User login
Recognizing, managing medical consequences of eating disorders in primary care
Eating disorders are debilitating biopsychosocial illnesses associated with serious medical illness and a high risk of death.1
Primary care physicians are often the first to see young women who have these problems, diagnose them, and start their evaluation and treatment.2–4 Many patients require acute medical interventions as well as long-term care for chronic medical issues. Therefore, primary care physicians play essential front-line and long-term roles in the multidisciplinary treatment team.
DEFINITIONS OF EATING DISORDERS HAVE CHANGED
Several problems existed in the category of eating disorders in the fourth edition of the Diagnostic and Statistical Manual of Mental Disorders (DSM-4) and in the DSM-4 Text Revision (DSM-4-TR). These problems have been addressed in the fifth edition (DSM-5), released in 2013.5
One problem in the earlier editions was that many patients referred for treatment of eating disorders—more than 50% in one study6—did not meet the criteria for anorexia nervosa or bulimia nervosa and thus had to be categorized as having “eating disorder not otherwise specified.” Further, the earlier editions did not recognize that young children and adolescent males can be affected.7
Eating disorders are now recognized as an equal-opportunity disease, with all ethnic and socioeconomic groups affected. Children can run into medical trouble with even a small amount of weight loss or falling off the growth curve. Moreover, children and adolescents do not “experience” their bodies in the same way adults do; they may lack the vocabulary for eating-disorder thoughts.
For these reasons, the definitions of eating disorders have changed in the DSM-5.5
Anorexia nervosa. Older editions of the DSM listed amenorrhea as a criterion. This has been eliminated in DSM-5, since amenorrhea does not necessarily predict medical risk or treatment outcome; also, it is not applicable to males or premenorrheal girls and postmenopausal women.8 In addition, the requirement of low weight is now defined in the context of “age, sex, developmental trajectory, and physical health,” rather than the old threshold of 85% of expected weight.9
What remains unchanged is that anorexia nervosa is still characterized by self-starvation in order to maintain an abnormally low body weight, along with an intense fear of being fat and a disturbed self-image.
Bulimia nervosa. In both the old and the new editions of the DSM, bulimia nervosa is characterized by episodes of binge eating followed by inappropriate compensatory behaviors to avoid weight gain, such as vomiting, laxative abuse, diuretic abuse, and overexercise. In DSM-5, bulimia nervosa no longer has subtypes and requires only one binge per week with compensatory behavior, for at least 3 months. This change was based on the finding that there is no clear difference in psychopathology or treatment outcome between patients with one and two binge-purge episodes a week.10
“Eating disorder not otherwise specified” was a wastebasket category, lumping all those who did not meet the criteria for anorexia nervosa or bulimia nervosa or who did not neatly fit into a specific category.10 In DSM-5, subcategories were designed to help distinguish different treatment needs and outcomes between various subtypes.
Binge-eating disorder, one of the new subcategories, is characterized by binge eating without inappropriate compensatory behaviors.9 Patients with binge-eating disorder are often obese, have greater functional impairment, and are more likely to develop components of metabolic syndrome than obese patients without eating disorders.11
Avoidant/restrictive food intake disorder is another new DSM-5 diagnosis, characterized by failure to meet nutritional needs for reasons other than weight control. Reasons include disinterest in eating, dislike of sensory characteristics of food, or avoidance of consequences of eating. This disorder replaces the category “feeding disorder of infancy or early childhood,” since the condition can also occur in adolescents and adults.12
Other new diagnoses are:
- Atypical anorexia nervosa (if the patient is not underweight)
- Purging disorder
- Subthreshold bulimia nervosa (if the patient has < 1 episode per week or has had them for < 3 months)
- Subthreshold binge eating disorder (< 1 time a week or < 3 months)
- Night eating syndrome
- Pica and rumination disorder.
Regardless of the diagnostic label, the medical evaluation and treatment of anyone with an eating disorder should be tailored to the specific behaviors of the eating disorder. Medical complications can be subdivided into those from starvation, from purging, and from refeeding.
MEDICAL COMPLICATIONS OF STARVATION
Cardiovascular effects of starvation
Malnutrition and starvation have multiple adverse effects on the heart.
Electrophysiologic effects. Sinus bradycardia (< 60 bpm) and hypotension are common cardiac manifestations of starvation.13 Bradycardia has been attributed to an adaptive increase in parasympathetic vagal tone.14 QTc prolongation is also seen in patients with malnutrition.15
Together, these electrocardiographic abnormalities predispose the patient to ventricular arrhythmia and sudden cardiac death.16 The risk of ventricular arrhythmia is particularly relevant when treating psychiatric symptoms, since antipsychotics and tricyclic antidepressants are among several drug classes that can cause further QTc prolongation (Table 1).17,18
In patients with QTc prolongation, bradycardia, or both, the standard of care involves acute hospitalization for refeeding using continuous telemetric monitoring until normal rhythm is restored and the heart rate is above 40 at night and 50 by day.4,19
Structural changes. Starvation also causes structural changes in the heart. Loss of lean body mass can reduce cardiac muscle mass, compromise cardiac output, and lead to mitral valve prolapse.20 These changes are fully reversible with restored nutrition and regaining of heart mass.21,22
Effects of starvation on the brain
Starvation can affect brain structure and cognitive function. Undernourished patients have reduced volumes of white and gray matter, a change that can occur within months. Cortical volumes may increase with weight gain, but a reduction in gray matter volume may not be completely reversible.23
Furthermore, starvation impairs cognitive functions that are needed to stop eating-disorder behaviors; namely, decision-making, emotional control, regulation of appetite, and reward path-ways. Therefore, undernourished patients may not have sufficient insight into the disease to be able to make the best choices for recovery. This finding lends support for using the Maudsley method in adolescents, in which parents take control of their child’s eating until the child can maintain a healthy weight.24
Gastrointestinal consequences of starvation
Patients with malnutrition have prolonged gastric emptying and colonic transit time with solid foods.25 They often complain of early satiety, abdominal pain, bloating, and constipation, all symptoms that complicate the refeeding process. A prokinetic such as metoclopramide (Reglan), given 1 hour before meals and at bedtime, may provide some relief from gastrointestinal symptoms.26
Patients may also experience transient lactose or fructose intolerance after prolonged starvation. Taking a lactase supplement (eg, Lactaid 1–10 tabs) before consuming dairy products and dextrose (contained in candies such as Smarties) before eating fruit or fructose-containing foods can sometimes partially relieve symptoms. In general, gastrointestinal function returns over time as nutritional status improves.
Patients with severe or prolonged starvation can develop steatosis accompanied by elevated levels of aspartate aminotransferase (AST) and alanine aminotransferase (ALT). In reports of starvation-induced steatosis, liver enzyme levels rapidly normalize with nutritional rehabilitation.27
Endocrine consequences of starvation
Amenorrhea. Dysregulation of the hypothalamic-pituitary-gonadal axis is a major endocrine complication of nutritional in-sufficiency. Weight loss disrupts the normal pulsatile secretion of gonadotropin-releasing hormone, reduces secretion of luteinizing hormone and follicle-stimulating hormone, and decreases estrogen levels.28 Leptin deficiency likely plays a role in suppressing gonadotropin secretion with subsequent development of amenorrhea. With weight gain, levels of leptin and gonadotropins normalize and menstruation eventually returns.29,30
Hypothyroidism. Starvation can also lead to dysregulation of the hypothalamic-pituitary-thyroid axis. Typically, the concentration of triiodothyronine (T3) is reduced, the ratio of thyroxine (T4) to T3 is elevated, and thyroid-stimulating hormone (TSH) is close to or within the normal range, creating a euthyroid sick syndrome. In eating disorders, this thyroid disturbance is a result of starvation and resolves with weight restoration. Therefore, thyroid hormone replacement therapy is not medically indicated.28
Osteoporosis. Amenorrhea resulting from low estrogen levels in undernourished patients can raise the risk of osteoporosis and fractures, particularly in patients with a low body mass index. Osteopenia results from a negative balance between bone deposition and resorption.
Lack of bone deposition can be especially problematic when disordered eating occurs during peak bone mass development, ie, ages 11 to 14 for girls, and ages 15 to 17 for boys.31,32 Even a 5% to 10% decrease in bone deposition can result in significant risk of osteopenia.33 However, after age 30, bone resorption is a greater contributor.34
Does hormone therapy correct bone loss? Given the association between estrogen deficiency and bone loss, estrogen supplementation was expected to be an effective treatment for bone loss in patients with eating disorders.35 Also, the restoration of menses through hormone replacement may give underweight patients a false sense of achieving a “healthy” weight.36
Golden et al37 prospectively studied 50 adolescents and found no significant difference in bone mineral density at 1 year of follow-up between patients treated with estrogen and those who received only standard nutritional therapy. However, increased bone mineral density was achieved in adolescents with anorexia nervosa treated with transdermally administered estrogen dosed to mimic physiologic pubertal levels.38
Klibanski et al39 found that hormone therapy resulted in a 4% gain in bone density in an extremely low-weight subset of women with anorexia nervosa (< 70% of ideal body weight), whereas similar patients in the control group lost 20%. However, in all groups, only weight gain correlated with bone gain in women who were within 70% of their ideal body weight.
Divasta et al40 evaluated 60 girls and women ages 13 to 27 with anorexia nervosa, randomized to receive either placebo or dehydroepiandrosterone combined with an estrogen-progestin oral contraceptive, and followed for 18 months. As in the study by Klibanski et al,39 bone loss was prevented in the treatment group, but significant bone gain occurred only in the context of weight gain.
The bottom line is that only weight gain has resulted in significant increases in bone density in patients with anorexia nervosa, and hormone therapy without weight gain has not been shown to increase bone density effectively in this population. Although calcium and vitamin D in oral therapeutic doses through foods or through supplementation are required for bone gain, the combination is not enough to augment bone density in the absence of weight gain.37 Although not curative, weight gain is currently the best option for treating bone loss, and no single pharmacologic treatment is effective.
COMPLICATIONS OF PURGING
Oral complications of purging
Patients who purge by vomiting are at risk of complications from exposure of the esophagus, pharynx, and mouth to acidic gastric contents.
Dental problems. Over time, contact with gastric acid wears down enamel on the lingual and occlusal surfaces of teeth, resulting in dental caries and periodontal disease. Until they can give up purging, patients should be instructed to rinse with mouthwash or water immediately after vomiting to reduce the acidity in the mouth.41,42 We recommend that patients not brush their teeth after vomiting, because brushing can deliver acid to otherwise unreachable surfaces and thus worsen tooth erosion. For patients who are determined to brush after vomiting, a bicarbonate toothpaste might mitigate harm.42
Sialadenosis (hypertrophy of the salivary glands) is another consequence of repeated vomiting, with elevated salivary amylase. Both the size of the glands and the salivary amylase level generally normalize on their own after vomiting is stopped, but parotitis can take up to a year to resolve. Similar to smoker’s cough, parotitis may acutely worsen when the patient abruptly stops vomiting and may worsen before it improves.
To reduce discomfort, patients can use hot compresses or sugarless hard candies.44 However, the latter should not be substituted as a chronic habit in a patient with disordered eating. Patients need to be reassured that the swelling is not permanent, since they often interpret it as having fat cheeks (the “chipmunk sign”).
Hypokalemia, metabolic alkalosis, renal dysfunction
Chronic vomiting can cause electrolyte and acid-base imbalances, the most worrisome of which is hypokalemia. With repeated vomiting, loss of potassium and gastric acid causes metabolic alkalosis with hypokalemia, hypochloremia, and hypomagnesemia. Loss of water and the resultant volume contraction activates the renin-angiotensin-aldosterone system, and elevated aldosterone further decreases serum potassium.
In patients with eating disorders, who often have other factors contributing to electrolyte imbalance, vomiting-induced hypokalemia heightens the risk of cardiac arrhythmias.43
Hypokalemia can also cause rhabdomyolysis and kidney damage.41,43 Prolonged hypokalemia and reduced kidney perfusion in the setting of volume depletion causes acute kidney injury and impaired concentrating ability of the renal tubules. Hypovolemia can cause prerenal azotemia and increases the risk for nephrolithiasis and nephrocalcinosis.44,45
When a patient stops vomiting, elevated aldosterone from prior hypovolemia results in water retention and can manifest in significant edema associated with hypochloremic alkalosis. This condition, known as pseudo-Bartter syndrome, usually resolves without treatment. In the meantime, salt restriction and leg elevation can help reduce edema.26
Laxative abuse: A mode of purging
Many patients with eating disorders abuse laxatives to lose weight or to prevent weight gain. Believing that laxatives will prevent calorie absorption, patients commonly take them to compensate for caloric intake (eg, during a binge episode). The immediate weight loss, albeit artificial, is highly reinforcing for an eating-disorder patient. In some cases, patients with eating disorders also abuse laxatives to self-treat the constipation that results from chronic starvation.46
Over time, tolerance to laxatives develops, and patients use increasingly larger doses. This can lead to activation of the renin-angiotensin-aldosterone system.47 Patients interpret the resultant edema as true weight gain and again take laxatives to get rid of it. If laxatives are stopped abruptly, the patient may need inpatient and outpatient support for the resultant fluid shifts.
Gastrointestinal complications of laxative abuse include reflex hypofunction of the bowel, malabsorption, steatorrhea, and gastrointestinal bleeding.47 Reflex hypofunction during laxative withdrawal is a consequence of the bowel becoming tolerant of laxatives.48 Cathartic colon syndrome is a rare complication characterized by loss of the normal haustral markings and slowed or absent peristalsis in segments of the colon.49
Systemically, the major risk of laxative abuse relates to electrolyte and acid-base imbalance. Loss of potassium and water in the stool can cause hypokalemia and metabolic alkalosis.48 The disturbances caused by laxative abuse are similar to those caused by vomiting and diuretic use and have the same treatment.
The most important component of treating laxative abuse is giving patients realistic expectations to help them tolerate temporary discomfort and to help manage the edema and fluid shifts that can happen acutely with shifting of fluid into the intracellular space. In extreme cases, this may need to be managed in the hospital. To help relieve the initial anxiety, doctors should emphasize that any bloating the patient experiences is not true weight gain and will go away within a few days to weeks. In addition, explaining that laxatives reduce nutrient absorption only minimally may lessen the temptation to resume taking them.48
Diuretic abuse: Another form of purging
Diuretic abuse is yet another mode of purging, with its own set of medical complications. Like laxatives, diuretics are not effective weight-loss agents, and the weight reduction they cause is only temporary.
As with vomiting, there is a compensatory activation of the renin-angiotensin-aldosterone system, and therefore subsequent fluid intake will lead to water retention, which encourages further diuretic use.41 Diuretics can also contribute to hypokalemia, hypomagnesemia, hypochloremia, and metabolic alkalosis.
Ipecac abuse can lead to heart failure
Ipecac syrup has long been used to induce vomiting, but this practice has become much less common since ipecac has become harder to obtain in the United States.50 The emetine base contained in ipecac binds irreversibly to cardiac and skeletal muscle. With continued use, irreversible cardiomyopathy develops and can lead to heart failure. Treatment should include supportive care and immediate cessation of ipecac use.
Diabetic patients may skip insulin to lose weight
Patients with diabetes, especially those with type 1 that begins in childhood, are at greater risk of eating disorders over time.51 They may withhold insulin to lose weight, a practice referred to in the nonmedical literature as “diabulimia,” and they seem particularly more likely to develop bulimia nervosa than those without diabetes.52
The medical prognosis is poor for patients with diabetes who develop eating disorders and do not receive intensive treatment.51 In addition, if a diabetic patient on an insulin pump becomes depressed in addition to having an eating disorder, careful monitoring for suicidal thoughts and a rapid follow-up with mental health services are in order.
REFEEDING SYNDROME
When refeeding is started, a high glucose load stimulates insulin secretion, resulting in cellular uptake of phosphorus along with potassium, magnesium, and glucose. In addition, total body phosphorus is depleted by the increased demand for adenosine triphosphate and 2,3-diphosphoglycerate for cellular metabolism.
When liver enzyme levels increase, the astute clinician will closely monitor the patient for evidence of refeeding syndrome. In a child, adolescent, or young adult, the standard of care is inpatient monitoring for acute stabilization.4,19
Hypophosphatemia is the hallmark of refeeding syndrome, although hypomagnesemia, hypokalemia, and hypoglycemia can also occur.53 In addition, sodium and water retention can lead to fluid overload, with shifting of fluid into the intracellular space, resulting in dependent edema.
Cardiovascular complications are the most worrisome manifestations of refeeding syndrome. Electrolyte shifts and increased fluid volume can cause arrhythmias and heart failure. Furthermore, severely undernourished patients may have reduced myocardial mass as well as electrocardiographic abnormalities associated with starvation, which further increase their vulnerability to electrolyte shifts and fluid retention during refeeding.15
Other manifestations of refeeding syndrome include delirium, seizures, rhabdomyolysis, and respiratory failure. In the most extreme cases, refeeding syndrome causes sudden death.53
Fortunately, refeeding syndrome is easily preventable and treatable when recognized early. Electrolytes and cardiovascular and renal function must be carefully monitored, especially during the first week of nutritional restoration.53 In patients with extremely low body mass (< 70% of ideal body weight) or with precipitous weight loss, close monitoring of the complete metabolic panel including electrolytes, AST, ALT, calcium, magnesium, and phosphorus may be required to detect changes that can affect cardiac status. Specific suggestions for refeeding are discussed below and in Table 2.45
ACUTE CARE OF PATIENTS WITH EATING DISORDERS
Refeeding in the inpatient setting
The decision to hospitalize an eating-disorder patient is based on the current or potential risk of serious medical complications and the likelihood of success at home. Medical criteria for hospital admission are outlined in Table 3.4,54
In refeeding undernourished patients, the challenge is to maximize weight gain while preventing refeeding syndrome. Undernourished patients are generally hypometabolic at baseline but become hypermetabolic once refeeding begins.
How many calories should refeeding start with? The traditional principle of “start low and go slow” has been recently challenged.55 Starting at 1,200 kcal/day or less in the typical patient can result in failure to gain weight or even in weight loss in the first week of refeeding.56 The goal is to achieve a weight gain of 0.2 kg/day while the patient is in the hospital. Thus, we start higher, and to date we have seen no cases of life-threatening refeeding syndrome. In all patients who need hospitalization or who are beginning the refeeding process as outpatients, caloric intake should be started at 1,500 to 2,000 kcal/day.45,57 However, for exceptionally low-weight patients, intake may be started lower.
In Australia, patients are started at 1,900 kcal/day.56 All patients in one program there receive nasogastric feeding initially in an intensive care unit and then are moved to a regular nursing floor where they graduate to full oral feeding as they improve cardiovascularly and behaviorally. In the United States, some programs use nasogastric feeding at night for caloric restoration; our program and others use nasogastric feeding as a behavioral modification strategy for patients who refuse food or supplements by mouth.
Phosphorus supplementation. Many centers give phosphorus supplements preventively. In our center, we give potassium phosphate (Neutra-Phos) 500 mg orally twice daily for 5 days, and we have seen no life-threatening cases of refeeding syndrome with that regimen. Other centers give phosphorus supplements in a dose of 250 mg orally twice a day for 5 days, while still others only supplement phosphorus reactively once a deficit has been identified. The latter method requires daily blood draws for monitoring and is reactive rather than proactive. Further studies can help clarify the optimal dosing and timing of phosphorus supplementation.
Managing fluid balance. Fluid-loading these patients may tip them over the edge into refeeding syndrome. Except in cases of shock, patients with eating disorders should not be given intravenous fluids, as it is safer to rehydrate and feed them orally. Electrolyte imbalances can be corrected orally with no need for intravenous supplementation. To avoid fluid overload, fluids can be started at 1,500 mL to 2,000 mL per day, with strict monitoring of intake and output. Fluids are liberalized if ALT and AST levels remain normal and to gradually correct orthostatic hypotension; caloric fluids are ideal to help address energy needs and improve bradycardia.
Laboratory monitoring. On admission, a urinalysis, complete blood cell count, complete metabolic panel, TSH, erythrocyte sedimentation rate, serum magnesium, and phosphorus should be obtained.26 In addition, continuous electrocardiographic recording should begin on admission.45 Inpatient use of a telemetry bed helps identify extreme tachycardia with arrhythmia, as well as profound bradycardia.45,56
Some protocols call for daily laboratory monitoring, although that degree of testing is less cost-effective. If initial results are normal, clinical judgment can be used on when to repeat laboratory evaluation. For instance, patients with edema require repeat complete metabolic panels to assess for elevated ALT and AST, electrolyte imbalances, and other abnormalities.
Signs of refeeding syndrome include tachycardia, hepatosplenomegaly, peripheral edema, altered mental status, and electrolyte disturbances, specifically, acute or severe hypophosphatemia or hypokalemia.26,45 If refeeding syndrome is suspected, the rate of caloric intake should be reduced or not advanced, fluid intake should be urgently reassessed for volume overload, and supportive care with close monitoring should be provided.
