Cardiac implantable electronic device infection

Article Type
Changed
Mon, 10/01/2018 - 14:32
Display Headline
Cardiac implantable electronic device infection

Cardiac implantable electronic devices (CIEDs) have become common tools to improve the quality of life and longevity of patients with cardiac disease over the last few decades.1–4 CIEDs include implantable cardioverter defibrillators (ICDs), permanent pacemakers, biventricular pacemakers providing cardiac resynchronization therapy with or without a defibrillator, subcutaneous ICDs, and implantable loop recorders. With increasing approved indications, the number of CIEDs implanted each year continues to grow. This, paired with the aging population of patients receiving devices and their medical complexity, has led to a corresponding increase in device-related complications.2,3 One of the most serious complications is CIED infection, which leads to significant morbidity and death. These infections also represent a significant cost burden to the healthcare system, with treatment costs for a CIED infection estimated at over $146,000 in 2008.5

SCOPE OF THE PROBLEM

More than half a million permanent pacemakers and ICDs are implanted each year in the United States, with more than 4 million implanted between 1993 and 2008.5 The risk of infection is 0.5% to 1%, for a first-time implantation and 1% to 5% for a device replacement or upgrade.1,2,5–9 These infections can involve the generator pocket, bloodstream, or cardiac structures, leading to infective endocarditis.10 The timing of CIED infection appears to be bimodal in distribution: early infections usually occur as a result of the implantation procedure itself, whereas late infections occur in patients who are generally unwell or because of an insidious process that eventually crosses a threshold of clinical significance.3,11,12

Incidence and risk factors

Klug et al13 investigated the incidence rate and risk factors of CIED infection prospectively in a large cohort of patients from 44 centers who underwent CIED implantation. Of 6,319 procedures, 4,465 were first implants and the other 1,854 were a replacement or revision; 42 patients (0.68%) developed CIED infection by 12 months after the procedure, and the incidence of infection in replacement or revision cases was nearly twice the rate found in first implants.13 Risk factors for CIED infection included renal failure, heart failure, diabetes, and fever within last 24 hours before CIED implantation.14 The Implantable Cardiac Pulse Generator Replacement (REPLACE) registry found the 6-month incidence rate of CIED infection to be 1.4% after CIED replacement.6

Recently, there has been concern that the rate of newly infected CIEDs has outpaced the rate of newly implanted ones.5,15 Voigt et al15 reported a 12% increase in the rate of CIED implantation from 2004 to 2006 and an out-of-proportion 57% increase in the rate of CIED infection. A review from 2011 confirmed these findings, showing the annual CIED implantation incidence increased an average of 4.7% per year between 1993 and 2008.5 This was probably driven by clinical trials that broadened the indications for ICD implantation for primary prevention.16–19 Between 1993 and 2008, the rate of newly implanted devices increased by 96%, while the rate for newly infected CIEDs increased by 210%; the majority of this increase occurred after 2004.5 The study showed that comorbidities in patients receiving CIEDs increased sharply starting in 2004—alluding to the contribution of comorbid medical conditions such as renal failure, respiratory failure, heart failure, and diabetes to infection risk.5

However, a major obstacle to defining the true incidence rate of CIED infection is the lack of a clear denominator. CIED infection is not limited to the first few months after implantation. In fact, over half of these patients present more than 1 year after the last CIED intervention.12 Therefore, the number of patients at risk continues to grow each year and includes patients who underwent implantation that year or before, making it very difficult to compare infection rates. Additionally, the lack of a clear definition of CIED infection and the variations in duration of follow-up in different studies make it difficult to accurately assess the incidence of CIED infection.

PATHOGENESIS

Pathogens identified in 816 patients with lead extraction or device removal for CIED infection
A CIED can become infected at the time of implantation or pocket revision. The infection can then track along the endovascular portion of the leads resulting in endovascular infection and possibly endocarditis. A CIED can also become infected as a result of the hematogenous seeding of the leads or pocket during an episode of bacteremia. Most of these infections (70%) are caused by staphylococcal species, and many are becoming resistant to methicillin.12 Other species include gram-negative organisms (9%), enterococci (4.2%), streptococci (2.5%), and fungi (1%) (Table 1). Despite clear evidence of clinical CIED infection, the cultures remain negative in about 13% of cases, perhaps because of the unfortunately common practice of starting antibiotic therapy before obtaining cultures or because of the need to incubate culture samples for a longer duration.12 A longer incubation time is particularly important for infections involving Proprionibacterium acnes, an aerobic gram-positive rod commonly associated with acne vulgaris.20

 

 

DIAGNOSIS

Prompt and accurate diagnosis of CIED infection is critical as it allows for early management with antibiotic therapy and device removal. As the number of CIED implantations increases, providers on the front lines—emergency, family practice, and internal medicine physicians—will play an increasing role in recognizing and diagnosing CIED infection. Patients with CIED infection present with a range of signs and symptoms including fever, chills, erythema, swelling, drainage, tenderness, malaise, erosion, and warmth of the skin overlying the generator pocket.2 In 55% of cases, patients present with localized pocket infection, while the remaining patients have signs of an endovascular infection without obvious pocket involvement.12 Localized pocket infection is more common during the first year after device implantation. CIED-associated endovascular infections occur more commonly in patients with multiple comorbidities including diabetes, renal failure, prior heart valve operation, rheumatic heart disease, and prior bloodstream infection.2 Despite the theoretical divide in CIED infections (endovascular vs pocket), overlap is common: many patients with pocket infection show evidence of bacteremia and vegetations on the leads.

