The $64,000 Question/

Article Type
Changed
Wed, 05/23/2018 - 14:49
Display Headline
The $64,000 question

A 40‐year‐old Sudanese man was admitted due to worsening abdominal pain with recurrent ascites. He had a history of hepatitis B (HBV) infection and diabetes. He previously drank 3 beers per day on the weekends, but he had not consumed alcohol in over a year. He was born in Sudan but lived in Egypt most of his adult life; he immigrated to the United States 6 years previously. He was hospitalized out of state 9 months ago for a swollen abdomen and underwent an exploratory laparotomy that reportedly was unremarkable except for ascites.

Portal hypertension due to liver disease is the most common cause of ascites. This patient has a known risk factor for liver disease (history of HBV infection). Although his reported alcohol consumption is low, there is a synergistic effect on liver injury in the setting of chronic hepatitis. Abdominal pain in the setting of ascites needs to be urgently evaluated to exclude spontaneous bacterial peritonitis (SBP). Also, because chronic HBV infection is the major risk factor for hepatocellular carcinoma in the world, malignant ascites is in the differential. Hepatic vascular thrombosis and tuberculous peritonitis (given the patient's country of origin and travel history) also should be considered. The most appropriate initial test would be a diagnostic paracentesis to support or exclude the presence of SBP and direct the evaluation toward liver disease or other less‐common causes of ascites.

The patient was seen as an outpatient 5 months prior to admission with transient fever and joint pains. Laboratory studies at that visit were notable for a serum albumin of 3.2 g/dL (normal 3.55), 2.4 g of predicted 24‐hour protein on urinalysis (normal <30 mg per 24 hours), creatinine of 0.5 mg/dL (normal 0.81.3), and a positive hepatitis B surface antibody. The working diagnosis was a nonspecific viral syndrome and his symptoms resolved without treatment. One month later, he developed ascites and mild lower extremity edema. Additional laboratory studies at that time showed a normocytic anemia with hemoglobin 11.7 g/dL (normal 13.517.5) and leukopenia with white blood cell count of 2.4 109/L (normal 3.510.5), neutrophil count of 1.45 109/L (normal 1.77.0), and lymphocyte count of 0.58 109/L (normal 0.902.90). Transaminases, serum bilirubin, prothrombin time, alpha fetoprotein, and peripheral blood smear were normal. Human immunodeficiency virus antibody screen and QuantiFERON‐TB assay were negative. Hemoglobin A1c was 6.2% (normal 4.06.0). Repeat urinalysis demonstrated 883 mg of predicted 24‐hour protein. Computed tomography (CT) of the abdomen showed a large amount of intra‐abdominal ascites; the liver and spleen were normal, and there were no varices or other evidence of portal hypertension. Echocardiogram was normal except for a small inferior vena cava (IVC) and a mildly increased right ventricular systolic pressure of 32 mm Hg (systolic blood pressure 98 mm Hg). Due to the indeterminate cause for the patient's ascites, referral was made for gastroenterology evaluation with consideration for a paracentesis.

Cirrhotic ascites seems less likely. Postsinusoidal causes of portal hypertension (eg, cardiomyopathy) are also less likely given the absence of suggestive findings on echocardiography. Malignant ascites also appears less probable in the absence of suggestive findings such as mass lesions, lymphadenopathy, or peritoneal carcinomatosis on CT imaging. The suspicion for tuberculous peritonitis is lower with the negative QuantiFERON‐TB test. Hypoalbuminemia, normocytic anemia, leukopenia, and proteinuria all suggest a systemic inflammatory condition (eg, systemic lupus erythematosus [SLE]) with inflammatory serositis causing ascites). Nephrotic syndrome can cause hypoalbuminemia, edema, and ascites, but his total urine protein losses of <3.5 grams per 24 hours are not in keeping with this diagnosis. Other uncommon causes of ascites such as chylous ascites have not yet been excluded. The most appropriate next step remains ascitic fluid analysis.

A paracentesis yielded 7.8 L of clear‐yellow fluid and improvement in his abdominal discomfort. Analysis showed 224 total nucleated cells/L with 2% neutrophils, 57% lymphocytes, and 37% monocytes. Ascites total protein was 3.8 g/dL and glucose was 55 mg/dL. Gram stain and culture were negative, and cytology was negative for malignancy but showed lymphocytes, plasma cells, monocytes, and reactive mesothelial cells interpreted as consistent with chronic inflammation. The serum‐ascites albumin gradient (SAAG) was not obtained.

With a low leukocyte count and a paucity of neutrophils, this is not SBP. The ascites fluid did not have a chylous appearance. The SAAG, which can distinguish between portal hypertensive and nonportal hypertensive causes for ascites using a cutoff of 1.1 g/dL, was not done. The total protein was high, arguing against cirrhosis. High protein ascites with a high SAAG would suggest a posthepatic source of portal hypertension (eg, Budd‐Chiari syndrome, constrictive pericarditis). High protein ascites with a low SAAG would suggest an inflammatory or malignant source of ascites. The relative lymphocytosis in the ascites fluid suggests an inflammatory process, but is a nonspecific finding. The negative cytology does not completely exclude a malignancy, but given the absence of findings on the CT, malignant ascites is less likely.

Three months before admission, the patient underwent a repeat large‐volume paracentesis and a liver biopsy. The biopsy showed ectopic portal vein branches consistent with hepatoportal sclerosis, but no actual sclerosis was identified. The pathologist concluded that the findings suggested noncirrhotic portal hypertension due to a vascular in‐flow abnormality. Abdominal ultrasound with Doppler was unremarkable other than slightly increased echogenicity of the liver. Magnetic resonance (MR) angiogram showed narrowing of the intra‐abdominal IVC at the level of the diaphragm. Because of concern that hepatic congestion from high pressures in the narrowed IVC was leading to poor vascular inflow as suggested by the biopsy findings, an inferior vena cavagram was performed. This study was normal, although no transhepatic pressure measurements were obtained. Three stool specimens and 2 urine specimens were negative for parasites. The patient required repeat large‐volume paracenteses monthly. SBP was again ruled out, but no other diagnostic labs were obtained. He had anorexia with poor oral intake each time his abdomen became distended.

The patient was started on furosemide 1 month prior to admission to the hospital but had only a slight improvement in the ascites. His other medications included insulin, tamsulosin, and hydrocodone‐acetaminophen. Five days prior to admission, he underwent a diagnostic laparoscopy, which showed only ascites and small adhesions to the anterior abdominal wall. There was no visual evidence of malignancy, and the surgeon commented that the liver was normal. No additional biopsies were obtained.

The liver biopsy findings could be seen in noncirrhotic portal hypertension, although this diagnosis would be unlikely without splenomegaly, varices, or other signs of portal hypertension. However, 2 possible etiologies for noncirrhotic portal hypertension in this patient would be hepatic congestion from the narrowed IVC (although the normal IVC study argues against this) and hepatic schistosomiasis. Schistosomiasis is an important cause of noncirrhotic portal hypertension in endemic areas like this patient's country of origin, but the negative stool and urine studies, combined with the lack of granulomas or fibrosis seen on biopsy, make this condition unlikely.

Systemic amyloidosis (primary or secondary) could also be a cause of ascites and could present with multiorgan involvement (diarrhea and nephrotic syndrome). Amyloid deposits would have probably been seen in the liver biopsy, if present, but may not have been apparent unless specific stains (Congo red) were performed.

Evaluation for systemic, inflammatory autoimmune processes is indicated. Serum autoantibodies (anti‐nuclear antibody [ANA] and extractable nuclear antigens), and a serum and 24‐hour urine protein electrophoresis would be appropriate diagnostic tests. Peritoneal biopsies would have been helpful to assess for serosal diseases.

The patient subsequently developed acute right‐sided abdominal pain requiring urgent evaluation and admission to the hospital. He was initially assessed by a general surgeon, who found no evidence of postoperative complications. His temperature was 36.7C, blood pressure 105/64, heart rate 82, respiratory rate 16, and oxygen saturation 97% on room air. He appeared chronically ill, but he was in no distress and he had a normal mental status. Cardiac exam was normal except for mild jugular venous distension. He had mild bibasilar lung crackles. His abdomen was distended with superficial abdominal tenderness and a fluid wave, but he had normal bowel sounds and no peritoneal signs. He had mild scrotal edema but no peripheral edema. Joint exam did not suggest synovitis and there were no rashes or oral ulcers. Lactate was 0.9 mmol/L (normal 0.62.3), albumin was 2.6 g/dL, and prealbumin was 9 mg/dL (normal 1938). Erythrocyte sedimentation rate and C‐reactive protein were 46 mm/hour (normal <22) and 33.1 mg/L (normal 8), respectively. He had a normocytic anemia and leukopenia. Liver tests and routine chemistries were normal. Serum protein electrophoresis indicated no monoclonal protein. Complete 24‐hour urine collection showed 1.2 g of protein (normal <102 mg). Paracentesis of 3.4 L demonstrated 227 total nucleated cells/L with 2% neutrophils. Following the fluid removal, he had improvement in his pain, which he felt was related to the ascites rather than the recent surgery. Ascites total protein was 3.9 g/dL and ascites albumin was 1.7 g/dL. Ascites culture was negative for infection. Serum Schistosoma immunoglobulin G (IgG) antibody was positive at 3.53 (normal <1.00).

Further history revealed prior episodes of polyarticular joint pain and swelling in his hands and knees 5 years before admission. At that time, he reported a diffuse, pruritic, papular body rash. In addition, he noticed that his fingertips and toes turned white with cold exposure.

Importantly, surgical and infectious complications have been excluded. High protein ascites with a low SAAG of 0.9 suggests an inflammatory source of ascites. The follow‐up clinical data (arthritis, normocytic anemia, leukopenia, rash, Raynaud's phenomenon) suggest a systemic inflammatory syndrome such as SLE, with accompanying serositis. Serologic testing for autoantibodies would be recommended. Peritoneal biopsies, if obtained, may have demonstrated chronic, inflammatory infiltrate (nonspecific) or leukocytoclastic vasculitis (strongly supportive).

ANA enzyme immunoassay was >12 U (normal 1.0 U). Extractable nuclear antigens revealed positive autoantibodies for anti‐SSA, anti‐SSB, and anti‐ribosomal P. Moreover, double‐stranded DNA IgG antibody was 120 IU/mL (normal <30 IU/mL) and C3, C4, and total complement levels were low.

The clinical data support a diagnosis of SLE with serositis. Treatment of the underlying connective tissue disease will typically result in resolution of the ascites; diuretic therapy is generally ineffective.

In consultation with rheumatology and gastroenterology specialists, the diagnosis of SLE was made based on criteria of serositis, persistent leukopenia, arthritis, renal disease (proteinuria), positive ANA, elevated ds‐DNA antibodies, and hypocomplementemia. MR imaging of the abdominal vasculature demonstrated no evidence of vasculitis. The patient was given intravenous methylprednisolone 1 g daily for 3 days followed by high‐dose oral corticosteroids with a gradual taper. He was also started on mycophenolate mofetil as a steroid‐sparing medication (which was later changed to leflunomide due to persistent leukopenia) and hydroxychloroquine. His isolated positive Schistosoma IgG antibody in the absence of other findings was consistent with past exposure or infection. The infectious disease specialist felt there was no evidence of active schistosomiasis, but recommended treatment with a single dose of praziquantel due to the potential benefit with low risk of side effects. The patient had ongoing improvement following dismissal. He had 1 additional paracentesis of 4.1 L, 10 days after his hospitalization, and his ascites and proteinuria resolved. At the 5‐year follow‐up visit, there had been no recurrence of abdominal ascites or abdominal pain. He remains on low‐dose prednisone at 5 mg daily, leflunomide, and hydroxychloroquine.

COMMENTARY

This patient had recurrent ascites with 29.6 L removed over the 4 months prior to admission and an additional 3.4 L during his hospitalization. His outpatient providers initially considered a portal hypertensive etiology of his ascites due to his history of HBV and prior alcohol use. They also appropriately investigated for a possible infectious process. They next directed their evaluation toward the liver biopsy findings, which raised concern for a vascular inflow abnormality. However, the evaluation could have been performed more rapidly and far more cost‐efficiently had a diagnostic paracentesis with calculation of the SAAG been performed early in the evaluation.

The SAAG, which was first described in 1983 by Par and colleagues, is a parameter reflecting the oncotic pressure gradient between the vascular bed and the interstitial splanchnic or ascitic fluid. [1] In the classic study by Runyon and colleagues, a SAAG difference of 1.1 g/dL correctly differentiated causes of ascites due to portal hypertension from those that were not due to portal hypertension 96.7% of the time. [2] Conditions such as nephrotic syndrome, peritoneal carcinomatosis, and serositis (lupus peritonitis) can cause ascites in patients without portal hypertension.

Serositis in the form of pleuritis and/or pericarditis is a common feature of SLE, and ascites has been described in 8% to 11% of SLE patients.[3] However, massive ascites due to lupus peritonitis as a presenting symptom is rare.[4] More common causes of ascites in the setting of SLE include nephrotic syndrome, heart failure, protein‐losing enteropathy, constrictive pericarditis, Budd‐Chiari syndrome, indolent infections such as tuberculosis, and chylous ascites.[5, 6, 7] Of note, lupus peritonitis may be chronic or acute. Chronic ascites develops insidiously with few manifestations of active lupus and may be painless, whereas ascites from acute lupus peritonitis typically develops rapidly and presents with acute abdominal pain and other signs of increased lupus activity.[3, 5, 6, 8, 9]

Ascites from lupus peritonitis may be due to marked serosal exudative accumulation with reduced absorptive capacity in the peritoneum.[3, 4, 10] Other possible causes include peritoneal inflammation from deposition of immune complexes or vasculitis of peritoneal vessels and visceral serous membranes.[4, 9, 11] Although subserosal and submucosal vasculitis have been found in acute ascites, chronic ascites may be related to scarring from vasculitis and serosal inflammation leading to poor venous and lymph drainage.[9] Ascitic fluid characteristics from lupus peritonitis include a SAAG <1.1, presence of white blood cells anywhere in a broad range from 10 to 1630/L, and a range of fluid protein from 3.4 to 4.7 mg/dL.[3] Although not tested in this patient, findings of low complement levels, positive ANA, and elevated anti‐DNA antibody in the ascitic fluid would be supportive of lupus peritonitis, but not specific.[5, 9, 12] Lupus erythematosus cells are occasionally found in the ascitic fluid, but do not rule out other causes of ascites.[9] On retrospective analysis, lupus erythematosus cells were not seen in this patient's pathology specimens.

Treatment of lupus peritonitis and ascites is with high‐dose glucocorticoid therapy, but many patients may need a second immunosuppressant, possibly because of impaired peritoneal circulation from chronic inflammation leading to decreased drug delivery.[13, 14] Chronic ascites may be recalcitrant to systemic glucocorticoids,[3] so a possible alternative therapy is intraperitoneal injection of triamcinolone, which successfully treated massive ascites in a patient who did not respond to oral glucocorticoid treatment.[13] Although ascites may be refractory in some patients, those with chronic lupus peritonitis can generally achieve remission, yet the overall prognosis depends on the presence and severity of multiorgan involvement from SLE. As with any SLE patient, there are also risks of infection from immunosuppression and increased cardiovascular risks.

This patient's evaluation and treatment could have been expedited if he had undergone a paracenteses with determination of the SAAG early in his workup. It is not known why the SAAG was not obtained despite multiple outpatient visits and paracenteses, his history of HBV, and prior alcohol use. This may have been simply an unfortunate oversight. Alternatively, it may have been that his outpatient providers focused on tantalizing clues such as his country of origin, which led to concern for schistosomiasis, and the biopsy findings suggestive of a vascular inflow abnormality that led to further extensive testing. In so doing, the clinicians committed several diagnostic errors, including multiple alternatives bias, anchoring, and confirmation bias.[15] As a result, the patient accrued excess charges of $64,000 from multiple tests, laparoscopic surgery, and 2 hospitalizations. This case highlights how cognitive errors introduce costly variability into patient care, especially when a simple and accurate test is at the beginning of the decision tree.

CLINICAL TEACHING POINTS

  1. Diagnostic paracentesis, with calculation of the serum‐ascites albumin gradient, should be the first test in the workup for ascites and can distinguish portal hypertensive causes from nonportal hypertensive causes.
  2. Ascites related to SLE can be acute or chronic and caused by bowel infarction, perforation, pancreatitis, mesenteric vasculitis, nephrotic syndrome, heart failure, protein‐losing enteropathy, constrictive pericarditis, lupus peritonitis, Budd‐Chiari syndrome, or serositis (lupus peritonitis).
  3. Ascites caused by lupus peritonitis is rare. Once treated, management should be directed toward keeping the SLE in remission.

ACKNOWLEDGMENTS

Disclosure: Nothing to report.

Files
References
  1. Paré P, Talbot J, Hoefs JC. Serum‐ascites albumin concentration gradient: a physiologic approach to the differential diagnosis of ascites. Gastroenterology. 1983;85(2):240244.
  2. Runyon BA, Montano AA, Akriviadis EA, et al. The serum‐ascites albumin gradient is superior to the exudate‐transudate concept in the differential diagnosis of ascites. Ann Intern Med. 1992;117:215220.
  3. Forouhar‐Graff H, Dennis‐Yawingu KA, Parke AL. Insidious onset of massive painless ascites as initial manifestation of systemic lupus erythematosus. Lupus. 2011;20:754757.
  4. Weinstein JP, Noyer CM. Rapid onset of massive ascites as the initial presentation of systemic lupus erythematosus. Am J Gastroenterol. 2000;95:302303.
  5. Ebert EC, Hagspiel KD. Gastrointestinal and hepatic manifestations of systemic lupus erythematosus. J Clin Gastroenterol. 2011;45:436441.
  6. Prasad S, Abujam B, Lawrence A, Aggarwal A. Massive ascites as a presenting feature of lupus. Int J Rheum Dis. 2012;15:e15e16.
  7. Lee CK, Han JM, Lee KN, et al. Concurrent occurrence of chylothorax, chylous ascites, and protein‐losing enteropathy in systemic lupus erythematosus. J Rheumatol. 2002;29:13301333.
  8. Richer O, Ulinski T, Lemelle I, et al. Abdominal manifestations in childhood‐onset systemic lupus erythematosus. Ann Rheum Dis. 2007;66:174178.
  9. Schousboe JT, Koch AE, Chang RW. Chronic lupus peritonitis with ascites: review of the literature with a case report. Semin Arthritis Rheum. 1988;18:121126.
  10. Salomon P, Mayer L. Nonhepatic Gastrointestinal Manifestations of Systemic Lupus Erythematosus. London, United Kingdom: Churchill Livingstone; 1987:747760.
  11. Pott Júnior H, Neto AA, Teixeira MAB, Provenza JR. Ascites due to lupus peritonitis: a rare form of onset of systemic lupus erythematosus. Rev Bras Reumatol. 2012;52(1):113119.
  12. Trock D, Volnea A, Wolk J, Majoros A. New‐onset lupus presenting as serositis in an 80‐year‐old woman: does a high‐titer ANA in pleural, pericardial, or peritoneal fluid help confirm the diagnosis? J Clin Rheum.2005:11(5):292293.
  13. Zhou QG, Yang XB, Hou FF, Zhang X. Successful treatment of massive ascites with intraperitoneal administration of a steroid in a case of systemic lupus erythematosus. Lupus. 2009;18:740742.
  14. Ito H, Nanamiya W, Kuroda N, et al. Chronic lupus peritonitis with massive ascites at elderly onset: case report and review of the literature. Intern Med. 2002;41:10561061.
  15. Croskerry P. The Importance of cognitive errors in diagnosis and strategies to minimize them. Acad Med. 2003;78:775780.
Article PDF
Issue
Journal of Hospital Medicine - 10(2)
Page Number
112-115
Sections
Files
Files
Article PDF
Article PDF

A 40‐year‐old Sudanese man was admitted due to worsening abdominal pain with recurrent ascites. He had a history of hepatitis B (HBV) infection and diabetes. He previously drank 3 beers per day on the weekends, but he had not consumed alcohol in over a year. He was born in Sudan but lived in Egypt most of his adult life; he immigrated to the United States 6 years previously. He was hospitalized out of state 9 months ago for a swollen abdomen and underwent an exploratory laparotomy that reportedly was unremarkable except for ascites.

Portal hypertension due to liver disease is the most common cause of ascites. This patient has a known risk factor for liver disease (history of HBV infection). Although his reported alcohol consumption is low, there is a synergistic effect on liver injury in the setting of chronic hepatitis. Abdominal pain in the setting of ascites needs to be urgently evaluated to exclude spontaneous bacterial peritonitis (SBP). Also, because chronic HBV infection is the major risk factor for hepatocellular carcinoma in the world, malignant ascites is in the differential. Hepatic vascular thrombosis and tuberculous peritonitis (given the patient's country of origin and travel history) also should be considered. The most appropriate initial test would be a diagnostic paracentesis to support or exclude the presence of SBP and direct the evaluation toward liver disease or other less‐common causes of ascites.

The patient was seen as an outpatient 5 months prior to admission with transient fever and joint pains. Laboratory studies at that visit were notable for a serum albumin of 3.2 g/dL (normal 3.55), 2.4 g of predicted 24‐hour protein on urinalysis (normal <30 mg per 24 hours), creatinine of 0.5 mg/dL (normal 0.81.3), and a positive hepatitis B surface antibody. The working diagnosis was a nonspecific viral syndrome and his symptoms resolved without treatment. One month later, he developed ascites and mild lower extremity edema. Additional laboratory studies at that time showed a normocytic anemia with hemoglobin 11.7 g/dL (normal 13.517.5) and leukopenia with white blood cell count of 2.4 109/L (normal 3.510.5), neutrophil count of 1.45 109/L (normal 1.77.0), and lymphocyte count of 0.58 109/L (normal 0.902.90). Transaminases, serum bilirubin, prothrombin time, alpha fetoprotein, and peripheral blood smear were normal. Human immunodeficiency virus antibody screen and QuantiFERON‐TB assay were negative. Hemoglobin A1c was 6.2% (normal 4.06.0). Repeat urinalysis demonstrated 883 mg of predicted 24‐hour protein. Computed tomography (CT) of the abdomen showed a large amount of intra‐abdominal ascites; the liver and spleen were normal, and there were no varices or other evidence of portal hypertension. Echocardiogram was normal except for a small inferior vena cava (IVC) and a mildly increased right ventricular systolic pressure of 32 mm Hg (systolic blood pressure 98 mm Hg). Due to the indeterminate cause for the patient's ascites, referral was made for gastroenterology evaluation with consideration for a paracentesis.

Cirrhotic ascites seems less likely. Postsinusoidal causes of portal hypertension (eg, cardiomyopathy) are also less likely given the absence of suggestive findings on echocardiography. Malignant ascites also appears less probable in the absence of suggestive findings such as mass lesions, lymphadenopathy, or peritoneal carcinomatosis on CT imaging. The suspicion for tuberculous peritonitis is lower with the negative QuantiFERON‐TB test. Hypoalbuminemia, normocytic anemia, leukopenia, and proteinuria all suggest a systemic inflammatory condition (eg, systemic lupus erythematosus [SLE]) with inflammatory serositis causing ascites). Nephrotic syndrome can cause hypoalbuminemia, edema, and ascites, but his total urine protein losses of <3.5 grams per 24 hours are not in keeping with this diagnosis. Other uncommon causes of ascites such as chylous ascites have not yet been excluded. The most appropriate next step remains ascitic fluid analysis.

A paracentesis yielded 7.8 L of clear‐yellow fluid and improvement in his abdominal discomfort. Analysis showed 224 total nucleated cells/L with 2% neutrophils, 57% lymphocytes, and 37% monocytes. Ascites total protein was 3.8 g/dL and glucose was 55 mg/dL. Gram stain and culture were negative, and cytology was negative for malignancy but showed lymphocytes, plasma cells, monocytes, and reactive mesothelial cells interpreted as consistent with chronic inflammation. The serum‐ascites albumin gradient (SAAG) was not obtained.

With a low leukocyte count and a paucity of neutrophils, this is not SBP. The ascites fluid did not have a chylous appearance. The SAAG, which can distinguish between portal hypertensive and nonportal hypertensive causes for ascites using a cutoff of 1.1 g/dL, was not done. The total protein was high, arguing against cirrhosis. High protein ascites with a high SAAG would suggest a posthepatic source of portal hypertension (eg, Budd‐Chiari syndrome, constrictive pericarditis). High protein ascites with a low SAAG would suggest an inflammatory or malignant source of ascites. The relative lymphocytosis in the ascites fluid suggests an inflammatory process, but is a nonspecific finding. The negative cytology does not completely exclude a malignancy, but given the absence of findings on the CT, malignant ascites is less likely.

Three months before admission, the patient underwent a repeat large‐volume paracentesis and a liver biopsy. The biopsy showed ectopic portal vein branches consistent with hepatoportal sclerosis, but no actual sclerosis was identified. The pathologist concluded that the findings suggested noncirrhotic portal hypertension due to a vascular in‐flow abnormality. Abdominal ultrasound with Doppler was unremarkable other than slightly increased echogenicity of the liver. Magnetic resonance (MR) angiogram showed narrowing of the intra‐abdominal IVC at the level of the diaphragm. Because of concern that hepatic congestion from high pressures in the narrowed IVC was leading to poor vascular inflow as suggested by the biopsy findings, an inferior vena cavagram was performed. This study was normal, although no transhepatic pressure measurements were obtained. Three stool specimens and 2 urine specimens were negative for parasites. The patient required repeat large‐volume paracenteses monthly. SBP was again ruled out, but no other diagnostic labs were obtained. He had anorexia with poor oral intake each time his abdomen became distended.

The patient was started on furosemide 1 month prior to admission to the hospital but had only a slight improvement in the ascites. His other medications included insulin, tamsulosin, and hydrocodone‐acetaminophen. Five days prior to admission, he underwent a diagnostic laparoscopy, which showed only ascites and small adhesions to the anterior abdominal wall. There was no visual evidence of malignancy, and the surgeon commented that the liver was normal. No additional biopsies were obtained.

The liver biopsy findings could be seen in noncirrhotic portal hypertension, although this diagnosis would be unlikely without splenomegaly, varices, or other signs of portal hypertension. However, 2 possible etiologies for noncirrhotic portal hypertension in this patient would be hepatic congestion from the narrowed IVC (although the normal IVC study argues against this) and hepatic schistosomiasis. Schistosomiasis is an important cause of noncirrhotic portal hypertension in endemic areas like this patient's country of origin, but the negative stool and urine studies, combined with the lack of granulomas or fibrosis seen on biopsy, make this condition unlikely.

Systemic amyloidosis (primary or secondary) could also be a cause of ascites and could present with multiorgan involvement (diarrhea and nephrotic syndrome). Amyloid deposits would have probably been seen in the liver biopsy, if present, but may not have been apparent unless specific stains (Congo red) were performed.

Evaluation for systemic, inflammatory autoimmune processes is indicated. Serum autoantibodies (anti‐nuclear antibody [ANA] and extractable nuclear antigens), and a serum and 24‐hour urine protein electrophoresis would be appropriate diagnostic tests. Peritoneal biopsies would have been helpful to assess for serosal diseases.

The patient subsequently developed acute right‐sided abdominal pain requiring urgent evaluation and admission to the hospital. He was initially assessed by a general surgeon, who found no evidence of postoperative complications. His temperature was 36.7C, blood pressure 105/64, heart rate 82, respiratory rate 16, and oxygen saturation 97% on room air. He appeared chronically ill, but he was in no distress and he had a normal mental status. Cardiac exam was normal except for mild jugular venous distension. He had mild bibasilar lung crackles. His abdomen was distended with superficial abdominal tenderness and a fluid wave, but he had normal bowel sounds and no peritoneal signs. He had mild scrotal edema but no peripheral edema. Joint exam did not suggest synovitis and there were no rashes or oral ulcers. Lactate was 0.9 mmol/L (normal 0.62.3), albumin was 2.6 g/dL, and prealbumin was 9 mg/dL (normal 1938). Erythrocyte sedimentation rate and C‐reactive protein were 46 mm/hour (normal <22) and 33.1 mg/L (normal 8), respectively. He had a normocytic anemia and leukopenia. Liver tests and routine chemistries were normal. Serum protein electrophoresis indicated no monoclonal protein. Complete 24‐hour urine collection showed 1.2 g of protein (normal <102 mg). Paracentesis of 3.4 L demonstrated 227 total nucleated cells/L with 2% neutrophils. Following the fluid removal, he had improvement in his pain, which he felt was related to the ascites rather than the recent surgery. Ascites total protein was 3.9 g/dL and ascites albumin was 1.7 g/dL. Ascites culture was negative for infection. Serum Schistosoma immunoglobulin G (IgG) antibody was positive at 3.53 (normal <1.00).

Further history revealed prior episodes of polyarticular joint pain and swelling in his hands and knees 5 years before admission. At that time, he reported a diffuse, pruritic, papular body rash. In addition, he noticed that his fingertips and toes turned white with cold exposure.

Importantly, surgical and infectious complications have been excluded. High protein ascites with a low SAAG of 0.9 suggests an inflammatory source of ascites. The follow‐up clinical data (arthritis, normocytic anemia, leukopenia, rash, Raynaud's phenomenon) suggest a systemic inflammatory syndrome such as SLE, with accompanying serositis. Serologic testing for autoantibodies would be recommended. Peritoneal biopsies, if obtained, may have demonstrated chronic, inflammatory infiltrate (nonspecific) or leukocytoclastic vasculitis (strongly supportive).

ANA enzyme immunoassay was >12 U (normal 1.0 U). Extractable nuclear antigens revealed positive autoantibodies for anti‐SSA, anti‐SSB, and anti‐ribosomal P. Moreover, double‐stranded DNA IgG antibody was 120 IU/mL (normal <30 IU/mL) and C3, C4, and total complement levels were low.

The clinical data support a diagnosis of SLE with serositis. Treatment of the underlying connective tissue disease will typically result in resolution of the ascites; diuretic therapy is generally ineffective.

In consultation with rheumatology and gastroenterology specialists, the diagnosis of SLE was made based on criteria of serositis, persistent leukopenia, arthritis, renal disease (proteinuria), positive ANA, elevated ds‐DNA antibodies, and hypocomplementemia. MR imaging of the abdominal vasculature demonstrated no evidence of vasculitis. The patient was given intravenous methylprednisolone 1 g daily for 3 days followed by high‐dose oral corticosteroids with a gradual taper. He was also started on mycophenolate mofetil as a steroid‐sparing medication (which was later changed to leflunomide due to persistent leukopenia) and hydroxychloroquine. His isolated positive Schistosoma IgG antibody in the absence of other findings was consistent with past exposure or infection. The infectious disease specialist felt there was no evidence of active schistosomiasis, but recommended treatment with a single dose of praziquantel due to the potential benefit with low risk of side effects. The patient had ongoing improvement following dismissal. He had 1 additional paracentesis of 4.1 L, 10 days after his hospitalization, and his ascites and proteinuria resolved. At the 5‐year follow‐up visit, there had been no recurrence of abdominal ascites or abdominal pain. He remains on low‐dose prednisone at 5 mg daily, leflunomide, and hydroxychloroquine.

COMMENTARY

This patient had recurrent ascites with 29.6 L removed over the 4 months prior to admission and an additional 3.4 L during his hospitalization. His outpatient providers initially considered a portal hypertensive etiology of his ascites due to his history of HBV and prior alcohol use. They also appropriately investigated for a possible infectious process. They next directed their evaluation toward the liver biopsy findings, which raised concern for a vascular inflow abnormality. However, the evaluation could have been performed more rapidly and far more cost‐efficiently had a diagnostic paracentesis with calculation of the SAAG been performed early in the evaluation.

The SAAG, which was first described in 1983 by Par and colleagues, is a parameter reflecting the oncotic pressure gradient between the vascular bed and the interstitial splanchnic or ascitic fluid. [1] In the classic study by Runyon and colleagues, a SAAG difference of 1.1 g/dL correctly differentiated causes of ascites due to portal hypertension from those that were not due to portal hypertension 96.7% of the time. [2] Conditions such as nephrotic syndrome, peritoneal carcinomatosis, and serositis (lupus peritonitis) can cause ascites in patients without portal hypertension.

Serositis in the form of pleuritis and/or pericarditis is a common feature of SLE, and ascites has been described in 8% to 11% of SLE patients.[3] However, massive ascites due to lupus peritonitis as a presenting symptom is rare.[4] More common causes of ascites in the setting of SLE include nephrotic syndrome, heart failure, protein‐losing enteropathy, constrictive pericarditis, Budd‐Chiari syndrome, indolent infections such as tuberculosis, and chylous ascites.[5, 6, 7] Of note, lupus peritonitis may be chronic or acute. Chronic ascites develops insidiously with few manifestations of active lupus and may be painless, whereas ascites from acute lupus peritonitis typically develops rapidly and presents with acute abdominal pain and other signs of increased lupus activity.[3, 5, 6, 8, 9]

Ascites from lupus peritonitis may be due to marked serosal exudative accumulation with reduced absorptive capacity in the peritoneum.[3, 4, 10] Other possible causes include peritoneal inflammation from deposition of immune complexes or vasculitis of peritoneal vessels and visceral serous membranes.[4, 9, 11] Although subserosal and submucosal vasculitis have been found in acute ascites, chronic ascites may be related to scarring from vasculitis and serosal inflammation leading to poor venous and lymph drainage.[9] Ascitic fluid characteristics from lupus peritonitis include a SAAG <1.1, presence of white blood cells anywhere in a broad range from 10 to 1630/L, and a range of fluid protein from 3.4 to 4.7 mg/dL.[3] Although not tested in this patient, findings of low complement levels, positive ANA, and elevated anti‐DNA antibody in the ascitic fluid would be supportive of lupus peritonitis, but not specific.[5, 9, 12] Lupus erythematosus cells are occasionally found in the ascitic fluid, but do not rule out other causes of ascites.[9] On retrospective analysis, lupus erythematosus cells were not seen in this patient's pathology specimens.

Treatment of lupus peritonitis and ascites is with high‐dose glucocorticoid therapy, but many patients may need a second immunosuppressant, possibly because of impaired peritoneal circulation from chronic inflammation leading to decreased drug delivery.[13, 14] Chronic ascites may be recalcitrant to systemic glucocorticoids,[3] so a possible alternative therapy is intraperitoneal injection of triamcinolone, which successfully treated massive ascites in a patient who did not respond to oral glucocorticoid treatment.[13] Although ascites may be refractory in some patients, those with chronic lupus peritonitis can generally achieve remission, yet the overall prognosis depends on the presence and severity of multiorgan involvement from SLE. As with any SLE patient, there are also risks of infection from immunosuppression and increased cardiovascular risks.

This patient's evaluation and treatment could have been expedited if he had undergone a paracenteses with determination of the SAAG early in his workup. It is not known why the SAAG was not obtained despite multiple outpatient visits and paracenteses, his history of HBV, and prior alcohol use. This may have been simply an unfortunate oversight. Alternatively, it may have been that his outpatient providers focused on tantalizing clues such as his country of origin, which led to concern for schistosomiasis, and the biopsy findings suggestive of a vascular inflow abnormality that led to further extensive testing. In so doing, the clinicians committed several diagnostic errors, including multiple alternatives bias, anchoring, and confirmation bias.[15] As a result, the patient accrued excess charges of $64,000 from multiple tests, laparoscopic surgery, and 2 hospitalizations. This case highlights how cognitive errors introduce costly variability into patient care, especially when a simple and accurate test is at the beginning of the decision tree.

CLINICAL TEACHING POINTS

  1. Diagnostic paracentesis, with calculation of the serum‐ascites albumin gradient, should be the first test in the workup for ascites and can distinguish portal hypertensive causes from nonportal hypertensive causes.
  2. Ascites related to SLE can be acute or chronic and caused by bowel infarction, perforation, pancreatitis, mesenteric vasculitis, nephrotic syndrome, heart failure, protein‐losing enteropathy, constrictive pericarditis, lupus peritonitis, Budd‐Chiari syndrome, or serositis (lupus peritonitis).
  3. Ascites caused by lupus peritonitis is rare. Once treated, management should be directed toward keeping the SLE in remission.

ACKNOWLEDGMENTS

Disclosure: Nothing to report.

A 40‐year‐old Sudanese man was admitted due to worsening abdominal pain with recurrent ascites. He had a history of hepatitis B (HBV) infection and diabetes. He previously drank 3 beers per day on the weekends, but he had not consumed alcohol in over a year. He was born in Sudan but lived in Egypt most of his adult life; he immigrated to the United States 6 years previously. He was hospitalized out of state 9 months ago for a swollen abdomen and underwent an exploratory laparotomy that reportedly was unremarkable except for ascites.

Portal hypertension due to liver disease is the most common cause of ascites. This patient has a known risk factor for liver disease (history of HBV infection). Although his reported alcohol consumption is low, there is a synergistic effect on liver injury in the setting of chronic hepatitis. Abdominal pain in the setting of ascites needs to be urgently evaluated to exclude spontaneous bacterial peritonitis (SBP). Also, because chronic HBV infection is the major risk factor for hepatocellular carcinoma in the world, malignant ascites is in the differential. Hepatic vascular thrombosis and tuberculous peritonitis (given the patient's country of origin and travel history) also should be considered. The most appropriate initial test would be a diagnostic paracentesis to support or exclude the presence of SBP and direct the evaluation toward liver disease or other less‐common causes of ascites.

The patient was seen as an outpatient 5 months prior to admission with transient fever and joint pains. Laboratory studies at that visit were notable for a serum albumin of 3.2 g/dL (normal 3.55), 2.4 g of predicted 24‐hour protein on urinalysis (normal <30 mg per 24 hours), creatinine of 0.5 mg/dL (normal 0.81.3), and a positive hepatitis B surface antibody. The working diagnosis was a nonspecific viral syndrome and his symptoms resolved without treatment. One month later, he developed ascites and mild lower extremity edema. Additional laboratory studies at that time showed a normocytic anemia with hemoglobin 11.7 g/dL (normal 13.517.5) and leukopenia with white blood cell count of 2.4 109/L (normal 3.510.5), neutrophil count of 1.45 109/L (normal 1.77.0), and lymphocyte count of 0.58 109/L (normal 0.902.90). Transaminases, serum bilirubin, prothrombin time, alpha fetoprotein, and peripheral blood smear were normal. Human immunodeficiency virus antibody screen and QuantiFERON‐TB assay were negative. Hemoglobin A1c was 6.2% (normal 4.06.0). Repeat urinalysis demonstrated 883 mg of predicted 24‐hour protein. Computed tomography (CT) of the abdomen showed a large amount of intra‐abdominal ascites; the liver and spleen were normal, and there were no varices or other evidence of portal hypertension. Echocardiogram was normal except for a small inferior vena cava (IVC) and a mildly increased right ventricular systolic pressure of 32 mm Hg (systolic blood pressure 98 mm Hg). Due to the indeterminate cause for the patient's ascites, referral was made for gastroenterology evaluation with consideration for a paracentesis.

Cirrhotic ascites seems less likely. Postsinusoidal causes of portal hypertension (eg, cardiomyopathy) are also less likely given the absence of suggestive findings on echocardiography. Malignant ascites also appears less probable in the absence of suggestive findings such as mass lesions, lymphadenopathy, or peritoneal carcinomatosis on CT imaging. The suspicion for tuberculous peritonitis is lower with the negative QuantiFERON‐TB test. Hypoalbuminemia, normocytic anemia, leukopenia, and proteinuria all suggest a systemic inflammatory condition (eg, systemic lupus erythematosus [SLE]) with inflammatory serositis causing ascites). Nephrotic syndrome can cause hypoalbuminemia, edema, and ascites, but his total urine protein losses of <3.5 grams per 24 hours are not in keeping with this diagnosis. Other uncommon causes of ascites such as chylous ascites have not yet been excluded. The most appropriate next step remains ascitic fluid analysis.

A paracentesis yielded 7.8 L of clear‐yellow fluid and improvement in his abdominal discomfort. Analysis showed 224 total nucleated cells/L with 2% neutrophils, 57% lymphocytes, and 37% monocytes. Ascites total protein was 3.8 g/dL and glucose was 55 mg/dL. Gram stain and culture were negative, and cytology was negative for malignancy but showed lymphocytes, plasma cells, monocytes, and reactive mesothelial cells interpreted as consistent with chronic inflammation. The serum‐ascites albumin gradient (SAAG) was not obtained.

With a low leukocyte count and a paucity of neutrophils, this is not SBP. The ascites fluid did not have a chylous appearance. The SAAG, which can distinguish between portal hypertensive and nonportal hypertensive causes for ascites using a cutoff of 1.1 g/dL, was not done. The total protein was high, arguing against cirrhosis. High protein ascites with a high SAAG would suggest a posthepatic source of portal hypertension (eg, Budd‐Chiari syndrome, constrictive pericarditis). High protein ascites with a low SAAG would suggest an inflammatory or malignant source of ascites. The relative lymphocytosis in the ascites fluid suggests an inflammatory process, but is a nonspecific finding. The negative cytology does not completely exclude a malignancy, but given the absence of findings on the CT, malignant ascites is less likely.

Three months before admission, the patient underwent a repeat large‐volume paracentesis and a liver biopsy. The biopsy showed ectopic portal vein branches consistent with hepatoportal sclerosis, but no actual sclerosis was identified. The pathologist concluded that the findings suggested noncirrhotic portal hypertension due to a vascular in‐flow abnormality. Abdominal ultrasound with Doppler was unremarkable other than slightly increased echogenicity of the liver. Magnetic resonance (MR) angiogram showed narrowing of the intra‐abdominal IVC at the level of the diaphragm. Because of concern that hepatic congestion from high pressures in the narrowed IVC was leading to poor vascular inflow as suggested by the biopsy findings, an inferior vena cavagram was performed. This study was normal, although no transhepatic pressure measurements were obtained. Three stool specimens and 2 urine specimens were negative for parasites. The patient required repeat large‐volume paracenteses monthly. SBP was again ruled out, but no other diagnostic labs were obtained. He had anorexia with poor oral intake each time his abdomen became distended.

The patient was started on furosemide 1 month prior to admission to the hospital but had only a slight improvement in the ascites. His other medications included insulin, tamsulosin, and hydrocodone‐acetaminophen. Five days prior to admission, he underwent a diagnostic laparoscopy, which showed only ascites and small adhesions to the anterior abdominal wall. There was no visual evidence of malignancy, and the surgeon commented that the liver was normal. No additional biopsies were obtained.

The liver biopsy findings could be seen in noncirrhotic portal hypertension, although this diagnosis would be unlikely without splenomegaly, varices, or other signs of portal hypertension. However, 2 possible etiologies for noncirrhotic portal hypertension in this patient would be hepatic congestion from the narrowed IVC (although the normal IVC study argues against this) and hepatic schistosomiasis. Schistosomiasis is an important cause of noncirrhotic portal hypertension in endemic areas like this patient's country of origin, but the negative stool and urine studies, combined with the lack of granulomas or fibrosis seen on biopsy, make this condition unlikely.

Systemic amyloidosis (primary or secondary) could also be a cause of ascites and could present with multiorgan involvement (diarrhea and nephrotic syndrome). Amyloid deposits would have probably been seen in the liver biopsy, if present, but may not have been apparent unless specific stains (Congo red) were performed.

Evaluation for systemic, inflammatory autoimmune processes is indicated. Serum autoantibodies (anti‐nuclear antibody [ANA] and extractable nuclear antigens), and a serum and 24‐hour urine protein electrophoresis would be appropriate diagnostic tests. Peritoneal biopsies would have been helpful to assess for serosal diseases.

The patient subsequently developed acute right‐sided abdominal pain requiring urgent evaluation and admission to the hospital. He was initially assessed by a general surgeon, who found no evidence of postoperative complications. His temperature was 36.7C, blood pressure 105/64, heart rate 82, respiratory rate 16, and oxygen saturation 97% on room air. He appeared chronically ill, but he was in no distress and he had a normal mental status. Cardiac exam was normal except for mild jugular venous distension. He had mild bibasilar lung crackles. His abdomen was distended with superficial abdominal tenderness and a fluid wave, but he had normal bowel sounds and no peritoneal signs. He had mild scrotal edema but no peripheral edema. Joint exam did not suggest synovitis and there were no rashes or oral ulcers. Lactate was 0.9 mmol/L (normal 0.62.3), albumin was 2.6 g/dL, and prealbumin was 9 mg/dL (normal 1938). Erythrocyte sedimentation rate and C‐reactive protein were 46 mm/hour (normal <22) and 33.1 mg/L (normal 8), respectively. He had a normocytic anemia and leukopenia. Liver tests and routine chemistries were normal. Serum protein electrophoresis indicated no monoclonal protein. Complete 24‐hour urine collection showed 1.2 g of protein (normal <102 mg). Paracentesis of 3.4 L demonstrated 227 total nucleated cells/L with 2% neutrophils. Following the fluid removal, he had improvement in his pain, which he felt was related to the ascites rather than the recent surgery. Ascites total protein was 3.9 g/dL and ascites albumin was 1.7 g/dL. Ascites culture was negative for infection. Serum Schistosoma immunoglobulin G (IgG) antibody was positive at 3.53 (normal <1.00).

Further history revealed prior episodes of polyarticular joint pain and swelling in his hands and knees 5 years before admission. At that time, he reported a diffuse, pruritic, papular body rash. In addition, he noticed that his fingertips and toes turned white with cold exposure.

Importantly, surgical and infectious complications have been excluded. High protein ascites with a low SAAG of 0.9 suggests an inflammatory source of ascites. The follow‐up clinical data (arthritis, normocytic anemia, leukopenia, rash, Raynaud's phenomenon) suggest a systemic inflammatory syndrome such as SLE, with accompanying serositis. Serologic testing for autoantibodies would be recommended. Peritoneal biopsies, if obtained, may have demonstrated chronic, inflammatory infiltrate (nonspecific) or leukocytoclastic vasculitis (strongly supportive).

ANA enzyme immunoassay was >12 U (normal 1.0 U). Extractable nuclear antigens revealed positive autoantibodies for anti‐SSA, anti‐SSB, and anti‐ribosomal P. Moreover, double‐stranded DNA IgG antibody was 120 IU/mL (normal <30 IU/mL) and C3, C4, and total complement levels were low.

The clinical data support a diagnosis of SLE with serositis. Treatment of the underlying connective tissue disease will typically result in resolution of the ascites; diuretic therapy is generally ineffective.

In consultation with rheumatology and gastroenterology specialists, the diagnosis of SLE was made based on criteria of serositis, persistent leukopenia, arthritis, renal disease (proteinuria), positive ANA, elevated ds‐DNA antibodies, and hypocomplementemia. MR imaging of the abdominal vasculature demonstrated no evidence of vasculitis. The patient was given intravenous methylprednisolone 1 g daily for 3 days followed by high‐dose oral corticosteroids with a gradual taper. He was also started on mycophenolate mofetil as a steroid‐sparing medication (which was later changed to leflunomide due to persistent leukopenia) and hydroxychloroquine. His isolated positive Schistosoma IgG antibody in the absence of other findings was consistent with past exposure or infection. The infectious disease specialist felt there was no evidence of active schistosomiasis, but recommended treatment with a single dose of praziquantel due to the potential benefit with low risk of side effects. The patient had ongoing improvement following dismissal. He had 1 additional paracentesis of 4.1 L, 10 days after his hospitalization, and his ascites and proteinuria resolved. At the 5‐year follow‐up visit, there had been no recurrence of abdominal ascites or abdominal pain. He remains on low‐dose prednisone at 5 mg daily, leflunomide, and hydroxychloroquine.

COMMENTARY

This patient had recurrent ascites with 29.6 L removed over the 4 months prior to admission and an additional 3.4 L during his hospitalization. His outpatient providers initially considered a portal hypertensive etiology of his ascites due to his history of HBV and prior alcohol use. They also appropriately investigated for a possible infectious process. They next directed their evaluation toward the liver biopsy findings, which raised concern for a vascular inflow abnormality. However, the evaluation could have been performed more rapidly and far more cost‐efficiently had a diagnostic paracentesis with calculation of the SAAG been performed early in the evaluation.

The SAAG, which was first described in 1983 by Par and colleagues, is a parameter reflecting the oncotic pressure gradient between the vascular bed and the interstitial splanchnic or ascitic fluid. [1] In the classic study by Runyon and colleagues, a SAAG difference of 1.1 g/dL correctly differentiated causes of ascites due to portal hypertension from those that were not due to portal hypertension 96.7% of the time. [2] Conditions such as nephrotic syndrome, peritoneal carcinomatosis, and serositis (lupus peritonitis) can cause ascites in patients without portal hypertension.

Serositis in the form of pleuritis and/or pericarditis is a common feature of SLE, and ascites has been described in 8% to 11% of SLE patients.[3] However, massive ascites due to lupus peritonitis as a presenting symptom is rare.[4] More common causes of ascites in the setting of SLE include nephrotic syndrome, heart failure, protein‐losing enteropathy, constrictive pericarditis, Budd‐Chiari syndrome, indolent infections such as tuberculosis, and chylous ascites.[5, 6, 7] Of note, lupus peritonitis may be chronic or acute. Chronic ascites develops insidiously with few manifestations of active lupus and may be painless, whereas ascites from acute lupus peritonitis typically develops rapidly and presents with acute abdominal pain and other signs of increased lupus activity.[3, 5, 6, 8, 9]

Ascites from lupus peritonitis may be due to marked serosal exudative accumulation with reduced absorptive capacity in the peritoneum.[3, 4, 10] Other possible causes include peritoneal inflammation from deposition of immune complexes or vasculitis of peritoneal vessels and visceral serous membranes.[4, 9, 11] Although subserosal and submucosal vasculitis have been found in acute ascites, chronic ascites may be related to scarring from vasculitis and serosal inflammation leading to poor venous and lymph drainage.[9] Ascitic fluid characteristics from lupus peritonitis include a SAAG <1.1, presence of white blood cells anywhere in a broad range from 10 to 1630/L, and a range of fluid protein from 3.4 to 4.7 mg/dL.[3] Although not tested in this patient, findings of low complement levels, positive ANA, and elevated anti‐DNA antibody in the ascitic fluid would be supportive of lupus peritonitis, but not specific.[5, 9, 12] Lupus erythematosus cells are occasionally found in the ascitic fluid, but do not rule out other causes of ascites.[9] On retrospective analysis, lupus erythematosus cells were not seen in this patient's pathology specimens.

Treatment of lupus peritonitis and ascites is with high‐dose glucocorticoid therapy, but many patients may need a second immunosuppressant, possibly because of impaired peritoneal circulation from chronic inflammation leading to decreased drug delivery.[13, 14] Chronic ascites may be recalcitrant to systemic glucocorticoids,[3] so a possible alternative therapy is intraperitoneal injection of triamcinolone, which successfully treated massive ascites in a patient who did not respond to oral glucocorticoid treatment.[13] Although ascites may be refractory in some patients, those with chronic lupus peritonitis can generally achieve remission, yet the overall prognosis depends on the presence and severity of multiorgan involvement from SLE. As with any SLE patient, there are also risks of infection from immunosuppression and increased cardiovascular risks.

This patient's evaluation and treatment could have been expedited if he had undergone a paracenteses with determination of the SAAG early in his workup. It is not known why the SAAG was not obtained despite multiple outpatient visits and paracenteses, his history of HBV, and prior alcohol use. This may have been simply an unfortunate oversight. Alternatively, it may have been that his outpatient providers focused on tantalizing clues such as his country of origin, which led to concern for schistosomiasis, and the biopsy findings suggestive of a vascular inflow abnormality that led to further extensive testing. In so doing, the clinicians committed several diagnostic errors, including multiple alternatives bias, anchoring, and confirmation bias.[15] As a result, the patient accrued excess charges of $64,000 from multiple tests, laparoscopic surgery, and 2 hospitalizations. This case highlights how cognitive errors introduce costly variability into patient care, especially when a simple and accurate test is at the beginning of the decision tree.

CLINICAL TEACHING POINTS

  1. Diagnostic paracentesis, with calculation of the serum‐ascites albumin gradient, should be the first test in the workup for ascites and can distinguish portal hypertensive causes from nonportal hypertensive causes.
  2. Ascites related to SLE can be acute or chronic and caused by bowel infarction, perforation, pancreatitis, mesenteric vasculitis, nephrotic syndrome, heart failure, protein‐losing enteropathy, constrictive pericarditis, lupus peritonitis, Budd‐Chiari syndrome, or serositis (lupus peritonitis).
  3. Ascites caused by lupus peritonitis is rare. Once treated, management should be directed toward keeping the SLE in remission.

ACKNOWLEDGMENTS

Disclosure: Nothing to report.

