications to prevent or treat delirium. One hypothesis is that delirium may result from cholinergic neurotransmission imbalance and therefore, treatment with cholinesterase inhibitors may be helpful in delirium. Four placebo-controlled studies have recently investigated the use of cholinesterase inhibitors (3 rivastigmine, 1 donepezil) in hospitalized elders to prevent or treat delirium.238–241 Two of these studies focused on prevention and 2 focused on treatment.238,241 Outcomes included incidence of delirium, delirium duration, and hospital length of stay. No study showed any benefit in the treated group compared with placebo. One study was halted early as the treatment group showed a higher mortality rate (22%) compared with the placebo group (5%) and delirium duration was longer in the treated group.240 There are also several recent studies exploring the use of antipsychotics to treat or prevent delirium. These drugs must be used with caution given the associated increased risk of death with antipsychotics (OR 1.5, p 5 .02).242 A small study (n 5 42) of quetiapine versus placebo for treatment of delirium showed a more rapid recovery for the treatment group.243 A larger (n 5 400) prevention trial in postoperative elders showed a reduction in incident delirium with olanzapine, but also a concerning increase in severity of delirium in the treated group.244 A meta-analysis found that for the treatment group, haloperidol and atypical antipsychotics were beneficial in reducing the intensity and duration of delirium symptoms.245 Given this conflicting data and awareness of the risks associated with antipsychotics, routine use of antipsychotics for delirium would not be advised. Until further data clarifies the benefit, use of antipsychotics for delirium treatment should be limited to the most severe cases after exhausting nonpharmacological options. Nonpharmacological strategies and risk factor modification remain the standard of care for delirium treatment and prevention. Multifactorial hospital-based interventions have demonstrated benefit in reducing incidence and severity of delirium. In a study of 199 postoperative hospitalized patients age . 70, a program of medical management, rehabilitation and comprehensive geriatric assessment reduced incident delirium from 75–55% (p 5 .003). The duration of delirium was also reduced (5.0 versus 10.2 day; p 5 .009) and length of hospitalization declined from 28 versus 38 days (p 5 .028).246 In summary, for screening and managing delirium in hospitalized and institutionalized older adults, the following points should be kept in mind:  Delirium carries a high risk of functional decline, rehospitalization, institutionalization, and mortality  Delirium is frequently underrecognized by health care professionals 624 Messinger-Rapport et al

 New screening tools may assist in classifying disease severity and disease course  Subsyndromal delirium may precede the full diagnosis of delirium  Cholinesterase inhibitors are not effective in preventing or treating delirium  Evidence is mixed on the benefit of antipsychotics for prevention and treatment of delirium

POLYPHARMACY Polypharmacy is more than just ‘‘too many’’ medications. Although traditionally defined as the use of multiple medications or more medications than are clinically indicated, polypharmacy also includes the use of: Inappropriate classes/ agents, inappropriate dosing, failure to stop treatment, failure to start treatment, and medication with high risk for drug– drug/drug–disease interactions. Polypharmacy can lead to adverse drug reactions or events, increased cost, reduced patient compliance, and a higher likelihood of administration errors. Adverse Drug Events Adverse drug events (ADEs) are defined as any harm that results from the use of a medication (Figure 1). These include adverse drug reactions (harmful effects from the drug at normal doses and during normal use) and preventable events such as overdoses, inappropriate prescribing and medication errors. Fortunately, most medication errors and inappropriate prescribing do not result in an adverse drug event. Some do not result in any appreciable harm and others are caught as a ‘‘near miss’’ before harm occurs. The evidence is clear that, in older adult populations, as the number of medications increases, the risk for adverse drug events increases. Falls are a classic example of such a relationship. Chen and colleagues studied the rate of falls and fall-related events in elders using 7 or more medications. Compared to those using fewer medications, those taking

Fig. 1. Unwanted medication effects. JAMDA – November 2011

more than 7 medications had 1.3 times the risk of sustaining a fall-related fracture (HR 1.3; 1.04–1.62, p 5 .02).247 Exactly how many is too many medications? Studies clearly demonstrate that the more medications taken, the higher the risk of ADEs. A study of 335 nursing home residents found that the rate of ADEs was 1.2 per person/year for those taking 9 or more medications, compared with 0.53 per person/year in those taking fewer than 9 medications.248 Research in a Veterans Affairs population also showed that 9 medications was a cutoff for adverse events. Compared to those receiving regular care, a group of men receiving a targeted medication reduction program had a decrease in number of medications from 13.1 to 8.2 and a 30% reduction in hospitalizations (p \ .0002).249 As the number of medications increases, compliance with appropriate use declines. A study of patients who were randomized to a tight pharmacological disease-based control had an increase of 1.1 medications per person (compared with 0.6 medications in the control group) but a significant decline in compliance with suggested medication usage (OR 0.84; 95% CI 0.78–0.91, p \ .001).250 In the past 20 years, much work had been done to define and identify inappropriate medication use in older adults. In 1991, Mark Beers and 12 experts used the Delphi method to create a list containing 19 medications/classes which were potentially inappropriate for use in older adults in the nursing home setting. The ‘‘Beers List’’ as it is called was a stepping stone from which other groups created and modified ‘‘inappropriate’’ medication lists for older adults in a variety of clinical settings.251 The Beers List The Beers List underwent its first revision in 1997 to include 28 different medications or classes of medications considered to be high risk for use in older adults. A third iteration, including 48 medications/classes, was introduced in 2003. In this version, medications were classified as inappropriate ‘‘independent of diagnosis or condition’’ and potentially inappropriate when ‘‘considering diagnosis or condition’’ that could result in adverse drug-disease events.252,253 Clinicians and researchers have used various versions of the Beers List, applied to different care settings, to measure outcomes and trends in medication prescribing. Stuart et al measured the impact of the Beers List in community dwelling elders between 1995 and 1999. 7600 medication profiles were reviewed. A statistically significant drop in use of Beers List medications was seen over time (25% versus 21%, p \ .05).254A larger database analysis of ambulatory elders found similar results with 21% having filled a high risk Beers medication in the preceding year.255 A later study in 2003 of assisted living residents found that 16% were taking at least 1 medication on the Beers List and nearly all subjects were on 5 or more medications.256 Due to concerns about the generalizability of the Beers List outside of long-term care, Zhan and colleagues convened a panel of experts with the purpose of identifying drugs that should be avoided in the community-dwelling elderly population and which could be used to monitor medication usage in UPDATES FROM THE AMDA MEETING

Table 6.

Medication Appropriateness Index

Criterion

Weighted Score

Are there significant drug-drug interactions? Are there significant drug-disease interactions? Is there an indication for the drug? Is the drug effective for the indication? Is there unnecessary duplication with other drugs? Is the duration of therapy acceptable Is the dosage correct? Are the directions correct? Are the directions practical? Is the drug the least expensive alternative?

2 2 3 3 1 1 2 2 1 1

U.S. Medicare beneficiaries. This group identified 33 medications which were classified as always inappropriate, rarely appropriate, or having some indications. Data collected from a 1996 survey using Zhan criteria found 21% of communitydwelling elders were using 1 of 33 potentially risky medications.257 Despite the wide popularity of the Beers List, expert panel guidelines are known to possess significant methodological limitations. There have been concerns about nursing home surveyors using this list as a regulatory tool in the long-term care setting, a purpose for which it was not designed. AMDA thus developed a position statement regarding the use of the Beers List criteria when applied to the long-term care setting.258 This statement indicates that, ‘‘The Beers list is a helpful general guide regarding potentially inappropriate medication use of medications for older adults, but it must be use in conjunction with a patient-centered care process.Ultimately, decisions about medication prescribing must be clinically based and consider the patient’s total clinical picture, including the entire medication regimen, history of medication use, comorbidities, functional status, and prognosis. Checklist approaches should not substitute for the necessary steps in the care process for appropirate prescribing.’’