Pocket infection after placement of a cardiac implantable electronic device can present as erythema and drainage (A); swelling, skin necrosis, and eschar formation (B); and erythema, swelling, and bullae formation (C).
Figure 1. Pocket infection after placement of a cardiac implantable electronic device can present as erythema and drainage (A); swelling, skin necrosis, and eschar formation (B); and erythema, swelling, and bullae formation (C).
Physical examination of the pocket is critical as it may reveal visible signs of infection and support the diagnosis of localized pocket infection (Figure 1). Blood cultures are essential and should be collected before starting antibiotic therapy. Culture results assist in the diagnosis of CIED infection and also help identify the microorganism involved, and this information helps tailor the choice and duration of antibiotic therapy. Echocardiography (transthoracic and transesophageal) can assist the clinician in the diagnosis of CIED infection but requires careful interpretation because some patients with no signs or symptoms of infection can have small fibrinous strands or thrombi attached to the CIED leads.14 These findings should only be interpreted in correlation to the clinical presentation.

Diagnosing pocket infection from the physical examination can be difficult due to the often subtle manifestations of the underlying pathophysiology and because visible changes to the pocket can occur over weeks and months. Furthermore, differentiating superficial infection, hematoma, seroma, and allergic reactions from deep pocket infection can be challenging. In cases when the diagnosis is not clear and there are no systemic findings of infection, conservative management with close follow-up is reasonable. Similarly, the diagnosis of endovascular infection is sometimes delayed because the symptoms are not very specific or because of a lack of awareness of the presence of a CIED and its role in endovascular infection.

MANAGEMENT

A multidisciplinary approach involving cardiology, infectious disease, electrophysiology, and cardiothoracic surgery teams is required to optimize outcomes in patients with CIED infection. CIED infection is particularly difficult to treat with antibiotic therapy alone because it involves infection of an implanted device and an associated biofilm that is resistant to the effects of antibiotics. Once infection is confirmed, antibiotic therapy serves as an adjunct to the complete removal of the hardware. Most patients receive 2 weeks of intravenous antibiotics after removal of an infected CIED, with longer courses for patients with Staphylococcus aureus infection or documented endocarditis.21

Infectious disease consultation is paramount in order to choose the appropriate type and duration of antibiotic therapy. Conservative approaches that involve using antibiotics alone or incomplete system removal have high failure rates with high rates of morbidity and mortality.13,21–28 However, chronic antibiotic suppressive therapy may be considered as a palliative measure for patients who are not candidates for lead extraction.

DEVICE REMOVAL

Confirmation of CIED infection is a class I indication for device removal and the patient should be referred to an electrophysiologist. Transvenous lead extraction (TLE) is a percutaneous procedure performed by the electrophysiologist in the electrophysiology laboratory or hybrid operating room with cardiothoracic surgery support, and it is generally performed under general anesthesia with invasive hemodynamic monitoring. After opening and debriding the infected pocket, the generator is disconnected from the leads. After the lead tips are unscrewed from the myocardium, gentle traction is applied to determine if the leads can easily be removed. If traction is unsuccessful, additional tools (both powered or mechanical sheaths) are used to complete the lead extraction29; the goal is to lyse and free the fibrotic attachments between parallel leads and between the leads and vessel wall or the myocardium. Once the lead is freed from the adhesions it can be removed safely.

In a patient with endocarditis after cardiac implantable electronic device placement, transthoracic echocardiography shows a large vegetation near the right atrium, right ventricle, and across the tricuspid valve.
Figure 2. In a patient with endocarditis after cardiac implantable electronic device placement, transthoracic echocardiography shows a large vegetation (V) near the right atrium (RA), right ventricle (RV), and across the tricuspid valve (TV). This required surgical extraction of the organized vegetation along with the device and leads.
The incidence of major complications with lead extraction is low (1.8%), but the procedure can be life-threatening.30 Major complications include cardiac avulsion, vascular laceration, pericardial effusion, tamponade, hemothorax, valve injury, and death during the procedure.30 Risk factors for major complications with TLE include renal failure, low body mass index, and the presence of a defibrillator coil on the lead.30,31 In a large cohort of more than 3,000 patients requiring 6,000 TLE procedures at our tertiary care center, the incidence of catastrophic complications that required emergency cardiac surgery or vascular intervention was 0.8%.32 Many of these patients were rescued through emergency surgical repair of a venous laceration or cardiac perforation but still had an in-hospital mortality rate of 36%. Surgical lead extraction is usually performed if percutaneous lead extraction has failed, if epicardial leads are present, if large vegetations are attached to the leads, or if surgery is warranted for valvular involvement with endocarditis (Figure 2).14

 

 

REIMPLANTATION

The need for reimplantation after removal of an infected CIED should be thought about before the extraction. In general, extracting an infected CIED should be viewed as an opportunity to reassess the need for the device. Almost one-third of patients who undergo extraction of infected CIED do not require immediate reimplantation.2 This could be due to reversal of the initial indication, emergence of new clinical conditions, patient preference, or the lack of an absolute indication. If reimplantation is necessary, the new device is typically placed on the opposite side of the chest from the previously infected pocket site after blood cultures are negative for at least 72 hours.21