References
  1. Paré P, Talbot J, Hoefs JC. Serum‐ascites albumin concentration gradient: a physiologic approach to the differential diagnosis of ascites. Gastroenterology. 1983;85(2):240244.
  2. Runyon BA, Montano AA, Akriviadis EA, et al. The serum‐ascites albumin gradient is superior to the exudate‐transudate concept in the differential diagnosis of ascites. Ann Intern Med. 1992;117:215220.
  3. Forouhar‐Graff H, Dennis‐Yawingu KA, Parke AL. Insidious onset of massive painless ascites as initial manifestation of systemic lupus erythematosus. Lupus. 2011;20:754757.
  4. Weinstein JP, Noyer CM. Rapid onset of massive ascites as the initial presentation of systemic lupus erythematosus. Am J Gastroenterol. 2000;95:302303.
  5. Ebert EC, Hagspiel KD. Gastrointestinal and hepatic manifestations of systemic lupus erythematosus. J Clin Gastroenterol. 2011;45:436441.
  6. Prasad S, Abujam B, Lawrence A, Aggarwal A. Massive ascites as a presenting feature of lupus. Int J Rheum Dis. 2012;15:e15e16.
  7. Lee CK, Han JM, Lee KN, et al. Concurrent occurrence of chylothorax, chylous ascites, and protein‐losing enteropathy in systemic lupus erythematosus. J Rheumatol. 2002;29:13301333.
  8. Richer O, Ulinski T, Lemelle I, et al. Abdominal manifestations in childhood‐onset systemic lupus erythematosus. Ann Rheum Dis. 2007;66:174178.
  9. Schousboe JT, Koch AE, Chang RW. Chronic lupus peritonitis with ascites: review of the literature with a case report. Semin Arthritis Rheum. 1988;18:121126.
  10. Salomon P, Mayer L. Nonhepatic Gastrointestinal Manifestations of Systemic Lupus Erythematosus. London, United Kingdom: Churchill Livingstone; 1987:747760.
  11. Pott Júnior H, Neto AA, Teixeira MAB, Provenza JR. Ascites due to lupus peritonitis: a rare form of onset of systemic lupus erythematosus. Rev Bras Reumatol. 2012;52(1):113119.
  12. Trock D, Volnea A, Wolk J, Majoros A. New‐onset lupus presenting as serositis in an 80‐year‐old woman: does a high‐titer ANA in pleural, pericardial, or peritoneal fluid help confirm the diagnosis? J Clin Rheum.2005:11(5):292293.
  13. Zhou QG, Yang XB, Hou FF, Zhang X. Successful treatment of massive ascites with intraperitoneal administration of a steroid in a case of systemic lupus erythematosus. Lupus. 2009;18:740742.
  14. Ito H, Nanamiya W, Kuroda N, et al. Chronic lupus peritonitis with massive ascites at elderly onset: case report and review of the literature. Intern Med. 2002;41:10561061.
  15. Croskerry P. The Importance of cognitive errors in diagnosis and strategies to minimize them. Acad Med. 2003;78:775780.
References
  1. Paré P, Talbot J, Hoefs JC. Serum‐ascites albumin concentration gradient: a physiologic approach to the differential diagnosis of ascites. Gastroenterology. 1983;85(2):240244.
  2. Runyon BA, Montano AA, Akriviadis EA, et al. The serum‐ascites albumin gradient is superior to the exudate‐transudate concept in the differential diagnosis of ascites. Ann Intern Med. 1992;117:215220.
  3. Forouhar‐Graff H, Dennis‐Yawingu KA, Parke AL. Insidious onset of massive painless ascites as initial manifestation of systemic lupus erythematosus. Lupus. 2011;20:754757.
  4. Weinstein JP, Noyer CM. Rapid onset of massive ascites as the initial presentation of systemic lupus erythematosus. Am J Gastroenterol. 2000;95:302303.
  5. Ebert EC, Hagspiel KD. Gastrointestinal and hepatic manifestations of systemic lupus erythematosus. J Clin Gastroenterol. 2011;45:436441.
  6. Prasad S, Abujam B, Lawrence A, Aggarwal A. Massive ascites as a presenting feature of lupus. Int J Rheum Dis. 2012;15:e15e16.
  7. Lee CK, Han JM, Lee KN, et al. Concurrent occurrence of chylothorax, chylous ascites, and protein‐losing enteropathy in systemic lupus erythematosus. J Rheumatol. 2002;29:13301333.
  8. Richer O, Ulinski T, Lemelle I, et al. Abdominal manifestations in childhood‐onset systemic lupus erythematosus. Ann Rheum Dis. 2007;66:174178.
  9. Schousboe JT, Koch AE, Chang RW. Chronic lupus peritonitis with ascites: review of the literature with a case report. Semin Arthritis Rheum. 1988;18:121126.
  10. Salomon P, Mayer L. Nonhepatic Gastrointestinal Manifestations of Systemic Lupus Erythematosus. London, United Kingdom: Churchill Livingstone; 1987:747760.
  11. Pott Júnior H, Neto AA, Teixeira MAB, Provenza JR. Ascites due to lupus peritonitis: a rare form of onset of systemic lupus erythematosus. Rev Bras Reumatol. 2012;52(1):113119.
  12. Trock D, Volnea A, Wolk J, Majoros A. New‐onset lupus presenting as serositis in an 80‐year‐old woman: does a high‐titer ANA in pleural, pericardial, or peritoneal fluid help confirm the diagnosis? J Clin Rheum.2005:11(5):292293.
  13. Zhou QG, Yang XB, Hou FF, Zhang X. Successful treatment of massive ascites with intraperitoneal administration of a steroid in a case of systemic lupus erythematosus. Lupus. 2009;18:740742.
  14. Ito H, Nanamiya W, Kuroda N, et al. Chronic lupus peritonitis with massive ascites at elderly onset: case report and review of the literature. Intern Med. 2002;41:10561061.
  15. Croskerry P. The Importance of cognitive errors in diagnosis and strategies to minimize them. Acad Med. 2003;78:775780.
Issue
Journal of Hospital Medicine - 10(2)
Issue
Journal of Hospital Medicine - 10(2)
Page Number
112-115
Page Number
112-115
Article Type
Display Headline
The $64,000 question
Display Headline
The $64,000 question
Sections
Article Source
© 2014 Society of Hospital Medicine
Disallow All Ads
Correspondence Location
Address for correspondence and reprint requests: A. Scott Keller, MD, Division of Hospital Internal Medicine, Mayo Clinic, 200 1st Street SW, Rochester, MN 55905; Telephone: 507‐255‐8043; Fax: 507‐255‐9189; E‐mail: [email protected]
Content Gating
Gated (full article locked unless allowed per User)
Gating Strategy
First Peek Free
Article PDF Media
Media Files

In‐Hospital Stroke Alerts

Article Type
Changed
Mon, 01/02/2017 - 19:34
Display Headline
Code stroke: Multicenter experience with in‐hospital stroke alerts

Acute change in neurologic status in a hospitalized patient is an emergency requiring timely coordinated evaluation. To address this need, many hospitals have created a mechanism for in‐hospital stroke alerts utilizing generalized rapid response teams or specialized stroke teams.[1, 2, 3] The common purpose is to quickly diagnose new ischemic stroke within the time window for thrombolytic therapy.

Even when acute change in neurologic status is not due to brain ischemia, it may represent a new metabolic disturbance or reflect developing serious systemic illness. Sepsis, hypoglycemia, cardiac arrhythmia, respiratory failure, severe electrolyte disturbances, seizures, or delirium may first manifest as a change in neurologic status.

Prior research on stroke alerts has largely focused on patients who present from the community to the emergency department (ED).[4, 5, 6, 7, 8] Patients who develop acute neurologic symptoms during hospitalization have different risk factors and exposures compared to patients in the community.[9] This study represents the experience of a multistate quality improvement initiative for in‐hospital stroke. We characterize etiologies for symptoms triggering in‐hospital stroke alerts and thrombolytic treatment for in‐hospital strokes.

PATIENTS AND METHODS

The National Stroke Association's (NSA) initiative, Improving In‐Hospital Stroke Response: A Team‐based Quality Improvement Program, included data collection for all in‐hospital stroke alerts over a 12‐month period.[10] Six Joint Commission certified primary stroke centers from Michigan, South Carolina, Pennsylvania, Colorado, Washington, and North Carolina completed the 1‐year quality improvement initiative. One additional site withdrew from the program after the first quarter and was not included in this analysis. Sites prospectively reported deidentified patient‐level data on all adult in‐hospital stroke alerts from July 2010 to June 2011 to the NSA. At all sites, any provider could activate the in‐hospital stroke response system. Stroke alerts were evaluated by a rapid response team with stroke training. The providers on the stroke rapid response team varied between sites. A nurse with stroke training was 1 of the first responders on the stroke response team at all sites.

The NSA in‐hospital stroke‐alert criteria included the following symptoms occurring in the last 24‐hours, even if they resolved: (1) sudden numbness or weakness of the face, arm or leg, especially on 1 side of the body; (2) sudden confusion, trouble speaking or understanding; (3) sudden trouble seeing in 1 or both eyes; (4) sudden trouble walking, dizziness, loss of balance or coordination; and (5) sudden, severe headache with no known cause. Hospitals reported location, service, age, sex, race, symptoms triggering the stroke alert, free text entry of final clinical diagnosis following the completion of stroke alert evaluation, treatment with intravenous or intra‐arterial/mechanical thrombolysis, and any contraindications to intravenous thrombolysis. We categorized stroke mimics using the responses in the final diagnosis field after the data collection period was complete. Strokes were categorized as ischemic stroke, transient ischemic attack (TIA), or intracranial hemorrhage (intraparenchymal, intraventricular, epidural, subdural, or subarachnoid). Stroke mimics were subdivided according to the categories in Table 1. Lack of certainty in the final diagnosis was handled by creating a category of possible TIA, which includes alternative diagnosis versus TIA or the qualifier possible before TIA. Patients with final diagnoses unable to be determined were classified as stroke mimics. Institutional review board exemption was obtained for the deidentified prospective data registry of this quality‐improvement program.

Final Diagnosis Following In‐Hospital Stroke Alert
Diagnosis No. (N=393) %
  • NOTE: Abbreviations: NOS, not otherwise specified; TIA, transient ischemic attack.

Ischemic stroke 167 42.5%
TIA (definite, probable, or likely) 27 6.9%
TIA (possible or versus a mimic) 7 1.8%
Syncope, hypotension, presyncope, bradycardia 23 5.9%
Seizure 23 5.9%
Delirium/encephalopathy/acute confusional state/dementia 23 5.9%
Stroke mimic NOS 21 5.3%
Other (examples include Parkinson's crisis, musculoskeletal, primary ophthalmologic diagnosis, or cardiovascular ischemia) 17 4.3%
Final diagnosis uncertain 16 4.1%
Medication effect (sedation due to narcotics, limb weakness due to epidural anesthetic, pupil dilation from ipratropium) 15 3.8%
Metabolic (hypoglycemia, electrolyte abnormality, hypercarbia, acid/base disorders, respiratory failure) 12 3.1%
Intracranial hemorrhage (intraparenchymal hemorrhage, subarachnoid hemorrhage, subdural hematoma) 11 2.8%
Conversion disorder/psychiatric/functional/medically unexplained symptoms 7 1.8%
Old deficit due to remote stroke 6 1.5%
Peripheral neuropathy (Bell's palsy, cranial nerve palsy, compression neuropathy) 6 1.5%
Sepsis/emnfection 5 1.3%
Migraine 4 1.0%
Peripheral vestibular dysfunction 3 0.8%

RESULTS

During the 12‐month data collection period, 393 in‐hospital stroke alerts were reported to the NSA. Hospitals reported an average of 65.5 in‐hospital stroke alerts (range, 27156; standard deviation 46.8) (Table 2). Median age was 70 years (range, 18 to >89 years, interquartile range [IQR], 6280 years). Of the stoke alert patients, 52.8% were female, 81.7% were white, 12.7% were black, 2.9% were Hispanic, and 2.7% were other or were unable to be determined. The most common primary services were medicine/hospitalist (36.4%), cardiology (19.5%), cardiothoracic/vascular surgery (13%), and orthopedic surgery (8.6%).

Participating Site Characteristics
All Six Sites Site A Site B Site C Site D Site E Site F
  • NOTE: Abbreviations: IQR, interquartile range. *Lower limit of confidence range represents estimate if all possible transient ischemic attack and final diagnosis unknown represented true ischemic strokes. Upper limit of confidence range represents estimate if all possible transient ischemic attack and final diagnosis uncertain represented stroke mimics.

No. of stroke alerts 393 156 72 50 49 39 27
Median age, y, (IQR 25th to 75th percentile), no. with data for this demographic 70.0 (6280) 376 71.0 (63.081.0) 156 68.0 (58.879.3) 72 76.5 (65.585.0) 50 71.0 (63.078.5) 48 75.0 (58.584.5) 23 77.0 (66.084.5) 27
Sex, % female, no. with data for this demographic 52.8%, 377 48.7%, 156 63.9%, 72 52%, 50 49.0%, 49 52.2%, 23 55.6%, 27
Race, no. (%)
White 308 (81.7%) 146 (93.6%) 40 (55.6%) 47 (94%) 39 (80.0%) 15 (65.2%) 21 (77.8%)
Black or African American 48 (12.7%) 3 (1.9%) 32 (44.4%) 1 (2%) 6 (12.2%) 0 (0%) 6 (22.2%)
Hispanic 11 (2.9%) 3 (1.9%) 0 (0%) 1 (2%) 1 (2.0%) 6 (26.1%) 0 (0%)
Other or unable to determine 10 (2.7%) 4 (2.6%) 0 (0%) 1 (2%) 3 (6.1%) 2 (8.7%) 0 (0%)
No. with data for this demographic 377 156 72 50 49 23 27
Service caring for patient, no. (%)
General medicine 123 (36.4%) 44 (32.1%) 29 (40.3%) 21 (46.7%) 11 (22.9%) 7 (77.7%) 11 (40.7%)
Cardiology 66 (19.5%) 36 (26.3%) 11 (15.3%) 10 (22.2%) 9 (18.8%) 0 (0%) 0 (0%)
Cardiothoracic/vascular surgery 44 (13.0%) 21 (15.3%) 8 (11.1%) 3 (6.7%) 11 (22.9%) 0 (0%) 1 (3.7%)
Orthopedic surgery 29 (8.6%) 17 (12.4%) 4 (5.6%) 3 (6.7%) 2 (4.2%) 0 (0%) 3 (11.1%)
Family practice 13 (3.8%) 2 (1.5%) 1 (1.4%) 1 (2.2%) 0 (0%) 0 (0%) 9 (33.3%)
Pulmonology/critical care 11 (3.3%) 4 (2.9%) 4 (5.6%) 2 (4.4%) 1 (2.1%) 0 (0%) 0 (0%)
General surgery 11 (3.3%) 4 (2.9%) 1 (1.4%) 3 (6.7%) 2 (4.2%) 0 (0%) 1 (3.7%)
Other 41 (12.1%) 9 (6.6%) 14 (19.4%) 2 (4.4%) 12 (25.0%) 2 (22.2) 2 (7.4%)
No. with data for this demographic 338 137 72 45 48 9 27
In‐hospital stroke alert mimic rate
Percent stroke mimics(confidence range)* 46.1% (42.0%47.8%) 48.7% (42.9%51.3%) 50.0% (50.0%50.0%) 28.0% (28.0%30.0%) 42.9% (36.7%46.9%) 66.7% (56.4%66.7%) 29.6% (29.6%29.6%)

Of the stroke alert patients, 167 (42.5%) were found to have ischemic stroke, 27 (6.9%) TIA, 11 (2.8%) intracranial hemorrhage, and 7 (1.8%) had TIA possible or considered along with a stroke mimic in the final diagnosis. The stroke mimic rate was 46.1%, with a confidence range of 42.0% to 47.8% depending on the true pathologic cause of the alerts in the categories possible TIA and final diagnosis uncertain. Participating hospitals had an alarm rate for stroke mimics ranging from 28.0% to 66.7% (median, 45.8%; IQR, 32.9%49.7%) (Table 2). The most common stroke mimics were seizure, hypotension, and delirium (Table 1). Data were available on symptoms that triggered the alert in 373 (94.9%) of cases. Eighteen alerts (4.8%) were for symptoms clearly not included in the NSA stroke alert criteria. The final diagnosis was acute ischemic stroke/TIA or intracranial hemorrhage in 4 of these 18 (22.2%) nonconforming alerts. If alerts called for a decrease in consciousness were also considered nonconforming, then 67 alerts (18.0%) could be categorized as nonconforming. However, 24 of these 67 alerts (35.8%) had a final diagnosis of acute ischemic stroke/TIA or intracranial hemorrhage.

For 194 patients with a final diagnosis of ischemic stroke or TIA, intravenous thrombolysis alone was used for 16 in‐hospital stroke patients (8.2%), 20 received intra‐arterial/mechanical thrombolysis alone (10.3%), and 2 patients received both (1%) (Table 3). No patient with a stroke mimic received thrombolysis.

In‐Hospital Stroke Thrombolysis Rates and Contraindications
  • NOTE: Abbreviations: IA, intra‐arterial; IV, intravenous; TIA, transient ischemic attack; tPA, tissue plasminogen activator. *Definitions for IV exclusions. Multiple: any time more than 1 valid contraindication to IV tPA was listed. Examples would include: recent myocardial infarction on anticoagulation, out of time window and recent myocardial infarction, recent stroke, and advanced age with high National Institute of Health Stroke Scale, no clear onset time, and history of hemorrhagic stroke. Time based: if the sole listed contraindication related to time from onset of brain ischemia. Examples include outside of treatment window, time delay, subacute strokes on imaging, or unknown time last known normal. Medical contraindications: examples include arterial‐venous malformation noted on computed tomography scan, history of recent stroke, history of recent myocardial infarction, gastrointestinal bleeding, or hematuria. Surgical/procedural: recent surgery such as femoral bypass, coronary artery bypass, orthopedic surgery, bowel resection, or invasive procedure such as thoracentesis, arterial puncture at noncompressable site, or cardiac catheterization. Contraindication not otherwise specified: contraindication to IV thrombolysis present but no specific contraindication listed. Minor or improving symptoms: examples include low scores on the National Institute of Health Stroke Scale or rapid improvement in symptoms. Anticoagulation: IV thrombolysis contraindicated due to use of anticoagulation product. Examples include use of warfarin with elevated international normalized ratio or treatment with therapeutic heparin or low‐molecular‐weight heparin. Other: if contraindication was listed but did not meet approved list of contraindications or if no contraindication to IV thrombolysis was listed but the patient was treated only with intra‐arterial or mechanical thrombolysis. Examples include epistaxis or diabetic retinopathy or basilar artery thrombosis treated with IA thrombolysis. Goals of care: patient preferences or goals represent the reason for not considering thrombolysis or if patient/family declined thrombolysis. Examples include comfort measures only status or family declined. Missing: field for contraindication left blank or notated as unable to determine. Seizure at onset of symptoms: for patients with final diagnosis of stroke this would represent onset seizures rather than seizure mimicking stroke, but at the time of the initial stroke alert the seizure was felt to be a contraindication to thrombolysis.

Treatment of stroke alerts with final diagnosis of ischemic stroke or TIA, no. (%), n=194
Treated with IV thrombolysis alone 16 (8.2%)
Treated with IA or mechanical thrombolysis alone 20 (10.3%)
Treated with both IV and IA/mechanical thrombolysis 2 (1.0%)
Contraindication to IV thrombolysis for patients not treated with IV thrombolysis, no. (%), n=176*
Multiple 42 (23.9%)
Time based 27 (15.3%)
Medical 25 (14.2%)
Contraindication not otherwise specified 24 (13.6%)
Surgical/procedural 20 (11.4%)
Minor or rapidly improving symptoms 19 (10.8%)
Anticoagulation 7 (4.0%)
Other 4 (2.3%)
Goals of care 3 (1.7%)
Data unavailable 3 (1.7%)
Seizure at onset of symptoms 2 (1.1%)

DISCUSSION

Given the protean manifestations of brain ischemia, and significant symptom overlap with many mimics, stroke alert criteria casts a wide net in order not to miss or delay evaluation and treatment of true brain ischemia. Time is critical given the association of improved outcomes with more rapid delivery of treatment.[11] The inevitable consequence of the combination of time pressure and clinical uncertainty based solely on physical exam will be alerts due to stroke mimics. Our analysis reveals many of these alternative diagnoses also require urgent evaluation and treatment.

Prior research has found a large proportion of in‐hospital stroke alerts are not for cerebrovascular events.[1, 4, 12] We observed an average of 46.1% of in‐hospital stroke alerts were due to mimics. This rate is substantially higher than described in studies of stroke mimics in the ED.[7, 13, 14] The largest analysis over a 10‐year period from 2 hospitals in Washington found a 30% stroke mimic rate and concluded that in‐hospital location for symptom onset was a statistically significant predictor of being a mimic rather than a cerebrovascular event.[4] One single‐center trial in North Carolina found markedly higher mimic rates for in‐hospital stroke alerts (73%) versus ED stroke alerts (49%).[12] Assessment of neurologic symptoms is challenging in patients already hospitalized for acute medical conditions. The interaction of systemic illness, medications, and surgery seen in the hospital setting may make it more difficult to distinguish between cerebrovascular events and their many mimics.

Interpretation of NSA criteria for calling a stroke code likely varied within and between sites, and inter‐rater reliability of physical signs was not assessed, which is a limitation of the data. Observed rates of stroke for alerts that did not conform to the NSA criteria suggest that clinical judgment remains valuable. Final diagnoses were assigned by the stroke programs, and reliability of this assessment was not evaluated. Sites were not asked to use a specific categorization scheme to group final diagnoses. This analysis was limited to stroke centers with existing infrastructure to respond to stroke alerts and participated in an explicit quality‐improvement initiative on in‐hospital stroke response. Mimic and thrombolysis treatment rates may be different for hospitals without this stroke expertise.

Clinical uncertainty as to final diagnosis was addressed with the inclusion of confidence intervals accounting for potential misdiagnosis of the events in the categories of possible TIA or in the cases where the final diagnosis was unknown. Other studies have categorized TIA versus an alternative diagnosis as stroke mimic, and so our methodology is expected to yield a conservative estimate of the stroke mimic rate. Delirium is often a multifactorial phenomenon, so there may be an element of overlap between this category and other more specific mimic etiologies such as infection, hypotension, metabolic, or medication effect.

This initiative did not have the ability to assess the false negative rate of stroke team activation (failure to identify stroke symptoms in time for acute evaluation). It is not possible to calculate the sensitivity of stroke alerts in each center or conclude the optimal rate of false alarms. The finding of inter‐institutional variability in stroke alerts due to true brain ischemia could be explained by differences in staff education, systematic differences in the patient populations cared for among hospitals, or variation in institutional acceptance of having activated the stroke response team for cases with lower pretest probability of stroke. Sensitivity of alert criteria is more important than specificity, given the consequences of missing a potentially treatable emergent condition.

In conclusion, in this multi‐institution analysis of in‐hospital stroke alerts, a substantial proportion of in‐hospital strokes received thrombolytic therapy. Almost half of stroke alerts will not be for stroke or TIA. For many patients in our study, a change in neurologic status represented a harbinger of a change in general medical condition (hemorrhage, hypotension, hypoglycemia, or respiratory failure). Rapid response systems used for stroke in the hospital need to be trained and prepared to respond to a variety of acute medical conditions that extend beyond ischemic stroke.

Acknowledgements

This work was possible through the National Stroke Association's (NSA) In‐hospital Stroke Quality Improvement Initiative and NSA staff members including Jane Staller, MEd, Miranda N. Bretz, MS, and Amy K. Jensen.

Disclosures: This quality improvement project was funded by an educational grant to the National Stroke Association from Genentech, Inc. and Penumbra, Inc. The funding organizations had no role in the design, content, or preparation of this manuscript. The authors report no conflicts of interest.

Files
References
  1. Cumbler E, Anderson T, Neumann R, Jones W, Brega K. Stroke alert program improves recognition and evaluation time of in‐hospital ischemic stroke. J Stroke Cerebrovasc Dis. 2010;19:494496.
  2. Nolan S, Naylor G, Burns M. Code gray—an organized approach to inpatient stroke. Crit Care Nurs Q. 2003;26:296302.
  3. Daly M, Orto V, Wood C. ID, Stat: rapid response to in‐hospital stroke patients. Nurs Manage. 2009;40:3438.
  4. Merino JG, Luby M, Benson RT, et al. Predictors of acute stroke mimics in 8187 patients referred to a stroke service. J Stroke Cerebrovasc Dis. 2013;22:e397e403.
  5. Forster A, Griebe M, Wolf ME, Szabo K, Hennerici MG, Kern R. How to identify stroke mimics in patients eligible for intravenous thrombolysis? J Neurol. 2012;259:13471353.
  6. Hand PJ, Kwan J, Lindley RI, Dennis MS, Wardlaw JM. Distinguishing between stroke and mimic at the bedside: The Brain Attack Study. Stroke. 2006;37:769775.
  7. Hemmen TM, Meyer BC, McClean TL, Lyden PD. Identification of nonischemic stroke mimics among 411 code strokes at the University of California, San Diego, Stroke Center. J Stroke Cerebrovasc Dis. 2008;17:2325.
  8. Tobin WO, Hentz JG, Bobrow BJ, Demaerschalk BM. Identification of stroke mimics in the emergency department setting. J Brain Dis. 2009;1:1922.
  9. Park JH, Cho HJ, Kim DW, et al. Comparison of the characteristics for in‐hospital and out‐of‐hospital ischaemic strokes. Eur J Neur. 2009;16:582588.
  10. National Stroke Association. Improving in‐hospital stroke through quality improvement interventions webinar. Available at: http://www.stroke.org/we‐can‐help/healthcare‐professionals/improve‐your‐skills/pre‐hospital‐acute‐stroke‐programs‐4. Accessed December 18, 2014.
  11. Saver JL, Fonarow GC, Smith EE, et al. Time to treatment with intravenous tissue plasminogen activator and outcome from acute ischemic stroke. JAMA. 2013;309:24802488.
  12. Husseini NE, Goldstein LB. “Code Stroke”: hospitalized versus emergency department patients. J Stroke Cerebrovasc Dis. 2013;22:345348.
  13. Harbison J, Hossain O, Jenkinson D, et al. Diagnostic accuracy of stroke referrals from primary care, emergency room physicians, and ambulance staff using the face arm speech test. Stroke. 2003;34:7176.
  14. Heckmann JG, Stadter M, Dütsch M, Handschu R, Rauch C, Neundörfer B. Hospitalization of non‐stroke patients in a stroke unit [in German]. Dtsch Med Wochenschr. 2004;129:731735.
Article PDF
Issue
Journal of Hospital Medicine - 10(3)
Page Number
179-183
Sections
Files
Files
Article PDF
Article PDF

Acute change in neurologic status in a hospitalized patient is an emergency requiring timely coordinated evaluation. To address this need, many hospitals have created a mechanism for in‐hospital stroke alerts utilizing generalized rapid response teams or specialized stroke teams.[1, 2, 3] The common purpose is to quickly diagnose new ischemic stroke within the time window for thrombolytic therapy.

Even when acute change in neurologic status is not due to brain ischemia, it may represent a new metabolic disturbance or reflect developing serious systemic illness. Sepsis, hypoglycemia, cardiac arrhythmia, respiratory failure, severe electrolyte disturbances, seizures, or delirium may first manifest as a change in neurologic status.

Prior research on stroke alerts has largely focused on patients who present from the community to the emergency department (ED).[4, 5, 6, 7, 8] Patients who develop acute neurologic symptoms during hospitalization have different risk factors and exposures compared to patients in the community.[9] This study represents the experience of a multistate quality improvement initiative for in‐hospital stroke. We characterize etiologies for symptoms triggering in‐hospital stroke alerts and thrombolytic treatment for in‐hospital strokes.

PATIENTS AND METHODS

The National Stroke Association's (NSA) initiative, Improving In‐Hospital Stroke Response: A Team‐based Quality Improvement Program, included data collection for all in‐hospital stroke alerts over a 12‐month period.[10] Six Joint Commission certified primary stroke centers from Michigan, South Carolina, Pennsylvania, Colorado, Washington, and North Carolina completed the 1‐year quality improvement initiative. One additional site withdrew from the program after the first quarter and was not included in this analysis. Sites prospectively reported deidentified patient‐level data on all adult in‐hospital stroke alerts from July 2010 to June 2011 to the NSA. At all sites, any provider could activate the in‐hospital stroke response system. Stroke alerts were evaluated by a rapid response team with stroke training. The providers on the stroke rapid response team varied between sites. A nurse with stroke training was 1 of the first responders on the stroke response team at all sites.

The NSA in‐hospital stroke‐alert criteria included the following symptoms occurring in the last 24‐hours, even if they resolved: (1) sudden numbness or weakness of the face, arm or leg, especially on 1 side of the body; (2) sudden confusion, trouble speaking or understanding; (3) sudden trouble seeing in 1 or both eyes; (4) sudden trouble walking, dizziness, loss of balance or coordination; and (5) sudden, severe headache with no known cause. Hospitals reported location, service, age, sex, race, symptoms triggering the stroke alert, free text entry of final clinical diagnosis following the completion of stroke alert evaluation, treatment with intravenous or intra‐arterial/mechanical thrombolysis, and any contraindications to intravenous thrombolysis. We categorized stroke mimics using the responses in the final diagnosis field after the data collection period was complete. Strokes were categorized as ischemic stroke, transient ischemic attack (TIA), or intracranial hemorrhage (intraparenchymal, intraventricular, epidural, subdural, or subarachnoid). Stroke mimics were subdivided according to the categories in Table 1. Lack of certainty in the final diagnosis was handled by creating a category of possible TIA, which includes alternative diagnosis versus TIA or the qualifier possible before TIA. Patients with final diagnoses unable to be determined were classified as stroke mimics. Institutional review board exemption was obtained for the deidentified prospective data registry of this quality‐improvement program.

Final Diagnosis Following In‐Hospital Stroke Alert
Diagnosis No. (N=393) %
  • NOTE: Abbreviations: NOS, not otherwise specified; TIA, transient ischemic attack.

Ischemic stroke 167 42.5%
TIA (definite, probable, or likely) 27 6.9%
TIA (possible or versus a mimic) 7 1.8%
Syncope, hypotension, presyncope, bradycardia 23 5.9%
Seizure 23 5.9%
Delirium/encephalopathy/acute confusional state/dementia 23 5.9%
Stroke mimic NOS 21 5.3%
Other (examples include Parkinson's crisis, musculoskeletal, primary ophthalmologic diagnosis, or cardiovascular ischemia) 17 4.3%
Final diagnosis uncertain 16 4.1%
Medication effect (sedation due to narcotics, limb weakness due to epidural anesthetic, pupil dilation from ipratropium) 15 3.8%
Metabolic (hypoglycemia, electrolyte abnormality, hypercarbia, acid/base disorders, respiratory failure) 12 3.1%
Intracranial hemorrhage (intraparenchymal hemorrhage, subarachnoid hemorrhage, subdural hematoma) 11 2.8%
Conversion disorder/psychiatric/functional/medically unexplained symptoms 7 1.8%
Old deficit due to remote stroke 6 1.5%
Peripheral neuropathy (Bell's palsy, cranial nerve palsy, compression neuropathy) 6 1.5%
Sepsis/emnfection 5 1.3%
Migraine 4 1.0%
Peripheral vestibular dysfunction 3 0.8%

RESULTS

During the 12‐month data collection period, 393 in‐hospital stroke alerts were reported to the NSA. Hospitals reported an average of 65.5 in‐hospital stroke alerts (range, 27156; standard deviation 46.8) (Table 2). Median age was 70 years (range, 18 to >89 years, interquartile range [IQR], 6280 years). Of the stoke alert patients, 52.8% were female, 81.7% were white, 12.7% were black, 2.9% were Hispanic, and 2.7% were other or were unable to be determined. The most common primary services were medicine/hospitalist (36.4%), cardiology (19.5%), cardiothoracic/vascular surgery (13%), and orthopedic surgery (8.6%).

Participating Site Characteristics
All Six Sites Site A Site B Site C Site D Site E Site F
  • NOTE: Abbreviations: IQR, interquartile range. *Lower limit of confidence range represents estimate if all possible transient ischemic attack and final diagnosis unknown represented true ischemic strokes. Upper limit of confidence range represents estimate if all possible transient ischemic attack and final diagnosis uncertain represented stroke mimics.

No. of stroke alerts 393 156 72 50 49 39 27
Median age, y, (IQR 25th to 75th percentile), no. with data for this demographic 70.0 (6280) 376 71.0 (63.081.0) 156 68.0 (58.879.3) 72 76.5 (65.585.0) 50 71.0 (63.078.5) 48 75.0 (58.584.5) 23 77.0 (66.084.5) 27
Sex, % female, no. with data for this demographic 52.8%, 377 48.7%, 156 63.9%, 72 52%, 50 49.0%, 49 52.2%, 23 55.6%, 27
Race, no. (%)
White 308 (81.7%) 146 (93.6%) 40 (55.6%) 47 (94%) 39 (80.0%) 15 (65.2%) 21 (77.8%)
Black or African American 48 (12.7%) 3 (1.9%) 32 (44.4%) 1 (2%) 6 (12.2%) 0 (0%) 6 (22.2%)
Hispanic 11 (2.9%) 3 (1.9%) 0 (0%) 1 (2%) 1 (2.0%) 6 (26.1%) 0 (0%)
Other or unable to determine 10 (2.7%) 4 (2.6%) 0 (0%) 1 (2%) 3 (6.1%) 2 (8.7%) 0 (0%)
No. with data for this demographic 377 156 72 50 49 23 27
Service caring for patient, no. (%)
General medicine 123 (36.4%) 44 (32.1%) 29 (40.3%) 21 (46.7%) 11 (22.9%) 7 (77.7%) 11 (40.7%)
Cardiology 66 (19.5%) 36 (26.3%) 11 (15.3%) 10 (22.2%) 9 (18.8%) 0 (0%) 0 (0%)
Cardiothoracic/vascular surgery 44 (13.0%) 21 (15.3%) 8 (11.1%) 3 (6.7%) 11 (22.9%) 0 (0%) 1 (3.7%)
Orthopedic surgery 29 (8.6%) 17 (12.4%) 4 (5.6%) 3 (6.7%) 2 (4.2%) 0 (0%) 3 (11.1%)
Family practice 13 (3.8%) 2 (1.5%) 1 (1.4%) 1 (2.2%) 0 (0%) 0 (0%) 9 (33.3%)
Pulmonology/critical care 11 (3.3%) 4 (2.9%) 4 (5.6%) 2 (4.4%) 1 (2.1%) 0 (0%) 0 (0%)
General surgery 11 (3.3%) 4 (2.9%) 1 (1.4%) 3 (6.7%) 2 (4.2%) 0 (0%) 1 (3.7%)
Other 41 (12.1%) 9 (6.6%) 14 (19.4%) 2 (4.4%) 12 (25.0%) 2 (22.2) 2 (7.4%)
No. with data for this demographic 338 137 72 45 48 9 27
In‐hospital stroke alert mimic rate
Percent stroke mimics(confidence range)* 46.1% (42.0%47.8%) 48.7% (42.9%51.3%) 50.0% (50.0%50.0%) 28.0% (28.0%30.0%) 42.9% (36.7%46.9%) 66.7% (56.4%66.7%) 29.6% (29.6%29.6%)

Of the stroke alert patients, 167 (42.5%) were found to have ischemic stroke, 27 (6.9%) TIA, 11 (2.8%) intracranial hemorrhage, and 7 (1.8%) had TIA possible or considered along with a stroke mimic in the final diagnosis. The stroke mimic rate was 46.1%, with a confidence range of 42.0% to 47.8% depending on the true pathologic cause of the alerts in the categories possible TIA and final diagnosis uncertain. Participating hospitals had an alarm rate for stroke mimics ranging from 28.0% to 66.7% (median, 45.8%; IQR, 32.9%49.7%) (Table 2). The most common stroke mimics were seizure, hypotension, and delirium (Table 1). Data were available on symptoms that triggered the alert in 373 (94.9%) of cases. Eighteen alerts (4.8%) were for symptoms clearly not included in the NSA stroke alert criteria. The final diagnosis was acute ischemic stroke/TIA or intracranial hemorrhage in 4 of these 18 (22.2%) nonconforming alerts. If alerts called for a decrease in consciousness were also considered nonconforming, then 67 alerts (18.0%) could be categorized as nonconforming. However, 24 of these 67 alerts (35.8%) had a final diagnosis of acute ischemic stroke/TIA or intracranial hemorrhage.

For 194 patients with a final diagnosis of ischemic stroke or TIA, intravenous thrombolysis alone was used for 16 in‐hospital stroke patients (8.2%), 20 received intra‐arterial/mechanical thrombolysis alone (10.3%), and 2 patients received both (1%) (Table 3). No patient with a stroke mimic received thrombolysis.

In‐Hospital Stroke Thrombolysis Rates and Contraindications
  • NOTE: Abbreviations: IA, intra‐arterial; IV, intravenous; TIA, transient ischemic attack; tPA, tissue plasminogen activator. *Definitions for IV exclusions. Multiple: any time more than 1 valid contraindication to IV tPA was listed. Examples would include: recent myocardial infarction on anticoagulation, out of time window and recent myocardial infarction, recent stroke, and advanced age with high National Institute of Health Stroke Scale, no clear onset time, and history of hemorrhagic stroke. Time based: if the sole listed contraindication related to time from onset of brain ischemia. Examples include outside of treatment window, time delay, subacute strokes on imaging, or unknown time last known normal. Medical contraindications: examples include arterial‐venous malformation noted on computed tomography scan, history of recent stroke, history of recent myocardial infarction, gastrointestinal bleeding, or hematuria. Surgical/procedural: recent surgery such as femoral bypass, coronary artery bypass, orthopedic surgery, bowel resection, or invasive procedure such as thoracentesis, arterial puncture at noncompressable site, or cardiac catheterization. Contraindication not otherwise specified: contraindication to IV thrombolysis present but no specific contraindication listed. Minor or improving symptoms: examples include low scores on the National Institute of Health Stroke Scale or rapid improvement in symptoms. Anticoagulation: IV thrombolysis contraindicated due to use of anticoagulation product. Examples include use of warfarin with elevated international normalized ratio or treatment with therapeutic heparin or low‐molecular‐weight heparin. Other: if contraindication was listed but did not meet approved list of contraindications or if no contraindication to IV thrombolysis was listed but the patient was treated only with intra‐arterial or mechanical thrombolysis. Examples include epistaxis or diabetic retinopathy or basilar artery thrombosis treated with IA thrombolysis. Goals of care: patient preferences or goals represent the reason for not considering thrombolysis or if patient/family declined thrombolysis. Examples include comfort measures only status or family declined. Missing: field for contraindication left blank or notated as unable to determine. Seizure at onset of symptoms: for patients with final diagnosis of stroke this would represent onset seizures rather than seizure mimicking stroke, but at the time of the initial stroke alert the seizure was felt to be a contraindication to thrombolysis.

Treatment of stroke alerts with final diagnosis of ischemic stroke or TIA, no. (%), n=194
Treated with IV thrombolysis alone 16 (8.2%)
Treated with IA or mechanical thrombolysis alone 20 (10.3%)
Treated with both IV and IA/mechanical thrombolysis 2 (1.0%)
Contraindication to IV thrombolysis for patients not treated with IV thrombolysis, no. (%), n=176*
Multiple 42 (23.9%)
Time based 27 (15.3%)
Medical 25 (14.2%)
Contraindication not otherwise specified 24 (13.6%)
Surgical/procedural 20 (11.4%)
Minor or rapidly improving symptoms 19 (10.8%)
Anticoagulation 7 (4.0%)
Other 4 (2.3%)
Goals of care 3 (1.7%)
Data unavailable 3 (1.7%)
Seizure at onset of symptoms 2 (1.1%)

DISCUSSION

Given the protean manifestations of brain ischemia, and significant symptom overlap with many mimics, stroke alert criteria casts a wide net in order not to miss or delay evaluation and treatment of true brain ischemia. Time is critical given the association of improved outcomes with more rapid delivery of treatment.[11] The inevitable consequence of the combination of time pressure and clinical uncertainty based solely on physical exam will be alerts due to stroke mimics. Our analysis reveals many of these alternative diagnoses also require urgent evaluation and treatment.

Prior research has found a large proportion of in‐hospital stroke alerts are not for cerebrovascular events.[1, 4, 12] We observed an average of 46.1% of in‐hospital stroke alerts were due to mimics. This rate is substantially higher than described in studies of stroke mimics in the ED.[7, 13, 14] The largest analysis over a 10‐year period from 2 hospitals in Washington found a 30% stroke mimic rate and concluded that in‐hospital location for symptom onset was a statistically significant predictor of being a mimic rather than a cerebrovascular event.[4] One single‐center trial in North Carolina found markedly higher mimic rates for in‐hospital stroke alerts (73%) versus ED stroke alerts (49%).[12] Assessment of neurologic symptoms is challenging in patients already hospitalized for acute medical conditions. The interaction of systemic illness, medications, and surgery seen in the hospital setting may make it more difficult to distinguish between cerebrovascular events and their many mimics.

Interpretation of NSA criteria for calling a stroke code likely varied within and between sites, and inter‐rater reliability of physical signs was not assessed, which is a limitation of the data. Observed rates of stroke for alerts that did not conform to the NSA criteria suggest that clinical judgment remains valuable. Final diagnoses were assigned by the stroke programs, and reliability of this assessment was not evaluated. Sites were not asked to use a specific categorization scheme to group final diagnoses. This analysis was limited to stroke centers with existing infrastructure to respond to stroke alerts and participated in an explicit quality‐improvement initiative on in‐hospital stroke response. Mimic and thrombolysis treatment rates may be different for hospitals without this stroke expertise.

Clinical uncertainty as to final diagnosis was addressed with the inclusion of confidence intervals accounting for potential misdiagnosis of the events in the categories of possible TIA or in the cases where the final diagnosis was unknown. Other studies have categorized TIA versus an alternative diagnosis as stroke mimic, and so our methodology is expected to yield a conservative estimate of the stroke mimic rate. Delirium is often a multifactorial phenomenon, so there may be an element of overlap between this category and other more specific mimic etiologies such as infection, hypotension, metabolic, or medication effect.

This initiative did not have the ability to assess the false negative rate of stroke team activation (failure to identify stroke symptoms in time for acute evaluation). It is not possible to calculate the sensitivity of stroke alerts in each center or conclude the optimal rate of false alarms. The finding of inter‐institutional variability in stroke alerts due to true brain ischemia could be explained by differences in staff education, systematic differences in the patient populations cared for among hospitals, or variation in institutional acceptance of having activated the stroke response team for cases with lower pretest probability of stroke. Sensitivity of alert criteria is more important than specificity, given the consequences of missing a potentially treatable emergent condition.

In conclusion, in this multi‐institution analysis of in‐hospital stroke alerts, a substantial proportion of in‐hospital strokes received thrombolytic therapy. Almost half of stroke alerts will not be for stroke or TIA. For many patients in our study, a change in neurologic status represented a harbinger of a change in general medical condition (hemorrhage, hypotension, hypoglycemia, or respiratory failure). Rapid response systems used for stroke in the hospital need to be trained and prepared to respond to a variety of acute medical conditions that extend beyond ischemic stroke.

Acknowledgements

This work was possible through the National Stroke Association's (NSA) In‐hospital Stroke Quality Improvement Initiative and NSA staff members including Jane Staller, MEd, Miranda N. Bretz, MS, and Amy K. Jensen.

Disclosures: This quality improvement project was funded by an educational grant to the National Stroke Association from Genentech, Inc. and Penumbra, Inc. The funding organizations had no role in the design, content, or preparation of this manuscript. The authors report no conflicts of interest.

Acute change in neurologic status in a hospitalized patient is an emergency requiring timely coordinated evaluation. To address this need, many hospitals have created a mechanism for in‐hospital stroke alerts utilizing generalized rapid response teams or specialized stroke teams.[1, 2, 3] The common purpose is to quickly diagnose new ischemic stroke within the time window for thrombolytic therapy.

Even when acute change in neurologic status is not due to brain ischemia, it may represent a new metabolic disturbance or reflect developing serious systemic illness. Sepsis, hypoglycemia, cardiac arrhythmia, respiratory failure, severe electrolyte disturbances, seizures, or delirium may first manifest as a change in neurologic status.

Prior research on stroke alerts has largely focused on patients who present from the community to the emergency department (ED).[4, 5, 6, 7, 8] Patients who develop acute neurologic symptoms during hospitalization have different risk factors and exposures compared to patients in the community.[9] This study represents the experience of a multistate quality improvement initiative for in‐hospital stroke. We characterize etiologies for symptoms triggering in‐hospital stroke alerts and thrombolytic treatment for in‐hospital strokes.

PATIENTS AND METHODS

The National Stroke Association's (NSA) initiative, Improving In‐Hospital Stroke Response: A Team‐based Quality Improvement Program, included data collection for all in‐hospital stroke alerts over a 12‐month period.[10] Six Joint Commission certified primary stroke centers from Michigan, South Carolina, Pennsylvania, Colorado, Washington, and North Carolina completed the 1‐year quality improvement initiative. One additional site withdrew from the program after the first quarter and was not included in this analysis. Sites prospectively reported deidentified patient‐level data on all adult in‐hospital stroke alerts from July 2010 to June 2011 to the NSA. At all sites, any provider could activate the in‐hospital stroke response system. Stroke alerts were evaluated by a rapid response team with stroke training. The providers on the stroke rapid response team varied between sites. A nurse with stroke training was 1 of the first responders on the stroke response team at all sites.

The NSA in‐hospital stroke‐alert criteria included the following symptoms occurring in the last 24‐hours, even if they resolved: (1) sudden numbness or weakness of the face, arm or leg, especially on 1 side of the body; (2) sudden confusion, trouble speaking or understanding; (3) sudden trouble seeing in 1 or both eyes; (4) sudden trouble walking, dizziness, loss of balance or coordination; and (5) sudden, severe headache with no known cause. Hospitals reported location, service, age, sex, race, symptoms triggering the stroke alert, free text entry of final clinical diagnosis following the completion of stroke alert evaluation, treatment with intravenous or intra‐arterial/mechanical thrombolysis, and any contraindications to intravenous thrombolysis. We categorized stroke mimics using the responses in the final diagnosis field after the data collection period was complete. Strokes were categorized as ischemic stroke, transient ischemic attack (TIA), or intracranial hemorrhage (intraparenchymal, intraventricular, epidural, subdural, or subarachnoid). Stroke mimics were subdivided according to the categories in Table 1. Lack of certainty in the final diagnosis was handled by creating a category of possible TIA, which includes alternative diagnosis versus TIA or the qualifier possible before TIA. Patients with final diagnoses unable to be determined were classified as stroke mimics. Institutional review board exemption was obtained for the deidentified prospective data registry of this quality‐improvement program.

Final Diagnosis Following In‐Hospital Stroke Alert
Diagnosis No. (N=393) %
  • NOTE: Abbreviations: NOS, not otherwise specified; TIA, transient ischemic attack.

Ischemic stroke 167 42.5%
TIA (definite, probable, or likely) 27 6.9%
TIA (possible or versus a mimic) 7 1.8%
Syncope, hypotension, presyncope, bradycardia 23 5.9%
Seizure 23 5.9%
Delirium/encephalopathy/acute confusional state/dementia 23 5.9%
Stroke mimic NOS 21 5.3%
Other (examples include Parkinson's crisis, musculoskeletal, primary ophthalmologic diagnosis, or cardiovascular ischemia) 17 4.3%
Final diagnosis uncertain 16 4.1%
Medication effect (sedation due to narcotics, limb weakness due to epidural anesthetic, pupil dilation from ipratropium) 15 3.8%
Metabolic (hypoglycemia, electrolyte abnormality, hypercarbia, acid/base disorders, respiratory failure) 12 3.1%
Intracranial hemorrhage (intraparenchymal hemorrhage, subarachnoid hemorrhage, subdural hematoma) 11 2.8%
Conversion disorder/psychiatric/functional/medically unexplained symptoms 7 1.8%
Old deficit due to remote stroke 6 1.5%
Peripheral neuropathy (Bell's palsy, cranial nerve palsy, compression neuropathy) 6 1.5%
Sepsis/emnfection 5 1.3%
Migraine 4 1.0%
Peripheral vestibular dysfunction 3 0.8%

RESULTS

During the 12‐month data collection period, 393 in‐hospital stroke alerts were reported to the NSA. Hospitals reported an average of 65.5 in‐hospital stroke alerts (range, 27156; standard deviation 46.8) (Table 2). Median age was 70 years (range, 18 to >89 years, interquartile range [IQR], 6280 years). Of the stoke alert patients, 52.8% were female, 81.7% were white, 12.7% were black, 2.9% were Hispanic, and 2.7% were other or were unable to be determined. The most common primary services were medicine/hospitalist (36.4%), cardiology (19.5%), cardiothoracic/vascular surgery (13%), and orthopedic surgery (8.6%).

Participating Site Characteristics
All Six Sites Site A Site B Site C Site D Site E Site F
  • NOTE: Abbreviations: IQR, interquartile range. *Lower limit of confidence range represents estimate if all possible transient ischemic attack and final diagnosis unknown represented true ischemic strokes. Upper limit of confidence range represents estimate if all possible transient ischemic attack and final diagnosis uncertain represented stroke mimics.

No. of stroke alerts 393 156 72 50 49 39 27
Median age, y, (IQR 25th to 75th percentile), no. with data for this demographic 70.0 (6280) 376 71.0 (63.081.0) 156 68.0 (58.879.3) 72 76.5 (65.585.0) 50 71.0 (63.078.5) 48 75.0 (58.584.5) 23 77.0 (66.084.5) 27
Sex, % female, no. with data for this demographic 52.8%, 377 48.7%, 156 63.9%, 72 52%, 50 49.0%, 49 52.2%, 23 55.6%, 27
Race, no. (%)
White 308 (81.7%) 146 (93.6%) 40 (55.6%) 47 (94%) 39 (80.0%) 15 (65.2%) 21 (77.8%)
Black or African American 48 (12.7%) 3 (1.9%) 32 (44.4%) 1 (2%) 6 (12.2%) 0 (0%) 6 (22.2%)
Hispanic 11 (2.9%) 3 (1.9%) 0 (0%) 1 (2%) 1 (2.0%) 6 (26.1%) 0 (0%)
Other or unable to determine 10 (2.7%) 4 (2.6%) 0 (0%) 1 (2%) 3 (6.1%) 2 (8.7%) 0 (0%)
No. with data for this demographic 377 156 72 50 49 23 27
Service caring for patient, no. (%)
General medicine 123 (36.4%) 44 (32.1%) 29 (40.3%) 21 (46.7%) 11 (22.9%) 7 (77.7%) 11 (40.7%)
Cardiology 66 (19.5%) 36 (26.3%) 11 (15.3%) 10 (22.2%) 9 (18.8%) 0 (0%) 0 (0%)
Cardiothoracic/vascular surgery 44 (13.0%) 21 (15.3%) 8 (11.1%) 3 (6.7%) 11 (22.9%) 0 (0%) 1 (3.7%)
Orthopedic surgery 29 (8.6%) 17 (12.4%) 4 (5.6%) 3 (6.7%) 2 (4.2%) 0 (0%) 3 (11.1%)
Family practice 13 (3.8%) 2 (1.5%) 1 (1.4%) 1 (2.2%) 0 (0%) 0 (0%) 9 (33.3%)
Pulmonology/critical care 11 (3.3%) 4 (2.9%) 4 (5.6%) 2 (4.4%) 1 (2.1%) 0 (0%) 0 (0%)
General surgery 11 (3.3%) 4 (2.9%) 1 (1.4%) 3 (6.7%) 2 (4.2%) 0 (0%) 1 (3.7%)
Other 41 (12.1%) 9 (6.6%) 14 (19.4%) 2 (4.4%) 12 (25.0%) 2 (22.2) 2 (7.4%)
No. with data for this demographic 338 137 72 45 48 9 27
In‐hospital stroke alert mimic rate
Percent stroke mimics(confidence range)* 46.1% (42.0%47.8%) 48.7% (42.9%51.3%) 50.0% (50.0%50.0%) 28.0% (28.0%30.0%) 42.9% (36.7%46.9%) 66.7% (56.4%66.7%) 29.6% (29.6%29.6%)

Of the stroke alert patients, 167 (42.5%) were found to have ischemic stroke, 27 (6.9%) TIA, 11 (2.8%) intracranial hemorrhage, and 7 (1.8%) had TIA possible or considered along with a stroke mimic in the final diagnosis. The stroke mimic rate was 46.1%, with a confidence range of 42.0% to 47.8% depending on the true pathologic cause of the alerts in the categories possible TIA and final diagnosis uncertain. Participating hospitals had an alarm rate for stroke mimics ranging from 28.0% to 66.7% (median, 45.8%; IQR, 32.9%49.7%) (Table 2). The most common stroke mimics were seizure, hypotension, and delirium (Table 1). Data were available on symptoms that triggered the alert in 373 (94.9%) of cases. Eighteen alerts (4.8%) were for symptoms clearly not included in the NSA stroke alert criteria. The final diagnosis was acute ischemic stroke/TIA or intracranial hemorrhage in 4 of these 18 (22.2%) nonconforming alerts. If alerts called for a decrease in consciousness were also considered nonconforming, then 67 alerts (18.0%) could be categorized as nonconforming. However, 24 of these 67 alerts (35.8%) had a final diagnosis of acute ischemic stroke/TIA or intracranial hemorrhage.

For 194 patients with a final diagnosis of ischemic stroke or TIA, intravenous thrombolysis alone was used for 16 in‐hospital stroke patients (8.2%), 20 received intra‐arterial/mechanical thrombolysis alone (10.3%), and 2 patients received both (1%) (Table 3). No patient with a stroke mimic received thrombolysis.

In‐Hospital Stroke Thrombolysis Rates and Contraindications
  • NOTE: Abbreviations: IA, intra‐arterial; IV, intravenous; TIA, transient ischemic attack; tPA, tissue plasminogen activator. *Definitions for IV exclusions. Multiple: any time more than 1 valid contraindication to IV tPA was listed. Examples would include: recent myocardial infarction on anticoagulation, out of time window and recent myocardial infarction, recent stroke, and advanced age with high National Institute of Health Stroke Scale, no clear onset time, and history of hemorrhagic stroke. Time based: if the sole listed contraindication related to time from onset of brain ischemia. Examples include outside of treatment window, time delay, subacute strokes on imaging, or unknown time last known normal. Medical contraindications: examples include arterial‐venous malformation noted on computed tomography scan, history of recent stroke, history of recent myocardial infarction, gastrointestinal bleeding, or hematuria. Surgical/procedural: recent surgery such as femoral bypass, coronary artery bypass, orthopedic surgery, bowel resection, or invasive procedure such as thoracentesis, arterial puncture at noncompressable site, or cardiac catheterization. Contraindication not otherwise specified: contraindication to IV thrombolysis present but no specific contraindication listed. Minor or improving symptoms: examples include low scores on the National Institute of Health Stroke Scale or rapid improvement in symptoms. Anticoagulation: IV thrombolysis contraindicated due to use of anticoagulation product. Examples include use of warfarin with elevated international normalized ratio or treatment with therapeutic heparin or low‐molecular‐weight heparin. Other: if contraindication was listed but did not meet approved list of contraindications or if no contraindication to IV thrombolysis was listed but the patient was treated only with intra‐arterial or mechanical thrombolysis. Examples include epistaxis or diabetic retinopathy or basilar artery thrombosis treated with IA thrombolysis. Goals of care: patient preferences or goals represent the reason for not considering thrombolysis or if patient/family declined thrombolysis. Examples include comfort measures only status or family declined. Missing: field for contraindication left blank or notated as unable to determine. Seizure at onset of symptoms: for patients with final diagnosis of stroke this would represent onset seizures rather than seizure mimicking stroke, but at the time of the initial stroke alert the seizure was felt to be a contraindication to thrombolysis.

Treatment of stroke alerts with final diagnosis of ischemic stroke or TIA, no. (%), n=194
Treated with IV thrombolysis alone 16 (8.2%)
Treated with IA or mechanical thrombolysis alone 20 (10.3%)
Treated with both IV and IA/mechanical thrombolysis 2 (1.0%)
Contraindication to IV thrombolysis for patients not treated with IV thrombolysis, no. (%), n=176*
Multiple 42 (23.9%)
Time based 27 (15.3%)
Medical 25 (14.2%)
Contraindication not otherwise specified 24 (13.6%)
Surgical/procedural 20 (11.4%)
Minor or rapidly improving symptoms 19 (10.8%)
Anticoagulation 7 (4.0%)
Other 4 (2.3%)
Goals of care 3 (1.7%)
Data unavailable 3 (1.7%)
Seizure at onset of symptoms 2 (1.1%)

DISCUSSION

Given the protean manifestations of brain ischemia, and significant symptom overlap with many mimics, stroke alert criteria casts a wide net in order not to miss or delay evaluation and treatment of true brain ischemia. Time is critical given the association of improved outcomes with more rapid delivery of treatment.[11] The inevitable consequence of the combination of time pressure and clinical uncertainty based solely on physical exam will be alerts due to stroke mimics. Our analysis reveals many of these alternative diagnoses also require urgent evaluation and treatment.

Prior research has found a large proportion of in‐hospital stroke alerts are not for cerebrovascular events.[1, 4, 12] We observed an average of 46.1% of in‐hospital stroke alerts were due to mimics. This rate is substantially higher than described in studies of stroke mimics in the ED.[7, 13, 14] The largest analysis over a 10‐year period from 2 hospitals in Washington found a 30% stroke mimic rate and concluded that in‐hospital location for symptom onset was a statistically significant predictor of being a mimic rather than a cerebrovascular event.[4] One single‐center trial in North Carolina found markedly higher mimic rates for in‐hospital stroke alerts (73%) versus ED stroke alerts (49%).[12] Assessment of neurologic symptoms is challenging in patients already hospitalized for acute medical conditions. The interaction of systemic illness, medications, and surgery seen in the hospital setting may make it more difficult to distinguish between cerebrovascular events and their many mimics.