New Measures of Polypharmacy In the past 10 years, several new methods have been developed for measuring and classifying inappropriate medication. These methods are frequently compared with the Beers List, often by measuring the ability to predict the occurrence of adverse drug events. The Medication Appropriateness Index (MAI) is a 10question weighted scale that assigns an individual patient risk score based on a medication profile (Table 6). Using the MAI and the Beers Criteria as risk measures, an elderly Veteran cohort was assessed for ADEs. 14% of 238 patients had an ADE during the study period. 49% were taking a Beers List medication and 99% met at least 1 MAI criterion. The authors concluded that the MAI was a better scale for predicting the potential risk of ADEs than was the Beers List.259 Messinger-Rapport et al 625

In the United Kingdom, the START (Screening Tool to Alert doctors to Right Treatments) and STOPP (Screening Tool of Older People’s potentially inappropriate Prescriptions) tools have been developed to guide clinicians in medication management of older adults. These tools are quite extensive and include over 75 organ system-based medication recommendations.260 A study of over 1000 Irish elders did not find that the START or STOPP tools identified potential medication problems any better than did the Beers List.260 Reducing Polypharmacy Reducing polypharmacy requires not only an awareness of potentially inappropriate medications (by whatever criterion used) but a process by which a patient’s medication list can be reviewed, discussed, and modified as appropriate. This is best accomplished in a collaborative partnership that may include the patient, pharmacist, physician, family, and nurse. Education is crucial as many members of this care team may be unaware of the potential risks of certain medications in older adults. Education and team based approaches can reduce the use of high risk medication. When a consultant pharmacist advised physicians of high-risk prescribing patterns, the use of inappropriate medication dropped.257,261,262 A team-based geriatric inpatient unit that focused on medication management was able to reduce the number of medications and the use of high-risk medications.263 An electronic medical record

626 Messinger-Rapport et al

which flags high risk medication and provides warnings for drug–drug interactions has significantly reduced inappropriate prescribing in older adults.264–266 Engaging the patient in discussions of medication risk and in appropriate use can have profound potential benefits. When the physician and patient partnered in completing a Medication Risk Assessment Form and a Medication Review Checklist in the ambulatory setting, 88% of patient medication profiles were found to have at least 1 medication problem. The clinician acted to modify the medication profile in 88% of these cases.267 In summary, polypharmacy remains a significant finding in older adults throughout the healthcare system. Despite over 2 decades of using the Beers List, over 20% of elders continue to use medications which may be potentially inappropriate. New screening tools for identifying high risk medications have been recently developed. Tools such as the MAI, STOPP, START, and Zahn Criteria may be better suited in some cases to identify potential medication problems and risk of adverse drug events. Despite the criterion posed in these screening tools, appropriate prescribing must account for patient disease, needs and wishes. It is clear that educational and team-based interventions to reduce polypharmacy are effective in a variety of clinical settings. REFERENCES References available at www.jamda.com with your JAMDA login.

JAMDA – November 2011

REFERENCES 1. Morley JE. Osteoporosis and fragility fractures. J Am Med Dir Assoc 2011;12:389–392. 2. Miles RW. Hip fracture reduction in northwest Arkansas nursing homes. J Am Med Dir Assoc 2008;9:449–453. 3. Gloth FM 3rd, Simonson W. Osteoporosis is underdiagnosed in skilled nursing facilities: a large-scale heel BMD screening study. J Am Med Dir Assoc 2008;9:190–193. 4. Kamel HK. Update on Osteoporosis management in long-term care: Focus on bisphosphonates. J Am Med Dir Assoc 2007;8:434–440. 5. Heinrich S, Rapp K, Rissmann U, et al. Service use and costs of incident femoral fractures in nursing home residents in Germany: The Bavarian fall and fracture prevention project (BG(2)P(2)). J Am Med Dir Assoc 2011;12:459–466. 6. Young Y, Xiong K, Pruzek RM, Brant LJ. Examining heterogeneity of functional recovery among older adults with hip fractures. J Am Med Dir Assoc 2010;11:132–139. 7. Curtis JR, Arora T, Matthews RS, et al. Is withholding osteoporosis medication after fracture sometimes rational? A comparison of the risk for second fracture versus death. J Am Med Dir Assoc 2010;11: 584–591. 8. Jalbert JJ, Eaton CB, Miller SC, et al. Antipsychotic use and the risk of hip fracture among older adults afflicted with dementia. J Am Med Dir Assoc 2010;11:120–127. 9. Lapcevic WA, French DD, Campbell RR. All-cause mortality rates of hip fractures treated in the VHA: Do they differ from Medicare facilities? J Am Med Dir Assoc 2010;11:116–119. 10. Zimmerman S, Magaziner J, Birge SJ, et al. Adherence to hip protectors and implications for U.S. long-term care settings. J Am Med Dir Assoc 2010;11:106–115. 11. Nelson HD, Haney EM, Chou R, et al. Screening for Osteoporosis: Systematic Review to Update the 2002 U.S. Preventive Services Task Force Recommendation. Rockville, MD: Agency for Healthcare Research and Quality, U.S. Department of Health and Human Services; July 2010. Number 77. 12. Miller PD. Diagnosis and treatment of osteoporosis in chronic renal disease. Semin Nephrol 2009;29(2):144–155. 13. Black DM, Kelly MP, Genant HK, et al. Bisphosphonates and fractures of the subtrochanteric or diaphyseal femur. N Engl J Med 2010;362(19): 1761–1771. 14. Park-Wyllie LY, Mamdani MM, Juurlink DN, et al. Bisphosphonate use and the risk of subtrochanteric or femoral shaft fractures in older women. JAMA 2011;305(8):783–789. 15. Cummings SR, San Martin J, McClung MR, et al. Denosumab for prevention of fractures in postmenopausal women with osteoporosis. N Engl J Med 2009;361(8):756–765. 16. Smith MR, Egerdie B, Hernandez Toriz N, et al. Denosumab in men receiving androgen-deprivation therapy for prostate cancer. N Engl J Med 2009;361(8):745–755. 17. Islam T, Peiris P, Copeland RJ, et al. Vitamin D: Lessons from the veterans population. J Am Med Dir Assoc 2011;12:257–262. 18. Demontiero O, Herrmann M, Duque G. Supplementation with vitamin D and calcium in long-term care residents. J Am Med Dir Assoc 2011; 12:190–194. 19. Grant WB, Peiris AN. Possible role of serum 25-hydroxyvitamin D in black-white health disparities in the United States. J Am Med Dir Assoc 2010;11:617–628. 20. Gaddipati VC, Kuriacose R, Copeland R, et al. Vitamin D deficiency: an increasing concern in peripheral arterial disease. J Am Med Dir Assoc 2010;11:308–311. 21. Gau JT. Prevalence of vitamin D deficiency/insufficiency practice patterns in nursing homes. J Am Med Dir Assoc 2010;11:296. 22. Peiris AN, Bailery B, Manning T, et al. Testing for vitamin D deficiency in veterans–is there a seasonal bias? J Am Med Dir Assoc 2010;11: 128–131. 23. Braddy KK, Imam SN, Palla KR, et al. Vitamin D deficiency/insuffiency practice patterns in a veterans Health Administration long-term care UPDATES FROM THE AMDA MEETING

24. 25.

26.

27.

28.

29.

30.

31.

32. 33.

34.

35.

36. 37.

38.

39. 40.

41. 42.

43.

44.