KNOWLEDGE SAVES LIVES
Eating disorders can lead to potentially life-threatening medical complications that require attentive care by the primary care clinician and subspecialist. Without thoughtful consideration, it is easy for even a caring medical team to unintentionally enable patients with these illnesses or to cause active harm in the case of underrecognized pathology.58
Acute medical stabilization on an inpatient unit trained to recognize pathology and treat sequelae can be lifesaving. Arming patients and families with medical knowledge, as provided in the Academy for Eating Disorders’ brochure, “Critical Points for Early Recognition and Medical Risk Management in the Care of Individuals with Eating Disorders”59 can help save patients’ lives.
- Arcelus J, Mitchell AJ, Wales J, Nielsen S. Mortality rates in patients with anorexia nervosa and other eating disorders. A meta-analysis of 36 studies. Arch Gen Psychiatry 2011; 68:724–731.
- Walsh JM, Wheat ME, Freund K. Detection, evaluation, and treatment of eating disorders the role of the primary care physician. J Gen Intern Med 2000; 15:577–590.
- American Academy of Pediatrics; Committee on Adolescence. Identifying and treating eating disorders. Pediatrics 2003; 111:204–211.
- Rosen DS; American Academy of Pediatrics Committee on Adolescence. Identification and management of eating disorders in children and adolescents. Pediatrics 2010; 126:1240–1253.
- American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders. 5th edition. Arlington, VA: American Psychiatric Publishing, Incorporated; 2013.
- Eddy KT, Celio Doyle A, Hoste RR, Herzog DB, le Grange D. Eating disorder not otherwise specified in adolescents. J Am Acad Child Adolesc Psychiatry 2008; 47:156–164.
- Muise AM, Stein DG, Arbess G. Eating disorders in adolescent boys: a review of the adolescent and young adult literature. J Adolesc Health 2003; 33:427–435.
- Attia E, Roberto CA. Should amenorrhea be a diagnostic criterion for anorexia nervosa? Int J Eat Disord 2009; 42:581–589.
- American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders, fifth edition. http://dsm.psychiatryonline.org/content.aspx?bookid=556§ionid=41101776#103439089. Accessed January 31, 2014.
- Wilfley DE, Bishop ME, Wilson GT, Agras WS. Classification of eating disorders: toward DSM-V. Int J Eat Disord 2007; 40:S123–S129.
- Wonderlich SA, Gordon KH, Mitchell JE, Crosby RD, Engel SG. The validity and clinical utility of binge eating disorder. Int J Eat Disord 2009; 42:687–705.
- Ornstein RM, Rosen DS, Mammel KA, et al. Distribution of eating disorders in children and adolescents using the proposed DSM-5 criteria for feeding and eating disorders. J Adolesc Health 2013: 53:303–305.
- Winston AP, Stafford PJ. Cardiovascular effects of anorexia nervosa. Eur Eat Disord Rev 2000; 8:117–125.
- Galetta F, Franzoni F, Prattichizzo F, Rolla M, Santoro G, Pentimone F. Heart rate variability and left ventricular diastolic function in anorexia nervosa. J Adolesc Health 2003; 32:416–421.
- McCallum K, Bermudez O, Ohlemeyer C, Tyson E, Portilla M, Ferdman B. How should the clinician evaluate and manage the cardiovascular complications of anorexia nervosa? Eat Disord 2006; 14:73–80.
- Akhtar M. Clinical spectrum of ventricular tachycardia. Circulation 1990; 82:1561–1573.
- Beach SR, Celano CM, Noseworthy PA, Januzzi JL, Huffman JC. QTc prolongation, torsades de pointes, and psychotropic medications. Psychosomatics 2013; 54:1–13.
- The University of Arizona Center for Education and Research on Therapeutics. QT Drug Lists. http://crediblemeds.org/everyone/compos-ite-list-all-qtdrugs/?rf=US. Accessed January 31, 2014.
- Rome ES, Ammerman S. Medical complications of eating disorders: an update. J Adolesc Health 2003; 33:418–426.
- Romano C, Chinali M, Pasanisi F, et al. Reduced hemodynamic load and cardiac hypotrophy in patients with anorexia nervosa. Am J Clin Nutr 2003; 77:308–312.
- Shamim T, Golden NH, Arden M, Filiberto L, Shenker IR. Resolution of vital sign instability: an objective measure of medical stability in anorexia nervosa. J Adolesc Health 2003; 32:73–77.
- Mont L, Castro J, Herreros B, et al. Reversibility of cardiac abnormalities in adolescents with anorexia nervosa after weight recovery. J Am Acad Child Adolesc Psychiatry 2003; 42:808–813.
- Roberto CA, Mayer LE, Brickman AM, et al. Brain tissue volume changes following weight gain in adults with anorexia nervosa. Int J Eat Disord 2011; 44:406–411.
- Treasure J, Russell G. The case for early intervention in anorexia nervosa: theoretical exploration of maintaining factors. Br J Psychiatry 2011; 199:5–7.
- Hadley SJ, Walsh BT. Gastrointestinal disturbances in anorexia nervosa and bulimia nervosa. Curr Drug Targets CNS Neurol Disord 2003; 2:1–9.
- Yager J, Andersen AE. Clinical practice. Anorexia nervosa. N Engl J Med 2005; 353:1481–1488.
- De Caprio C, Alfano A, Senatore I, Zarrella L, Pasanisi F, Contaldo F. Severe acute liver damage in anorexia nervosa: two case reports. Nutrition 2006; 22:572–575.
- Lawson EA, Klibanski A. Endocrine abnormalities in anorexia nervosa. Nat Clin Pract Endocrinol Metab 2008; 4:407–414.
- Holtkamp K, Mika C, Grzella I, et al. Reproductive function during weight gain in anorexia nervosa. Leptin represents a metabolic gate to gonadotropin secretion. J Neural Transm 2003; 110:427–435.
- Golden NH, Jacobson MS, Schebendach J, Solanto MV, Hertz SM, Shenker IR. Resumption of menses in anorexia nervosa. Arch Pediatr Adolesc Med 1997; 151:16–21.
- Soyka LA, Misra M, Frenchman A, et al. Abnormal bone mineral accrual in adolescent girls with anorexia nervosa. J Clin Endocrinol Metab 2002; 87:4177–4185.
- Misra M, Klibanski A. Bone metabolism in adolescents with anorexia nervosa. J Endocrinol Invest 2011; 34:324–332.
- Recker RR, Davies KM, Hinders SM, Heaney RP, Stegman MR, Kimmel DB. Bone gain in young adult women. JAMA 1992; 268:2403–2408.
- Biller BM, Saxe V, Herzog DB, Rosenthal DI, Holzman S, Klibanski A. Mechanisms of osteoporosis in adult and adolescent women with anorexia nervosa. J Clin Endocrinol Metab 1989; 68:548–554.
- Hergenroeder AC, Smith EO, Shypailo R, Jones LA, Klish WJ, Ellis K. Bone mineral changes in young women with hypothalamic amenorrhea treated with oral contraceptives, medroxyprogesterone, or placebo over 12 months. Am J Obstet Gynecol 1997; 176:1017–1025.
- Sim LA, McGovern L, Elamin MB, Swiglo BA, Erwin PJ, Montori VM. Effect on bone health of estrogen preparations in premenopausal women with anorexia nervosa: a systematic review and meta-analyses. Int J Eat Disord 2010; 43:218–225.
- Golden NH, Lanzkowsky L, Schebendach J, Palestro CJ, Jacobson MS, Shenker IR. The effect of estrogen-progestin treatment on bone mineral density in anorexia nervosa. J Pediatr Adolesc Gynecol 2002; 15:135–143.
- Misra M, Katzman D, Miller KK, et al. Physiologic estrogen replacement increases bone density in adolescent girls with anorexia nervosa. J Bone Miner Res 2011; 26:2430–2438.
- Klibanski A, Biller BM, Schoenfeld DA, Herzog DB, Saxe VC. The effects of estrogen administration on trabecular bone loss in young women with anorexia nervosa. J Clin Endocrinol Metab 1995; 80:898–904.
- Divasta AD, Feldman HA, Giancaterino C, Rosen CJ, Leboff MS, Gordon CM. The effect of gonadal and adrenal steroid therapy on skeletal health in adolescents and young women with anorexia nervosa. Metabolism 2012; 61:1010–1020.
- Mehler PS. Medical complications of bulimia nervosa and their treatments. Int J Eat Disord 2011; 44:95–104.
- Milosevic A. Eating disorders and the dentist. Br Dent J 1999; 186:109–113.
- Greenfeld D, Mickley D, Quinlan DM, Roloff P. Hypokalemia in outpatients with eating disorders. Am J Psychiatry 1995; 152:60–63.
- Bouquegneau A, Dubois BE, Krzesinski JM, Delanaye P. Anorexia nervosa and the kidney. Am J Kidney Dis 2012; 60:299–307.
- Auron M, Rome E. Anorexia nervosa and bulimia nervosa: what the hospitalist needs to know about CPT 269.9, or nutritional insufficiency. ACP Hospitalist 2011 Sept:28–45.
- Steffen KJ, Mitchell JE, Roerig JL, Lancaster KL. The eating disorders medicine cabinet revisited: a clinician’s guide to ipecac and laxatives. Int J Eat Disord 2007; 40:360–368.
- Roerig JL, Steffen KJ, Mitchell JE, Zunker C. Laxative abuse: epidemiology, diagnosis and management. Drugs 2010; 70:1487–1503.
- Mitchell JE, Boutacoff LI. Laxative abuse complicating bulimia: medical and treatment implications. Int J Eat Disord 1986; 5:325–334.
- Joo JS, Ehrenpreis ED, Gonzalez L, et al. Alterations in colonic anatomy induced by chronic stimulant laxatives: the cathartic colon revisited. J Clin Gastroenterol 1998; 26:283–286.
- Drugs.com. Ipecac syrup. www.drugs.com/monograph/ipecac-syrup.html. Accessed January 31, 2014.
- Peveler RC, Bryden KS, Neil HA, et al. The relationship of disordered eating habits and attitudes to clinical outcomes in young adult females with type 1 diabetes. Diabetes Care 2005; 28:84–88.
- Mannucci E, Rotella F, Ricca V, Moretti S, Placidi GF, Rotella CM. Eating disorders in patients with type 1 diabetes: a meta-analysis. J Endocrinol Invest 2005; 28:417–419.
- Crook MA, Hally V, Panteli JV. The importance of the refeeding syndrome. Nutrition 2001; 17:632–637.
- Fisher M, Golden NH, Katzman DK, et al. Eating disorders in adolescents: a background paper. J Adolesc Health 1995; 16:420–437.
- Kohn MR, Madden S, Clarke SD. Refeeding in anorexia nervosa: increased safety and efficiency through understanding the pathophysiology of protein calorie malnutrition. Curr Opin Pediatr 2011; 23:390–394.
- Garber AK, Michihata N, Hetnal K, Shafer MA, Moscicki AB. A prospective examination of weight gain in hospitalized adolescents with anorexia nervosa on a recommended refeeding protocol. J Adolesc Health 2012; 50:24–29.
- Whitelaw M, Gilbertson H, Lam PY, Sawyer SM. Does aggressive refeeding in hospitalized adolescents with anorexia nervosa result in increased hypophosphatemia? J Adolesc Health 2010; 46:577–582.
- Treasure J, Crane A, McKnight R, Buchanan E, Wolfe M. First do no harm: iatrogenic maintaining factors in anorexia nervosa. Eur Eat Disord Rev 2011; 19:296–302.
- Academy for Eating Disorders (AED). Critical points for early recognition and medical risk management in the care of individuals with eating disorders. http://www.aedweb.org/AM/Template.cfm?Section=Medical_Care_Standards&Template=/CM/ContentDisplay.cfm&ContentID=2413. Accessed January 31, 2014.
Eating disorders are debilitating biopsychosocial illnesses associated with serious medical illness and a high risk of death.1
Primary care physicians are often the first to see young women who have these problems, diagnose them, and start their evaluation and treatment.2–4 Many patients require acute medical interventions as well as long-term care for chronic medical issues. Therefore, primary care physicians play essential front-line and long-term roles in the multidisciplinary treatment team.
DEFINITIONS OF EATING DISORDERS HAVE CHANGED
Several problems existed in the category of eating disorders in the fourth edition of the Diagnostic and Statistical Manual of Mental Disorders (DSM-4) and in the DSM-4 Text Revision (DSM-4-TR). These problems have been addressed in the fifth edition (DSM-5), released in 2013.5
One problem in the earlier editions was that many patients referred for treatment of eating disorders—more than 50% in one study6—did not meet the criteria for anorexia nervosa or bulimia nervosa and thus had to be categorized as having “eating disorder not otherwise specified.” Further, the earlier editions did not recognize that young children and adolescent males can be affected.7
Eating disorders are now recognized as an equal-opportunity disease, with all ethnic and socioeconomic groups affected. Children can run into medical trouble with even a small amount of weight loss or falling off the growth curve. Moreover, children and adolescents do not “experience” their bodies in the same way adults do; they may lack the vocabulary for eating-disorder thoughts.
For these reasons, the definitions of eating disorders have changed in the DSM-5.5
Anorexia nervosa. Older editions of the DSM listed amenorrhea as a criterion. This has been eliminated in DSM-5, since amenorrhea does not necessarily predict medical risk or treatment outcome; also, it is not applicable to males or premenorrheal girls and postmenopausal women.8 In addition, the requirement of low weight is now defined in the context of “age, sex, developmental trajectory, and physical health,” rather than the old threshold of 85% of expected weight.9
What remains unchanged is that anorexia nervosa is still characterized by self-starvation in order to maintain an abnormally low body weight, along with an intense fear of being fat and a disturbed self-image.
Bulimia nervosa. In both the old and the new editions of the DSM, bulimia nervosa is characterized by episodes of binge eating followed by inappropriate compensatory behaviors to avoid weight gain, such as vomiting, laxative abuse, diuretic abuse, and overexercise. In DSM-5, bulimia nervosa no longer has subtypes and requires only one binge per week with compensatory behavior, for at least 3 months. This change was based on the finding that there is no clear difference in psychopathology or treatment outcome between patients with one and two binge-purge episodes a week.10
“Eating disorder not otherwise specified” was a wastebasket category, lumping all those who did not meet the criteria for anorexia nervosa or bulimia nervosa or who did not neatly fit into a specific category.10 In DSM-5, subcategories were designed to help distinguish different treatment needs and outcomes between various subtypes.
Binge-eating disorder, one of the new subcategories, is characterized by binge eating without inappropriate compensatory behaviors.9 Patients with binge-eating disorder are often obese, have greater functional impairment, and are more likely to develop components of metabolic syndrome than obese patients without eating disorders.11
Avoidant/restrictive food intake disorder is another new DSM-5 diagnosis, characterized by failure to meet nutritional needs for reasons other than weight control. Reasons include disinterest in eating, dislike of sensory characteristics of food, or avoidance of consequences of eating. This disorder replaces the category “feeding disorder of infancy or early childhood,” since the condition can also occur in adolescents and adults.12
Other new diagnoses are:
- Atypical anorexia nervosa (if the patient is not underweight)
- Purging disorder
- Subthreshold bulimia nervosa (if the patient has < 1 episode per week or has had them for < 3 months)
- Subthreshold binge eating disorder (< 1 time a week or < 3 months)
- Night eating syndrome
- Pica and rumination disorder.
Regardless of the diagnostic label, the medical evaluation and treatment of anyone with an eating disorder should be tailored to the specific behaviors of the eating disorder. Medical complications can be subdivided into those from starvation, from purging, and from refeeding.
MEDICAL COMPLICATIONS OF STARVATION
Cardiovascular effects of starvation
Malnutrition and starvation have multiple adverse effects on the heart.
Electrophysiologic effects. Sinus bradycardia (< 60 bpm) and hypotension are common cardiac manifestations of starvation.13 Bradycardia has been attributed to an adaptive increase in parasympathetic vagal tone.14 QTc prolongation is also seen in patients with malnutrition.15
Together, these electrocardiographic abnormalities predispose the patient to ventricular arrhythmia and sudden cardiac death.16 The risk of ventricular arrhythmia is particularly relevant when treating psychiatric symptoms, since antipsychotics and tricyclic antidepressants are among several drug classes that can cause further QTc prolongation (Table 1).17,18
In patients with QTc prolongation, bradycardia, or both, the standard of care involves acute hospitalization for refeeding using continuous telemetric monitoring until normal rhythm is restored and the heart rate is above 40 at night and 50 by day.4,19
Structural changes. Starvation also causes structural changes in the heart. Loss of lean body mass can reduce cardiac muscle mass, compromise cardiac output, and lead to mitral valve prolapse.20 These changes are fully reversible with restored nutrition and regaining of heart mass.21,22
Effects of starvation on the brain
Starvation can affect brain structure and cognitive function. Undernourished patients have reduced volumes of white and gray matter, a change that can occur within months. Cortical volumes may increase with weight gain, but a reduction in gray matter volume may not be completely reversible.23
Furthermore, starvation impairs cognitive functions that are needed to stop eating-disorder behaviors; namely, decision-making, emotional control, regulation of appetite, and reward path-ways. Therefore, undernourished patients may not have sufficient insight into the disease to be able to make the best choices for recovery. This finding lends support for using the Maudsley method in adolescents, in which parents take control of their child’s eating until the child can maintain a healthy weight.24
Gastrointestinal consequences of starvation
Patients with malnutrition have prolonged gastric emptying and colonic transit time with solid foods.25 They often complain of early satiety, abdominal pain, bloating, and constipation, all symptoms that complicate the refeeding process. A prokinetic such as metoclopramide (Reglan), given 1 hour before meals and at bedtime, may provide some relief from gastrointestinal symptoms.26
Patients may also experience transient lactose or fructose intolerance after prolonged starvation. Taking a lactase supplement (eg, Lactaid 1–10 tabs) before consuming dairy products and dextrose (contained in candies such as Smarties) before eating fruit or fructose-containing foods can sometimes partially relieve symptoms. In general, gastrointestinal function returns over time as nutritional status improves.
Patients with severe or prolonged starvation can develop steatosis accompanied by elevated levels of aspartate aminotransferase (AST) and alanine aminotransferase (ALT). In reports of starvation-induced steatosis, liver enzyme levels rapidly normalize with nutritional rehabilitation.27
Endocrine consequences of starvation
Amenorrhea. Dysregulation of the hypothalamic-pituitary-gonadal axis is a major endocrine complication of nutritional in-sufficiency. Weight loss disrupts the normal pulsatile secretion of gonadotropin-releasing hormone, reduces secretion of luteinizing hormone and follicle-stimulating hormone, and decreases estrogen levels.28 Leptin deficiency likely plays a role in suppressing gonadotropin secretion with subsequent development of amenorrhea. With weight gain, levels of leptin and gonadotropins normalize and menstruation eventually returns.29,30
Hypothyroidism. Starvation can also lead to dysregulation of the hypothalamic-pituitary-thyroid axis. Typically, the concentration of triiodothyronine (T3) is reduced, the ratio of thyroxine (T4) to T3 is elevated, and thyroid-stimulating hormone (TSH) is close to or within the normal range, creating a euthyroid sick syndrome. In eating disorders, this thyroid disturbance is a result of starvation and resolves with weight restoration. Therefore, thyroid hormone replacement therapy is not medically indicated.28
Osteoporosis. Amenorrhea resulting from low estrogen levels in undernourished patients can raise the risk of osteoporosis and fractures, particularly in patients with a low body mass index. Osteopenia results from a negative balance between bone deposition and resorption.
Lack of bone deposition can be especially problematic when disordered eating occurs during peak bone mass development, ie, ages 11 to 14 for girls, and ages 15 to 17 for boys.31,32 Even a 5% to 10% decrease in bone deposition can result in significant risk of osteopenia.33 However, after age 30, bone resorption is a greater contributor.34
Does hormone therapy correct bone loss? Given the association between estrogen deficiency and bone loss, estrogen supplementation was expected to be an effective treatment for bone loss in patients with eating disorders.35 Also, the restoration of menses through hormone replacement may give underweight patients a false sense of achieving a “healthy” weight.36
Golden et al37 prospectively studied 50 adolescents and found no significant difference in bone mineral density at 1 year of follow-up between patients treated with estrogen and those who received only standard nutritional therapy. However, increased bone mineral density was achieved in adolescents with anorexia nervosa treated with transdermally administered estrogen dosed to mimic physiologic pubertal levels.38
Klibanski et al39 found that hormone therapy resulted in a 4% gain in bone density in an extremely low-weight subset of women with anorexia nervosa (< 70% of ideal body weight), whereas similar patients in the control group lost 20%. However, in all groups, only weight gain correlated with bone gain in women who were within 70% of their ideal body weight.