Interpretation of NSA criteria for calling a stroke code likely varied within and between sites, and inter‐rater reliability of physical signs was not assessed, which is a limitation of the data. Observed rates of stroke for alerts that did not conform to the NSA criteria suggest that clinical judgment remains valuable. Final diagnoses were assigned by the stroke programs, and reliability of this assessment was not evaluated. Sites were not asked to use a specific categorization scheme to group final diagnoses. This analysis was limited to stroke centers with existing infrastructure to respond to stroke alerts and participated in an explicit quality‐improvement initiative on in‐hospital stroke response. Mimic and thrombolysis treatment rates may be different for hospitals without this stroke expertise.

Clinical uncertainty as to final diagnosis was addressed with the inclusion of confidence intervals accounting for potential misdiagnosis of the events in the categories of possible TIA or in the cases where the final diagnosis was unknown. Other studies have categorized TIA versus an alternative diagnosis as stroke mimic, and so our methodology is expected to yield a conservative estimate of the stroke mimic rate. Delirium is often a multifactorial phenomenon, so there may be an element of overlap between this category and other more specific mimic etiologies such as infection, hypotension, metabolic, or medication effect.

This initiative did not have the ability to assess the false negative rate of stroke team activation (failure to identify stroke symptoms in time for acute evaluation). It is not possible to calculate the sensitivity of stroke alerts in each center or conclude the optimal rate of false alarms. The finding of inter‐institutional variability in stroke alerts due to true brain ischemia could be explained by differences in staff education, systematic differences in the patient populations cared for among hospitals, or variation in institutional acceptance of having activated the stroke response team for cases with lower pretest probability of stroke. Sensitivity of alert criteria is more important than specificity, given the consequences of missing a potentially treatable emergent condition.

In conclusion, in this multi‐institution analysis of in‐hospital stroke alerts, a substantial proportion of in‐hospital strokes received thrombolytic therapy. Almost half of stroke alerts will not be for stroke or TIA. For many patients in our study, a change in neurologic status represented a harbinger of a change in general medical condition (hemorrhage, hypotension, hypoglycemia, or respiratory failure). Rapid response systems used for stroke in the hospital need to be trained and prepared to respond to a variety of acute medical conditions that extend beyond ischemic stroke.

Acknowledgements

This work was possible through the National Stroke Association's (NSA) In‐hospital Stroke Quality Improvement Initiative and NSA staff members including Jane Staller, MEd, Miranda N. Bretz, MS, and Amy K. Jensen.

Disclosures: This quality improvement project was funded by an educational grant to the National Stroke Association from Genentech, Inc. and Penumbra, Inc. The funding organizations had no role in the design, content, or preparation of this manuscript. The authors report no conflicts of interest.

References
  1. Cumbler E, Anderson T, Neumann R, Jones W, Brega K. Stroke alert program improves recognition and evaluation time of in‐hospital ischemic stroke. J Stroke Cerebrovasc Dis. 2010;19:494496.
  2. Nolan S, Naylor G, Burns M. Code gray—an organized approach to inpatient stroke. Crit Care Nurs Q. 2003;26:296302.
  3. Daly M, Orto V, Wood C. ID, Stat: rapid response to in‐hospital stroke patients. Nurs Manage. 2009;40:3438.
  4. Merino JG, Luby M, Benson RT, et al. Predictors of acute stroke mimics in 8187 patients referred to a stroke service. J Stroke Cerebrovasc Dis. 2013;22:e397e403.
  5. Forster A, Griebe M, Wolf ME, Szabo K, Hennerici MG, Kern R. How to identify stroke mimics in patients eligible for intravenous thrombolysis? J Neurol. 2012;259:13471353.
  6. Hand PJ, Kwan J, Lindley RI, Dennis MS, Wardlaw JM. Distinguishing between stroke and mimic at the bedside: The Brain Attack Study. Stroke. 2006;37:769775.
  7. Hemmen TM, Meyer BC, McClean TL, Lyden PD. Identification of nonischemic stroke mimics among 411 code strokes at the University of California, San Diego, Stroke Center. J Stroke Cerebrovasc Dis. 2008;17:2325.
  8. Tobin WO, Hentz JG, Bobrow BJ, Demaerschalk BM. Identification of stroke mimics in the emergency department setting. J Brain Dis. 2009;1:1922.
  9. Park JH, Cho HJ, Kim DW, et al. Comparison of the characteristics for in‐hospital and out‐of‐hospital ischaemic strokes. Eur J Neur. 2009;16:582588.
  10. National Stroke Association. Improving in‐hospital stroke through quality improvement interventions webinar. Available at: http://www.stroke.org/we‐can‐help/healthcare‐professionals/improve‐your‐skills/pre‐hospital‐acute‐stroke‐programs‐4. Accessed December 18, 2014.
  11. Saver JL, Fonarow GC, Smith EE, et al. Time to treatment with intravenous tissue plasminogen activator and outcome from acute ischemic stroke. JAMA. 2013;309:24802488.
  12. Husseini NE, Goldstein LB. “Code Stroke”: hospitalized versus emergency department patients. J Stroke Cerebrovasc Dis. 2013;22:345348.
  13. Harbison J, Hossain O, Jenkinson D, et al. Diagnostic accuracy of stroke referrals from primary care, emergency room physicians, and ambulance staff using the face arm speech test. Stroke. 2003;34:7176.
  14. Heckmann JG, Stadter M, Dütsch M, Handschu R, Rauch C, Neundörfer B. Hospitalization of non‐stroke patients in a stroke unit [in German]. Dtsch Med Wochenschr. 2004;129:731735.
References
  1. Cumbler E, Anderson T, Neumann R, Jones W, Brega K. Stroke alert program improves recognition and evaluation time of in‐hospital ischemic stroke. J Stroke Cerebrovasc Dis. 2010;19:494496.
  2. Nolan S, Naylor G, Burns M. Code gray—an organized approach to inpatient stroke. Crit Care Nurs Q. 2003;26:296302.
  3. Daly M, Orto V, Wood C. ID, Stat: rapid response to in‐hospital stroke patients. Nurs Manage. 2009;40:3438.
  4. Merino JG, Luby M, Benson RT, et al. Predictors of acute stroke mimics in 8187 patients referred to a stroke service. J Stroke Cerebrovasc Dis. 2013;22:e397e403.
  5. Forster A, Griebe M, Wolf ME, Szabo K, Hennerici MG, Kern R. How to identify stroke mimics in patients eligible for intravenous thrombolysis? J Neurol. 2012;259:13471353.
  6. Hand PJ, Kwan J, Lindley RI, Dennis MS, Wardlaw JM. Distinguishing between stroke and mimic at the bedside: The Brain Attack Study. Stroke. 2006;37:769775.
  7. Hemmen TM, Meyer BC, McClean TL, Lyden PD. Identification of nonischemic stroke mimics among 411 code strokes at the University of California, San Diego, Stroke Center. J Stroke Cerebrovasc Dis. 2008;17:2325.
  8. Tobin WO, Hentz JG, Bobrow BJ, Demaerschalk BM. Identification of stroke mimics in the emergency department setting. J Brain Dis. 2009;1:1922.
  9. Park JH, Cho HJ, Kim DW, et al. Comparison of the characteristics for in‐hospital and out‐of‐hospital ischaemic strokes. Eur J Neur. 2009;16:582588.
  10. National Stroke Association. Improving in‐hospital stroke through quality improvement interventions webinar. Available at: http://www.stroke.org/we‐can‐help/healthcare‐professionals/improve‐your‐skills/pre‐hospital‐acute‐stroke‐programs‐4. Accessed December 18, 2014.
  11. Saver JL, Fonarow GC, Smith EE, et al. Time to treatment with intravenous tissue plasminogen activator and outcome from acute ischemic stroke. JAMA. 2013;309:24802488.
  12. Husseini NE, Goldstein LB. “Code Stroke”: hospitalized versus emergency department patients. J Stroke Cerebrovasc Dis. 2013;22:345348.
  13. Harbison J, Hossain O, Jenkinson D, et al. Diagnostic accuracy of stroke referrals from primary care, emergency room physicians, and ambulance staff using the face arm speech test. Stroke. 2003;34:7176.
  14. Heckmann JG, Stadter M, Dütsch M, Handschu R, Rauch C, Neundörfer B. Hospitalization of non‐stroke patients in a stroke unit [in German]. Dtsch Med Wochenschr. 2004;129:731735.
Issue
Journal of Hospital Medicine - 10(3)
Issue
Journal of Hospital Medicine - 10(3)
Page Number
179-183
Page Number
179-183
Article Type
Display Headline
Code stroke: Multicenter experience with in‐hospital stroke alerts
Display Headline
Code stroke: Multicenter experience with in‐hospital stroke alerts
Sections
Article Source
© 2014 Society of Hospital Medicine
Disallow All Ads
Correspondence Location
Address for correspondence and reprint requests: Ethan Cumbler, MD, Associate Professor, Department of Medicine, University of Colorado School of Medicine, Anschutz Medical Campus, 12401 E. 17th Ave., Mail Stop F782, Aurora, CO 80045; Telephone: 702‐848‐4289; Fax: 720‐848‐4293; E‐mail: [email protected]
Content Gating
Gated (full article locked unless allowed per User)
Gating Strategy
First Peek Free
Article PDF Media
Media Files

ADA’s revised diabetes 'standards' broaden statin use

Statin treatment benefits most diabetes patients
Article Type
Changed
Tue, 05/03/2022 - 15:45
Display Headline
ADA’s revised diabetes 'standards' broaden statin use

Most patients with diabetes should receive at least a moderate statin dosage regardless of their cardiovascular disease risk profile, according to the American Diabetes Association’s annual update to standards for managing patients with diabetes.

“Standards of Medical Care in Diabetes–2015” also shifts the ADA’s official recommendation on assessing patients for statin treatment from a decision based on blood levels of low density lipoprotein (LDL) cholesterol to a risk-based assessment. That change brings the ADA’s position in line with the approach advocated in late 2013 by guidelines from the American College of Cardiology (ACC) and the American Heart Association (AHA) (J. Am. Coll. Cardiol. 2014;63:2889-934).

The ADA released the revised standards online Dec. 23.

The statin use recommendation is “a major change, a fairly big change in how we provide care, although not that big a change in what most patients are prescribed,” said Dr. Richard W. Grant, a primary care physician and researcher at Kaiser Permanente Northern California in Oakland and chair of the ADA’s Professional Practice Committee, the 14-member panel that produced the revised standards.

Dr. Richard W. Grant

“We agreed [with the 2013 ACC and AHA lipid guidelines] that the decision to start a statin should be based on a patient’s cardiovascular disease risk, and it turns out that nearly every patient with type 2 diabetes should be on a statin,” Dr. Grant said in an interview.

The revised standards recommend a “moderate” statin dosage for patients with diabetes who are aged 40-75 years, as well as those who are older than 75 years even if they have no other cardiovascular disease risk factors (Diabetes Care 2015;38:S1-S94).

The dosage should be intensified to “high” for patients with diagnosed cardiovascular disease, and for patients aged 40-75 years with other cardiovascular disease risk factors. For patients older than 75 years with cardiovascular disease risk factors, the new revision calls for either a moderate or high dosage.

However, for patients younger than 40 years with no cardiovascular disease or risk factors, the revised standards call for no statin treatment, a moderate or high dosage for patients younger than 40 years with risk factors, and a high dosage for those with cardiovascular disease.

The ADA’s recommendation for no statin treatment of the youngest and lowest-risk patients with diabetes is somewhat at odds with the 2013 ACC and AHA recommendations. For this patient group, those recommendations said, “statin therapy should be individualized on the basis of considerations of atherosclerotic cardiovascular disease risk-reduction benefits, the potential for adverse effects and drug-drug interactions, and patient preferences.”

The new standards revision contains several other changes, including:

• The recommended goal diastolic blood pressure for patients with diabetes was revised to less than 90 mm Hg, an increase from the 80–mm Hg target that had been in place. That change follows a revision in the ADA’s 2014 standards that increased the systolic blood pressure target to less than 140 mm Hg.

Changing the diastolic target to less than 90 mm Hg was primarily a matter of following the best evidence that exists in the literature, Dr. Grant said, because only lower-grade evidence supports a target of less than 80 mm Hg.

The revised standards also note that the new targets of less than 140/90 mm Hg put the standards “ in harmonization” with the 2014 recommendations of the panel originally assembled at the Eighth Joint National Committee (JAMA 2014;311:507-20).

• The recommended blood glucose target when measured before eating is now 80-130 mg/dL, with the lower limit increased from 70 mg/dL. That change reflects new data that correlate blood glucose levels with blood levels of hemoglobin A1c.

• The revision sets the body mass index cutpoint for screening overweight or obese Asian Americans at 23 kg/m2, an increase from the prior cutpoint of 25 kg/m2.

• A new section devoted to managing patients with diabetes during pregnancy draws together information that previously had been scattered throughout the standards document, Dr. Grant explained. The section discusses gestational diabetes management, as well as managing women who had preexisting type 1 or type 2 diabetes prior to becoming pregnant.

Dr. Grant had no disclosures.

[email protected]

On Twitter @mitchelzoler

References

Click for Credit Link
Body

The efficacy of a moderate statin dosage for primary prevention of cardiovascular disease events in patients age 40-75 years with type 2 diabetes and no other risk factors was clearly established a decade ago by results from the Collaborative Atorvastatin Diabetes Study (CARDS) (Lancet 2004;364:685-96).

No prospective, randomized study has proved the efficacy of statin treatment in patients younger than 40 years with diabetes and no other risk factors; but we see increasing numbers of these patients, and they, too, are at high risk for cardiovascular disease events. I agree with the 2013 recommendation from the American College of Cardiology and American Heart Association that statin treatment should be discussed and in many cases started for these younger, lower-risk patients who still face an important cardiovascular disease risk from their diabetes alone.

Changing the target diastolic blood pressure to less than 90 mm Hg is also consistent with existing evidence. A few years ago, I wrote in an editorial that some prior blood pressure targets for patients with diabetes had been set too low (Circulation 2011;123:2776-8).

There is no evidence that patients with diabetes will benefit from a diastolic blood pressure target that is lower than less than 90 mm Hg, and an overly aggressive approach to blood pressure reduction potentially can cause adverse events. Elderly patients with diabetes often have “silent” coronary artery disease, and if their diastolic pressure goes too low, they can have inadequate coronary perfusion that will cause coronary ischemia.

Dr. Prakash Deedwania

But the diastolic blood pressure target also needs individualization. Some patients, such as those with Asian ethnicity, may benefit from the greater stroke reduction achieved with more aggressive blood pressure reduction.

Aspirin use in patients with diabetes and no other cardiovascular disease risk factors has been controversial, but recent evidence from the Japanese Primary Prevention Project suggests it does not benefit patients with diabetes, even if they may also have hypertension, dyslipidemia, or both. About a third of the patients aged 60-85 years enrolled in this Japanese study had diabetes, more than 70% had dyslipidemia, and 85% had hypertension. But despite this background, daily low-dose aspirin did not reduce the incidence of atherosclerotic cardiovascular disease events during 5 years of follow-up of more than 14,000 randomized patients (JAMA 2014;312:2510-20).

Dr. Prakash C. Deedwania is professor of medicine at the University of California, San Francisco, and director of cardiology at the VA Central California Health Care System in Fresno. He made these comments in an interview. He has served as a consultant to several drug companies that market statins.

Author and Disclosure Information

Publications
Topics
Legacy Keywords
diabetes, American Diabetes Association, Standards of Medical Care, Grant, Deedwania
Sections
Click for Credit Link
Click for Credit Link
Author and Disclosure Information

Author and Disclosure Information

Body

The efficacy of a moderate statin dosage for primary prevention of cardiovascular disease events in patients age 40-75 years with type 2 diabetes and no other risk factors was clearly established a decade ago by results from the Collaborative Atorvastatin Diabetes Study (CARDS) (Lancet 2004;364:685-96).

No prospective, randomized study has proved the efficacy of statin treatment in patients younger than 40 years with diabetes and no other risk factors; but we see increasing numbers of these patients, and they, too, are at high risk for cardiovascular disease events. I agree with the 2013 recommendation from the American College of Cardiology and American Heart Association that statin treatment should be discussed and in many cases started for these younger, lower-risk patients who still face an important cardiovascular disease risk from their diabetes alone.

Changing the target diastolic blood pressure to less than 90 mm Hg is also consistent with existing evidence. A few years ago, I wrote in an editorial that some prior blood pressure targets for patients with diabetes had been set too low (Circulation 2011;123:2776-8).

There is no evidence that patients with diabetes will benefit from a diastolic blood pressure target that is lower than less than 90 mm Hg, and an overly aggressive approach to blood pressure reduction potentially can cause adverse events. Elderly patients with diabetes often have “silent” coronary artery disease, and if their diastolic pressure goes too low, they can have inadequate coronary perfusion that will cause coronary ischemia.

Dr. Prakash Deedwania

But the diastolic blood pressure target also needs individualization. Some patients, such as those with Asian ethnicity, may benefit from the greater stroke reduction achieved with more aggressive blood pressure reduction.

Aspirin use in patients with diabetes and no other cardiovascular disease risk factors has been controversial, but recent evidence from the Japanese Primary Prevention Project suggests it does not benefit patients with diabetes, even if they may also have hypertension, dyslipidemia, or both. About a third of the patients aged 60-85 years enrolled in this Japanese study had diabetes, more than 70% had dyslipidemia, and 85% had hypertension. But despite this background, daily low-dose aspirin did not reduce the incidence of atherosclerotic cardiovascular disease events during 5 years of follow-up of more than 14,000 randomized patients (JAMA 2014;312:2510-20).

Dr. Prakash C. Deedwania is professor of medicine at the University of California, San Francisco, and director of cardiology at the VA Central California Health Care System in Fresno. He made these comments in an interview. He has served as a consultant to several drug companies that market statins.

Body

The efficacy of a moderate statin dosage for primary prevention of cardiovascular disease events in patients age 40-75 years with type 2 diabetes and no other risk factors was clearly established a decade ago by results from the Collaborative Atorvastatin Diabetes Study (CARDS) (Lancet 2004;364:685-96).

No prospective, randomized study has proved the efficacy of statin treatment in patients younger than 40 years with diabetes and no other risk factors; but we see increasing numbers of these patients, and they, too, are at high risk for cardiovascular disease events. I agree with the 2013 recommendation from the American College of Cardiology and American Heart Association that statin treatment should be discussed and in many cases started for these younger, lower-risk patients who still face an important cardiovascular disease risk from their diabetes alone.

Changing the target diastolic blood pressure to less than 90 mm Hg is also consistent with existing evidence. A few years ago, I wrote in an editorial that some prior blood pressure targets for patients with diabetes had been set too low (Circulation 2011;123:2776-8).

There is no evidence that patients with diabetes will benefit from a diastolic blood pressure target that is lower than less than 90 mm Hg, and an overly aggressive approach to blood pressure reduction potentially can cause adverse events. Elderly patients with diabetes often have “silent” coronary artery disease, and if their diastolic pressure goes too low, they can have inadequate coronary perfusion that will cause coronary ischemia.

Dr. Prakash Deedwania

But the diastolic blood pressure target also needs individualization. Some patients, such as those with Asian ethnicity, may benefit from the greater stroke reduction achieved with more aggressive blood pressure reduction.

Aspirin use in patients with diabetes and no other cardiovascular disease risk factors has been controversial, but recent evidence from the Japanese Primary Prevention Project suggests it does not benefit patients with diabetes, even if they may also have hypertension, dyslipidemia, or both. About a third of the patients aged 60-85 years enrolled in this Japanese study had diabetes, more than 70% had dyslipidemia, and 85% had hypertension. But despite this background, daily low-dose aspirin did not reduce the incidence of atherosclerotic cardiovascular disease events during 5 years of follow-up of more than 14,000 randomized patients (JAMA 2014;312:2510-20).

Dr. Prakash C. Deedwania is professor of medicine at the University of California, San Francisco, and director of cardiology at the VA Central California Health Care System in Fresno. He made these comments in an interview. He has served as a consultant to several drug companies that market statins.

Title
Statin treatment benefits most diabetes patients
Statin treatment benefits most diabetes patients

Most patients with diabetes should receive at least a moderate statin dosage regardless of their cardiovascular disease risk profile, according to the American Diabetes Association’s annual update to standards for managing patients with diabetes.

“Standards of Medical Care in Diabetes–2015” also shifts the ADA’s official recommendation on assessing patients for statin treatment from a decision based on blood levels of low density lipoprotein (LDL) cholesterol to a risk-based assessment. That change brings the ADA’s position in line with the approach advocated in late 2013 by guidelines from the American College of Cardiology (ACC) and the American Heart Association (AHA) (J. Am. Coll. Cardiol. 2014;63:2889-934).

The ADA released the revised standards online Dec. 23.

The statin use recommendation is “a major change, a fairly big change in how we provide care, although not that big a change in what most patients are prescribed,” said Dr. Richard W. Grant, a primary care physician and researcher at Kaiser Permanente Northern California in Oakland and chair of the ADA’s Professional Practice Committee, the 14-member panel that produced the revised standards.

Dr. Richard W. Grant

“We agreed [with the 2013 ACC and AHA lipid guidelines] that the decision to start a statin should be based on a patient’s cardiovascular disease risk, and it turns out that nearly every patient with type 2 diabetes should be on a statin,” Dr. Grant said in an interview.

The revised standards recommend a “moderate” statin dosage for patients with diabetes who are aged 40-75 years, as well as those who are older than 75 years even if they have no other cardiovascular disease risk factors (Diabetes Care 2015;38:S1-S94).

The dosage should be intensified to “high” for patients with diagnosed cardiovascular disease, and for patients aged 40-75 years with other cardiovascular disease risk factors. For patients older than 75 years with cardiovascular disease risk factors, the new revision calls for either a moderate or high dosage.

However, for patients younger than 40 years with no cardiovascular disease or risk factors, the revised standards call for no statin treatment, a moderate or high dosage for patients younger than 40 years with risk factors, and a high dosage for those with cardiovascular disease.

The ADA’s recommendation for no statin treatment of the youngest and lowest-risk patients with diabetes is somewhat at odds with the 2013 ACC and AHA recommendations. For this patient group, those recommendations said, “statin therapy should be individualized on the basis of considerations of atherosclerotic cardiovascular disease risk-reduction benefits, the potential for adverse effects and drug-drug interactions, and patient preferences.”

The new standards revision contains several other changes, including:

• The recommended goal diastolic blood pressure for patients with diabetes was revised to less than 90 mm Hg, an increase from the 80–mm Hg target that had been in place. That change follows a revision in the ADA’s 2014 standards that increased the systolic blood pressure target to less than 140 mm Hg.

Changing the diastolic target to less than 90 mm Hg was primarily a matter of following the best evidence that exists in the literature, Dr. Grant said, because only lower-grade evidence supports a target of less than 80 mm Hg.

The revised standards also note that the new targets of less than 140/90 mm Hg put the standards “ in harmonization” with the 2014 recommendations of the panel originally assembled at the Eighth Joint National Committee (JAMA 2014;311:507-20).

• The recommended blood glucose target when measured before eating is now 80-130 mg/dL, with the lower limit increased from 70 mg/dL. That change reflects new data that correlate blood glucose levels with blood levels of hemoglobin A1c.

• The revision sets the body mass index cutpoint for screening overweight or obese Asian Americans at 23 kg/m2, an increase from the prior cutpoint of 25 kg/m2.

• A new section devoted to managing patients with diabetes during pregnancy draws together information that previously had been scattered throughout the standards document, Dr. Grant explained. The section discusses gestational diabetes management, as well as managing women who had preexisting type 1 or type 2 diabetes prior to becoming pregnant.

Dr. Grant had no disclosures.

[email protected]

On Twitter @mitchelzoler

Most patients with diabetes should receive at least a moderate statin dosage regardless of their cardiovascular disease risk profile, according to the American Diabetes Association’s annual update to standards for managing patients with diabetes.

“Standards of Medical Care in Diabetes–2015” also shifts the ADA’s official recommendation on assessing patients for statin treatment from a decision based on blood levels of low density lipoprotein (LDL) cholesterol to a risk-based assessment. That change brings the ADA’s position in line with the approach advocated in late 2013 by guidelines from the American College of Cardiology (ACC) and the American Heart Association (AHA) (J. Am. Coll. Cardiol. 2014;63:2889-934).

The ADA released the revised standards online Dec. 23.

The statin use recommendation is “a major change, a fairly big change in how we provide care, although not that big a change in what most patients are prescribed,” said Dr. Richard W. Grant, a primary care physician and researcher at Kaiser Permanente Northern California in Oakland and chair of the ADA’s Professional Practice Committee, the 14-member panel that produced the revised standards.

Dr. Richard W. Grant

“We agreed [with the 2013 ACC and AHA lipid guidelines] that the decision to start a statin should be based on a patient’s cardiovascular disease risk, and it turns out that nearly every patient with type 2 diabetes should be on a statin,” Dr. Grant said in an interview.

The revised standards recommend a “moderate” statin dosage for patients with diabetes who are aged 40-75 years, as well as those who are older than 75 years even if they have no other cardiovascular disease risk factors (Diabetes Care 2015;38:S1-S94).

The dosage should be intensified to “high” for patients with diagnosed cardiovascular disease, and for patients aged 40-75 years with other cardiovascular disease risk factors. For patients older than 75 years with cardiovascular disease risk factors, the new revision calls for either a moderate or high dosage.

However, for patients younger than 40 years with no cardiovascular disease or risk factors, the revised standards call for no statin treatment, a moderate or high dosage for patients younger than 40 years with risk factors, and a high dosage for those with cardiovascular disease.

The ADA’s recommendation for no statin treatment of the youngest and lowest-risk patients with diabetes is somewhat at odds with the 2013 ACC and AHA recommendations. For this patient group, those recommendations said, “statin therapy should be individualized on the basis of considerations of atherosclerotic cardiovascular disease risk-reduction benefits, the potential for adverse effects and drug-drug interactions, and patient preferences.”

The new standards revision contains several other changes, including:

• The recommended goal diastolic blood pressure for patients with diabetes was revised to less than 90 mm Hg, an increase from the 80–mm Hg target that had been in place. That change follows a revision in the ADA’s 2014 standards that increased the systolic blood pressure target to less than 140 mm Hg.

Changing the diastolic target to less than 90 mm Hg was primarily a matter of following the best evidence that exists in the literature, Dr. Grant said, because only lower-grade evidence supports a target of less than 80 mm Hg.

The revised standards also note that the new targets of less than 140/90 mm Hg put the standards “ in harmonization” with the 2014 recommendations of the panel originally assembled at the Eighth Joint National Committee (JAMA 2014;311:507-20).

• The recommended blood glucose target when measured before eating is now 80-130 mg/dL, with the lower limit increased from 70 mg/dL. That change reflects new data that correlate blood glucose levels with blood levels of hemoglobin A1c.

• The revision sets the body mass index cutpoint for screening overweight or obese Asian Americans at 23 kg/m2, an increase from the prior cutpoint of 25 kg/m2.

• A new section devoted to managing patients with diabetes during pregnancy draws together information that previously had been scattered throughout the standards document, Dr. Grant explained. The section discusses gestational diabetes management, as well as managing women who had preexisting type 1 or type 2 diabetes prior to becoming pregnant.

Dr. Grant had no disclosures.

[email protected]

On Twitter @mitchelzoler

References

References

Publications
Publications
Topics
Article Type
Display Headline
ADA’s revised diabetes 'standards' broaden statin use
Display Headline
ADA’s revised diabetes 'standards' broaden statin use
Legacy Keywords
diabetes, American Diabetes Association, Standards of Medical Care, Grant, Deedwania
Legacy Keywords
diabetes, American Diabetes Association, Standards of Medical Care, Grant, Deedwania
Sections
Article Source

FROM DIABETES CARE

PURLs Copyright

Inside the Article

Consider a mandibular positioning device to alleviate sleep-disordered breathing

Article Type
Changed
Tue, 12/11/2018 - 15:18
Display Headline
Consider a mandibular positioning device to alleviate sleep-disordered breathing

Snoring, snorting, gasping, and obstruc­tive sleep apnea are caused by col­lapse of the pharyngeal airway during sleep.1 Pathophysiology includes a combi­nation of anatomical and physiological vari­ables.1 Common anatomical predisposing conditions include abnormalities of pharyn­geal, lingual, and dental arches; physiologi­cal concerns are advancing age, male sex, obesity, use of sedatives, body positioning, and reduced muscle tone during rapid eye movement sleep. Coexistence of anatomic and physiological elements can produce significant narrowing of the upper airway.

Comorbidities include vascular, meta­bolic, and psychiatric conditions. As many as one-third of people with symptoms of sleep apnea report depressed mood; approx­imately 10% of these patients meet criteria for moderate or severe depression.2

In short, sleep-disordered breathing has a globally negative effect on mental health.


When should you consider obtaining a sleep apnea study?

Refer patients for a sleep study when snor­ing, snorting, gasping, or pauses in breathing occur during sleep, or in the case of daytime sleepiness, fatigue, or unrefreshing sleep that cannot be explained by another medical or psychiatric illness.2 A sleep specialist can determine the most appropriate intervention for sleep-disordered breathing.

An apneic event is characterized by complete cessation of airflow; hypopnea is a partially compromised airway. In either event, at least a 3% decrease in oxygen saturation occurs for at least 10 seconds.3 A diagnosis of obstructive sleep apnea or hypopnea is required when polysomnography reveals either of:
   • ≥5 episodes of apnea or hypopnea, or both, per hour of sleep, with symptoms of a rhythmic breathing disturbance or daytime sleepiness or fatigue
   • ≥15 episodes of apnea or hypopnea, or both, per hour of sleep, regardless of accom­panying symptoms.2


What are the treatment options?
 
   • Continuous positive airway pressure (CPAP) machines.
   • Surgical procedures include adeno-tonsillectomy in children and surgical maxilla-mandibular advancement or pala­tal implants for adults.
   • A novel implantable electrical stimu­lation device stimulates the hypoglossal nerve, which activates the genioglossus muscle, thus moving the tongue forward to open the airway.
   • An anterior mandibular positioning (AMP) device increases the diameter of the retroglossal space by preventing posterior movement of the mandible and tongue, thereby limiting encroachment on the air­way diameter and reducing the potential for collapse.1-4


When should you recommend an AMP device?

Consider recommending an AMP device to treat sleep-disordered breathing when (1) lifestyle changes, such as sleep hygiene, weight loss, and stopping sedatives, do not work and (2) a CPAP machine or a surgical procedure is contraindicated or has been ineffective.1 An AMP device can minimize snoring and relieve airway obstruction, especially in patients with supine position-related apnea.4 To keep the airway open in non-supine position-related cases, an AMP device might be indicated in addition to CPAP delivered nasally.1

This plastic oral appliance is either a 1- or 2-piece design, and looks and is sized simi­larly to an athletic mouth-protection guard or an oral anti-bruxism tooth-protection appliance. It is affixed to the mandible and maxillary arches by clasps (Figure).




An AMP device often is most beneficial for supine-dependent sleep apnea patients and those with loud snoring, without sleep apnea.4 Response is best in young adults and in patients who have a low body mass index, are free of sedatives, and have appropriate cephalometrics of the oral, dental, or pha­ryngeal anatomy. Improved sleep architec­ture, continuous sleep with less snoring, and increased daytime alertness are observed in patients who respond to an AMP device.

An AMP device is contraindicated when the device cannot be affixed to the dental arches and in some patients with an anatom­ical or pain-related temporomandibular joint disorder.5 The device is easy to use, nonin­vasive, readily accessible, and less expensive than alternatives.3


How can you help maintain treatment adherence?
AMP devices can induce adverse effects, including dental pain or discomfort through orthodontic alterations; patient reports and follow-up can yield detection and device adjustments can alleviate such problems. Adherence generally is good, with complaints usually limited to minor tooth discomfort, occlusive changes, and increased or decreased salivation.5 In our clinical experience, many patients find these devices comfortable and easy to use, but might complain of feeling awkward when wearing them.

Changes in occlusion can occur during long-term treatment with an AMP device. Proper fitting is essential to facilitate a more open airway and the ability to speak and drink fluids, and to maintain safety, even if vomiting occurs while the device is in place.

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

References


1. Epstein LJ, Kristo D, Strollo PJ, et al. Clinical guideline for the evaluation, management and long-term care of obstructive sleep apnea in adults. J Clin Sleep Med. 2009;5(3):263-276.
2. Diagnostic and statistical manual of mental disorders, 5th ed. Washington, DC: American Psychiatric Association; 2013.
3. de Britto Teixeira AO, Abi-Ramia LB, de Oliveira Almeida MA. Treatment of obstructive sleep apnea with oral appliances. Prog Orthod. 2013;14:10.
4. Marklund M, Stenlund H, Franklin K. Mandibular advancement devices in 630 men and women with obstructive sleep apnea and snoring: tolerability and predictors of treatment success. Chest. 2004;125(4):1270-1278.
5. Ferguson KA, Cartwright R, Rogers R, et al. Oral appliances for snoring and obstructive sleep apnea: a review. Sleep. 2006;29(2):244-262.

Article PDF
Author and Disclosure Information

Anoop Narahari, MD
Observer Physician

Shama Panjwani, MD
Observer Physician

Amandeep Bains, MD
Observer Physician

Steven Lippmann, MD
Professor
Department of Psychiatry and Behavioral Sciences
University of Louisville School of Medicine
Louisville, Kentucky

Issue
Current Psychiatry - 14(1)
Publications
Topics
Page Number
50-51
Legacy Keywords
mandibular positioning device, sleep-disordered breathing, sleep disordered breathing, sleep disorders, sleep apnea, snoring, snorting, gasping, obstructive sleep apnea
Sections
Author and Disclosure Information

Anoop Narahari, MD
Observer Physician

Shama Panjwani, MD
Observer Physician

Amandeep Bains, MD
Observer Physician

Steven Lippmann, MD
Professor
Department of Psychiatry and Behavioral Sciences
University of Louisville School of Medicine
Louisville, Kentucky

Author and Disclosure Information

Anoop Narahari, MD
Observer Physician

Shama Panjwani, MD
Observer Physician

Amandeep Bains, MD
Observer Physician

Steven Lippmann, MD
Professor
Department of Psychiatry and Behavioral Sciences
University of Louisville School of Medicine
Louisville, Kentucky

Article PDF
Article PDF

Snoring, snorting, gasping, and obstruc­tive sleep apnea are caused by col­lapse of the pharyngeal airway during sleep.1 Pathophysiology includes a combi­nation of anatomical and physiological vari­ables.1 Common anatomical predisposing conditions include abnormalities of pharyn­geal, lingual, and dental arches; physiologi­cal concerns are advancing age, male sex, obesity, use of sedatives, body positioning, and reduced muscle tone during rapid eye movement sleep. Coexistence of anatomic and physiological elements can produce significant narrowing of the upper airway.

Comorbidities include vascular, meta­bolic, and psychiatric conditions. As many as one-third of people with symptoms of sleep apnea report depressed mood; approx­imately 10% of these patients meet criteria for moderate or severe depression.2

In short, sleep-disordered breathing has a globally negative effect on mental health.


When should you consider obtaining a sleep apnea study?

Refer patients for a sleep study when snor­ing, snorting, gasping, or pauses in breathing occur during sleep, or in the case of daytime sleepiness, fatigue, or unrefreshing sleep that cannot be explained by another medical or psychiatric illness.2 A sleep specialist can determine the most appropriate intervention for sleep-disordered breathing.

An apneic event is characterized by complete cessation of airflow; hypopnea is a partially compromised airway. In either event, at least a 3% decrease in oxygen saturation occurs for at least 10 seconds.3 A diagnosis of obstructive sleep apnea or hypopnea is required when polysomnography reveals either of:
   • ≥5 episodes of apnea or hypopnea, or both, per hour of sleep, with symptoms of a rhythmic breathing disturbance or daytime sleepiness or fatigue
   • ≥15 episodes of apnea or hypopnea, or both, per hour of sleep, regardless of accom­panying symptoms.2


What are the treatment options?
 
   • Continuous positive airway pressure (CPAP) machines.
   • Surgical procedures include adeno-tonsillectomy in children and surgical maxilla-mandibular advancement or pala­tal implants for adults.
   • A novel implantable electrical stimu­lation device stimulates the hypoglossal nerve, which activates the genioglossus muscle, thus moving the tongue forward to open the airway.
   • An anterior mandibular positioning (AMP) device increases the diameter of the retroglossal space by preventing posterior movement of the mandible and tongue, thereby limiting encroachment on the air­way diameter and reducing the potential for collapse.1-4


When should you recommend an AMP device?

Consider recommending an AMP device to treat sleep-disordered breathing when (1) lifestyle changes, such as sleep hygiene, weight loss, and stopping sedatives, do not work and (2) a CPAP machine or a surgical procedure is contraindicated or has been ineffective.1 An AMP device can minimize snoring and relieve airway obstruction, especially in patients with supine position-related apnea.4 To keep the airway open in non-supine position-related cases, an AMP device might be indicated in addition to CPAP delivered nasally.1

This plastic oral appliance is either a 1- or 2-piece design, and looks and is sized simi­larly to an athletic mouth-protection guard or an oral anti-bruxism tooth-protection appliance. It is affixed to the mandible and maxillary arches by clasps (Figure).




An AMP device often is most beneficial for supine-dependent sleep apnea patients and those with loud snoring, without sleep apnea.4 Response is best in young adults and in patients who have a low body mass index, are free of sedatives, and have appropriate cephalometrics of the oral, dental, or pha­ryngeal anatomy. Improved sleep architec­ture, continuous sleep with less snoring, and increased daytime alertness are observed in patients who respond to an AMP device.

An AMP device is contraindicated when the device cannot be affixed to the dental arches and in some patients with an anatom­ical or pain-related temporomandibular joint disorder.5 The device is easy to use, nonin­vasive, readily accessible, and less expensive than alternatives.3


How can you help maintain treatment adherence?
AMP devices can induce adverse effects, including dental pain or discomfort through orthodontic alterations; patient reports and follow-up can yield detection and device adjustments can alleviate such problems. Adherence generally is good, with complaints usually limited to minor tooth discomfort, occlusive changes, and increased or decreased salivation.5 In our clinical experience, many patients find these devices comfortable and easy to use, but might complain of feeling awkward when wearing them.

Changes in occlusion can occur during long-term treatment with an AMP device. Proper fitting is essential to facilitate a more open airway and the ability to speak and drink fluids, and to maintain safety, even if vomiting occurs while the device is in place.

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

Snoring, snorting, gasping, and obstruc­tive sleep apnea are caused by col­lapse of the pharyngeal airway during sleep.1 Pathophysiology includes a combi­nation of anatomical and physiological vari­ables.1 Common anatomical predisposing conditions include abnormalities of pharyn­geal, lingual, and dental arches; physiologi­cal concerns are advancing age, male sex, obesity, use of sedatives, body positioning, and reduced muscle tone during rapid eye movement sleep. Coexistence of anatomic and physiological elements can produce significant narrowing of the upper airway.

Comorbidities include vascular, meta­bolic, and psychiatric conditions. As many as one-third of people with symptoms of sleep apnea report depressed mood; approx­imately 10% of these patients meet criteria for moderate or severe depression.2

In short, sleep-disordered breathing has a globally negative effect on mental health.


When should you consider obtaining a sleep apnea study?

Refer patients for a sleep study when snor­ing, snorting, gasping, or pauses in breathing occur during sleep, or in the case of daytime sleepiness, fatigue, or unrefreshing sleep that cannot be explained by another medical or psychiatric illness.2 A sleep specialist can determine the most appropriate intervention for sleep-disordered breathing.

An apneic event is characterized by complete cessation of airflow; hypopnea is a partially compromised airway. In either event, at least a 3% decrease in oxygen saturation occurs for at least 10 seconds.3 A diagnosis of obstructive sleep apnea or hypopnea is required when polysomnography reveals either of:
   • ≥5 episodes of apnea or hypopnea, or both, per hour of sleep, with symptoms of a rhythmic breathing disturbance or daytime sleepiness or fatigue
   • ≥15 episodes of apnea or hypopnea, or both, per hour of sleep, regardless of accom­panying symptoms.2


What are the treatment options?
 
   • Continuous positive airway pressure (CPAP) machines.
   • Surgical procedures include adeno-tonsillectomy in children and surgical maxilla-mandibular advancement or pala­tal implants for adults.
   • A novel implantable electrical stimu­lation device stimulates the hypoglossal nerve, which activates the genioglossus muscle, thus moving the tongue forward to open the airway.
   • An anterior mandibular positioning (AMP) device increases the diameter of the retroglossal space by preventing posterior movement of the mandible and tongue, thereby limiting encroachment on the air­way diameter and reducing the potential for collapse.1-4


When should you recommend an AMP device?

Consider recommending an AMP device to treat sleep-disordered breathing when (1) lifestyle changes, such as sleep hygiene, weight loss, and stopping sedatives, do not work and (2) a CPAP machine or a surgical procedure is contraindicated or has been ineffective.1 An AMP device can minimize snoring and relieve airway obstruction, especially in patients with supine position-related apnea.4 To keep the airway open in non-supine position-related cases, an AMP device might be indicated in addition to CPAP delivered nasally.1

This plastic oral appliance is either a 1- or 2-piece design, and looks and is sized simi­larly to an athletic mouth-protection guard or an oral anti-bruxism tooth-protection appliance. It is affixed to the mandible and maxillary arches by clasps (Figure).




An AMP device often is most beneficial for supine-dependent sleep apnea patients and those with loud snoring, without sleep apnea.4 Response is best in young adults and in patients who have a low body mass index, are free of sedatives, and have appropriate cephalometrics of the oral, dental, or pha­ryngeal anatomy. Improved sleep architec­ture, continuous sleep with less snoring, and increased daytime alertness are observed in patients who respond to an AMP device.

An AMP device is contraindicated when the device cannot be affixed to the dental arches and in some patients with an anatom­ical or pain-related temporomandibular joint disorder.5 The device is easy to use, nonin­vasive, readily accessible, and less expensive than alternatives.3


How can you help maintain treatment adherence?
AMP devices can induce adverse effects, including dental pain or discomfort through orthodontic alterations; patient reports and follow-up can yield detection and device adjustments can alleviate such problems. Adherence generally is good, with complaints usually limited to minor tooth discomfort, occlusive changes, and increased or decreased salivation.5 In our clinical experience, many patients find these devices comfortable and easy to use, but might complain of feeling awkward when wearing them.

Changes in occlusion can occur during long-term treatment with an AMP device. Proper fitting is essential to facilitate a more open airway and the ability to speak and drink fluids, and to maintain safety, even if vomiting occurs while the device is in place.

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

References


1. Epstein LJ, Kristo D, Strollo PJ, et al. Clinical guideline for the evaluation, management and long-term care of obstructive sleep apnea in adults. J Clin Sleep Med. 2009;5(3):263-276.
2. Diagnostic and statistical manual of mental disorders, 5th ed. Washington, DC: American Psychiatric Association; 2013.
3. de Britto Teixeira AO, Abi-Ramia LB, de Oliveira Almeida MA. Treatment of obstructive sleep apnea with oral appliances. Prog Orthod. 2013;14:10.
4. Marklund M, Stenlund H, Franklin K. Mandibular advancement devices in 630 men and women with obstructive sleep apnea and snoring: tolerability and predictors of treatment success. Chest. 2004;125(4):1270-1278.
5. Ferguson KA, Cartwright R, Rogers R, et al. Oral appliances for snoring and obstructive sleep apnea: a review. Sleep. 2006;29(2):244-262.

References


1. Epstein LJ, Kristo D, Strollo PJ, et al. Clinical guideline for the evaluation, management and long-term care of obstructive sleep apnea in adults. J Clin Sleep Med. 2009;5(3):263-276.
2. Diagnostic and statistical manual of mental disorders, 5th ed. Washington, DC: American Psychiatric Association; 2013.
3. de Britto Teixeira AO, Abi-Ramia LB, de Oliveira Almeida MA. Treatment of obstructive sleep apnea with oral appliances. Prog Orthod. 2013;14:10.
4. Marklund M, Stenlund H, Franklin K. Mandibular advancement devices in 630 men and women with obstructive sleep apnea and snoring: tolerability and predictors of treatment success. Chest. 2004;125(4):1270-1278.
5. Ferguson KA, Cartwright R, Rogers R, et al. Oral appliances for snoring and obstructive sleep apnea: a review. Sleep. 2006;29(2):244-262.

Issue
Current Psychiatry - 14(1)
Issue
Current Psychiatry - 14(1)
Page Number
50-51
Page Number
50-51
Publications
Publications
Topics
Article Type
Display Headline
Consider a mandibular positioning device to alleviate sleep-disordered breathing
Display Headline
Consider a mandibular positioning device to alleviate sleep-disordered breathing
Legacy Keywords
mandibular positioning device, sleep-disordered breathing, sleep disordered breathing, sleep disorders, sleep apnea, snoring, snorting, gasping, obstructive sleep apnea
Legacy Keywords
mandibular positioning device, sleep-disordered breathing, sleep disordered breathing, sleep disorders, sleep apnea, snoring, snorting, gasping, obstructive sleep apnea
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media

Young, pregnant, ataxic—and jilted

Article Type
Changed
Tue, 12/11/2018 - 15:29
Display Headline
Young, pregnant, ataxic—and jilted

CASE Difficulty walking
Ms. M, age 15, is a pregnant, Spanish-speaking Guatemalan woman who is brought to obstet­rics triage in a large academic medical center at 35 weeks gestational age. She complains of dizziness, tinnitus, left orbital headache, and difficulty walking.

The neurology service finds profound trun­cal ataxia, astasia-abasia, and buckling of the knees; a normal brain and spine MRI are not consistent with a neurologic etiology. Otolaryngology service evaluates Ms. M to rule out a cholesteatoma and suggests a head CT and endoscopy, which are normal.

Ms. M’s symptoms resolve after 3 days, although the gait disturbances persist. When no clear cause is found for her difficulty walk­ing, the psychiatry service is consulted to evalu­ate whether an underlying psychiatric disorder is contributing to symptoms.


What could be causing Ms. M’s symptoms?

   a) malingering
   b) factitious disorder
   c) undiagnosed neurologic disorder
   d) conversion disorder


The authors’ observations

Women are vulnerable to a variety of psy­chiatric illnesses during pregnancy1 that have deleterious effects on mother, baby, and family.2-6 Although there is a burgeoning literature on affective and anxiety disorders occurring in pregnancy, there is a dearth of information about somatoform disorders.


HISTORY
Abandonment

Ms. M reports that, although her boyfriend deserted her after learning about the unexpected pregnancy, she will welcome the baby and looks forward to motherhood. She seems unaware of the responsibilities of being a mother.

Ms. M acknowledges a history of depression and self-harm a few years earlier, yet says she feels better now and thinks that psychiatric care is unnecessary. Because she does not endorse a history of trauma or symptoms suggesting an affective, anxiety, or psychotic illness, the psy­chiatrist does not recommend treatment with psychotropic medication. 

At age 5, Ms. M’s parents sent her to the United States with her aunt, hoping that she would have a better life than she would have had in Guatemala. Her aunt reports that Ms. M initially had difficulty adjusting to life in the United States without her parents, yet she has made substantial strides over the years and is now quite accustomed to the country. Her aunt describes Ms. M as an independent high school student who earns good grades.

During the interview, the psychiatrist observes that Ms. M exhibits childlike manner­isms, including sleeping with stuffed toys and coloring in Disney books with crayons. She also is indifferent to her gait difficulty, pregnancy, and psychosocial stressors. Her affect is incon­sistent with the content of her speech and she is alexithymic.

Ms. M’s aunt reports that her niece is becom­ing more dependent on her, which is not con­sistent with her baseline. Her aunt also notes that several years earlier, Ms. M’s nephew was diagnosed with a cholesteatoma after he pre­sented with similar symptoms.

The combination of (1) Ms. M’s clinical pre­sentation, which was causing her significant impairment in her social functioning, (2) the incompatibility of symptoms with any recog­nized neurologic and medical disease, and (3) prior family experience with cholesteatoma leads the consulting psychiatrist to suspect conversion disorder. Ms. M’s alexithymia, indif­ference to her symptoms, and recent aban­donment by the baby’s father also support a conversion disorder diagnosis.

From a psychodynamic perspective, the ataxia appears to be her way of protecting her­self from the abandonment she is experiencing by being left again to “stand alone” by her boy­friend as she had been when her parents sent her to the United States. Her regressive behav­ior could be her way of securing her aunt’s love and support.


The authors’ observations
This is the first case of psychogenic gait dis­turbance during pregnancy described in the literature. Authors have reported on pseu­dotoxemia,7 hyperemesis gravidarum,8 and pesudocyesis,9 yet there is a paucity of infor­mation on psychogenic gait disturbance during pregnancy. Ms. M’s case elucidates many of the clinical quandaries that occur when managing psychiatric illness—and, more specifically, conversion disorder— during pregnancy. Many women are hesi­tant to seek psychiatric treatment during pregnancy because of shame, stigma, and fear of loss of personal or parental rights10,11; it is not surprising that emotionally dis­tressed women communicate their feel­ings or troubled thoughts through physical symptoms.


Likely diagnosis

Conversion disorder is the presence of neurologic symptoms in the absence of a neurologic diagnosis that fully explains those symptoms. Conversion disorder, previously known as hysteria, is called functional neurologic symptom disorder in DSM-5 (Box).12 Symptoms are not feigned; instead, they represent “conver­sion” of emotional distress into neurologic symptoms.13,14 Although misdiagnosing conversion disorder in patients with true neurologic disease is uncommon, clini­cians often are uncomfortable making the diagnosis until all medical causes have been ruled out.14 It is not always possible to find a psychological explanation for conversion disorder, but a history of child­hood abuse, particularly sexual abuse, could play a role.14




Because of the variety of presenta­tions, clinicians in all specialties should be familiar with somatoform disorders; this is especially important in obstetrics and gynecology because women are more likely than men to develop these disorders.15 It is important to consider that Ms. M is a teen­ager and somatoform disorders can present differently in adults. The diagnostic process should include a diligent somatic workup and a personal and social history to identify the patient’s developmental tasks, stressors, and coping style.15

 

 


How would you treat Ms. M?

   a) destigmatize psychiatric illness and provide psychoeducation regarding treat­ment benefits
   b) identify and treat any comorbid psychiat­ric disorders
   c) maintain a proactive and multidisci­plinary approach that includes assess­ment of psychosocial stressors and psychodynamic factors, particularly those related to the pregnancy
  d) all of the above


TREATMENT
Close follow-up

The psychiatrist recommends continued close psychiatric follow-up as well as multidisci­plinary involvement, including physical ther­apy, neurology, and obstetrics.

Ms. M initially is resistant to psychiatric follow-up because she says that “people on the street” told her that, if she saw a psychia­trist, her baby would be taken away. After the psychiatrist explains that it is unlikely her baby would be taken away, Ms. M immediately appears relieved, smiles, and readily agrees to outpatient psychotherapy.

Over the next 24 hours, she continues to work with a physical therapist and her gait sig­nificantly improves. She is discharged home 2 days later with a walking aid (Zimmer frame) for assistance.

Four days later, however, Ms. M is readmit­ted with worsening ataxia. Her aunt reports that, at home, Ms. M’s regressed behaviors are worsening; she is sleeping in bed with her and had several episodes of enuresis at home.

Ms. M continues to deny psychiatric symp­toms or anxiety about the delivery. Although she shows some improvement when work­ing with physical therapists, they note that Ms. M is still unable to ambulate or stand on her own. The psychiatrist is increasingly con­cerned about her regressed behavior and con­tinued ataxia.

A family meeting is held and the psychia­trist and social worker educate Ms. M and her aunt about conversion disorder, including how some emotionally distressed women communicate their feelings or troubled thoughts through physical symptoms and how that may apply to Ms. M. During the meeting, the team also destigmatizes psy­chiatric illness and treatment and provides psychoeducation regarding its benefits. The psychiatrist and social worker also provide a psychodynamic interpretation that her ataxia could be a way of protecting herself against the abandonment she is experiencing by being left to “stand alone” by her boyfriend— as she had been when her parents sent her to the United States, and that her behavior could be her way of securing her aunt’s love and support.Ms. M and her aunt both readily agree with this interpretation. The aunt notes that her niece is more anxious about motherhood than she acknowledges and is concerned that Ms. M expects her to be the primary care­giver for the baby. Those present note that Ms. M is becoming increasingly dependent on her aunt, and that it is important for her to retain her independence, especially once she becomes a mother.

Ms. M immediately begins to display more affect; she smiles and reports feeling relieved. Similar to the previous admission, her gait sig­nificantly improves over the next 2 days and she is discharged home with a walking aid.


The authors’ observations

A broad differential diagnosis and early multidisciplinary involvement might facilitate earlier diagnosis and treatment.16 Assessment of psychosocial stressors in the patient’s personal and family life, includ­ing circumstances around the pregnancy and the meaning of motherhood, as well as investigation of what the patient may gain from the sick role, are paramount. In Ms. M’s case, cultural background, sepa­ration from her parents at a young age, and recent abandonment by her boyfriend have contributed to her inability to “stand alone,” which manifested as ataxia. Young age, regressed behavior, and her minimi­zation of stressors also point to her dif­ficulty acknowledging and coping with psychosocial stressors.

Successful delivery of the diagnosis is key to treatment success. After building a therapeutic alliance, a multidisciplinary discussion should take place that allows the patient to understand the diagnosis and treatment plan.17,18 The patient and family should be reassured that the fetus is healthy and all organic causes of symptoms have been investigated.17 Although man­agement of conversion disorder during pregnancy is similar to that in non-preg­nant women, several additional avenues of investigation should be considered:
   • Explore the psychodynamic basis of the disorder and the role of the pregnancy and motherhood.
   • Identify any comorbid psychiatric disorders, particularly those specific to pregnancy or the postpartum period.
   • Because of the shame and stigma associated with seeking psychiatric treat­ment during pregnancy,10,11 it is impera­tive to destigmatize treatment and provide psychoeducation regarding its benefits.

A treatment plan can then be developed that involves psychotherapy, psychoedu­cation, stress management, and, when appropriate, pharmacotherapy.17

Providing psychoeducation about postpartum depression and other perina­tal psychiatric illness could be beneficial. Physical therapy often is culturally accept­able and can help re-establish healthy patterns of motor function.19 Ms. M’s gait  showed some improvement with physical therapy as part of the multidisciplinary approach, which also should include a thorough medical workup. Appropriate psychiatric treatment can help patients give up the sick role and return to their previous level of functioning.17

 

 

Maintain close communication with the outpatient perinatal care team as they mon­itor the patient’s parenting capacity. The outpatient perinatal care team also should engage pregnant or postpartum women in prioritizing their emotional well-being and encourage outpatient mental health treatment. Despite a dearth of data on the regressive symptoms and prognosis for future pregnancies, it is important to mon­itor maternal capacity and discuss the pos­sibility of symptom recurrence.


OUTCOME
Healthy baby

Three days later, Ms. M returns in labor with improved gait yet still using a walking aid. She has a normal vaginal delivery of a healthy baby boy at 37 weeks’ gestational age.

After the birth, Ms. M reports feeling well and enjoying motherhood, and denies psy­chiatric symptoms. She is ambulating without assistance within hours of delivery. This spon­taneous resolution of symptoms could have been because of the psychodynamically ori­ented multidisciplinary approach to her care, which may have helped her realize that she did not have to “stand alone” as she embarked on motherhood.

Before being discharged home, Ms. M and her aunt meet with the inpatient obstetric social worker to assess Ms. M’s ability to care for the baby and discuss the importance of continued emotional support. The social worker does not contact the Department of Children and Families because Ms. M is walking independently and not endorsing or exhibiting regressive behaviors. Ms. M also reports that she will ask her aunt to take care of the baby should ataxia recur. Her aunt reassures the social workers that she will encourage Ms. M to attend outpatient psy­chotherapy and will contact the social worker if she becomes concerned about Ms. M’s or the baby’s well-being.