CIED INFECTION MORTALITY

Despite proper management with CIED removal supported by antibiotic therapy, CIED infection carries a high risk of death. The 30-day mortality is estimated to be between 5% and 6%.33 In a large case series of 412 CIED extractions, there were 19 in-hospital deaths. Of these 19 deaths, 2 were related to the extraction itself with the other 17 related to sepsis, multiorgan failure, stroke, renal failure, or heart failure.2 The 1-year mortality rate is also increased for this population; recent data show 1-year mortality rates of 8% to 17% despite device removal and antibiotic therapy.2,34,35 This increased mortality rate was also demonstrated in a large cohort of Medicare patients undergoing CIED procedures.36 Medicare patients with CIED infection had double the risk of death at 1 year compared with patients without infection.36

Risk factors for death at 1 year include worse baseline functional status, renal failure, and type of infection; eg, endovascular infection carries a risk of death 2 times higher than pocket infection.37

PREVENTION

Because CIED infection carries significant short-term and long-term mortality rates despite optimal management, the best strategy is prevention. Preventing CIED infection begins with the decision to implant a device with careful assessment of the indication, the timing of the procedure, and the patient’s clinical status. CIED procedures are performed under strict sterile surgical techniques with great attention to the incision and proper closure. Surgical data favor the use of chlorhexidine-alcohol solutions for skin preparation compared with povidone-iodine solutions to prevent both superficial and deep surgical wound infections.38 However, recent studies showed no significant difference between the 2 preparation methods in reducing rates of CIED infection.39,40 In individuals colonized with S aureus, the risk of CIED infection can be reduced using a body wash containing chlorhexidine and a nasal spray containing mupirocin.41,42

Preoperative antibiotics

The use of preoperative antibiotics has been shown to reduce the risk of infection.43 In a large prospective cohort of patients undergoing a de novo or secondary CIED procedure, the use of perioperative antibiotics was negatively associated with the risk of CIED infection.13 This was later confirmed by a double-blind randomized trial of 1,000 patients undergoing permanent pacemaker or ICD initial implantation or generator replacement. This study was stopped prematurely as the use of antibiotics was clearly associated with a lower risk of CIED infection.44 Therefore, prophylaxis with an antibiotic active against staphylococci before the incision is made is a class I indication to prevent infection.1

Currently, no data support giving prophylactic antibiotics after the procedure; however, the Prevention of Arrhythmia Device Infection Trial (PADIT) is currently comparing the risk of infection with conventional preoperative antibiotics vs a regimen of pre- and post-procedure antibiotics (clinicaltrial.gov: NCT01628666).

Hemostasis

Adequate hemostasis is critical, since the risk of CIED infection is 7 times greater with formation of a hematoma.45 Heparin products, especially low-molecular-weight heparin, should be avoided at the time of CIED implantation. In patients at high risk for thromboembolism who are on warfarin therapy, the continuation of warfarin is associated with a lower incidence of hematoma compared with bridging with heparin in patients undergoing CIED procedures.46 Therefore, if anticoagulation can be withheld, it is better to stop the anticoagulant before the procedure. When this is not possible or when it carries significant risk (eg, a patient with a mechanical mitral valve who needs a CIED implantation), it is better to maintain the patient on warfarin therapy with a therapeutic international normalized ratio rather than bridging with heparin products.

Antibacterial envelop and new devices

TYRX
Figure 3. The TYRX absorbable antibacterial envelope is a mesh coated with the antibiotics rifampin and minocycline, which elute off the mesh within approximately 7 days. The mesh is completely absorbed into the body in about 9 weeks.
A new development in the prevention of CIED infection is the TYRX absorbable antibacterial envelope (Medtronic Inc.) (Figure 3), a multifilament knitted mesh coated with the antibiotics rifampin and minocycline, which are released in the device pocket over 7 days. The first-generation envelope was nonabsorbable; the new product uses a fully bioabsorbable polymer that dissolves within 9 weeks. Data from nonrandomized studies using mainly the nonabsorbable version showed favorable outcomes in reducing the rate of CIED infections.47,48 The World-wide Randomized Antibiotic Envelope Infection Prevention Trial (WRAP-IT) is a large randomized clinical trial assessing the efficacy of the absorbable envelope in reducing CIED infection rates in patients undergoing CIED replacement or upgrade.49

A leadless pacemaker in the right ventricle.
Figure 4. A leadless pacemaker in the right ventricle. The left atrial appendage exclusion clip is present.
The development of new cardiac devices carries the potential of reducing certain types of infection. The subcutaneous ICD is an entirely subcutaneous system with no endovascular component, and therefore it can prevent endovascular infection, especially in patients at high risk of infection (eg, patients on hemodialysis).50 On the other hand, the leadless pacemaker is a single-chamber pacemaker deployed percutaneously in the right ventricle without the need for a pocket, thereby eliminating the risk of pocket infection (Figure 4).51,52 Whether the risk of endovascular infection will be reduced is not yet known.

 

 

CONCLUSION

CIED infection is a major complication that carries significant risk of morbidity and death. Early diagnosis and referral to a multidisciplinary treatment team is crucial to increasing the possibility of a cure. While device extraction has risks, it is nevertheless typically required for complete resolution of the infection. Large clinical trials are under way to address current knowledge gaps about CIED infection, including our understanding of the true incidence rate, risk factors, and efficacy of various implantation techniques. Future trends to minimize the risk of CIED infection include better screening, better diagnostic tools, new devices with fewer or no leads, longer battery life to minimize the need for additional procedures, and the use of supportive tools and products to minimize the risk of infection.