45.

population: a retrospective analysis. J Am Med Dir Assoc 2009;10: 653–657. Drinka PJ, Krause PF, Nest LJ, et al. Determinants of vitamin D levels in nursing home residents. J Am Med Dir Assoc 2007;8:76–79. Hamid Z, Riggs A, Spencer T, et al. Vitamin D deficiency in residents of academic long-term care facilities despite having been prescribed vitamin D. J Am Med Dir Assoc 2007;8:71–75. Bischoff-Ferrari HA, Willett WC, Wong JB, et al. Prevention of nonvertebral fractures with oral vitamin D and dose dependency: A meta-analysis of randomized controlled trials. Arch Intern Med 2009; 169(6):551–561. Sanders KM, Stuart AL, Williamson EJ, et al. Annual high-dose oral vitamin D and falls and fractures in older women: a randomized controlled trial. JAMA 2010;303(18):1815–1822. Curhan GC, Willett WC, Speizer FE, et al. Comparison of dietary calcium with supplemental calcium and other nutrients as factors affecting the risk for kidney stones in women. Ann Intern Med 1997;126(7): 497–504. Giovannucci E, Liu Y, Stampfer MJ, Willett WC. A prospective study of calcium intake and incident and fatal prostate cancer. Cancer Epidemiol Biomark Prev 2006;15(2):203–210. Bolland MJ, Avenell A, Baron JA, et al. Effect of calcium supplements on risk of myocardial infarction and cardiovascular events: meta-analysis. BMJ 2010;341:c3691. Bolland MJ, Grey A, Avenell A, et al. Calcium supplements with or without vitamin D and risk of cardiovascular events: reanalysis of the Women’s Health Initiative limited access dataset and meta-analysis. BMJ 2011;342:d2040. Reid IR, Bolland MJ, Avenell A, et al. Cardiovascular effects of calcium supplementation. Osteoporo Int 2011;22(6):1649–1658. Anderson GL, Limacher M, Assaf AR, et al. Effects of conjugated equine estrogen in postmenopausal women with hysterectomy: the Women’s Health Initiative randomized controlled trial [see comment]. JAMA 2004;291(14):1701–1712. Rossouw JE, Anderson GL, Prentice RL, et al. Risk and benefits of estrogen plus progestin in healthy postmenopausal women: principal results from the Women’s Health Initiative Randomized Controlled Trial. JAMA 2002;288(3):321–333. Shumaker SA, Legault C, Thal L, et al. Estrogen plus progestin and the incidence of dementia and mild cognitive impairment in postmenopausal women: the Women’s Health Initiative Memory Study: a randomized controlled trial [comment]. JAMA 2003;289(20):2651–2662. Kanis JA, McCloskey EV, Johansson H, et al. Development and use of FRAX in osteoporosis. Osteoporo Int 2010;21(Suppl 2):S407–S413. Zhang X, Decker FH, Luo H, et al. Trends in the prevalence and comorbidities of diabetes in nursing home residents in the United States: 1995-2004. J Am Geriatr Soc 2010;58:724–730. Gerstel C, Zarate Laguines M, Vischer UM. Fecal incontinence resolved using metformin withdrawal. J Am Geriatr Soc 2011;59: 756–757. Mazza AD, Morley JE. Update on diabetes in the elderly and the application of current therapeutics. J Am Med Dir Assoc 2007;8:489–492. Lu FP, Lin KP, Kuo HK. Diabetes and the risk of multi-system phenotypes: a systematic review and meta-analysis. PLoS One 2009;4: e4144. Epub 2009 Jan 7. Meyers RM, Reger L. Diabetes management in long-term care facilities: a practical guide. J Am Med Dir Assoc 2009;10:589. Luchsinger JA, Palmas W, Teresi JA, et al. Improved diabetes control in the elderly delays global cognitive decline. J Nutr Health Aging 2011; 15:445–449. van der Maarel-Wierink CD, Vanobbergen JN, Bronkhorst EM, et al. Risk factors for aspiration pneumonia in frail older people: a systematic literature review. J Am Med Dir Assoc 2011;12:344–354. Chen LK, Peng LN, Lin MH, et al. Diabetes mellitus, glycemic control, and pneumonia in long-term care facilities: a 2-year, prospective cohort study. J Am Med Dir Assoc 2011;12:33–37. Schwartz AV, Vittinghoff E, Bauer DC, et al. Association of BMD and FRAX score with risk of fracture in older adults with type 2 diabetes. JAMA 2011;305:2184–2192. Messinger-Rapport et al

626.e1

46. Rosenthal MJ, Fajardo M, Gilmore S, et al. Hospitalization and mortality of diabetes in older adults. A 3-year prospective study. Diabetes Care 1998;21:231–235. 47. Miller DK, Lui LY, Perry HM 3rd, et al. Reported and measured physical functioning in older inner-city diabetic African Americans. J Gerontol A Biol Sci Med Sci 1999;54:M230–M236. 48. Morley JE. Diabetes and aging: epidemiologic overview. Clin Geriatr Med 2008;24:395–405. 49. Vischer UM. Glycemic targets for elderly diabetic patients need reappraisal. J Am Med Dir Assoc 2011;12:9–11. 50. Bourdel-Marchasson I, Doucet J, Bauduceau B, et al. Key priorities in managing glucose control in older people with diabetes. J Nutr Health Aging 2009;13:685–691. 51. Paolisso G, Sinclair AJ. Effective clinical practice for older people with type 2 diabetes. Glucose regulation is important but other strategies also need to be included. J Nutr Health Aging 2010;14:3–4. 52. Morley JE. Hypertension: Is it overtreated in the elderly? J Am Med Dir Assoc 2010;11:147–152. 53. Morley JE. Editorial: Postprandial hypotension—the ultimate Big Mac Attack. J Gerontol A Biol Sci Med Sic 2001;56:M741–M743. 54. Iwanczyk L, Weintraub NT, Rubenstein LZ. Orthostatic hypotension in the nursing home setting. J Am Med Dir Assoc 2006;7:163–167. 55. Gentilcore D, Vanis L, Wishart JM, et al. The alpha-glucosidase inhibitor, acarbose, attenuates the blood pressure and splanchnic blood flow responses to intraduodenal sucrose in older adults. J Gerontol A Biol Sci Med Sci 2011. May 31 [Epub ahead of print]. 56. Lee A. Preventive treatment with acarbose in diabetic reactive hypoglycemia. J Am Med Dir Assoc 2010;11:377–378. 57. Maxwell CS, Wood RJ. Update on vitamin D and type 2 diabetes. Nutr Rev 2011;69:291–295. 58. Morley JE. Vitamin D redux. J Am Med Dir Assoc 2009;10:591–592. 59. Jin HY, Joung SJ, Park JH, et al. The effect of alpha-lipoic acid on symptoms and skin blood flow in diabetic neuropathy. Diabet Med 2007;24: 1034–1038. 60. Foster TS. Efficacy and safety of alpha-lipoic acid supplementation in the treatment of symptomatic diabetic neuropathy. Diabetes Educ 2007;33:111–117. 61. Bouillet B, Vaillant G, Petit JM, et al. Are elderly patients with diabetes being overtreated in French long-term-care homes? Diabetes Metab 2010;36:272–277. 62. Mooradian AD, Osterweil D, Petrasek D, et al. Diabetes mellitus in elderly nursing home patients. A survey of clinical characteristics and management. J Am Geriatr Soc 1988;36:391–396. 63. Joseph J, Koka M, Aronow WS. Prevalence of a hemoglobin A1C less than 7.0%, of a blood pressure less than 130/80 mm Hg, and of a serum low-density lipoprotein cholesterol less than 100 mg/dL in older patients with diabetes mellitus in an academic nursing home. J Am Med Dir Assoc 2008;9:51–54. 64. Gersteain HC, Miller ME, Genuth S, et al., ACCORD Study Group. Long-term effects of intensive glucose lowering on cardiovascular outcomes. N Engl J Med 2011;364:818–828. 65. Patel A, MacMahon S, Chalmers J, et al., ADVANCE Collaborative Group. Intensive blood glucose control and vascular outcomes in patients with type 2 diabetes. N Engl J Med 2008;358:2560–2572. 66. Duckworth W, Abraira C, Moritz T, et al. Glucose control and vascular complications in veterans with type 2 diabetes. N Engl J Med 2009;360: 129–139. 67. Currie CJ, Peters JR, Rynan A, et al. Survival as a function of HbA(1c) in people with type 2 diabetes: a retrospective cohort study. Lancet 2010;375:481–489. 68. Nelson JM, Dufraux K, Cook PF. The relationship between glycemic control and falls in older adults. J Am Geriatr Soc 2007;55:2041–2044. 69. Wedick NM, Barrett-Connor E, Knoke JD, et al. The relationship between weight loss and all-cause mortality in older men and women with and without diabetes mellitus: The Rancho Bernardo study. J Am Geriatr Soc 2002;50:1810–1815. 70. Coulston AM, Mandelbaum D, Reaven GM. Dietary management of nursing home residents with non-insulin-dependent diabetes mellitus. Am J Clin Nutr 1990;51:67–71. 626.e2 Messinger-Rapport et al