During her postpartum obstetric visit, Ms. M is walking independently and does not exhibit or endorse neurologic symptoms. The social worker provides psychoeducation about the importance of outpatient psycho­therapy and schedules an initial appointment; Ms. M does not attend outpatient psychother­apy after discharge.

Bottom Line
Consider conversion disorder in obstetric patients who present with ataxia without a neurologic cause. Management involves a proactive and multidisciplinary approach that includes a thorough medical workup and assessment of psychosocial stressors and psychodynamic factors, particularly those related to the pregnancy. Early identification and delivery of the diagnosis, destigmatization, and provision of appropriate psychiatric treatment can facilitate treatment success.

Disclosures
Dr. Byatt has received grant funding/support for this project from the National Center for Research Resources and the National Center for Advancing Translational Sciences, National Institutes of Health, through Grant KL2TR000160. The content is solely the responsibility of the authors and does not necessarily represent the official views of the NIH. Dr. Toor reports no financial relationships with any company whose products are mentioned in this article or manufacturers of competing products.

References


1. Vesga-Lopez O, Blanco C, Keyes K, et al. Psychiatric disorders in pregnant and postpartum women in the United States. Arch Gen Psychiatry. 2008;65(7):805-815.
2. Britton HL, Gronwaldt V, Britton JR. Maternal postpartum behaviors and mother-infant relationship during the first year of life. J Pediatr. 2001;138(6):905-909.
3. Deave T, Heron J, Evans J, et al. The impact of maternal depression in pregnancy on early child development. BJOG. 2008;115(8):1043-1051.
4. Paulson JF, Keefe HA, Leiferman JA. Early parental depression and child language development. J Child Psychol Psychiatry. 2009;50(3):254-262.
5. Zuckerman B, Amaro H, Bauchner H, et al. Depressive symptoms during pregnancy: relationship to poor health behaviors. Am J Obstet Gynecol. 1989;160(5 pt 1):1107-1111.
6. Forman DR, O’Hara MW, Stuart S, et al. Effective treatment for postpartum depression is not sufficient to improve the developing mother-child relationship. Dev Psychopathol. 2007;19(2):585-602.
7. Brady WJ Jr, Huff JS. Pseudotoxemia: new onset psychogenic seizure in third trimester pregnancy. J Emerg Med. 1997;15(6):815-820.
8. el-Mallakh RS, Liebowitz NR, Hale MS. Hyperemesis gravidarum as conversion disorder. J Nerv Ment Dis. 1990; 178(10):655-659.
9. Paulman PM, Sadat A. Pseudocyesis. J Fam Pract. 1990;30(5):575-576.
10. Dennis CL, Chung-Lee L. Postpartum depression help-seeking barriers and maternal treatment p: a qualitative systematic review. Birth. 2006;33(4):323-331.
11. Byatt N, Simas TA, Lundquist RS, et al. Strategies for improving perinatal depression treatment in North American outpatient obstetric settings. J Psychosom Obstetr Gynaecol. 2012;33(4):143-161.
12. Diagnostic and statistical manual of mental disorders, 5th ed. Washington, DC: American Psychiatric Association; 2013.
13. Feinstein A. Conversion disorder: advances in our understanding. CMAJ. 2011;183(8):915-920.
14. Nicholson TR, Stone J, Kanaan RA. Conversion disorder: a problematic diagnosis. J Neurol Neurosurg Psychiatry. 2011;82(11):1267-1273.
15. Bitzer J. Somatization disorders in obstetrics and gynecology. Arch Womens Mental health, 2003;6(2):99-107.
16. Smith HE, Rynning RE, Okafor C, et al. Evaluation of neurologic deficit without apparent cause: the importance of a multidisciplinary approach. J Spinal Cord Med. 2007;30(5):509-517.
17. Hinson VK, Haren WB. Psychogenic movement disorders. Lancet Neurol. 2006;5(8):695-700.
18. Oyama O, Paltoo C, Greengold J. Somatoform disorders. Am Fam Physician. 2007;76(9):1333-1338.
19. Ness D. Physical therapy management for conversion disorder: case series. J Neurol Phys Ther. 2007;31(1):30-39.

Article PDF
Author and Disclosure Information

Nancy Byatt, DO, MBA
Assistant Professor of Psychiatry and Obstetrics and Gynecology
University of Massachusetts Medical School
Psychiatrist
Psychosomatic Medicine
Women’s Mental Health Specialty Clinic
UMass Memorial Medical Center
Worcester, Massachusetts


Ramanpreet Toor, MD
Acting Assistant Professor
Department of Psychiatry and Behavioral Sciences
University of Washington
Seattle, Washington

Issue
Current Psychiatry - 14(1)
Publications
Topics
Page Number
44-49
Legacy Keywords
pregnant, truncal ataxia, astasia-abasia, somatic disorders, conversion disorder, neurologic symptoms
Sections
Author and Disclosure Information

Nancy Byatt, DO, MBA
Assistant Professor of Psychiatry and Obstetrics and Gynecology
University of Massachusetts Medical School
Psychiatrist
Psychosomatic Medicine
Women’s Mental Health Specialty Clinic
UMass Memorial Medical Center
Worcester, Massachusetts


Ramanpreet Toor, MD
Acting Assistant Professor
Department of Psychiatry and Behavioral Sciences
University of Washington
Seattle, Washington

Author and Disclosure Information

Nancy Byatt, DO, MBA
Assistant Professor of Psychiatry and Obstetrics and Gynecology
University of Massachusetts Medical School
Psychiatrist
Psychosomatic Medicine
Women’s Mental Health Specialty Clinic
UMass Memorial Medical Center
Worcester, Massachusetts


Ramanpreet Toor, MD
Acting Assistant Professor
Department of Psychiatry and Behavioral Sciences
University of Washington
Seattle, Washington

Article PDF
Article PDF

CASE Difficulty walking
Ms. M, age 15, is a pregnant, Spanish-speaking Guatemalan woman who is brought to obstet­rics triage in a large academic medical center at 35 weeks gestational age. She complains of dizziness, tinnitus, left orbital headache, and difficulty walking.

The neurology service finds profound trun­cal ataxia, astasia-abasia, and buckling of the knees; a normal brain and spine MRI are not consistent with a neurologic etiology. Otolaryngology service evaluates Ms. M to rule out a cholesteatoma and suggests a head CT and endoscopy, which are normal.

Ms. M’s symptoms resolve after 3 days, although the gait disturbances persist. When no clear cause is found for her difficulty walk­ing, the psychiatry service is consulted to evalu­ate whether an underlying psychiatric disorder is contributing to symptoms.


What could be causing Ms. M’s symptoms?

   a) malingering
   b) factitious disorder
   c) undiagnosed neurologic disorder
   d) conversion disorder


The authors’ observations

Women are vulnerable to a variety of psy­chiatric illnesses during pregnancy1 that have deleterious effects on mother, baby, and family.2-6 Although there is a burgeoning literature on affective and anxiety disorders occurring in pregnancy, there is a dearth of information about somatoform disorders.


HISTORY
Abandonment

Ms. M reports that, although her boyfriend deserted her after learning about the unexpected pregnancy, she will welcome the baby and looks forward to motherhood. She seems unaware of the responsibilities of being a mother.

Ms. M acknowledges a history of depression and self-harm a few years earlier, yet says she feels better now and thinks that psychiatric care is unnecessary. Because she does not endorse a history of trauma or symptoms suggesting an affective, anxiety, or psychotic illness, the psy­chiatrist does not recommend treatment with psychotropic medication. 

At age 5, Ms. M’s parents sent her to the United States with her aunt, hoping that she would have a better life than she would have had in Guatemala. Her aunt reports that Ms. M initially had difficulty adjusting to life in the United States without her parents, yet she has made substantial strides over the years and is now quite accustomed to the country. Her aunt describes Ms. M as an independent high school student who earns good grades.

During the interview, the psychiatrist observes that Ms. M exhibits childlike manner­isms, including sleeping with stuffed toys and coloring in Disney books with crayons. She also is indifferent to her gait difficulty, pregnancy, and psychosocial stressors. Her affect is incon­sistent with the content of her speech and she is alexithymic.

Ms. M’s aunt reports that her niece is becom­ing more dependent on her, which is not con­sistent with her baseline. Her aunt also notes that several years earlier, Ms. M’s nephew was diagnosed with a cholesteatoma after he pre­sented with similar symptoms.

The combination of (1) Ms. M’s clinical pre­sentation, which was causing her significant impairment in her social functioning, (2) the incompatibility of symptoms with any recog­nized neurologic and medical disease, and (3) prior family experience with cholesteatoma leads the consulting psychiatrist to suspect conversion disorder. Ms. M’s alexithymia, indif­ference to her symptoms, and recent aban­donment by the baby’s father also support a conversion disorder diagnosis.

From a psychodynamic perspective, the ataxia appears to be her way of protecting her­self from the abandonment she is experiencing by being left again to “stand alone” by her boy­friend as she had been when her parents sent her to the United States. Her regressive behav­ior could be her way of securing her aunt’s love and support.


The authors’ observations
This is the first case of psychogenic gait dis­turbance during pregnancy described in the literature. Authors have reported on pseu­dotoxemia,7 hyperemesis gravidarum,8 and pesudocyesis,9 yet there is a paucity of infor­mation on psychogenic gait disturbance during pregnancy. Ms. M’s case elucidates many of the clinical quandaries that occur when managing psychiatric illness—and, more specifically, conversion disorder— during pregnancy. Many women are hesi­tant to seek psychiatric treatment during pregnancy because of shame, stigma, and fear of loss of personal or parental rights10,11; it is not surprising that emotionally dis­tressed women communicate their feel­ings or troubled thoughts through physical symptoms.


Likely diagnosis

Conversion disorder is the presence of neurologic symptoms in the absence of a neurologic diagnosis that fully explains those symptoms. Conversion disorder, previously known as hysteria, is called functional neurologic symptom disorder in DSM-5 (Box).12 Symptoms are not feigned; instead, they represent “conver­sion” of emotional distress into neurologic symptoms.13,14 Although misdiagnosing conversion disorder in patients with true neurologic disease is uncommon, clini­cians often are uncomfortable making the diagnosis until all medical causes have been ruled out.14 It is not always possible to find a psychological explanation for conversion disorder, but a history of child­hood abuse, particularly sexual abuse, could play a role.14




Because of the variety of presenta­tions, clinicians in all specialties should be familiar with somatoform disorders; this is especially important in obstetrics and gynecology because women are more likely than men to develop these disorders.15 It is important to consider that Ms. M is a teen­ager and somatoform disorders can present differently in adults. The diagnostic process should include a diligent somatic workup and a personal and social history to identify the patient’s developmental tasks, stressors, and coping style.15

 

 


How would you treat Ms. M?

   a) destigmatize psychiatric illness and provide psychoeducation regarding treat­ment benefits
   b) identify and treat any comorbid psychiat­ric disorders
   c) maintain a proactive and multidisci­plinary approach that includes assess­ment of psychosocial stressors and psychodynamic factors, particularly those related to the pregnancy
  d) all of the above


TREATMENT
Close follow-up

The psychiatrist recommends continued close psychiatric follow-up as well as multidisci­plinary involvement, including physical ther­apy, neurology, and obstetrics.

Ms. M initially is resistant to psychiatric follow-up because she says that “people on the street” told her that, if she saw a psychia­trist, her baby would be taken away. After the psychiatrist explains that it is unlikely her baby would be taken away, Ms. M immediately appears relieved, smiles, and readily agrees to outpatient psychotherapy.

Over the next 24 hours, she continues to work with a physical therapist and her gait sig­nificantly improves. She is discharged home 2 days later with a walking aid (Zimmer frame) for assistance.

Four days later, however, Ms. M is readmit­ted with worsening ataxia. Her aunt reports that, at home, Ms. M’s regressed behaviors are worsening; she is sleeping in bed with her and had several episodes of enuresis at home.

Ms. M continues to deny psychiatric symp­toms or anxiety about the delivery. Although she shows some improvement when work­ing with physical therapists, they note that Ms. M is still unable to ambulate or stand on her own. The psychiatrist is increasingly con­cerned about her regressed behavior and con­tinued ataxia.

A family meeting is held and the psychia­trist and social worker educate Ms. M and her aunt about conversion disorder, including how some emotionally distressed women communicate their feelings or troubled thoughts through physical symptoms and how that may apply to Ms. M. During the meeting, the team also destigmatizes psy­chiatric illness and treatment and provides psychoeducation regarding its benefits. The psychiatrist and social worker also provide a psychodynamic interpretation that her ataxia could be a way of protecting herself against the abandonment she is experiencing by being left to “stand alone” by her boyfriend— as she had been when her parents sent her to the United States, and that her behavior could be her way of securing her aunt’s love and support.Ms. M and her aunt both readily agree with this interpretation. The aunt notes that her niece is more anxious about motherhood than she acknowledges and is concerned that Ms. M expects her to be the primary care­giver for the baby. Those present note that Ms. M is becoming increasingly dependent on her aunt, and that it is important for her to retain her independence, especially once she becomes a mother.

Ms. M immediately begins to display more affect; she smiles and reports feeling relieved. Similar to the previous admission, her gait sig­nificantly improves over the next 2 days and she is discharged home with a walking aid.


The authors’ observations

A broad differential diagnosis and early multidisciplinary involvement might facilitate earlier diagnosis and treatment.16 Assessment of psychosocial stressors in the patient’s personal and family life, includ­ing circumstances around the pregnancy and the meaning of motherhood, as well as investigation of what the patient may gain from the sick role, are paramount. In Ms. M’s case, cultural background, sepa­ration from her parents at a young age, and recent abandonment by her boyfriend have contributed to her inability to “stand alone,” which manifested as ataxia. Young age, regressed behavior, and her minimi­zation of stressors also point to her dif­ficulty acknowledging and coping with psychosocial stressors.

Successful delivery of the diagnosis is key to treatment success. After building a therapeutic alliance, a multidisciplinary discussion should take place that allows the patient to understand the diagnosis and treatment plan.17,18 The patient and family should be reassured that the fetus is healthy and all organic causes of symptoms have been investigated.17 Although man­agement of conversion disorder during pregnancy is similar to that in non-preg­nant women, several additional avenues of investigation should be considered:
   • Explore the psychodynamic basis of the disorder and the role of the pregnancy and motherhood.
   • Identify any comorbid psychiatric disorders, particularly those specific to pregnancy or the postpartum period.
   • Because of the shame and stigma associated with seeking psychiatric treat­ment during pregnancy,10,11 it is impera­tive to destigmatize treatment and provide psychoeducation regarding its benefits.

A treatment plan can then be developed that involves psychotherapy, psychoedu­cation, stress management, and, when appropriate, pharmacotherapy.17

Providing psychoeducation about postpartum depression and other perina­tal psychiatric illness could be beneficial. Physical therapy often is culturally accept­able and can help re-establish healthy patterns of motor function.19 Ms. M’s gait  showed some improvement with physical therapy as part of the multidisciplinary approach, which also should include a thorough medical workup. Appropriate psychiatric treatment can help patients give up the sick role and return to their previous level of functioning.17

 

 

Maintain close communication with the outpatient perinatal care team as they mon­itor the patient’s parenting capacity. The outpatient perinatal care team also should engage pregnant or postpartum women in prioritizing their emotional well-being and encourage outpatient mental health treatment. Despite a dearth of data on the regressive symptoms and prognosis for future pregnancies, it is important to mon­itor maternal capacity and discuss the pos­sibility of symptom recurrence.


OUTCOME
Healthy baby

Three days later, Ms. M returns in labor with improved gait yet still using a walking aid. She has a normal vaginal delivery of a healthy baby boy at 37 weeks’ gestational age.

After the birth, Ms. M reports feeling well and enjoying motherhood, and denies psy­chiatric symptoms. She is ambulating without assistance within hours of delivery. This spon­taneous resolution of symptoms could have been because of the psychodynamically ori­ented multidisciplinary approach to her care, which may have helped her realize that she did not have to “stand alone” as she embarked on motherhood.

Before being discharged home, Ms. M and her aunt meet with the inpatient obstetric social worker to assess Ms. M’s ability to care for the baby and discuss the importance of continued emotional support. The social worker does not contact the Department of Children and Families because Ms. M is walking independently and not endorsing or exhibiting regressive behaviors. Ms. M also reports that she will ask her aunt to take care of the baby should ataxia recur. Her aunt reassures the social workers that she will encourage Ms. M to attend outpatient psy­chotherapy and will contact the social worker if she becomes concerned about Ms. M’s or the baby’s well-being.

During her postpartum obstetric visit, Ms. M is walking independently and does not exhibit or endorse neurologic symptoms. The social worker provides psychoeducation about the importance of outpatient psycho­therapy and schedules an initial appointment; Ms. M does not attend outpatient psychother­apy after discharge.

Bottom Line
Consider conversion disorder in obstetric patients who present with ataxia without a neurologic cause. Management involves a proactive and multidisciplinary approach that includes a thorough medical workup and assessment of psychosocial stressors and psychodynamic factors, particularly those related to the pregnancy. Early identification and delivery of the diagnosis, destigmatization, and provision of appropriate psychiatric treatment can facilitate treatment success.

Disclosures
Dr. Byatt has received grant funding/support for this project from the National Center for Research Resources and the National Center for Advancing Translational Sciences, National Institutes of Health, through Grant KL2TR000160. The content is solely the responsibility of the authors and does not necessarily represent the official views of the NIH. Dr. Toor reports no financial relationships with any company whose products are mentioned in this article or manufacturers of competing products.

CASE Difficulty walking
Ms. M, age 15, is a pregnant, Spanish-speaking Guatemalan woman who is brought to obstet­rics triage in a large academic medical center at 35 weeks gestational age. She complains of dizziness, tinnitus, left orbital headache, and difficulty walking.

The neurology service finds profound trun­cal ataxia, astasia-abasia, and buckling of the knees; a normal brain and spine MRI are not consistent with a neurologic etiology. Otolaryngology service evaluates Ms. M to rule out a cholesteatoma and suggests a head CT and endoscopy, which are normal.

Ms. M’s symptoms resolve after 3 days, although the gait disturbances persist. When no clear cause is found for her difficulty walk­ing, the psychiatry service is consulted to evalu­ate whether an underlying psychiatric disorder is contributing to symptoms.


What could be causing Ms. M’s symptoms?

   a) malingering
   b) factitious disorder
   c) undiagnosed neurologic disorder
   d) conversion disorder


The authors’ observations

Women are vulnerable to a variety of psy­chiatric illnesses during pregnancy1 that have deleterious effects on mother, baby, and family.2-6 Although there is a burgeoning literature on affective and anxiety disorders occurring in pregnancy, there is a dearth of information about somatoform disorders.


HISTORY
Abandonment

Ms. M reports that, although her boyfriend deserted her after learning about the unexpected pregnancy, she will welcome the baby and looks forward to motherhood. She seems unaware of the responsibilities of being a mother.

Ms. M acknowledges a history of depression and self-harm a few years earlier, yet says she feels better now and thinks that psychiatric care is unnecessary. Because she does not endorse a history of trauma or symptoms suggesting an affective, anxiety, or psychotic illness, the psy­chiatrist does not recommend treatment with psychotropic medication. 

At age 5, Ms. M’s parents sent her to the United States with her aunt, hoping that she would have a better life than she would have had in Guatemala. Her aunt reports that Ms. M initially had difficulty adjusting to life in the United States without her parents, yet she has made substantial strides over the years and is now quite accustomed to the country. Her aunt describes Ms. M as an independent high school student who earns good grades.

During the interview, the psychiatrist observes that Ms. M exhibits childlike manner­isms, including sleeping with stuffed toys and coloring in Disney books with crayons. She also is indifferent to her gait difficulty, pregnancy, and psychosocial stressors. Her affect is incon­sistent with the content of her speech and she is alexithymic.

Ms. M’s aunt reports that her niece is becom­ing more dependent on her, which is not con­sistent with her baseline. Her aunt also notes that several years earlier, Ms. M’s nephew was diagnosed with a cholesteatoma after he pre­sented with similar symptoms.

The combination of (1) Ms. M’s clinical pre­sentation, which was causing her significant impairment in her social functioning, (2) the incompatibility of symptoms with any recog­nized neurologic and medical disease, and (3) prior family experience with cholesteatoma leads the consulting psychiatrist to suspect conversion disorder. Ms. M’s alexithymia, indif­ference to her symptoms, and recent aban­donment by the baby’s father also support a conversion disorder diagnosis.

From a psychodynamic perspective, the ataxia appears to be her way of protecting her­self from the abandonment she is experiencing by being left again to “stand alone” by her boy­friend as she had been when her parents sent her to the United States. Her regressive behav­ior could be her way of securing her aunt’s love and support.


The authors’ observations
This is the first case of psychogenic gait dis­turbance during pregnancy described in the literature. Authors have reported on pseu­dotoxemia,7 hyperemesis gravidarum,8 and pesudocyesis,9 yet there is a paucity of infor­mation on psychogenic gait disturbance during pregnancy. Ms. M’s case elucidates many of the clinical quandaries that occur when managing psychiatric illness—and, more specifically, conversion disorder— during pregnancy. Many women are hesi­tant to seek psychiatric treatment during pregnancy because of shame, stigma, and fear of loss of personal or parental rights10,11; it is not surprising that emotionally dis­tressed women communicate their feel­ings or troubled thoughts through physical symptoms.


Likely diagnosis

Conversion disorder is the presence of neurologic symptoms in the absence of a neurologic diagnosis that fully explains those symptoms. Conversion disorder, previously known as hysteria, is called functional neurologic symptom disorder in DSM-5 (Box).12 Symptoms are not feigned; instead, they represent “conver­sion” of emotional distress into neurologic symptoms.13,14 Although misdiagnosing conversion disorder in patients with true neurologic disease is uncommon, clini­cians often are uncomfortable making the diagnosis until all medical causes have been ruled out.14 It is not always possible to find a psychological explanation for conversion disorder, but a history of child­hood abuse, particularly sexual abuse, could play a role.14




Because of the variety of presenta­tions, clinicians in all specialties should be familiar with somatoform disorders; this is especially important in obstetrics and gynecology because women are more likely than men to develop these disorders.15 It is important to consider that Ms. M is a teen­ager and somatoform disorders can present differently in adults. The diagnostic process should include a diligent somatic workup and a personal and social history to identify the patient’s developmental tasks, stressors, and coping style.15

 

 


How would you treat Ms. M?

   a) destigmatize psychiatric illness and provide psychoeducation regarding treat­ment benefits
   b) identify and treat any comorbid psychiat­ric disorders
   c) maintain a proactive and multidisci­plinary approach that includes assess­ment of psychosocial stressors and psychodynamic factors, particularly those related to the pregnancy
  d) all of the above


TREATMENT
Close follow-up

The psychiatrist recommends continued close psychiatric follow-up as well as multidisci­plinary involvement, including physical ther­apy, neurology, and obstetrics.

Ms. M initially is resistant to psychiatric follow-up because she says that “people on the street” told her that, if she saw a psychia­trist, her baby would be taken away. After the psychiatrist explains that it is unlikely her baby would be taken away, Ms. M immediately appears relieved, smiles, and readily agrees to outpatient psychotherapy.

Over the next 24 hours, she continues to work with a physical therapist and her gait sig­nificantly improves. She is discharged home 2 days later with a walking aid (Zimmer frame) for assistance.

Four days later, however, Ms. M is readmit­ted with worsening ataxia. Her aunt reports that, at home, Ms. M’s regressed behaviors are worsening; she is sleeping in bed with her and had several episodes of enuresis at home.

Ms. M continues to deny psychiatric symp­toms or anxiety about the delivery. Although she shows some improvement when work­ing with physical therapists, they note that Ms. M is still unable to ambulate or stand on her own. The psychiatrist is increasingly con­cerned about her regressed behavior and con­tinued ataxia.

A family meeting is held and the psychia­trist and social worker educate Ms. M and her aunt about conversion disorder, including how some emotionally distressed women communicate their feelings or troubled thoughts through physical symptoms and how that may apply to Ms. M. During the meeting, the team also destigmatizes psy­chiatric illness and treatment and provides psychoeducation regarding its benefits. The psychiatrist and social worker also provide a psychodynamic interpretation that her ataxia could be a way of protecting herself against the abandonment she is experiencing by being left to “stand alone” by her boyfriend— as she had been when her parents sent her to the United States, and that her behavior could be her way of securing her aunt’s love and support.Ms. M and her aunt both readily agree with this interpretation. The aunt notes that her niece is more anxious about motherhood than she acknowledges and is concerned that Ms. M expects her to be the primary care­giver for the baby. Those present note that Ms. M is becoming increasingly dependent on her aunt, and that it is important for her to retain her independence, especially once she becomes a mother.

Ms. M immediately begins to display more affect; she smiles and reports feeling relieved. Similar to the previous admission, her gait sig­nificantly improves over the next 2 days and she is discharged home with a walking aid.


The authors’ observations

A broad differential diagnosis and early multidisciplinary involvement might facilitate earlier diagnosis and treatment.16 Assessment of psychosocial stressors in the patient’s personal and family life, includ­ing circumstances around the pregnancy and the meaning of motherhood, as well as investigation of what the patient may gain from the sick role, are paramount. In Ms. M’s case, cultural background, sepa­ration from her parents at a young age, and recent abandonment by her boyfriend have contributed to her inability to “stand alone,” which manifested as ataxia. Young age, regressed behavior, and her minimi­zation of stressors also point to her dif­ficulty acknowledging and coping with psychosocial stressors.

Successful delivery of the diagnosis is key to treatment success. After building a therapeutic alliance, a multidisciplinary discussion should take place that allows the patient to understand the diagnosis and treatment plan.17,18 The patient and family should be reassured that the fetus is healthy and all organic causes of symptoms have been investigated.17 Although man­agement of conversion disorder during pregnancy is similar to that in non-preg­nant women, several additional avenues of investigation should be considered:
   • Explore the psychodynamic basis of the disorder and the role of the pregnancy and motherhood.
   • Identify any comorbid psychiatric disorders, particularly those specific to pregnancy or the postpartum period.
   • Because of the shame and stigma associated with seeking psychiatric treat­ment during pregnancy,10,11 it is impera­tive to destigmatize treatment and provide psychoeducation regarding its benefits.

A treatment plan can then be developed that involves psychotherapy, psychoedu­cation, stress management, and, when appropriate, pharmacotherapy.17

Providing psychoeducation about postpartum depression and other perina­tal psychiatric illness could be beneficial. Physical therapy often is culturally accept­able and can help re-establish healthy patterns of motor function.19 Ms. M’s gait  showed some improvement with physical therapy as part of the multidisciplinary approach, which also should include a thorough medical workup. Appropriate psychiatric treatment can help patients give up the sick role and return to their previous level of functioning.17

 

 

Maintain close communication with the outpatient perinatal care team as they mon­itor the patient’s parenting capacity. The outpatient perinatal care team also should engage pregnant or postpartum women in prioritizing their emotional well-being and encourage outpatient mental health treatment. Despite a dearth of data on the regressive symptoms and prognosis for future pregnancies, it is important to mon­itor maternal capacity and discuss the pos­sibility of symptom recurrence.


OUTCOME
Healthy baby

Three days later, Ms. M returns in labor with improved gait yet still using a walking aid. She has a normal vaginal delivery of a healthy baby boy at 37 weeks’ gestational age.

After the birth, Ms. M reports feeling well and enjoying motherhood, and denies psy­chiatric symptoms. She is ambulating without assistance within hours of delivery. This spon­taneous resolution of symptoms could have been because of the psychodynamically ori­ented multidisciplinary approach to her care, which may have helped her realize that she did not have to “stand alone” as she embarked on motherhood.

Before being discharged home, Ms. M and her aunt meet with the inpatient obstetric social worker to assess Ms. M’s ability to care for the baby and discuss the importance of continued emotional support. The social worker does not contact the Department of Children and Families because Ms. M is walking independently and not endorsing or exhibiting regressive behaviors. Ms. M also reports that she will ask her aunt to take care of the baby should ataxia recur. Her aunt reassures the social workers that she will encourage Ms. M to attend outpatient psy­chotherapy and will contact the social worker if she becomes concerned about Ms. M’s or the baby’s well-being.

During her postpartum obstetric visit, Ms. M is walking independently and does not exhibit or endorse neurologic symptoms. The social worker provides psychoeducation about the importance of outpatient psycho­therapy and schedules an initial appointment; Ms. M does not attend outpatient psychother­apy after discharge.

Bottom Line
Consider conversion disorder in obstetric patients who present with ataxia without a neurologic cause. Management involves a proactive and multidisciplinary approach that includes a thorough medical workup and assessment of psychosocial stressors and psychodynamic factors, particularly those related to the pregnancy. Early identification and delivery of the diagnosis, destigmatization, and provision of appropriate psychiatric treatment can facilitate treatment success.

Disclosures
Dr. Byatt has received grant funding/support for this project from the National Center for Research Resources and the National Center for Advancing Translational Sciences, National Institutes of Health, through Grant KL2TR000160. The content is solely the responsibility of the authors and does not necessarily represent the official views of the NIH. Dr. Toor reports no financial relationships with any company whose products are mentioned in this article or manufacturers of competing products.

References


1. Vesga-Lopez O, Blanco C, Keyes K, et al. Psychiatric disorders in pregnant and postpartum women in the United States. Arch Gen Psychiatry. 2008;65(7):805-815.
2. Britton HL, Gronwaldt V, Britton JR. Maternal postpartum behaviors and mother-infant relationship during the first year of life. J Pediatr. 2001;138(6):905-909.
3. Deave T, Heron J, Evans J, et al. The impact of maternal depression in pregnancy on early child development. BJOG. 2008;115(8):1043-1051.
4. Paulson JF, Keefe HA, Leiferman JA. Early parental depression and child language development. J Child Psychol Psychiatry. 2009;50(3):254-262.
5. Zuckerman B, Amaro H, Bauchner H, et al. Depressive symptoms during pregnancy: relationship to poor health behaviors. Am J Obstet Gynecol. 1989;160(5 pt 1):1107-1111.
6. Forman DR, O’Hara MW, Stuart S, et al. Effective treatment for postpartum depression is not sufficient to improve the developing mother-child relationship. Dev Psychopathol. 2007;19(2):585-602.
7. Brady WJ Jr, Huff JS. Pseudotoxemia: new onset psychogenic seizure in third trimester pregnancy. J Emerg Med. 1997;15(6):815-820.
8. el-Mallakh RS, Liebowitz NR, Hale MS. Hyperemesis gravidarum as conversion disorder. J Nerv Ment Dis. 1990; 178(10):655-659.
9. Paulman PM, Sadat A. Pseudocyesis. J Fam Pract. 1990;30(5):575-576.
10. Dennis CL, Chung-Lee L. Postpartum depression help-seeking barriers and maternal treatment p: a qualitative systematic review. Birth. 2006;33(4):323-331.
11. Byatt N, Simas TA, Lundquist RS, et al. Strategies for improving perinatal depression treatment in North American outpatient obstetric settings. J Psychosom Obstetr Gynaecol. 2012;33(4):143-161.
12. Diagnostic and statistical manual of mental disorders, 5th ed. Washington, DC: American Psychiatric Association; 2013.
13. Feinstein A. Conversion disorder: advances in our understanding. CMAJ. 2011;183(8):915-920.
14. Nicholson TR, Stone J, Kanaan RA. Conversion disorder: a problematic diagnosis. J Neurol Neurosurg Psychiatry. 2011;82(11):1267-1273.
15. Bitzer J. Somatization disorders in obstetrics and gynecology. Arch Womens Mental health, 2003;6(2):99-107.
16. Smith HE, Rynning RE, Okafor C, et al. Evaluation of neurologic deficit without apparent cause: the importance of a multidisciplinary approach. J Spinal Cord Med. 2007;30(5):509-517.
17. Hinson VK, Haren WB. Psychogenic movement disorders. Lancet Neurol. 2006;5(8):695-700.
18. Oyama O, Paltoo C, Greengold J. Somatoform disorders. Am Fam Physician. 2007;76(9):1333-1338.
19. Ness D. Physical therapy management for conversion disorder: case series. J Neurol Phys Ther. 2007;31(1):30-39.

References


1. Vesga-Lopez O, Blanco C, Keyes K, et al. Psychiatric disorders in pregnant and postpartum women in the United States. Arch Gen Psychiatry. 2008;65(7):805-815.
2. Britton HL, Gronwaldt V, Britton JR. Maternal postpartum behaviors and mother-infant relationship during the first year of life. J Pediatr. 2001;138(6):905-909.
3. Deave T, Heron J, Evans J, et al. The impact of maternal depression in pregnancy on early child development. BJOG. 2008;115(8):1043-1051.
4. Paulson JF, Keefe HA, Leiferman JA. Early parental depression and child language development. J Child Psychol Psychiatry. 2009;50(3):254-262.
5. Zuckerman B, Amaro H, Bauchner H, et al. Depressive symptoms during pregnancy: relationship to poor health behaviors. Am J Obstet Gynecol. 1989;160(5 pt 1):1107-1111.
6. Forman DR, O’Hara MW, Stuart S, et al. Effective treatment for postpartum depression is not sufficient to improve the developing mother-child relationship. Dev Psychopathol. 2007;19(2):585-602.
7. Brady WJ Jr, Huff JS. Pseudotoxemia: new onset psychogenic seizure in third trimester pregnancy. J Emerg Med. 1997;15(6):815-820.
8. el-Mallakh RS, Liebowitz NR, Hale MS. Hyperemesis gravidarum as conversion disorder. J Nerv Ment Dis. 1990; 178(10):655-659.
9. Paulman PM, Sadat A. Pseudocyesis. J Fam Pract. 1990;30(5):575-576.
10. Dennis CL, Chung-Lee L. Postpartum depression help-seeking barriers and maternal treatment p: a qualitative systematic review. Birth. 2006;33(4):323-331.
11. Byatt N, Simas TA, Lundquist RS, et al. Strategies for improving perinatal depression treatment in North American outpatient obstetric settings. J Psychosom Obstetr Gynaecol. 2012;33(4):143-161.
12. Diagnostic and statistical manual of mental disorders, 5th ed. Washington, DC: American Psychiatric Association; 2013.
13. Feinstein A. Conversion disorder: advances in our understanding. CMAJ. 2011;183(8):915-920.
14. Nicholson TR, Stone J, Kanaan RA. Conversion disorder: a problematic diagnosis. J Neurol Neurosurg Psychiatry. 2011;82(11):1267-1273.
15. Bitzer J. Somatization disorders in obstetrics and gynecology. Arch Womens Mental health, 2003;6(2):99-107.
16. Smith HE, Rynning RE, Okafor C, et al. Evaluation of neurologic deficit without apparent cause: the importance of a multidisciplinary approach. J Spinal Cord Med. 2007;30(5):509-517.
17. Hinson VK, Haren WB. Psychogenic movement disorders. Lancet Neurol. 2006;5(8):695-700.
18. Oyama O, Paltoo C, Greengold J. Somatoform disorders. Am Fam Physician. 2007;76(9):1333-1338.
19. Ness D. Physical therapy management for conversion disorder: case series. J Neurol Phys Ther. 2007;31(1):30-39.

Issue
Current Psychiatry - 14(1)
Issue
Current Psychiatry - 14(1)
Page Number
44-49
Page Number
44-49
Publications
Publications
Topics
Article Type
Display Headline
Young, pregnant, ataxic—and jilted
Display Headline
Young, pregnant, ataxic—and jilted
Legacy Keywords
pregnant, truncal ataxia, astasia-abasia, somatic disorders, conversion disorder, neurologic symptoms
Legacy Keywords
pregnant, truncal ataxia, astasia-abasia, somatic disorders, conversion disorder, neurologic symptoms
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media

Woman, 32, With Crusty Red Blisters

Article Type
Changed
Tue, 06/04/2019 - 15:22
Display Headline
Woman, 32, With Crusty Red Blisters

A 32-year-old Korean woman presented with a rash on her scalp, face, palms, soles, and genital region and with sores in the oral cavity. The blisters were red and flat with some crusting, particularly on the scalp and face. The patient described the blisters as very painful, adding that it hurt to walk, grasp objects, and drink fluids. Associated symptoms included painful urination, sore throat, malaise, and fever of up to 103°F. She was taking acetaminophen and ibuprofen to alleviate the fever and pain.

 

Medical history was unremarkable. Social history was negative for recent changes in sexual partner or travel to foreign countries.

Physical examination revealed numerous flat, erythematous lesions. Lesions on the face and scalp had developed a weeping, honey-colored crust (see Figure 1 and Figure 2). The lesions were tender to the touch, particularly on the palms and soles.

Further questioning revealed that the patient’s 18-month-old son had exhibited similar symptoms two to three days prior to her illness.

Continue for differential diagnosis >>

 

 

DIFFERENTIAL DIAGNOSIS
Because multiple bacterial and viral diseases manifest in this fashion, the differential diagnosis included the following ­disorders:

Erythema multiforme. This skin condition may result from an allergic or hypersensitivity reaction to certain drugs or from infections. Infections that can cause erythema multiforme include herpes simplex virus and mycoplasma. Patients present with lesions on the palms (see Figure 3), soles, extremities, face, or trunk. The lesions can appear as a nodule, papule, macule, or vesicle. Initially, in the mild form, the lesions may appear as hives or target-shaped rashes, occurring on the face and acral surfaces. A severe form of erythema multiforme, known as Stevens-Johnson syndrome, is characterized by rash, mucosal involvement, and systemic symptoms.1

Herpes zoster. This viral infection is caused by the varicella-zoster virus, which also causes chicken pox. The virus lies dormant within a single sensory ganglion and may reappear as shingles along the dermatome of that nerve. Patients may experience burning or shooting pain with tingling or itching before the rash appears; vesicular lesions with erythematous bases appear days later. The rash occurs unilaterally on the body or face and does not cross the midline. A viral culture may be obtained for identification.2

Herpetic gingivostomatitis. This infection is most commonly caused by herpes simplex virus type 1, the same virus that causes cold sores. Patients may present with ulcerations along the buccal mucosa and gums. The infection manifests with systemic symptoms, including malaise, fever, irritability, and cervical adenopathy. A viral culture will identify the etiology.3

Impetigo. This skin infection is typically caused by bacteria, predominantly Staphylococcus aureus, Streptococcus pyogenes, or a combination. Infections generally occur after a break in the skin surface. The most common presentation is a rash that spreads to different parts of the body after scratching. Skin lesions can occur on the face, lips, or extremities. Initially vesicular, the lesions generally form a honey-colored crust after fluid discharge. The clinician should take a skin or fluid sample from the lesion to culture, which may identify the pathogen.4

Syphilis. This sexually transmitted infection is caused by the spirochete Treponema pallidum. In primary syphilis, patients can develop a painless sore, or chancre, on the genitals, rectal area, or mouth. If left untreated, the disease can progress to secondary syphilis, manifesting as a pale pink or reddish maculopapular rash on the palms and soles. The rash can be associated with fever, sore throat, myalgia, and fatigue. It is important to rule out syphilis because, left untreated, it can lead to cardiac and neurologic complications. Screening tests include VDRL and the rapid plasma reagin (RPR) test, both of which assess for antibodies to the organism, and dark field microscopy of ulcerations to identify the organism.5

Also included in the differential diagnosis for the patient’s symptoms was hand-foot-mouth disease (HFMD), discussed below.

Next page: Discussion >>

 

 

DISCUSSION
HFMD is an acute viral illness most often affecting children younger than 5 and occurring in summer to early fall months. HFMD manifests with fever and papulovesicular eruptions; lesions often appear in the oral cavity first, spreading to the palms, soles, and buttocks. Route of transmission is usually fecal-oral or through respiratory droplets, oral secretions, or direct contact with fluid-filled vesicles.6-8 The highly contagious nature of the virus causes it to spread to close contacts and family members and leads to outbreaks in schools and daycare centers.9,10

In the United States, the most common etiology of HFMD is coxsackievirus A16.7 Another causative agent, enterovirus 71 (EV71), has been found responsible for HFMD epidemics in southeast Asia and Australia.11,12 Recently, the coxsackievirus A6 (CV A6) strain has been linked to outbreaks of HFMD.7,9 This strain may produce an atypical manifestation of skin lesions on the face, trunk, and extremities. The lesions may also appear larger than usual and have a vesiculobullous rather than the more typical papulovesicular appearance. The course of the illness differs in severity depending on the strain of the virus causing HFMD.9

CLINICAL PRESENTATION
The acute phase of HFMD typically begins with prodromal symptoms such as fever, malaise, and sore throat. Erythematous ulcerations usually appear in the oral cavity first (see Figure 4) and often cause symptoms such as sore throat, dysphagia, or dryness. As the disease progresses, cutaneous lesions spread to the face, extremities, interdigital areas (see Figure 5), trunk, and perianal area (which may cause dysuria). The lesions may initially appear as erythematous macules or papules, transforming to vesicles as the disease progresses. The mucocutaneous lesions are usually asymptomatic but can be tender to touch or pressure and may leak fluid.7,10

In only a few cases—caused by CV A6—have lesions in the scalp been reported; the mechanism of action is unknown.10 Instances of lesions invading the nails have been reported, causing desquamation and shedding. This condition is known as onychomadesis.9,11

Continue for diagnosis >>

 

 

DIAGNOSIS
Serologic testing and viral cultures can identify the exact strain of virus causing HFMD and are particularly useful in unusual presentations. Polymerase chain reaction (PCR) testing yields a high sensitivity and specificity for the causative agent. Histologic examination of skin biopsies may show lymphocytic infiltrates and areas of degeneration along the epidermis. However, most cases are diagnosed based on clinical presentation alone.6,12

TREATMENT AND MANAGEMENT
Management of HFMD is primarily symptomatic, consisting of supportive care that includes use of antipyretics, NSAIDs, and adequate fluid intake to prevent dehydration. The disease is usually self-limited, resolving within seven to 10 days without sequelae.6,9 Aseptic meningitis and other severe complications (especially pulmonary and neurologic), most often associated with EV71 infection, can occur in vulnerable populations, including elderly, pregnant, and immunocompromised patients.9,11 Because the virus is excreted directly from palmar lesions onto the hands, proper hygiene and handwashing techniques offer an exceptionally strong protective effect, preventing transmission and reducing morbidity.8

PATIENT OUTCOME
Based on clinical findings and patient history, the patient was diagnosed with HFMD, which she contracted from her son. Laboratory testing and viral cultures were deemed unnecessary in this case. Treatment was symptomatic, and her skin lesions resolved in one to two weeks.

At follow-up, the patient stated her skin lesions resolved completely without leaving any scars. She also indicated that her nails peeled and shed approximately four weeks after diagnosis, but began to regrow normally four months after diagnosis.

CONCLUSION
Clinicians need to recognize that, although it is uncommon outside the pediatric population, HFMD may occur in adults with intact immune systems. The presentation of HFMD in adults may be atypical, including cutaneous lesions in the scalp and shedding of the nails several weeks after diagnosis. Depending on the viral strain involved, adult patients may have more severe illness and may take longer to recover. Therefore, early diagnosis is important to help prevent the spread of infection and reduce the severity of complications.

REFERENCES
1. Patel NN, Patel DN. Erythema multiforme syndrome. Am J Med. 2009;122(7):623-625.
2. Bader MS. Herpes zoster: diagnostic, therapeutic, and preventive approaches. Postgrad Med. 2013;125(5):78-91.
3. Avci O, Ertam I. Viral infections of the face. Clin Derm. 2014;32:715-733.
4. Hartman-Adams H, Banvard C, Juckett G. Impetigo: diagnosis and treatment. Am Fam Physician. 2014;90(4):229-235.
5. Markle W, Conti T, Kad M. Sexually transmitted diseases. Prim Care Clin Office Pract. 2013;40:557-587.
6. Shin JU, Oh SH, Lee JH. A case of hand-foot-mouth disease in an immunocompetent adult. Ann Dermatol. 2010;22(2):216-218.
7. CDC. Notes from the field: severe hand, foot, and mouth disease associated with coxsackievirus A6 - Alabama, Connecticut, California, and Nevada, November 2011-February 2012. MMWR Morb Mortal Wkly Rep. 2012;61(12):213-214.
8. Ruan F, Yang T, Ma H, et al. Risk factors for hand, foot, and mouth disease and herpangina and the preventive effect of hand-washing. Pediatrics. 2011;127(4):e898-e904.
9. Kaminska K, Martinetti G, Lucchini R, et al. Coxsackievirus A6 and hand, foot and mouth Disease: three case reports of familial child-to-immunocompetent adult transmission and a literature review. Case Rep Dermatol. 2013;5(2):203-209.
10. Lønnberg AS, Elberling J, Fischer TK, Skov L. Two cases of hand, foot, and mouth disease involving the scalp. Acta Derm Venereol. 2013;93(4):467-468.
11. Osterback R, Vuorinen T, Linna M, et al. Coxsackievirus A6 and hand, foot, and mouth disease, Finland. Emerging Infect Dis. 2009;15(9):1485-1488.
12. Shea YF, Chan CY, Hung IFN, Chan KH. Hand, foot and mouth disease in an immunocompetent adult due to Coxsackievirus A6. Hong Kong Med J. 2013;19(3):262-264.

Article PDF
Author and Disclosure Information

 

Purvi Amin, MPA, PA-C, Lisa Daitch, MPAS, PA-C

Purvi Amin is in private practice near Orlando. Lisa Daitch is an Associate Professor at Georgia Regents University in Augusta.

Issue
Clinician Reviews - 25(1)
Publications
Topics
Page Number
37,40-42
Legacy Keywords
facial lesions, red blisters, erythema multiforme, herpes zoster, herpetic gingivostomatis, impetigo, syphilis, hand-foot-mouth disease, HFMD
Sections
Author and Disclosure Information

 

Purvi Amin, MPA, PA-C, Lisa Daitch, MPAS, PA-C

Purvi Amin is in private practice near Orlando. Lisa Daitch is an Associate Professor at Georgia Regents University in Augusta.

Author and Disclosure Information

 

Purvi Amin, MPA, PA-C, Lisa Daitch, MPAS, PA-C

Purvi Amin is in private practice near Orlando. Lisa Daitch is an Associate Professor at Georgia Regents University in Augusta.

Article PDF
Article PDF
Related Articles

A 32-year-old Korean woman presented with a rash on her scalp, face, palms, soles, and genital region and with sores in the oral cavity. The blisters were red and flat with some crusting, particularly on the scalp and face. The patient described the blisters as very painful, adding that it hurt to walk, grasp objects, and drink fluids. Associated symptoms included painful urination, sore throat, malaise, and fever of up to 103°F. She was taking acetaminophen and ibuprofen to alleviate the fever and pain.

 

Medical history was unremarkable. Social history was negative for recent changes in sexual partner or travel to foreign countries.

Physical examination revealed numerous flat, erythematous lesions. Lesions on the face and scalp had developed a weeping, honey-colored crust (see Figure 1 and Figure 2). The lesions were tender to the touch, particularly on the palms and soles.

Further questioning revealed that the patient’s 18-month-old son had exhibited similar symptoms two to three days prior to her illness.

Continue for differential diagnosis >>

 

 

DIFFERENTIAL DIAGNOSIS
Because multiple bacterial and viral diseases manifest in this fashion, the differential diagnosis included the following ­disorders:

Erythema multiforme. This skin condition may result from an allergic or hypersensitivity reaction to certain drugs or from infections. Infections that can cause erythema multiforme include herpes simplex virus and mycoplasma. Patients present with lesions on the palms (see Figure 3), soles, extremities, face, or trunk. The lesions can appear as a nodule, papule, macule, or vesicle. Initially, in the mild form, the lesions may appear as hives or target-shaped rashes, occurring on the face and acral surfaces. A severe form of erythema multiforme, known as Stevens-Johnson syndrome, is characterized by rash, mucosal involvement, and systemic symptoms.1

Herpes zoster. This viral infection is caused by the varicella-zoster virus, which also causes chicken pox. The virus lies dormant within a single sensory ganglion and may reappear as shingles along the dermatome of that nerve. Patients may experience burning or shooting pain with tingling or itching before the rash appears; vesicular lesions with erythematous bases appear days later. The rash occurs unilaterally on the body or face and does not cross the midline. A viral culture may be obtained for identification.2

Herpetic gingivostomatitis. This infection is most commonly caused by herpes simplex virus type 1, the same virus that causes cold sores. Patients may present with ulcerations along the buccal mucosa and gums. The infection manifests with systemic symptoms, including malaise, fever, irritability, and cervical adenopathy. A viral culture will identify the etiology.3

Impetigo. This skin infection is typically caused by bacteria, predominantly Staphylococcus aureus, Streptococcus pyogenes, or a combination. Infections generally occur after a break in the skin surface. The most common presentation is a rash that spreads to different parts of the body after scratching. Skin lesions can occur on the face, lips, or extremities. Initially vesicular, the lesions generally form a honey-colored crust after fluid discharge. The clinician should take a skin or fluid sample from the lesion to culture, which may identify the pathogen.4

Syphilis. This sexually transmitted infection is caused by the spirochete Treponema pallidum. In primary syphilis, patients can develop a painless sore, or chancre, on the genitals, rectal area, or mouth. If left untreated, the disease can progress to secondary syphilis, manifesting as a pale pink or reddish maculopapular rash on the palms and soles. The rash can be associated with fever, sore throat, myalgia, and fatigue. It is important to rule out syphilis because, left untreated, it can lead to cardiac and neurologic complications. Screening tests include VDRL and the rapid plasma reagin (RPR) test, both of which assess for antibodies to the organism, and dark field microscopy of ulcerations to identify the organism.5

Also included in the differential diagnosis for the patient’s symptoms was hand-foot-mouth disease (HFMD), discussed below.

Next page: Discussion >>

 

 

DISCUSSION
HFMD is an acute viral illness most often affecting children younger than 5 and occurring in summer to early fall months. HFMD manifests with fever and papulovesicular eruptions; lesions often appear in the oral cavity first, spreading to the palms, soles, and buttocks. Route of transmission is usually fecal-oral or through respiratory droplets, oral secretions, or direct contact with fluid-filled vesicles.6-8 The highly contagious nature of the virus causes it to spread to close contacts and family members and leads to outbreaks in schools and daycare centers.9,10

In the United States, the most common etiology of HFMD is coxsackievirus A16.7 Another causative agent, enterovirus 71 (EV71), has been found responsible for HFMD epidemics in southeast Asia and Australia.11,12 Recently, the coxsackievirus A6 (CV A6) strain has been linked to outbreaks of HFMD.7,9 This strain may produce an atypical manifestation of skin lesions on the face, trunk, and extremities. The lesions may also appear larger than usual and have a vesiculobullous rather than the more typical papulovesicular appearance. The course of the illness differs in severity depending on the strain of the virus causing HFMD.9

CLINICAL PRESENTATION
The acute phase of HFMD typically begins with prodromal symptoms such as fever, malaise, and sore throat. Erythematous ulcerations usually appear in the oral cavity first (see Figure 4) and often cause symptoms such as sore throat, dysphagia, or dryness. As the disease progresses, cutaneous lesions spread to the face, extremities, interdigital areas (see Figure 5), trunk, and perianal area (which may cause dysuria). The lesions may initially appear as erythematous macules or papules, transforming to vesicles as the disease progresses. The mucocutaneous lesions are usually asymptomatic but can be tender to touch or pressure and may leak fluid.7,10

In only a few cases—caused by CV A6—have lesions in the scalp been reported; the mechanism of action is unknown.10 Instances of lesions invading the nails have been reported, causing desquamation and shedding. This condition is known as onychomadesis.9,11

Continue for diagnosis >>

 

 

DIAGNOSIS
Serologic testing and viral cultures can identify the exact strain of virus causing HFMD and are particularly useful in unusual presentations. Polymerase chain reaction (PCR) testing yields a high sensitivity and specificity for the causative agent. Histologic examination of skin biopsies may show lymphocytic infiltrates and areas of degeneration along the epidermis. However, most cases are diagnosed based on clinical presentation alone.6,12

TREATMENT AND MANAGEMENT
Management of HFMD is primarily symptomatic, consisting of supportive care that includes use of antipyretics, NSAIDs, and adequate fluid intake to prevent dehydration. The disease is usually self-limited, resolving within seven to 10 days without sequelae.6,9 Aseptic meningitis and other severe complications (especially pulmonary and neurologic), most often associated with EV71 infection, can occur in vulnerable populations, including elderly, pregnant, and immunocompromised patients.9,11 Because the virus is excreted directly from palmar lesions onto the hands, proper hygiene and handwashing techniques offer an exceptionally strong protective effect, preventing transmission and reducing morbidity.8

PATIENT OUTCOME
Based on clinical findings and patient history, the patient was diagnosed with HFMD, which she contracted from her son. Laboratory testing and viral cultures were deemed unnecessary in this case. Treatment was symptomatic, and her skin lesions resolved in one to two weeks.

At follow-up, the patient stated her skin lesions resolved completely without leaving any scars. She also indicated that her nails peeled and shed approximately four weeks after diagnosis, but began to regrow normally four months after diagnosis.

CONCLUSION
Clinicians need to recognize that, although it is uncommon outside the pediatric population, HFMD may occur in adults with intact immune systems. The presentation of HFMD in adults may be atypical, including cutaneous lesions in the scalp and shedding of the nails several weeks after diagnosis. Depending on the viral strain involved, adult patients may have more severe illness and may take longer to recover. Therefore, early diagnosis is important to help prevent the spread of infection and reduce the severity of complications.

REFERENCES
1. Patel NN, Patel DN. Erythema multiforme syndrome. Am J Med. 2009;122(7):623-625.
2. Bader MS. Herpes zoster: diagnostic, therapeutic, and preventive approaches. Postgrad Med. 2013;125(5):78-91.
3. Avci O, Ertam I. Viral infections of the face. Clin Derm. 2014;32:715-733.
4. Hartman-Adams H, Banvard C, Juckett G. Impetigo: diagnosis and treatment. Am Fam Physician. 2014;90(4):229-235.
5. Markle W, Conti T, Kad M. Sexually transmitted diseases. Prim Care Clin Office Pract. 2013;40:557-587.
6. Shin JU, Oh SH, Lee JH. A case of hand-foot-mouth disease in an immunocompetent adult. Ann Dermatol. 2010;22(2):216-218.
7. CDC. Notes from the field: severe hand, foot, and mouth disease associated with coxsackievirus A6 - Alabama, Connecticut, California, and Nevada, November 2011-February 2012. MMWR Morb Mortal Wkly Rep. 2012;61(12):213-214.
8. Ruan F, Yang T, Ma H, et al. Risk factors for hand, foot, and mouth disease and herpangina and the preventive effect of hand-washing. Pediatrics. 2011;127(4):e898-e904.
9. Kaminska K, Martinetti G, Lucchini R, et al. Coxsackievirus A6 and hand, foot and mouth Disease: three case reports of familial child-to-immunocompetent adult transmission and a literature review. Case Rep Dermatol. 2013;5(2):203-209.
10. Lønnberg AS, Elberling J, Fischer TK, Skov L. Two cases of hand, foot, and mouth disease involving the scalp. Acta Derm Venereol. 2013;93(4):467-468.
11. Osterback R, Vuorinen T, Linna M, et al. Coxsackievirus A6 and hand, foot, and mouth disease, Finland. Emerging Infect Dis. 2009;15(9):1485-1488.
12. Shea YF, Chan CY, Hung IFN, Chan KH. Hand, foot and mouth disease in an immunocompetent adult due to Coxsackievirus A6. Hong Kong Med J. 2013;19(3):262-264.