References
  1. Baddour LM, Epstein AE, Erickson CC, et al; American Heart Association Rheumatic Fever, Endocarditis, and Kawasaki Disease Committee of the Council on Cardiovascular Disease in the Young; Council on Cardiovascular Surgery and Anesthesia; Council on Cardiovascular Nursing; Council on Clinical Cardiology; and the Interdisciplinary Council on Quality of Care and Outcomes Research. Update on cardiovascular implantable electronic device infections and their management: a scientific statement from the American Heart Association. Circulation 2010; 121:458–477.
  2. Tarakji KG, Chan EJ, Cantillon DJ, et al. Cardiac implantable electronic device infections: presentation, management, and patient outcomes. Heart Rhythm 2010; 7:1043–1047.
  3. Baddour LM. Cardiac device infection—or not. Circulation 2010; 121:1686–1687.
  4. Kusumoto FM, Schoenfeld MH, Wilkoff BL, et al. 2017 HRS expert consensus statement on cardiovascular implantable electronic device lead management and extraction. Heart Rhythm 2017; Sept 15. pii: S1547-5271(17)31080-9. doi:10.1016/j.hrthm.2017.09.001. [Epub ahead of print]
  5. Greenspon AJ, Patel JD, Lau E, et al. 16-Year trends in the infection burden for pacemakers and implantable cardioverter-defibrillators in the United States: 1993 to 2008. J Am Coll Cardiol 2011; 58:1001–1006.
  6. Poole JE, Gleva MJ, Mela T, et al; REPLACE Registry Investigators. Complication rates associated with pacemaker or implantable cardioverter-defibrillator generator replacements and upgrade procedures: results from the REPLACE registry. Circulation 2010; 122:1553–1561.
  7. Mela T, McGovern BA, Garan H, et al. Long-term infection rates associated with the pectoral versus abdominal approach to cardioverter-defibrillator implants. Am J Cardiol 2001; 88:750–753.
  8. de Bie MK, van Rees JB, Thijssen J, et al. Cardiac device infections are associated with a significant mortality risk. Heart Rhythm 2012; 9:494–498.
  9. Polyzos KA, Konstantelias AA, Falagas ME. Risk factors for cardiac implantable electronic device infection: a systematic review and meta-analysis. Europace 2015; 17:767–777.
  10. Deharo J-C, Quatre A, Mancini J, et al. Long-term outcomes following infection of cardiac implantable electronic devices: a prospective matched cohort study. Heart 2012; 98:724–731.
  11. Sohail MR, Hussain S, Le KY, et al; Mayo Cardiovascular Infections Study Group. Risk factors associated with early- versus late-onset implantable cardioverter-defibrillator infections. J Interv Card Electrophysiol 2011; 31:171–183.
  12. Hussein AA, Baghdy Y, Wazni OM, et al. Microbiology of cardiac implantable electronic device infections. JACC Clin Electrophysiol 2016; 2:498–505.
  13. Klug D, Balde M, Pavin D, et al; PEOPLE Study Group. Risk factors related to infections of implanted pacemakers and cardioverter-defibrillators: results of a large prospective study. Circulation 2007; 116:1349–1355.
  14. Tarakji KG, Wilkoff BL. Management of cardiac implantable electronic device infections: the challenges of understanding the scope of the problem and its associated mortality. Expert Rev Cardiovasc Ther 2013; 11:607–616.
  15. Voigt A, Shalaby A, Saba S. Continued rise in rates of cardiovascular implantable electronic device infections in the United States: temporal trends and causative insights. Pacing Clin Electrophysiol 2010; 33:414–419.
  16. Bardy GH, Lee KL, Mark DB, et al; Sudden Cardiac Death in Heart Failure Trial (SCD-HeFT) Investigators. Amiodarone or an implantable cardioverter-defibrillator for congestive heart failure. N Engl J Med 2005; 352:225–237.
  17. Moss AJ, Zareba W, Hall WJ, et al; Multicenter Automatic Defibrillator Implantation Trial II Investigators. Prophylactic implantation of a defibrillator in patients with myocardial infarction and reduced ejection fraction. N Engl J Med 2002; 346:877–883.
  18. Kadish A, Dyer A, Daubert JP, et al; Defibrillators in Non-Ischemic Cardiomyopathy Treatment Evaluation (DEFINITE) Investigators. Prophylactic defibrillator implantation in patients with nonischemic dilated cardiomyopathy. N Engl J Med 2004; 350:2151–2158.
  19. Buxton AE, Lee KL, Fisher JD, Josephson ME, Prystowsky EN, Hafley G; Multicenter Unsustained Tachycardia Trial Investigators. A randomized study of the prevention of sudden death in patients with coronary artery disease. N Engl J Med 1999; 341:1882–1890.
  20. Abdulmassih R, Makadia J, Como J, Paulson M, Min Z, Bhanot N. Propionibacterium acnes: Time-to-positivity in standard bacterial culture from different anatomical sites. J Clin Med Res 2016; 8:916–918.
  21. Sohail MR, Uslan DZ, Khan AH, et al. Management and outcome of permanent pacemaker and implantable cardioverter-defibrillator infections. J Am Coll Cardiol 2007; 49:1851–1859.
  22. Cacoub P, Leprince P, Nataf P, et al. Pacemaker infective endocarditis. Am J Cardiol 1998; 82:480–484.
  23. Chua JD, Wilkoff BL, Lee I, Juratli N, Longworth DL, Gordon SM. Diagnosis and management of infections involving implantable electrophysiologic cardiac devices. Ann Intern Med 2000; 133:604–608.
  24. Bracke FA, Meijer A, van Gelder LM. Pacemaker lead complications: when is extraction appropriate and what can we learn from published data? Heart 2001; 85:254–259.
  25. Camus C, Leport C, Raffi F, Michelet C, Cartier F, Vilde JL. Sustained bacteremia in 26 patients with a permanent endocardial pacemaker: assessment of wire removal. Clin Infect Dis 1993; 17:46–55.
  26. Molina JE. Undertreatment and overtreatment of patients with infected antiarrhythmic implantable devices. Ann Thorac Surg 1997; 63:504–509.