71. Tariq SH, Karcic E, Thomas DR, et al. The use of a no-concentratedsweets diet in the management of type 2 diabetes in nursing homes. J Am Diet Assoc 2001;101:1463–1466. 72. Dorner B, Friedrich EK, Posthauer ME. Practice paper of the american dietetic association: Individualized nutrition approaches for older adults in health care communities. J Am Diet Assoc 2010;110(10): 1554–1563. 73. Umpierre D, Ribeiro PA, Kramer CK, et al. Physical activity advice only or structured exercise training and association with HbA1c levels in type 2 diabetes: a systematic review and meta-analysis. JAMA 2011; 305:1790–1799. 74. Morley JE. The elderly type 2 diabetic patient: special considerations. Diabet Med 1998;(15 Suppl 4):S41–S46. 75. Colberg SR, Somma CT, Sechrist SR. Physical activity participation may offset some of the negative impact of diabetes on cognitive function. J Am Med Dir Assoc 2008;9:434–438. 76. Aman E, Thomas DR. Supervised exercise reduces agitation in severely cognitively impaired persons. J Am Med Dir Assoc 2009;10:271–276. 77. Rolland Y, Abellan van Kan G, Vellas B. Physical activity and Alzheimer’s disease: from prevention to therapeutic perspectives. J Am Med Dir Assoc 2008;9:390–405. 78. Colberg SR, Zarrabi L, Bennington L, et al. Postprandial walking is better for lowering the glycemic effect of dinner than pre-dinner exercise in type 2 diabetic individuals. J Am Med Dir Assoc 2009;10:394–397. 79. Nylen ES, Kokkinos P, Myers J, et al. Prognostic effect of exercise capacity on mortality in older adults with diabetes mellitus. J Am Geriatr Soc 2010;58:1850–1854. 80. Morley JE, Mooradian AD, Rosenthal MJ, et al. Diabetes mellitus in elderly patients. Is it different? Am J Med 1987;83:533–544. 81. Holman R. Metformin as first choice in oral diabetes treatment: The UKPDS experience. J Annu Diabetol Hotel Dieu 2007;13–20. 82. Kumar VB, Bernardo AE, Vyas K, et al. Effect of metformin on nitric oxide synthase in genetically obese (ob/ob) mice. Life Sci 2001;69: 2789–3799. 83. Lee A, Morley JE. Metformin decreases food consumption and induces weight loss in subjects with obesity with type II non-insulin-dependent diabetes. Obes Res 1998;6:47–53. 84. Messinger-Rapport BJ, Thomas DR, et al. Clinical update on nursing home medicine: 2008. J Am Med Dir Assoc 2008;9:460–475. 85. Viccica G, Francucci CM, Marcocci C. The role of PPARy for the osteoblastic differentiation. J Endocrinol Invest 2010;33(7 Suppl):9–12. 86. Vestergaard P. Bone metabolism in type 2 diabetes and role of thiazolidinediones. Curr Opin Endocrinol Diabetes Obes 2009;16:125–131. 87. Kahn SE, Zinman B, Lachin JM, et al. Rosiglitazone-associated fractures in type 2 diabetes: an analysis from a Diabetes Outcome Progression Trial (ADOPT). Diabetes Care 2008;31:845–851. 88. Lee A, Patrick P, Wishart J, et al. The effects of miglitol on glucagonlike peptide-1 secretion and appetite sensations in obese type 2 diabetics. Diabetes Obes Metab 2002;4:329–335. 89. DeLeon MJ, Chandurkar V, Albert SG, et al. Glucagon-like peptide-1 response to acarbose in elderly type 2 diabetic subjects. Diabetes Res Clin Pract 2002;56:101–106. 90. Stafford S, Elahi D, Meneilly GS. Effect of the dipeptidyl peptidase-4 inhibitor sitagliptin in older adults with type 2 diabetes mellitus. J Am Geriatr Soc 2011;1148–1149. 91. Schejter YD, Turvall E, Ackerman Z. Characteristics of patients with sulphonurea-induced hypoglycemia. J Am Med Dir Assoc 2010; doi:10.1016/j.jamda.2010.07.014. 92. Pandya N, Nathanson E. Managing diabetes in long-term care facilities: benefits of switching from human insulin to insulin analogs. J Am Med Dir Assoc 2010;11:171–178. 93. Yki-Jarvinen H, Kauppinen-Makelin R, Tiikkainen M, et al. Insulin glargine or nph combined with metformin in type 2 diabetes: The LANMET study. Diabetologia 2006;49(3):442–451. 94. Fritsche A, Schweitzer MA, Haring HU. Glimepiride combined with morning insulin glargine, bedtime neutral protamine hagedorn insulin, or bedtime insulin glargine in patients with type 2 diabetes. A randomized, controlled trial. Ann Intern Med 2003;138(12):952–959. JAMDA – November 2011

95. Rosenstock J, Schwartz SL, Clark CM Jr, et al. Basal insulin therapy in type 2 diabetes: 28-week comparison of insulin glargine (HOE 901) and NPH insulin. Diabetes Care 2001;24(4):631–636. 96. Kumari J, Dharmarajan TS. Insulin glargine induced persistent intractable hypoglycemia, with variable presentations in older diabetic patients: an experience of 4 cases. J Am Med Dir Assoc 2009;10:672–673. 97. Morley JE. Polypharmacy in the nursing home. J Am Med Dir Assoc 2009;10:289–291. 98. Fitzgerald SP, Bean NG. An analysis of the interactions between individual comorbidities and their treatments—implications for guidelines and polypharmacy. J Am Med Dir Assoc 2010;11:475–484. 99. Gokce Kutsal Y, Barak A, Atalay A, et al. Polypharmacy in the elderly: a multicenter study. J Am Med Dir Assoc 2009;10:486–490. 100. Messinger-Rapport BJ, Morley JE, Thomas DR, et al. Clinical update on nursing home medicine: 2010. J Am Med Dir Assoc 2010;11:543–566. 101. Chisholm JW, Goldfine AB, Dhalia AK, et al. Effect of ranolazine on A1C and glucose levels in hyperglycemic patients with non-ST elevation acute coronary syndrome. Diabetes Care 2010;33:1163–1168. 102. Strojek K, Yoon K, Hruba V, et al. Effect of dapagliflozin in patients with type 2 diabetes who have inadequate glycaemic control with glimepiride: a randomized, 24-week, double-blind, placebo-controlled trial. Diabetes Obes Metab 2011. Jun 15 [Epu ahead of print]. 103. Feldman SM, Rosen R, DeStasio J. Status of diabetes management in the nursing home setting in 2008: a retrospective chart review and epidemiology study of diabetic nursing home residents and nursing home initiatives in diabetes management. J Am Med Dir Assoc 2009;10: 354–360. 104. Gerstein HC, Miller ME, Byington RP, et al. Effects of intensive glucose lowering in type 2 diabetes. N Engl J Med 2008;358(24):2545–2559. 105. Patel A, MacMahon S, Chalmers J, et al. Intensive blood glucose control and vascular outcomes in patients with type 2 diabetes. N Engl J Med 2008;358(24):2560–2572. 106. Reaven PD, Moritz TE, Schwenke DC, et al. Intensive glucose lowering therapy reduces cardiovascular disease events in VADT participants with lower calcified coronary atherosclerosis. Diabetes Aug 3 2009. 107. Monnier L, Colette C. Contributions of fasting and postprandial glucose to hemoglobin A1c. Endocr Pract 2006;12(Suppl 1):42–46. 108. Pandya N, Thompson S, Sambamoorthi U. The prevalence and persistence of sliding scale insulin use among newly admitted elderly nursing home residents with diabetes mellitus. J Am Med Dir Assoc 2008;9(9): 663–669. 109. Castellanos VH, Andrews YN. Inherent flaws in a method of estimating meal intake commonly used in long-term-care facilities. J Am Diet Assoc 2002;102(6):826–830. 110. Thomas DR, Ashmen W, Morley JE, et al. Nutritional management in long-term care: development of a clinical guideline. Council for Nutritional Strategies in Long-Term Care. Am J Clin Nutr 2002;75:308–313. 111. Morley JE. Anorexia and weight loss in older persons. J Gerontol A Biol Sci Med Sci 2003;58:131–137. 112. Kalantar-Zadeh K, Horwich TB, Oreopoulos A, et al. Risk factor paradox in wasting diseases. Curr Opin Clin Nutr Metab Care 2007;10: 433–442. 113. Karcic E, Philpot C, Morley JE. Treating malnutrition with megestrol acetate: literature review and review of our experience. J Nutr Health Aging 2002;6:191–200. 114. Payette H. Nutrition as a determinant of functional autonomy and quality of life in aging: a research program. Can J Physiol Pharmacol 2005; 83:1061–1070. 115. Ensrud KE, Ewing SK, Stone KL, et al. Intentional and unintentional weight loss increase bone loss and hip fracture risk in older women. J Am Geriatr Soc 2003;51:1740–1747. 116. Morley JE. Undernutrition: a major problem in nursing homes. J Am Med Dir Assoc 2011;12:243–246. 117. Tolson D, Rolland Y, Andrieu S, et al. International Association of Gerontology and Geriatrics: A global agenda for clinical research and quality of care in nursing homes. J Am Med Dir Assoc 2011;12: 184–189. 118. Morley JE. Anorexia, weight loss, and frailty. J Am Med Dir Assoc 2010; 11:225–228. UPDATES FROM THE AMDA MEETING