A 32-year-old Korean woman presented with a rash on her scalp, face, palms, soles, and genital region and with sores in the oral cavity. The blisters were red and flat with some crusting, particularly on the scalp and face. The patient described the blisters as very painful, adding that it hurt to walk, grasp objects, and drink fluids. Associated symptoms included painful urination, sore throat, malaise, and fever of up to 103°F. She was taking acetaminophen and ibuprofen to alleviate the fever and pain.

 

Medical history was unremarkable. Social history was negative for recent changes in sexual partner or travel to foreign countries.

Physical examination revealed numerous flat, erythematous lesions. Lesions on the face and scalp had developed a weeping, honey-colored crust (see Figure 1 and Figure 2). The lesions were tender to the touch, particularly on the palms and soles.

Further questioning revealed that the patient’s 18-month-old son had exhibited similar symptoms two to three days prior to her illness.

Continue for differential diagnosis >>

 

 

DIFFERENTIAL DIAGNOSIS
Because multiple bacterial and viral diseases manifest in this fashion, the differential diagnosis included the following ­disorders:

Erythema multiforme. This skin condition may result from an allergic or hypersensitivity reaction to certain drugs or from infections. Infections that can cause erythema multiforme include herpes simplex virus and mycoplasma. Patients present with lesions on the palms (see Figure 3), soles, extremities, face, or trunk. The lesions can appear as a nodule, papule, macule, or vesicle. Initially, in the mild form, the lesions may appear as hives or target-shaped rashes, occurring on the face and acral surfaces. A severe form of erythema multiforme, known as Stevens-Johnson syndrome, is characterized by rash, mucosal involvement, and systemic symptoms.1

Herpes zoster. This viral infection is caused by the varicella-zoster virus, which also causes chicken pox. The virus lies dormant within a single sensory ganglion and may reappear as shingles along the dermatome of that nerve. Patients may experience burning or shooting pain with tingling or itching before the rash appears; vesicular lesions with erythematous bases appear days later. The rash occurs unilaterally on the body or face and does not cross the midline. A viral culture may be obtained for identification.2

Herpetic gingivostomatitis. This infection is most commonly caused by herpes simplex virus type 1, the same virus that causes cold sores. Patients may present with ulcerations along the buccal mucosa and gums. The infection manifests with systemic symptoms, including malaise, fever, irritability, and cervical adenopathy. A viral culture will identify the etiology.3

Impetigo. This skin infection is typically caused by bacteria, predominantly Staphylococcus aureus, Streptococcus pyogenes, or a combination. Infections generally occur after a break in the skin surface. The most common presentation is a rash that spreads to different parts of the body after scratching. Skin lesions can occur on the face, lips, or extremities. Initially vesicular, the lesions generally form a honey-colored crust after fluid discharge. The clinician should take a skin or fluid sample from the lesion to culture, which may identify the pathogen.4

Syphilis. This sexually transmitted infection is caused by the spirochete Treponema pallidum. In primary syphilis, patients can develop a painless sore, or chancre, on the genitals, rectal area, or mouth. If left untreated, the disease can progress to secondary syphilis, manifesting as a pale pink or reddish maculopapular rash on the palms and soles. The rash can be associated with fever, sore throat, myalgia, and fatigue. It is important to rule out syphilis because, left untreated, it can lead to cardiac and neurologic complications. Screening tests include VDRL and the rapid plasma reagin (RPR) test, both of which assess for antibodies to the organism, and dark field microscopy of ulcerations to identify the organism.5

Also included in the differential diagnosis for the patient’s symptoms was hand-foot-mouth disease (HFMD), discussed below.

Next page: Discussion >>

 

 

DISCUSSION
HFMD is an acute viral illness most often affecting children younger than 5 and occurring in summer to early fall months. HFMD manifests with fever and papulovesicular eruptions; lesions often appear in the oral cavity first, spreading to the palms, soles, and buttocks. Route of transmission is usually fecal-oral or through respiratory droplets, oral secretions, or direct contact with fluid-filled vesicles.6-8 The highly contagious nature of the virus causes it to spread to close contacts and family members and leads to outbreaks in schools and daycare centers.9,10

In the United States, the most common etiology of HFMD is coxsackievirus A16.7 Another causative agent, enterovirus 71 (EV71), has been found responsible for HFMD epidemics in southeast Asia and Australia.11,12 Recently, the coxsackievirus A6 (CV A6) strain has been linked to outbreaks of HFMD.7,9 This strain may produce an atypical manifestation of skin lesions on the face, trunk, and extremities. The lesions may also appear larger than usual and have a vesiculobullous rather than the more typical papulovesicular appearance. The course of the illness differs in severity depending on the strain of the virus causing HFMD.9

CLINICAL PRESENTATION
The acute phase of HFMD typically begins with prodromal symptoms such as fever, malaise, and sore throat. Erythematous ulcerations usually appear in the oral cavity first (see Figure 4) and often cause symptoms such as sore throat, dysphagia, or dryness. As the disease progresses, cutaneous lesions spread to the face, extremities, interdigital areas (see Figure 5), trunk, and perianal area (which may cause dysuria). The lesions may initially appear as erythematous macules or papules, transforming to vesicles as the disease progresses. The mucocutaneous lesions are usually asymptomatic but can be tender to touch or pressure and may leak fluid.7,10

In only a few cases—caused by CV A6—have lesions in the scalp been reported; the mechanism of action is unknown.10 Instances of lesions invading the nails have been reported, causing desquamation and shedding. This condition is known as onychomadesis.9,11

Continue for diagnosis >>

 

 

DIAGNOSIS
Serologic testing and viral cultures can identify the exact strain of virus causing HFMD and are particularly useful in unusual presentations. Polymerase chain reaction (PCR) testing yields a high sensitivity and specificity for the causative agent. Histologic examination of skin biopsies may show lymphocytic infiltrates and areas of degeneration along the epidermis. However, most cases are diagnosed based on clinical presentation alone.6,12

TREATMENT AND MANAGEMENT
Management of HFMD is primarily symptomatic, consisting of supportive care that includes use of antipyretics, NSAIDs, and adequate fluid intake to prevent dehydration. The disease is usually self-limited, resolving within seven to 10 days without sequelae.6,9 Aseptic meningitis and other severe complications (especially pulmonary and neurologic), most often associated with EV71 infection, can occur in vulnerable populations, including elderly, pregnant, and immunocompromised patients.9,11 Because the virus is excreted directly from palmar lesions onto the hands, proper hygiene and handwashing techniques offer an exceptionally strong protective effect, preventing transmission and reducing morbidity.8

PATIENT OUTCOME
Based on clinical findings and patient history, the patient was diagnosed with HFMD, which she contracted from her son. Laboratory testing and viral cultures were deemed unnecessary in this case. Treatment was symptomatic, and her skin lesions resolved in one to two weeks.

At follow-up, the patient stated her skin lesions resolved completely without leaving any scars. She also indicated that her nails peeled and shed approximately four weeks after diagnosis, but began to regrow normally four months after diagnosis.

CONCLUSION
Clinicians need to recognize that, although it is uncommon outside the pediatric population, HFMD may occur in adults with intact immune systems. The presentation of HFMD in adults may be atypical, including cutaneous lesions in the scalp and shedding of the nails several weeks after diagnosis. Depending on the viral strain involved, adult patients may have more severe illness and may take longer to recover. Therefore, early diagnosis is important to help prevent the spread of infection and reduce the severity of complications.

REFERENCES
1. Patel NN, Patel DN. Erythema multiforme syndrome. Am J Med. 2009;122(7):623-625.
2. Bader MS. Herpes zoster: diagnostic, therapeutic, and preventive approaches. Postgrad Med. 2013;125(5):78-91.
3. Avci O, Ertam I. Viral infections of the face. Clin Derm. 2014;32:715-733.
4. Hartman-Adams H, Banvard C, Juckett G. Impetigo: diagnosis and treatment. Am Fam Physician. 2014;90(4):229-235.
5. Markle W, Conti T, Kad M. Sexually transmitted diseases. Prim Care Clin Office Pract. 2013;40:557-587.
6. Shin JU, Oh SH, Lee JH. A case of hand-foot-mouth disease in an immunocompetent adult. Ann Dermatol. 2010;22(2):216-218.
7. CDC. Notes from the field: severe hand, foot, and mouth disease associated with coxsackievirus A6 - Alabama, Connecticut, California, and Nevada, November 2011-February 2012. MMWR Morb Mortal Wkly Rep. 2012;61(12):213-214.
8. Ruan F, Yang T, Ma H, et al. Risk factors for hand, foot, and mouth disease and herpangina and the preventive effect of hand-washing. Pediatrics. 2011;127(4):e898-e904.
9. Kaminska K, Martinetti G, Lucchini R, et al. Coxsackievirus A6 and hand, foot and mouth Disease: three case reports of familial child-to-immunocompetent adult transmission and a literature review. Case Rep Dermatol. 2013;5(2):203-209.
10. Lønnberg AS, Elberling J, Fischer TK, Skov L. Two cases of hand, foot, and mouth disease involving the scalp. Acta Derm Venereol. 2013;93(4):467-468.
11. Osterback R, Vuorinen T, Linna M, et al. Coxsackievirus A6 and hand, foot, and mouth disease, Finland. Emerging Infect Dis. 2009;15(9):1485-1488.
12. Shea YF, Chan CY, Hung IFN, Chan KH. Hand, foot and mouth disease in an immunocompetent adult due to Coxsackievirus A6. Hong Kong Med J. 2013;19(3):262-264.

Issue
Clinician Reviews - 25(1)
Issue
Clinician Reviews - 25(1)
Page Number
37,40-42
Page Number
37,40-42
Publications
Publications
Topics
Article Type
Display Headline
Woman, 32, With Crusty Red Blisters
Display Headline
Woman, 32, With Crusty Red Blisters
Legacy Keywords
facial lesions, red blisters, erythema multiforme, herpes zoster, herpetic gingivostomatis, impetigo, syphilis, hand-foot-mouth disease, HFMD
Legacy Keywords
facial lesions, red blisters, erythema multiforme, herpes zoster, herpetic gingivostomatis, impetigo, syphilis, hand-foot-mouth disease, HFMD
Sections
Disallow All Ads
Alternative CME
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Article PDF Media

Second of 2 parts: The mysteries of psychiatry maintenance of certification, further unraveled

Article Type
Changed
Thu, 03/28/2019 - 15:33
Display Headline
Second of 2 parts: The mysteries of psychiatry maintenance of certification, further unraveled

To recap what I discussed in Part 1 of this article (December 2014): As part of a trend across all medical specialty boards, the American Board of Psychiatry and Neurology (ABPN) instituted a recertification pro­cess for all new general psychiatry certifications, starting October 1, 1994.1 In 2000, the specialties that comprise the American Board of Medical Specialties (ABMS) agreed to develop a comprehensive maintenance of certification (MOC) process to demonstrate ongoing learning and com­petency beyond what can be captured by a recertification examination. All ABMS member boards now use a 4-part process for recertification.

A great deal of professional and personal importance has been attached to maintaining one’s general and sub­specialty certifications. To that end, the 2 parts of this article highlight current ABPN MOC requirements and provide resources for understanding, tracking, and completing the self-assessment (SA) and performance-in-practice (PIP) components.

In this installment, I examine 3 components of MOC:
   • continuing medical education (CME), including SA requirements
   • improvement in medical practice (PIP)
   • continuous maintenance of certification (C-MOC)

In addition to this review, all physicians who are subject to MOC should download and read the 20-page revised MOC Program booklet v. 2.1 (May 2014).2

Continuing medical education
The CME requirement is clear: All diplo­mate physicians must accrue, on average, 30 Category-1 CME credits a year; the CME must be relevant to the specialty or subspecialty in which the diplomate prac­tices.3 For physicians who hold >1 ABPN certificates, the total CME requirement is the same; CME credits can be applied across each specialty and subspecialty.

The May 2014 MOC revision states that, for physicians who certified or recertified between 2005 and 2011 and who applied for the 2015 examination in 2014, the required CME credit total is 270.2 For all subsequent years of certification or recertification, including 2012, diplomates are enrolled in C-MOC, which is described below.2

To even out the accrual of CME credits across the prior 10 years, ABPN mandates that, for diplomates who certified or recer­tified between 2005 and 2011, one hundred fifty of the CME credits be accrued in the 5 years before they apply for the examina­tion. Diplomates in C-MOC should accrue, on average, 30 CME credits a year in each of the 3-year blocks (ie, 90 units in each block).2


Self-assessment

SA is a specific form of CME that is designed to provide comprehensive test-based feedback on knowledge acquired, to enhance the learning process.4 SA CME feedback must include:
   • the correct answer to each test question
   • recommended literature resources for each question
   • performance compared to peers on each question.

Given the structured nature of SA activi­ties, beginning January 1, 2014, one must use only ABPN-approved SA products (see Related Resources for a list of APBN-approved SA products).5

Table 1 and Table 2 outline SA require­ments for, respectively, physicians who certified or recertified from 2005 through 2011, and those who certified or recertified in 2012 (and later). The SA requirement increases after 2011 to 24 credits in each 3-year block (8 credits a year, on average).2 Multiple SA activities can be used to fulfill the credit requirement of each 3-year block.




Note: Credits accrued by performing SA activities count toward the CME credit total.


Improvement in medical practice, or PIP

Physicians who are active clinically must complete PIP modules. Each module comprises peer or patient feedback plus a clinical aspect. The May 2014 MOC revi­sion simplified the feedback process to mandate peer or patient feedback—but not both, as required previously.2 For the feedback PIP module, the physician selects 5 peers or patients to complete review forms, examines the results, and creates a plan of improvement. An exception to this “rule of 5” applies to diplomates who have a supervisor capable of evaluating all gen­eral competencies, defined below.

Related Resources
provides a link to ABPN-created forms.

Within 24 months, but not sooner than 1 month, 5 peers or patients (or 1 appli­cable supervisor) are selected to complete review forms; changes in practice are noted. The same peers or patients might be selected for a second review. As noted in Table 1 and Table 2, the number of PIP modules is fewer for physicians who certi­fied or recertified between 2005 and 2011; from 2012 onward, 1 PIP clinical module is required in each 3-year block.2

There are 6 ABPN-approved feedback module options, of which the diplomate must choose 1 in any given block2:
   • 5 patient surveys
   • 5 peer evaluations of general competenciesa
   • 5 resident evaluations of general competenciesa  
   • 360° evaluation of general competencies,a with 5 respondents
   • institutional peer review of general competencies,a with 5 respondents
   • 1 supervisor evaluation of general competencies.a

 

 

aGeneral competencies include patient care; practice-based learning and improvement; professionalism; medical knowledge; interpersonal and communication skills; and system-based practices.

Although many institutions have a quality improvement (QI) program, that program must be approved by the Multi-Specialty MOC Portfolio Approval Program sponsored by ABMS for a clinician to receive credit for 1 PIP clinical module. If the approved QI program includes patient or peer feedback (eg, a survey), the diplo­ mate can receive credit for 1 PIP feedback module.2

For the clinical PIP module, the physician selects 5 charts for review and examines them based on criteria found in an ABPN-approved (starting in 2014) PIP product. (Related Resources provides a link to this list.) After reviewing the initial 5 charts, a plan for improvement is created. Within 24 months, but no sooner than 1 month, 5 charts are again selected and reviewed, and changes in practice are noted. The same charts can be selected for the second review.

As noted in Table 1 and Table 2, the number of PIP modules is fewer for those who initially certified or recertified between 2005 and 2011; from 2012 onward, 1 PIP clinical module is required in each 3-year block.2


The C-MOC process

Physicians who certified or recertified in 2012, or who will certify or recertify after that year, are enrolled automatically in C-MOC.6,7 The purpose of C-MOC is to keep diplomates on track to fulfill the higher level of SA requirements that began with this group; this is done by mandating use of the ABPN Physician Folios system. As shown in Table 2, there is no longer a 10-year cycle; instead, there are continuous 3-year stages, within which each diplomate must accrue 90 CME cred­its (on average, 30 credits a year), 24 SA credits (on average, 8 a year), 1 PIP clinical module, and 1 PIP feedback module.6,7

The first 3-year block of C-MOC require­ments will be waived for physicians who complete Accreditation Council on Graduate Medical Education–accredited or ABPN-approved subspecialty training in 2012 or later—if they pass the corre­sponding ABPN subspecialty examination during the first 3-year block of enrollment in C-MOC.2 For diplomates enrolled in C-MOC, failure to track progress of each 3-year block, via the ABPN Physician Folios system, has significant consequences: Those who do not complete the first stage of the program by the end of 3 years will be listed on the ABPN Web site as “certified— not meeting MOC requirements.” Those who do not complete 2 stages by the end of 6 years will be listed as “not certified.”2

Cognitive exam still in place. The only remnant of the old 10-year cycle is the requirement to pass the cognitive examina­tion every 10 years, although the exam can be taken earlier if the diplomate wishes. If all requirements are met and one does not sit for, or fails, the exam, the ABPN Web site will report the diplomate as “not meet­ing MOC requirements.” One can retake the exam within 1 year of the failed or missed exam, but a subsequent failure or missed exam will result in being listed as “not certified.”2

Fee structure. Instead of a single fee paid at the time of the exam(s), physicians in the C-MOC program pay an annual fee that covers participation in ABPN Physician Folios and 1 exam in a 10-year period. Fewer than 10 years of participation, or applying for a combined examination (for diplomates who hold multiple certifica­tions), requires an additional fee.7 


Bottom Line

Maintenance of certification (MOC) is manageable, although it requires you to be familiar with its various elements. Those elements include continuing medical education (CME requirements); the additional self-assessment component of CME; performance-in-practice modules; and continuous maintenance of certification. The MOC program booklet of the American Board of Psychiatry and Neurology provides all necessary details.

Disclosure
Dr. Meyer reports no financial relationships with any company whose products are mentioned in this article or with manufacturers of competing products.

References


1. Faulkner LR, Tivnan PW, Winstead DK, et al. The ABPN Maintenance of Certification Program for psychiatrists: past history, current status, and future directions. Acad Psychiatry. 2008;32(3):241-248.
2. Maintenance of Certification Program. American Board of Psychiatry and Neurology Inc. http://www.abpn.com/ downloads/moc/moc_web_doc.pdf. Published May 2014. Accessed August 25, 2014.
3. Faulkner LR, Vondrak PA. Frequently asked questions about maintenance of certification (MOC). J Clin Psychiatry. 2010;71(5):632-633.
4. Ebert MH, Faulkner L, Stubbe DE, et al. Maintenance of certification in psychiatry. J Clin Psychiatry. 2009;70(10):e39.
5. Approved MOC Products. American Board of Psychiatry and Neurology Inc. http://www.abpn.com/moc_products. asp. Accessed August 25, 2014.
6. Continuous MOC (C-MOC). American Board of Psychiatry and Neurology Inc. http://www.abpn.com/downloads/ moc/ContinuousCertificationApproach_0311.pdf. Accessed August 25, 2014.
7. C-MOC Program Overview. American Board of Psychiatry and Neurology Inc. http://www.abpn.com/downloads/ moc/moc-handouts-CMOC-051314.pdf. Published May 13, 2014. Accessed August 25, 2014.

Article PDF
Author and Disclosure Information

Jonathan M. Meyer, MD
Psychopharmacology Consultant
California Department of State Hospitals
Medical Director
Mental Health Intensive Case Management
VA San Diego Healthcare System
San Diego, California
Assistant Clinical Professor of Psychiatry
University of California, San Diego
San Diego, California
Associate Clinical Professor of Psychiatry
Loma Linda University
Loma Linda, California

Issue
Current Psychiatry - 14(1)
Publications
Topics
Page Number
37-41
Legacy Keywords
MOC, maintenance of certification, MOC credit requirements, American Board of Psychiatry and Neurology (ABPN)
Sections
Author and Disclosure Information

Jonathan M. Meyer, MD
Psychopharmacology Consultant
California Department of State Hospitals
Medical Director
Mental Health Intensive Case Management
VA San Diego Healthcare System
San Diego, California
Assistant Clinical Professor of Psychiatry
University of California, San Diego
San Diego, California
Associate Clinical Professor of Psychiatry
Loma Linda University
Loma Linda, California

Author and Disclosure Information

Jonathan M. Meyer, MD
Psychopharmacology Consultant
California Department of State Hospitals
Medical Director
Mental Health Intensive Case Management
VA San Diego Healthcare System
San Diego, California
Assistant Clinical Professor of Psychiatry
University of California, San Diego
San Diego, California
Associate Clinical Professor of Psychiatry
Loma Linda University
Loma Linda, California

Article PDF
Article PDF

To recap what I discussed in Part 1 of this article (December 2014): As part of a trend across all medical specialty boards, the American Board of Psychiatry and Neurology (ABPN) instituted a recertification pro­cess for all new general psychiatry certifications, starting October 1, 1994.1 In 2000, the specialties that comprise the American Board of Medical Specialties (ABMS) agreed to develop a comprehensive maintenance of certification (MOC) process to demonstrate ongoing learning and com­petency beyond what can be captured by a recertification examination. All ABMS member boards now use a 4-part process for recertification.

A great deal of professional and personal importance has been attached to maintaining one’s general and sub­specialty certifications. To that end, the 2 parts of this article highlight current ABPN MOC requirements and provide resources for understanding, tracking, and completing the self-assessment (SA) and performance-in-practice (PIP) components.

In this installment, I examine 3 components of MOC:
   • continuing medical education (CME), including SA requirements
   • improvement in medical practice (PIP)
   • continuous maintenance of certification (C-MOC)

In addition to this review, all physicians who are subject to MOC should download and read the 20-page revised MOC Program booklet v. 2.1 (May 2014).2

Continuing medical education
The CME requirement is clear: All diplo­mate physicians must accrue, on average, 30 Category-1 CME credits a year; the CME must be relevant to the specialty or subspecialty in which the diplomate prac­tices.3 For physicians who hold >1 ABPN certificates, the total CME requirement is the same; CME credits can be applied across each specialty and subspecialty.

The May 2014 MOC revision states that, for physicians who certified or recertified between 2005 and 2011 and who applied for the 2015 examination in 2014, the required CME credit total is 270.2 For all subsequent years of certification or recertification, including 2012, diplomates are enrolled in C-MOC, which is described below.2

To even out the accrual of CME credits across the prior 10 years, ABPN mandates that, for diplomates who certified or recer­tified between 2005 and 2011, one hundred fifty of the CME credits be accrued in the 5 years before they apply for the examina­tion. Diplomates in C-MOC should accrue, on average, 30 CME credits a year in each of the 3-year blocks (ie, 90 units in each block).2


Self-assessment

SA is a specific form of CME that is designed to provide comprehensive test-based feedback on knowledge acquired, to enhance the learning process.4 SA CME feedback must include:
   • the correct answer to each test question
   • recommended literature resources for each question
   • performance compared to peers on each question.

Given the structured nature of SA activi­ties, beginning January 1, 2014, one must use only ABPN-approved SA products (see Related Resources for a list of APBN-approved SA products).5

Table 1 and Table 2 outline SA require­ments for, respectively, physicians who certified or recertified from 2005 through 2011, and those who certified or recertified in 2012 (and later). The SA requirement increases after 2011 to 24 credits in each 3-year block (8 credits a year, on average).2 Multiple SA activities can be used to fulfill the credit requirement of each 3-year block.




Note: Credits accrued by performing SA activities count toward the CME credit total.


Improvement in medical practice, or PIP

Physicians who are active clinically must complete PIP modules. Each module comprises peer or patient feedback plus a clinical aspect. The May 2014 MOC revi­sion simplified the feedback process to mandate peer or patient feedback—but not both, as required previously.2 For the feedback PIP module, the physician selects 5 peers or patients to complete review forms, examines the results, and creates a plan of improvement. An exception to this “rule of 5” applies to diplomates who have a supervisor capable of evaluating all gen­eral competencies, defined below.

Related Resources
provides a link to ABPN-created forms.

Within 24 months, but not sooner than 1 month, 5 peers or patients (or 1 appli­cable supervisor) are selected to complete review forms; changes in practice are noted. The same peers or patients might be selected for a second review. As noted in Table 1 and Table 2, the number of PIP modules is fewer for physicians who certi­fied or recertified between 2005 and 2011; from 2012 onward, 1 PIP clinical module is required in each 3-year block.2

There are 6 ABPN-approved feedback module options, of which the diplomate must choose 1 in any given block2:
   • 5 patient surveys
   • 5 peer evaluations of general competenciesa
   • 5 resident evaluations of general competenciesa  
   • 360° evaluation of general competencies,a with 5 respondents
   • institutional peer review of general competencies,a with 5 respondents
   • 1 supervisor evaluation of general competencies.a

 

 

aGeneral competencies include patient care; practice-based learning and improvement; professionalism; medical knowledge; interpersonal and communication skills; and system-based practices.

Although many institutions have a quality improvement (QI) program, that program must be approved by the Multi-Specialty MOC Portfolio Approval Program sponsored by ABMS for a clinician to receive credit for 1 PIP clinical module. If the approved QI program includes patient or peer feedback (eg, a survey), the diplo­ mate can receive credit for 1 PIP feedback module.2

For the clinical PIP module, the physician selects 5 charts for review and examines them based on criteria found in an ABPN-approved (starting in 2014) PIP product. (Related Resources provides a link to this list.) After reviewing the initial 5 charts, a plan for improvement is created. Within 24 months, but no sooner than 1 month, 5 charts are again selected and reviewed, and changes in practice are noted. The same charts can be selected for the second review.

As noted in Table 1 and Table 2, the number of PIP modules is fewer for those who initially certified or recertified between 2005 and 2011; from 2012 onward, 1 PIP clinical module is required in each 3-year block.2


The C-MOC process

Physicians who certified or recertified in 2012, or who will certify or recertify after that year, are enrolled automatically in C-MOC.6,7 The purpose of C-MOC is to keep diplomates on track to fulfill the higher level of SA requirements that began with this group; this is done by mandating use of the ABPN Physician Folios system. As shown in Table 2, there is no longer a 10-year cycle; instead, there are continuous 3-year stages, within which each diplomate must accrue 90 CME cred­its (on average, 30 credits a year), 24 SA credits (on average, 8 a year), 1 PIP clinical module, and 1 PIP feedback module.6,7

The first 3-year block of C-MOC require­ments will be waived for physicians who complete Accreditation Council on Graduate Medical Education–accredited or ABPN-approved subspecialty training in 2012 or later—if they pass the corre­sponding ABPN subspecialty examination during the first 3-year block of enrollment in C-MOC.2 For diplomates enrolled in C-MOC, failure to track progress of each 3-year block, via the ABPN Physician Folios system, has significant consequences: Those who do not complete the first stage of the program by the end of 3 years will be listed on the ABPN Web site as “certified— not meeting MOC requirements.” Those who do not complete 2 stages by the end of 6 years will be listed as “not certified.”2

Cognitive exam still in place. The only remnant of the old 10-year cycle is the requirement to pass the cognitive examina­tion every 10 years, although the exam can be taken earlier if the diplomate wishes. If all requirements are met and one does not sit for, or fails, the exam, the ABPN Web site will report the diplomate as “not meet­ing MOC requirements.” One can retake the exam within 1 year of the failed or missed exam, but a subsequent failure or missed exam will result in being listed as “not certified.”2

Fee structure. Instead of a single fee paid at the time of the exam(s), physicians in the C-MOC program pay an annual fee that covers participation in ABPN Physician Folios and 1 exam in a 10-year period. Fewer than 10 years of participation, or applying for a combined examination (for diplomates who hold multiple certifica­tions), requires an additional fee.7 


Bottom Line

Maintenance of certification (MOC) is manageable, although it requires you to be familiar with its various elements. Those elements include continuing medical education (CME requirements); the additional self-assessment component of CME; performance-in-practice modules; and continuous maintenance of certification. The MOC program booklet of the American Board of Psychiatry and Neurology provides all necessary details.

Disclosure
Dr. Meyer reports no financial relationships with any company whose products are mentioned in this article or with manufacturers of competing products.

To recap what I discussed in Part 1 of this article (December 2014): As part of a trend across all medical specialty boards, the American Board of Psychiatry and Neurology (ABPN) instituted a recertification pro­cess for all new general psychiatry certifications, starting October 1, 1994.1 In 2000, the specialties that comprise the American Board of Medical Specialties (ABMS) agreed to develop a comprehensive maintenance of certification (MOC) process to demonstrate ongoing learning and com­petency beyond what can be captured by a recertification examination. All ABMS member boards now use a 4-part process for recertification.

A great deal of professional and personal importance has been attached to maintaining one’s general and sub­specialty certifications. To that end, the 2 parts of this article highlight current ABPN MOC requirements and provide resources for understanding, tracking, and completing the self-assessment (SA) and performance-in-practice (PIP) components.

In this installment, I examine 3 components of MOC:
   • continuing medical education (CME), including SA requirements
   • improvement in medical practice (PIP)
   • continuous maintenance of certification (C-MOC)

In addition to this review, all physicians who are subject to MOC should download and read the 20-page revised MOC Program booklet v. 2.1 (May 2014).2

Continuing medical education
The CME requirement is clear: All diplo­mate physicians must accrue, on average, 30 Category-1 CME credits a year; the CME must be relevant to the specialty or subspecialty in which the diplomate prac­tices.3 For physicians who hold >1 ABPN certificates, the total CME requirement is the same; CME credits can be applied across each specialty and subspecialty.

The May 2014 MOC revision states that, for physicians who certified or recertified between 2005 and 2011 and who applied for the 2015 examination in 2014, the required CME credit total is 270.2 For all subsequent years of certification or recertification, including 2012, diplomates are enrolled in C-MOC, which is described below.2

To even out the accrual of CME credits across the prior 10 years, ABPN mandates that, for diplomates who certified or recer­tified between 2005 and 2011, one hundred fifty of the CME credits be accrued in the 5 years before they apply for the examina­tion. Diplomates in C-MOC should accrue, on average, 30 CME credits a year in each of the 3-year blocks (ie, 90 units in each block).2


Self-assessment

SA is a specific form of CME that is designed to provide comprehensive test-based feedback on knowledge acquired, to enhance the learning process.4 SA CME feedback must include:
   • the correct answer to each test question
   • recommended literature resources for each question
   • performance compared to peers on each question.

Given the structured nature of SA activi­ties, beginning January 1, 2014, one must use only ABPN-approved SA products (see Related Resources for a list of APBN-approved SA products).5

Table 1 and Table 2 outline SA require­ments for, respectively, physicians who certified or recertified from 2005 through 2011, and those who certified or recertified in 2012 (and later). The SA requirement increases after 2011 to 24 credits in each 3-year block (8 credits a year, on average).2 Multiple SA activities can be used to fulfill the credit requirement of each 3-year block.




Note: Credits accrued by performing SA activities count toward the CME credit total.


Improvement in medical practice, or PIP

Physicians who are active clinically must complete PIP modules. Each module comprises peer or patient feedback plus a clinical aspect. The May 2014 MOC revi­sion simplified the feedback process to mandate peer or patient feedback—but not both, as required previously.2 For the feedback PIP module, the physician selects 5 peers or patients to complete review forms, examines the results, and creates a plan of improvement. An exception to this “rule of 5” applies to diplomates who have a supervisor capable of evaluating all gen­eral competencies, defined below.

Related Resources
provides a link to ABPN-created forms.

Within 24 months, but not sooner than 1 month, 5 peers or patients (or 1 appli­cable supervisor) are selected to complete review forms; changes in practice are noted. The same peers or patients might be selected for a second review. As noted in Table 1 and Table 2, the number of PIP modules is fewer for physicians who certi­fied or recertified between 2005 and 2011; from 2012 onward, 1 PIP clinical module is required in each 3-year block.2

There are 6 ABPN-approved feedback module options, of which the diplomate must choose 1 in any given block2:
   • 5 patient surveys
   • 5 peer evaluations of general competenciesa
   • 5 resident evaluations of general competenciesa  
   • 360° evaluation of general competencies,a with 5 respondents
   • institutional peer review of general competencies,a with 5 respondents
   • 1 supervisor evaluation of general competencies.a

 

 

aGeneral competencies include patient care; practice-based learning and improvement; professionalism; medical knowledge; interpersonal and communication skills; and system-based practices.

Although many institutions have a quality improvement (QI) program, that program must be approved by the Multi-Specialty MOC Portfolio Approval Program sponsored by ABMS for a clinician to receive credit for 1 PIP clinical module. If the approved QI program includes patient or peer feedback (eg, a survey), the diplo­ mate can receive credit for 1 PIP feedback module.2

For the clinical PIP module, the physician selects 5 charts for review and examines them based on criteria found in an ABPN-approved (starting in 2014) PIP product. (Related Resources provides a link to this list.) After reviewing the initial 5 charts, a plan for improvement is created. Within 24 months, but no sooner than 1 month, 5 charts are again selected and reviewed, and changes in practice are noted. The same charts can be selected for the second review.

As noted in Table 1 and Table 2, the number of PIP modules is fewer for those who initially certified or recertified between 2005 and 2011; from 2012 onward, 1 PIP clinical module is required in each 3-year block.2


The C-MOC process

Physicians who certified or recertified in 2012, or who will certify or recertify after that year, are enrolled automatically in C-MOC.6,7 The purpose of C-MOC is to keep diplomates on track to fulfill the higher level of SA requirements that began with this group; this is done by mandating use of the ABPN Physician Folios system. As shown in Table 2, there is no longer a 10-year cycle; instead, there are continuous 3-year stages, within which each diplomate must accrue 90 CME cred­its (on average, 30 credits a year), 24 SA credits (on average, 8 a year), 1 PIP clinical module, and 1 PIP feedback module.6,7

The first 3-year block of C-MOC require­ments will be waived for physicians who complete Accreditation Council on Graduate Medical Education–accredited or ABPN-approved subspecialty training in 2012 or later—if they pass the corre­sponding ABPN subspecialty examination during the first 3-year block of enrollment in C-MOC.2 For diplomates enrolled in C-MOC, failure to track progress of each 3-year block, via the ABPN Physician Folios system, has significant consequences: Those who do not complete the first stage of the program by the end of 3 years will be listed on the ABPN Web site as “certified— not meeting MOC requirements.” Those who do not complete 2 stages by the end of 6 years will be listed as “not certified.”2

Cognitive exam still in place. The only remnant of the old 10-year cycle is the requirement to pass the cognitive examina­tion every 10 years, although the exam can be taken earlier if the diplomate wishes. If all requirements are met and one does not sit for, or fails, the exam, the ABPN Web site will report the diplomate as “not meet­ing MOC requirements.” One can retake the exam within 1 year of the failed or missed exam, but a subsequent failure or missed exam will result in being listed as “not certified.”2

Fee structure. Instead of a single fee paid at the time of the exam(s), physicians in the C-MOC program pay an annual fee that covers participation in ABPN Physician Folios and 1 exam in a 10-year period. Fewer than 10 years of participation, or applying for a combined examination (for diplomates who hold multiple certifica­tions), requires an additional fee.7 


Bottom Line

Maintenance of certification (MOC) is manageable, although it requires you to be familiar with its various elements. Those elements include continuing medical education (CME requirements); the additional self-assessment component of CME; performance-in-practice modules; and continuous maintenance of certification. The MOC program booklet of the American Board of Psychiatry and Neurology provides all necessary details.

Disclosure
Dr. Meyer reports no financial relationships with any company whose products are mentioned in this article or with manufacturers of competing products.

References


1. Faulkner LR, Tivnan PW, Winstead DK, et al. The ABPN Maintenance of Certification Program for psychiatrists: past history, current status, and future directions. Acad Psychiatry. 2008;32(3):241-248.
2. Maintenance of Certification Program. American Board of Psychiatry and Neurology Inc. http://www.abpn.com/ downloads/moc/moc_web_doc.pdf. Published May 2014. Accessed August 25, 2014.
3. Faulkner LR, Vondrak PA. Frequently asked questions about maintenance of certification (MOC). J Clin Psychiatry. 2010;71(5):632-633.
4. Ebert MH, Faulkner L, Stubbe DE, et al. Maintenance of certification in psychiatry. J Clin Psychiatry. 2009;70(10):e39.
5. Approved MOC Products. American Board of Psychiatry and Neurology Inc. http://www.abpn.com/moc_products. asp. Accessed August 25, 2014.
6. Continuous MOC (C-MOC). American Board of Psychiatry and Neurology Inc. http://www.abpn.com/downloads/ moc/ContinuousCertificationApproach_0311.pdf. Accessed August 25, 2014.
7. C-MOC Program Overview. American Board of Psychiatry and Neurology Inc. http://www.abpn.com/downloads/ moc/moc-handouts-CMOC-051314.pdf. Published May 13, 2014. Accessed August 25, 2014.

References


1. Faulkner LR, Tivnan PW, Winstead DK, et al. The ABPN Maintenance of Certification Program for psychiatrists: past history, current status, and future directions. Acad Psychiatry. 2008;32(3):241-248.
2. Maintenance of Certification Program. American Board of Psychiatry and Neurology Inc. http://www.abpn.com/ downloads/moc/moc_web_doc.pdf. Published May 2014. Accessed August 25, 2014.
3. Faulkner LR, Vondrak PA. Frequently asked questions about maintenance of certification (MOC). J Clin Psychiatry. 2010;71(5):632-633.
4. Ebert MH, Faulkner L, Stubbe DE, et al. Maintenance of certification in psychiatry. J Clin Psychiatry. 2009;70(10):e39.
5. Approved MOC Products. American Board of Psychiatry and Neurology Inc. http://www.abpn.com/moc_products. asp. Accessed August 25, 2014.
6. Continuous MOC (C-MOC). American Board of Psychiatry and Neurology Inc. http://www.abpn.com/downloads/ moc/ContinuousCertificationApproach_0311.pdf. Accessed August 25, 2014.
7. C-MOC Program Overview. American Board of Psychiatry and Neurology Inc. http://www.abpn.com/downloads/ moc/moc-handouts-CMOC-051314.pdf. Published May 13, 2014. Accessed August 25, 2014.

Issue
Current Psychiatry - 14(1)
Issue
Current Psychiatry - 14(1)
Page Number
37-41
Page Number
37-41
Publications
Publications
Topics
Article Type
Display Headline
Second of 2 parts: The mysteries of psychiatry maintenance of certification, further unraveled
Display Headline
Second of 2 parts: The mysteries of psychiatry maintenance of certification, further unraveled
Legacy Keywords
MOC, maintenance of certification, MOC credit requirements, American Board of Psychiatry and Neurology (ABPN)
Legacy Keywords
MOC, maintenance of certification, MOC credit requirements, American Board of Psychiatry and Neurology (ABPN)
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media

Choosing a treatment for disruptive, impulse-control, and conduct disorders

Article Type
Changed
Fri, 01/25/2019 - 14:39
Display Headline
Choosing a treatment for disruptive, impulse-control, and conduct disorders

Chronic  disruptive and impulsive behaviors are significant concerns for psychiatric clinicians because of their persistence and potential legal ramifications. To date, few studies have assessed treatment options for pyromania, oppositional defiant disorder (ODD), intermittent explosive disorder (IED), kleptomania, and conduct disorder (CD).

This article reviews the literature on the treatment of these disorders, focusing primarily on randomized, controlled studies. Because of the lack of clinical studies for these disorders, however, case studies and open tri­als are mentioned for reference. Summaries of supported medication and psychological interventions are provided for each disorder.


Categorizing impulse-control disorders
The DSM-5 created a new chapter on disruptive, impulse control, and conduct disorders that brought together disorders previously classified as disorders usually first diagnosed in infancy, childhood, or adolescence (ODD, CD) and impulse-control disorders not elsewhere classified. These disorders are unified by the presence of difficult, disruptive, aggressive, or antisocial behavior. Disruptive, aggressive, or antisocial behavior usu­ally is a multifaceted behavior, often associated with physical or verbal injury to self, others, or objects or with violating the rights of others. These behaviors can appear in several forms and can be defensive, premedi­tated, or impulsive.

Despite a high prevalence in the general population1 and in psychi­atric cohorts,2 disruptive and impulse-control disorders have been rela­tively understudied. Controlled trials of treatments do not exist for many impulse-control disorders, and there are no FDA-approved medications for any of these disorders.
 

Oppositional defiant disorder
Irritability, anger, defiance, and temper are specific descriptors of ODD. ODD seems to be a developmental antecedent for some youth with CD, suggesting that these dis­orders could reflect different stages of a spectrum of disruptive behavior. Transient oppositional behavior is common among children and adolescents, but ODD occurs in 1% to 11% of youth.3 The disorder is more prevalent among boys before puberty and has an equal sex prevalence in young people after puberty.

Regrettably, most ODD research has included patients with comorbidities, most commonly attention-deficit/hyperactivity disorder (ADHD). Because of this limita­tion, the drugs and programs discussed below are drawn from meta-analyses and review articles.

Pharmacotherapy. No medications have been FDA-approved for ODD. Studies assess­ing ODD have employed a variety of meth­odologies, not all of which are double-blind. The meta-analyses and reviews cited in this section include both randomized and open trials, and should be interpreted as such.

Stimulants are commonly used to treat ODD because of a high comorbidity rate with ADHD, and these drugs have improved ODD symptoms in randomized trials.4 Methylphenidate and d-amphetamine have shown some efficacy in trials of ODD and CD.5-7 These medications are most commonly used when ODD is complicated by ADHD symptoms.

Antipsychotics also have been used to treat ODD, with the largest body of research suggesting that risperidone has some effi­cacy. Risperidone usually is considered a second- or third-line option because it has been associated with adverse effects in chil­dren and adolescents and requires caution in younger populations, despite its potential efficacy.4,8-10

Alpha-2 agonists—clonidine and guanfa­cine—have shown some efficacy in treating ODD but have not been studied extensively. Studies of clonidine, however, often have grouped ODD, CD, and ADHD, which lim­its our understanding of this medication for ODD alone.4,5,11

Atomoxetine has been studied for ODD, but its efficacy is limited, with different meta-analyses finding distinct results regarding efficacy. One explanation for these dispa­rate findings is that improvements in oppo­sitional symptoms may be secondary to improvement in ADHD symptoms.7,12-14

Psychological treatments. As noted for pharmacotherapy, this section provides gen­eral information on empirically studied ther­apies. A series of meta-analyses have been included for further review, but are not iso­lated to randomized, controlled studies.

Individual therapy has shown consistent improvements in ODD. Examples include behavior modification therapy and parent-child interaction therapy. These sessions emphasize skills to manage outbursts and erratic emotionality. Emotion regulation and behavior and social skills training have shown significant reductions in target mea­sures. Some of these programs incorporate both patient and parent components.15-17

Family/teacher training programs such as “Helping the Noncompliant Child” and the “Triple P” have yielded significant improve­ments. These programs focus on ways to manage the child’s oppositional behavior at home and in the classroom, as well as strate­gies to limit positive reinforcement for prob­lem behaviors.17-20

Group programs have shown some effi­cacy with ODD. These programs cover a wide number of needs and intents. Examples include the “Incredible Years” program and the Community Parent Education Program. Research has found that these programs show some efficacy as preemptive measures to reduce the rate of ODD among adolescents.

Conclusions. A number of treatment options for ODD have shown some efficacy. However, many of these options have only been studied in patients with comorbid ADHD, which limits current knowledge about ODD as a distinct disorder.

 

 


Intermittent explosive disorder
IED is defined by recurrent, significant out­bursts of aggression, often leading to assaul­tive acts against people or property, which are disproportionate to outside stressors and are not better explained by another psy­chiatric diagnosis. Research suggests IED is common, with 6.3% of a community sample meeting criteria for lifetime IED.21

IED symptoms tend to start in adolescence and appear to be chronic.21,22 People with IED regard their behavior as distressing and prob­lematic.22 Outbursts generally are short-lived (usually <30 minutes) and frequent (multiple times a month22). Legal and occupational dif­ficulties are common.22

Pharmacotherapy. Data on drug treatment for IED comes for a small set of double-blind studies (Table). Although pharmacotherapies have been studied for treating aggression, impulsivity, and vio­lent behavior, only 5 controlled studies are specific to IED.


A double-blind, randomized, placebo-controlled trial of fluoxetine in 100 par­ticipants with IED found that fluoxetine produced a sustained reduction in aggression and irritability as early as the second week of treatment. Full or partial remission of impul­sive aggressive behaviors occurred in 46% of fluoxetine-treated subjects. These findings have been supported by studies assessing other samples of aggressive patients, but not specifically IED.23,24 Another treatment study found that oxcarbazepine produced signifi­cant improvements in IED symptom severity, specifically on impulsive aggression.25

In a randomized, double-blind, placebo-controlled study, 96 participants with Cluster B personality disorders, 116 with IED, and 34 with posttraumatic stress disorder were assigned to divalproex sodium or placebo for 12 weeks. Using an intent-to-treat analysis, divalproex had no significant influence on aggression in patients with IED.26 Similarly, a study assessing levetiracetam for IED did not show any improvements to measures of impulsive aggression.27

Psychological treatments. The only available study on psychological treatments for IED found that patients receiving active cognitive-behavioral therapy (CBT) or group therapy showed significant improvements compared with waitlist controls. These improvements spanned several target symptoms of IED.28

Conclusions. Although there is a paucity of treatment studies for IED, fluoxetine may be an effective treatment based on available studies, and oxcarbazepine has shown some preliminary efficacy. CBT also has shown some initial efficacy in reducing symptom severity in IED.


Conduct disorder
The essential feature of CD is a repetitive and persistent pattern of behavior in which the basic rights of others or social norms are vio­lated.3 These behaviors can entail:
   • aggressive conduct that causes or threatens harm to others or to animals
   • nonaggressive behavior resulting in property damage
   • deceitfulness or theft
   • serious violation of rules.

Prevalence among the general population is 2% to 10%. The disorder is more common among boys than girls.3

Pharmacotherapy. No medication is FDA-approved to treat CD. Fifteen con­trolled studies have examined medica­tions in patients with CD (Table), although a number of these included a high rate of comorbid ADHD.

To date, 7 studies have shown efficacy with lithium for patients with CD.29-35 A number of trials assessing lithium also included a treatment condition with halo­peridol, which showed significant improve­ment.29,30,33,34 Both lithium and haloperidol were associated with select deficits on cog­nitive tests, suggesting that there may be risks associated with these medications.

Preliminary double-blind results have indicated that methylphenidate, risperi­done, quetiapine, molindone, thioridazine, and carbamazepine might be effective options for treating CD.36-43 The evidence for these medications is limited and addi­tional studies are needed to replicate initial findings.

Three studies of divalproex sodium have shown some efficacy in randomized stud­ies comparing high and low dosages of the drug.40-42 Because these studies did not include a placebo, additional studies are necessary to corroborate these findings.

Psychological treatments. Several forms of behavioral, family-based, and school-based therapies have been found effective in randomized trials. Specifically, behavioral therapy and parental skills training have shown consistent benefits for patients and their families. As with ODD, parental train­ing programs for CD focus on parents’ skill acquisition to help manage outbursts and aggressive behavior. These treatments often follow a similar course to those used for other externalizing and disruptive disorders.44-46

Conclusions. Based on evidence, psychother­apy and some pharmacotherapies (eg, lith­ium) could be considered first-line treatment options for CD. Psychotherapy programs have shown efficacy in reducing aggression in high-risk groups.44 Lithium or antipsychot­ics could be useful for patients who do not respond sufficiently to psychotherapy. The risk of cognitive deficits with lithium and antipsychotics should be weighed against potential benefits of these medications.33,34


Kleptomania
Kleptomania is characterized by repetitive, poorly controlled stealing of items that are not needed for personal use. Kleptomania often begins in late adolescence or early adulthood.47 The course of the illness gen­erally is chronic, with waxing and waning symptoms. Women are twice as likely as men to suffer from kleptomania.48 People with kleptomania frequently hoard, discard, or return stolen items.47

 

 

Most people with kleptomania try unsuc­cessfully to stop stealing, which often leads to feelings of shame and guilt.48 Many (64% to 87%) have been arrested because of their stealing behavior47; a smaller percentage (15% to 23%) have been incarcerated.48 Suicide attempts are common among these patients.49

Pharmacotherapy. There has been only 1 randomized, placebo-controlled study of pharmacotherapy for kleptomania (Table). An 8-week, double-blind, placebo-controlled trial was conducted to evaluate the safety and efficacy of oral naltrexone, 50 to 150 mg/d, in 25 patients with kleptomania. Those taking naltrexone had a significantly greater reduc­tion in total score than those taking placebo on the Yale-Brown Obsessive Compulsive Scale Modified for Kleptomania; in stealing urges; and in stealing behavior. The mean effective dosage of naltrexone was 116.7 (± 44.4) mg/d.50

Naltrexone was well tolerated, with mini­mal nausea, and did not cause elevation of liver enzymes.

There is one available open-label study with a double-blind discontinuation phase assessing the efficacy of escitalopram for kleptomania. Continuation of escitalopram during the blinded discontinuation phase did produce lower relapse rates.51

Psychological treatments. There are no con­trolled studies of psychological treatments for kleptomania. Case reports suggest that cognitive and behavioral therapies might be effective:
   • A young man who underwent 7 ses­sions of covert sensitization, combined with exposure and response prevention, over a 4-month period was able to reduce his steal­ing frequency.52
   • In another case, a young woman underwent 5 weekly sessions when she was instructed to practice covert sensitiza­tion whenever she had an urge to steal. She remained in remission for 14 months with only a single lapse in behavior and with no reported urges to steal.53
  
• In 2 patients, imaginal desensitization in fourteen 15-minutes sessions over 5 days resulted in complete remission of symptoms for a 2-year period.54

Conclusions. The single controlled study of naltrexone for kleptomania suggests that naltrexone might be a beneficial treatment for this disorder. No controlled trials of psy­chosocial interventions have been reported. The current psychological research is based primarily on case reports.

This state of affairs likely is because of (1) the low prevalence of kleptomania and (2) clinical difficulties in treating patients involved in illegal activities. Nevertheless, there is a need for systematic studies of treat­ing this disorder; such studies could involve collaboration across multiple treatment cen­ters because of the disorder’s low prevalence.


Pyromania
Pyromania is characterized by (1) deliberate and purposeful fire setting on >1 occasion; (2) tension or affective arousal before the act; (3) fascination with, interest in, curiosity about, or attraction to fire and its situational con­texts; and (4) pleasure, gratification, or relief when setting fires or when witnessing or par­ticipating in their aftermath.3

Although pyromania is thought to be a disorder primarily affecting men, recent research suggests that the sex ratio is equal among adults and may be slightly higher among adolescent females. Mean age of onset usually is late adoles­cence. Pyromania appears to be chronic if untreated.55

Urges to set fires are common and the fire setting is almost always pleasurable. Severe distress follows the fire setting, and persons with pyromania report significant functional impairment. High rates of co-occurring psy­chiatric disorders (depression, substance use disorders, other impulse-control dis­orders) are common among persons with pyromania.55

Pharmacotherapy. There are no random­ized, controlled clinical trials examining pharmacotherapy for treating pyromania. There are no FDA-approved medications for pyromania.

In case reports, medications that have shown benefit in treating pyromania include topiramate, escitalopram, sertraline, fluox­etine, lithium, and a combination of olan­zapine and sodium valproate. An equal number of medications have shown no ben­efit: fluoxetine, valproic acid, lithium, sertra­line, olanzapine, escitalopram, citalopram, and clonazepam. A case report of an 18-year-old man with pyromania described success­fully using a combination of topiramate with 3 weeks of daily CBT to achieve significant symptom improvement.56,57

Pyromania is a largely unrecognized dis­order that causes significant psychological, social, and legal repercussions. Because few persons with pyromania volunteer informa­tion regarding fire-setting, it is important that clinicians recognize the disorder and screen patients appropriately. Various treatments have been helpful in case studies, but more research on the etiology and treatment of the disorder is needed.56,57


Conclusions based on the literature
In disruptive, impulse-control, and conduct disorders, the systematic study of treatment efficacy and tolerability is in its infancy. With few controlled studies published, it is not possible to make treatment recommendations with confidence. There are no FDA-approved drugs for treating any of these disorders.

Nonetheless, specific psychotherapies and drug therapies offer promising options, but often are based on small studies, often in patient populations with prominent comor­bidities, and have not been replicated by independent investigators. For all of these disorders, issues such as which psycho­therapy or medication to use and the ideal duration of treatment cannot be sufficiently addressed with the available data.

 

 

In conjunction with emerging epidemio­logical data supporting a relatively high prevalence of disruptive, impulse-control, and conduct disorders, the small amount of data regarding effective treatments highlights the clinical need for additional research.


Bottom Line
Empirically supported treatment options for impulse-control disorders currently are limited, because only select disorders have been studied across multiple trials. New research is needed to confirm possible treatment options and identify effective psychotherapeutic and pharmacological treatment alternatives.
 

Related Resources
• Grant JE. Impulse control disorders: a clinician’s guide to un­derstanding and treating behavioral addictions. New York, NY: W. W. Norton & Company; 2008.
• Grant JE, Kim SW. Stop me because I can’t stop myself: tak­ing control of impulsive behavior. New York, NY: McGraw- Hill; 2003.
• American Academy of Child and Adolescent Psychiatry. Conduct disorder resource center. http://www.aacap.org/AACAP/FamiliesandYouth/ResourceCenters/ConductDisorderResourceCenter/Home.aspx.


Drug Brand Names
Atomoxetine • Strattera                      Methylphenidate • Ritalin
Carbamazepine • Tegretol                  Molindone • Moban
Citalopram • Celexa                            Naltrexone • ReVia
Clonazepam • Klonopin                      Olanzapine • Zyprexa
Clonidine • Catapres                           Oxcarbazepine • Trileptal
D-amphetamine • Dexedrine               Quetiapine • Seroquel
Divalproex sodium • Depakote            Risperidone • Risperdal
Escitalopram • Lexapro                       Sertraline • Zoloft
Fluoxetine • Prozac                             Sodium valproate • Depacon
Guanfacine • Intuniv                           Thioridazine • Mellaril
Haloperidol • Haldol                             Topiramate • Topamax
Levetiracetam • Keppra                       Valproic acid • Depakote
Lithium • Eskalith, Lithobid  

 

Disclosures
Dr. Grant receives grant or research support from Brainsway, Forest Pharmaceuticals, and Roche Pharmaceuticals. Mr. Leppink reports no financial relationship with any company whose products are mentioned in this article or with competing products.