  27. Viganego F, O’Donoghue S, Eldadah Z, et al. Effect of early diagnosis and treatment with percutaneous lead extraction on survival in patients with cardiac device infections. Am J Cardiol 2012; 109:1466–1471.
  28. Le KY, Sohail MR, Friedman PA, et al; Mayo Cardiovascular Infections Study Group. Impact of timing of device removal on mortality in patients with cardiovascular implantable electronic device infections. Heart Rhythm 2011; 8:1678–1685.
  29. Wazni O, Wilkoff BL. Considerations for cardiac device lead extraction. Nat Rev Cardiol 2016; 13:221–229.
  30. Brunner MP, Cronin EM, Duarte VE, et al. Clinical predictors of adverse patient outcomes in an experience of more than 5000 chronic endovascular pacemaker and defibrillator lead extractions. Heart Rhythm 2014; 11:799–805.
  31. Wazni O, Epstein LM, Carrillo RG, et al. Lead extraction in the contemporary setting: the LExICon study: an observational retrospective study of consecutive laser lead extractions. J Am Coll Cardiol 2010; 55:579–586.
  32. Brunner MP, Cronin EM, Wazni O, et al. Outcomes of patients requiring emergent surgical or endovascular intervention for catastrophic complications during transvenous lead extraction. Heart Rhythm 2014; 11:419–425.
  33. Habib A, Le KY, Baddour LM, et al; for the Mayo Cardiovascular Infections Study Group. Predictors of mortality in patients with cardiovascular implantable electronic device infections. Am J Cardiol 2013; 111:874–879.
  34. Baman TS, Gupta SK, Valle JA, Yamada E. Risk factors for mortality in patients with cardiac device-related infection. Circ Arrhythm Electrophysiol 2009; 2:129–134.
  35. Deckx S, Marynissen T, Rega F, et al. Predictors of 30-day and 1-year mortality after transvenous lead extraction: a single-centre experience. Europace 2014; 16:1218–1225.
  36. Sohail MR, Henrikson CA, Braid-Forbes MJ, Forbes KF, Lerner DJ. Increased long-term mortality in patients with cardiovascular implantable electronic device infections. Pacing Clin Electrophysiol 2015; 38:231–239.
  37. Tarakji KG, Wazni OM, Harb S, Hsu A, Saliba W, Wilkoff BL. Risk factors for 1-year mortality among patients with cardiac implantable electronic device infection undergoing transvenous lead extraction: the impact of the infection type and the presence of vegetation on survival. Europace 2014; 16:1490–1495.
  38. Darouiche RO, Wall MJ Jr., Itani KM, et al. Chlorhexidine—alcohol versus povidone—iodine for surgical-site antisepsis. N Engl J Med 2010; 362:18–26.
  39. Qintar M, Zardkoohi O, Hammadah M, et al. The impact of changing antiseptic skin preparation agent used for cardiac implantable electronic device (CIED) procedures on the risk of infection. Pacing Clin Electrophysiol 2015; 38:240–246.
  40. Da Costa A, Tulane C, Dauphinot V, et al. Preoperative skin antiseptics for prevention of cardiac implantable electronic device infections: a historical-controlled interventional trial comparing aqueous against alcoholic povidone-iodine solutions. Europace 2015; 17:1092–1098.
  41. Padfield GJ, Steinberg C, Bennett MT, et al. Preventing cardiac implantable electronic device infections. Heart Rhythm 2015; 12:2344–2356.
  42. Bode LGM, Kluytmans JAJW, Wertheim HFL, et al. Preventing surgical-site infections in nasal carriers of Staphylococcus aureus. N Engl J Med 2010; 362:9–17.
  43. Da Costa A, Kirkorian G, Cucherat M, et al. Antibiotic prophylaxis for permanent pacemaker implantation: a meta-analysis. Circulation 1998; 97:1796–1801.
  44. de Oliveira JC, Martinelli M, Nishioka SADO, et al. Efficacy of antibiotic prophylaxis before the implantation of pacemakers and cardioverter-defibrillators: results of a large, prospective, randomized, double-blinded, placebo-controlled trial. Circ Arrhythm Electrophysiol 2009; 2:29–34.
  45. Essebag V, Verma A, Healey JS, et al; BRUISE CONTROL Investigators. Clinically significant pocket hematoma increases long-term risk of device infection: BRUISE CONTROL INFECTION study. J Am Coll Cardiol 2016; 67:1300–1308.
  46. Birnie DH, Healey JS, Wells GA, et al; BRUISE CONTROL Investigators. Pacemaker or defibrillator surgery without interruption of anticoagulation. N Engl J Med 2013; 368:2084–2093.
  47. Henrikson CA, Sohail MR, Acosta H, et al. Antibacterial envelope is associated with low infection rates after implantable cardioverter-defibrillator and cardiac resynchronization therapy device replacement: results of the Citadel and Centurion studies. 2017 http://dx.doi.org/10.1016/j.jacep.2017.02.016
  48. Mittal S, Shaw RE, Michel K, et al. Cardiac implantable electronic device infections: incidence, risk factors, and the effect of the AigisRx antibacterial envelope. Heart Rhythm 2014; 11:595–601.
  49. Tarakji KG, Mittal S, Kennergren C, et al. Worldwide Randomized Antibiotic EnveloPe Infection PrevenTion Trial (WRAP-IT). Am Heart J 2016; 180:12–B21.
  50. Burke MC, Gold MR, Knight BP, et al. Safety and efficacy of the totally subcutaneous implantable defibrillator: 2-year results from a pooled analysis of the IDE study and EFFORTLESS registry. J Am Coll Cardiol 2015; 65:1605–1615.
  51. Reddy VY, Exner DV, Cantillon DJ, et al; LEADLESS II Study Investigators. Percutaneous implantation of an entirely intracardiac leadless pacemaker. N Engl J Med 2015; 373:1125–1135.
  52. Reynolds D, Duray GZ, Omar R, et al; Micra Transcatheter Pacing Study Group. A leadless intracardiac transcatheter pacing system. N Engl J Med 2016; 374:533–541.
Article PDF
Author and Disclosure Information