119. Murphy C. The chemical senses and nutrition in older adults. J Nutr Elder 2008;27:247–265. 120. Jones KL, Doran SM, Hveem K, et al. Relation between postprandial satiation and antral area in normal subjects. Am J Clin Nutr 1997;66: 127–132. 121. Sturm K, Parker B, Wishart J, et al. Energy intake and appetite are related to antral area in healthy young and older subjects. Am J Clin Nutr 2004;80:656–667. 122. MacIntosh CG, Morley JE, Wishart J, et al. Effect of exogenous cholecystokinin (CCK)-8 on food intake and plasma CCK, leptin, and insulin concetrations in older and young adults: evidence for increased CCK activity as a cause of the anorexia of aging. J Clin Endocrinol Metab 2001;86:5830–5837. 123. Sih R, Morley JE, Kaiser FE, et al. Testosterone replacement in older hypogonadal men: a 12-month randomized controlled trial. J Clin Endocrinol Metab 1997;82:1661–1667. 124. Gosnell BA, Levine AS, Morley JE. The effects of aging on opioid modulation of feeding in rats. Life Sci 1983;32:2793–2799. 125. Morley JE, Levine AS. The pharmacology of eating behavior. Annu Rev Pharmacol Toxicol 1985;25:127–146. 126. Cornali C, Franzoni S, Frisoni GB, et al. Anorexia as an independent predictor of mortality. J Am Geriatr Soc 2005;53:354–355. 127. Morley JE. Weight loss in older persons: new therapeutic approaches. Curr Pharm Des 2007;13:3637–3647. 128. Wilson MM, Thomas DR, Rubenstein LZ, et al. Appetite assessment: simple appetite questionnaire predicts weight loss in communitydwelling adults and nursing home residents. Am J Clin Nutr 2005;82: 1074–1081. 129. Morley JE. Assessment of malnutrition in older persons: A focus on the Mini Nutritional Assessment. J Nutr Health Aging 2011;15:87–90. 130. Vellas B, Villars H, Abellan G, et al. Overview of the MNA—Its history and challenges. J Nutr Health Aging 2006;10:456–463. 131. Charlton KE, Nichols C, Bowden S, et al. Older rehabilitation patients are at high risk of malnutrition: evidence from a large Australian database. J Nutr Health Aging 2010;14:622–628. 132. Kaiser MJ, Bauer JM, Ramsch C, et al. Validation of the Mini Nutritional Assessment short-form (MNA-SF): A practical tool for identification of nutritional status. J Nutr Health Aging 2009;13:782–788. 133. Abellan van Kan G, Rolland Y, Bergman H, et al. The I.A.N.A. Task Force on frailty assessment of older people in clinical practice. J Nutr Health Aging 2008;12:29–37. 134. Abellan van Kan G, Rolland YM, Morley JE, et al. Frailty: Toward a clinical definition. J Am Med Dir Assoc 2008;9:71–72. 135. Omran ML, Morley JE. Assessment of protein energy malnutrition in older persons, Part II: Laboratory evaluation. Nutrition 2000;16: 131–140. 136. Morley JE, Kraenzle D. Causes of weight loss in a community nursing home. J Am Geriatr Soc 1988;36:487–491. 137. Morley JE. Depression in nursing home residents. J Am Med Dir Assoc 2010;11:301–303. 138. Cabrera MA, Mesas AE, Garcia AR, et al. Malnutrition and depression among community-dwelling elderly people. J Am Med Dir Assoc 2007; 8:582–584. 139. Argiles JM, Anker SD, Evans WJ, et al. Consensus on cachexia definitions. J Am Med Dir Assoc 2010;11:229–230. 140. Evans WJ, Morley JE, Argiles J, et al. Cachexia: A new definition. Clin Nutr 2008;27:793–799. 141. Morley JE, Thomas DR, Wilson MM. Cachexia: pathophysiology and clinical relevance. Am J Clin Nutr 2006;83:735–743. 142. Morley JE, Thomas DR. Cachexia: new advances in the management of wasting diseases. J Am Med Dir Assoc 2008;9:205–210. 143. von Haehling S, Anker SD. Cachexia as a major underestimated and unmet medical need: facts and numbers. J Cachex Sarcopenia Muscle 2010;1:1–5. 144. von Haehling S, Anker SD. Cardio-renal anemia syndrome. Contrib Nephrol 2011;171:266–273. 145. Yeh SS, Blackwood K, Schuster MW. The cytokine basis of cachexia and its treatment: are they ready for prime time? J Am Med Dir Assoc 2008;9:219–236. Messinger-Rapport et al