Divasta et al40 evaluated 60 girls and women ages 13 to 27 with anorexia nervosa, randomized to receive either placebo or dehydroepiandrosterone combined with an estrogen-progestin oral contraceptive, and followed for 18 months. As in the study by Klibanski et al,39 bone loss was prevented in the treatment group, but significant bone gain occurred only in the context of weight gain.
The bottom line is that only weight gain has resulted in significant increases in bone density in patients with anorexia nervosa, and hormone therapy without weight gain has not been shown to increase bone density effectively in this population. Although calcium and vitamin D in oral therapeutic doses through foods or through supplementation are required for bone gain, the combination is not enough to augment bone density in the absence of weight gain.37 Although not curative, weight gain is currently the best option for treating bone loss, and no single pharmacologic treatment is effective.
COMPLICATIONS OF PURGING
Oral complications of purging
Patients who purge by vomiting are at risk of complications from exposure of the esophagus, pharynx, and mouth to acidic gastric contents.
Dental problems. Over time, contact with gastric acid wears down enamel on the lingual and occlusal surfaces of teeth, resulting in dental caries and periodontal disease. Until they can give up purging, patients should be instructed to rinse with mouthwash or water immediately after vomiting to reduce the acidity in the mouth.41,42 We recommend that patients not brush their teeth after vomiting, because brushing can deliver acid to otherwise unreachable surfaces and thus worsen tooth erosion. For patients who are determined to brush after vomiting, a bicarbonate toothpaste might mitigate harm.42
Sialadenosis (hypertrophy of the salivary glands) is another consequence of repeated vomiting, with elevated salivary amylase. Both the size of the glands and the salivary amylase level generally normalize on their own after vomiting is stopped, but parotitis can take up to a year to resolve. Similar to smoker’s cough, parotitis may acutely worsen when the patient abruptly stops vomiting and may worsen before it improves.
To reduce discomfort, patients can use hot compresses or sugarless hard candies.44 However, the latter should not be substituted as a chronic habit in a patient with disordered eating. Patients need to be reassured that the swelling is not permanent, since they often interpret it as having fat cheeks (the “chipmunk sign”).
Hypokalemia, metabolic alkalosis, renal dysfunction
Chronic vomiting can cause electrolyte and acid-base imbalances, the most worrisome of which is hypokalemia. With repeated vomiting, loss of potassium and gastric acid causes metabolic alkalosis with hypokalemia, hypochloremia, and hypomagnesemia. Loss of water and the resultant volume contraction activates the renin-angiotensin-aldosterone system, and elevated aldosterone further decreases serum potassium.
In patients with eating disorders, who often have other factors contributing to electrolyte imbalance, vomiting-induced hypokalemia heightens the risk of cardiac arrhythmias.43
Hypokalemia can also cause rhabdomyolysis and kidney damage.41,43 Prolonged hypokalemia and reduced kidney perfusion in the setting of volume depletion causes acute kidney injury and impaired concentrating ability of the renal tubules. Hypovolemia can cause prerenal azotemia and increases the risk for nephrolithiasis and nephrocalcinosis.44,45
When a patient stops vomiting, elevated aldosterone from prior hypovolemia results in water retention and can manifest in significant edema associated with hypochloremic alkalosis. This condition, known as pseudo-Bartter syndrome, usually resolves without treatment. In the meantime, salt restriction and leg elevation can help reduce edema.26
Laxative abuse: A mode of purging
Many patients with eating disorders abuse laxatives to lose weight or to prevent weight gain. Believing that laxatives will prevent calorie absorption, patients commonly take them to compensate for caloric intake (eg, during a binge episode). The immediate weight loss, albeit artificial, is highly reinforcing for an eating-disorder patient. In some cases, patients with eating disorders also abuse laxatives to self-treat the constipation that results from chronic starvation.46
Over time, tolerance to laxatives develops, and patients use increasingly larger doses. This can lead to activation of the renin-angiotensin-aldosterone system.47 Patients interpret the resultant edema as true weight gain and again take laxatives to get rid of it. If laxatives are stopped abruptly, the patient may need inpatient and outpatient support for the resultant fluid shifts.
Gastrointestinal complications of laxative abuse include reflex hypofunction of the bowel, malabsorption, steatorrhea, and gastrointestinal bleeding.47 Reflex hypofunction during laxative withdrawal is a consequence of the bowel becoming tolerant of laxatives.48 Cathartic colon syndrome is a rare complication characterized by loss of the normal haustral markings and slowed or absent peristalsis in segments of the colon.49
Systemically, the major risk of laxative abuse relates to electrolyte and acid-base imbalance. Loss of potassium and water in the stool can cause hypokalemia and metabolic alkalosis.48 The disturbances caused by laxative abuse are similar to those caused by vomiting and diuretic use and have the same treatment.
The most important component of treating laxative abuse is giving patients realistic expectations to help them tolerate temporary discomfort and to help manage the edema and fluid shifts that can happen acutely with shifting of fluid into the intracellular space. In extreme cases, this may need to be managed in the hospital. To help relieve the initial anxiety, doctors should emphasize that any bloating the patient experiences is not true weight gain and will go away within a few days to weeks. In addition, explaining that laxatives reduce nutrient absorption only minimally may lessen the temptation to resume taking them.48
Diuretic abuse: Another form of purging
Diuretic abuse is yet another mode of purging, with its own set of medical complications. Like laxatives, diuretics are not effective weight-loss agents, and the weight reduction they cause is only temporary.
As with vomiting, there is a compensatory activation of the renin-angiotensin-aldosterone system, and therefore subsequent fluid intake will lead to water retention, which encourages further diuretic use.41 Diuretics can also contribute to hypokalemia, hypomagnesemia, hypochloremia, and metabolic alkalosis.
Ipecac abuse can lead to heart failure
Ipecac syrup has long been used to induce vomiting, but this practice has become much less common since ipecac has become harder to obtain in the United States.50 The emetine base contained in ipecac binds irreversibly to cardiac and skeletal muscle. With continued use, irreversible cardiomyopathy develops and can lead to heart failure. Treatment should include supportive care and immediate cessation of ipecac use.
Diabetic patients may skip insulin to lose weight
Patients with diabetes, especially those with type 1 that begins in childhood, are at greater risk of eating disorders over time.51 They may withhold insulin to lose weight, a practice referred to in the nonmedical literature as “diabulimia,” and they seem particularly more likely to develop bulimia nervosa than those without diabetes.52
The medical prognosis is poor for patients with diabetes who develop eating disorders and do not receive intensive treatment.51 In addition, if a diabetic patient on an insulin pump becomes depressed in addition to having an eating disorder, careful monitoring for suicidal thoughts and a rapid follow-up with mental health services are in order.
REFEEDING SYNDROME
When refeeding is started, a high glucose load stimulates insulin secretion, resulting in cellular uptake of phosphorus along with potassium, magnesium, and glucose. In addition, total body phosphorus is depleted by the increased demand for adenosine triphosphate and 2,3-diphosphoglycerate for cellular metabolism.
When liver enzyme levels increase, the astute clinician will closely monitor the patient for evidence of refeeding syndrome. In a child, adolescent, or young adult, the standard of care is inpatient monitoring for acute stabilization.4,19
Hypophosphatemia is the hallmark of refeeding syndrome, although hypomagnesemia, hypokalemia, and hypoglycemia can also occur.53 In addition, sodium and water retention can lead to fluid overload, with shifting of fluid into the intracellular space, resulting in dependent edema.
Cardiovascular complications are the most worrisome manifestations of refeeding syndrome. Electrolyte shifts and increased fluid volume can cause arrhythmias and heart failure. Furthermore, severely undernourished patients may have reduced myocardial mass as well as electrocardiographic abnormalities associated with starvation, which further increase their vulnerability to electrolyte shifts and fluid retention during refeeding.15
Other manifestations of refeeding syndrome include delirium, seizures, rhabdomyolysis, and respiratory failure. In the most extreme cases, refeeding syndrome causes sudden death.53
Fortunately, refeeding syndrome is easily preventable and treatable when recognized early. Electrolytes and cardiovascular and renal function must be carefully monitored, especially during the first week of nutritional restoration.53 In patients with extremely low body mass (< 70% of ideal body weight) or with precipitous weight loss, close monitoring of the complete metabolic panel including electrolytes, AST, ALT, calcium, magnesium, and phosphorus may be required to detect changes that can affect cardiac status. Specific suggestions for refeeding are discussed below and in Table 2.45
ACUTE CARE OF PATIENTS WITH EATING DISORDERS
Refeeding in the inpatient setting
The decision to hospitalize an eating-disorder patient is based on the current or potential risk of serious medical complications and the likelihood of success at home. Medical criteria for hospital admission are outlined in Table 3.4,54
In refeeding undernourished patients, the challenge is to maximize weight gain while preventing refeeding syndrome. Undernourished patients are generally hypometabolic at baseline but become hypermetabolic once refeeding begins.
How many calories should refeeding start with? The traditional principle of “start low and go slow” has been recently challenged.55 Starting at 1,200 kcal/day or less in the typical patient can result in failure to gain weight or even in weight loss in the first week of refeeding.56 The goal is to achieve a weight gain of 0.2 kg/day while the patient is in the hospital. Thus, we start higher, and to date we have seen no cases of life-threatening refeeding syndrome. In all patients who need hospitalization or who are beginning the refeeding process as outpatients, caloric intake should be started at 1,500 to 2,000 kcal/day.45,57 However, for exceptionally low-weight patients, intake may be started lower.
In Australia, patients are started at 1,900 kcal/day.56 All patients in one program there receive nasogastric feeding initially in an intensive care unit and then are moved to a regular nursing floor where they graduate to full oral feeding as they improve cardiovascularly and behaviorally. In the United States, some programs use nasogastric feeding at night for caloric restoration; our program and others use nasogastric feeding as a behavioral modification strategy for patients who refuse food or supplements by mouth.
Phosphorus supplementation. Many centers give phosphorus supplements preventively. In our center, we give potassium phosphate (Neutra-Phos) 500 mg orally twice daily for 5 days, and we have seen no life-threatening cases of refeeding syndrome with that regimen. Other centers give phosphorus supplements in a dose of 250 mg orally twice a day for 5 days, while still others only supplement phosphorus reactively once a deficit has been identified. The latter method requires daily blood draws for monitoring and is reactive rather than proactive. Further studies can help clarify the optimal dosing and timing of phosphorus supplementation.
Managing fluid balance. Fluid-loading these patients may tip them over the edge into refeeding syndrome. Except in cases of shock, patients with eating disorders should not be given intravenous fluids, as it is safer to rehydrate and feed them orally. Electrolyte imbalances can be corrected orally with no need for intravenous supplementation. To avoid fluid overload, fluids can be started at 1,500 mL to 2,000 mL per day, with strict monitoring of intake and output. Fluids are liberalized if ALT and AST levels remain normal and to gradually correct orthostatic hypotension; caloric fluids are ideal to help address energy needs and improve bradycardia.
Laboratory monitoring. On admission, a urinalysis, complete blood cell count, complete metabolic panel, TSH, erythrocyte sedimentation rate, serum magnesium, and phosphorus should be obtained.26 In addition, continuous electrocardiographic recording should begin on admission.45 Inpatient use of a telemetry bed helps identify extreme tachycardia with arrhythmia, as well as profound bradycardia.45,56
Some protocols call for daily laboratory monitoring, although that degree of testing is less cost-effective. If initial results are normal, clinical judgment can be used on when to repeat laboratory evaluation. For instance, patients with edema require repeat complete metabolic panels to assess for elevated ALT and AST, electrolyte imbalances, and other abnormalities.
Signs of refeeding syndrome include tachycardia, hepatosplenomegaly, peripheral edema, altered mental status, and electrolyte disturbances, specifically, acute or severe hypophosphatemia or hypokalemia.26,45 If refeeding syndrome is suspected, the rate of caloric intake should be reduced or not advanced, fluid intake should be urgently reassessed for volume overload, and supportive care with close monitoring should be provided.
KNOWLEDGE SAVES LIVES
Eating disorders can lead to potentially life-threatening medical complications that require attentive care by the primary care clinician and subspecialist. Without thoughtful consideration, it is easy for even a caring medical team to unintentionally enable patients with these illnesses or to cause active harm in the case of underrecognized pathology.58
Acute medical stabilization on an inpatient unit trained to recognize pathology and treat sequelae can be lifesaving. Arming patients and families with medical knowledge, as provided in the Academy for Eating Disorders’ brochure, “Critical Points for Early Recognition and Medical Risk Management in the Care of Individuals with Eating Disorders”59 can help save patients’ lives.
Eating disorders are debilitating biopsychosocial illnesses associated with serious medical illness and a high risk of death.1
Primary care physicians are often the first to see young women who have these problems, diagnose them, and start their evaluation and treatment.2–4 Many patients require acute medical interventions as well as long-term care for chronic medical issues. Therefore, primary care physicians play essential front-line and long-term roles in the multidisciplinary treatment team.
DEFINITIONS OF EATING DISORDERS HAVE CHANGED
Several problems existed in the category of eating disorders in the fourth edition of the Diagnostic and Statistical Manual of Mental Disorders (DSM-4) and in the DSM-4 Text Revision (DSM-4-TR). These problems have been addressed in the fifth edition (DSM-5), released in 2013.5
One problem in the earlier editions was that many patients referred for treatment of eating disorders—more than 50% in one study6—did not meet the criteria for anorexia nervosa or bulimia nervosa and thus had to be categorized as having “eating disorder not otherwise specified.” Further, the earlier editions did not recognize that young children and adolescent males can be affected.7
Eating disorders are now recognized as an equal-opportunity disease, with all ethnic and socioeconomic groups affected. Children can run into medical trouble with even a small amount of weight loss or falling off the growth curve. Moreover, children and adolescents do not “experience” their bodies in the same way adults do; they may lack the vocabulary for eating-disorder thoughts.
For these reasons, the definitions of eating disorders have changed in the DSM-5.5
Anorexia nervosa. Older editions of the DSM listed amenorrhea as a criterion. This has been eliminated in DSM-5, since amenorrhea does not necessarily predict medical risk or treatment outcome; also, it is not applicable to males or premenorrheal girls and postmenopausal women.8 In addition, the requirement of low weight is now defined in the context of “age, sex, developmental trajectory, and physical health,” rather than the old threshold of 85% of expected weight.9
What remains unchanged is that anorexia nervosa is still characterized by self-starvation in order to maintain an abnormally low body weight, along with an intense fear of being fat and a disturbed self-image.
Bulimia nervosa. In both the old and the new editions of the DSM, bulimia nervosa is characterized by episodes of binge eating followed by inappropriate compensatory behaviors to avoid weight gain, such as vomiting, laxative abuse, diuretic abuse, and overexercise. In DSM-5, bulimia nervosa no longer has subtypes and requires only one binge per week with compensatory behavior, for at least 3 months. This change was based on the finding that there is no clear difference in psychopathology or treatment outcome between patients with one and two binge-purge episodes a week.10
“Eating disorder not otherwise specified” was a wastebasket category, lumping all those who did not meet the criteria for anorexia nervosa or bulimia nervosa or who did not neatly fit into a specific category.10 In DSM-5, subcategories were designed to help distinguish different treatment needs and outcomes between various subtypes.
Binge-eating disorder, one of the new subcategories, is characterized by binge eating without inappropriate compensatory behaviors.9 Patients with binge-eating disorder are often obese, have greater functional impairment, and are more likely to develop components of metabolic syndrome than obese patients without eating disorders.11
Avoidant/restrictive food intake disorder is another new DSM-5 diagnosis, characterized by failure to meet nutritional needs for reasons other than weight control. Reasons include disinterest in eating, dislike of sensory characteristics of food, or avoidance of consequences of eating. This disorder replaces the category “feeding disorder of infancy or early childhood,” since the condition can also occur in adolescents and adults.12
Other new diagnoses are:
- Atypical anorexia nervosa (if the patient is not underweight)
- Purging disorder
- Subthreshold bulimia nervosa (if the patient has < 1 episode per week or has had them for < 3 months)
- Subthreshold binge eating disorder (< 1 time a week or < 3 months)
- Night eating syndrome
- Pica and rumination disorder.
Regardless of the diagnostic label, the medical evaluation and treatment of anyone with an eating disorder should be tailored to the specific behaviors of the eating disorder. Medical complications can be subdivided into those from starvation, from purging, and from refeeding.
MEDICAL COMPLICATIONS OF STARVATION
Cardiovascular effects of starvation
Malnutrition and starvation have multiple adverse effects on the heart.
Electrophysiologic effects. Sinus bradycardia (< 60 bpm) and hypotension are common cardiac manifestations of starvation.13 Bradycardia has been attributed to an adaptive increase in parasympathetic vagal tone.14 QTc prolongation is also seen in patients with malnutrition.15
Together, these electrocardiographic abnormalities predispose the patient to ventricular arrhythmia and sudden cardiac death.16 The risk of ventricular arrhythmia is particularly relevant when treating psychiatric symptoms, since antipsychotics and tricyclic antidepressants are among several drug classes that can cause further QTc prolongation (Table 1).17,18
In patients with QTc prolongation, bradycardia, or both, the standard of care involves acute hospitalization for refeeding using continuous telemetric monitoring until normal rhythm is restored and the heart rate is above 40 at night and 50 by day.4,19
Structural changes. Starvation also causes structural changes in the heart. Loss of lean body mass can reduce cardiac muscle mass, compromise cardiac output, and lead to mitral valve prolapse.20 These changes are fully reversible with restored nutrition and regaining of heart mass.21,22
Effects of starvation on the brain
Starvation can affect brain structure and cognitive function. Undernourished patients have reduced volumes of white and gray matter, a change that can occur within months. Cortical volumes may increase with weight gain, but a reduction in gray matter volume may not be completely reversible.23
Furthermore, starvation impairs cognitive functions that are needed to stop eating-disorder behaviors; namely, decision-making, emotional control, regulation of appetite, and reward path-ways. Therefore, undernourished patients may not have sufficient insight into the disease to be able to make the best choices for recovery. This finding lends support for using the Maudsley method in adolescents, in which parents take control of their child’s eating until the child can maintain a healthy weight.24
Gastrointestinal consequences of starvation
Patients with malnutrition have prolonged gastric emptying and colonic transit time with solid foods.25 They often complain of early satiety, abdominal pain, bloating, and constipation, all symptoms that complicate the refeeding process. A prokinetic such as metoclopramide (Reglan), given 1 hour before meals and at bedtime, may provide some relief from gastrointestinal symptoms.26
Patients may also experience transient lactose or fructose intolerance after prolonged starvation. Taking a lactase supplement (eg, Lactaid 1–10 tabs) before consuming dairy products and dextrose (contained in candies such as Smarties) before eating fruit or fructose-containing foods can sometimes partially relieve symptoms. In general, gastrointestinal function returns over time as nutritional status improves.
Patients with severe or prolonged starvation can develop steatosis accompanied by elevated levels of aspartate aminotransferase (AST) and alanine aminotransferase (ALT). In reports of starvation-induced steatosis, liver enzyme levels rapidly normalize with nutritional rehabilitation.27
Endocrine consequences of starvation
Amenorrhea. Dysregulation of the hypothalamic-pituitary-gonadal axis is a major endocrine complication of nutritional in-sufficiency. Weight loss disrupts the normal pulsatile secretion of gonadotropin-releasing hormone, reduces secretion of luteinizing hormone and follicle-stimulating hormone, and decreases estrogen levels.28 Leptin deficiency likely plays a role in suppressing gonadotropin secretion with subsequent development of amenorrhea. With weight gain, levels of leptin and gonadotropins normalize and menstruation eventually returns.29,30
Hypothyroidism. Starvation can also lead to dysregulation of the hypothalamic-pituitary-thyroid axis. Typically, the concentration of triiodothyronine (T3) is reduced, the ratio of thyroxine (T4) to T3 is elevated, and thyroid-stimulating hormone (TSH) is close to or within the normal range, creating a euthyroid sick syndrome. In eating disorders, this thyroid disturbance is a result of starvation and resolves with weight restoration. Therefore, thyroid hormone replacement therapy is not medically indicated.28
Osteoporosis. Amenorrhea resulting from low estrogen levels in undernourished patients can raise the risk of osteoporosis and fractures, particularly in patients with a low body mass index. Osteopenia results from a negative balance between bone deposition and resorption.
Lack of bone deposition can be especially problematic when disordered eating occurs during peak bone mass development, ie, ages 11 to 14 for girls, and ages 15 to 17 for boys.31,32 Even a 5% to 10% decrease in bone deposition can result in significant risk of osteopenia.33 However, after age 30, bone resorption is a greater contributor.34
Does hormone therapy correct bone loss? Given the association between estrogen deficiency and bone loss, estrogen supplementation was expected to be an effective treatment for bone loss in patients with eating disorders.35 Also, the restoration of menses through hormone replacement may give underweight patients a false sense of achieving a “healthy” weight.36
Golden et al37 prospectively studied 50 adolescents and found no significant difference in bone mineral density at 1 year of follow-up between patients treated with estrogen and those who received only standard nutritional therapy. However, increased bone mineral density was achieved in adolescents with anorexia nervosa treated with transdermally administered estrogen dosed to mimic physiologic pubertal levels.38
Klibanski et al39 found that hormone therapy resulted in a 4% gain in bone density in an extremely low-weight subset of women with anorexia nervosa (< 70% of ideal body weight), whereas similar patients in the control group lost 20%. However, in all groups, only weight gain correlated with bone gain in women who were within 70% of their ideal body weight.