References


1. Kessler RC, Berglund P, Demler O, et al. Lifetime prevalence and age-of-onset distributions of DSM-IV disorders in the National Comorbidity Survey Replication. Arch Gen Psychiatry. 2005;62(6):593-602.
2. Grant JE, Levine L, Kim D, et al. Impulse control disorders in adult psychiatric inpatients. Am J Psychiatry. 2005;162(11):2184-2188.
3. Diagnostic and statistical manual of mental disorders, 5th ed. Washington, DC: American Psychiatric Association; 2013.
4. Turgay A. Psychopharmacological treatment of oppositional defiant disorder. CNS Drugs. 2009;23(1):1-17.
5. Hazell P. Review of attention-deficit/hyperactivity disorder comorbid with oppositional defiant disorder. Australas Psychiatry. 2010;18(6):556-559.
6. Burke JD, Loeber R, Birmaher B. Oppositional defiant disorder and conduct disorder: a review of the past 10 years, part II. J Am Acad Child Adolesc Psychiatry. 2002; 41(11):1275-1293.
7. Connor DF, Steeber J, McBurnett K. A review of attention-deficit/hyperactivity disorder complicated by symptoms of oppositional defiant disorder or conduct disorder. J Dev Behav Pediatr. 2010;31(5):427-440.
8. Aman MG, Bukstein OG, Gadow KD, et al. What does risperidone add to parent training and stimulant for severe aggression in child attention-deficit/hyperactivity disorder? J Am Acad Child Adolesc Psychiatry. 2014;53(1):47-60.e1.
9. Loy JH, Merry SN, Hetrick SE, et al. Atypical antipsychotics for disruptive behavior disorders in children and youths. Cochrane Database Syst Rev. 2012;9:CD008559.
10. Gadow KD, Arnold LE, Molina BS, et al. Risperidone added to parent training and stimulant medication: effects on attention-deficit/hyperactivity disorder, oppositional defiant disorder, conduct disorder, and peer aggression. J Am Acad Child Adolesc Psychiatry. 2014;53(9):948-959.e1.
12. Signorovitch J, Erder MH, Xie J, et al. Comparative effectiveness research using matching-adjusted indirect comparison: an application to treatment with guanfacine extended release or atomoxetine in children with attention-deficit/hyperactivity disorder and comorbid oppositional defiant disorder. Pharmacoepidemiol Drug Saf. 2012;21(suppl 2):130-137.
13. Bangs ME, Hazell P, Danckaerts M, et al; Atomoxetine ADHD/ODD Study Group. Atomoxetine for the treatment of attention-deficit/hyperactivity disorder and oppositional defiant disorder. Pediatrics. 2008;121(2):e314-e320.
14. Biederman J, Spencer TJ, Newcorn JH, et al. Effect of comorbid symptoms of oppositional defiant disorder on responses to atomoxetine in children with ADHD: a meta-analysis of controlled clinical trial data. Psychopharmacology (Berl). 2007;190(1):31-41.
15. Miller NV, Haas SM, Waschbusch DA, et al. Behavior therapy and callous-unemotional traits: effects of a pilot study examining modified behavioral contingencies on child behavior. Behav Ther. 2014;45(5):606-618.
16. Hamilton SS, Armando J. Oppositional defiant disorder. Am Fam Physician. 2008;78(7):861-866.
17. Steiner H, Remsing L; Work Group on Quality Issues. Practice parameter for the assessment and treatment of children and adolescents with oppositional defiant disorder. J Am Acad Child Adolesc Psychiatry. 2007;46(1):126-141.
18. Winther J, Carlsson A, Vance A. A pilot study of a school-based prevention and early intervention program to reduce oppositional defiant disorder/conduct disorder. Early Interv Psychiatry. 2014;8(2):181-189.
19. Plueck J, Eichelberger I, Hautmann C, et al. Effectiveness of a teacher-based indicated prevention program for preschool children with externalizing problem behavior [published online April 22, 2014]. Prev Sci. doi: 10.1007/s11121-014- 0487-x.
20. Dretzke J, Frew E, Davenport C, et al. The effectiveness and cost-effectiveness of parent training/education programmes for the treatment of conduct disorder, including oppositional defiant disorder, in children. Health Tech Assess. 2005;9(50):iii, ix-x, 1-233.
21. Coccaro EF, Schmidt CA, Samuels JF, et al. Lifetime and 1-month prevalence rates of intermittent explosive disorder in a community sample. J Clin Psychiatry. 2004;65(6):820-824.
22. McElroy SL, Soutullo CA, Beckman DA, et al. DSM-IV intermittent explosive disorder: a report of 27 cases. J Clin Psychiatry. 1998;59(4):203-210; quiz 211.
23. Coccaro EF, Lee RJ, Kavoussi RJ. A double-blind, randomized, placebo-controlled trial of fluoxetine in patients with intermittent explosive disorder. J Clin Psychiatry. 2009;70(5):653-662.
24. Coccaro EF. Intermittent explosive disorder as a disorder of impulsive aggression for DSM-5. Am J Psychiatry. 2012;169(6):577-588.
25. Mattes JA. Oxcarbazepine in patients with impulsive aggression: a double-blind, placebo-controlled trial. J Clin Psychopharmacol. 2005;25(6):575-579.
26. Hollander E, Tracy KA, Swann AC, et al. Divalproex in the treatment of impulsive aggression: efficacy in cluster B personality disorders. Neuropsychopharmacology. 2003;28(6):1186-1197.
27. Mattes JA. Levetiracetam in patients with impulsive aggression: a double-blind, placebo-controlled trial. J Clin Psychiatry. 2008;69(2):310-315.
28. McCloskey MS, Noblett KL, Deffenbacher JL, et al. Cognitive-behavioral therapy for intermittent explosive disorder: a pilot randomized clinical trial. J Consult Clin Psychol. 2008;76(5):876-886.
29. Campbell M, Small AM, Green WH, et al. Behavioral efficacy of haloperidol and lithium carbonate. A comparison in hospitalized aggressive children with conduct disorder. Arch Gen Psychiatry. 1984;41(7):650-656.
30. Campbell M, Adams PB, Small AM, et al. Lithium in hospitalized aggressive children with conduct disorder: a double-blind and placebo-controlled study. J Am Acad Child Adolesc Psychiatry. 1995;34(4):445-453.
31. Malone RP, Simpson GM. Psychopharmacology: use of placebos in clinical trials involving children and adolescents. Psychiatr Serv. 1998;49(11):1413-1414, 1417.
32. Malone RP, Delaney MA, Luebbert JF, et al. A double-blind placebo-controlled study of lithium in hospitalized aggressive children and adolescents with conduct disorder. Arch Gen Psychiatry. 2000;57(7):649-654.
33. Platt JE, Campbell M, Green WH, et al. Effects of lithium carbonate and haloperidol on cognition in aggressive hospitalized school-age children. J Clin Psychopharmacol. 1981;1(1):8-13.
34. Platt JE, Campbell M, Green WH, et al. Cognitive effects of lithium carbonate and haloperidol in treatment-resistant aggressive children. Arch Gen Psychiatry. 1984;41(7):657-662.
35. Rifkin A, Karajgi B, Dicker R, et al. Lithium treatment of conduct disorders in adolescents. Am J Psychiatry. 1997;154(4):554-555.
36. Cueva JE, Overall JE, Small AM, et al. Carbamazepine in aggressive children with conduct disorder: a double-blind and placebo-controlled study. J Am Acad Child Adolesc Psychiatry. 1996;35(4):480-490.
37. Findling RL, McNamara NK, Branicky LA, et al. A double-blind pilot study of risperidone in the treatment of conduct disorder. J Am Acad Child Adolesc Psychiatry. 2000;39(4):509-516.
38. Connor DF, McLaughlin TJ, Jeffers-Terry M. Randomized controlled pilot study of quetiapine in the treatment of adolescent conduct disorder. J Child Adolesc Psychopharmacol. 2008;18(2):140-156.
39. Greenhill LL, Solomon M, Pleak R, et al. Molindone hydrochloride treatment of hospitalized children with conduct disorder. J Clin Psychiatry. 1985;46(8 pt 2):20-25.
40. Khanzode LA, Saxena K, Kraemer H, et al. Efficacy profiles of psychopharmacology: divalproex sodium in conduct disorder. Child Psychiatry Hum Dev. 2006;37(1):55-64.
41. Padhy R, Saxena K, Remsing L, et al. Symptomatic response to divalproex in subtypes of conduct disorder. Child Psychiatry Hum Dev. 2011;42(5):584-593.
42. Steiner H, Petersen ML, Saxena K, et al. Divalproex sodium for the treatment of conduct disorder: a randomized controlled clinical trial. J Clin Psychiatry. 2003;64(10):1183-1191.
43. Klein RG, Abikoff H, Klass E, et al. Clinical efficacy of methylphenidate in conduct disorder with and without attention deficit hyperactivity disorder. Arch Gen Psychiatry. 1997;54(12):1073-1080.
44. Heneggeler SW, Sheidow AJ. Empirically supported family-based treatments for conduct disorder and delinquency in adolescents. J Marital Fam Ther. 2012;38(1):30-58.
45. Lochman JE, Powell NP, Boxmeyer CL, et al. Cognitive-behavioral therapy for externalizing disorder in children and adolescents. Child Adolesc Psychiatr Clin N Am. 2011;20(2):305-318.
46. Furlong M, McGilloway S, Bywater T, et al. Behavioural and cognitive-behavioural group-based parenting programmes for early-onset conduct problems in children aged 3 to 12 years. Cochrane Database Syst Rev. 2012;2:CD008225.
47. McElroy SL, Pope HG Jr, Hudson JI, et al. Kleptomania: a report of 20 cases. Am J Psychiatry. 1991;148(5):652-657.
48. Grant JE, Kim SW. Clinical characteristics and associated psychopathology of 22 patients with kleptomania. Compr Psychiatry. 2002;43(5):378-384.
49. Odlaug BL, Grant JE, Kim SW. Suicide attempts in 107 adolescents and adults with kleptomania. Arch Suicide Res. 2012;16(4):348-359.
50. Grant JE, Kim SW, Odlaug BL. A double-blind, placebo-controlled study of the opiate antagonist, naltrexone, in the treatment of kleptomania. Biol Psychiatry. 2009;65(7): 600-606.
51. Koran LM, Aboujaoude EN, Gamel NN. Escitalopram treatment of kleptomania: an open-label trial followed by double-blind discontinuation. J Clin Psychiatry. 2007;68(3):422-427.
52. Guidry LS. Use of a covert punishing contingency in compulsive stealing. J Behav Therapy Exp Psychiatry. 1975;6(2):169.
53. Gauthier J, Pellerin D. Management of compulsive shoplifting through covert sensitization. J Behav Therapy Exp Psychiatry. 1982;13(1):73-75.
54. McConaghy N, Blaszczynski A. Imaginal desensitization: a cost-effective treatment in two shop-lifters and a binge-eater resistant to previous therapy. Aus N Z J Psychiatry. 1988;22(1):78-82.
55. Grant JE, Won Kim S. Clinical characteristics and psychiatric comorbidity of pyromania. J Clin Psychiatry. 2007;68(11):1717-1722.
56. Grant JE, Odlaug B. Assessment and treatment of pyromania. In: Oxford handbook of impulse control disorders. Grant JE, Potenza MN, eds. Oxford, United Kingdom: Oxford University Press; 2012:353-359.
57. Dell’Osso B, Altamura AC, Allen A, et al. Epidemiologic and clinical updates on impulse control disorders: a critical review. Eur Arch Psychiatry Clin Neurosci. 2006;256(8):464-475.

Article PDF
Author and Disclosure Information

 

Jon E. Grant, JD, MD, MPH
Professor
Department of Psychiatry & Behavioral Neuroscience
University of Chicago, Pritzker School of Medicine
Chicago, Illinois


Eric W. Leppink, BA
Research Specialist
University of Chicago Hospital
Department of Psychiatry & Behavioral Neuroscience
Chicago, Illinois

Issue
Current Psychiatry - 14(1)
Publications
Topics
Page Number
29-36
Legacy Keywords
impulse-control disorders, conduct disorders, impulse control, disruptive disorders, kleptomania, oppositional disorder, intermittent explosive disorder, ADHD, attention deficit/hyperactivity disorder, pyromania
Sections
Author and Disclosure Information

 

Jon E. Grant, JD, MD, MPH
Professor
Department of Psychiatry & Behavioral Neuroscience
University of Chicago, Pritzker School of Medicine
Chicago, Illinois


Eric W. Leppink, BA
Research Specialist
University of Chicago Hospital
Department of Psychiatry & Behavioral Neuroscience
Chicago, Illinois

Author and Disclosure Information

 

Jon E. Grant, JD, MD, MPH
Professor
Department of Psychiatry & Behavioral Neuroscience
University of Chicago, Pritzker School of Medicine
Chicago, Illinois


Eric W. Leppink, BA
Research Specialist
University of Chicago Hospital
Department of Psychiatry & Behavioral Neuroscience
Chicago, Illinois

Article PDF
Article PDF

Chronic  disruptive and impulsive behaviors are significant concerns for psychiatric clinicians because of their persistence and potential legal ramifications. To date, few studies have assessed treatment options for pyromania, oppositional defiant disorder (ODD), intermittent explosive disorder (IED), kleptomania, and conduct disorder (CD).

This article reviews the literature on the treatment of these disorders, focusing primarily on randomized, controlled studies. Because of the lack of clinical studies for these disorders, however, case studies and open tri­als are mentioned for reference. Summaries of supported medication and psychological interventions are provided for each disorder.


Categorizing impulse-control disorders
The DSM-5 created a new chapter on disruptive, impulse control, and conduct disorders that brought together disorders previously classified as disorders usually first diagnosed in infancy, childhood, or adolescence (ODD, CD) and impulse-control disorders not elsewhere classified. These disorders are unified by the presence of difficult, disruptive, aggressive, or antisocial behavior. Disruptive, aggressive, or antisocial behavior usu­ally is a multifaceted behavior, often associated with physical or verbal injury to self, others, or objects or with violating the rights of others. These behaviors can appear in several forms and can be defensive, premedi­tated, or impulsive.

Despite a high prevalence in the general population1 and in psychi­atric cohorts,2 disruptive and impulse-control disorders have been rela­tively understudied. Controlled trials of treatments do not exist for many impulse-control disorders, and there are no FDA-approved medications for any of these disorders.
 

Oppositional defiant disorder
Irritability, anger, defiance, and temper are specific descriptors of ODD. ODD seems to be a developmental antecedent for some youth with CD, suggesting that these dis­orders could reflect different stages of a spectrum of disruptive behavior. Transient oppositional behavior is common among children and adolescents, but ODD occurs in 1% to 11% of youth.3 The disorder is more prevalent among boys before puberty and has an equal sex prevalence in young people after puberty.

Regrettably, most ODD research has included patients with comorbidities, most commonly attention-deficit/hyperactivity disorder (ADHD). Because of this limita­tion, the drugs and programs discussed below are drawn from meta-analyses and review articles.

Pharmacotherapy. No medications have been FDA-approved for ODD. Studies assess­ing ODD have employed a variety of meth­odologies, not all of which are double-blind. The meta-analyses and reviews cited in this section include both randomized and open trials, and should be interpreted as such.

Stimulants are commonly used to treat ODD because of a high comorbidity rate with ADHD, and these drugs have improved ODD symptoms in randomized trials.4 Methylphenidate and d-amphetamine have shown some efficacy in trials of ODD and CD.5-7 These medications are most commonly used when ODD is complicated by ADHD symptoms.

Antipsychotics also have been used to treat ODD, with the largest body of research suggesting that risperidone has some effi­cacy. Risperidone usually is considered a second- or third-line option because it has been associated with adverse effects in chil­dren and adolescents and requires caution in younger populations, despite its potential efficacy.4,8-10

Alpha-2 agonists—clonidine and guanfa­cine—have shown some efficacy in treating ODD but have not been studied extensively. Studies of clonidine, however, often have grouped ODD, CD, and ADHD, which lim­its our understanding of this medication for ODD alone.4,5,11

Atomoxetine has been studied for ODD, but its efficacy is limited, with different meta-analyses finding distinct results regarding efficacy. One explanation for these dispa­rate findings is that improvements in oppo­sitional symptoms may be secondary to improvement in ADHD symptoms.7,12-14

Psychological treatments. As noted for pharmacotherapy, this section provides gen­eral information on empirically studied ther­apies. A series of meta-analyses have been included for further review, but are not iso­lated to randomized, controlled studies.

Individual therapy has shown consistent improvements in ODD. Examples include behavior modification therapy and parent-child interaction therapy. These sessions emphasize skills to manage outbursts and erratic emotionality. Emotion regulation and behavior and social skills training have shown significant reductions in target mea­sures. Some of these programs incorporate both patient and parent components.15-17

Family/teacher training programs such as “Helping the Noncompliant Child” and the “Triple P” have yielded significant improve­ments. These programs focus on ways to manage the child’s oppositional behavior at home and in the classroom, as well as strate­gies to limit positive reinforcement for prob­lem behaviors.17-20

Group programs have shown some effi­cacy with ODD. These programs cover a wide number of needs and intents. Examples include the “Incredible Years” program and the Community Parent Education Program. Research has found that these programs show some efficacy as preemptive measures to reduce the rate of ODD among adolescents.

Conclusions. A number of treatment options for ODD have shown some efficacy. However, many of these options have only been studied in patients with comorbid ADHD, which limits current knowledge about ODD as a distinct disorder.

 

 


Intermittent explosive disorder
IED is defined by recurrent, significant out­bursts of aggression, often leading to assaul­tive acts against people or property, which are disproportionate to outside stressors and are not better explained by another psy­chiatric diagnosis. Research suggests IED is common, with 6.3% of a community sample meeting criteria for lifetime IED.21

IED symptoms tend to start in adolescence and appear to be chronic.21,22 People with IED regard their behavior as distressing and prob­lematic.22 Outbursts generally are short-lived (usually <30 minutes) and frequent (multiple times a month22). Legal and occupational dif­ficulties are common.22

Pharmacotherapy. Data on drug treatment for IED comes for a small set of double-blind studies (Table). Although pharmacotherapies have been studied for treating aggression, impulsivity, and vio­lent behavior, only 5 controlled studies are specific to IED.


A double-blind, randomized, placebo-controlled trial of fluoxetine in 100 par­ticipants with IED found that fluoxetine produced a sustained reduction in aggression and irritability as early as the second week of treatment. Full or partial remission of impul­sive aggressive behaviors occurred in 46% of fluoxetine-treated subjects. These findings have been supported by studies assessing other samples of aggressive patients, but not specifically IED.23,24 Another treatment study found that oxcarbazepine produced signifi­cant improvements in IED symptom severity, specifically on impulsive aggression.25

In a randomized, double-blind, placebo-controlled study, 96 participants with Cluster B personality disorders, 116 with IED, and 34 with posttraumatic stress disorder were assigned to divalproex sodium or placebo for 12 weeks. Using an intent-to-treat analysis, divalproex had no significant influence on aggression in patients with IED.26 Similarly, a study assessing levetiracetam for IED did not show any improvements to measures of impulsive aggression.27

Psychological treatments. The only available study on psychological treatments for IED found that patients receiving active cognitive-behavioral therapy (CBT) or group therapy showed significant improvements compared with waitlist controls. These improvements spanned several target symptoms of IED.28

Conclusions. Although there is a paucity of treatment studies for IED, fluoxetine may be an effective treatment based on available studies, and oxcarbazepine has shown some preliminary efficacy. CBT also has shown some initial efficacy in reducing symptom severity in IED.


Conduct disorder
The essential feature of CD is a repetitive and persistent pattern of behavior in which the basic rights of others or social norms are vio­lated.3 These behaviors can entail:
   • aggressive conduct that causes or threatens harm to others or to animals
   • nonaggressive behavior resulting in property damage
   • deceitfulness or theft
   • serious violation of rules.

Prevalence among the general population is 2% to 10%. The disorder is more common among boys than girls.3

Pharmacotherapy. No medication is FDA-approved to treat CD. Fifteen con­trolled studies have examined medica­tions in patients with CD (Table), although a number of these included a high rate of comorbid ADHD.

To date, 7 studies have shown efficacy with lithium for patients with CD.29-35 A number of trials assessing lithium also included a treatment condition with halo­peridol, which showed significant improve­ment.29,30,33,34 Both lithium and haloperidol were associated with select deficits on cog­nitive tests, suggesting that there may be risks associated with these medications.

Preliminary double-blind results have indicated that methylphenidate, risperi­done, quetiapine, molindone, thioridazine, and carbamazepine might be effective options for treating CD.36-43 The evidence for these medications is limited and addi­tional studies are needed to replicate initial findings.

Three studies of divalproex sodium have shown some efficacy in randomized stud­ies comparing high and low dosages of the drug.40-42 Because these studies did not include a placebo, additional studies are necessary to corroborate these findings.

Psychological treatments. Several forms of behavioral, family-based, and school-based therapies have been found effective in randomized trials. Specifically, behavioral therapy and parental skills training have shown consistent benefits for patients and their families. As with ODD, parental train­ing programs for CD focus on parents’ skill acquisition to help manage outbursts and aggressive behavior. These treatments often follow a similar course to those used for other externalizing and disruptive disorders.44-46

Conclusions. Based on evidence, psychother­apy and some pharmacotherapies (eg, lith­ium) could be considered first-line treatment options for CD. Psychotherapy programs have shown efficacy in reducing aggression in high-risk groups.44 Lithium or antipsychot­ics could be useful for patients who do not respond sufficiently to psychotherapy. The risk of cognitive deficits with lithium and antipsychotics should be weighed against potential benefits of these medications.33,34


Kleptomania
Kleptomania is characterized by repetitive, poorly controlled stealing of items that are not needed for personal use. Kleptomania often begins in late adolescence or early adulthood.47 The course of the illness gen­erally is chronic, with waxing and waning symptoms. Women are twice as likely as men to suffer from kleptomania.48 People with kleptomania frequently hoard, discard, or return stolen items.47

 

 

Most people with kleptomania try unsuc­cessfully to stop stealing, which often leads to feelings of shame and guilt.48 Many (64% to 87%) have been arrested because of their stealing behavior47; a smaller percentage (15% to 23%) have been incarcerated.48 Suicide attempts are common among these patients.49

Pharmacotherapy. There has been only 1 randomized, placebo-controlled study of pharmacotherapy for kleptomania (Table). An 8-week, double-blind, placebo-controlled trial was conducted to evaluate the safety and efficacy of oral naltrexone, 50 to 150 mg/d, in 25 patients with kleptomania. Those taking naltrexone had a significantly greater reduc­tion in total score than those taking placebo on the Yale-Brown Obsessive Compulsive Scale Modified for Kleptomania; in stealing urges; and in stealing behavior. The mean effective dosage of naltrexone was 116.7 (± 44.4) mg/d.50

Naltrexone was well tolerated, with mini­mal nausea, and did not cause elevation of liver enzymes.

There is one available open-label study with a double-blind discontinuation phase assessing the efficacy of escitalopram for kleptomania. Continuation of escitalopram during the blinded discontinuation phase did produce lower relapse rates.51

Psychological treatments. There are no con­trolled studies of psychological treatments for kleptomania. Case reports suggest that cognitive and behavioral therapies might be effective:
   • A young man who underwent 7 ses­sions of covert sensitization, combined with exposure and response prevention, over a 4-month period was able to reduce his steal­ing frequency.52
   • In another case, a young woman underwent 5 weekly sessions when she was instructed to practice covert sensitiza­tion whenever she had an urge to steal. She remained in remission for 14 months with only a single lapse in behavior and with no reported urges to steal.53
  
• In 2 patients, imaginal desensitization in fourteen 15-minutes sessions over 5 days resulted in complete remission of symptoms for a 2-year period.54

Conclusions. The single controlled study of naltrexone for kleptomania suggests that naltrexone might be a beneficial treatment for this disorder. No controlled trials of psy­chosocial interventions have been reported. The current psychological research is based primarily on case reports.

This state of affairs likely is because of (1) the low prevalence of kleptomania and (2) clinical difficulties in treating patients involved in illegal activities. Nevertheless, there is a need for systematic studies of treat­ing this disorder; such studies could involve collaboration across multiple treatment cen­ters because of the disorder’s low prevalence.


Pyromania
Pyromania is characterized by (1) deliberate and purposeful fire setting on >1 occasion; (2) tension or affective arousal before the act; (3) fascination with, interest in, curiosity about, or attraction to fire and its situational con­texts; and (4) pleasure, gratification, or relief when setting fires or when witnessing or par­ticipating in their aftermath.3

Although pyromania is thought to be a disorder primarily affecting men, recent research suggests that the sex ratio is equal among adults and may be slightly higher among adolescent females. Mean age of onset usually is late adoles­cence. Pyromania appears to be chronic if untreated.55

Urges to set fires are common and the fire setting is almost always pleasurable. Severe distress follows the fire setting, and persons with pyromania report significant functional impairment. High rates of co-occurring psy­chiatric disorders (depression, substance use disorders, other impulse-control dis­orders) are common among persons with pyromania.55

Pharmacotherapy. There are no random­ized, controlled clinical trials examining pharmacotherapy for treating pyromania. There are no FDA-approved medications for pyromania.

In case reports, medications that have shown benefit in treating pyromania include topiramate, escitalopram, sertraline, fluox­etine, lithium, and a combination of olan­zapine and sodium valproate. An equal number of medications have shown no ben­efit: fluoxetine, valproic acid, lithium, sertra­line, olanzapine, escitalopram, citalopram, and clonazepam. A case report of an 18-year-old man with pyromania described success­fully using a combination of topiramate with 3 weeks of daily CBT to achieve significant symptom improvement.56,57

Pyromania is a largely unrecognized dis­order that causes significant psychological, social, and legal repercussions. Because few persons with pyromania volunteer informa­tion regarding fire-setting, it is important that clinicians recognize the disorder and screen patients appropriately. Various treatments have been helpful in case studies, but more research on the etiology and treatment of the disorder is needed.56,57


Conclusions based on the literature
In disruptive, impulse-control, and conduct disorders, the systematic study of treatment efficacy and tolerability is in its infancy. With few controlled studies published, it is not possible to make treatment recommendations with confidence. There are no FDA-approved drugs for treating any of these disorders.

Nonetheless, specific psychotherapies and drug therapies offer promising options, but often are based on small studies, often in patient populations with prominent comor­bidities, and have not been replicated by independent investigators. For all of these disorders, issues such as which psycho­therapy or medication to use and the ideal duration of treatment cannot be sufficiently addressed with the available data.

 

 

In conjunction with emerging epidemio­logical data supporting a relatively high prevalence of disruptive, impulse-control, and conduct disorders, the small amount of data regarding effective treatments highlights the clinical need for additional research.


Bottom Line
Empirically supported treatment options for impulse-control disorders currently are limited, because only select disorders have been studied across multiple trials. New research is needed to confirm possible treatment options and identify effective psychotherapeutic and pharmacological treatment alternatives.
 

Related Resources
• Grant JE. Impulse control disorders: a clinician’s guide to un­derstanding and treating behavioral addictions. New York, NY: W. W. Norton & Company; 2008.
• Grant JE, Kim SW. Stop me because I can’t stop myself: tak­ing control of impulsive behavior. New York, NY: McGraw- Hill; 2003.
• American Academy of Child and Adolescent Psychiatry. Conduct disorder resource center. http://www.aacap.org/AACAP/FamiliesandYouth/ResourceCenters/ConductDisorderResourceCenter/Home.aspx.


Drug Brand Names
Atomoxetine • Strattera                      Methylphenidate • Ritalin
Carbamazepine • Tegretol                  Molindone • Moban
Citalopram • Celexa                            Naltrexone • ReVia
Clonazepam • Klonopin                      Olanzapine • Zyprexa
Clonidine • Catapres                           Oxcarbazepine • Trileptal
D-amphetamine • Dexedrine               Quetiapine • Seroquel
Divalproex sodium • Depakote            Risperidone • Risperdal
Escitalopram • Lexapro                       Sertraline • Zoloft
Fluoxetine • Prozac                             Sodium valproate • Depacon
Guanfacine • Intuniv                           Thioridazine • Mellaril
Haloperidol • Haldol                             Topiramate • Topamax
Levetiracetam • Keppra                       Valproic acid • Depakote
Lithium • Eskalith, Lithobid  

 

Disclosures
Dr. Grant receives grant or research support from Brainsway, Forest Pharmaceuticals, and Roche Pharmaceuticals. Mr. Leppink reports no financial relationship with any company whose products are mentioned in this article or with competing products.

Chronic  disruptive and impulsive behaviors are significant concerns for psychiatric clinicians because of their persistence and potential legal ramifications. To date, few studies have assessed treatment options for pyromania, oppositional defiant disorder (ODD), intermittent explosive disorder (IED), kleptomania, and conduct disorder (CD).

This article reviews the literature on the treatment of these disorders, focusing primarily on randomized, controlled studies. Because of the lack of clinical studies for these disorders, however, case studies and open tri­als are mentioned for reference. Summaries of supported medication and psychological interventions are provided for each disorder.


Categorizing impulse-control disorders
The DSM-5 created a new chapter on disruptive, impulse control, and conduct disorders that brought together disorders previously classified as disorders usually first diagnosed in infancy, childhood, or adolescence (ODD, CD) and impulse-control disorders not elsewhere classified. These disorders are unified by the presence of difficult, disruptive, aggressive, or antisocial behavior. Disruptive, aggressive, or antisocial behavior usu­ally is a multifaceted behavior, often associated with physical or verbal injury to self, others, or objects or with violating the rights of others. These behaviors can appear in several forms and can be defensive, premedi­tated, or impulsive.

Despite a high prevalence in the general population1 and in psychi­atric cohorts,2 disruptive and impulse-control disorders have been rela­tively understudied. Controlled trials of treatments do not exist for many impulse-control disorders, and there are no FDA-approved medications for any of these disorders.
 

Oppositional defiant disorder
Irritability, anger, defiance, and temper are specific descriptors of ODD. ODD seems to be a developmental antecedent for some youth with CD, suggesting that these dis­orders could reflect different stages of a spectrum of disruptive behavior. Transient oppositional behavior is common among children and adolescents, but ODD occurs in 1% to 11% of youth.3 The disorder is more prevalent among boys before puberty and has an equal sex prevalence in young people after puberty.

Regrettably, most ODD research has included patients with comorbidities, most commonly attention-deficit/hyperactivity disorder (ADHD). Because of this limita­tion, the drugs and programs discussed below are drawn from meta-analyses and review articles.

Pharmacotherapy. No medications have been FDA-approved for ODD. Studies assess­ing ODD have employed a variety of meth­odologies, not all of which are double-blind. The meta-analyses and reviews cited in this section include both randomized and open trials, and should be interpreted as such.

Stimulants are commonly used to treat ODD because of a high comorbidity rate with ADHD, and these drugs have improved ODD symptoms in randomized trials.4 Methylphenidate and d-amphetamine have shown some efficacy in trials of ODD and CD.5-7 These medications are most commonly used when ODD is complicated by ADHD symptoms.

Antipsychotics also have been used to treat ODD, with the largest body of research suggesting that risperidone has some effi­cacy. Risperidone usually is considered a second- or third-line option because it has been associated with adverse effects in chil­dren and adolescents and requires caution in younger populations, despite its potential efficacy.4,8-10

Alpha-2 agonists—clonidine and guanfa­cine—have shown some efficacy in treating ODD but have not been studied extensively. Studies of clonidine, however, often have grouped ODD, CD, and ADHD, which lim­its our understanding of this medication for ODD alone.4,5,11

Atomoxetine has been studied for ODD, but its efficacy is limited, with different meta-analyses finding distinct results regarding efficacy. One explanation for these dispa­rate findings is that improvements in oppo­sitional symptoms may be secondary to improvement in ADHD symptoms.7,12-14

Psychological treatments. As noted for pharmacotherapy, this section provides gen­eral information on empirically studied ther­apies. A series of meta-analyses have been included for further review, but are not iso­lated to randomized, controlled studies.

Individual therapy has shown consistent improvements in ODD. Examples include behavior modification therapy and parent-child interaction therapy. These sessions emphasize skills to manage outbursts and erratic emotionality. Emotion regulation and behavior and social skills training have shown significant reductions in target mea­sures. Some of these programs incorporate both patient and parent components.15-17

Family/teacher training programs such as “Helping the Noncompliant Child” and the “Triple P” have yielded significant improve­ments. These programs focus on ways to manage the child’s oppositional behavior at home and in the classroom, as well as strate­gies to limit positive reinforcement for prob­lem behaviors.17-20

Group programs have shown some effi­cacy with ODD. These programs cover a wide number of needs and intents. Examples include the “Incredible Years” program and the Community Parent Education Program. Research has found that these programs show some efficacy as preemptive measures to reduce the rate of ODD among adolescents.

Conclusions. A number of treatment options for ODD have shown some efficacy. However, many of these options have only been studied in patients with comorbid ADHD, which limits current knowledge about ODD as a distinct disorder.

 

 


Intermittent explosive disorder
IED is defined by recurrent, significant out­bursts of aggression, often leading to assaul­tive acts against people or property, which are disproportionate to outside stressors and are not better explained by another psy­chiatric diagnosis. Research suggests IED is common, with 6.3% of a community sample meeting criteria for lifetime IED.21

IED symptoms tend to start in adolescence and appear to be chronic.21,22 People with IED regard their behavior as distressing and prob­lematic.22 Outbursts generally are short-lived (usually <30 minutes) and frequent (multiple times a month22). Legal and occupational dif­ficulties are common.22

Pharmacotherapy. Data on drug treatment for IED comes for a small set of double-blind studies (Table). Although pharmacotherapies have been studied for treating aggression, impulsivity, and vio­lent behavior, only 5 controlled studies are specific to IED.


A double-blind, randomized, placebo-controlled trial of fluoxetine in 100 par­ticipants with IED found that fluoxetine produced a sustained reduction in aggression and irritability as early as the second week of treatment. Full or partial remission of impul­sive aggressive behaviors occurred in 46% of fluoxetine-treated subjects. These findings have been supported by studies assessing other samples of aggressive patients, but not specifically IED.23,24 Another treatment study found that oxcarbazepine produced signifi­cant improvements in IED symptom severity, specifically on impulsive aggression.25

In a randomized, double-blind, placebo-controlled study, 96 participants with Cluster B personality disorders, 116 with IED, and 34 with posttraumatic stress disorder were assigned to divalproex sodium or placebo for 12 weeks. Using an intent-to-treat analysis, divalproex had no significant influence on aggression in patients with IED.26 Similarly, a study assessing levetiracetam for IED did not show any improvements to measures of impulsive aggression.27

Psychological treatments. The only available study on psychological treatments for IED found that patients receiving active cognitive-behavioral therapy (CBT) or group therapy showed significant improvements compared with waitlist controls. These improvements spanned several target symptoms of IED.28

Conclusions. Although there is a paucity of treatment studies for IED, fluoxetine may be an effective treatment based on available studies, and oxcarbazepine has shown some preliminary efficacy. CBT also has shown some initial efficacy in reducing symptom severity in IED.


Conduct disorder
The essential feature of CD is a repetitive and persistent pattern of behavior in which the basic rights of others or social norms are vio­lated.3 These behaviors can entail:
   • aggressive conduct that causes or threatens harm to others or to animals
   • nonaggressive behavior resulting in property damage
   • deceitfulness or theft
   • serious violation of rules.

Prevalence among the general population is 2% to 10%. The disorder is more common among boys than girls.3

Pharmacotherapy. No medication is FDA-approved to treat CD. Fifteen con­trolled studies have examined medica­tions in patients with CD (Table), although a number of these included a high rate of comorbid ADHD.

To date, 7 studies have shown efficacy with lithium for patients with CD.29-35 A number of trials assessing lithium also included a treatment condition with halo­peridol, which showed significant improve­ment.29,30,33,34 Both lithium and haloperidol were associated with select deficits on cog­nitive tests, suggesting that there may be risks associated with these medications.

Preliminary double-blind results have indicated that methylphenidate, risperi­done, quetiapine, molindone, thioridazine, and carbamazepine might be effective options for treating CD.36-43 The evidence for these medications is limited and addi­tional studies are needed to replicate initial findings.

Three studies of divalproex sodium have shown some efficacy in randomized stud­ies comparing high and low dosages of the drug.40-42 Because these studies did not include a placebo, additional studies are necessary to corroborate these findings.

Psychological treatments. Several forms of behavioral, family-based, and school-based therapies have been found effective in randomized trials. Specifically, behavioral therapy and parental skills training have shown consistent benefits for patients and their families. As with ODD, parental train­ing programs for CD focus on parents’ skill acquisition to help manage outbursts and aggressive behavior. These treatments often follow a similar course to those used for other externalizing and disruptive disorders.44-46

Conclusions. Based on evidence, psychother­apy and some pharmacotherapies (eg, lith­ium) could be considered first-line treatment options for CD. Psychotherapy programs have shown efficacy in reducing aggression in high-risk groups.44 Lithium or antipsychot­ics could be useful for patients who do not respond sufficiently to psychotherapy. The risk of cognitive deficits with lithium and antipsychotics should be weighed against potential benefits of these medications.33,34


Kleptomania
Kleptomania is characterized by repetitive, poorly controlled stealing of items that are not needed for personal use. Kleptomania often begins in late adolescence or early adulthood.47 The course of the illness gen­erally is chronic, with waxing and waning symptoms. Women are twice as likely as men to suffer from kleptomania.48 People with kleptomania frequently hoard, discard, or return stolen items.47

 

 

Most people with kleptomania try unsuc­cessfully to stop stealing, which often leads to feelings of shame and guilt.48 Many (64% to 87%) have been arrested because of their stealing behavior47; a smaller percentage (15% to 23%) have been incarcerated.48 Suicide attempts are common among these patients.49

Pharmacotherapy. There has been only 1 randomized, placebo-controlled study of pharmacotherapy for kleptomania (Table). An 8-week, double-blind, placebo-controlled trial was conducted to evaluate the safety and efficacy of oral naltrexone, 50 to 150 mg/d, in 25 patients with kleptomania. Those taking naltrexone had a significantly greater reduc­tion in total score than those taking placebo on the Yale-Brown Obsessive Compulsive Scale Modified for Kleptomania; in stealing urges; and in stealing behavior. The mean effective dosage of naltrexone was 116.7 (± 44.4) mg/d.50

Naltrexone was well tolerated, with mini­mal nausea, and did not cause elevation of liver enzymes.

There is one available open-label study with a double-blind discontinuation phase assessing the efficacy of escitalopram for kleptomania. Continuation of escitalopram during the blinded discontinuation phase did produce lower relapse rates.51

Psychological treatments. There are no con­trolled studies of psychological treatments for kleptomania. Case reports suggest that cognitive and behavioral therapies might be effective:
   • A young man who underwent 7 ses­sions of covert sensitization, combined with exposure and response prevention, over a 4-month period was able to reduce his steal­ing frequency.52
   • In another case, a young woman underwent 5 weekly sessions when she was instructed to practice covert sensitiza­tion whenever she had an urge to steal. She remained in remission for 14 months with only a single lapse in behavior and with no reported urges to steal.53
  
• In 2 patients, imaginal desensitization in fourteen 15-minutes sessions over 5 days resulted in complete remission of symptoms for a 2-year period.54

Conclusions. The single controlled study of naltrexone for kleptomania suggests that naltrexone might be a beneficial treatment for this disorder. No controlled trials of psy­chosocial interventions have been reported. The current psychological research is based primarily on case reports.

This state of affairs likely is because of (1) the low prevalence of kleptomania and (2) clinical difficulties in treating patients involved in illegal activities. Nevertheless, there is a need for systematic studies of treat­ing this disorder; such studies could involve collaboration across multiple treatment cen­ters because of the disorder’s low prevalence.


Pyromania
Pyromania is characterized by (1) deliberate and purposeful fire setting on >1 occasion; (2) tension or affective arousal before the act; (3) fascination with, interest in, curiosity about, or attraction to fire and its situational con­texts; and (4) pleasure, gratification, or relief when setting fires or when witnessing or par­ticipating in their aftermath.3

Although pyromania is thought to be a disorder primarily affecting men, recent research suggests that the sex ratio is equal among adults and may be slightly higher among adolescent females. Mean age of onset usually is late adoles­cence. Pyromania appears to be chronic if untreated.55

Urges to set fires are common and the fire setting is almost always pleasurable. Severe distress follows the fire setting, and persons with pyromania report significant functional impairment. High rates of co-occurring psy­chiatric disorders (depression, substance use disorders, other impulse-control dis­orders) are common among persons with pyromania.55

Pharmacotherapy. There are no random­ized, controlled clinical trials examining pharmacotherapy for treating pyromania. There are no FDA-approved medications for pyromania.

In case reports, medications that have shown benefit in treating pyromania include topiramate, escitalopram, sertraline, fluox­etine, lithium, and a combination of olan­zapine and sodium valproate. An equal number of medications have shown no ben­efit: fluoxetine, valproic acid, lithium, sertra­line, olanzapine, escitalopram, citalopram, and clonazepam. A case report of an 18-year-old man with pyromania described success­fully using a combination of topiramate with 3 weeks of daily CBT to achieve significant symptom improvement.56,57

Pyromania is a largely unrecognized dis­order that causes significant psychological, social, and legal repercussions. Because few persons with pyromania volunteer informa­tion regarding fire-setting, it is important that clinicians recognize the disorder and screen patients appropriately. Various treatments have been helpful in case studies, but more research on the etiology and treatment of the disorder is needed.56,57


Conclusions based on the literature
In disruptive, impulse-control, and conduct disorders, the systematic study of treatment efficacy and tolerability is in its infancy. With few controlled studies published, it is not possible to make treatment recommendations with confidence. There are no FDA-approved drugs for treating any of these disorders.

Nonetheless, specific psychotherapies and drug therapies offer promising options, but often are based on small studies, often in patient populations with prominent comor­bidities, and have not been replicated by independent investigators. For all of these disorders, issues such as which psycho­therapy or medication to use and the ideal duration of treatment cannot be sufficiently addressed with the available data.

 

 

In conjunction with emerging epidemio­logical data supporting a relatively high prevalence of disruptive, impulse-control, and conduct disorders, the small amount of data regarding effective treatments highlights the clinical need for additional research.


Bottom Line
Empirically supported treatment options for impulse-control disorders currently are limited, because only select disorders have been studied across multiple trials. New research is needed to confirm possible treatment options and identify effective psychotherapeutic and pharmacological treatment alternatives.
 

Related Resources
• Grant JE. Impulse control disorders: a clinician’s guide to un­derstanding and treating behavioral addictions. New York, NY: W. W. Norton & Company; 2008.
• Grant JE, Kim SW. Stop me because I can’t stop myself: tak­ing control of impulsive behavior. New York, NY: McGraw- Hill; 2003.
• American Academy of Child and Adolescent Psychiatry. Conduct disorder resource center. http://www.aacap.org/AACAP/FamiliesandYouth/ResourceCenters/ConductDisorderResourceCenter/Home.aspx.


Drug Brand Names
Atomoxetine • Strattera                      Methylphenidate • Ritalin
Carbamazepine • Tegretol                  Molindone • Moban
Citalopram • Celexa                            Naltrexone • ReVia
Clonazepam • Klonopin                      Olanzapine • Zyprexa
Clonidine • Catapres                           Oxcarbazepine • Trileptal
D-amphetamine • Dexedrine               Quetiapine • Seroquel
Divalproex sodium • Depakote            Risperidone • Risperdal
Escitalopram • Lexapro                       Sertraline • Zoloft
Fluoxetine • Prozac                             Sodium valproate • Depacon
Guanfacine • Intuniv                           Thioridazine • Mellaril
Haloperidol • Haldol                             Topiramate • Topamax
Levetiracetam • Keppra                       Valproic acid • Depakote
Lithium • Eskalith, Lithobid  

 

Disclosures
Dr. Grant receives grant or research support from Brainsway, Forest Pharmaceuticals, and Roche Pharmaceuticals. Mr. Leppink reports no financial relationship with any company whose products are mentioned in this article or with competing products.

References


1. Kessler RC, Berglund P, Demler O, et al. Lifetime prevalence and age-of-onset distributions of DSM-IV disorders in the National Comorbidity Survey Replication. Arch Gen Psychiatry. 2005;62(6):593-602.
2. Grant JE, Levine L, Kim D, et al. Impulse control disorders in adult psychiatric inpatients. Am J Psychiatry. 2005;162(11):2184-2188.
3. Diagnostic and statistical manual of mental disorders, 5th ed. Washington, DC: American Psychiatric Association; 2013.
4. Turgay A. Psychopharmacological treatment of oppositional defiant disorder. CNS Drugs. 2009;23(1):1-17.
5. Hazell P. Review of attention-deficit/hyperactivity disorder comorbid with oppositional defiant disorder. Australas Psychiatry. 2010;18(6):556-559.
6. Burke JD, Loeber R, Birmaher B. Oppositional defiant disorder and conduct disorder: a review of the past 10 years, part II. J Am Acad Child Adolesc Psychiatry. 2002; 41(11):1275-1293.
7. Connor DF, Steeber J, McBurnett K. A review of attention-deficit/hyperactivity disorder complicated by symptoms of oppositional defiant disorder or conduct disorder. J Dev Behav Pediatr. 2010;31(5):427-440.
8. Aman MG, Bukstein OG, Gadow KD, et al. What does risperidone add to parent training and stimulant for severe aggression in child attention-deficit/hyperactivity disorder? J Am Acad Child Adolesc Psychiatry. 2014;53(1):47-60.e1.
9. Loy JH, Merry SN, Hetrick SE, et al. Atypical antipsychotics for disruptive behavior disorders in children and youths. Cochrane Database Syst Rev. 2012;9:CD008559.
10. Gadow KD, Arnold LE, Molina BS, et al. Risperidone added to parent training and stimulant medication: effects on attention-deficit/hyperactivity disorder, oppositional defiant disorder, conduct disorder, and peer aggression. J Am Acad Child Adolesc Psychiatry. 2014;53(9):948-959.e1.
12. Signorovitch J, Erder MH, Xie J, et al. Comparative effectiveness research using matching-adjusted indirect comparison: an application to treatment with guanfacine extended release or atomoxetine in children with attention-deficit/hyperactivity disorder and comorbid oppositional defiant disorder. Pharmacoepidemiol Drug Saf. 2012;21(suppl 2):130-137.
13. Bangs ME, Hazell P, Danckaerts M, et al; Atomoxetine ADHD/ODD Study Group. Atomoxetine for the treatment of attention-deficit/hyperactivity disorder and oppositional defiant disorder. Pediatrics. 2008;121(2):e314-e320.
14. Biederman J, Spencer TJ, Newcorn JH, et al. Effect of comorbid symptoms of oppositional defiant disorder on responses to atomoxetine in children with ADHD: a meta-analysis of controlled clinical trial data. Psychopharmacology (Berl). 2007;190(1):31-41.
15. Miller NV, Haas SM, Waschbusch DA, et al. Behavior therapy and callous-unemotional traits: effects of a pilot study examining modified behavioral contingencies on child behavior. Behav Ther. 2014;45(5):606-618.
16. Hamilton SS, Armando J. Oppositional defiant disorder. Am Fam Physician. 2008;78(7):861-866.
17. Steiner H, Remsing L; Work Group on Quality Issues. Practice parameter for the assessment and treatment of children and adolescents with oppositional defiant disorder. J Am Acad Child Adolesc Psychiatry. 2007;46(1):126-141.
18. Winther J, Carlsson A, Vance A. A pilot study of a school-based prevention and early intervention program to reduce oppositional defiant disorder/conduct disorder. Early Interv Psychiatry. 2014;8(2):181-189.
19. Plueck J, Eichelberger I, Hautmann C, et al. Effectiveness of a teacher-based indicated prevention program for preschool children with externalizing problem behavior [published online April 22, 2014]. Prev Sci. doi: 10.1007/s11121-014- 0487-x.
20. Dretzke J, Frew E, Davenport C, et al. The effectiveness and cost-effectiveness of parent training/education programmes for the treatment of conduct disorder, including oppositional defiant disorder, in children. Health Tech Assess. 2005;9(50):iii, ix-x, 1-233.
21. Coccaro EF, Schmidt CA, Samuels JF, et al. Lifetime and 1-month prevalence rates of intermittent explosive disorder in a community sample. J Clin Psychiatry. 2004;65(6):820-824.
22. McElroy SL, Soutullo CA, Beckman DA, et al. DSM-IV intermittent explosive disorder: a report of 27 cases. J Clin Psychiatry. 1998;59(4):203-210; quiz 211.
23. Coccaro EF, Lee RJ, Kavoussi RJ. A double-blind, randomized, placebo-controlled trial of fluoxetine in patients with intermittent explosive disorder. J Clin Psychiatry. 2009;70(5):653-662.
24. Coccaro EF. Intermittent explosive disorder as a disorder of impulsive aggression for DSM-5. Am J Psychiatry. 2012;169(6):577-588.
25. Mattes JA. Oxcarbazepine in patients with impulsive aggression: a double-blind, placebo-controlled trial. J Clin Psychopharmacol. 2005;25(6):575-579.
26. Hollander E, Tracy KA, Swann AC, et al. Divalproex in the treatment of impulsive aggression: efficacy in cluster B personality disorders. Neuropsychopharmacology. 2003;28(6):1186-1197.
27. Mattes JA. Levetiracetam in patients with impulsive aggression: a double-blind, placebo-controlled trial. J Clin Psychiatry. 2008;69(2):310-315.
28. McCloskey MS, Noblett KL, Deffenbacher JL, et al. Cognitive-behavioral therapy for intermittent explosive disorder: a pilot randomized clinical trial. J Consult Clin Psychol. 2008;76(5):876-886.
29. Campbell M, Small AM, Green WH, et al. Behavioral efficacy of haloperidol and lithium carbonate. A comparison in hospitalized aggressive children with conduct disorder. Arch Gen Psychiatry. 1984;41(7):650-656.
30. Campbell M, Adams PB, Small AM, et al. Lithium in hospitalized aggressive children with conduct disorder: a double-blind and placebo-controlled study. J Am Acad Child Adolesc Psychiatry. 1995;34(4):445-453.
31. Malone RP, Simpson GM. Psychopharmacology: use of placebos in clinical trials involving children and adolescents. Psychiatr Serv. 1998;49(11):1413-1414, 1417.
32. Malone RP, Delaney MA, Luebbert JF, et al. A double-blind placebo-controlled study of lithium in hospitalized aggressive children and adolescents with conduct disorder. Arch Gen Psychiatry. 2000;57(7):649-654.
33. Platt JE, Campbell M, Green WH, et al. Effects of lithium carbonate and haloperidol on cognition in aggressive hospitalized school-age children. J Clin Psychopharmacol. 1981;1(1):8-13.
34. Platt JE, Campbell M, Green WH, et al. Cognitive effects of lithium carbonate and haloperidol in treatment-resistant aggressive children. Arch Gen Psychiatry. 1984;41(7):657-662.
35. Rifkin A, Karajgi B, Dicker R, et al. Lithium treatment of conduct disorders in adolescents. Am J Psychiatry. 1997;154(4):554-555.
36. Cueva JE, Overall JE, Small AM, et al. Carbamazepine in aggressive children with conduct disorder: a double-blind and placebo-controlled study. J Am Acad Child Adolesc Psychiatry. 1996;35(4):480-490.
37. Findling RL, McNamara NK, Branicky LA, et al. A double-blind pilot study of risperidone in the treatment of conduct disorder. J Am Acad Child Adolesc Psychiatry. 2000;39(4):509-516.
38. Connor DF, McLaughlin TJ, Jeffers-Terry M. Randomized controlled pilot study of quetiapine in the treatment of adolescent conduct disorder. J Child Adolesc Psychopharmacol. 2008;18(2):140-156.
39. Greenhill LL, Solomon M, Pleak R, et al. Molindone hydrochloride treatment of hospitalized children with conduct disorder. J Clin Psychiatry. 1985;46(8 pt 2):20-25.
40. Khanzode LA, Saxena K, Kraemer H, et al. Efficacy profiles of psychopharmacology: divalproex sodium in conduct disorder. Child Psychiatry Hum Dev. 2006;37(1):55-64.
41. Padhy R, Saxena K, Remsing L, et al. Symptomatic response to divalproex in subtypes of conduct disorder. Child Psychiatry Hum Dev. 2011;42(5):584-593.
42. Steiner H, Petersen ML, Saxena K, et al. Divalproex sodium for the treatment of conduct disorder: a randomized controlled clinical trial. J Clin Psychiatry. 2003;64(10):1183-1191.
43. Klein RG, Abikoff H, Klass E, et al. Clinical efficacy of methylphenidate in conduct disorder with and without attention deficit hyperactivity disorder. Arch Gen Psychiatry. 1997;54(12):1073-1080.
44. Heneggeler SW, Sheidow AJ. Empirically supported family-based treatments for conduct disorder and delinquency in adolescents. J Marital Fam Ther. 2012;38(1):30-58.
45. Lochman JE, Powell NP, Boxmeyer CL, et al. Cognitive-behavioral therapy for externalizing disorder in children and adolescents. Child Adolesc Psychiatr Clin N Am. 2011;20(2):305-318.
46. Furlong M, McGilloway S, Bywater T, et al. Behavioural and cognitive-behavioural group-based parenting programmes for early-onset conduct problems in children aged 3 to 12 years. Cochrane Database Syst Rev. 2012;2:CD008225.
47. McElroy SL, Pope HG Jr, Hudson JI, et al. Kleptomania: a report of 20 cases. Am J Psychiatry. 1991;148(5):652-657.
48. Grant JE, Kim SW. Clinical characteristics and associated psychopathology of 22 patients with kleptomania. Compr Psychiatry. 2002;43(5):378-384.
49. Odlaug BL, Grant JE, Kim SW. Suicide attempts in 107 adolescents and adults with kleptomania. Arch Suicide Res. 2012;16(4):348-359.
50. Grant JE, Kim SW, Odlaug BL. A double-blind, placebo-controlled study of the opiate antagonist, naltrexone, in the treatment of kleptomania. Biol Psychiatry. 2009;65(7): 600-606.
51. Koran LM, Aboujaoude EN, Gamel NN. Escitalopram treatment of kleptomania: an open-label trial followed by double-blind discontinuation. J Clin Psychiatry. 2007;68(3):422-427.
52. Guidry LS. Use of a covert punishing contingency in compulsive stealing. J Behav Therapy Exp Psychiatry. 1975;6(2):169.
53. Gauthier J, Pellerin D. Management of compulsive shoplifting through covert sensitization. J Behav Therapy Exp Psychiatry. 1982;13(1):73-75.
54. McConaghy N, Blaszczynski A. Imaginal desensitization: a cost-effective treatment in two shop-lifters and a binge-eater resistant to previous therapy. Aus N Z J Psychiatry. 1988;22(1):78-82.
55. Grant JE, Won Kim S. Clinical characteristics and psychiatric comorbidity of pyromania. J Clin Psychiatry. 2007;68(11):1717-1722.
56. Grant JE, Odlaug B. Assessment and treatment of pyromania. In: Oxford handbook of impulse control disorders. Grant JE, Potenza MN, eds. Oxford, United Kingdom: Oxford University Press; 2012:353-359.
57. Dell’Osso B, Altamura AC, Allen A, et al. Epidemiologic and clinical updates on impulse control disorders: a critical review. Eur Arch Psychiatry Clin Neurosci. 2006;256(8):464-475.