Cameron T. Lambert, MD
Department of Cardiovascular Medicine, Heart and Vascular Institute, Cleveland Clinic

Khaldoun G. Tarakji, MD, MPH
Department of Cardiovascular Medicine, Heart and Vascular Institute, Cleveland Clinic

Correspondence: Khaldoun G. Tarakji, MD, MPH, Department of Cardiovascular Medicine, Heart and Vascular Institute, J2-2, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195; [email protected]

Dr. Lambert reported no financial interests or relationships that pose a potential conflict of interest with this article. Dr. Tarakji reported that he receives consulting/advisory fees from Medtronic and AliveCor.

Publications
Page Number
47-53
Legacy Keywords
cardiac implantable electronic device infection, CIED infection, pacemaker, implantable cardioverter-defibrillator, MRSA, methicillin-resistant Staphylococcus aureus, VRE, vancomycin-resistant Enterococcus species, antibiotics, vegetation, Cameron Lamberg, Khaldoun Tarakji
Author and Disclosure Information

Cameron T. Lambert, MD
Department of Cardiovascular Medicine, Heart and Vascular Institute, Cleveland Clinic

Khaldoun G. Tarakji, MD, MPH
Department of Cardiovascular Medicine, Heart and Vascular Institute, Cleveland Clinic

Correspondence: Khaldoun G. Tarakji, MD, MPH, Department of Cardiovascular Medicine, Heart and Vascular Institute, J2-2, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195; [email protected]

Dr. Lambert reported no financial interests or relationships that pose a potential conflict of interest with this article. Dr. Tarakji reported that he receives consulting/advisory fees from Medtronic and AliveCor.

Author and Disclosure Information

Cameron T. Lambert, MD
Department of Cardiovascular Medicine, Heart and Vascular Institute, Cleveland Clinic

Khaldoun G. Tarakji, MD, MPH
Department of Cardiovascular Medicine, Heart and Vascular Institute, Cleveland Clinic

Correspondence: Khaldoun G. Tarakji, MD, MPH, Department of Cardiovascular Medicine, Heart and Vascular Institute, J2-2, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195; [email protected]

Dr. Lambert reported no financial interests or relationships that pose a potential conflict of interest with this article. Dr. Tarakji reported that he receives consulting/advisory fees from Medtronic and AliveCor.

Article PDF
Article PDF
Related Articles

Cardiac implantable electronic devices (CIEDs) have become common tools to improve the quality of life and longevity of patients with cardiac disease over the last few decades.1–4 CIEDs include implantable cardioverter defibrillators (ICDs), permanent pacemakers, biventricular pacemakers providing cardiac resynchronization therapy with or without a defibrillator, subcutaneous ICDs, and implantable loop recorders. With increasing approved indications, the number of CIEDs implanted each year continues to grow. This, paired with the aging population of patients receiving devices and their medical complexity, has led to a corresponding increase in device-related complications.2,3 One of the most serious complications is CIED infection, which leads to significant morbidity and death. These infections also represent a significant cost burden to the healthcare system, with treatment costs for a CIED infection estimated at over $146,000 in 2008.5

SCOPE OF THE PROBLEM

More than half a million permanent pacemakers and ICDs are implanted each year in the United States, with more than 4 million implanted between 1993 and 2008.5 The risk of infection is 0.5% to 1%, for a first-time implantation and 1% to 5% for a device replacement or upgrade.1,2,5–9 These infections can involve the generator pocket, bloodstream, or cardiac structures, leading to infective endocarditis.10 The timing of CIED infection appears to be bimodal in distribution: early infections usually occur as a result of the implantation procedure itself, whereas late infections occur in patients who are generally unwell or because of an insidious process that eventually crosses a threshold of clinical significance.3,11,12

Incidence and risk factors

Klug et al13 investigated the incidence rate and risk factors of CIED infection prospectively in a large cohort of patients from 44 centers who underwent CIED implantation. Of 6,319 procedures, 4,465 were first implants and the other 1,854 were a replacement or revision; 42 patients (0.68%) developed CIED infection by 12 months after the procedure, and the incidence of infection in replacement or revision cases was nearly twice the rate found in first implants.13 Risk factors for CIED infection included renal failure, heart failure, diabetes, and fever within last 24 hours before CIED implantation.14 The Implantable Cardiac Pulse Generator Replacement (REPLACE) registry found the 6-month incidence rate of CIED infection to be 1.4% after CIED replacement.6