Divasta et al40 evaluated 60 girls and women ages 13 to 27 with anorexia nervosa, randomized to receive either placebo or dehydroepiandrosterone combined with an estrogen-progestin oral contraceptive, and followed for 18 months. As in the study by Klibanski et al,39 bone loss was prevented in the treatment group, but significant bone gain occurred only in the context of weight gain.
The bottom line is that only weight gain has resulted in significant increases in bone density in patients with anorexia nervosa, and hormone therapy without weight gain has not been shown to increase bone density effectively in this population. Although calcium and vitamin D in oral therapeutic doses through foods or through supplementation are required for bone gain, the combination is not enough to augment bone density in the absence of weight gain.37 Although not curative, weight gain is currently the best option for treating bone loss, and no single pharmacologic treatment is effective.
COMPLICATIONS OF PURGING
Oral complications of purging
Patients who purge by vomiting are at risk of complications from exposure of the esophagus, pharynx, and mouth to acidic gastric contents.
Dental problems. Over time, contact with gastric acid wears down enamel on the lingual and occlusal surfaces of teeth, resulting in dental caries and periodontal disease. Until they can give up purging, patients should be instructed to rinse with mouthwash or water immediately after vomiting to reduce the acidity in the mouth.41,42 We recommend that patients not brush their teeth after vomiting, because brushing can deliver acid to otherwise unreachable surfaces and thus worsen tooth erosion. For patients who are determined to brush after vomiting, a bicarbonate toothpaste might mitigate harm.42
Sialadenosis (hypertrophy of the salivary glands) is another consequence of repeated vomiting, with elevated salivary amylase. Both the size of the glands and the salivary amylase level generally normalize on their own after vomiting is stopped, but parotitis can take up to a year to resolve. Similar to smoker’s cough, parotitis may acutely worsen when the patient abruptly stops vomiting and may worsen before it improves.
To reduce discomfort, patients can use hot compresses or sugarless hard candies.44 However, the latter should not be substituted as a chronic habit in a patient with disordered eating. Patients need to be reassured that the swelling is not permanent, since they often interpret it as having fat cheeks (the “chipmunk sign”).
Hypokalemia, metabolic alkalosis, renal dysfunction
Chronic vomiting can cause electrolyte and acid-base imbalances, the most worrisome of which is hypokalemia. With repeated vomiting, loss of potassium and gastric acid causes metabolic alkalosis with hypokalemia, hypochloremia, and hypomagnesemia. Loss of water and the resultant volume contraction activates the renin-angiotensin-aldosterone system, and elevated aldosterone further decreases serum potassium.
In patients with eating disorders, who often have other factors contributing to electrolyte imbalance, vomiting-induced hypokalemia heightens the risk of cardiac arrhythmias.43
Hypokalemia can also cause rhabdomyolysis and kidney damage.41,43 Prolonged hypokalemia and reduced kidney perfusion in the setting of volume depletion causes acute kidney injury and impaired concentrating ability of the renal tubules. Hypovolemia can cause prerenal azotemia and increases the risk for nephrolithiasis and nephrocalcinosis.44,45
When a patient stops vomiting, elevated aldosterone from prior hypovolemia results in water retention and can manifest in significant edema associated with hypochloremic alkalosis. This condition, known as pseudo-Bartter syndrome, usually resolves without treatment. In the meantime, salt restriction and leg elevation can help reduce edema.26
Laxative abuse: A mode of purging
Many patients with eating disorders abuse laxatives to lose weight or to prevent weight gain. Believing that laxatives will prevent calorie absorption, patients commonly take them to compensate for caloric intake (eg, during a binge episode). The immediate weight loss, albeit artificial, is highly reinforcing for an eating-disorder patient. In some cases, patients with eating disorders also abuse laxatives to self-treat the constipation that results from chronic starvation.46
Over time, tolerance to laxatives develops, and patients use increasingly larger doses. This can lead to activation of the renin-angiotensin-aldosterone system.47 Patients interpret the resultant edema as true weight gain and again take laxatives to get rid of it. If laxatives are stopped abruptly, the patient may need inpatient and outpatient support for the resultant fluid shifts.
Gastrointestinal complications of laxative abuse include reflex hypofunction of the bowel, malabsorption, steatorrhea, and gastrointestinal bleeding.47 Reflex hypofunction during laxative withdrawal is a consequence of the bowel becoming tolerant of laxatives.48 Cathartic colon syndrome is a rare complication characterized by loss of the normal haustral markings and slowed or absent peristalsis in segments of the colon.49
Systemically, the major risk of laxative abuse relates to electrolyte and acid-base imbalance. Loss of potassium and water in the stool can cause hypokalemia and metabolic alkalosis.48 The disturbances caused by laxative abuse are similar to those caused by vomiting and diuretic use and have the same treatment.
The most important component of treating laxative abuse is giving patients realistic expectations to help them tolerate temporary discomfort and to help manage the edema and fluid shifts that can happen acutely with shifting of fluid into the intracellular space. In extreme cases, this may need to be managed in the hospital. To help relieve the initial anxiety, doctors should emphasize that any bloating the patient experiences is not true weight gain and will go away within a few days to weeks. In addition, explaining that laxatives reduce nutrient absorption only minimally may lessen the temptation to resume taking them.48
Diuretic abuse: Another form of purging
Diuretic abuse is yet another mode of purging, with its own set of medical complications. Like laxatives, diuretics are not effective weight-loss agents, and the weight reduction they cause is only temporary.
As with vomiting, there is a compensatory activation of the renin-angiotensin-aldosterone system, and therefore subsequent fluid intake will lead to water retention, which encourages further diuretic use.41 Diuretics can also contribute to hypokalemia, hypomagnesemia, hypochloremia, and metabolic alkalosis.
Ipecac abuse can lead to heart failure
Ipecac syrup has long been used to induce vomiting, but this practice has become much less common since ipecac has become harder to obtain in the United States.50 The emetine base contained in ipecac binds irreversibly to cardiac and skeletal muscle. With continued use, irreversible cardiomyopathy develops and can lead to heart failure. Treatment should include supportive care and immediate cessation of ipecac use.
Diabetic patients may skip insulin to lose weight
Patients with diabetes, especially those with type 1 that begins in childhood, are at greater risk of eating disorders over time.51 They may withhold insulin to lose weight, a practice referred to in the nonmedical literature as “diabulimia,” and they seem particularly more likely to develop bulimia nervosa than those without diabetes.52
The medical prognosis is poor for patients with diabetes who develop eating disorders and do not receive intensive treatment.51 In addition, if a diabetic patient on an insulin pump becomes depressed in addition to having an eating disorder, careful monitoring for suicidal thoughts and a rapid follow-up with mental health services are in order.
REFEEDING SYNDROME
When refeeding is started, a high glucose load stimulates insulin secretion, resulting in cellular uptake of phosphorus along with potassium, magnesium, and glucose. In addition, total body phosphorus is depleted by the increased demand for adenosine triphosphate and 2,3-diphosphoglycerate for cellular metabolism.
When liver enzyme levels increase, the astute clinician will closely monitor the patient for evidence of refeeding syndrome. In a child, adolescent, or young adult, the standard of care is inpatient monitoring for acute stabilization.4,19
Hypophosphatemia is the hallmark of refeeding syndrome, although hypomagnesemia, hypokalemia, and hypoglycemia can also occur.53 In addition, sodium and water retention can lead to fluid overload, with shifting of fluid into the intracellular space, resulting in dependent edema.
Cardiovascular complications are the most worrisome manifestations of refeeding syndrome. Electrolyte shifts and increased fluid volume can cause arrhythmias and heart failure. Furthermore, severely undernourished patients may have reduced myocardial mass as well as electrocardiographic abnormalities associated with starvation, which further increase their vulnerability to electrolyte shifts and fluid retention during refeeding.15
Other manifestations of refeeding syndrome include delirium, seizures, rhabdomyolysis, and respiratory failure. In the most extreme cases, refeeding syndrome causes sudden death.53
Fortunately, refeeding syndrome is easily preventable and treatable when recognized early. Electrolytes and cardiovascular and renal function must be carefully monitored, especially during the first week of nutritional restoration.53 In patients with extremely low body mass (< 70% of ideal body weight) or with precipitous weight loss, close monitoring of the complete metabolic panel including electrolytes, AST, ALT, calcium, magnesium, and phosphorus may be required to detect changes that can affect cardiac status. Specific suggestions for refeeding are discussed below and in Table 2.45
ACUTE CARE OF PATIENTS WITH EATING DISORDERS
Refeeding in the inpatient setting
The decision to hospitalize an eating-disorder patient is based on the current or potential risk of serious medical complications and the likelihood of success at home. Medical criteria for hospital admission are outlined in Table 3.4,54
In refeeding undernourished patients, the challenge is to maximize weight gain while preventing refeeding syndrome. Undernourished patients are generally hypometabolic at baseline but become hypermetabolic once refeeding begins.
How many calories should refeeding start with? The traditional principle of “start low and go slow” has been recently challenged.55 Starting at 1,200 kcal/day or less in the typical patient can result in failure to gain weight or even in weight loss in the first week of refeeding.56 The goal is to achieve a weight gain of 0.2 kg/day while the patient is in the hospital. Thus, we start higher, and to date we have seen no cases of life-threatening refeeding syndrome. In all patients who need hospitalization or who are beginning the refeeding process as outpatients, caloric intake should be started at 1,500 to 2,000 kcal/day.45,57 However, for exceptionally low-weight patients, intake may be started lower.
In Australia, patients are started at 1,900 kcal/day.56 All patients in one program there receive nasogastric feeding initially in an intensive care unit and then are moved to a regular nursing floor where they graduate to full oral feeding as they improve cardiovascularly and behaviorally. In the United States, some programs use nasogastric feeding at night for caloric restoration; our program and others use nasogastric feeding as a behavioral modification strategy for patients who refuse food or supplements by mouth.
Phosphorus supplementation. Many centers give phosphorus supplements preventively. In our center, we give potassium phosphate (Neutra-Phos) 500 mg orally twice daily for 5 days, and we have seen no life-threatening cases of refeeding syndrome with that regimen. Other centers give phosphorus supplements in a dose of 250 mg orally twice a day for 5 days, while still others only supplement phosphorus reactively once a deficit has been identified. The latter method requires daily blood draws for monitoring and is reactive rather than proactive. Further studies can help clarify the optimal dosing and timing of phosphorus supplementation.
Managing fluid balance. Fluid-loading these patients may tip them over the edge into refeeding syndrome. Except in cases of shock, patients with eating disorders should not be given intravenous fluids, as it is safer to rehydrate and feed them orally. Electrolyte imbalances can be corrected orally with no need for intravenous supplementation. To avoid fluid overload, fluids can be started at 1,500 mL to 2,000 mL per day, with strict monitoring of intake and output. Fluids are liberalized if ALT and AST levels remain normal and to gradually correct orthostatic hypotension; caloric fluids are ideal to help address energy needs and improve bradycardia.
Laboratory monitoring. On admission, a urinalysis, complete blood cell count, complete metabolic panel, TSH, erythrocyte sedimentation rate, serum magnesium, and phosphorus should be obtained.26 In addition, continuous electrocardiographic recording should begin on admission.45 Inpatient use of a telemetry bed helps identify extreme tachycardia with arrhythmia, as well as profound bradycardia.45,56
Some protocols call for daily laboratory monitoring, although that degree of testing is less cost-effective. If initial results are normal, clinical judgment can be used on when to repeat laboratory evaluation. For instance, patients with edema require repeat complete metabolic panels to assess for elevated ALT and AST, electrolyte imbalances, and other abnormalities.
Signs of refeeding syndrome include tachycardia, hepatosplenomegaly, peripheral edema, altered mental status, and electrolyte disturbances, specifically, acute or severe hypophosphatemia or hypokalemia.26,45 If refeeding syndrome is suspected, the rate of caloric intake should be reduced or not advanced, fluid intake should be urgently reassessed for volume overload, and supportive care with close monitoring should be provided.
KNOWLEDGE SAVES LIVES
Eating disorders can lead to potentially life-threatening medical complications that require attentive care by the primary care clinician and subspecialist. Without thoughtful consideration, it is easy for even a caring medical team to unintentionally enable patients with these illnesses or to cause active harm in the case of underrecognized pathology.58
Acute medical stabilization on an inpatient unit trained to recognize pathology and treat sequelae can be lifesaving. Arming patients and families with medical knowledge, as provided in the Academy for Eating Disorders’ brochure, “Critical Points for Early Recognition and Medical Risk Management in the Care of Individuals with Eating Disorders”59 can help save patients’ lives.
- Arcelus J, Mitchell AJ, Wales J, Nielsen S. Mortality rates in patients with anorexia nervosa and other eating disorders. A meta-analysis of 36 studies. Arch Gen Psychiatry 2011; 68:724–731.
- Walsh JM, Wheat ME, Freund K. Detection, evaluation, and treatment of eating disorders the role of the primary care physician. J Gen Intern Med 2000; 15:577–590.
- American Academy of Pediatrics; Committee on Adolescence. Identifying and treating eating disorders. Pediatrics 2003; 111:204–211.
- Rosen DS; American Academy of Pediatrics Committee on Adolescence. Identification and management of eating disorders in children and adolescents. Pediatrics 2010; 126:1240–1253.
- American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders. 5th edition. Arlington, VA: American Psychiatric Publishing, Incorporated; 2013.
- Eddy KT, Celio Doyle A, Hoste RR, Herzog DB, le Grange D. Eating disorder not otherwise specified in adolescents. J Am Acad Child Adolesc Psychiatry 2008; 47:156–164.
- Muise AM, Stein DG, Arbess G. Eating disorders in adolescent boys: a review of the adolescent and young adult literature. J Adolesc Health 2003; 33:427–435.
- Attia E, Roberto CA. Should amenorrhea be a diagnostic criterion for anorexia nervosa? Int J Eat Disord 2009; 42:581–589.
- American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders, fifth edition. http://dsm.psychiatryonline.org/content.aspx?bookid=556§ionid=41101776#103439089. Accessed January 31, 2014.
- Wilfley DE, Bishop ME, Wilson GT, Agras WS. Classification of eating disorders: toward DSM-V. Int J Eat Disord 2007; 40:S123–S129.
- Wonderlich SA, Gordon KH, Mitchell JE, Crosby RD, Engel SG. The validity and clinical utility of binge eating disorder. Int J Eat Disord 2009; 42:687–705.
- Ornstein RM, Rosen DS, Mammel KA, et al. Distribution of eating disorders in children and adolescents using the proposed DSM-5 criteria for feeding and eating disorders. J Adolesc Health 2013: 53:303–305.
- Winston AP, Stafford PJ. Cardiovascular effects of anorexia nervosa. Eur Eat Disord Rev 2000; 8:117–125.
- Galetta F, Franzoni F, Prattichizzo F, Rolla M, Santoro G, Pentimone F. Heart rate variability and left ventricular diastolic function in anorexia nervosa. J Adolesc Health 2003; 32:416–421.
- McCallum K, Bermudez O, Ohlemeyer C, Tyson E, Portilla M, Ferdman B. How should the clinician evaluate and manage the cardiovascular complications of anorexia nervosa? Eat Disord 2006; 14:73–80.
- Akhtar M. Clinical spectrum of ventricular tachycardia. Circulation 1990; 82:1561–1573.
- Beach SR, Celano CM, Noseworthy PA, Januzzi JL, Huffman JC. QTc prolongation, torsades de pointes, and psychotropic medications. Psychosomatics 2013; 54:1–13.
- The University of Arizona Center for Education and Research on Therapeutics. QT Drug Lists. http://crediblemeds.org/everyone/compos-ite-list-all-qtdrugs/?rf=US. Accessed January 31, 2014.
- Rome ES, Ammerman S. Medical complications of eating disorders: an update. J Adolesc Health 2003; 33:418–426.
- Romano C, Chinali M, Pasanisi F, et al. Reduced hemodynamic load and cardiac hypotrophy in patients with anorexia nervosa. Am J Clin Nutr 2003; 77:308–312.
- Shamim T, Golden NH, Arden M, Filiberto L, Shenker IR. Resolution of vital sign instability: an objective measure of medical stability in anorexia nervosa. J Adolesc Health 2003; 32:73–77.
- Mont L, Castro J, Herreros B, et al. Reversibility of cardiac abnormalities in adolescents with anorexia nervosa after weight recovery. J Am Acad Child Adolesc Psychiatry 2003; 42:808–813.
- Roberto CA, Mayer LE, Brickman AM, et al. Brain tissue volume changes following weight gain in adults with anorexia nervosa. Int J Eat Disord 2011; 44:406–411.
- Treasure J, Russell G. The case for early intervention in anorexia nervosa: theoretical exploration of maintaining factors. Br J Psychiatry 2011; 199:5–7.
- Hadley SJ, Walsh BT. Gastrointestinal disturbances in anorexia nervosa and bulimia nervosa. Curr Drug Targets CNS Neurol Disord 2003; 2:1–9.
- Yager J, Andersen AE. Clinical practice. Anorexia nervosa. N Engl J Med 2005; 353:1481–1488.
- De Caprio C, Alfano A, Senatore I, Zarrella L, Pasanisi F, Contaldo F. Severe acute liver damage in anorexia nervosa: two case reports. Nutrition 2006; 22:572–575.
- Lawson EA, Klibanski A. Endocrine abnormalities in anorexia nervosa. Nat Clin Pract Endocrinol Metab 2008; 4:407–414.
- Holtkamp K, Mika C, Grzella I, et al. Reproductive function during weight gain in anorexia nervosa. Leptin represents a metabolic gate to gonadotropin secretion. J Neural Transm 2003; 110:427–435.
- Golden NH, Jacobson MS, Schebendach J, Solanto MV, Hertz SM, Shenker IR. Resumption of menses in anorexia nervosa. Arch Pediatr Adolesc Med 1997; 151:16–21.
- Soyka LA, Misra M, Frenchman A, et al. Abnormal bone mineral accrual in adolescent girls with anorexia nervosa. J Clin Endocrinol Metab 2002; 87:4177–4185.
- Misra M, Klibanski A. Bone metabolism in adolescents with anorexia nervosa. J Endocrinol Invest 2011; 34:324–332.
- Recker RR, Davies KM, Hinders SM, Heaney RP, Stegman MR, Kimmel DB. Bone gain in young adult women. JAMA 1992; 268:2403–2408.
- Biller BM, Saxe V, Herzog DB, Rosenthal DI, Holzman S, Klibanski A. Mechanisms of osteoporosis in adult and adolescent women with anorexia nervosa. J Clin Endocrinol Metab 1989; 68:548–554.
- Hergenroeder AC, Smith EO, Shypailo R, Jones LA, Klish WJ, Ellis K. Bone mineral changes in young women with hypothalamic amenorrhea treated with oral contraceptives, medroxyprogesterone, or placebo over 12 months. Am J Obstet Gynecol 1997; 176:1017–1025.
- Sim LA, McGovern L, Elamin MB, Swiglo BA, Erwin PJ, Montori VM. Effect on bone health of estrogen preparations in premenopausal women with anorexia nervosa: a systematic review and meta-analyses. Int J Eat Disord 2010; 43:218–225.
- Golden NH, Lanzkowsky L, Schebendach J, Palestro CJ, Jacobson MS, Shenker IR. The effect of estrogen-progestin treatment on bone mineral density in anorexia nervosa. J Pediatr Adolesc Gynecol 2002; 15:135–143.
- Misra M, Katzman D, Miller KK, et al. Physiologic estrogen replacement increases bone density in adolescent girls with anorexia nervosa. J Bone Miner Res 2011; 26:2430–2438.
- Klibanski A, Biller BM, Schoenfeld DA, Herzog DB, Saxe VC. The effects of estrogen administration on trabecular bone loss in young women with anorexia nervosa. J Clin Endocrinol Metab 1995; 80:898–904.
- Divasta AD, Feldman HA, Giancaterino C, Rosen CJ, Leboff MS, Gordon CM. The effect of gonadal and adrenal steroid therapy on skeletal health in adolescents and young women with anorexia nervosa. Metabolism 2012; 61:1010–1020.
- Mehler PS. Medical complications of bulimia nervosa and their treatments. Int J Eat Disord 2011; 44:95–104.
- Milosevic A. Eating disorders and the dentist. Br Dent J 1999; 186:109–113.