626.e3

146. von Haehling S, Morley JE, Anker SD. An overview of sarcopenia: facts and numbers on prevalence and clinical impact. J Cachex Sarcopenia Muscle 2010;1:129–133. 147. Morley JE, Baumgartner RN, Roubenoff R, et al. Sarcopenia. J Lab Clin Med 2001;137:231–243. 148. Morley JE, Abbatecola AM, Argiles JM, et al. Sarcopenia with limited mobility: An international consensus. J Am Med Dir Assoc 2011;12: 403–409. 149. Fielding RA, Vellas B, Evans WJ, et al. Sarcopenia: an undiagnosed condition in older adults. Current consensus definition: prevalence, etiology, and consequences. International Working Group on Sarcopenia. J Am Med Dir Assoc 2011;12:249–256. 150. Cruz-Jentoft AJ, Baevens JP, Bauer JM, et al. Sarcopenia: European consensus on definition and diagnosis: Report of the European Working Group on Sarcopenia in Older People. Age Ageing 2010;39:412–423. 151. Baumgartner RN, Wayne SJ, Waters DL, et al. Sarcopenic obesity predicts instrumental activities of daily living disability in the elderly. Obes Res 2004;12:1995–2004. 152. Rolland Y, Lauwers-Cances V, Cristini C, et al. Difficulties with physical function associated with obesity, sarcopenia, and sarcopenic-obesity in community-dwelling elderly women: the EPIDOS (EPIDemiologie de l’OSteoporose) Study. Am J Clin Nutr 2009;89:1895–1900. 153. Thomas DR, Cote TR, Lawhorne L, et al. Understanding clinical dehydration and its treatment. J Am Med Dir Assoc 2008;9:292–301. 154. Messinger-Rapport BJ, Thomas DR, Gammack JK, et al. Clinical update on nursing home medicine: 2009. J Am Med Dir Assoc 2009;10: 530–553. 155. Thomas DR, Ashmen W, Morley JE, et al. Nutritional management in long-term care: development of a clinical guideline. Council for Nutritional Strategies in Long-Term Care. J Gerontol A Biol Sci Med Sci 2000;55:M725–M734. 156. Nijs K, de Graaf C, van Staveren WA, et al. Malnutrition and mealtime ambiance in nursing homes. J Am Med Dir Assoc 2009;10:226–229. 157. Morley JE. Weight loss in the nursing home. J Am Med Dir Assoc 2007; 8:201–204. 158. Milne AC, Potter J, Vivanti A, et al. Protein and energy supplementation in elderly people at risk from malnutrition. Cochrane Database Syst Rev 2009;(2):CD003288. 159. Wilson MM, Punushothaman R, Morley JE. Effect of liquid dietary supplements on energy intake in the elderly. Am J Clin Nutr 2001;75: 944–947. 160. van Wetering CR, Hoogendoorn M, Broekhuizen R, et al. Efficacy and costs of nutritional rehabilitation in muscle-wasted patients with chronic obstructive pulmonary disease in a community-based setting: a prespecified subgroup analysis of the INTERCOM trial. J Am Med Dir Assoc 2010;11:179–187. 161. Neelemaat F, Bosmans JE, Thijs A, et al. Post-discharge nutritional support in malnourished elderly individuals improves functional limitations. J Am Med Dir Assoc 2011;12:295–301. 162. Morley JE, Argiles JM, Evans WJ, et al. Nutritional recommendations for the management of sarcopenia. J Am Med Dir Assoc 2010;11: 391–396. 163. Chapman IM, MacIntosh CG, Morley JE, et al. The anorexia of aging. Biogerontology 2002;3:67–71. 164. Yeh SS, Lovitt S, Schuster MW. Usage of megestrol acetate in the treatment of anorexia-cachexia syndrome in the elderly. J Nutr Health Aging 2009;13:448–454. 165. Yeh S, Wu SY, Levine DM, et al. Quality of life and stimulation of weight gain after treatment with megestrol acetate: correlation between cytokine levels and nutritional status, appetite in geriatric patients with wasting syndrome. J Nutr Health Aging 2000;4:246–251. 166. Wilson MM, Philpot C, Morley JE. Anorexia of aging in long-term care: is dronabinol an effective appetite stimulant?—a pilot study. J Nutr Health Aging 2007;11:195–198. 167. Jatoi A, Windschitl HE, Loprinzi CL, et al. Dronabinol versus megestrol acetate versus combination therapy for cancer-associated anorexia: A North Central Cancer Treatment Group study. J Clin Oncol 2002; 20:567–573. 626.e4 Messinger-Rapport et al

168. Wang C, Nieschlag E, Swerdloff R, et al. Investigation, treatment and monitoring of late-onset hypogonadism in males: ISA, ISSAM EAU, EAA and ASA recommendations. Eur J Endocrinol 2008;159:507–514. 169. Morley JE. Anabolic steroids and frailty. J Am Med Dir Assoc 2010;11: 533–536. 170. Chahla EJ, Hayek ME, Morley JE. Testosterone replacement therapy and cardiovascular risk factors modification. Aging Male 2011;14: 83–90. 171. Basaria S, Coviello AD, Travison TG, et al. Adverse events associated with testosterone administration. N Engl J Med 2010;363:109–122. 172. Chapman IM, Visvanathan R, Hammond AJ, et al. Effect of testosterone and a nutritional supplement, alone and in combination, on hospital admissions in undernourished older men and women. Am J Clin Nutr 2009;89:880–889. 173. Srinivas-Shankar U, Roberts SA, et al. Effects of testosterone on muscle strength, physical function, body composition, and quality of life in intermediate-frail and frail elderly men: a randomized, double-blind, placebo-controlled study. J Clin Endocrinol Metab 2010;95:639–650. 174. Bhasin S, Jasuja R. Selective androgen receptor modulators as function promoting therapies. Curr Opin Clin Nutr Metab Care 2009;12: 232–240. 175. Cheung WW, Mak RH. Ghrelin and its analogues as therapeutic agents for anorexia and cachexia in end-stage renal disease. Kidney Int 2009; 76:135–137. 176. Chasen M, Hirschman SZ, Bhargava R. Phase II study of the novel peptide—nucleic acid OHR118 in the management of cancer-related anorexia/cachexia. J Am Med Dir Assoc 2011;12:62–67. 177. Kuo S, Rhodes RL, Mitchell SL, et al. Natural history of feeding-tube use in nursing home residents with advanced dementia. J Am Med Dir Assoc 2009;10:264–270. 178. Gillick MR, Volandes AE. The standard of caring: why do we still use feeding tubes in patients with advanced dementia? J Am Med Dir Assoc 2008;9:364–367. 179. Beier MT. Warfarin monitoring woes: a glimmer of hope with pharmacogenomics. J Am Med Dir Assoc 2008;9:199–200. 180. Dharmarajan L, Sajjad W. Potentially lethal acetaminophen-warfarin interaction in an older adult: an under-recognized phenomenon? J Am Med Dir Assoc 2007;8:545–547. 181. Gurwitz JH. Pharmacogenomics: Is this the final chapter in the remarkable story of warfarin? J Am Med Dir Assoc 2011. May 13 [Epub ahead of print]. 182. Dharmarajan TS, Gupta A, Baig MA, et al. Warfarin: Implementing its safe use in hospitalized patients from nursing homes and community through a performance improvement initiative. J Am Med Dir Assoc 2011;12:613–614. 183. Aguilar MI, et al. Cochrane Database Syst Rev 2007;(3):CD00618. 184. Saxena R, Koudstaal PJ. Anticoagulants versus antiplatelet therapy for preventing stroke in patients with nonrheumatic atrial fibrillation and a history of stroke or transient ischemic attack. Cochrane Database Syst Rev 2006;(1):CD000187. 185. Gage BF, Waterman AD, Shannon W, et al. Validation of clinical classification schemes for predicting stroke: results from the National Registry of Atrial Fibrillation. JAMA 2001;285:2864–2870. 186. Connolly SJ, Ezekowitz MD, Yusuf S, et al. RE-LY Steering Committee and Investigators. Dabigatran versus warfarin in patients with atrial fibrillation. N Engl J Med 2009;361(12):1139–1151. 187. Man-Son-Hing M, et al. Choosing antithrombotic therapy for elderly patients with atrial fibrillation who are at risk for falls. Arch Intern Med 1999;159:677–685. 188. Massie BM, Krol WF, Ammon SE, et al. The Warfarin and Antiplatelet Therapy in Heart Failure trial (WATCH): rationale, design, and baseline patient characteristics. J Card Fail 2004;10(2):101–112. 189. Cleland JG, Findlay I, Jafri S, et al. The Warfarin/Aspirin Study in Heart failure (WASH): a randomized trial comparing antithrombotic strategies for patients with heart failure. Am Heart J 2004;148(1): 157–164. 190. Bhatt DL, Fox KA, Hacke W, et al., CHARISMA Investigators. Clopidogrel and aspirin versus aspirin alone for the prevention of atherothrombotic events. N Engl J Med 2006;354:1706–1717. JAMDA – November 2011