References


1. Kessler RC, Berglund P, Demler O, et al. Lifetime prevalence and age-of-onset distributions of DSM-IV disorders in the National Comorbidity Survey Replication. Arch Gen Psychiatry. 2005;62(6):593-602.
2. Grant JE, Levine L, Kim D, et al. Impulse control disorders in adult psychiatric inpatients. Am J Psychiatry. 2005;162(11):2184-2188.
3. Diagnostic and statistical manual of mental disorders, 5th ed. Washington, DC: American Psychiatric Association; 2013.
4. Turgay A. Psychopharmacological treatment of oppositional defiant disorder. CNS Drugs. 2009;23(1):1-17.
5. Hazell P. Review of attention-deficit/hyperactivity disorder comorbid with oppositional defiant disorder. Australas Psychiatry. 2010;18(6):556-559.
6. Burke JD, Loeber R, Birmaher B. Oppositional defiant disorder and conduct disorder: a review of the past 10 years, part II. J Am Acad Child Adolesc Psychiatry. 2002; 41(11):1275-1293.
7. Connor DF, Steeber J, McBurnett K. A review of attention-deficit/hyperactivity disorder complicated by symptoms of oppositional defiant disorder or conduct disorder. J Dev Behav Pediatr. 2010;31(5):427-440.
8. Aman MG, Bukstein OG, Gadow KD, et al. What does risperidone add to parent training and stimulant for severe aggression in child attention-deficit/hyperactivity disorder? J Am Acad Child Adolesc Psychiatry. 2014;53(1):47-60.e1.
9. Loy JH, Merry SN, Hetrick SE, et al. Atypical antipsychotics for disruptive behavior disorders in children and youths. Cochrane Database Syst Rev. 2012;9:CD008559.
10. Gadow KD, Arnold LE, Molina BS, et al. Risperidone added to parent training and stimulant medication: effects on attention-deficit/hyperactivity disorder, oppositional defiant disorder, conduct disorder, and peer aggression. J Am Acad Child Adolesc Psychiatry. 2014;53(9):948-959.e1.
12. Signorovitch J, Erder MH, Xie J, et al. Comparative effectiveness research using matching-adjusted indirect comparison: an application to treatment with guanfacine extended release or atomoxetine in children with attention-deficit/hyperactivity disorder and comorbid oppositional defiant disorder. Pharmacoepidemiol Drug Saf. 2012;21(suppl 2):130-137.
13. Bangs ME, Hazell P, Danckaerts M, et al; Atomoxetine ADHD/ODD Study Group. Atomoxetine for the treatment of attention-deficit/hyperactivity disorder and oppositional defiant disorder. Pediatrics. 2008;121(2):e314-e320.
14. Biederman J, Spencer TJ, Newcorn JH, et al. Effect of comorbid symptoms of oppositional defiant disorder on responses to atomoxetine in children with ADHD: a meta-analysis of controlled clinical trial data. Psychopharmacology (Berl). 2007;190(1):31-41.
15. Miller NV, Haas SM, Waschbusch DA, et al. Behavior therapy and callous-unemotional traits: effects of a pilot study examining modified behavioral contingencies on child behavior. Behav Ther. 2014;45(5):606-618.
16. Hamilton SS, Armando J. Oppositional defiant disorder. Am Fam Physician. 2008;78(7):861-866.
17. Steiner H, Remsing L; Work Group on Quality Issues. Practice parameter for the assessment and treatment of children and adolescents with oppositional defiant disorder. J Am Acad Child Adolesc Psychiatry. 2007;46(1):126-141.
18. Winther J, Carlsson A, Vance A. A pilot study of a school-based prevention and early intervention program to reduce oppositional defiant disorder/conduct disorder. Early Interv Psychiatry. 2014;8(2):181-189.
19. Plueck J, Eichelberger I, Hautmann C, et al. Effectiveness of a teacher-based indicated prevention program for preschool children with externalizing problem behavior [published online April 22, 2014]. Prev Sci. doi: 10.1007/s11121-014- 0487-x.
20. Dretzke J, Frew E, Davenport C, et al. The effectiveness and cost-effectiveness of parent training/education programmes for the treatment of conduct disorder, including oppositional defiant disorder, in children. Health Tech Assess. 2005;9(50):iii, ix-x, 1-233.
21. Coccaro EF, Schmidt CA, Samuels JF, et al. Lifetime and 1-month prevalence rates of intermittent explosive disorder in a community sample. J Clin Psychiatry. 2004;65(6):820-824.
22. McElroy SL, Soutullo CA, Beckman DA, et al. DSM-IV intermittent explosive disorder: a report of 27 cases. J Clin Psychiatry. 1998;59(4):203-210; quiz 211.
23. Coccaro EF, Lee RJ, Kavoussi RJ. A double-blind, randomized, placebo-controlled trial of fluoxetine in patients with intermittent explosive disorder. J Clin Psychiatry. 2009;70(5):653-662.
24. Coccaro EF. Intermittent explosive disorder as a disorder of impulsive aggression for DSM-5. Am J Psychiatry. 2012;169(6):577-588.
25. Mattes JA. Oxcarbazepine in patients with impulsive aggression: a double-blind, placebo-controlled trial. J Clin Psychopharmacol. 2005;25(6):575-579.
26. Hollander E, Tracy KA, Swann AC, et al. Divalproex in the treatment of impulsive aggression: efficacy in cluster B personality disorders. Neuropsychopharmacology. 2003;28(6):1186-1197.
27. Mattes JA. Levetiracetam in patients with impulsive aggression: a double-blind, placebo-controlled trial. J Clin Psychiatry. 2008;69(2):310-315.
28. McCloskey MS, Noblett KL, Deffenbacher JL, et al. Cognitive-behavioral therapy for intermittent explosive disorder: a pilot randomized clinical trial. J Consult Clin Psychol. 2008;76(5):876-886.
29. Campbell M, Small AM, Green WH, et al. Behavioral efficacy of haloperidol and lithium carbonate. A comparison in hospitalized aggressive children with conduct disorder. Arch Gen Psychiatry. 1984;41(7):650-656.
30. Campbell M, Adams PB, Small AM, et al. Lithium in hospitalized aggressive children with conduct disorder: a double-blind and placebo-controlled study. J Am Acad Child Adolesc Psychiatry. 1995;34(4):445-453.
31. Malone RP, Simpson GM. Psychopharmacology: use of placebos in clinical trials involving children and adolescents. Psychiatr Serv. 1998;49(11):1413-1414, 1417.
32. Malone RP, Delaney MA, Luebbert JF, et al. A double-blind placebo-controlled study of lithium in hospitalized aggressive children and adolescents with conduct disorder. Arch Gen Psychiatry. 2000;57(7):649-654.
33. Platt JE, Campbell M, Green WH, et al. Effects of lithium carbonate and haloperidol on cognition in aggressive hospitalized school-age children. J Clin Psychopharmacol. 1981;1(1):8-13.
34. Platt JE, Campbell M, Green WH, et al. Cognitive effects of lithium carbonate and haloperidol in treatment-resistant aggressive children. Arch Gen Psychiatry. 1984;41(7):657-662.
35. Rifkin A, Karajgi B, Dicker R, et al. Lithium treatment of conduct disorders in adolescents. Am J Psychiatry. 1997;154(4):554-555.
36. Cueva JE, Overall JE, Small AM, et al. Carbamazepine in aggressive children with conduct disorder: a double-blind and placebo-controlled study. J Am Acad Child Adolesc Psychiatry. 1996;35(4):480-490.
37. Findling RL, McNamara NK, Branicky LA, et al. A double-blind pilot study of risperidone in the treatment of conduct disorder. J Am Acad Child Adolesc Psychiatry. 2000;39(4):509-516.
38. Connor DF, McLaughlin TJ, Jeffers-Terry M. Randomized controlled pilot study of quetiapine in the treatment of adolescent conduct disorder. J Child Adolesc Psychopharmacol. 2008;18(2):140-156.
39. Greenhill LL, Solomon M, Pleak R, et al. Molindone hydrochloride treatment of hospitalized children with conduct disorder. J Clin Psychiatry. 1985;46(8 pt 2):20-25.
40. Khanzode LA, Saxena K, Kraemer H, et al. Efficacy profiles of psychopharmacology: divalproex sodium in conduct disorder. Child Psychiatry Hum Dev. 2006;37(1):55-64.
41. Padhy R, Saxena K, Remsing L, et al. Symptomatic response to divalproex in subtypes of conduct disorder. Child Psychiatry Hum Dev. 2011;42(5):584-593.
42. Steiner H, Petersen ML, Saxena K, et al. Divalproex sodium for the treatment of conduct disorder: a randomized controlled clinical trial. J Clin Psychiatry. 2003;64(10):1183-1191.
43. Klein RG, Abikoff H, Klass E, et al. Clinical efficacy of methylphenidate in conduct disorder with and without attention deficit hyperactivity disorder. Arch Gen Psychiatry. 1997;54(12):1073-1080.
44. Heneggeler SW, Sheidow AJ. Empirically supported family-based treatments for conduct disorder and delinquency in adolescents. J Marital Fam Ther. 2012;38(1):30-58.
45. Lochman JE, Powell NP, Boxmeyer CL, et al. Cognitive-behavioral therapy for externalizing disorder in children and adolescents. Child Adolesc Psychiatr Clin N Am. 2011;20(2):305-318.
46. Furlong M, McGilloway S, Bywater T, et al. Behavioural and cognitive-behavioural group-based parenting programmes for early-onset conduct problems in children aged 3 to 12 years. Cochrane Database Syst Rev. 2012;2:CD008225.
47. McElroy SL, Pope HG Jr, Hudson JI, et al. Kleptomania: a report of 20 cases. Am J Psychiatry. 1991;148(5):652-657.
48. Grant JE, Kim SW. Clinical characteristics and associated psychopathology of 22 patients with kleptomania. Compr Psychiatry. 2002;43(5):378-384.
49. Odlaug BL, Grant JE, Kim SW. Suicide attempts in 107 adolescents and adults with kleptomania. Arch Suicide Res. 2012;16(4):348-359.
50. Grant JE, Kim SW, Odlaug BL. A double-blind, placebo-controlled study of the opiate antagonist, naltrexone, in the treatment of kleptomania. Biol Psychiatry. 2009;65(7): 600-606.
51. Koran LM, Aboujaoude EN, Gamel NN. Escitalopram treatment of kleptomania: an open-label trial followed by double-blind discontinuation. J Clin Psychiatry. 2007;68(3):422-427.
52. Guidry LS. Use of a covert punishing contingency in compulsive stealing. J Behav Therapy Exp Psychiatry. 1975;6(2):169.
53. Gauthier J, Pellerin D. Management of compulsive shoplifting through covert sensitization. J Behav Therapy Exp Psychiatry. 1982;13(1):73-75.
54. McConaghy N, Blaszczynski A. Imaginal desensitization: a cost-effective treatment in two shop-lifters and a binge-eater resistant to previous therapy. Aus N Z J Psychiatry. 1988;22(1):78-82.
55. Grant JE, Won Kim S. Clinical characteristics and psychiatric comorbidity of pyromania. J Clin Psychiatry. 2007;68(11):1717-1722.
56. Grant JE, Odlaug B. Assessment and treatment of pyromania. In: Oxford handbook of impulse control disorders. Grant JE, Potenza MN, eds. Oxford, United Kingdom: Oxford University Press; 2012:353-359.
57. Dell’Osso B, Altamura AC, Allen A, et al. Epidemiologic and clinical updates on impulse control disorders: a critical review. Eur Arch Psychiatry Clin Neurosci. 2006;256(8):464-475.

Issue
Current Psychiatry - 14(1)
Issue
Current Psychiatry - 14(1)
Page Number
29-36
Page Number
29-36
Publications
Publications
Topics
Article Type
Display Headline
Choosing a treatment for disruptive, impulse-control, and conduct disorders
Display Headline
Choosing a treatment for disruptive, impulse-control, and conduct disorders
Legacy Keywords
impulse-control disorders, conduct disorders, impulse control, disruptive disorders, kleptomania, oppositional disorder, intermittent explosive disorder, ADHD, attention deficit/hyperactivity disorder, pyromania
Legacy Keywords
impulse-control disorders, conduct disorders, impulse control, disruptive disorders, kleptomania, oppositional disorder, intermittent explosive disorder, ADHD, attention deficit/hyperactivity disorder, pyromania
Sections
Disallow All Ads
Alternative CME
Use ProPublica
Article PDF Media

Suvorexant for sleep-onset insomnia or sleep-maintenance insomnia, or both

Article Type
Changed
Tue, 12/11/2018 - 15:36
Display Headline
Suvorexant for sleep-onset insomnia or sleep-maintenance insomnia, or both

Suvorexant, FDA-approved to treat insomnia, has demonstrated efficacy in helping patients with insomnia improve their ability to fall asleep and remain asleep (Table 1).1 This first-in-class compound represents a novel mechanism of action to promoting sleep that may avoid some prob­lems associated with other hypnotics.2




Clinical implications
Insomnia is among the most common clini­cal complaints in psychiatry and medicine. The FDA-approved insomnia medications include several benzodiazepine-receptor agonists (zolpidem, eszopiclone, zaleplon), a melatonin-receptor agonist (ramelteon), and a histamine-receptor antagonist (low-dose doxepin). Suvorexant joins these drugs and is an entirely novel compound that is the first orexin- (also called hypo­cretin) receptor antagonist approved by the FDA for any indication.

Through a highly targeted mechanism of action, suvorexant could enhance sleep for patients with insomnia, while maintain­ing an acceptable safety profile.3 The drug should help patients with chronic insom­nia, particularly those who have difficulty maintaining sleep—the sleep disturbance pattern that is most challenging to treat pharmacotherapeutically.

Because orexin antagonists have not been used outside of clinical trials, it is too soon to tell whether suvorexant will have the ideal real-world efficacy and safety profile to make it a first-line treatment for insomnia patients, or if it will be reserved for those who have failed a trial of several other treatments.4

In theory, the orexin antagonist approach to treating insomnia could represent a major advance that modulates the fundamental pathology of the disorder.5 The syndrome of chronic insomnia encompasses not just the nighttime sleep disturbance but also an assort­ment of daytime symptoms that can include fatigue, poor concentration, irritability, and decreased school or work performance but usually not sleepiness. This constellation of nighttime and daytime symptoms could be conceptualized as a manifestation of persis­tent CNS hyperarousal. Because the orexin system promotes and reinforces arousal, per­haps an orexin antagonist that dampens the level of orexin activity will ameliorate the full spectrum of insomnia symptoms—not sim­ply sedate patients.6


How suvorexant works
Suvorexant is a potent and reversible dual orexin-receptor antagonist. The orexin system, first described in 1998, has a key role in promoting and stabilizing wake­fulness.7 Evidence suggests that people with chronic insomnia exhibit a central hyperarousal that perpetuates their sleep difficulty. Accordingly, a targeted phar­maceutical approach that reduces orexin activity should facilitate sleep onset and sleep maintenance for these patients. It is well known that the regulation of sleep and wakefulness depends on the interaction of multiple nuclei within the hypothalamus. Orexinergic neurons in the perifornical-lateral hypothalamic region project widely in the CNS and have especially dense con­nections with wake-promoting cholinergic, serotonergic, noradrenergic, and histamin­ergic neurons.6

A precursor prepro-orexin peptide is split into 2 orexin neurotransmitters (orexin A and orexin B). These 2 orexins bind with 2 G-protein-coupled receptors (OX1R and OX2R) that have both overlapping and distinct distributions.7 Suvorexant is highly selective and has similar affinity for OX1R and OX2R, functioning as an antag­onist for both.8 Fundamentally, suvorexant enhances sleep by dampening the arous­ing wake drive.


Pharmacokinetics
Suvorexant is available as an immediate-release tablet with pharmacokinetic prop­erties that offer benefits for sleep onset and maintenance.9 Ingestion under fasting conditions results in a median time to maxi­mum concentration (Tmax) of approximately 2 hours, although the Tmax values vary widely from patient to patient (range 30 minutes to 6 hours). Although suvorexant can be taken with food, there is a modest absorption delay after a high-fat meal, resulting in a further Tmax delay of approximately 1.5 hours.

Suvorexant is primarily metabolized through the cytochrome P450 (CYP) 3A path­way, with limited contribution by CYP2C19. There are no active metabolites. The suvorex­ant blood level and risk of side effects will be higher with concomitant use of CYP3A inhibitors. The drug should not be adminis­tered with strong CYP3A inhibitors; the ini­tial dosage should be reduced with moderate CYP3A inhibitors. Concomitant use of strong CYP3A inducers can result in a low suvorex­ant level and reduced efficacy.

Suvorexant has little effect on other med­ications, although a person taking digoxin might experience intestinal P-glycoprotein inhibition with a slight rise in the digoxin level. In a patient taking both medica­tions, monitoring of the digoxin level is recommended.

The elimination half-life of suvorexant is approximately 12 hours, with a steady state in approximately 3 days. Because the half-life of suvorexant is moderately long for a sleep-promoting medication, use of the drug might be associated with residual sleepiness the morning after bedtime dosing. The risk for next-morning sleepiness or impairment should be minimized, however, when using the recommended dosages. Elimination is approximately two-thirds through feces and one-third in the urine.

Suvorexant metabolism can be affected by sex and body mass index. Females and obese people have a modestly elevated expo­sure to suvorexant, as reflected by the area under the curve and maximum concentra­tion (Cmax). These patients might not require dosage adjustments unless they are obese and female, in which case they should take a lower dosage.

Age and race have not been shown to influence suvorexant metabolism to a signifi­cant degree. Patients with renal impairment and those with mild or moderate hepatic impairment do not need dosage adjust­ment. Suvorexant has not been evaluated in patients with severe hepatic impairment.

 

 


Efficacy
Suvorexant showed significant evidence of improved sleep onset and sleep maintenance in patients with insomnia in clinical trials. The key efficacy clinical trials with insomnia patients included a phase-IIb dose-finding study,10 2 similar 3-month phase-III studies,11 and one 12-month phase-III safety study that incorporated efficacy outcomes.12 All these trials included subjective sleep measures and all except for the long-term safety study also incorporated polysomnographic assess­ment. The specific sleep laboratory outcomes were latency to persistent sleep (LPS), wake after the onset of persistent sleep (WASO), total sleep time (TST), and sleep efficiency (SE). Subjective sleep outcomes were time to sleep onset (sTSO), wake after sleep onset (sWASO), and total sleep time (sTST). Other exploratory endpoints also were assessed. These efficacy and safety studies mostly were performed at dosages considerably higher than those approved by the FDA.

The dose-finding (phase-IIb) trial was conducted with non-geriatric (age 18 to 64) patients with insomnia in a random­ized, double-blind, crossover design of two 4-week periods with subjects given a nightly placebo or suvorexant (10 mg, 20 mg, 40 mg, or 80 mg).10 Each of the 4 groups included approximately 60 subjects. The 2 co-primary endpoints were SE at Night 1 and the end of Week 4; secondary endpoints were LPS and WASO. Suvorexant was associated with dos­age-related improvements in SE and WASO compared with placebo at both time points. Carryover effects from the period-1 active drug group complicated the analysis of LPS.

The phase-III efficacy and safety trials were performed with 40 mg high dosage (HD) and 20 mg low dosage (LD) groups for adults and with 30 mg HD and 15 mg LD groups for geriatric (age ≥65) patients.11 Two similarly designed 3-month randomized, double-blind, placebo-controlled pivotal efficacy studies assessed objective and sub­jective sleep measures in 4 groups with non-geriatric (HD and LD) and geriatric (HD and LD) insomnia patients.

After baseline assessment, patients took nightly bedtime doses of placebo; suvorexant, 40 mg or 20 mg (non-geriatric individuals); or suvorexant, 30 mg or 15 mg (geriatric indi­viduals). All subjects kept a daily electronic diary and had polysomnographic recordings performed on Night 1, at the end of Month 1, and at the end of Month 3. Both the indi­vidual studies and combined analyses (2,030 subjects) showed that, in non-geriatric and geriatric patients, HD suvorexant resulted in significantly greater improvement in key subjective and objective measures through­out the study (Table 2,9 and Table 3,9), with the exception of a single LPS outcome in 1 study, compared with placebo. The LD dosages also demonstrated efficacy, but to a reduced extent.

Subjective sleep outcomes were assessed in a 1-year randomized, placebo-controlled trial with nightly placebo, suvorexant, 40 mg, for non-geriatric, or suvorexant, 30 mg, for geriatric insomnia patients.12 The 1-year phase was completed with 484 subjects. Key efficacy outcomes were sTST and sTSO changes from baseline during the first month of treatment. Compared with placebo, suvorexant dosages demonstrated significantly greater efficacy, improvements that were sustained throughout the year.

Clinical trials found suvorexant to be gen­erally safe and well tolerated.13 However, specific safety concerns led the FDA to approve the medication at dosages lower than those assessed in the phase-III studies.1

Somnolence was the most common adverse event in clinical trials. In the phase- IIb dose-finding study, somnolence was reported in <1% in the placebo group, but was associated with suvorexant in 2% of the 10 mg group, 5% with 20 mg, 12% with 40 mg, and 11% with 80 mg.9 In the phase-III combined analysis of the 3-month studies, somnolence was reported by 3% in the placebo group and 7% of non-geriatric patients taking 20 mg or geriatric patients taking 15 mg. Somnolence was reported in 8% of women and 3% of men taking the 15 mg or 20 mg dosage in these stud­ies. The 1-year study was performed only with higher suvorexant dosages (30 mg and 40 mg), in comparison with placebo. In this long-term trial, somnolence was reported by 13% of subjects taking suvorexant and 3% taking placebo.

Additional safety issues in trials included excessive daytime sleepiness, impaired driv­ing, suicidal ideation, sleep paralysis, hyp­nagogic/hypnopompic hallucinations, and cataplexy-like symptoms.9 Occurrences of these events are rare but have been reported more often among patients taking suvorex­ant than among those taking placebo.


Unique clinical issues
The U.S. Drug Enforcement Agency has categorized suvorexant as a Schedule IV controlled substance. Although there is no evidence of physiological dependence or withdrawal symptoms with suvorexant, studies with recreational substance abusers have shown that the likeability rating is simi­lar to that of zolpidem.13


Contraindication
Suvorexant is contraindicated in patients with narcolepsy.9 The underlying pathol­ogy of narcolepsy involves a marked reduction in orexin functioning with corre­sponding excessive sleepiness and related symptoms, such as cataplexy, hypnago­gic hallucinations, and sleep paralysis. Although suvorexant has not been evalu­ated in patients with narcolepsy, the drug might, hypothetically, put patients at higher risk of the full spectrum of narco­lepsy symptoms.

There are no other contraindications for suvorexant.


Dosing
Suvorexant should be taken no more than once a night within 30 minutes of bedtime and with at least 7 hours before the planned wake time.9 The recommended starting dosage is 10 mg. If this dosage is well toler­ated but insufficiently effective, the dosage can be increased to a maximum of 20 mg. The 5-mg dosage is recommended for indi­viduals taking a moderate CYP3A inhibitor. Generally, patients should take the lowest effective dosage.

 

 

There are no specified limitations on the duration of suvorexant use. There is no evidence of withdrawal effects when discontinuing the medication. Patients tak­ing suvorexant should be educated about possible next-day effects that might impair driving or other activities that require full mental alertness, especially if they are tak­ing the 20-mg dosage.


Bottom Line
Suvorexant is FDA-approved for treating sleep onset and sleep maintenance insomnia. The drug is a dual orexin-receptor antagonist, which targets persistent CNS hyperarousal. In clinical trials, suvorexant improved the ability to fall asleep and remain asleep in patients with insomnia. It is generally safe and well tolerated. However, these studies evaluated dosages higher than those approved by the FDA.

 

Related Resources
• Jacobson LH, Callander GE, Hoyer D. Suvorexant for the treatment of insomnia. Expert Rev Clin Pharmacol. 2014; 7(6):711-730.
• Neubauer DN. New and emerging pharmacotherapeutic approaches for insomnia. Int Rev Psychiatry. 2014;26(2): 214-224.


Drug Brand Names
Doxepin • Silenor             Suvorexant • Belsomra
Digoxin • Lanoxin             Zaleplon • Sonata
Eszopiclone • Lunesta       Zolpidem • Ambien,
Ramelteon • Rozerem            Edluar, Intermezzo

 

Disclosure
Dr. Neubauer is a consultant to Ferring Pharmaceuticals and Vanda Pharmaceuticals.

References


1. U.S. Food and Drug Administration. Survorexant (orexin receptor antagonist). For insomnia characterized by difficulties with sleep onset and/or maintenance. http:// www.fda.gov/downloads/AdvisoryCommittees/ CommitteesMeetingMaterials/Drugs/Peripheraland CentralNervousSystemDrugsAdvisoryCommittee/ UCM352969.pdf. Published May 22, 2013. Accessed November 24, 2014.
2. Mignot E. Sleep, sleep disorders and hypocretin (orexin). Sleep Med. 2004;5(suppl 1):S2-S8.
3. Nishino S. The hypocretin/orexin receptor: therapeutic prospective in sleep disorders. Expert Opin Investig Drugs. 2007;16(11):1785-1797.
4. Citrome L. Suvorexant for insomnia: a systematic review of the efficacy and safety profile for this newly approved hypnotic - what is the number needed to treat, number needed to harm and likelihood to be helped or harmed? Int J Clin Pract. 2014;68(12):1429-1441.
5. Winrow CJ, Gotter AL, Cox CD, et al. Promotion of sleep by suvorexant-a novel dual orexin receptor antagonist. J Neurogenet. 2011;25(1-2):52-61.
6. Saper CB, Chou TC, Scammell TE. The sleep switch: hypothalamic control of sleep and wakefulness. Trends Neurosci. 2001;24(12):726-731.
7. Sakurai T, Amemiya A, Ishii M, et al. Orexins and orexin receptors: a family of hypothalamic neuropeptides and G protein-coupled receptors that regulate feeding behavior. Cell. 1998;92(4):573-585.
8. Winrow CJ, Renger JJ. Discovery and development of orexin receptor antagonists as therapeutics for insomnia. Br J Pharmacol. 2014;171(2):283-293.
9. Belsomra [package insert]. Whitehouse Station, NJ: Merck; 2014.
10. Herring WJ, Snyder E, Budd K, et al. Orexin receptor antagonism for treatment of insomnia: a randomized clinical trial of suvorexant. Neurology. 2012;79(23):2265-2274.
11. Ivgy-May N, Snavely D, Minigh J, et al. Efficacy of suvorexant, an orexin receptor antagonist, in patients with primary insomnia: integrated results from 2 similarly designed phase 3 trials. Sleep. 2013;36(abstract supplement): A192.
12. Michelson D, Snyder E, Paradis E, et al. Safety and efficacy of suvorexant during 1-year treatment of insomnia with subsequent abrupt treatment discontinuation: a phase 3 randomised, double-blind, placebo-controlled trial. Lancet Neurol. 2014;13(5):461-471.
13. Merck Sharp and Dohme Corporation. Suvorexant advisory committee meeting briefing document. http:// www.fda.govdownloadsadvisorycommittees/committee smeetingmaterials/drugsperipheralandcentralnervous systemdrugsadvisorycommittee/ucm352970.pdf. Published May 22, 2013. Accessed November 24, 2014.

Article PDF
Author and Disclosure Information

 

David N. Neubauer, MD
Associate Professor
Department of Psychiatry
Johns Hopkins University School of Medicine
Baltimore, Maryland

Issue
Current Psychiatry - 14(1)
Publications
Topics
Page Number
19-24
Legacy Keywords
insomnia, sleep-onset insomnia, sleep-maintenance insomnia, survorexant, treatment for insomnia, insomnia treatment, sleep disorders
Sections
Author and Disclosure Information

 

David N. Neubauer, MD
Associate Professor
Department of Psychiatry
Johns Hopkins University School of Medicine
Baltimore, Maryland

Author and Disclosure Information

 

David N. Neubauer, MD
Associate Professor
Department of Psychiatry
Johns Hopkins University School of Medicine
Baltimore, Maryland

Article PDF
Article PDF

Suvorexant, FDA-approved to treat insomnia, has demonstrated efficacy in helping patients with insomnia improve their ability to fall asleep and remain asleep (Table 1).1 This first-in-class compound represents a novel mechanism of action to promoting sleep that may avoid some prob­lems associated with other hypnotics.2




Clinical implications
Insomnia is among the most common clini­cal complaints in psychiatry and medicine. The FDA-approved insomnia medications include several benzodiazepine-receptor agonists (zolpidem, eszopiclone, zaleplon), a melatonin-receptor agonist (ramelteon), and a histamine-receptor antagonist (low-dose doxepin). Suvorexant joins these drugs and is an entirely novel compound that is the first orexin- (also called hypo­cretin) receptor antagonist approved by the FDA for any indication.

Through a highly targeted mechanism of action, suvorexant could enhance sleep for patients with insomnia, while maintain­ing an acceptable safety profile.3 The drug should help patients with chronic insom­nia, particularly those who have difficulty maintaining sleep—the sleep disturbance pattern that is most challenging to treat pharmacotherapeutically.

Because orexin antagonists have not been used outside of clinical trials, it is too soon to tell whether suvorexant will have the ideal real-world efficacy and safety profile to make it a first-line treatment for insomnia patients, or if it will be reserved for those who have failed a trial of several other treatments.4

In theory, the orexin antagonist approach to treating insomnia could represent a major advance that modulates the fundamental pathology of the disorder.5 The syndrome of chronic insomnia encompasses not just the nighttime sleep disturbance but also an assort­ment of daytime symptoms that can include fatigue, poor concentration, irritability, and decreased school or work performance but usually not sleepiness. This constellation of nighttime and daytime symptoms could be conceptualized as a manifestation of persis­tent CNS hyperarousal. Because the orexin system promotes and reinforces arousal, per­haps an orexin antagonist that dampens the level of orexin activity will ameliorate the full spectrum of insomnia symptoms—not sim­ply sedate patients.6


How suvorexant works
Suvorexant is a potent and reversible dual orexin-receptor antagonist. The orexin system, first described in 1998, has a key role in promoting and stabilizing wake­fulness.7 Evidence suggests that people with chronic insomnia exhibit a central hyperarousal that perpetuates their sleep difficulty. Accordingly, a targeted phar­maceutical approach that reduces orexin activity should facilitate sleep onset and sleep maintenance for these patients. It is well known that the regulation of sleep and wakefulness depends on the interaction of multiple nuclei within the hypothalamus. Orexinergic neurons in the perifornical-lateral hypothalamic region project widely in the CNS and have especially dense con­nections with wake-promoting cholinergic, serotonergic, noradrenergic, and histamin­ergic neurons.6

A precursor prepro-orexin peptide is split into 2 orexin neurotransmitters (orexin A and orexin B). These 2 orexins bind with 2 G-protein-coupled receptors (OX1R and OX2R) that have both overlapping and distinct distributions.7 Suvorexant is highly selective and has similar affinity for OX1R and OX2R, functioning as an antag­onist for both.8 Fundamentally, suvorexant enhances sleep by dampening the arous­ing wake drive.


Pharmacokinetics
Suvorexant is available as an immediate-release tablet with pharmacokinetic prop­erties that offer benefits for sleep onset and maintenance.9 Ingestion under fasting conditions results in a median time to maxi­mum concentration (Tmax) of approximately 2 hours, although the Tmax values vary widely from patient to patient (range 30 minutes to 6 hours). Although suvorexant can be taken with food, there is a modest absorption delay after a high-fat meal, resulting in a further Tmax delay of approximately 1.5 hours.

Suvorexant is primarily metabolized through the cytochrome P450 (CYP) 3A path­way, with limited contribution by CYP2C19. There are no active metabolites. The suvorex­ant blood level and risk of side effects will be higher with concomitant use of CYP3A inhibitors. The drug should not be adminis­tered with strong CYP3A inhibitors; the ini­tial dosage should be reduced with moderate CYP3A inhibitors. Concomitant use of strong CYP3A inducers can result in a low suvorex­ant level and reduced efficacy.

Suvorexant has little effect on other med­ications, although a person taking digoxin might experience intestinal P-glycoprotein inhibition with a slight rise in the digoxin level. In a patient taking both medica­tions, monitoring of the digoxin level is recommended.

The elimination half-life of suvorexant is approximately 12 hours, with a steady state in approximately 3 days. Because the half-life of suvorexant is moderately long for a sleep-promoting medication, use of the drug might be associated with residual sleepiness the morning after bedtime dosing. The risk for next-morning sleepiness or impairment should be minimized, however, when using the recommended dosages. Elimination is approximately two-thirds through feces and one-third in the urine.

Suvorexant metabolism can be affected by sex and body mass index. Females and obese people have a modestly elevated expo­sure to suvorexant, as reflected by the area under the curve and maximum concentra­tion (Cmax). These patients might not require dosage adjustments unless they are obese and female, in which case they should take a lower dosage.

Age and race have not been shown to influence suvorexant metabolism to a signifi­cant degree. Patients with renal impairment and those with mild or moderate hepatic impairment do not need dosage adjust­ment. Suvorexant has not been evaluated in patients with severe hepatic impairment.

 

 


Efficacy
Suvorexant showed significant evidence of improved sleep onset and sleep maintenance in patients with insomnia in clinical trials. The key efficacy clinical trials with insomnia patients included a phase-IIb dose-finding study,10 2 similar 3-month phase-III studies,11 and one 12-month phase-III safety study that incorporated efficacy outcomes.12 All these trials included subjective sleep measures and all except for the long-term safety study also incorporated polysomnographic assess­ment. The specific sleep laboratory outcomes were latency to persistent sleep (LPS), wake after the onset of persistent sleep (WASO), total sleep time (TST), and sleep efficiency (SE). Subjective sleep outcomes were time to sleep onset (sTSO), wake after sleep onset (sWASO), and total sleep time (sTST). Other exploratory endpoints also were assessed. These efficacy and safety studies mostly were performed at dosages considerably higher than those approved by the FDA.

The dose-finding (phase-IIb) trial was conducted with non-geriatric (age 18 to 64) patients with insomnia in a random­ized, double-blind, crossover design of two 4-week periods with subjects given a nightly placebo or suvorexant (10 mg, 20 mg, 40 mg, or 80 mg).10 Each of the 4 groups included approximately 60 subjects. The 2 co-primary endpoints were SE at Night 1 and the end of Week 4; secondary endpoints were LPS and WASO. Suvorexant was associated with dos­age-related improvements in SE and WASO compared with placebo at both time points. Carryover effects from the period-1 active drug group complicated the analysis of LPS.

The phase-III efficacy and safety trials were performed with 40 mg high dosage (HD) and 20 mg low dosage (LD) groups for adults and with 30 mg HD and 15 mg LD groups for geriatric (age ≥65) patients.11 Two similarly designed 3-month randomized, double-blind, placebo-controlled pivotal efficacy studies assessed objective and sub­jective sleep measures in 4 groups with non-geriatric (HD and LD) and geriatric (HD and LD) insomnia patients.

After baseline assessment, patients took nightly bedtime doses of placebo; suvorexant, 40 mg or 20 mg (non-geriatric individuals); or suvorexant, 30 mg or 15 mg (geriatric indi­viduals). All subjects kept a daily electronic diary and had polysomnographic recordings performed on Night 1, at the end of Month 1, and at the end of Month 3. Both the indi­vidual studies and combined analyses (2,030 subjects) showed that, in non-geriatric and geriatric patients, HD suvorexant resulted in significantly greater improvement in key subjective and objective measures through­out the study (Table 2,9 and Table 3,9), with the exception of a single LPS outcome in 1 study, compared with placebo. The LD dosages also demonstrated efficacy, but to a reduced extent.

Subjective sleep outcomes were assessed in a 1-year randomized, placebo-controlled trial with nightly placebo, suvorexant, 40 mg, for non-geriatric, or suvorexant, 30 mg, for geriatric insomnia patients.12 The 1-year phase was completed with 484 subjects. Key efficacy outcomes were sTST and sTSO changes from baseline during the first month of treatment. Compared with placebo, suvorexant dosages demonstrated significantly greater efficacy, improvements that were sustained throughout the year.

Clinical trials found suvorexant to be gen­erally safe and well tolerated.13 However, specific safety concerns led the FDA to approve the medication at dosages lower than those assessed in the phase-III studies.1

Somnolence was the most common adverse event in clinical trials. In the phase- IIb dose-finding study, somnolence was reported in <1% in the placebo group, but was associated with suvorexant in 2% of the 10 mg group, 5% with 20 mg, 12% with 40 mg, and 11% with 80 mg.9 In the phase-III combined analysis of the 3-month studies, somnolence was reported by 3% in the placebo group and 7% of non-geriatric patients taking 20 mg or geriatric patients taking 15 mg. Somnolence was reported in 8% of women and 3% of men taking the 15 mg or 20 mg dosage in these stud­ies. The 1-year study was performed only with higher suvorexant dosages (30 mg and 40 mg), in comparison with placebo. In this long-term trial, somnolence was reported by 13% of subjects taking suvorexant and 3% taking placebo.

Additional safety issues in trials included excessive daytime sleepiness, impaired driv­ing, suicidal ideation, sleep paralysis, hyp­nagogic/hypnopompic hallucinations, and cataplexy-like symptoms.9 Occurrences of these events are rare but have been reported more often among patients taking suvorex­ant than among those taking placebo.


Unique clinical issues
The U.S. Drug Enforcement Agency has categorized suvorexant as a Schedule IV controlled substance. Although there is no evidence of physiological dependence or withdrawal symptoms with suvorexant, studies with recreational substance abusers have shown that the likeability rating is simi­lar to that of zolpidem.13


Contraindication
Suvorexant is contraindicated in patients with narcolepsy.9 The underlying pathol­ogy of narcolepsy involves a marked reduction in orexin functioning with corre­sponding excessive sleepiness and related symptoms, such as cataplexy, hypnago­gic hallucinations, and sleep paralysis. Although suvorexant has not been evalu­ated in patients with narcolepsy, the drug might, hypothetically, put patients at higher risk of the full spectrum of narco­lepsy symptoms.

There are no other contraindications for suvorexant.


Dosing
Suvorexant should be taken no more than once a night within 30 minutes of bedtime and with at least 7 hours before the planned wake time.9 The recommended starting dosage is 10 mg. If this dosage is well toler­ated but insufficiently effective, the dosage can be increased to a maximum of 20 mg. The 5-mg dosage is recommended for indi­viduals taking a moderate CYP3A inhibitor. Generally, patients should take the lowest effective dosage.

 

 

There are no specified limitations on the duration of suvorexant use. There is no evidence of withdrawal effects when discontinuing the medication. Patients tak­ing suvorexant should be educated about possible next-day effects that might impair driving or other activities that require full mental alertness, especially if they are tak­ing the 20-mg dosage.


Bottom Line
Suvorexant is FDA-approved for treating sleep onset and sleep maintenance insomnia. The drug is a dual orexin-receptor antagonist, which targets persistent CNS hyperarousal. In clinical trials, suvorexant improved the ability to fall asleep and remain asleep in patients with insomnia. It is generally safe and well tolerated. However, these studies evaluated dosages higher than those approved by the FDA.

 

Related Resources
• Jacobson LH, Callander GE, Hoyer D. Suvorexant for the treatment of insomnia. Expert Rev Clin Pharmacol. 2014; 7(6):711-730.
• Neubauer DN. New and emerging pharmacotherapeutic approaches for insomnia. Int Rev Psychiatry. 2014;26(2): 214-224.


Drug Brand Names
Doxepin • Silenor             Suvorexant • Belsomra
Digoxin • Lanoxin             Zaleplon • Sonata
Eszopiclone • Lunesta       Zolpidem • Ambien,
Ramelteon • Rozerem            Edluar, Intermezzo

 

Disclosure
Dr. Neubauer is a consultant to Ferring Pharmaceuticals and Vanda Pharmaceuticals.

Suvorexant, FDA-approved to treat insomnia, has demonstrated efficacy in helping patients with insomnia improve their ability to fall asleep and remain asleep (Table 1).1 This first-in-class compound represents a novel mechanism of action to promoting sleep that may avoid some prob­lems associated with other hypnotics.2




Clinical implications
Insomnia is among the most common clini­cal complaints in psychiatry and medicine. The FDA-approved insomnia medications include several benzodiazepine-receptor agonists (zolpidem, eszopiclone, zaleplon), a melatonin-receptor agonist (ramelteon), and a histamine-receptor antagonist (low-dose doxepin). Suvorexant joins these drugs and is an entirely novel compound that is the first orexin- (also called hypo­cretin) receptor antagonist approved by the FDA for any indication.

Through a highly targeted mechanism of action, suvorexant could enhance sleep for patients with insomnia, while maintain­ing an acceptable safety profile.3 The drug should help patients with chronic insom­nia, particularly those who have difficulty maintaining sleep—the sleep disturbance pattern that is most challenging to treat pharmacotherapeutically.

Because orexin antagonists have not been used outside of clinical trials, it is too soon to tell whether suvorexant will have the ideal real-world efficacy and safety profile to make it a first-line treatment for insomnia patients, or if it will be reserved for those who have failed a trial of several other treatments.4

In theory, the orexin antagonist approach to treating insomnia could represent a major advance that modulates the fundamental pathology of the disorder.5 The syndrome of chronic insomnia encompasses not just the nighttime sleep disturbance but also an assort­ment of daytime symptoms that can include fatigue, poor concentration, irritability, and decreased school or work performance but usually not sleepiness. This constellation of nighttime and daytime symptoms could be conceptualized as a manifestation of persis­tent CNS hyperarousal. Because the orexin system promotes and reinforces arousal, per­haps an orexin antagonist that dampens the level of orexin activity will ameliorate the full spectrum of insomnia symptoms—not sim­ply sedate patients.6


How suvorexant works
Suvorexant is a potent and reversible dual orexin-receptor antagonist. The orexin system, first described in 1998, has a key role in promoting and stabilizing wake­fulness.7 Evidence suggests that people with chronic insomnia exhibit a central hyperarousal that perpetuates their sleep difficulty. Accordingly, a targeted phar­maceutical approach that reduces orexin activity should facilitate sleep onset and sleep maintenance for these patients. It is well known that the regulation of sleep and wakefulness depends on the interaction of multiple nuclei within the hypothalamus. Orexinergic neurons in the perifornical-lateral hypothalamic region project widely in the CNS and have especially dense con­nections with wake-promoting cholinergic, serotonergic, noradrenergic, and histamin­ergic neurons.6

A precursor prepro-orexin peptide is split into 2 orexin neurotransmitters (orexin A and orexin B). These 2 orexins bind with 2 G-protein-coupled receptors (OX1R and OX2R) that have both overlapping and distinct distributions.7 Suvorexant is highly selective and has similar affinity for OX1R and OX2R, functioning as an antag­onist for both.8 Fundamentally, suvorexant enhances sleep by dampening the arous­ing wake drive.


Pharmacokinetics
Suvorexant is available as an immediate-release tablet with pharmacokinetic prop­erties that offer benefits for sleep onset and maintenance.9 Ingestion under fasting conditions results in a median time to maxi­mum concentration (Tmax) of approximately 2 hours, although the Tmax values vary widely from patient to patient (range 30 minutes to 6 hours). Although suvorexant can be taken with food, there is a modest absorption delay after a high-fat meal, resulting in a further Tmax delay of approximately 1.5 hours.

Suvorexant is primarily metabolized through the cytochrome P450 (CYP) 3A path­way, with limited contribution by CYP2C19. There are no active metabolites. The suvorex­ant blood level and risk of side effects will be higher with concomitant use of CYP3A inhibitors. The drug should not be adminis­tered with strong CYP3A inhibitors; the ini­tial dosage should be reduced with moderate CYP3A inhibitors. Concomitant use of strong CYP3A inducers can result in a low suvorex­ant level and reduced efficacy.

Suvorexant has little effect on other med­ications, although a person taking digoxin might experience intestinal P-glycoprotein inhibition with a slight rise in the digoxin level. In a patient taking both medica­tions, monitoring of the digoxin level is recommended.

The elimination half-life of suvorexant is approximately 12 hours, with a steady state in approximately 3 days. Because the half-life of suvorexant is moderately long for a sleep-promoting medication, use of the drug might be associated with residual sleepiness the morning after bedtime dosing. The risk for next-morning sleepiness or impairment should be minimized, however, when using the recommended dosages. Elimination is approximately two-thirds through feces and one-third in the urine.

Suvorexant metabolism can be affected by sex and body mass index. Females and obese people have a modestly elevated expo­sure to suvorexant, as reflected by the area under the curve and maximum concentra­tion (Cmax). These patients might not require dosage adjustments unless they are obese and female, in which case they should take a lower dosage.

Age and race have not been shown to influence suvorexant metabolism to a signifi­cant degree. Patients with renal impairment and those with mild or moderate hepatic impairment do not need dosage adjust­ment. Suvorexant has not been evaluated in patients with severe hepatic impairment.

 

 


Efficacy
Suvorexant showed significant evidence of improved sleep onset and sleep maintenance in patients with insomnia in clinical trials. The key efficacy clinical trials with insomnia patients included a phase-IIb dose-finding study,10 2 similar 3-month phase-III studies,11 and one 12-month phase-III safety study that incorporated efficacy outcomes.12 All these trials included subjective sleep measures and all except for the long-term safety study also incorporated polysomnographic assess­ment. The specific sleep laboratory outcomes were latency to persistent sleep (LPS), wake after the onset of persistent sleep (WASO), total sleep time (TST), and sleep efficiency (SE). Subjective sleep outcomes were time to sleep onset (sTSO), wake after sleep onset (sWASO), and total sleep time (sTST). Other exploratory endpoints also were assessed. These efficacy and safety studies mostly were performed at dosages considerably higher than those approved by the FDA.

The dose-finding (phase-IIb) trial was conducted with non-geriatric (age 18 to 64) patients with insomnia in a random­ized, double-blind, crossover design of two 4-week periods with subjects given a nightly placebo or suvorexant (10 mg, 20 mg, 40 mg, or 80 mg).10 Each of the 4 groups included approximately 60 subjects. The 2 co-primary endpoints were SE at Night 1 and the end of Week 4; secondary endpoints were LPS and WASO. Suvorexant was associated with dos­age-related improvements in SE and WASO compared with placebo at both time points. Carryover effects from the period-1 active drug group complicated the analysis of LPS.

The phase-III efficacy and safety trials were performed with 40 mg high dosage (HD) and 20 mg low dosage (LD) groups for adults and with 30 mg HD and 15 mg LD groups for geriatric (age ≥65) patients.11 Two similarly designed 3-month randomized, double-blind, placebo-controlled pivotal efficacy studies assessed objective and sub­jective sleep measures in 4 groups with non-geriatric (HD and LD) and geriatric (HD and LD) insomnia patients.

After baseline assessment, patients took nightly bedtime doses of placebo; suvorexant, 40 mg or 20 mg (non-geriatric individuals); or suvorexant, 30 mg or 15 mg (geriatric indi­viduals). All subjects kept a daily electronic diary and had polysomnographic recordings performed on Night 1, at the end of Month 1, and at the end of Month 3. Both the indi­vidual studies and combined analyses (2,030 subjects) showed that, in non-geriatric and geriatric patients, HD suvorexant resulted in significantly greater improvement in key subjective and objective measures through­out the study (Table 2,9 and Table 3,9), with the exception of a single LPS outcome in 1 study, compared with placebo. The LD dosages also demonstrated efficacy, but to a reduced extent.

Subjective sleep outcomes were assessed in a 1-year randomized, placebo-controlled trial with nightly placebo, suvorexant, 40 mg, for non-geriatric, or suvorexant, 30 mg, for geriatric insomnia patients.12 The 1-year phase was completed with 484 subjects. Key efficacy outcomes were sTST and sTSO changes from baseline during the first month of treatment. Compared with placebo, suvorexant dosages demonstrated significantly greater efficacy, improvements that were sustained throughout the year.

Clinical trials found suvorexant to be gen­erally safe and well tolerated.13 However, specific safety concerns led the FDA to approve the medication at dosages lower than those assessed in the phase-III studies.1

Somnolence was the most common adverse event in clinical trials. In the phase- IIb dose-finding study, somnolence was reported in <1% in the placebo group, but was associated with suvorexant in 2% of the 10 mg group, 5% with 20 mg, 12% with 40 mg, and 11% with 80 mg.9 In the phase-III combined analysis of the 3-month studies, somnolence was reported by 3% in the placebo group and 7% of non-geriatric patients taking 20 mg or geriatric patients taking 15 mg. Somnolence was reported in 8% of women and 3% of men taking the 15 mg or 20 mg dosage in these stud­ies. The 1-year study was performed only with higher suvorexant dosages (30 mg and 40 mg), in comparison with placebo. In this long-term trial, somnolence was reported by 13% of subjects taking suvorexant and 3% taking placebo.

Additional safety issues in trials included excessive daytime sleepiness, impaired driv­ing, suicidal ideation, sleep paralysis, hyp­nagogic/hypnopompic hallucinations, and cataplexy-like symptoms.9 Occurrences of these events are rare but have been reported more often among patients taking suvorex­ant than among those taking placebo.


Unique clinical issues
The U.S. Drug Enforcement Agency has categorized suvorexant as a Schedule IV controlled substance. Although there is no evidence of physiological dependence or withdrawal symptoms with suvorexant, studies with recreational substance abusers have shown that the likeability rating is simi­lar to that of zolpidem.13


Contraindication
Suvorexant is contraindicated in patients with narcolepsy.9 The underlying pathol­ogy of narcolepsy involves a marked reduction in orexin functioning with corre­sponding excessive sleepiness and related symptoms, such as cataplexy, hypnago­gic hallucinations, and sleep paralysis. Although suvorexant has not been evalu­ated in patients with narcolepsy, the drug might, hypothetically, put patients at higher risk of the full spectrum of narco­lepsy symptoms.

There are no other contraindications for suvorexant.


Dosing
Suvorexant should be taken no more than once a night within 30 minutes of bedtime and with at least 7 hours before the planned wake time.9 The recommended starting dosage is 10 mg. If this dosage is well toler­ated but insufficiently effective, the dosage can be increased to a maximum of 20 mg. The 5-mg dosage is recommended for indi­viduals taking a moderate CYP3A inhibitor. Generally, patients should take the lowest effective dosage.

 

 

There are no specified limitations on the duration of suvorexant use. There is no evidence of withdrawal effects when discontinuing the medication. Patients tak­ing suvorexant should be educated about possible next-day effects that might impair driving or other activities that require full mental alertness, especially if they are tak­ing the 20-mg dosage.


Bottom Line
Suvorexant is FDA-approved for treating sleep onset and sleep maintenance insomnia. The drug is a dual orexin-receptor antagonist, which targets persistent CNS hyperarousal. In clinical trials, suvorexant improved the ability to fall asleep and remain asleep in patients with insomnia. It is generally safe and well tolerated. However, these studies evaluated dosages higher than those approved by the FDA.

 

Related Resources
• Jacobson LH, Callander GE, Hoyer D. Suvorexant for the treatment of insomnia. Expert Rev Clin Pharmacol. 2014; 7(6):711-730.
• Neubauer DN. New and emerging pharmacotherapeutic approaches for insomnia. Int Rev Psychiatry. 2014;26(2): 214-224.


Drug Brand Names
Doxepin • Silenor             Suvorexant • Belsomra
Digoxin • Lanoxin             Zaleplon • Sonata
Eszopiclone • Lunesta       Zolpidem • Ambien,
Ramelteon • Rozerem            Edluar, Intermezzo

 

Disclosure
Dr. Neubauer is a consultant to Ferring Pharmaceuticals and Vanda Pharmaceuticals.

References


1. U.S. Food and Drug Administration. Survorexant (orexin receptor antagonist). For insomnia characterized by difficulties with sleep onset and/or maintenance. http:// www.fda.gov/downloads/AdvisoryCommittees/ CommitteesMeetingMaterials/Drugs/Peripheraland CentralNervousSystemDrugsAdvisoryCommittee/ UCM352969.pdf. Published May 22, 2013. Accessed November 24, 2014.
2. Mignot E. Sleep, sleep disorders and hypocretin (orexin). Sleep Med. 2004;5(suppl 1):S2-S8.
3. Nishino S. The hypocretin/orexin receptor: therapeutic prospective in sleep disorders. Expert Opin Investig Drugs. 2007;16(11):1785-1797.
4. Citrome L. Suvorexant for insomnia: a systematic review of the efficacy and safety profile for this newly approved hypnotic - what is the number needed to treat, number needed to harm and likelihood to be helped or harmed? Int J Clin Pract. 2014;68(12):1429-1441.
5. Winrow CJ, Gotter AL, Cox CD, et al. Promotion of sleep by suvorexant-a novel dual orexin receptor antagonist. J Neurogenet. 2011;25(1-2):52-61.
6. Saper CB, Chou TC, Scammell TE. The sleep switch: hypothalamic control of sleep and wakefulness. Trends Neurosci. 2001;24(12):726-731.
7. Sakurai T, Amemiya A, Ishii M, et al. Orexins and orexin receptors: a family of hypothalamic neuropeptides and G protein-coupled receptors that regulate feeding behavior. Cell. 1998;92(4):573-585.
8. Winrow CJ, Renger JJ. Discovery and development of orexin receptor antagonists as therapeutics for insomnia. Br J Pharmacol. 2014;171(2):283-293.
9. Belsomra [package insert]. Whitehouse Station, NJ: Merck; 2014.
10. Herring WJ, Snyder E, Budd K, et al. Orexin receptor antagonism for treatment of insomnia: a randomized clinical trial of suvorexant. Neurology. 2012;79(23):2265-2274.
11. Ivgy-May N, Snavely D, Minigh J, et al. Efficacy of suvorexant, an orexin receptor antagonist, in patients with primary insomnia: integrated results from 2 similarly designed phase 3 trials. Sleep. 2013;36(abstract supplement): A192.
12. Michelson D, Snyder E, Paradis E, et al. Safety and efficacy of suvorexant during 1-year treatment of insomnia with subsequent abrupt treatment discontinuation: a phase 3 randomised, double-blind, placebo-controlled trial. Lancet Neurol. 2014;13(5):461-471.
13. Merck Sharp and Dohme Corporation. Suvorexant advisory committee meeting briefing document. http:// www.fda.govdownloadsadvisorycommittees/committee smeetingmaterials/drugsperipheralandcentralnervous systemdrugsadvisorycommittee/ucm352970.pdf. Published May 22, 2013. Accessed November 24, 2014.

References


1. U.S. Food and Drug Administration. Survorexant (orexin receptor antagonist). For insomnia characterized by difficulties with sleep onset and/or maintenance. http:// www.fda.gov/downloads/AdvisoryCommittees/ CommitteesMeetingMaterials/Drugs/Peripheraland CentralNervousSystemDrugsAdvisoryCommittee/ UCM352969.pdf. Published May 22, 2013. Accessed November 24, 2014.
2. Mignot E. Sleep, sleep disorders and hypocretin (orexin). Sleep Med. 2004;5(suppl 1):S2-S8.
3. Nishino S. The hypocretin/orexin receptor: therapeutic prospective in sleep disorders. Expert Opin Investig Drugs. 2007;16(11):1785-1797.
4. Citrome L. Suvorexant for insomnia: a systematic review of the efficacy and safety profile for this newly approved hypnotic - what is the number needed to treat, number needed to harm and likelihood to be helped or harmed? Int J Clin Pract. 2014;68(12):1429-1441.
5. Winrow CJ, Gotter AL, Cox CD, et al. Promotion of sleep by suvorexant-a novel dual orexin receptor antagonist. J Neurogenet. 2011;25(1-2):52-61.
6. Saper CB, Chou TC, Scammell TE. The sleep switch: hypothalamic control of sleep and wakefulness. Trends Neurosci. 2001;24(12):726-731.
7. Sakurai T, Amemiya A, Ishii M, et al. Orexins and orexin receptors: a family of hypothalamic neuropeptides and G protein-coupled receptors that regulate feeding behavior. Cell. 1998;92(4):573-585.
8. Winrow CJ, Renger JJ. Discovery and development of orexin receptor antagonists as therapeutics for insomnia. Br J Pharmacol. 2014;171(2):283-293.
9. Belsomra [package insert]. Whitehouse Station, NJ: Merck; 2014.
10. Herring WJ, Snyder E, Budd K, et al. Orexin receptor antagonism for treatment of insomnia: a randomized clinical trial of suvorexant. Neurology. 2012;79(23):2265-2274.
11. Ivgy-May N, Snavely D, Minigh J, et al. Efficacy of suvorexant, an orexin receptor antagonist, in patients with primary insomnia: integrated results from 2 similarly designed phase 3 trials. Sleep. 2013;36(abstract supplement): A192.
12. Michelson D, Snyder E, Paradis E, et al. Safety and efficacy of suvorexant during 1-year treatment of insomnia with subsequent abrupt treatment discontinuation: a phase 3 randomised, double-blind, placebo-controlled trial. Lancet Neurol. 2014;13(5):461-471.
13. Merck Sharp and Dohme Corporation. Suvorexant advisory committee meeting briefing document. http:// www.fda.govdownloadsadvisorycommittees/committee smeetingmaterials/drugsperipheralandcentralnervous systemdrugsadvisorycommittee/ucm352970.pdf. Published May 22, 2013. Accessed November 24, 2014.