- Greenfeld D, Mickley D, Quinlan DM, Roloff P. Hypokalemia in outpatients with eating disorders. Am J Psychiatry 1995; 152:60–63.
- Bouquegneau A, Dubois BE, Krzesinski JM, Delanaye P. Anorexia nervosa and the kidney. Am J Kidney Dis 2012; 60:299–307.
- Auron M, Rome E. Anorexia nervosa and bulimia nervosa: what the hospitalist needs to know about CPT 269.9, or nutritional insufficiency. ACP Hospitalist 2011 Sept:28–45.
- Steffen KJ, Mitchell JE, Roerig JL, Lancaster KL. The eating disorders medicine cabinet revisited: a clinician’s guide to ipecac and laxatives. Int J Eat Disord 2007; 40:360–368.
- Roerig JL, Steffen KJ, Mitchell JE, Zunker C. Laxative abuse: epidemiology, diagnosis and management. Drugs 2010; 70:1487–1503.
- Mitchell JE, Boutacoff LI. Laxative abuse complicating bulimia: medical and treatment implications. Int J Eat Disord 1986; 5:325–334.
- Joo JS, Ehrenpreis ED, Gonzalez L, et al. Alterations in colonic anatomy induced by chronic stimulant laxatives: the cathartic colon revisited. J Clin Gastroenterol 1998; 26:283–286.
- Drugs.com. Ipecac syrup. www.drugs.com/monograph/ipecac-syrup.html. Accessed January 31, 2014.
- Peveler RC, Bryden KS, Neil HA, et al. The relationship of disordered eating habits and attitudes to clinical outcomes in young adult females with type 1 diabetes. Diabetes Care 2005; 28:84–88.
- Mannucci E, Rotella F, Ricca V, Moretti S, Placidi GF, Rotella CM. Eating disorders in patients with type 1 diabetes: a meta-analysis. J Endocrinol Invest 2005; 28:417–419.
- Crook MA, Hally V, Panteli JV. The importance of the refeeding syndrome. Nutrition 2001; 17:632–637.
- Fisher M, Golden NH, Katzman DK, et al. Eating disorders in adolescents: a background paper. J Adolesc Health 1995; 16:420–437.
- Kohn MR, Madden S, Clarke SD. Refeeding in anorexia nervosa: increased safety and efficiency through understanding the pathophysiology of protein calorie malnutrition. Curr Opin Pediatr 2011; 23:390–394.
- Garber AK, Michihata N, Hetnal K, Shafer MA, Moscicki AB. A prospective examination of weight gain in hospitalized adolescents with anorexia nervosa on a recommended refeeding protocol. J Adolesc Health 2012; 50:24–29.
- Whitelaw M, Gilbertson H, Lam PY, Sawyer SM. Does aggressive refeeding in hospitalized adolescents with anorexia nervosa result in increased hypophosphatemia? J Adolesc Health 2010; 46:577–582.
- Treasure J, Crane A, McKnight R, Buchanan E, Wolfe M. First do no harm: iatrogenic maintaining factors in anorexia nervosa. Eur Eat Disord Rev 2011; 19:296–302.
- Academy for Eating Disorders (AED). Critical points for early recognition and medical risk management in the care of individuals with eating disorders. http://www.aedweb.org/AM/Template.cfm?Section=Medical_Care_Standards&Template=/CM/ContentDisplay.cfm&ContentID=2413. Accessed January 31, 2014.
- Arcelus J, Mitchell AJ, Wales J, Nielsen S. Mortality rates in patients with anorexia nervosa and other eating disorders. A meta-analysis of 36 studies. Arch Gen Psychiatry 2011; 68:724–731.
- Walsh JM, Wheat ME, Freund K. Detection, evaluation, and treatment of eating disorders the role of the primary care physician. J Gen Intern Med 2000; 15:577–590.
- American Academy of Pediatrics; Committee on Adolescence. Identifying and treating eating disorders. Pediatrics 2003; 111:204–211.
- Rosen DS; American Academy of Pediatrics Committee on Adolescence. Identification and management of eating disorders in children and adolescents. Pediatrics 2010; 126:1240–1253.
- American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders. 5th edition. Arlington, VA: American Psychiatric Publishing, Incorporated; 2013.
- Eddy KT, Celio Doyle A, Hoste RR, Herzog DB, le Grange D. Eating disorder not otherwise specified in adolescents. J Am Acad Child Adolesc Psychiatry 2008; 47:156–164.
- Muise AM, Stein DG, Arbess G. Eating disorders in adolescent boys: a review of the adolescent and young adult literature. J Adolesc Health 2003; 33:427–435.
- Attia E, Roberto CA. Should amenorrhea be a diagnostic criterion for anorexia nervosa? Int J Eat Disord 2009; 42:581–589.
- American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders, fifth edition. http://dsm.psychiatryonline.org/content.aspx?bookid=556§ionid=41101776#103439089. Accessed January 31, 2014.
- Wilfley DE, Bishop ME, Wilson GT, Agras WS. Classification of eating disorders: toward DSM-V. Int J Eat Disord 2007; 40:S123–S129.
- Wonderlich SA, Gordon KH, Mitchell JE, Crosby RD, Engel SG. The validity and clinical utility of binge eating disorder. Int J Eat Disord 2009; 42:687–705.
- Ornstein RM, Rosen DS, Mammel KA, et al. Distribution of eating disorders in children and adolescents using the proposed DSM-5 criteria for feeding and eating disorders. J Adolesc Health 2013: 53:303–305.
- Winston AP, Stafford PJ. Cardiovascular effects of anorexia nervosa. Eur Eat Disord Rev 2000; 8:117–125.
- Galetta F, Franzoni F, Prattichizzo F, Rolla M, Santoro G, Pentimone F. Heart rate variability and left ventricular diastolic function in anorexia nervosa. J Adolesc Health 2003; 32:416–421.
- McCallum K, Bermudez O, Ohlemeyer C, Tyson E, Portilla M, Ferdman B. How should the clinician evaluate and manage the cardiovascular complications of anorexia nervosa? Eat Disord 2006; 14:73–80.
- Akhtar M. Clinical spectrum of ventricular tachycardia. Circulation 1990; 82:1561–1573.
- Beach SR, Celano CM, Noseworthy PA, Januzzi JL, Huffman JC. QTc prolongation, torsades de pointes, and psychotropic medications. Psychosomatics 2013; 54:1–13.
- The University of Arizona Center for Education and Research on Therapeutics. QT Drug Lists. http://crediblemeds.org/everyone/compos-ite-list-all-qtdrugs/?rf=US. Accessed January 31, 2014.
- Rome ES, Ammerman S. Medical complications of eating disorders: an update. J Adolesc Health 2003; 33:418–426.
- Romano C, Chinali M, Pasanisi F, et al. Reduced hemodynamic load and cardiac hypotrophy in patients with anorexia nervosa. Am J Clin Nutr 2003; 77:308–312.
- Shamim T, Golden NH, Arden M, Filiberto L, Shenker IR. Resolution of vital sign instability: an objective measure of medical stability in anorexia nervosa. J Adolesc Health 2003; 32:73–77.
- Mont L, Castro J, Herreros B, et al. Reversibility of cardiac abnormalities in adolescents with anorexia nervosa after weight recovery. J Am Acad Child Adolesc Psychiatry 2003; 42:808–813.
- Roberto CA, Mayer LE, Brickman AM, et al. Brain tissue volume changes following weight gain in adults with anorexia nervosa. Int J Eat Disord 2011; 44:406–411.
- Treasure J, Russell G. The case for early intervention in anorexia nervosa: theoretical exploration of maintaining factors. Br J Psychiatry 2011; 199:5–7.
- Hadley SJ, Walsh BT. Gastrointestinal disturbances in anorexia nervosa and bulimia nervosa. Curr Drug Targets CNS Neurol Disord 2003; 2:1–9.
- Yager J, Andersen AE. Clinical practice. Anorexia nervosa. N Engl J Med 2005; 353:1481–1488.
- De Caprio C, Alfano A, Senatore I, Zarrella L, Pasanisi F, Contaldo F. Severe acute liver damage in anorexia nervosa: two case reports. Nutrition 2006; 22:572–575.
- Lawson EA, Klibanski A. Endocrine abnormalities in anorexia nervosa. Nat Clin Pract Endocrinol Metab 2008; 4:407–414.
- Holtkamp K, Mika C, Grzella I, et al. Reproductive function during weight gain in anorexia nervosa. Leptin represents a metabolic gate to gonadotropin secretion. J Neural Transm 2003; 110:427–435.
- Golden NH, Jacobson MS, Schebendach J, Solanto MV, Hertz SM, Shenker IR. Resumption of menses in anorexia nervosa. Arch Pediatr Adolesc Med 1997; 151:16–21.
- Soyka LA, Misra M, Frenchman A, et al. Abnormal bone mineral accrual in adolescent girls with anorexia nervosa. J Clin Endocrinol Metab 2002; 87:4177–4185.
- Misra M, Klibanski A. Bone metabolism in adolescents with anorexia nervosa. J Endocrinol Invest 2011; 34:324–332.
- Recker RR, Davies KM, Hinders SM, Heaney RP, Stegman MR, Kimmel DB. Bone gain in young adult women. JAMA 1992; 268:2403–2408.
- Biller BM, Saxe V, Herzog DB, Rosenthal DI, Holzman S, Klibanski A. Mechanisms of osteoporosis in adult and adolescent women with anorexia nervosa. J Clin Endocrinol Metab 1989; 68:548–554.
- Hergenroeder AC, Smith EO, Shypailo R, Jones LA, Klish WJ, Ellis K. Bone mineral changes in young women with hypothalamic amenorrhea treated with oral contraceptives, medroxyprogesterone, or placebo over 12 months. Am J Obstet Gynecol 1997; 176:1017–1025.
- Sim LA, McGovern L, Elamin MB, Swiglo BA, Erwin PJ, Montori VM. Effect on bone health of estrogen preparations in premenopausal women with anorexia nervosa: a systematic review and meta-analyses. Int J Eat Disord 2010; 43:218–225.
- Golden NH, Lanzkowsky L, Schebendach J, Palestro CJ, Jacobson MS, Shenker IR. The effect of estrogen-progestin treatment on bone mineral density in anorexia nervosa. J Pediatr Adolesc Gynecol 2002; 15:135–143.
- Misra M, Katzman D, Miller KK, et al. Physiologic estrogen replacement increases bone density in adolescent girls with anorexia nervosa. J Bone Miner Res 2011; 26:2430–2438.
- Klibanski A, Biller BM, Schoenfeld DA, Herzog DB, Saxe VC. The effects of estrogen administration on trabecular bone loss in young women with anorexia nervosa. J Clin Endocrinol Metab 1995; 80:898–904.
- Divasta AD, Feldman HA, Giancaterino C, Rosen CJ, Leboff MS, Gordon CM. The effect of gonadal and adrenal steroid therapy on skeletal health in adolescents and young women with anorexia nervosa. Metabolism 2012; 61:1010–1020.
- Mehler PS. Medical complications of bulimia nervosa and their treatments. Int J Eat Disord 2011; 44:95–104.
- Milosevic A. Eating disorders and the dentist. Br Dent J 1999; 186:109–113.
- Greenfeld D, Mickley D, Quinlan DM, Roloff P. Hypokalemia in outpatients with eating disorders. Am J Psychiatry 1995; 152:60–63.
- Bouquegneau A, Dubois BE, Krzesinski JM, Delanaye P. Anorexia nervosa and the kidney. Am J Kidney Dis 2012; 60:299–307.
- Auron M, Rome E. Anorexia nervosa and bulimia nervosa: what the hospitalist needs to know about CPT 269.9, or nutritional insufficiency. ACP Hospitalist 2011 Sept:28–45.
- Steffen KJ, Mitchell JE, Roerig JL, Lancaster KL. The eating disorders medicine cabinet revisited: a clinician’s guide to ipecac and laxatives. Int J Eat Disord 2007; 40:360–368.
- Roerig JL, Steffen KJ, Mitchell JE, Zunker C. Laxative abuse: epidemiology, diagnosis and management. Drugs 2010; 70:1487–1503.
- Mitchell JE, Boutacoff LI. Laxative abuse complicating bulimia: medical and treatment implications. Int J Eat Disord 1986; 5:325–334.
- Joo JS, Ehrenpreis ED, Gonzalez L, et al. Alterations in colonic anatomy induced by chronic stimulant laxatives: the cathartic colon revisited. J Clin Gastroenterol 1998; 26:283–286.
- Drugs.com. Ipecac syrup. www.drugs.com/monograph/ipecac-syrup.html. Accessed January 31, 2014.
- Peveler RC, Bryden KS, Neil HA, et al. The relationship of disordered eating habits and attitudes to clinical outcomes in young adult females with type 1 diabetes. Diabetes Care 2005; 28:84–88.
- Mannucci E, Rotella F, Ricca V, Moretti S, Placidi GF, Rotella CM. Eating disorders in patients with type 1 diabetes: a meta-analysis. J Endocrinol Invest 2005; 28:417–419.
- Crook MA, Hally V, Panteli JV. The importance of the refeeding syndrome. Nutrition 2001; 17:632–637.
- Fisher M, Golden NH, Katzman DK, et al. Eating disorders in adolescents: a background paper. J Adolesc Health 1995; 16:420–437.
- Kohn MR, Madden S, Clarke SD. Refeeding in anorexia nervosa: increased safety and efficiency through understanding the pathophysiology of protein calorie malnutrition. Curr Opin Pediatr 2011; 23:390–394.
- Garber AK, Michihata N, Hetnal K, Shafer MA, Moscicki AB. A prospective examination of weight gain in hospitalized adolescents with anorexia nervosa on a recommended refeeding protocol. J Adolesc Health 2012; 50:24–29.
- Whitelaw M, Gilbertson H, Lam PY, Sawyer SM. Does aggressive refeeding in hospitalized adolescents with anorexia nervosa result in increased hypophosphatemia? J Adolesc Health 2010; 46:577–582.
- Treasure J, Crane A, McKnight R, Buchanan E, Wolfe M. First do no harm: iatrogenic maintaining factors in anorexia nervosa. Eur Eat Disord Rev 2011; 19:296–302.
- Academy for Eating Disorders (AED). Critical points for early recognition and medical risk management in the care of individuals with eating disorders. http://www.aedweb.org/AM/Template.cfm?Section=Medical_Care_Standards&Template=/CM/ContentDisplay.cfm&ContentID=2413. Accessed January 31, 2014.
KEY POINTS
- The fifth edition of the Diagnostic and Statistical Manual of Mental Disorders (DSM-5), released in 2013, has updated the criteria for some eating disorders and has added some new disorders.
- Starvation can cause cardiac, cerebral, gastrointestinal, and endocrine problems.
- Purging can lead to problems with oral health, electrolyte imbalances, and even renal failure.
- Refeeding poses the risk of refeeding syndrome, with fluid overload and electrolyte imbalances. Many patients undergoing refeeding are best managed in the hospital.
HM14 Special Report: Setting up Your QI Project for Success: Organizational Imperatives, Data Collection, Implementation Strategy
Quality improvement (QI) is about system change for the entire organization. In a session at HM14, Michelle Mourad, MD, and Nasim Afsar, MD, SFHM, outlined QI principles and a systematic step-wise process that can create and sustain change successfully in our hospitals.
QI principles include creating a sense of urgency, implementation, and sustainability. The seven steps for a successful QI project are as follows: understand the problem, convince others there is a problem, identify areas of improvement, prioritize a small test of change, devise a measurement strategy, measure change, and sustain change.
Understanding the problem includes creating a fishbone diagram that helps you identify the causes of your problem. Convincing others requires presentation of inspiring data and telling your story.
Dr. Mourad encouraged the audience to use personal anecdotes and focus on the ‘why,’ not the ‘what’ or ‘how’ to relate to others on a personal level. Identifying areas of improvement requires creation of a process map to identify obstacles that can be removed. “Your goal is to make systems work for people,” said Dr. Mourad. Ask people to tell you how the process will work better for them.
Prioritizing small tests of change requires identifying low-effort, high-impact tasks that will lead to easy wins. “Don’t do the thankless tasks that are high-effort with low impact,” said Dr. Afsar. She further encouraged physicians to perform small tests of change to help understand if their ideas are worthy of large scale implementation.
Plan-Do-Study-Act (PDSA) and A3 methodology are two methods by which a QI project can be organized for implementation. Devising a measurement strategy requires collecting data from appropriate sources in the hospital to ensure improved outcomes as a result of interventions performed. Outcomes, structure, and process are the areas in which results can be measured.
Coaching your team is an important part of motivating your team for continued progress. Change can be concretely measured and plotted over time via statistical process control charts (download SPC macros in Excel to plot graphs).
Lastly, sustaining change requires celebrating success. Dr. Afsar cautioned the audience to set appropriate expectations and goals to sustain system change. Further, once the change is set in motion, put the process ownership into the group (not on you). Creating data that is readily accessible and visible to the group helps them understand progress over time. Design the process to fit into natural workflow to make the process an easy transition that is sustainable long-term.
Key Points
• QI is a four legged stool: education, data audit and feedback, systems change, and culture change;
• Create a fishbone diagram to understand the cause and the effect;
• Create a sense of urgency by sharing inspirational data and finding your story and focusing on the why;
• Use process maps to identify areas of improvement;
• Prioritize those areas and implement small tests of change;
• Measure all outcomes and use statistical process control charts to demonstrate change;
• Motivate and coach people throughout the process change;
• Make data easily accessible to all members of the group to track progress; and
• Sustain change by setting realistic expectations and celebrating success.
Dr. Kanikkannan is Hospitalist Medical Director and Assistant Professor of Medicine at Rowan University School of Osteopathic Medicine and is a member of Team Hospitalist.
Quality improvement (QI) is about system change for the entire organization. In a session at HM14, Michelle Mourad, MD, and Nasim Afsar, MD, SFHM, outlined QI principles and a systematic step-wise process that can create and sustain change successfully in our hospitals.
QI principles include creating a sense of urgency, implementation, and sustainability. The seven steps for a successful QI project are as follows: understand the problem, convince others there is a problem, identify areas of improvement, prioritize a small test of change, devise a measurement strategy, measure change, and sustain change.
Understanding the problem includes creating a fishbone diagram that helps you identify the causes of your problem. Convincing others requires presentation of inspiring data and telling your story.
Dr. Mourad encouraged the audience to use personal anecdotes and focus on the ‘why,’ not the ‘what’ or ‘how’ to relate to others on a personal level. Identifying areas of improvement requires creation of a process map to identify obstacles that can be removed. “Your goal is to make systems work for people,” said Dr. Mourad. Ask people to tell you how the process will work better for them.
Prioritizing small tests of change requires identifying low-effort, high-impact tasks that will lead to easy wins. “Don’t do the thankless tasks that are high-effort with low impact,” said Dr. Afsar. She further encouraged physicians to perform small tests of change to help understand if their ideas are worthy of large scale implementation.
Plan-Do-Study-Act (PDSA) and A3 methodology are two methods by which a QI project can be organized for implementation. Devising a measurement strategy requires collecting data from appropriate sources in the hospital to ensure improved outcomes as a result of interventions performed. Outcomes, structure, and process are the areas in which results can be measured.
Coaching your team is an important part of motivating your team for continued progress. Change can be concretely measured and plotted over time via statistical process control charts (download SPC macros in Excel to plot graphs).
Lastly, sustaining change requires celebrating success. Dr. Afsar cautioned the audience to set appropriate expectations and goals to sustain system change. Further, once the change is set in motion, put the process ownership into the group (not on you). Creating data that is readily accessible and visible to the group helps them understand progress over time. Design the process to fit into natural workflow to make the process an easy transition that is sustainable long-term.
Key Points
• QI is a four legged stool: education, data audit and feedback, systems change, and culture change;
• Create a fishbone diagram to understand the cause and the effect;
• Create a sense of urgency by sharing inspirational data and finding your story and focusing on the why;
• Use process maps to identify areas of improvement;
• Prioritize those areas and implement small tests of change;
• Measure all outcomes and use statistical process control charts to demonstrate change;
• Motivate and coach people throughout the process change;
• Make data easily accessible to all members of the group to track progress; and
• Sustain change by setting realistic expectations and celebrating success.
Dr. Kanikkannan is Hospitalist Medical Director and Assistant Professor of Medicine at Rowan University School of Osteopathic Medicine and is a member of Team Hospitalist.
Quality improvement (QI) is about system change for the entire organization. In a session at HM14, Michelle Mourad, MD, and Nasim Afsar, MD, SFHM, outlined QI principles and a systematic step-wise process that can create and sustain change successfully in our hospitals.
QI principles include creating a sense of urgency, implementation, and sustainability. The seven steps for a successful QI project are as follows: understand the problem, convince others there is a problem, identify areas of improvement, prioritize a small test of change, devise a measurement strategy, measure change, and sustain change.
Understanding the problem includes creating a fishbone diagram that helps you identify the causes of your problem. Convincing others requires presentation of inspiring data and telling your story.