191. Hankey GJ, Hacke W, Easton JD, et al., CHARISMA Trial Investigators. Effect of clopidogrel on the rate and functional severity of stroke among high vascular risk patients: a prespecified substudy of the Clopidogrel for High Atherothrombotic Risk and Ischemic Stabilization, Management and Avoidance (CHARISMA) trial. Stroke 2010; 41(8):1679–1683. 192. Squizzato A, et al. Clopidogrel plus aspirin versus aspirin alone for preventing cardiovascular disease. Cochrane Database Syst Rev 2007;(3): CD005158. 193. Grines CL, Bonow RO, Casey DE Jr, et al., American Heart Association, American College of Cardiology, Society for Cardiovascular Angiography and Interventions, American College of Surgeons, American Dental Association, American College of Physicians. Prevention of premature discontinuation of dual antiplatelet therapy in patients with coronary artery stents. Circulation 2007;115:813–818. 194. Mehta SR, Yusuf S, Peters RJ, et al. Clopidogrel in Unstable angina to prevent Recurrent Events trial (CURE) Investigators. Effects of pretreatment with clopidogrel and aspirin followed by long-term therapy in patients undergoing percutaneous coronary intervention: the PCICURE study. Lancet 2001;358(9281):527–533. 195. Price MJ, Berger PB, Teirstein PS, et al., GRAVITAS Investigators. Standard- vs high-dose clopidogrel based on platelet function testing after percutaneous coronary intervention: the GRAVITAS randomized trial. JAMA 2011;305:1079. 196. Rostom A, Dube C, Wells GA, et al. Prevention of NSAID-induced gastroduodenal ulcers. Cochrane Database Syst Rev (11) 2010. 197. Cook DJ, Fuller HD, Guyatt GH, et al. Risk factors for gastrointestinal bleeding in critically ill patients. Canadian Critical Care Trials Group. N Engl J Med 1994;330(6):377–381. 198. Kantorova I, Svoboda P, Scheer P, et al. Stress ulcer prophylaxis in critically ill patients: a randomized controlled trial. Hepato-Gastroenterology 2004;51(57):757–761. 199. Yildizdas D, Yapicioglu H, Yilmaz HL. Occurrence of ventilatorassociated pneumonia in mechanically ventilated pediatric intensive care patients during stress ulcer prophylaxis with sucralfate, ranitidine, and omeprazole. J Crit Care 2002;17:240–245. 200. Lin PC, Chang CH, Hsu PI, et al. The efficacy and safety of proton pump inhibitors vs histamine-2 receptor antagonists for stress ulcer bleeding prophylaxis among critical care patients: A meta-analysis. Crit Care Med 2010;38:1197. 201. Barletta JF, Erstad BL, Fortune JB. Stress ulcer prophylaxis in trauma patients. Crit Care (London, England) 2002;6(6):526–530. 202. Lam NP, Le PD, Crawford SY, Patel S. National survey of stress ulcer prophylaxis. Crit Care Med 1999;27:98–103. 203. Spirt MJ, Stanley S. Update on stress ulcer prophylaxis in critically ill patients. Crit Care Nurse 2006;26:18–20, 22–28. 204. Martin RM, Lim AG, Kerry SM, et al. Trends in prescribing H2receptor antagonists and proton pump inhibitors in primary care. Aliment Pharmacol Ther 1998;12(8):797–805. 205. The Medical Letter on Drugs and Therapeutics, February 23, 2009 (Issue 1306) p. 13. 206. Bhatt DL, Cryer BL, Contant CF, et al., COGENT Investigators. Clopidogrel with or without omeprazole in coronary artery disease. N Engl J Med 2010;363(20):1909–1917. 207. Rodriguez LA, Ruigomez A, Wallander MA, et al. Acid-suppressive drugs and community-acquired pneumonia. Epidemiology 2009;20(6): 800–806. 208. Laheij RJ, Sturkenboom MC, Hassing RJ, et al. Risk of communityacquired pneumonia and use of gastric acid-suppressive drugs. JAMA 2004;292:1955–1960. 209. Gulmez SE, Holm A, Frederiksen H, et al. Use of proton pump inhibitors and the risk of community-acquired pneumonia: a populationbased case-control study. Arch Intern Med 2007;167:950–955. 210. Vestergaard P, Rejnmark L, Mosekilde L. Proton pump inhibitors, histamine H2 receptor antagonists, and other antacid medications and the risk of fracture. Calc Tiss Int 2006;79(2):76–83. 211. Targownik LE, Lix LM, Metge CJ, et al. Use of PPIs and risk of osteoporosis-related fracture. CMAJ 2008;179(4):319–326. UPDATES FROM THE AMDA MEETING

212. Dial S, Kezouh A, Dascal A, et al. Patterns of antibiotic use and risk of hospital admission because of Clostridium difficile infection. CMAJ 2008;179(8):767–772. 213. Kazakova SV, Ware K, Baughman B, et al. A hospital outbreak of diarrhea due to an emerging epidemic strain of Clostridium difficile. Arch Intern Med 2006;166(22):2518–2524. 214. Leonard J, Marshall JK, Moayyedi P. Systematic review of the risk of enteric infection in patients taking acid suppression. Am J Gastroenterol 2007;102:2047–2205. 215. Al-Tureihi FI, Hassoun A, Wolf-Klein G, et al. Albumin, length of stay, and proton pump inhibitors: key factors in Clostridium difficileassociated disease in nursing home patients. J Am Med Dir Assoc 2005;6(2):105–108. 216. McColl EL. Am J Gastroenterol 2009;104:S5–S9. 217. Hutchison C. Proton pump inhibitors suppress absorption of dietary non-heme iron in hereditary haemochromatosis. Gut 2007;56: 1291–1295. 218. Sharma VR, Brannon MA, Carloss EA. Effect of omeprazole on oral iron replacement in patients with iron deficiency anemia. South Med J 2004;97:887–889. 219. Hoover JG, et al. Use of intravenous proton-pump inhibitors in a teaching hospital practice. Dig Dis Sci 2009;54:1947–1952. 220. Bellelli G, Magnifico F, Trabucchi M. Outcomes at 12 months in a population of elderly patients discharged from a rehabilitation unit. J Am Med Dir Assoc 2008;9(1):55–64. 221. Witlox J, Eurelings LS, de Jonghe JF, et al. Delirium in elderly patients and the risk of postdischarge mortality, institutionalization, and dementia: a meta-analysis. JAMA 2010;304(4):443–451. 222. Guerini F, Frisoni GB, Morghen S, et al. Clinical instability as a predictor of negative outcomes among elderly patients admitted to a rehabilitation ward. J Am Med Dir Assoc 2010;11(6):443–448. 223. Bellelli G, Speciale S, Morghen S, et al. Are fluctuations in motor performance a diagnostic sign of delirium? J Am Med Dir Assoc 2011; 12:578–583. 224. Francis J, Martin D, Kapoor WN. A prospective study of delirium in hospitalized elderly. JAMA 1990;263(8):1097–1101. 225. Inouye SK, Viscoli CM, Horwitz RI, et al. A predictive model for delirium in hospitalized elderly medical patients based on admission characteristics. Ann Intern Med 1993;119(6):474–481. 226. Inouye SK. Delirium in hospitalized older patients: recognition and risk factors. J Geriatr Psychiatry Neurol 1998;11(3):118–125. discussion 157–158. 227. Lewis LM, Miller DK, Morley JE, et al. Unrecognized delirium in ED geriatric patients. Am J Emerg Med 1995;13(2):142–145. 228. Flaherty JH, Shay K, Weir C, et al. The development of a mental status vital sign for use across the spectrum of care. J Am Med Dir Assoc 2009; 10(6):379–380. 229. American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders. Revised 4th ed. Washington, DC: Author; 2000. 230. Inouye SK, van Dyck CH, Alessi CA, et al. Clarifying confusion: the confusion assessment method. A new method for detection of delirium. Ann Intern Med 1990;113(12):941–948. 231. Flaherty JH. Psychotherapeutic agents in older adults. Commonly prescribed and over-the-counter remedies: causes of confusion. Clin Geriatr Med 1998;14(1):101–127. 232. van Munster BC, Korevaar JC, Zwinderman AH, et al. The association between delirium and the apolipoprotein E epsilon 4 allele: new study results and a meta-analysis. Am J Geriatr Psychiatry 2009;17(10): 856–862. 233. van Munster BC, de Rooij SE, Yazdanpanah M, et al. The association of the dopamine transporter gene and the dopamine receptor 2 gene with delirium, a meta-analysis. Am J Med Genet B Neuropsychiatr Genet 2010;153B(2):648–655. 234. von Gunten A, Mosimann UP. Delirium upon admission to Swiss nursing homes: a cross-sectional study. Swiss Med Wkly 2010;140(25-26): 376–381. 235. Voyer P, Richard S, Doucet L, et al. Detecting delirium and subsyndromal delirium using different diagnostic criteria among demented longterm care residents. J Am Med Dir Assoc 2009;10(3):181–188. Messinger-Rapport et al