Issue
Current Psychiatry - 14(1)
Issue
Current Psychiatry - 14(1)
Page Number
19-24
Page Number
19-24
Publications
Publications
Topics
Article Type
Display Headline
Suvorexant for sleep-onset insomnia or sleep-maintenance insomnia, or both
Display Headline
Suvorexant for sleep-onset insomnia or sleep-maintenance insomnia, or both
Legacy Keywords
insomnia, sleep-onset insomnia, sleep-maintenance insomnia, survorexant, treatment for insomnia, insomnia treatment, sleep disorders
Legacy Keywords
insomnia, sleep-onset insomnia, sleep-maintenance insomnia, survorexant, treatment for insomnia, insomnia treatment, sleep disorders
Sections
Disallow All Ads
Alternative CME
Article PDF Media

Akathisia: Is restlessness a primary condition or an adverse drug effect?

Article Type
Changed
Tue, 12/11/2018 - 15:02
Display Headline
Akathisia: Is restlessness a primary condition or an adverse drug effect?

Akathisia—from the Greek for “inability to sit”—is a neuropsychiatric syndrome characterized by subjective and objective psychomotor restlessness. Patients typi­cally experience feelings of unease, inner restlessness mainly involving the legs, and a compulsion to move. Most engage in repetitive movement. They might swing or cross and uncross their legs, shift from one foot to the other, continuously pace, or persistently fidget.

In clinical settings, akathisia usually is a side effect of medi­cation. Antipsychotics, serotonin reuptake inhibitors, and buspirone are common triggers, but akathisia also has been associated with some antiemetics, preoperative sedatives, calcium channel blockers, and antivertigo agents. It also can be caused by withdrawal from an antipsychotic or related to a substance use disorder, especially cocaine. Akathisia can be acute or chronic, occurring in a tardive form with symptoms that last >6 months.1-3


Much isn’t known about drug-induced akathisia
Our understanding of the pathophysiology of akathisia is incomplete. Some have suggested that it results from an imbal­ance between the dopaminergic/cholinergic and dopaminer­gic/serotonergic systems4; others, that the cause is a mismatch between the core and the shell of the nucleus accumbens, due in part to overstimulation of the locus ceruleus.5

More recently, researchers established a positive asso­ciation between higher scores on the Liverpool University Neuroleptic Side Effects Rating Scale and D2/D3 receptor occupancy in the ventral striatum (nucleus accumbens and olfactory tubercle).6 The D2/D3 receptor occupancy model might explain withdrawal symptoms associated with cocaine,7 as well as rela­tive worsening of symptoms after tapering or discontinuing stimulants in attention-deficit/hyperactivity disorder (ADHD).


Elements of a clinical evaluation

When akathisia is suspected, evaluation by a clinician familiar with its phenom­enology is crucial. A validated tool, such as the Barnes Akathisia Rating Scale (at out cometracker.org/library/BAS.pdf) can aid in the detection and assessment of severity.8

In evaluating patients, keep in mind that the inner restlessness that characterizes akathisia can affect the trunk, hands, and arms, as well as the legs, and can cause dys­phoria and anxiety. Akathisia has been linked to an increased likelihood of developing sui­cidal ideation and behavior.9

Less common subjective symptoms include rage, fear, nausea, and worsening of psychotic symptoms. Because of its asso­ciation with aggression and agitation, drug-induced akathisia has been cited—with little success—as the basis for an insanity defense by people who have committed a violent act.10


Or is akathisia another psychiatric disorder?

Akathisia might go undetected for several reasons. One key factor: Its symptoms resem­ble and often overlap with those of other psy­chiatric disorders, such as mania, psychosis, agitated depression, and ADHD. In addition, akathisia often occurs concurrently with, and is masked by, akinesia, a common extrapy­ramidal side effect of many antipsychotics. Such patients might have the inner feeling of restlessness and urge to move but do not exhibit characteristic limb movements. In some cases, cognitive or intellectual limita­tions prevent patients from communicating the inner turmoil they feel.11

Medication nonadherence further compli­cates the picture, sometimes prompting a cli­nician to increase the dosage of the drug that is causing akathisia (Box 112).


Managing drug-induced akathisia
Akathisia usually resolves when the drug causing it is discontinued; decreasing the dosage might alleviate the symptoms. Whenever akathisia is detected, careful revision of the current drug regimen— substituting an antipsychotic with a lower prevalence of akathisia, for example— should be considered (Box 213-16). Treatment of drug-induced akathisia, which should be tailored to the patient’s psycho­pathology and comorbidities, is needed as well (Table17-25).



Beta blockers
, particularly propranolol, are considered first-line therapy for drug-induced akathisia, with a dosage of 20 to 40 mg twice daily used to relieve symptoms26 The effect can be explained by adrenergic terminals in the locus ceruleus and ending in the nucleus accumbens and prefrontal cor­tex stimulate β adrenoreceptors.5,27 Although multiple small studies and case reports26,28-32 support the use of beta blockers to treat drug-induced akathisia, the quality of evidence of their efficacy is controversial.12,21,27 Consider the risk of hypotension and bradycardia and be aware of contraindications for patients with asthma or diabetes.

Low-dose mirtazapine (15 mg/d) was found to be as effective as propranolol, 80 mg/d, in a placebo-controlled study, and to be more effective than a beta blocker in treating akathisia induced by a first-gener­ation antipsychotic. The authors concluded that both propranolol and mirtazapine should be first-line therapy.23 Others have suggested that these results be interpreted with caution because mirtazapine (at a higher dosage) has been linked to akathi­sia.33 Mirtazapine blocks α-adrenergic receptors, resulting in antagonism of 5-HT2 and 5-HT3 receptors and consequent enhancement of 5-HT1A serotonergic trans­mission.34 In one study, it was shown to reduce binding of the D2/D3 receptor ago­nist quinpirole.35

 

 

Serotonin antagonists and agonists. Blockade of 5-HT2 receptors can attenuate D2 blockade and mitigate akathisia symp­toms. Mianserin, 15 mg/d, can be helpful, and ritanserin, 5 to 20 mg/d, produced about a 50% reduction in akathisia symp­toms in 10 patients taking neuroleptics.36 Neither is available in the United States, however.

Cyproheptadine, a potent 5-HT2A and 5-HT2C antagonist with anticholinergic and antihistaminic action, improved akathisia symptoms in an open trial of 17 patients with antipsychotic-induced akathisia.37 The recommended dose is 8 to 16 mg/d.

A study using the selective inverse ago­nist pimavanserin (not FDA-approved) decreased akathisia in healthy volunteers taking haloperidol.14,24,33

Zolmitriptan, a 5-HT1D agonist, also can be used38; one study found that 7.5 mg/d of zolmitriptan is as effective as propranolol.39

A 2010 study showed a statistically signifi­cant improvement in 8 patients taking trazo­done, compared with 5 patients on placebo, all of whom met criteria for at least mild akathisia. Trazodone’s antiakathitic effect is attributed to its 5-HT2A antagonism.25

Anticholinergics. Traditionally, benztropine, biperiden, diphenhydramine, and trihexy­phenidyl have been used for prevention and treatment of extrapyramidal side effects. A Cochrane review concluded, however, that data are insufficient to support use of anticho­linergics for akathisia.40 Although multiple case reports have shown anticholinergics to be effective in treating drug-induced akathi­sia,12,17,33 their association with cognitive side effects suggests a need for caution.18

Benzodiazepines. Through their sedative and anxiolytic properties, benzodiazepines are thought to partially alleviate akathisia symptoms. Two small trials found clonaz­epam helpful for akathisia symptoms2,20; and 1 case report revealed that a patient with akathisia improved after coadministration of clonazepam and baclofen.41

Anticonvulsants. Valproic acid has not been found to be useful in antipsychotic-induced tardive akathisia.42 However, a case report described a patient with schizophrenia whose akathisia symptoms improved after the dosage of gabapentin was increased.43 Last, carbamazepine was found to be effec­tive in reducing akathisia symptoms in 3 patients with schizophrenia who were resis­tant to beta blockers, anticholinergics, anti­histaminergics, and benzodiazepines.19

α-adrenergic agonists. In an open trial, akathisia symptoms in 6 patients improved with clonidine, 0.2 to 0.8 mg/d.17 Speculation is that strong α1 antagonism might help pre­vent akathisia, which could be why this con­dition is not associated with iloperidone.44

D2 agonists. Akathisia and restless legs syndrome have similar pathophysiology,1,2 and patients with akathisia could ben­efit from D2 agonists such as cabergoline, pramipexole, rotigotine, and ropinirole. One case study revealed that a patient with aripiprazole-induced akathisia improved with ropinirole.45 D2 agonists can precipi­tate or worsen psychosis, however, and would be a relative contraindication in patients with psychotic disorders.22


Bottom Line

Failure to detect drug-induced akathisia can increase morbidity and delay recovery in patients undergoing psychiatric care. Knowing what to look for and how to tailor treatment to the needs of a given patient is an essential component of good care.

Related Resources
• Ferrando SJ, Eisendrath SJ. Adverse neuropsychiatric effects of dopamine antagonist medications. Misdiagnosis in the medical setting. Psychosomatics. 1991;32(4):426-432.
• Vinson DR. Diphenhydramine in the treatment of akathisia induced by prochlorperazine. J Emerg Med. 2004;26(3):265-270.


Drug Brand Names

Aripiprazole • Abilify                  Haloperidol • Haldol
Baclofen • Lioresal                     Iloperidone • Fanapt
Benztropine • Cogentin              Lurasidone • Latuda
Biperiden • Akineton                  Mirtazapine • Remeron
Buspirone • BuSpar                   Pramipexole • Mirapex
Cabergoline • Dostinex              Propranolol • Inderal
Carbamazepine • Tegretol          Quetiapine • Seroquel
Clonazepam • Klonopin              Ropinirole • Requip
Clonidine • Catapres                  Rotigotine • Neupro
Clozapine • Clozaril                    Trazodone • Desyrel, Oleptro
Cyproheptadine • Periactin          Trihexyphenidyl • Artane
Diphenhydramine • Benadryl       Valproic acid • Depakene
Gabapentin • Neurontin               Zolmitriptan • Zomig


Acknowledgement
Mandy Evans, MD, assisted with editing the manuscript of this article.

Disclosure
Dr. Forcen reports no financial relationships with any company whose products are mentioned in this article or with manufacturers of competing products.

References


1. Sachdev P. Akathisia and restless legs. Cambridge, United Kingdom: Cambridge University Press; 1995.
2. Sachdev P, Longragan C. The present status of akathisia. J Nerv Ment Dis. 1991;179(7):381-391.
3. Poyurovsky M, Hermesh H, Weizman A. Severe withdrawal akathisia following neuroleptic discontinuation successfully controlled by clozapine. Int Clin Psychopharmacol. 1996;11(4):283-286.
4. Poyurovsky M, Weizman A. Serotonin-based pharma-cotherapy for acute neuroleptic-induced akathisia: a new approach to an old problem. Br J Psychiatry. 2001;179:4-8.
5. Loonen AJ, Stahl SM. The mechanism of drug-induced akathisia. CNS Spectr. 2011;16(1):7-10.
6. Kim JH, Son YD, Kim HK, et al. Antipsychotic-associated mental side effects and their relationship to dopamine D2 receptor occupancy in striatal subdivisions: a high-resolution PET study with [11C]raclopride. J Clin Psychopharmacol. 2011;31(4):507-511.
7. Dailey JW, Fryer TD, Brichard L, et al. Nucleus accumbens D2/3 receptor predict trait impulsivity and cocaine reinforcement. Science. 2007;315(5816):1267-1270.
8. Barnes TR, Braude WM. Akathisia variants and tardive dyskinesia. Arch Gen Psychiatry. 1985;42(9):874-878.
9. Seemüller F, Schennach R, Mayr A, et al. Akathisia and suicidal ideation in first-episode schizophrenia. J Clin Psychopharmacol. 2012;32(5):694-698.
10. Leong GB, Silva JA. Neuroleptic-induced akathisia and violence: a review. J Forensic Sci. 2003;48(1):187-189.
11. Hirose S. The causes of underdiagnosing akathisia. Schizophr Bull. 2003;29(3):547-558.
12. Velligan DI, Weiden PJ, Sajatovic M, et al; Expert Consensus Panel on Adherence Problems in Serious and Persistent Mental Illness. The expert consensus guideline series: adherence problems in patients with serious and persistent mental illness. J Clin Psychiatry. 2009;70(suppl 4):S1-S46; quiz 47-48.
13. Citrome L. A review of the pharmacology, efficacy and tolerability of recently approved and upcoming oral antipsychotics: an evidence-based medicine approach. CNS Drugs. 2013;27(11):879-911.
14. Poyurovsky M. Acute antipsychotic-induced akathisia revisited. Br J Psychiatry. 2010;196(2):89-91.
15. Saltz BL, Robinson DG, Woerner MG. Recognizing and managing antipsychotic drug treatment side effects in the elderly. Prim Care Companion J Clin Psychiatry. 2004;6(suppl 2):14-19.
16. Lieberman JA, Stroup TS. The NIMH-CATIE Schizophrenia Study: what did we learn? Am J Psychiatry. 2011;168(8):770-775.
17. Zubenko GS, Cohen BM, Lipinski JF Jr, et al. Use of clonidine in treating neuroleptic-induced akathisia. Psychiatry Res. 1984;13(3):253-259.
18. Vinogradov S, Fisher M, Warm H, et al. The cognitive cost of anticholinergic burden: decreased response to cognitive training in schizophrenia. Am J Psychiatry. 2009;166(9):1055-1062.
19. Masui T, Kusumi I, Takahashi Y, et al. Efficacy of carbamazepine against neuroleptic-induced akathisia in treatment with perospirone: case series. Prog Neuropsychopharmacol Biol Psychiatry. 2005;29(2):343-346.
20. Lima AR, Soares-Weiser K, Bacaltchuk J, et al. Benzodiazepines for neuroleptic-induced acute akathisia. Cochrane Database Syst Rev. 2002;(1):CD001950.
21. Lima AR, Bacalcthuk J, Barnes TR, et al. Central action beta-blockers versus placebo for neuroleptic-induced acute akathisia. Cochrane Database Syst Rev. 2004;(4):CD001946.
22. Bilal L, Ching C. Cabergoline-induced psychosis in a patient with undiagnosed depression. J Neuropsychiatry Clin Neurosci. 2012;24(4):E54.
23. Poyurovsky M, Pashinian A, Weizman A, et al. Low-dose mirtazapine: a new option in the treatment of antipsychotic-induced akathisia. A randomized, double-blind, placebo- and propranolol-controlled trial. Biol Psychiatry.
2006;59(11):1071-1077.
24. Maidment I. Use of serotonin antagonists in the treatment of neuroleptic-induced akathisia. Psychiatric Bulletin. 2000;24(9):348-351.
25. Stryjer R, Rosenzcwaig S, Bar F, et al. Trazodone for the treatment of neuroleptic-induced akathisia: a placebo-controlled, double-blind, crossover study. Clin Neuropharmacol. 2010;33(5):219-222.
26. Dumon JP, Catteau J, Lanvin F, et al. Randomized, double-blind, crossover, placebo-controlled comparison of propranolol and betaxolol in the treatment of neuroleptic-induced akathisia. Am J Psychiatry. 1992;149(5):647-650.
27. van Waarde A, Vaalburg W, Doze P, et al. PET imaging of beta-adrenoceptors in the human brain: a realistic goal or a mirage? Curr Pharm Des. 2004;10(13):1519-1536.
28. Kurzthaler I, Hummer M, Kohl C, et al. Propranolol treatment of olanzapine-induced akathisia. Am J Psychiatry. 1997;154(9):1316.
29. Adler LA, Peselow E, Rosenthal MA, et al. A controlled comparison of the effects of propranolol, benztropine, and placebo on akathisia: an interim analysis. Psychopharmacol Bull. 1993;29(2):283-286.
30. Dorevitch A, Durst R, Ginath Y. Propranolol in the treatment of akathisia caused by antipsychotic drugs. South Med J. 1991;84(12):1505-1506.
31. Lipinski JF Jr, Zubenko GS, Cohen BM, et al. Propranolol in the treatment of neuroleptic-induced akathisia. Am J Psychiatry. 1984;141(3):412-415.
32. Adler L, Angrist B, Peselow E, et al. A controlled assessment of propranolol in the treatment of neuroleptic-induced akathisia. Br J Psychiatry. 1986;149:42-45.
33. Kumar R, Sachdev PS. Akathisia and second-generation antipsychotic drugs. Curr Opin Psychiatry. 2009;22(3):293-299.
34. Anttila SA, Leinonen EV. A review of the pharmacological and clinical profile of mirtazapine. CNS Drug Rev. 2001;7(3):249-264.
35. Rogóz Z, Wróbel A, Dlaboga D, et al. Effect of repeated treatment with mirtazapine on the central dopaminergic D2/D3 receptors. Pol J Pharmacol. 2002;54(4):381-389.
36. Miller CH, Fleischhacker WW, Ehrmann H, et al. Treatment of neuroleptic induced akathisia with the 5-HT2 antagonist ritanserin. Psychopharmacol Bull. 1990;26(3):373-376.
37. Weiss D, Aizenberg D, Hermesh H, et al. Cyproheptadine treatment in neuroleptic-induced akathisia. Br J Psychiatry. 1995;167(4):483-486.
38. Gross-Isseroff R, Magen A, Shiloh R, et al. The 5-HT1D receptor agonist zolmitriptan for neuroleptic-induced akathisia: an open label preliminary study. Int Clin Psychopharmacol. 2005;20(1):23-25.
39. Avital A, Gross-Isseroff R, Stryjer R, et al. Zolmitriptan compared to propranolol in the treatment of acute neuroleptic-induced akathisia: a comparative double-blind study. Eur Neuropsychopharmacol. 2009;19(7):476-482.
40. Rathbone J, Soares-Weiser K. Anticholinergics for neuroleptic-induced acute akathisia. Cochrane Database Syst Rev. 2006;(4):CD003727.
41. Sandyk R. Successful treatment of neuroleptic-induced akathisia with baclofen and clonazepam. A case report. Eur Neurol. 1985;24(4):286-288.
42. Miller CH, Fleischhacker W. Managing antipsychotic-induced acute and chronic akathisia. Drug Saf. 2000;22(1):73-81.
43. Pfeffer G, Chouinard G, Margolese HC. Gabapentin in the treatment of antipsychotic-induced akathisia in schizophrenia. Int Clin Psychopharmacol. 2005;20(3):179-181.
44. Stahl SM. Role of α1 adrenergic antagonism in the mechanism of action of iloperidone: reducing extrapyramidal symptoms. CNS Spectr. 2013;18(6):285-258.
45. Hettema JM, Ross DE. A case of aripiprazole-related tardive akathisia and its treatment with ropinirole. J Clin Psychiatry. 2007;68(11):1814-1815.

Article PDF
Author and Disclosure Information

Fernando Espi Forcen, MD
Fellow, Psychosomatic Medicine
Department of Psychiatry and Behavioral Sciences
Memorial Sloan Kettering Cancer Center
New York, New York

Issue
Current Psychiatry - 14(1)
Publications
Topics
Page Number
14-18
Legacy Keywords
antipsychotics, akathisia, adverse effects, restlessness, drug-induced akathisia
Sections
Author and Disclosure Information

Fernando Espi Forcen, MD
Fellow, Psychosomatic Medicine
Department of Psychiatry and Behavioral Sciences
Memorial Sloan Kettering Cancer Center
New York, New York

Author and Disclosure Information

Fernando Espi Forcen, MD
Fellow, Psychosomatic Medicine
Department of Psychiatry and Behavioral Sciences
Memorial Sloan Kettering Cancer Center
New York, New York

Article PDF
Article PDF

Akathisia—from the Greek for “inability to sit”—is a neuropsychiatric syndrome characterized by subjective and objective psychomotor restlessness. Patients typi­cally experience feelings of unease, inner restlessness mainly involving the legs, and a compulsion to move. Most engage in repetitive movement. They might swing or cross and uncross their legs, shift from one foot to the other, continuously pace, or persistently fidget.

In clinical settings, akathisia usually is a side effect of medi­cation. Antipsychotics, serotonin reuptake inhibitors, and buspirone are common triggers, but akathisia also has been associated with some antiemetics, preoperative sedatives, calcium channel blockers, and antivertigo agents. It also can be caused by withdrawal from an antipsychotic or related to a substance use disorder, especially cocaine. Akathisia can be acute or chronic, occurring in a tardive form with symptoms that last >6 months.1-3


Much isn’t known about drug-induced akathisia
Our understanding of the pathophysiology of akathisia is incomplete. Some have suggested that it results from an imbal­ance between the dopaminergic/cholinergic and dopaminer­gic/serotonergic systems4; others, that the cause is a mismatch between the core and the shell of the nucleus accumbens, due in part to overstimulation of the locus ceruleus.5

More recently, researchers established a positive asso­ciation between higher scores on the Liverpool University Neuroleptic Side Effects Rating Scale and D2/D3 receptor occupancy in the ventral striatum (nucleus accumbens and olfactory tubercle).6 The D2/D3 receptor occupancy model might explain withdrawal symptoms associated with cocaine,7 as well as rela­tive worsening of symptoms after tapering or discontinuing stimulants in attention-deficit/hyperactivity disorder (ADHD).


Elements of a clinical evaluation

When akathisia is suspected, evaluation by a clinician familiar with its phenom­enology is crucial. A validated tool, such as the Barnes Akathisia Rating Scale (at out cometracker.org/library/BAS.pdf) can aid in the detection and assessment of severity.8

In evaluating patients, keep in mind that the inner restlessness that characterizes akathisia can affect the trunk, hands, and arms, as well as the legs, and can cause dys­phoria and anxiety. Akathisia has been linked to an increased likelihood of developing sui­cidal ideation and behavior.9

Less common subjective symptoms include rage, fear, nausea, and worsening of psychotic symptoms. Because of its asso­ciation with aggression and agitation, drug-induced akathisia has been cited—with little success—as the basis for an insanity defense by people who have committed a violent act.10


Or is akathisia another psychiatric disorder?

Akathisia might go undetected for several reasons. One key factor: Its symptoms resem­ble and often overlap with those of other psy­chiatric disorders, such as mania, psychosis, agitated depression, and ADHD. In addition, akathisia often occurs concurrently with, and is masked by, akinesia, a common extrapy­ramidal side effect of many antipsychotics. Such patients might have the inner feeling of restlessness and urge to move but do not exhibit characteristic limb movements. In some cases, cognitive or intellectual limita­tions prevent patients from communicating the inner turmoil they feel.11

Medication nonadherence further compli­cates the picture, sometimes prompting a cli­nician to increase the dosage of the drug that is causing akathisia (Box 112).


Managing drug-induced akathisia
Akathisia usually resolves when the drug causing it is discontinued; decreasing the dosage might alleviate the symptoms. Whenever akathisia is detected, careful revision of the current drug regimen— substituting an antipsychotic with a lower prevalence of akathisia, for example— should be considered (Box 213-16). Treatment of drug-induced akathisia, which should be tailored to the patient’s psycho­pathology and comorbidities, is needed as well (Table17-25).



Beta blockers
, particularly propranolol, are considered first-line therapy for drug-induced akathisia, with a dosage of 20 to 40 mg twice daily used to relieve symptoms26 The effect can be explained by adrenergic terminals in the locus ceruleus and ending in the nucleus accumbens and prefrontal cor­tex stimulate β adrenoreceptors.5,27 Although multiple small studies and case reports26,28-32 support the use of beta blockers to treat drug-induced akathisia, the quality of evidence of their efficacy is controversial.12,21,27 Consider the risk of hypotension and bradycardia and be aware of contraindications for patients with asthma or diabetes.

Low-dose mirtazapine (15 mg/d) was found to be as effective as propranolol, 80 mg/d, in a placebo-controlled study, and to be more effective than a beta blocker in treating akathisia induced by a first-gener­ation antipsychotic. The authors concluded that both propranolol and mirtazapine should be first-line therapy.23 Others have suggested that these results be interpreted with caution because mirtazapine (at a higher dosage) has been linked to akathi­sia.33 Mirtazapine blocks α-adrenergic receptors, resulting in antagonism of 5-HT2 and 5-HT3 receptors and consequent enhancement of 5-HT1A serotonergic trans­mission.34 In one study, it was shown to reduce binding of the D2/D3 receptor ago­nist quinpirole.35

 

 

Serotonin antagonists and agonists. Blockade of 5-HT2 receptors can attenuate D2 blockade and mitigate akathisia symp­toms. Mianserin, 15 mg/d, can be helpful, and ritanserin, 5 to 20 mg/d, produced about a 50% reduction in akathisia symp­toms in 10 patients taking neuroleptics.36 Neither is available in the United States, however.

Cyproheptadine, a potent 5-HT2A and 5-HT2C antagonist with anticholinergic and antihistaminic action, improved akathisia symptoms in an open trial of 17 patients with antipsychotic-induced akathisia.37 The recommended dose is 8 to 16 mg/d.

A study using the selective inverse ago­nist pimavanserin (not FDA-approved) decreased akathisia in healthy volunteers taking haloperidol.14,24,33

Zolmitriptan, a 5-HT1D agonist, also can be used38; one study found that 7.5 mg/d of zolmitriptan is as effective as propranolol.39

A 2010 study showed a statistically signifi­cant improvement in 8 patients taking trazo­done, compared with 5 patients on placebo, all of whom met criteria for at least mild akathisia. Trazodone’s antiakathitic effect is attributed to its 5-HT2A antagonism.25

Anticholinergics. Traditionally, benztropine, biperiden, diphenhydramine, and trihexy­phenidyl have been used for prevention and treatment of extrapyramidal side effects. A Cochrane review concluded, however, that data are insufficient to support use of anticho­linergics for akathisia.40 Although multiple case reports have shown anticholinergics to be effective in treating drug-induced akathi­sia,12,17,33 their association with cognitive side effects suggests a need for caution.18

Benzodiazepines. Through their sedative and anxiolytic properties, benzodiazepines are thought to partially alleviate akathisia symptoms. Two small trials found clonaz­epam helpful for akathisia symptoms2,20; and 1 case report revealed that a patient with akathisia improved after coadministration of clonazepam and baclofen.41

Anticonvulsants. Valproic acid has not been found to be useful in antipsychotic-induced tardive akathisia.42 However, a case report described a patient with schizophrenia whose akathisia symptoms improved after the dosage of gabapentin was increased.43 Last, carbamazepine was found to be effec­tive in reducing akathisia symptoms in 3 patients with schizophrenia who were resis­tant to beta blockers, anticholinergics, anti­histaminergics, and benzodiazepines.19

α-adrenergic agonists. In an open trial, akathisia symptoms in 6 patients improved with clonidine, 0.2 to 0.8 mg/d.17 Speculation is that strong α1 antagonism might help pre­vent akathisia, which could be why this con­dition is not associated with iloperidone.44

D2 agonists. Akathisia and restless legs syndrome have similar pathophysiology,1,2 and patients with akathisia could ben­efit from D2 agonists such as cabergoline, pramipexole, rotigotine, and ropinirole. One case study revealed that a patient with aripiprazole-induced akathisia improved with ropinirole.45 D2 agonists can precipi­tate or worsen psychosis, however, and would be a relative contraindication in patients with psychotic disorders.22


Bottom Line

Failure to detect drug-induced akathisia can increase morbidity and delay recovery in patients undergoing psychiatric care. Knowing what to look for and how to tailor treatment to the needs of a given patient is an essential component of good care.

Related Resources
• Ferrando SJ, Eisendrath SJ. Adverse neuropsychiatric effects of dopamine antagonist medications. Misdiagnosis in the medical setting. Psychosomatics. 1991;32(4):426-432.
• Vinson DR. Diphenhydramine in the treatment of akathisia induced by prochlorperazine. J Emerg Med. 2004;26(3):265-270.


Drug Brand Names

Aripiprazole • Abilify                  Haloperidol • Haldol
Baclofen • Lioresal                     Iloperidone • Fanapt
Benztropine • Cogentin              Lurasidone • Latuda
Biperiden • Akineton                  Mirtazapine • Remeron
Buspirone • BuSpar                   Pramipexole • Mirapex
Cabergoline • Dostinex              Propranolol • Inderal
Carbamazepine • Tegretol          Quetiapine • Seroquel
Clonazepam • Klonopin              Ropinirole • Requip
Clonidine • Catapres                  Rotigotine • Neupro
Clozapine • Clozaril                    Trazodone • Desyrel, Oleptro
Cyproheptadine • Periactin          Trihexyphenidyl • Artane
Diphenhydramine • Benadryl       Valproic acid • Depakene
Gabapentin • Neurontin               Zolmitriptan • Zomig


Acknowledgement
Mandy Evans, MD, assisted with editing the manuscript of this article.

Disclosure
Dr. Forcen reports no financial relationships with any company whose products are mentioned in this article or with manufacturers of competing products.

Akathisia—from the Greek for “inability to sit”—is a neuropsychiatric syndrome characterized by subjective and objective psychomotor restlessness. Patients typi­cally experience feelings of unease, inner restlessness mainly involving the legs, and a compulsion to move. Most engage in repetitive movement. They might swing or cross and uncross their legs, shift from one foot to the other, continuously pace, or persistently fidget.

In clinical settings, akathisia usually is a side effect of medi­cation. Antipsychotics, serotonin reuptake inhibitors, and buspirone are common triggers, but akathisia also has been associated with some antiemetics, preoperative sedatives, calcium channel blockers, and antivertigo agents. It also can be caused by withdrawal from an antipsychotic or related to a substance use disorder, especially cocaine. Akathisia can be acute or chronic, occurring in a tardive form with symptoms that last >6 months.1-3


Much isn’t known about drug-induced akathisia
Our understanding of the pathophysiology of akathisia is incomplete. Some have suggested that it results from an imbal­ance between the dopaminergic/cholinergic and dopaminer­gic/serotonergic systems4; others, that the cause is a mismatch between the core and the shell of the nucleus accumbens, due in part to overstimulation of the locus ceruleus.5

More recently, researchers established a positive asso­ciation between higher scores on the Liverpool University Neuroleptic Side Effects Rating Scale and D2/D3 receptor occupancy in the ventral striatum (nucleus accumbens and olfactory tubercle).6 The D2/D3 receptor occupancy model might explain withdrawal symptoms associated with cocaine,7 as well as rela­tive worsening of symptoms after tapering or discontinuing stimulants in attention-deficit/hyperactivity disorder (ADHD).


Elements of a clinical evaluation

When akathisia is suspected, evaluation by a clinician familiar with its phenom­enology is crucial. A validated tool, such as the Barnes Akathisia Rating Scale (at out cometracker.org/library/BAS.pdf) can aid in the detection and assessment of severity.8

In evaluating patients, keep in mind that the inner restlessness that characterizes akathisia can affect the trunk, hands, and arms, as well as the legs, and can cause dys­phoria and anxiety. Akathisia has been linked to an increased likelihood of developing sui­cidal ideation and behavior.9

Less common subjective symptoms include rage, fear, nausea, and worsening of psychotic symptoms. Because of its asso­ciation with aggression and agitation, drug-induced akathisia has been cited—with little success—as the basis for an insanity defense by people who have committed a violent act.10


Or is akathisia another psychiatric disorder?

Akathisia might go undetected for several reasons. One key factor: Its symptoms resem­ble and often overlap with those of other psy­chiatric disorders, such as mania, psychosis, agitated depression, and ADHD. In addition, akathisia often occurs concurrently with, and is masked by, akinesia, a common extrapy­ramidal side effect of many antipsychotics. Such patients might have the inner feeling of restlessness and urge to move but do not exhibit characteristic limb movements. In some cases, cognitive or intellectual limita­tions prevent patients from communicating the inner turmoil they feel.11

Medication nonadherence further compli­cates the picture, sometimes prompting a cli­nician to increase the dosage of the drug that is causing akathisia (Box 112).


Managing drug-induced akathisia
Akathisia usually resolves when the drug causing it is discontinued; decreasing the dosage might alleviate the symptoms. Whenever akathisia is detected, careful revision of the current drug regimen— substituting an antipsychotic with a lower prevalence of akathisia, for example— should be considered (Box 213-16). Treatment of drug-induced akathisia, which should be tailored to the patient’s psycho­pathology and comorbidities, is needed as well (Table17-25).



Beta blockers
, particularly propranolol, are considered first-line therapy for drug-induced akathisia, with a dosage of 20 to 40 mg twice daily used to relieve symptoms26 The effect can be explained by adrenergic terminals in the locus ceruleus and ending in the nucleus accumbens and prefrontal cor­tex stimulate β adrenoreceptors.5,27 Although multiple small studies and case reports26,28-32 support the use of beta blockers to treat drug-induced akathisia, the quality of evidence of their efficacy is controversial.12,21,27 Consider the risk of hypotension and bradycardia and be aware of contraindications for patients with asthma or diabetes.

Low-dose mirtazapine (15 mg/d) was found to be as effective as propranolol, 80 mg/d, in a placebo-controlled study, and to be more effective than a beta blocker in treating akathisia induced by a first-gener­ation antipsychotic. The authors concluded that both propranolol and mirtazapine should be first-line therapy.23 Others have suggested that these results be interpreted with caution because mirtazapine (at a higher dosage) has been linked to akathi­sia.33 Mirtazapine blocks α-adrenergic receptors, resulting in antagonism of 5-HT2 and 5-HT3 receptors and consequent enhancement of 5-HT1A serotonergic trans­mission.34 In one study, it was shown to reduce binding of the D2/D3 receptor ago­nist quinpirole.35

 

 

Serotonin antagonists and agonists. Blockade of 5-HT2 receptors can attenuate D2 blockade and mitigate akathisia symp­toms. Mianserin, 15 mg/d, can be helpful, and ritanserin, 5 to 20 mg/d, produced about a 50% reduction in akathisia symp­toms in 10 patients taking neuroleptics.36 Neither is available in the United States, however.

Cyproheptadine, a potent 5-HT2A and 5-HT2C antagonist with anticholinergic and antihistaminic action, improved akathisia symptoms in an open trial of 17 patients with antipsychotic-induced akathisia.37 The recommended dose is 8 to 16 mg/d.

A study using the selective inverse ago­nist pimavanserin (not FDA-approved) decreased akathisia in healthy volunteers taking haloperidol.14,24,33

Zolmitriptan, a 5-HT1D agonist, also can be used38; one study found that 7.5 mg/d of zolmitriptan is as effective as propranolol.39

A 2010 study showed a statistically signifi­cant improvement in 8 patients taking trazo­done, compared with 5 patients on placebo, all of whom met criteria for at least mild akathisia. Trazodone’s antiakathitic effect is attributed to its 5-HT2A antagonism.25

Anticholinergics. Traditionally, benztropine, biperiden, diphenhydramine, and trihexy­phenidyl have been used for prevention and treatment of extrapyramidal side effects. A Cochrane review concluded, however, that data are insufficient to support use of anticho­linergics for akathisia.40 Although multiple case reports have shown anticholinergics to be effective in treating drug-induced akathi­sia,12,17,33 their association with cognitive side effects suggests a need for caution.18

Benzodiazepines. Through their sedative and anxiolytic properties, benzodiazepines are thought to partially alleviate akathisia symptoms. Two small trials found clonaz­epam helpful for akathisia symptoms2,20; and 1 case report revealed that a patient with akathisia improved after coadministration of clonazepam and baclofen.41

Anticonvulsants. Valproic acid has not been found to be useful in antipsychotic-induced tardive akathisia.42 However, a case report described a patient with schizophrenia whose akathisia symptoms improved after the dosage of gabapentin was increased.43 Last, carbamazepine was found to be effec­tive in reducing akathisia symptoms in 3 patients with schizophrenia who were resis­tant to beta blockers, anticholinergics, anti­histaminergics, and benzodiazepines.19

α-adrenergic agonists. In an open trial, akathisia symptoms in 6 patients improved with clonidine, 0.2 to 0.8 mg/d.17 Speculation is that strong α1 antagonism might help pre­vent akathisia, which could be why this con­dition is not associated with iloperidone.44

D2 agonists. Akathisia and restless legs syndrome have similar pathophysiology,1,2 and patients with akathisia could ben­efit from D2 agonists such as cabergoline, pramipexole, rotigotine, and ropinirole. One case study revealed that a patient with aripiprazole-induced akathisia improved with ropinirole.45 D2 agonists can precipi­tate or worsen psychosis, however, and would be a relative contraindication in patients with psychotic disorders.22


Bottom Line

Failure to detect drug-induced akathisia can increase morbidity and delay recovery in patients undergoing psychiatric care. Knowing what to look for and how to tailor treatment to the needs of a given patient is an essential component of good care.

Related Resources
• Ferrando SJ, Eisendrath SJ. Adverse neuropsychiatric effects of dopamine antagonist medications. Misdiagnosis in the medical setting. Psychosomatics. 1991;32(4):426-432.
• Vinson DR. Diphenhydramine in the treatment of akathisia induced by prochlorperazine. J Emerg Med. 2004;26(3):265-270.


Drug Brand Names

Aripiprazole • Abilify                  Haloperidol • Haldol
Baclofen • Lioresal                     Iloperidone • Fanapt
Benztropine • Cogentin              Lurasidone • Latuda
Biperiden • Akineton                  Mirtazapine • Remeron
Buspirone • BuSpar                   Pramipexole • Mirapex
Cabergoline • Dostinex              Propranolol • Inderal
Carbamazepine • Tegretol          Quetiapine • Seroquel
Clonazepam • Klonopin              Ropinirole • Requip
Clonidine • Catapres                  Rotigotine • Neupro
Clozapine • Clozaril                    Trazodone • Desyrel, Oleptro
Cyproheptadine • Periactin          Trihexyphenidyl • Artane
Diphenhydramine • Benadryl       Valproic acid • Depakene
Gabapentin • Neurontin               Zolmitriptan • Zomig


Acknowledgement
Mandy Evans, MD, assisted with editing the manuscript of this article.

Disclosure
Dr. Forcen reports no financial relationships with any company whose products are mentioned in this article or with manufacturers of competing products.

References


1. Sachdev P. Akathisia and restless legs. Cambridge, United Kingdom: Cambridge University Press; 1995.
2. Sachdev P, Longragan C. The present status of akathisia. J Nerv Ment Dis. 1991;179(7):381-391.
3. Poyurovsky M, Hermesh H, Weizman A. Severe withdrawal akathisia following neuroleptic discontinuation successfully controlled by clozapine. Int Clin Psychopharmacol. 1996;11(4):283-286.
4. Poyurovsky M, Weizman A. Serotonin-based pharma-cotherapy for acute neuroleptic-induced akathisia: a new approach to an old problem. Br J Psychiatry. 2001;179:4-8.
5. Loonen AJ, Stahl SM. The mechanism of drug-induced akathisia. CNS Spectr. 2011;16(1):7-10.
6. Kim JH, Son YD, Kim HK, et al. Antipsychotic-associated mental side effects and their relationship to dopamine D2 receptor occupancy in striatal subdivisions: a high-resolution PET study with [11C]raclopride. J Clin Psychopharmacol. 2011;31(4):507-511.
7. Dailey JW, Fryer TD, Brichard L, et al. Nucleus accumbens D2/3 receptor predict trait impulsivity and cocaine reinforcement. Science. 2007;315(5816):1267-1270.
8. Barnes TR, Braude WM. Akathisia variants and tardive dyskinesia. Arch Gen Psychiatry. 1985;42(9):874-878.
9. Seemüller F, Schennach R, Mayr A, et al. Akathisia and suicidal ideation in first-episode schizophrenia. J Clin Psychopharmacol. 2012;32(5):694-698.
10. Leong GB, Silva JA. Neuroleptic-induced akathisia and violence: a review. J Forensic Sci. 2003;48(1):187-189.
11. Hirose S. The causes of underdiagnosing akathisia. Schizophr Bull. 2003;29(3):547-558.
12. Velligan DI, Weiden PJ, Sajatovic M, et al; Expert Consensus Panel on Adherence Problems in Serious and Persistent Mental Illness. The expert consensus guideline series: adherence problems in patients with serious and persistent mental illness. J Clin Psychiatry. 2009;70(suppl 4):S1-S46; quiz 47-48.
13. Citrome L. A review of the pharmacology, efficacy and tolerability of recently approved and upcoming oral antipsychotics: an evidence-based medicine approach. CNS Drugs. 2013;27(11):879-911.
14. Poyurovsky M. Acute antipsychotic-induced akathisia revisited. Br J Psychiatry. 2010;196(2):89-91.
15. Saltz BL, Robinson DG, Woerner MG. Recognizing and managing antipsychotic drug treatment side effects in the elderly. Prim Care Companion J Clin Psychiatry. 2004;6(suppl 2):14-19.
16. Lieberman JA, Stroup TS. The NIMH-CATIE Schizophrenia Study: what did we learn? Am J Psychiatry. 2011;168(8):770-775.
17. Zubenko GS, Cohen BM, Lipinski JF Jr, et al. Use of clonidine in treating neuroleptic-induced akathisia. Psychiatry Res. 1984;13(3):253-259.
18. Vinogradov S, Fisher M, Warm H, et al. The cognitive cost of anticholinergic burden: decreased response to cognitive training in schizophrenia. Am J Psychiatry. 2009;166(9):1055-1062.
19. Masui T, Kusumi I, Takahashi Y, et al. Efficacy of carbamazepine against neuroleptic-induced akathisia in treatment with perospirone: case series. Prog Neuropsychopharmacol Biol Psychiatry. 2005;29(2):343-346.
20. Lima AR, Soares-Weiser K, Bacaltchuk J, et al. Benzodiazepines for neuroleptic-induced acute akathisia. Cochrane Database Syst Rev. 2002;(1):CD001950.
21. Lima AR, Bacalcthuk J, Barnes TR, et al. Central action beta-blockers versus placebo for neuroleptic-induced acute akathisia. Cochrane Database Syst Rev. 2004;(4):CD001946.
22. Bilal L, Ching C. Cabergoline-induced psychosis in a patient with undiagnosed depression. J Neuropsychiatry Clin Neurosci. 2012;24(4):E54.
23. Poyurovsky M, Pashinian A, Weizman A, et al. Low-dose mirtazapine: a new option in the treatment of antipsychotic-induced akathisia. A randomized, double-blind, placebo- and propranolol-controlled trial. Biol Psychiatry.
2006;59(11):1071-1077.
24. Maidment I. Use of serotonin antagonists in the treatment of neuroleptic-induced akathisia. Psychiatric Bulletin. 2000;24(9):348-351.
25. Stryjer R, Rosenzcwaig S, Bar F, et al. Trazodone for the treatment of neuroleptic-induced akathisia: a placebo-controlled, double-blind, crossover study. Clin Neuropharmacol. 2010;33(5):219-222.
26. Dumon JP, Catteau J, Lanvin F, et al. Randomized, double-blind, crossover, placebo-controlled comparison of propranolol and betaxolol in the treatment of neuroleptic-induced akathisia. Am J Psychiatry. 1992;149(5):647-650.
27. van Waarde A, Vaalburg W, Doze P, et al. PET imaging of beta-adrenoceptors in the human brain: a realistic goal or a mirage? Curr Pharm Des. 2004;10(13):1519-1536.
28. Kurzthaler I, Hummer M, Kohl C, et al. Propranolol treatment of olanzapine-induced akathisia. Am J Psychiatry. 1997;154(9):1316.
29. Adler LA, Peselow E, Rosenthal MA, et al. A controlled comparison of the effects of propranolol, benztropine, and placebo on akathisia: an interim analysis. Psychopharmacol Bull. 1993;29(2):283-286.
30. Dorevitch A, Durst R, Ginath Y. Propranolol in the treatment of akathisia caused by antipsychotic drugs. South Med J. 1991;84(12):1505-1506.
31. Lipinski JF Jr, Zubenko GS, Cohen BM, et al. Propranolol in the treatment of neuroleptic-induced akathisia. Am J Psychiatry. 1984;141(3):412-415.
32. Adler L, Angrist B, Peselow E, et al. A controlled assessment of propranolol in the treatment of neuroleptic-induced akathisia. Br J Psychiatry. 1986;149:42-45.
33. Kumar R, Sachdev PS. Akathisia and second-generation antipsychotic drugs. Curr Opin Psychiatry. 2009;22(3):293-299.
34. Anttila SA, Leinonen EV. A review of the pharmacological and clinical profile of mirtazapine. CNS Drug Rev. 2001;7(3):249-264.
35. Rogóz Z, Wróbel A, Dlaboga D, et al. Effect of repeated treatment with mirtazapine on the central dopaminergic D2/D3 receptors. Pol J Pharmacol. 2002;54(4):381-389.
36. Miller CH, Fleischhacker WW, Ehrmann H, et al. Treatment of neuroleptic induced akathisia with the 5-HT2 antagonist ritanserin. Psychopharmacol Bull. 1990;26(3):373-376.
37. Weiss D, Aizenberg D, Hermesh H, et al. Cyproheptadine treatment in neuroleptic-induced akathisia. Br J Psychiatry. 1995;167(4):483-486.
38. Gross-Isseroff R, Magen A, Shiloh R, et al. The 5-HT1D receptor agonist zolmitriptan for neuroleptic-induced akathisia: an open label preliminary study. Int Clin Psychopharmacol. 2005;20(1):23-25.
39. Avital A, Gross-Isseroff R, Stryjer R, et al. Zolmitriptan compared to propranolol in the treatment of acute neuroleptic-induced akathisia: a comparative double-blind study. Eur Neuropsychopharmacol. 2009;19(7):476-482.
40. Rathbone J, Soares-Weiser K. Anticholinergics for neuroleptic-induced acute akathisia. Cochrane Database Syst Rev. 2006;(4):CD003727.
41. Sandyk R. Successful treatment of neuroleptic-induced akathisia with baclofen and clonazepam. A case report. Eur Neurol. 1985;24(4):286-288.
42. Miller CH, Fleischhacker W. Managing antipsychotic-induced acute and chronic akathisia. Drug Saf. 2000;22(1):73-81.
43. Pfeffer G, Chouinard G, Margolese HC. Gabapentin in the treatment of antipsychotic-induced akathisia in schizophrenia. Int Clin Psychopharmacol. 2005;20(3):179-181.
44. Stahl SM. Role of α1 adrenergic antagonism in the mechanism of action of iloperidone: reducing extrapyramidal symptoms. CNS Spectr. 2013;18(6):285-258.
45. Hettema JM, Ross DE. A case of aripiprazole-related tardive akathisia and its treatment with ropinirole. J Clin Psychiatry. 2007;68(11):1814-1815.

References


1. Sachdev P. Akathisia and restless legs. Cambridge, United Kingdom: Cambridge University Press; 1995.
2. Sachdev P, Longragan C. The present status of akathisia. J Nerv Ment Dis. 1991;179(7):381-391.
3. Poyurovsky M, Hermesh H, Weizman A. Severe withdrawal akathisia following neuroleptic discontinuation successfully controlled by clozapine. Int Clin Psychopharmacol. 1996;11(4):283-286.
4. Poyurovsky M, Weizman A. Serotonin-based pharma-cotherapy for acute neuroleptic-induced akathisia: a new approach to an old problem. Br J Psychiatry. 2001;179:4-8.
5. Loonen AJ, Stahl SM. The mechanism of drug-induced akathisia. CNS Spectr. 2011;16(1):7-10.
6. Kim JH, Son YD, Kim HK, et al. Antipsychotic-associated mental side effects and their relationship to dopamine D2 receptor occupancy in striatal subdivisions: a high-resolution PET study with [11C]raclopride. J Clin Psychopharmacol. 2011;31(4):507-511.
7. Dailey JW, Fryer TD, Brichard L, et al. Nucleus accumbens D2/3 receptor predict trait impulsivity and cocaine reinforcement. Science. 2007;315(5816):1267-1270.
8. Barnes TR, Braude WM. Akathisia variants and tardive dyskinesia. Arch Gen Psychiatry. 1985;42(9):874-878.
9. Seemüller F, Schennach R, Mayr A, et al. Akathisia and suicidal ideation in first-episode schizophrenia. J Clin Psychopharmacol. 2012;32(5):694-698.
10. Leong GB, Silva JA. Neuroleptic-induced akathisia and violence: a review. J Forensic Sci. 2003;48(1):187-189.
11. Hirose S. The causes of underdiagnosing akathisia. Schizophr Bull. 2003;29(3):547-558.
12. Velligan DI, Weiden PJ, Sajatovic M, et al; Expert Consensus Panel on Adherence Problems in Serious and Persistent Mental Illness. The expert consensus guideline series: adherence problems in patients with serious and persistent mental illness. J Clin Psychiatry. 2009;70(suppl 4):S1-S46; quiz 47-48.
13. Citrome L. A review of the pharmacology, efficacy and tolerability of recently approved and upcoming oral antipsychotics: an evidence-based medicine approach. CNS Drugs. 2013;27(11):879-911.
14. Poyurovsky M. Acute antipsychotic-induced akathisia revisited. Br J Psychiatry. 2010;196(2):89-91.
15. Saltz BL, Robinson DG, Woerner MG. Recognizing and managing antipsychotic drug treatment side effects in the elderly. Prim Care Companion J Clin Psychiatry. 2004;6(suppl 2):14-19.
16. Lieberman JA, Stroup TS. The NIMH-CATIE Schizophrenia Study: what did we learn? Am J Psychiatry. 2011;168(8):770-775.
17. Zubenko GS, Cohen BM, Lipinski JF Jr, et al. Use of clonidine in treating neuroleptic-induced akathisia. Psychiatry Res. 1984;13(3):253-259.
18. Vinogradov S, Fisher M, Warm H, et al. The cognitive cost of anticholinergic burden: decreased response to cognitive training in schizophrenia. Am J Psychiatry. 2009;166(9):1055-1062.
19. Masui T, Kusumi I, Takahashi Y, et al. Efficacy of carbamazepine against neuroleptic-induced akathisia in treatment with perospirone: case series. Prog Neuropsychopharmacol Biol Psychiatry. 2005;29(2):343-346.
20. Lima AR, Soares-Weiser K, Bacaltchuk J, et al. Benzodiazepines for neuroleptic-induced acute akathisia. Cochrane Database Syst Rev. 2002;(1):CD001950.
21. Lima AR, Bacalcthuk J, Barnes TR, et al. Central action beta-blockers versus placebo for neuroleptic-induced acute akathisia. Cochrane Database Syst Rev. 2004;(4):CD001946.
22. Bilal L, Ching C. Cabergoline-induced psychosis in a patient with undiagnosed depression. J Neuropsychiatry Clin Neurosci. 2012;24(4):E54.
23. Poyurovsky M, Pashinian A, Weizman A, et al. Low-dose mirtazapine: a new option in the treatment of antipsychotic-induced akathisia. A randomized, double-blind, placebo- and propranolol-controlled trial. Biol Psychiatry.
2006;59(11):1071-1077.
24. Maidment I. Use of serotonin antagonists in the treatment of neuroleptic-induced akathisia. Psychiatric Bulletin. 2000;24(9):348-351.
25. Stryjer R, Rosenzcwaig S, Bar F, et al. Trazodone for the treatment of neuroleptic-induced akathisia: a placebo-controlled, double-blind, crossover study. Clin Neuropharmacol. 2010;33(5):219-222.
26. Dumon JP, Catteau J, Lanvin F, et al. Randomized, double-blind, crossover, placebo-controlled comparison of propranolol and betaxolol in the treatment of neuroleptic-induced akathisia. Am J Psychiatry. 1992;149(5):647-650.
27. van Waarde A, Vaalburg W, Doze P, et al. PET imaging of beta-adrenoceptors in the human brain: a realistic goal or a mirage? Curr Pharm Des. 2004;10(13):1519-1536.
28. Kurzthaler I, Hummer M, Kohl C, et al. Propranolol treatment of olanzapine-induced akathisia. Am J Psychiatry. 1997;154(9):1316.
29. Adler LA, Peselow E, Rosenthal MA, et al. A controlled comparison of the effects of propranolol, benztropine, and placebo on akathisia: an interim analysis. Psychopharmacol Bull. 1993;29(2):283-286.
30. Dorevitch A, Durst R, Ginath Y. Propranolol in the treatment of akathisia caused by antipsychotic drugs. South Med J. 1991;84(12):1505-1506.
31. Lipinski JF Jr, Zubenko GS, Cohen BM, et al. Propranolol in the treatment of neuroleptic-induced akathisia. Am J Psychiatry. 1984;141(3):412-415.
32. Adler L, Angrist B, Peselow E, et al. A controlled assessment of propranolol in the treatment of neuroleptic-induced akathisia. Br J Psychiatry. 1986;149:42-45.
33. Kumar R, Sachdev PS. Akathisia and second-generation antipsychotic drugs. Curr Opin Psychiatry. 2009;22(3):293-299.
34. Anttila SA, Leinonen EV. A review of the pharmacological and clinical profile of mirtazapine. CNS Drug Rev. 2001;7(3):249-264.
35. Rogóz Z, Wróbel A, Dlaboga D, et al. Effect of repeated treatment with mirtazapine on the central dopaminergic D2/D3 receptors. Pol J Pharmacol. 2002;54(4):381-389.
36. Miller CH, Fleischhacker WW, Ehrmann H, et al. Treatment of neuroleptic induced akathisia with the 5-HT2 antagonist ritanserin. Psychopharmacol Bull. 1990;26(3):373-376.
37. Weiss D, Aizenberg D, Hermesh H, et al. Cyproheptadine treatment in neuroleptic-induced akathisia. Br J Psychiatry. 1995;167(4):483-486.
38. Gross-Isseroff R, Magen A, Shiloh R, et al. The 5-HT1D receptor agonist zolmitriptan for neuroleptic-induced akathisia: an open label preliminary study. Int Clin Psychopharmacol. 2005;20(1):23-25.
39. Avital A, Gross-Isseroff R, Stryjer R, et al. Zolmitriptan compared to propranolol in the treatment of acute neuroleptic-induced akathisia: a comparative double-blind study. Eur Neuropsychopharmacol. 2009;19(7):476-482.
40. Rathbone J, Soares-Weiser K. Anticholinergics for neuroleptic-induced acute akathisia. Cochrane Database Syst Rev. 2006;(4):CD003727.
41. Sandyk R. Successful treatment of neuroleptic-induced akathisia with baclofen and clonazepam. A case report. Eur Neurol. 1985;24(4):286-288.
42. Miller CH, Fleischhacker W. Managing antipsychotic-induced acute and chronic akathisia. Drug Saf. 2000;22(1):73-81.
43. Pfeffer G, Chouinard G, Margolese HC. Gabapentin in the treatment of antipsychotic-induced akathisia in schizophrenia. Int Clin Psychopharmacol. 2005;20(3):179-181.
44. Stahl SM. Role of α1 adrenergic antagonism in the mechanism of action of iloperidone: reducing extrapyramidal symptoms. CNS Spectr. 2013;18(6):285-258.
45. Hettema JM, Ross DE. A case of aripiprazole-related tardive akathisia and its treatment with ropinirole. J Clin Psychiatry. 2007;68(11):1814-1815.

Issue
Current Psychiatry - 14(1)
Issue
Current Psychiatry - 14(1)
Page Number
14-18
Page Number
14-18
Publications
Publications
Topics
Article Type
Display Headline
Akathisia: Is restlessness a primary condition or an adverse drug effect?
Display Headline
Akathisia: Is restlessness a primary condition or an adverse drug effect?
Legacy Keywords
antipsychotics, akathisia, adverse effects, restlessness, drug-induced akathisia
Legacy Keywords
antipsychotics, akathisia, adverse effects, restlessness, drug-induced akathisia
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media