Dr. Mourad encouraged the audience to use personal anecdotes and focus on the ‘why,’ not the ‘what’ or ‘how’ to relate to others on a personal level. Identifying areas of improvement requires creation of a process map to identify obstacles that can be removed. “Your goal is to make systems work for people,” said Dr. Mourad. Ask people to tell you how the process will work better for them.
Prioritizing small tests of change requires identifying low-effort, high-impact tasks that will lead to easy wins. “Don’t do the thankless tasks that are high-effort with low impact,” said Dr. Afsar. She further encouraged physicians to perform small tests of change to help understand if their ideas are worthy of large scale implementation.
Plan-Do-Study-Act (PDSA) and A3 methodology are two methods by which a QI project can be organized for implementation. Devising a measurement strategy requires collecting data from appropriate sources in the hospital to ensure improved outcomes as a result of interventions performed. Outcomes, structure, and process are the areas in which results can be measured.
Coaching your team is an important part of motivating your team for continued progress. Change can be concretely measured and plotted over time via statistical process control charts (download SPC macros in Excel to plot graphs).
Lastly, sustaining change requires celebrating success. Dr. Afsar cautioned the audience to set appropriate expectations and goals to sustain system change. Further, once the change is set in motion, put the process ownership into the group (not on you). Creating data that is readily accessible and visible to the group helps them understand progress over time. Design the process to fit into natural workflow to make the process an easy transition that is sustainable long-term.
Key Points
• QI is a four legged stool: education, data audit and feedback, systems change, and culture change;
• Create a fishbone diagram to understand the cause and the effect;
• Create a sense of urgency by sharing inspirational data and finding your story and focusing on the why;
• Use process maps to identify areas of improvement;
• Prioritize those areas and implement small tests of change;
• Measure all outcomes and use statistical process control charts to demonstrate change;
• Motivate and coach people throughout the process change;
• Make data easily accessible to all members of the group to track progress; and
• Sustain change by setting realistic expectations and celebrating success.
Dr. Kanikkannan is Hospitalist Medical Director and Assistant Professor of Medicine at Rowan University School of Osteopathic Medicine and is a member of Team Hospitalist.
Arthroscopic Treatment of Femoroacetabular Impingement
Stigma is a family affair
Each year, 60 million Americans experience mental illness. Across the United States, each year, regardless of race, age, religion, or economic status, mental illness affects the lives of one in four adults and one in 10 children. This means that someone in every family has mental illness.
Most of our patients probably don’t tell anyone that they or one of their family members has mental illness They probably are doing what most of us do: Pretend it’s not there. Why? Because the stigma of mental illness is pervasive and destructive. What can we do to decrease the stigma?
The word stigma is derived from Greek and means "to mark the body." The bearer of the mark, or the stigma, is avoided and shunned. This practice has continued through the ages. In medieval times, if a person had a mental illness, he or she was thought to be possessed by demons and viewed as weak. Today, people with mental illness are viewed as menacing, deviant, unpredictable, incompetent, or even dangerous. It is entirely reasonable then, that we would want to avoid the stigma of mental illness. However, this prejudice against mental illness must be challenged.
Mental illness accounts for increased morbidity and mortality as well as lifetime disability. The World Health Organization (WHO) estimates that neuropsychiatric disorders are the leading cause of disability in the United States, followed by cardiovascular and circulatory diseases, and neoplasms. The neuropsychiatric disorders category, which includes mental and behavioral disorders, accounts for 13.6% of total U.S. disability-adjusted life years (DALYs). Neurological disorders account for 5.1% of total U.S. DALYs.
Impact on the family
Not only does stigma affect individuals, it affects family members as well. Family members suffer from SBA, or stigma by association (Brit. J. Psych. 2002;181:494-8), also known as courtesy stigma (Social. Psychiatry Psychiatr. Epidemiol. 2003;38:593-602). Families share stigma because families share a genetic heritage. Families share stigma by assuming responsibility for their family members’ behaviors. Families share stigma because they are seen as having common motivations (J. Pers. Soc. Psychol. 2012;102:224-41).
SBA causes psychological distress in family members (Rehabil. Psychol. 2013;58:73-80; J. Nerv. Ment. Dis. 1987;175:4-11; Br. J. Psychiatry 2002;181:494-8; and Schizophr. Bull. 1998;24:115-126).
Psychological complaints, such as brooding, inner unrest, and irritability, and physical complaints, such as insomnia, fatigue, and neck and shoulder pain, have been attributed to the psychological distress of SBA. Family members may avoid social interactions and conceal their relationship to the family member who is mentally ill (Acad. Psychiatry 2008;32:87-91). They might psychologically distance themselves from a relative with mental illness.
SBA varies by disease type, family role, and age. The greatest SBA is associated with drug dependence. These family members are blamed for the illness, held responsible for relapse, and viewed as incompetent. In the study of Patrick W. Corrigan, Psy.D., (J. Fam. Psychol. 2006;20:239-46), family members report feelings of "contamination" and shame. Severe depression or panic and phobias engender less stigma. More educated people are less likely to report feelings of stigma.
According to Dr. Corrigan, SBA varies by family role: Parents are blamed for causing the child’s mental illness, siblings are blamed for not ensuring that relatives with mental illness adhere to treatment plans, and children are fearful of being "contaminated" with the mental illness of their parent. The closer the relationship, the less the stigma is perceived as defining the person. Family closeness can reduce stigma (The Gerontologist 2012;52:89-97). Regarding age, a British study showed that the highest stigma is reported in the 16- to 19-year-old age group (Br. J. Psychiatry 2000;177:4-7).
Psychiatry as a profession has not helped diminish stigma. It is not uncommon to hear psychiatrists assign blame to parents or spouses. Psychiatrists often believe that the family has a role in the patient’s illness. How many spouses have been told they are "codependent" with the implication that they have somehow "caused the illness"? What can we do diminish stigma?
Fighting stigma
Fighting stigma means confronting stigma (Advances in Psychiatric Treatment 2000;6:65-72). Most efforts worldwide have begun with the idea of educating people about mental illness. These efforts, focused on promoting mental illness as a biological illness, have had limited success and in some situations actually increased stigma (Acta Psychiatr. Scand. 2012; 125:440-52). The answer may lie in targeted education: specific facts for specific groups.
For example, young couples with children become less fearful after education targeted specifically for them (Br. J. Psychiatry 1996;168:191-8). Antistigma campaigns are common throughout the world. The websites of most professional psychiatric organizations, such as the American Psychiatric Association, the Royal College of Psychiatrists, and the College of Psychiatrists of Ireland, provide information about antistigma campaigns. Organizations often partner with mental health charities. Antistigma efforts also focus on publishing articles about stigma as the Lancet did in a series a few years ago (1998;352:1048). It is unclear whether these efforts reduce stigma. Dr. Corrigan suggests that meeting people who have mental illness weakens the tendency to link mental illness and violence (Psychiatric Rehabilitation Skills 2002;6: 312-34).
The current consensus is that antistigma campaigns should focus on the competence of people with mental illness. In this vein, the Scottish Mental Health Arts & Film Festival highlights the contributions that people with mental illness make to society. The festival, which began in 2007, also sponsors a contest for films that depict people with mental illness in realistic, holistic ways. In 2013, the festival drew 12,000 attendees and sparked 120 newspaper articles that emphasized the fact that people with mental illness are generally active, useful members of society.
Meanwhile, a Canadian antistigma campaign tells the stories of people with mental illness and provides evidence of the competence of these people. The APA’s public service video series, "A Healthy Minds Minute," features celebrities and prominent figures calling for equal access to quality care, and insurance coverage for people with mental illness and substance use disorders.
What do we do to reduce stigma? Psychiatrists such as William Beardslee have written about their personal experience of a family member with mental illness. A member of the Association of Family Psychiatrists, Julie Totten, lost her brother to suicide and in response, she developed an organization called Families for Depression Awareness, which is devoted to reducing the stigma of mental illness. For me, it is my personal campaign to say: "One in four means that someone in everyone’s family has mental illness."
What more can we do?
• Speak up when you hear or see stigma.
• Stress the normalcy of people who have mental illness.
• Come out of the closet on behalf of yourself or a family member.
• Include people who acknowledge they suffer from mental illness in antistigma campaigns.
• Discuss the role of stigma with patients and their families. Ask "How has stigma affected you as a family? In what ways has your family helped reduce the stigma of your mental illness?"
• Encourage attendance at support groups, such as NAMI (the National Alliance on Mental Illness).
• Embrace your family member or yourself: Look for personal qualities that wipe out stigma.
• Don’t allow people to stigmatize patients: It might be your family member they are talking about.
• Talk positively about respecting our patients.
• Start a conversation to reduce stigma.
• Remember that fighting stigma means confronting stigma.
Dr. Heru is an associate professor of psychiatry at the University of Colorado at Denver, Aurora. She has been a member of the Association of Family Psychiatrists since 2002 and currently serves as the organization’s treasurer. She is the author of a new book, "Working With Families in Medical Settings" (New York: Routledge, 2013).
Each year, 60 million Americans experience mental illness. Across the United States, each year, regardless of race, age, religion, or economic status, mental illness affects the lives of one in four adults and one in 10 children. This means that someone in every family has mental illness.
Most of our patients probably don’t tell anyone that they or one of their family members has mental illness They probably are doing what most of us do: Pretend it’s not there. Why? Because the stigma of mental illness is pervasive and destructive. What can we do to decrease the stigma?
The word stigma is derived from Greek and means "to mark the body." The bearer of the mark, or the stigma, is avoided and shunned. This practice has continued through the ages. In medieval times, if a person had a mental illness, he or she was thought to be possessed by demons and viewed as weak. Today, people with mental illness are viewed as menacing, deviant, unpredictable, incompetent, or even dangerous. It is entirely reasonable then, that we would want to avoid the stigma of mental illness. However, this prejudice against mental illness must be challenged.
Mental illness accounts for increased morbidity and mortality as well as lifetime disability. The World Health Organization (WHO) estimates that neuropsychiatric disorders are the leading cause of disability in the United States, followed by cardiovascular and circulatory diseases, and neoplasms. The neuropsychiatric disorders category, which includes mental and behavioral disorders, accounts for 13.6% of total U.S. disability-adjusted life years (DALYs). Neurological disorders account for 5.1% of total U.S. DALYs.
Impact on the family
Not only does stigma affect individuals, it affects family members as well. Family members suffer from SBA, or stigma by association (Brit. J. Psych. 2002;181:494-8), also known as courtesy stigma (Social. Psychiatry Psychiatr. Epidemiol. 2003;38:593-602). Families share stigma because families share a genetic heritage. Families share stigma by assuming responsibility for their family members’ behaviors. Families share stigma because they are seen as having common motivations (J. Pers. Soc. Psychol. 2012;102:224-41).
SBA causes psychological distress in family members (Rehabil. Psychol. 2013;58:73-80; J. Nerv. Ment. Dis. 1987;175:4-11; Br. J. Psychiatry 2002;181:494-8; and Schizophr. Bull. 1998;24:115-126).
Psychological complaints, such as brooding, inner unrest, and irritability, and physical complaints, such as insomnia, fatigue, and neck and shoulder pain, have been attributed to the psychological distress of SBA. Family members may avoid social interactions and conceal their relationship to the family member who is mentally ill (Acad. Psychiatry 2008;32:87-91). They might psychologically distance themselves from a relative with mental illness.
SBA varies by disease type, family role, and age. The greatest SBA is associated with drug dependence. These family members are blamed for the illness, held responsible for relapse, and viewed as incompetent. In the study of Patrick W. Corrigan, Psy.D., (J. Fam. Psychol. 2006;20:239-46), family members report feelings of "contamination" and shame. Severe depression or panic and phobias engender less stigma. More educated people are less likely to report feelings of stigma.
According to Dr. Corrigan, SBA varies by family role: Parents are blamed for causing the child’s mental illness, siblings are blamed for not ensuring that relatives with mental illness adhere to treatment plans, and children are fearful of being "contaminated" with the mental illness of their parent. The closer the relationship, the less the stigma is perceived as defining the person. Family closeness can reduce stigma (The Gerontologist 2012;52:89-97). Regarding age, a British study showed that the highest stigma is reported in the 16- to 19-year-old age group (Br. J. Psychiatry 2000;177:4-7).
Psychiatry as a profession has not helped diminish stigma. It is not uncommon to hear psychiatrists assign blame to parents or spouses. Psychiatrists often believe that the family has a role in the patient’s illness. How many spouses have been told they are "codependent" with the implication that they have somehow "caused the illness"? What can we do diminish stigma?
Fighting stigma
Fighting stigma means confronting stigma (Advances in Psychiatric Treatment 2000;6:65-72). Most efforts worldwide have begun with the idea of educating people about mental illness. These efforts, focused on promoting mental illness as a biological illness, have had limited success and in some situations actually increased stigma (Acta Psychiatr. Scand. 2012; 125:440-52). The answer may lie in targeted education: specific facts for specific groups.
For example, young couples with children become less fearful after education targeted specifically for them (Br. J. Psychiatry 1996;168:191-8). Antistigma campaigns are common throughout the world. The websites of most professional psychiatric organizations, such as the American Psychiatric Association, the Royal College of Psychiatrists, and the College of Psychiatrists of Ireland, provide information about antistigma campaigns. Organizations often partner with mental health charities. Antistigma efforts also focus on publishing articles about stigma as the Lancet did in a series a few years ago (1998;352:1048). It is unclear whether these efforts reduce stigma. Dr. Corrigan suggests that meeting people who have mental illness weakens the tendency to link mental illness and violence (Psychiatric Rehabilitation Skills 2002;6: 312-34).
The current consensus is that antistigma campaigns should focus on the competence of people with mental illness. In this vein, the Scottish Mental Health Arts & Film Festival highlights the contributions that people with mental illness make to society. The festival, which began in 2007, also sponsors a contest for films that depict people with mental illness in realistic, holistic ways. In 2013, the festival drew 12,000 attendees and sparked 120 newspaper articles that emphasized the fact that people with mental illness are generally active, useful members of society.
Meanwhile, a Canadian antistigma campaign tells the stories of people with mental illness and provides evidence of the competence of these people. The APA’s public service video series, "A Healthy Minds Minute," features celebrities and prominent figures calling for equal access to quality care, and insurance coverage for people with mental illness and substance use disorders.
What do we do to reduce stigma? Psychiatrists such as William Beardslee have written about their personal experience of a family member with mental illness. A member of the Association of Family Psychiatrists, Julie Totten, lost her brother to suicide and in response, she developed an organization called Families for Depression Awareness, which is devoted to reducing the stigma of mental illness. For me, it is my personal campaign to say: "One in four means that someone in everyone’s family has mental illness."
What more can we do?
• Speak up when you hear or see stigma.
• Stress the normalcy of people who have mental illness.
• Come out of the closet on behalf of yourself or a family member.
• Include people who acknowledge they suffer from mental illness in antistigma campaigns.
• Discuss the role of stigma with patients and their families. Ask "How has stigma affected you as a family? In what ways has your family helped reduce the stigma of your mental illness?"
• Encourage attendance at support groups, such as NAMI (the National Alliance on Mental Illness).
• Embrace your family member or yourself: Look for personal qualities that wipe out stigma.
• Don’t allow people to stigmatize patients: It might be your family member they are talking about.
• Talk positively about respecting our patients.
• Start a conversation to reduce stigma.
• Remember that fighting stigma means confronting stigma.
Dr. Heru is an associate professor of psychiatry at the University of Colorado at Denver, Aurora. She has been a member of the Association of Family Psychiatrists since 2002 and currently serves as the organization’s treasurer. She is the author of a new book, "Working With Families in Medical Settings" (New York: Routledge, 2013).
Each year, 60 million Americans experience mental illness. Across the United States, each year, regardless of race, age, religion, or economic status, mental illness affects the lives of one in four adults and one in 10 children. This means that someone in every family has mental illness.
Most of our patients probably don’t tell anyone that they or one of their family members has mental illness They probably are doing what most of us do: Pretend it’s not there. Why? Because the stigma of mental illness is pervasive and destructive. What can we do to decrease the stigma?
The word stigma is derived from Greek and means "to mark the body." The bearer of the mark, or the stigma, is avoided and shunned. This practice has continued through the ages. In medieval times, if a person had a mental illness, he or she was thought to be possessed by demons and viewed as weak. Today, people with mental illness are viewed as menacing, deviant, unpredictable, incompetent, or even dangerous. It is entirely reasonable then, that we would want to avoid the stigma of mental illness. However, this prejudice against mental illness must be challenged.
Mental illness accounts for increased morbidity and mortality as well as lifetime disability. The World Health Organization (WHO) estimates that neuropsychiatric disorders are the leading cause of disability in the United States, followed by cardiovascular and circulatory diseases, and neoplasms. The neuropsychiatric disorders category, which includes mental and behavioral disorders, accounts for 13.6% of total U.S. disability-adjusted life years (DALYs). Neurological disorders account for 5.1% of total U.S. DALYs.
Impact on the family
Not only does stigma affect individuals, it affects family members as well. Family members suffer from SBA, or stigma by association (Brit. J. Psych. 2002;181:494-8), also known as courtesy stigma (Social. Psychiatry Psychiatr. Epidemiol. 2003;38:593-602). Families share stigma because families share a genetic heritage. Families share stigma by assuming responsibility for their family members’ behaviors. Families share stigma because they are seen as having common motivations (J. Pers. Soc. Psychol. 2012;102:224-41).
SBA causes psychological distress in family members (Rehabil. Psychol. 2013;58:73-80; J. Nerv. Ment. Dis. 1987;175:4-11; Br. J. Psychiatry 2002;181:494-8; and Schizophr. Bull. 1998;24:115-126).
Psychological complaints, such as brooding, inner unrest, and irritability, and physical complaints, such as insomnia, fatigue, and neck and shoulder pain, have been attributed to the psychological distress of SBA. Family members may avoid social interactions and conceal their relationship to the family member who is mentally ill (Acad. Psychiatry 2008;32:87-91). They might psychologically distance themselves from a relative with mental illness.
SBA varies by disease type, family role, and age. The greatest SBA is associated with drug dependence. These family members are blamed for the illness, held responsible for relapse, and viewed as incompetent. In the study of Patrick W. Corrigan, Psy.D., (J. Fam. Psychol. 2006;20:239-46), family members report feelings of "contamination" and shame. Severe depression or panic and phobias engender less stigma. More educated people are less likely to report feelings of stigma.
According to Dr. Corrigan, SBA varies by family role: Parents are blamed for causing the child’s mental illness, siblings are blamed for not ensuring that relatives with mental illness adhere to treatment plans, and children are fearful of being "contaminated" with the mental illness of their parent. The closer the relationship, the less the stigma is perceived as defining the person. Family closeness can reduce stigma (The Gerontologist 2012;52:89-97). Regarding age, a British study showed that the highest stigma is reported in the 16- to 19-year-old age group (Br. J. Psychiatry 2000;177:4-7).
Psychiatry as a profession has not helped diminish stigma. It is not uncommon to hear psychiatrists assign blame to parents or spouses. Psychiatrists often believe that the family has a role in the patient’s illness. How many spouses have been told they are "codependent" with the implication that they have somehow "caused the illness"? What can we do diminish stigma?
Fighting stigma
Fighting stigma means confronting stigma (Advances in Psychiatric Treatment 2000;6:65-72). Most efforts worldwide have begun with the idea of educating people about mental illness. These efforts, focused on promoting mental illness as a biological illness, have had limited success and in some situations actually increased stigma (Acta Psychiatr. Scand. 2012; 125:440-52). The answer may lie in targeted education: specific facts for specific groups.
For example, young couples with children become less fearful after education targeted specifically for them (Br. J. Psychiatry 1996;168:191-8). Antistigma campaigns are common throughout the world. The websites of most professional psychiatric organizations, such as the American Psychiatric Association, the Royal College of Psychiatrists, and the College of Psychiatrists of Ireland, provide information about antistigma campaigns. Organizations often partner with mental health charities. Antistigma efforts also focus on publishing articles about stigma as the Lancet did in a series a few years ago (1998;352:1048). It is unclear whether these efforts reduce stigma. Dr. Corrigan suggests that meeting people who have mental illness weakens the tendency to link mental illness and violence (Psychiatric Rehabilitation Skills 2002;6: 312-34).
The current consensus is that antistigma campaigns should focus on the competence of people with mental illness. In this vein, the Scottish Mental Health Arts & Film Festival highlights the contributions that people with mental illness make to society. The festival, which began in 2007, also sponsors a contest for films that depict people with mental illness in realistic, holistic ways. In 2013, the festival drew 12,000 attendees and sparked 120 newspaper articles that emphasized the fact that people with mental illness are generally active, useful members of society.
Meanwhile, a Canadian antistigma campaign tells the stories of people with mental illness and provides evidence of the competence of these people. The APA’s public service video series, "A Healthy Minds Minute," features celebrities and prominent figures calling for equal access to quality care, and insurance coverage for people with mental illness and substance use disorders.