Recently, there has been concern that the rate of newly infected CIEDs has outpaced the rate of newly implanted ones.5,15 Voigt et al15 reported a 12% increase in the rate of CIED implantation from 2004 to 2006 and an out-of-proportion 57% increase in the rate of CIED infection. A review from 2011 confirmed these findings, showing the annual CIED implantation incidence increased an average of 4.7% per year between 1993 and 2008.5 This was probably driven by clinical trials that broadened the indications for ICD implantation for primary prevention.16–19 Between 1993 and 2008, the rate of newly implanted devices increased by 96%, while the rate for newly infected CIEDs increased by 210%; the majority of this increase occurred after 2004.5 The study showed that comorbidities in patients receiving CIEDs increased sharply starting in 2004—alluding to the contribution of comorbid medical conditions such as renal failure, respiratory failure, heart failure, and diabetes to infection risk.5

However, a major obstacle to defining the true incidence rate of CIED infection is the lack of a clear denominator. CIED infection is not limited to the first few months after implantation. In fact, over half of these patients present more than 1 year after the last CIED intervention.12 Therefore, the number of patients at risk continues to grow each year and includes patients who underwent implantation that year or before, making it very difficult to compare infection rates. Additionally, the lack of a clear definition of CIED infection and the variations in duration of follow-up in different studies make it difficult to accurately assess the incidence of CIED infection.

PATHOGENESIS

Pathogens identified in 816 patients with lead extraction or device removal for CIED infection
A CIED can become infected at the time of implantation or pocket revision. The infection can then track along the endovascular portion of the leads resulting in endovascular infection and possibly endocarditis. A CIED can also become infected as a result of the hematogenous seeding of the leads or pocket during an episode of bacteremia. Most of these infections (70%) are caused by staphylococcal species, and many are becoming resistant to methicillin.12 Other species include gram-negative organisms (9%), enterococci (4.2%), streptococci (2.5%), and fungi (1%) (Table 1). Despite clear evidence of clinical CIED infection, the cultures remain negative in about 13% of cases, perhaps because of the unfortunately common practice of starting antibiotic therapy before obtaining cultures or because of the need to incubate culture samples for a longer duration.12 A longer incubation time is particularly important for infections involving Proprionibacterium acnes, an aerobic gram-positive rod commonly associated with acne vulgaris.20

 

 

DIAGNOSIS

Prompt and accurate diagnosis of CIED infection is critical as it allows for early management with antibiotic therapy and device removal. As the number of CIED implantations increases, providers on the front lines—emergency, family practice, and internal medicine physicians—will play an increasing role in recognizing and diagnosing CIED infection. Patients with CIED infection present with a range of signs and symptoms including fever, chills, erythema, swelling, drainage, tenderness, malaise, erosion, and warmth of the skin overlying the generator pocket.2 In 55% of cases, patients present with localized pocket infection, while the remaining patients have signs of an endovascular infection without obvious pocket involvement.12 Localized pocket infection is more common during the first year after device implantation. CIED-associated endovascular infections occur more commonly in patients with multiple comorbidities including diabetes, renal failure, prior heart valve operation, rheumatic heart disease, and prior bloodstream infection.2 Despite the theoretical divide in CIED infections (endovascular vs pocket), overlap is common: many patients with pocket infection show evidence of bacteremia and vegetations on the leads.

Pocket infection after placement of a cardiac implantable electronic device can present as erythema and drainage (A); swelling, skin necrosis, and eschar formation (B); and erythema, swelling, and bullae formation (C).
Figure 1. Pocket infection after placement of a cardiac implantable electronic device can present as erythema and drainage (A); swelling, skin necrosis, and eschar formation (B); and erythema, swelling, and bullae formation (C).
Physical examination of the pocket is critical as it may reveal visible signs of infection and support the diagnosis of localized pocket infection (Figure 1). Blood cultures are essential and should be collected before starting antibiotic therapy. Culture results assist in the diagnosis of CIED infection and also help identify the microorganism involved, and this information helps tailor the choice and duration of antibiotic therapy. Echocardiography (transthoracic and transesophageal) can assist the clinician in the diagnosis of CIED infection but requires careful interpretation because some patients with no signs or symptoms of infection can have small fibrinous strands or thrombi attached to the CIED leads.14 These findings should only be interpreted in correlation to the clinical presentation.

Diagnosing pocket infection from the physical examination can be difficult due to the often subtle manifestations of the underlying pathophysiology and because visible changes to the pocket can occur over weeks and months. Furthermore, differentiating superficial infection, hematoma, seroma, and allergic reactions from deep pocket infection can be challenging. In cases when the diagnosis is not clear and there are no systemic findings of infection, conservative management with close follow-up is reasonable. Similarly, the diagnosis of endovascular infection is sometimes delayed because the symptoms are not very specific or because of a lack of awareness of the presence of a CIED and its role in endovascular infection.

MANAGEMENT

A multidisciplinary approach involving cardiology, infectious disease, electrophysiology, and cardiothoracic surgery teams is required to optimize outcomes in patients with CIED infection. CIED infection is particularly difficult to treat with antibiotic therapy alone because it involves infection of an implanted device and an associated biofilm that is resistant to the effects of antibiotics. Once infection is confirmed, antibiotic therapy serves as an adjunct to the complete removal of the hardware. Most patients receive 2 weeks of intravenous antibiotics after removal of an infected CIED, with longer courses for patients with Staphylococcus aureus infection or documented endocarditis.21

Infectious disease consultation is paramount in order to choose the appropriate type and duration of antibiotic therapy. Conservative approaches that involve using antibiotics alone or incomplete system removal have high failure rates with high rates of morbidity and mortality.13,21–28 However, chronic antibiotic suppressive therapy may be considered as a palliative measure for patients who are not candidates for lead extraction.