626.e5

236. Dosa D, Intrator O, McNicoll L, et al. Preliminary derivation of a Nursing Home Confusion Assessment Method based on data from the Minimum Data Set. J Am Geriatr Soc 2007;55(7):1099–1105. 237. Kiely DK, Jones RN, Bergmann MA, et al. Association between psychomotor activity delirium subtypes and mortality among newly admitted post-acute facility patients. J Gerontol A Biol Sci Med Sci 2007; 62(2):174–179. 238. Gamberini M, Bolliger D, Lurati Buse GA, et al. Rivastigmine for the prevention of postoperative delirium in elderly patients undergoing elective cardiac surgery–a randomized controlled trial. Crit Care Med 2009;37(5):1762–1768. 239. Overshott R, Vernon M, Morris J, et al. Rivastigmine in the treatment of delirium in older people: a pilot study. Int Psychogeriatr 2010;22(5): 812–818. 240. van Eijk MM, Roes KC, Honing ML, et al. Effect of rivastigmine as an adjunct to usual care with haloperidol on duration of delirium and mortality in critically ill patients: a multicentre, double-blind, placebo-controlled randomised trial. Lancet 2010;376(9755):1829–1837. 241. Sampson EL, Raven PR, Ndhlovu PN, et al. A randomized, doubleblind, placebo-controlled trial of donepezil hydrochloride (Aricept) for reducing the incidence of postoperative delirium after elective total hip replacement. Int J Geriatr Psychiatry 2007;22(4):343–349. 242. Schneider LS, Dagerman KS, Insel P. Risk of death with atypical antipsychotic drug treatment for dementia: meta-analysis of randomized placebo-controlled trials. JAMA 2005;294(15):1934–1943. 243. Tahir TA, Eeles E, Karapareddy V, et al. A randomized controlled trial of quetiapine versus placebo in the treatment of delirium. Psychosom Res 2010;69(5):485–490. 244. Larsen KA, Kelly SE, Stern TA, et al. Administration of olanzapine to prevent postoperative delirium in elderly joint-replacement patients: a randomized, controlled trial. Psychosomatics 2010;51(5):409–418. 245. Lonergan E, Britton AM, Luxenberg J, et al. Antipsychotics for delirium. Cochrane Database Syst Rev 2007;(2):CD005594. 246. Lundstr€ om M, Olofsson B, Stenvall M, et al. Postoperative delirium in old patients with femoral neck fracture: a randomized intervention study. Aging Clin Exp Res 2007;19(3):178–1786. 247. Chen JS, Simpson JM, March LM, et al. Fracture risk assessment in frail older people using clinical risk factors. Age Ageing 2008;37(5): 536–541. 248. Nguyen JK, Fouts MM, Kotabe SE, et al. Polypharmacy as a risk factor for adverse drug reactions in geriatric nursing home residents. Am J Geriatr Pharmacother 2006;4:36–41. 249. Morley JE. Conundrums of polypharmacy. Aging Successfully 2003; 13(2):1–2. 250. Van Bruggen R, Gorter K, Stolk RP, et al. Refill adherence and polypharmacy among patients with type 2 diabetes in general practice. Pharmacoepidemiol Drug Saf 2009;18(11):983–991. 251. Beers MH, Ouslander JG, Rollingher I, et al. Explicit criteria for determining inappropriate medication use in nursing home residents. Arch Intern Med 1991;151(9):1825–1832.

626.e6 Messinger-Rapport et al

252. Beers MH. Explicit criteria for determining potentially inappropriate medication use by the elderly. An update. Arch Intern Med 1997; 157(14):1531–1536. 253. Fick DM, Cooper JW, Wade WE, et al. Updating the Beers criteria for potentially inappropriate medication use in older adults: results of a US consensus panel of experts. Arch Intern Med 2003;163(22):2716–2724. 254. Stuart B, Kamal-Bahl S, Briesacher B, et al. Trends in the prescription of inappropriate drugs for the elderly between 1995 and 1999. Am J Geriatr Pharmacother 2003;1(2):61–74. 255. Curtis LH, Østbye T, Sendersky C, et al. Inappropriate prescribing for elderly Americans in a large outpatient population. Arch Intern Med 2004;164(15):1621–1625. 256. Sloane PD, Zimmerman S, Brown LC, et al. Inappropriate medication prescribing in residential care/assisted living facilities. J Am Geriatr Soc 2002;50(6):1001–1011. 257. Zhan C, Sangl J, Bierman AS, et al. Potentially inappropriate medication use in the community-dwelling elderly: findings from the 1996 Medical Expenditure Panel Survey. JAMA 2001;286(22):2823–2829. 258. Swagerty D, Brickley R; American Medical Directors Association and American Society of Consultant Pharmacists joint position statement on the Beers List of Potentially Inappropriate Medications in Older Adults. J Am Med Dir Assoc 2005;6(1):80–86. 259. Lund BC, Carnahan RM, Egge JA, et al. Inappropriate prescribing predicts adverse drug events in older adults. Ann Pharmacother 2010; 44(6):957–963. 260. Ryan C, O’Mahony D, Kennedy J, et al. Potentially inappropriate prescribing in an Irish elderly population in primary care. Br J Clin Pharmacol 2009;68(6):936–947. 261. Bregnhøj L, Thirstrup S, Kristensen MB, et al. Combined intervention programme reduces inappropriate prescribing in elderly patients exposed to polypharmacy in primary care. Eur J Clin Pharmacol 2009; 65(2):199–207. 262. Vinks TH, Egberts TC, de Lange TM, et al. Pharmacist-based medication review reduces potential drug-related problems in the elderly: the SMOG controlled trial. Drugs Aging 2009;26(2):123–133. 263. Laroche ML, Charmes JP, Nouaille Y, et al. Impact of hospitalisation in an acute medical geriatric unit on potentially inappropriate medication use. Drugs Aging 2006;23(1):49–59. 264. Terrell KM, Perkins AJ, Dexter PR, et al. Computerized decision support to reduce potentially inappropriate prescribing to older emergency department patients: a randomized, controlled trial. J Am Geriatr Soc 2009;57(8):1388–1394. 265. Simon SR, Smith DH, Feldstein AC, et al. Computerized prescribing alerts and group academic detailing to reduce the use of potentially inappropriate medications in older people. J Am Geriatr Soc 2006;54(6): 963–968. 266. Raebel MA, Charles J, Dugan J, et al. Randomized trial to improve prescribing safety in ambulatory elderly patients. J Am Geriatr Soc 2007; 55(7):977–985. 267. Pit SW, Byles JE, Cockburn J. Medication Review: Patient selection and general practitioner’s report of drug-related problems and actions taken in elderly Australians. J Am Geriatr Soc 2007;55(6):927–934.

JAMDA – November 2011