What do we do to reduce stigma? Psychiatrists such as William Beardslee have written about their personal experience of a family member with mental illness. A member of the Association of Family Psychiatrists, Julie Totten, lost her brother to suicide and in response, she developed an organization called Families for Depression Awareness, which is devoted to reducing the stigma of mental illness. For me, it is my personal campaign to say: "One in four means that someone in everyone’s family has mental illness."
What more can we do?
• Speak up when you hear or see stigma.
• Stress the normalcy of people who have mental illness.
• Come out of the closet on behalf of yourself or a family member.
• Include people who acknowledge they suffer from mental illness in antistigma campaigns.
• Discuss the role of stigma with patients and their families. Ask "How has stigma affected you as a family? In what ways has your family helped reduce the stigma of your mental illness?"
• Encourage attendance at support groups, such as NAMI (the National Alliance on Mental Illness).
• Embrace your family member or yourself: Look for personal qualities that wipe out stigma.
• Don’t allow people to stigmatize patients: It might be your family member they are talking about.
• Talk positively about respecting our patients.
• Start a conversation to reduce stigma.
• Remember that fighting stigma means confronting stigma.
Dr. Heru is an associate professor of psychiatry at the University of Colorado at Denver, Aurora. She has been a member of the Association of Family Psychiatrists since 2002 and currently serves as the organization’s treasurer. She is the author of a new book, "Working With Families in Medical Settings" (New York: Routledge, 2013).
Monosomal karyotype, high prognostic risk score predicted transplantation failure
Monosomal karyotype and high prognostic risk according to the revised International Prognostic Scoring System are independent predictors of relapse and mortality in patients with myelodysplastic syndrome or oligoblastic acute myeloid leukemia who undergo allogeneic hematopoietic stem cell transplantation, according to findings from the GITMO (Gruppo Italiano Trapianto di Midollo Osseo) registry.
Treatment failure after allogeneic hematopoietic stem cell transplantation may be from transplant complications or relapse. To understand the predictors of failure, investigators studied outcomes in 519 patients with myelodysplastic syndrome or oligoblastic acute myeloid leukemia who underwent hematopoietic stem cell transplantation between 2000 and 2011.
Those with monosomal karyotype had a 49% relapse rate and a 10% 5-year overall survival rate; both rates were significantly worse, compared with patients without monosomal karyotype (P less than .001 for each). Those considered high or very-high risk based on the International Prognostic Scoring System (IPSS-R), had 39% and 23% 5-year overall survival, respectively, and 23% and 39% relapse rates, respectively (P less than .001 in all cases vs. patients not at high or very-high risk), Dr. Matteo G. Della Porta of Fondazione IRCCS Policlinico San Matteo, Pavia, Italy and colleagues reported on behalf of the GITMO.
Age of 50 years or older and high hematopoietic cell transplantation-comorbidity index scores were independent predictors of nonrelapse mortality (P = .02; P = .017, respectively), they found (Blood 2014 [doi:10.1182/blood-2013-12-542720]).
Accounting for various combinations of patients’ ages, IPSS-R category, monosomal karyotype, and high hematopoietic cell transplantation–comorbidity index, the 5-year probability of survival after allogeneic hematopoietic stem cell transplantation ranged from 0 to 94%. The analyses performed reinforce the concept that allogenic hematopoietic stem cell transplantation – the only potentially curative treatment for MDS – "offers optimal eradication of myelodysplastic hematopoiesis when the procedure is performed before MDS patients progress to advanced disease stages," the investigators concluded.
The investigators reported having no disclosures.
Monosomal karyotype and high prognostic risk according to the revised International Prognostic Scoring System are independent predictors of relapse and mortality in patients with myelodysplastic syndrome or oligoblastic acute myeloid leukemia who undergo allogeneic hematopoietic stem cell transplantation, according to findings from the GITMO (Gruppo Italiano Trapianto di Midollo Osseo) registry.
Treatment failure after allogeneic hematopoietic stem cell transplantation may be from transplant complications or relapse. To understand the predictors of failure, investigators studied outcomes in 519 patients with myelodysplastic syndrome or oligoblastic acute myeloid leukemia who underwent hematopoietic stem cell transplantation between 2000 and 2011.
Those with monosomal karyotype had a 49% relapse rate and a 10% 5-year overall survival rate; both rates were significantly worse, compared with patients without monosomal karyotype (P less than .001 for each). Those considered high or very-high risk based on the International Prognostic Scoring System (IPSS-R), had 39% and 23% 5-year overall survival, respectively, and 23% and 39% relapse rates, respectively (P less than .001 in all cases vs. patients not at high or very-high risk), Dr. Matteo G. Della Porta of Fondazione IRCCS Policlinico San Matteo, Pavia, Italy and colleagues reported on behalf of the GITMO.
Age of 50 years or older and high hematopoietic cell transplantation-comorbidity index scores were independent predictors of nonrelapse mortality (P = .02; P = .017, respectively), they found (Blood 2014 [doi:10.1182/blood-2013-12-542720]).
Accounting for various combinations of patients’ ages, IPSS-R category, monosomal karyotype, and high hematopoietic cell transplantation–comorbidity index, the 5-year probability of survival after allogeneic hematopoietic stem cell transplantation ranged from 0 to 94%. The analyses performed reinforce the concept that allogenic hematopoietic stem cell transplantation – the only potentially curative treatment for MDS – "offers optimal eradication of myelodysplastic hematopoiesis when the procedure is performed before MDS patients progress to advanced disease stages," the investigators concluded.
The investigators reported having no disclosures.
Monosomal karyotype and high prognostic risk according to the revised International Prognostic Scoring System are independent predictors of relapse and mortality in patients with myelodysplastic syndrome or oligoblastic acute myeloid leukemia who undergo allogeneic hematopoietic stem cell transplantation, according to findings from the GITMO (Gruppo Italiano Trapianto di Midollo Osseo) registry.
Treatment failure after allogeneic hematopoietic stem cell transplantation may be from transplant complications or relapse. To understand the predictors of failure, investigators studied outcomes in 519 patients with myelodysplastic syndrome or oligoblastic acute myeloid leukemia who underwent hematopoietic stem cell transplantation between 2000 and 2011.
Those with monosomal karyotype had a 49% relapse rate and a 10% 5-year overall survival rate; both rates were significantly worse, compared with patients without monosomal karyotype (P less than .001 for each). Those considered high or very-high risk based on the International Prognostic Scoring System (IPSS-R), had 39% and 23% 5-year overall survival, respectively, and 23% and 39% relapse rates, respectively (P less than .001 in all cases vs. patients not at high or very-high risk), Dr. Matteo G. Della Porta of Fondazione IRCCS Policlinico San Matteo, Pavia, Italy and colleagues reported on behalf of the GITMO.
Age of 50 years or older and high hematopoietic cell transplantation-comorbidity index scores were independent predictors of nonrelapse mortality (P = .02; P = .017, respectively), they found (Blood 2014 [doi:10.1182/blood-2013-12-542720]).
Accounting for various combinations of patients’ ages, IPSS-R category, monosomal karyotype, and high hematopoietic cell transplantation–comorbidity index, the 5-year probability of survival after allogeneic hematopoietic stem cell transplantation ranged from 0 to 94%. The analyses performed reinforce the concept that allogenic hematopoietic stem cell transplantation – the only potentially curative treatment for MDS – "offers optimal eradication of myelodysplastic hematopoiesis when the procedure is performed before MDS patients progress to advanced disease stages," the investigators concluded.
The investigators reported having no disclosures.
FROM BLOOD
Major finding: Patients with a monosomal karyotype had a 49% relapse rate and a 10% 5-year overall survival rate, and those considered high or very-high risk based on the IPSS-R, had 39% and 23% 5-year overall survival, respectively, and 23% and 39% relapse rates, respectively.
Data source: An analysis of GITMO registry data.
Disclosures: The investigators reported having no disclosures.
Cell-cycle inhibitor entinostat appeared to block azacitidine action
Coadministration with entinostat – a cell-cycle inhibitor – appears to decrease hematologic responsiveness to azacitidine treatment for high-risk myelodysplastic syndrome, according to data from an open-label, phase II randomized trial.
An earlier, phase I pilot study had suggested that the combination was effective and tolerable, however this phase II study in 149 patients (97 patients with myelodysplastic syndrome and 52 patients with acute myeloid leukemia) showed a lower overall hematologic response and lower median overall survival in the combination arm, compared with the azacitidine-only arm, said Dr. Thomas Prebet, who was at the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, when the work was done, but is now at Institut Paoli Calmettes, Marseille, France, and his colleagues.
They also performed genome-wide methylation studies on 99 specimens, finding that while demethylation in the combination arm was trending toward overall demethylation, it was significantly reduced, compared with the single agent arm, suggesting the entinostat was actually blocking the action of the azacitidine (J. Clin. Oncol. 2014 March 24 [doi:10.1200/JCO.2013.50.3102]).
The authors did note that the lower-dose, 10-day schedule of azacitidine, which was developed specifically for this phase II trial, appeared to double the rate of hematologic normalization, compared with that observed in a previous study using the standard 7-day schedule, suggesting that the longer schedule was more effective.
Some authors reported being consultants for and/or receiving research funding from various pharmaceutical companies. The study was partly supported by grants from the Leukemia and Lymphoma Society of America and Fulbright Franco-American Commission/Foundation.
Coadministration with entinostat – a cell-cycle inhibitor – appears to decrease hematologic responsiveness to azacitidine treatment for high-risk myelodysplastic syndrome, according to data from an open-label, phase II randomized trial.
An earlier, phase I pilot study had suggested that the combination was effective and tolerable, however this phase II study in 149 patients (97 patients with myelodysplastic syndrome and 52 patients with acute myeloid leukemia) showed a lower overall hematologic response and lower median overall survival in the combination arm, compared with the azacitidine-only arm, said Dr. Thomas Prebet, who was at the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, when the work was done, but is now at Institut Paoli Calmettes, Marseille, France, and his colleagues.
They also performed genome-wide methylation studies on 99 specimens, finding that while demethylation in the combination arm was trending toward overall demethylation, it was significantly reduced, compared with the single agent arm, suggesting the entinostat was actually blocking the action of the azacitidine (J. Clin. Oncol. 2014 March 24 [doi:10.1200/JCO.2013.50.3102]).
The authors did note that the lower-dose, 10-day schedule of azacitidine, which was developed specifically for this phase II trial, appeared to double the rate of hematologic normalization, compared with that observed in a previous study using the standard 7-day schedule, suggesting that the longer schedule was more effective.
Some authors reported being consultants for and/or receiving research funding from various pharmaceutical companies. The study was partly supported by grants from the Leukemia and Lymphoma Society of America and Fulbright Franco-American Commission/Foundation.
Coadministration with entinostat – a cell-cycle inhibitor – appears to decrease hematologic responsiveness to azacitidine treatment for high-risk myelodysplastic syndrome, according to data from an open-label, phase II randomized trial.
An earlier, phase I pilot study had suggested that the combination was effective and tolerable, however this phase II study in 149 patients (97 patients with myelodysplastic syndrome and 52 patients with acute myeloid leukemia) showed a lower overall hematologic response and lower median overall survival in the combination arm, compared with the azacitidine-only arm, said Dr. Thomas Prebet, who was at the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, when the work was done, but is now at Institut Paoli Calmettes, Marseille, France, and his colleagues.
They also performed genome-wide methylation studies on 99 specimens, finding that while demethylation in the combination arm was trending toward overall demethylation, it was significantly reduced, compared with the single agent arm, suggesting the entinostat was actually blocking the action of the azacitidine (J. Clin. Oncol. 2014 March 24 [doi:10.1200/JCO.2013.50.3102]).
The authors did note that the lower-dose, 10-day schedule of azacitidine, which was developed specifically for this phase II trial, appeared to double the rate of hematologic normalization, compared with that observed in a previous study using the standard 7-day schedule, suggesting that the longer schedule was more effective.
Some authors reported being consultants for and/or receiving research funding from various pharmaceutical companies. The study was partly supported by grants from the Leukemia and Lymphoma Society of America and Fulbright Franco-American Commission/Foundation.
FROM THE JOURNAL OF CLINICAL ONCOLOGY
Major finding: The addition of cell-cycle inhibitor entinostat to a 10-day schedule of treatment with azacitidine led to a lower overall hematologic response and lower median overall survival in the patients with high-risk myelodysplastic syndromes, compared with treatment with azacitidine alone.
Data source: An open-label, phase II randomized trial in 149 patients (97 patients with myelodysplastic syndrome and 52 patients with acute myeloid leukemia).
Disclosures: Some authors reported being consultants for and/or receiving research funding from various pharmaceutical companies. The study was partly supported by grants from the Leukemia and Lymphoma Society of America and Fulbright Franco-American Commission/Foundation.
EC approves SC formulation of rituximab
The European Commission (EC) has approved a subcutaneous (SC) formulation of rituximab (MabThera) to treat patients with follicular lymphoma or diffuse large B-cell lymphoma.
This formulation allows for 5-minute administration, a significant decrease over the 2.5-hour infusion time required to administer intravenous (IV) rituximab.
The drug’s maker, Roche, plans to begin launching SC rituximab in a number of European markets this year.
The EC’s approval of this formulation was primarily based on data from the SABRINA trial, which was recently published in The Lancet Oncology and funded by Roche.
In this phase 3 trial, researchers compared 3-week cycles of fixed-dose, SC rituximab to IV rituximab. They enrolled 127 patients with previously untreated, grade 1-3a, CD20-positive follicular lymphoma.
Patients were randomized to receive IV rituximab (375 mg/m2) or SC rituximab (1400 mg). After randomization, they received 1 induction dose of IV rituximab in cycle 1 and then their allocated treatment for cycles 2 through 8. Patients with a complete or partial response continued their treatment as maintenance every 8 weeks.
The study’s primary endpoint was the ratio of observed rituximab serum trough concentrations (Ctrough) between the 2 groups at cycle 7.
Pharmacokinetic data were available for 75% of patients (48/64) in the IV arm and 86% of the patients (54/63) in the SC arm.
An analysis of these data suggested SC rituximab was non-inferior to the IV formulation. The geometric mean Ctrough was 83.13 μg/mL in the IV arm and 134.58 μg/mL in the SC arm (ratio, 1.62).
The rate of adverse events was similar between the 2 arms, occurring in 88% (57/65) of patients in the IV arm and 92% (57/62) of patients in the SC arm. Grade 3 or higher adverse events occurred in 46% (n=30) and 47% (n=29) of patients, respectively.
The most common grade 3 or higher adverse event in both arms was neutropenia. It occurred in 22% (n=14) of patients in the IV arm and 26% (n=16) in the SC arm.
Adverse events related to administration were mostly grade 1-2. And they occurred more often in the SC arm than in the IV arm, in 50% (n=31) and 32% (n=21) of patients, respectively.
The researchers said these results suggest the SC formulation of rituximab is non-inferior to the IV formulation and poses no new safety concerns.
The European Commission (EC) has approved a subcutaneous (SC) formulation of rituximab (MabThera) to treat patients with follicular lymphoma or diffuse large B-cell lymphoma.
This formulation allows for 5-minute administration, a significant decrease over the 2.5-hour infusion time required to administer intravenous (IV) rituximab.
The drug’s maker, Roche, plans to begin launching SC rituximab in a number of European markets this year.
The EC’s approval of this formulation was primarily based on data from the SABRINA trial, which was recently published in The Lancet Oncology and funded by Roche.
In this phase 3 trial, researchers compared 3-week cycles of fixed-dose, SC rituximab to IV rituximab. They enrolled 127 patients with previously untreated, grade 1-3a, CD20-positive follicular lymphoma.
Patients were randomized to receive IV rituximab (375 mg/m2) or SC rituximab (1400 mg). After randomization, they received 1 induction dose of IV rituximab in cycle 1 and then their allocated treatment for cycles 2 through 8. Patients with a complete or partial response continued their treatment as maintenance every 8 weeks.
The study’s primary endpoint was the ratio of observed rituximab serum trough concentrations (Ctrough) between the 2 groups at cycle 7.
Pharmacokinetic data were available for 75% of patients (48/64) in the IV arm and 86% of the patients (54/63) in the SC arm.
An analysis of these data suggested SC rituximab was non-inferior to the IV formulation. The geometric mean Ctrough was 83.13 μg/mL in the IV arm and 134.58 μg/mL in the SC arm (ratio, 1.62).
The rate of adverse events was similar between the 2 arms, occurring in 88% (57/65) of patients in the IV arm and 92% (57/62) of patients in the SC arm. Grade 3 or higher adverse events occurred in 46% (n=30) and 47% (n=29) of patients, respectively.
The most common grade 3 or higher adverse event in both arms was neutropenia. It occurred in 22% (n=14) of patients in the IV arm and 26% (n=16) in the SC arm.
Adverse events related to administration were mostly grade 1-2. And they occurred more often in the SC arm than in the IV arm, in 50% (n=31) and 32% (n=21) of patients, respectively.
The researchers said these results suggest the SC formulation of rituximab is non-inferior to the IV formulation and poses no new safety concerns.
The European Commission (EC) has approved a subcutaneous (SC) formulation of rituximab (MabThera) to treat patients with follicular lymphoma or diffuse large B-cell lymphoma.
This formulation allows for 5-minute administration, a significant decrease over the 2.5-hour infusion time required to administer intravenous (IV) rituximab.
The drug’s maker, Roche, plans to begin launching SC rituximab in a number of European markets this year.
The EC’s approval of this formulation was primarily based on data from the SABRINA trial, which was recently published in The Lancet Oncology and funded by Roche.
In this phase 3 trial, researchers compared 3-week cycles of fixed-dose, SC rituximab to IV rituximab. They enrolled 127 patients with previously untreated, grade 1-3a, CD20-positive follicular lymphoma.
Patients were randomized to receive IV rituximab (375 mg/m2) or SC rituximab (1400 mg). After randomization, they received 1 induction dose of IV rituximab in cycle 1 and then their allocated treatment for cycles 2 through 8. Patients with a complete or partial response continued their treatment as maintenance every 8 weeks.
The study’s primary endpoint was the ratio of observed rituximab serum trough concentrations (Ctrough) between the 2 groups at cycle 7.
Pharmacokinetic data were available for 75% of patients (48/64) in the IV arm and 86% of the patients (54/63) in the SC arm.
An analysis of these data suggested SC rituximab was non-inferior to the IV formulation. The geometric mean Ctrough was 83.13 μg/mL in the IV arm and 134.58 μg/mL in the SC arm (ratio, 1.62).
The rate of adverse events was similar between the 2 arms, occurring in 88% (57/65) of patients in the IV arm and 92% (57/62) of patients in the SC arm. Grade 3 or higher adverse events occurred in 46% (n=30) and 47% (n=29) of patients, respectively.
The most common grade 3 or higher adverse event in both arms was neutropenia. It occurred in 22% (n=14) of patients in the IV arm and 26% (n=16) in the SC arm.
Adverse events related to administration were mostly grade 1-2. And they occurred more often in the SC arm than in the IV arm, in 50% (n=31) and 32% (n=21) of patients, respectively.
The researchers said these results suggest the SC formulation of rituximab is non-inferior to the IV formulation and poses no new safety concerns.
ALL cells don’t survive SSC culture method
Results of a pilot study suggest safe transplantation of spermatogonial stem cells (SSCs) may be possible, potentially bringing us one step closer to ensuring fertility preservation in young boys with cancer.
Investigators found they could culture a large amount of SSCs using testicular tissue from boys with acute lymphoblastic leukemia (ALL).
And the ALL cells did not survive the culture process.
The researchers reported these results in Fertility and Sterility.
“Our study addressed an important safety issue—whether cancer cells that might be present in testicular tissue samples can survive the process to replicate the sperm-producing stem cells,” said study author Hooman Sadri-Ardekani, MD, PhD, of Wake Forest Baptist Medical Center in Winston-Salem, North Carolina.
“This is an important consideration because of the potential to re-introduce cancer into the patient. The research, which involved one of the most common childhood cancers, shows that the cancer cells were eliminated. Based on these findings, we recommend that all boys with cancer be offered the option of storing testicular tissue for possible future clinical use.”
Previous research had shown that up to 30% of boys with ALL had cancer cells in their testicular tissue. In several studies, researchers attempted to eliminate cancer cells from biopsy tissue, but the results varied.
So Dr Sadri-Ardekani and his colleagues decided to investigate whether cancer cells would survive the protocol they had developed to reproduce SSCs from a small tissue biopsy.
This process multiplies the original SSCs by 18,000-fold so there are enough cells to transplant back into the patient when he reaches adulthood.
The investigators tested the method using samples from 3 ALL patients. The team cultured the ALL cells alone and at various concentrations in combination with testicular cells.
The ALL cells cultured alone did not survive beyond 14 days. But the testicular cells cultured in parallel survived and continue to proliferate well after 8 weeks.
At 10 to 16 days, the ALL cells cultured in combination had begun to die off. The cells were undetectable in cultures from 2 of the patients that had an initial ALL concentration of 0.04%, 0.4%, or 4%.
At 20 to 26 days, ALL cells were undetectable in all cultures, even those with an initial concentration of 40% ALL cells.
“This pilot study showed that the culture system not only allowed for efficient propagation of sperm stem cells but also eliminated ALL cells,” Dr Sadri-Ardekani said.
SSC transplants have not yet been attempted in humans, but they have been performed successfully in several species of animals, including monkeys.
Dr Sadri-Ardekani noted that before we can perform SSC transplants in patients, additional research is needed, particularly, research investigating whether other types of leukemia cells will also be eliminated in the cell-propagation process.
Results of a pilot study suggest safe transplantation of spermatogonial stem cells (SSCs) may be possible, potentially bringing us one step closer to ensuring fertility preservation in young boys with cancer.
Investigators found they could culture a large amount of SSCs using testicular tissue from boys with acute lymphoblastic leukemia (ALL).
And the ALL cells did not survive the culture process.
The researchers reported these results in Fertility and Sterility.
“Our study addressed an important safety issue—whether cancer cells that might be present in testicular tissue samples can survive the process to replicate the sperm-producing stem cells,” said study author Hooman Sadri-Ardekani, MD, PhD, of Wake Forest Baptist Medical Center in Winston-Salem, North Carolina.
“This is an important consideration because of the potential to re-introduce cancer into the patient. The research, which involved one of the most common childhood cancers, shows that the cancer cells were eliminated. Based on these findings, we recommend that all boys with cancer be offered the option of storing testicular tissue for possible future clinical use.”
Previous research had shown that up to 30% of boys with ALL had cancer cells in their testicular tissue. In several studies, researchers attempted to eliminate cancer cells from biopsy tissue, but the results varied.
So Dr Sadri-Ardekani and his colleagues decided to investigate whether cancer cells would survive the protocol they had developed to reproduce SSCs from a small tissue biopsy.
This process multiplies the original SSCs by 18,000-fold so there are enough cells to transplant back into the patient when he reaches adulthood.
The investigators tested the method using samples from 3 ALL patients. The team cultured the ALL cells alone and at various concentrations in combination with testicular cells.
The ALL cells cultured alone did not survive beyond 14 days. But the testicular cells cultured in parallel survived and continue to proliferate well after 8 weeks.
At 10 to 16 days, the ALL cells cultured in combination had begun to die off. The cells were undetectable in cultures from 2 of the patients that had an initial ALL concentration of 0.04%, 0.4%, or 4%.
At 20 to 26 days, ALL cells were undetectable in all cultures, even those with an initial concentration of 40% ALL cells.
“This pilot study showed that the culture system not only allowed for efficient propagation of sperm stem cells but also eliminated ALL cells,” Dr Sadri-Ardekani said.
SSC transplants have not yet been attempted in humans, but they have been performed successfully in several species of animals, including monkeys.
Dr Sadri-Ardekani noted that before we can perform SSC transplants in patients, additional research is needed, particularly, research investigating whether other types of leukemia cells will also be eliminated in the cell-propagation process.
Results of a pilot study suggest safe transplantation of spermatogonial stem cells (SSCs) may be possible, potentially bringing us one step closer to ensuring fertility preservation in young boys with cancer.
Investigators found they could culture a large amount of SSCs using testicular tissue from boys with acute lymphoblastic leukemia (ALL).
And the ALL cells did not survive the culture process.
The researchers reported these results in Fertility and Sterility.
“Our study addressed an important safety issue—whether cancer cells that might be present in testicular tissue samples can survive the process to replicate the sperm-producing stem cells,” said study author Hooman Sadri-Ardekani, MD, PhD, of Wake Forest Baptist Medical Center in Winston-Salem, North Carolina.
“This is an important consideration because of the potential to re-introduce cancer into the patient. The research, which involved one of the most common childhood cancers, shows that the cancer cells were eliminated. Based on these findings, we recommend that all boys with cancer be offered the option of storing testicular tissue for possible future clinical use.”
Previous research had shown that up to 30% of boys with ALL had cancer cells in their testicular tissue. In several studies, researchers attempted to eliminate cancer cells from biopsy tissue, but the results varied.
So Dr Sadri-Ardekani and his colleagues decided to investigate whether cancer cells would survive the protocol they had developed to reproduce SSCs from a small tissue biopsy.
This process multiplies the original SSCs by 18,000-fold so there are enough cells to transplant back into the patient when he reaches adulthood.
The investigators tested the method using samples from 3 ALL patients. The team cultured the ALL cells alone and at various concentrations in combination with testicular cells.
The ALL cells cultured alone did not survive beyond 14 days. But the testicular cells cultured in parallel survived and continue to proliferate well after 8 weeks.
At 10 to 16 days, the ALL cells cultured in combination had begun to die off. The cells were undetectable in cultures from 2 of the patients that had an initial ALL concentration of 0.04%, 0.4%, or 4%.
At 20 to 26 days, ALL cells were undetectable in all cultures, even those with an initial concentration of 40% ALL cells.
“This pilot study showed that the culture system not only allowed for efficient propagation of sperm stem cells but also eliminated ALL cells,” Dr Sadri-Ardekani said.
SSC transplants have not yet been attempted in humans, but they have been performed successfully in several species of animals, including monkeys.
Dr Sadri-Ardekani noted that before we can perform SSC transplants in patients, additional research is needed, particularly, research investigating whether other types of leukemia cells will also be eliminated in the cell-propagation process.
CDC reports more cases of Heartland virus disease
Credit: CDC
Health officials have reported 6 new cases of Heartland virus disease—5 in Missouri and 1 in Tennessee.
These cases, discovered in 2012 and 2013, add to the 2 cases discovered in 2009 and are described in the Centers for Disease Control and Prevention (CDC) Morbidity and Mortality Weekly Report.
The Heartland virus was first reported in 2 farmers in northwestern Missouri who were hospitalized in 2009 with what was thought to be ehrlichiosis, a tick-borne disease.
However, the patients failed to improve with treatment, and testing failed to confirm ehlrlichiosis.
Working with state and local partners, the CDC eventually identified the cause of the men’s illness: a previously unknown phlebovirus, now dubbed the Heartland virus.
Although we do not know for certain how patients are infected with the virus, research has suggested that ticks, namely lone star ticks, transmit it.
Ongoing investigations have uncovered 6 more cases of Heartland virus disease. All of the patients were white men older than 50 years of age. Five of them reported tick bites in the days or weeks before they fell ill.
Their symptoms started in May to September and included fever, fatigue, loss of appetite, headache, nausea, and muscle pain. The patients also had leukopenia and thrombocytopenia.
Four of the 6 patients were hospitalized. And 1 man, who suffered from other health conditions, died. It is not known if the Heartland virus was the cause of death or how much it contributed to his death.
The CDC has been working with the Missouri and Tennessee state health departments and other federal agencies to advance our understanding of Heartland virus disease by learning more about the patients who were infected, their illness, and their exposure to ticks.
The CDC aims to determine the symptoms and severity of the disease, where it is found, how people are being infected, and how to prevent infections.
CDC studies to date have shown that the Heartland virus is carried by lone star ticks, which are primarily found in the southeastern and eastern US.
Researchers hope additional studies can confirm whether ticks can spread the virus and reveal which other insects or animals may be involved in the transmission cycle. The CDC is also looking for the Heartland virus in other parts of the country to understand how widely it may be distributed.
“During the past 2 years, CDC has worked closely with state health departments, hospitals, and many experts from universities and other federal agencies to learn more about Heartland virus,” said Roger Nasci, PhD, chief of the CDC’s Arboviral Diseases Branch.
“By gathering information about the disease Heartland virus causes, and about how it’s spread to people, we hope to better understand the potential impact on the public’s health and how we can help protect people from this virus.”
The CDC developed the blood tests used to confirm the new cases of Heartland virus disease. CDC teams are working to further validate these tests and develop additional tests. The researchers hope to develop a diagnostic test that public health laboratories could use to test for the virus.
There is no specific treatment or vaccine for Heartland virus disease. However, supportive therapies such as intravenous fluids and fever reducers can relieve some symptoms of Heartland disease.
To reduce the risk of Heartland and other vector-borne diseases, the CDC recommends:
- Avoiding wooded and bushy areas with high grass and leaf litter
- Using insect repellent when outdoors
- Using products that contain permethrin on clothing
- Conducting a full-body tick check after spending time outdoors
- Bathing as soon as possible after coming indoors to wash off and more easily find any ticks
- Examining gear and pets, as ticks can “ride” into the home and attach to a person later.
For more information on the Heartland virus, visit the CDC website.
Credit: CDC
Health officials have reported 6 new cases of Heartland virus disease—5 in Missouri and 1 in Tennessee.
These cases, discovered in 2012 and 2013, add to the 2 cases discovered in 2009 and are described in the Centers for Disease Control and Prevention (CDC) Morbidity and Mortality Weekly Report.
The Heartland virus was first reported in 2 farmers in northwestern Missouri who were hospitalized in 2009 with what was thought to be ehrlichiosis, a tick-borne disease.
However, the patients failed to improve with treatment, and testing failed to confirm ehlrlichiosis.
Working with state and local partners, the CDC eventually identified the cause of the men’s illness: a previously unknown phlebovirus, now dubbed the Heartland virus.
Although we do not know for certain how patients are infected with the virus, research has suggested that ticks, namely lone star ticks, transmit it.
Ongoing investigations have uncovered 6 more cases of Heartland virus disease. All of the patients were white men older than 50 years of age. Five of them reported tick bites in the days or weeks before they fell ill.
Their symptoms started in May to September and included fever, fatigue, loss of appetite, headache, nausea, and muscle pain. The patients also had leukopenia and thrombocytopenia.
Four of the 6 patients were hospitalized. And 1 man, who suffered from other health conditions, died. It is not known if the Heartland virus was the cause of death or how much it contributed to his death.
The CDC has been working with the Missouri and Tennessee state health departments and other federal agencies to advance our understanding of Heartland virus disease by learning more about the patients who were infected, their illness, and their exposure to ticks.
The CDC aims to determine the symptoms and severity of the disease, where it is found, how people are being infected, and how to prevent infections.
CDC studies to date have shown that the Heartland virus is carried by lone star ticks, which are primarily found in the southeastern and eastern US.
Researchers hope additional studies can confirm whether ticks can spread the virus and reveal which other insects or animals may be involved in the transmission cycle. The CDC is also looking for the Heartland virus in other parts of the country to understand how widely it may be distributed.
“During the past 2 years, CDC has worked closely with state health departments, hospitals, and many experts from universities and other federal agencies to learn more about Heartland virus,” said Roger Nasci, PhD, chief of the CDC’s Arboviral Diseases Branch.
“By gathering information about the disease Heartland virus causes, and about how it’s spread to people, we hope to better understand the potential impact on the public’s health and how we can help protect people from this virus.”
The CDC developed the blood tests used to confirm the new cases of Heartland virus disease. CDC teams are working to further validate these tests and develop additional tests. The researchers hope to develop a diagnostic test that public health laboratories could use to test for the virus.
There is no specific treatment or vaccine for Heartland virus disease. However, supportive therapies such as intravenous fluids and fever reducers can relieve some symptoms of Heartland disease.
To reduce the risk of Heartland and other vector-borne diseases, the CDC recommends:
- Avoiding wooded and bushy areas with high grass and leaf litter
- Using insect repellent when outdoors
- Using products that contain permethrin on clothing
- Conducting a full-body tick check after spending time outdoors
- Bathing as soon as possible after coming indoors to wash off and more easily find any ticks
- Examining gear and pets, as ticks can “ride” into the home and attach to a person later.
For more information on the Heartland virus, visit the CDC website.
Credit: CDC
Health officials have reported 6 new cases of Heartland virus disease—5 in Missouri and 1 in Tennessee.
These cases, discovered in 2012 and 2013, add to the 2 cases discovered in 2009 and are described in the Centers for Disease Control and Prevention (CDC) Morbidity and Mortality Weekly Report.
The Heartland virus was first reported in 2 farmers in northwestern Missouri who were hospitalized in 2009 with what was thought to be ehrlichiosis, a tick-borne disease.
However, the patients failed to improve with treatment, and testing failed to confirm ehlrlichiosis.
Working with state and local partners, the CDC eventually identified the cause of the men’s illness: a previously unknown phlebovirus, now dubbed the Heartland virus.
Although we do not know for certain how patients are infected with the virus, research has suggested that ticks, namely lone star ticks, transmit it.
Ongoing investigations have uncovered 6 more cases of Heartland virus disease. All of the patients were white men older than 50 years of age. Five of them reported tick bites in the days or weeks before they fell ill.
Their symptoms started in May to September and included fever, fatigue, loss of appetite, headache, nausea, and muscle pain. The patients also had leukopenia and thrombocytopenia.
Four of the 6 patients were hospitalized. And 1 man, who suffered from other health conditions, died. It is not known if the Heartland virus was the cause of death or how much it contributed to his death.
The CDC has been working with the Missouri and Tennessee state health departments and other federal agencies to advance our understanding of Heartland virus disease by learning more about the patients who were infected, their illness, and their exposure to ticks.
The CDC aims to determine the symptoms and severity of the disease, where it is found, how people are being infected, and how to prevent infections.
CDC studies to date have shown that the Heartland virus is carried by lone star ticks, which are primarily found in the southeastern and eastern US.
Researchers hope additional studies can confirm whether ticks can spread the virus and reveal which other insects or animals may be involved in the transmission cycle. The CDC is also looking for the Heartland virus in other parts of the country to understand how widely it may be distributed.
“During the past 2 years, CDC has worked closely with state health departments, hospitals, and many experts from universities and other federal agencies to learn more about Heartland virus,” said Roger Nasci, PhD, chief of the CDC’s Arboviral Diseases Branch.
“By gathering information about the disease Heartland virus causes, and about how it’s spread to people, we hope to better understand the potential impact on the public’s health and how we can help protect people from this virus.”
The CDC developed the blood tests used to confirm the new cases of Heartland virus disease. CDC teams are working to further validate these tests and develop additional tests. The researchers hope to develop a diagnostic test that public health laboratories could use to test for the virus.
There is no specific treatment or vaccine for Heartland virus disease. However, supportive therapies such as intravenous fluids and fever reducers can relieve some symptoms of Heartland disease.
To reduce the risk of Heartland and other vector-borne diseases, the CDC recommends:
- Avoiding wooded and bushy areas with high grass and leaf litter
- Using insect repellent when outdoors
- Using products that contain permethrin on clothing
- Conducting a full-body tick check after spending time outdoors
- Bathing as soon as possible after coming indoors to wash off and more easily find any ticks
- Examining gear and pets, as ticks can “ride” into the home and attach to a person later.
For more information on the Heartland virus, visit the CDC website.
FDA approves factor for hemophilia B
The US Food and Drug Administration (FDA) has approved a recombinant factor IX Fc fusion protein (rFIXFc, Alprolix) for use in adults and children with hemophilia B.
The product is intended to help control, prevent, or reduce the frequency of bleeding episodes that can occur day-to-day in patients with hemophilia B, as well as manage bleeding in patients undergoing surgery.
rFIXFc is made by fusing factor IX to the Fc portion of the IgG1 protein. Scientists believe this enables the product to use a naturally occurring pathway to prolong the time the therapy remains in the body.
The FDA’s approval is the second worldwide approval of rFIXFc. The product was recently authorized for use in Canada.
Researchers evaluated the safety, efficacy, and pharmacokinetics of rFIXFc in the phase 3 B-LONG study, which was funded by the product’s developers, Biogen Idec and Sobi.
Investigators tested rFIXFc in 123 males with hemophilia B who were at least 12 years of age. The patients were assigned to 1 of 4 treatment arms: weekly prophylaxis, individualized-interval prophylaxis, on-demand treatment to control bleeding, and perioperative management.
The overall median annualized bleeding rates were 2.95 for the weekly prophylaxis arm, 1.38 for the individualized-interval prophylaxis arm, and 17.69 in the on-demand treatment arm.
The overall median dosing interval with individualized-interval prophylaxis was 12.5 days. During the last 6 months of the study, the median dosing interval was 13.8 days.
Of the patients who received rFIXFc for perioperative management, 85.7% required a single injection of the product to maintain hemostasis during their operation. The median dose was 90.9 IU/kg per injection.
Most patients required 1 to 2 injections of the product the day before and the day of surgery. And most required 2 to 3 injections from days 1 to 3 after surgery.
The most common adverse events associated with rFIXFc (an incidence of ≥ 5% in a pooled analysis of treatment arms 1, 2, and 3) were nasopharyngitis, influenza, arthralgia, upper respiratory tract infection, hypertension, and headache.
The results of this study were released by Biogen Idec and Sobi in September 2012, presented at ISTH 2013, and published in NEJM in December 2013.
The US Food and Drug Administration (FDA) has approved a recombinant factor IX Fc fusion protein (rFIXFc, Alprolix) for use in adults and children with hemophilia B.
The product is intended to help control, prevent, or reduce the frequency of bleeding episodes that can occur day-to-day in patients with hemophilia B, as well as manage bleeding in patients undergoing surgery.
rFIXFc is made by fusing factor IX to the Fc portion of the IgG1 protein. Scientists believe this enables the product to use a naturally occurring pathway to prolong the time the therapy remains in the body.
The FDA’s approval is the second worldwide approval of rFIXFc. The product was recently authorized for use in Canada.
Researchers evaluated the safety, efficacy, and pharmacokinetics of rFIXFc in the phase 3 B-LONG study, which was funded by the product’s developers, Biogen Idec and Sobi.
Investigators tested rFIXFc in 123 males with hemophilia B who were at least 12 years of age. The patients were assigned to 1 of 4 treatment arms: weekly prophylaxis, individualized-interval prophylaxis, on-demand treatment to control bleeding, and perioperative management.
The overall median annualized bleeding rates were 2.95 for the weekly prophylaxis arm, 1.38 for the individualized-interval prophylaxis arm, and 17.69 in the on-demand treatment arm.
The overall median dosing interval with individualized-interval prophylaxis was 12.5 days. During the last 6 months of the study, the median dosing interval was 13.8 days.
Of the patients who received rFIXFc for perioperative management, 85.7% required a single injection of the product to maintain hemostasis during their operation. The median dose was 90.9 IU/kg per injection.
Most patients required 1 to 2 injections of the product the day before and the day of surgery. And most required 2 to 3 injections from days 1 to 3 after surgery.
The most common adverse events associated with rFIXFc (an incidence of ≥ 5% in a pooled analysis of treatment arms 1, 2, and 3) were nasopharyngitis, influenza, arthralgia, upper respiratory tract infection, hypertension, and headache.
The results of this study were released by Biogen Idec and Sobi in September 2012, presented at ISTH 2013, and published in NEJM in December 2013.
The US Food and Drug Administration (FDA) has approved a recombinant factor IX Fc fusion protein (rFIXFc, Alprolix) for use in adults and children with hemophilia B.
The product is intended to help control, prevent, or reduce the frequency of bleeding episodes that can occur day-to-day in patients with hemophilia B, as well as manage bleeding in patients undergoing surgery.
rFIXFc is made by fusing factor IX to the Fc portion of the IgG1 protein. Scientists believe this enables the product to use a naturally occurring pathway to prolong the time the therapy remains in the body.
The FDA’s approval is the second worldwide approval of rFIXFc. The product was recently authorized for use in Canada.
Researchers evaluated the safety, efficacy, and pharmacokinetics of rFIXFc in the phase 3 B-LONG study, which was funded by the product’s developers, Biogen Idec and Sobi.
Investigators tested rFIXFc in 123 males with hemophilia B who were at least 12 years of age. The patients were assigned to 1 of 4 treatment arms: weekly prophylaxis, individualized-interval prophylaxis, on-demand treatment to control bleeding, and perioperative management.
The overall median annualized bleeding rates were 2.95 for the weekly prophylaxis arm, 1.38 for the individualized-interval prophylaxis arm, and 17.69 in the on-demand treatment arm.
The overall median dosing interval with individualized-interval prophylaxis was 12.5 days. During the last 6 months of the study, the median dosing interval was 13.8 days.
Of the patients who received rFIXFc for perioperative management, 85.7% required a single injection of the product to maintain hemostasis during their operation. The median dose was 90.9 IU/kg per injection.
Most patients required 1 to 2 injections of the product the day before and the day of surgery. And most required 2 to 3 injections from days 1 to 3 after surgery.
The most common adverse events associated with rFIXFc (an incidence of ≥ 5% in a pooled analysis of treatment arms 1, 2, and 3) were nasopharyngitis, influenza, arthralgia, upper respiratory tract infection, hypertension, and headache.
The results of this study were released by Biogen Idec and Sobi in September 2012, presented at ISTH 2013, and published in NEJM in December 2013.