DEVICE REMOVAL

Confirmation of CIED infection is a class I indication for device removal and the patient should be referred to an electrophysiologist. Transvenous lead extraction (TLE) is a percutaneous procedure performed by the electrophysiologist in the electrophysiology laboratory or hybrid operating room with cardiothoracic surgery support, and it is generally performed under general anesthesia with invasive hemodynamic monitoring. After opening and debriding the infected pocket, the generator is disconnected from the leads. After the lead tips are unscrewed from the myocardium, gentle traction is applied to determine if the leads can easily be removed. If traction is unsuccessful, additional tools (both powered or mechanical sheaths) are used to complete the lead extraction29; the goal is to lyse and free the fibrotic attachments between parallel leads and between the leads and vessel wall or the myocardium. Once the lead is freed from the adhesions it can be removed safely.

In a patient with endocarditis after cardiac implantable electronic device placement, transthoracic echocardiography shows a large vegetation near the right atrium, right ventricle, and across the tricuspid valve.
Figure 2. In a patient with endocarditis after cardiac implantable electronic device placement, transthoracic echocardiography shows a large vegetation (V) near the right atrium (RA), right ventricle (RV), and across the tricuspid valve (TV). This required surgical extraction of the organized vegetation along with the device and leads.
The incidence of major complications with lead extraction is low (1.8%), but the procedure can be life-threatening.30 Major complications include cardiac avulsion, vascular laceration, pericardial effusion, tamponade, hemothorax, valve injury, and death during the procedure.30 Risk factors for major complications with TLE include renal failure, low body mass index, and the presence of a defibrillator coil on the lead.30,31 In a large cohort of more than 3,000 patients requiring 6,000 TLE procedures at our tertiary care center, the incidence of catastrophic complications that required emergency cardiac surgery or vascular intervention was 0.8%.32 Many of these patients were rescued through emergency surgical repair of a venous laceration or cardiac perforation but still had an in-hospital mortality rate of 36%. Surgical lead extraction is usually performed if percutaneous lead extraction has failed, if epicardial leads are present, if large vegetations are attached to the leads, or if surgery is warranted for valvular involvement with endocarditis (Figure 2).14

 

 

REIMPLANTATION

The need for reimplantation after removal of an infected CIED should be thought about before the extraction. In general, extracting an infected CIED should be viewed as an opportunity to reassess the need for the device. Almost one-third of patients who undergo extraction of infected CIED do not require immediate reimplantation.2 This could be due to reversal of the initial indication, emergence of new clinical conditions, patient preference, or the lack of an absolute indication. If reimplantation is necessary, the new device is typically placed on the opposite side of the chest from the previously infected pocket site after blood cultures are negative for at least 72 hours.21

CIED INFECTION MORTALITY

Despite proper management with CIED removal supported by antibiotic therapy, CIED infection carries a high risk of death. The 30-day mortality is estimated to be between 5% and 6%.33 In a large case series of 412 CIED extractions, there were 19 in-hospital deaths. Of these 19 deaths, 2 were related to the extraction itself with the other 17 related to sepsis, multiorgan failure, stroke, renal failure, or heart failure.2 The 1-year mortality rate is also increased for this population; recent data show 1-year mortality rates of 8% to 17% despite device removal and antibiotic therapy.2,34,35 This increased mortality rate was also demonstrated in a large cohort of Medicare patients undergoing CIED procedures.36 Medicare patients with CIED infection had double the risk of death at 1 year compared with patients without infection.36

Risk factors for death at 1 year include worse baseline functional status, renal failure, and type of infection; eg, endovascular infection carries a risk of death 2 times higher than pocket infection.37

PREVENTION

Because CIED infection carries significant short-term and long-term mortality rates despite optimal management, the best strategy is prevention. Preventing CIED infection begins with the decision to implant a device with careful assessment of the indication, the timing of the procedure, and the patient’s clinical status. CIED procedures are performed under strict sterile surgical techniques with great attention to the incision and proper closure. Surgical data favor the use of chlorhexidine-alcohol solutions for skin preparation compared with povidone-iodine solutions to prevent both superficial and deep surgical wound infections.38 However, recent studies showed no significant difference between the 2 preparation methods in reducing rates of CIED infection.39,40 In individuals colonized with S aureus, the risk of CIED infection can be reduced using a body wash containing chlorhexidine and a nasal spray containing mupirocin.41,42

Preoperative antibiotics

The use of preoperative antibiotics has been shown to reduce the risk of infection.43 In a large prospective cohort of patients undergoing a de novo or secondary CIED procedure, the use of perioperative antibiotics was negatively associated with the risk of CIED infection.13 This was later confirmed by a double-blind randomized trial of 1,000 patients undergoing permanent pacemaker or ICD initial implantation or generator replacement. This study was stopped prematurely as the use of antibiotics was clearly associated with a lower risk of CIED infection.44 Therefore, prophylaxis with an antibiotic active against staphylococci before the incision is made is a class I indication to prevent infection.1

Currently, no data support giving prophylactic antibiotics after the procedure; however, the Prevention of Arrhythmia Device Infection Trial (PADIT) is currently comparing the risk of infection with conventional preoperative antibiotics vs a regimen of pre- and post-procedure antibiotics (clinicaltrial.gov: NCT01628666).

Hemostasis

Adequate hemostasis is critical, since the risk of CIED infection is 7 times greater with formation of a hematoma.45 Heparin products, especially low-molecular-weight heparin, should be avoided at the time of CIED implantation. In patients at high risk for thromboembolism who are on warfarin therapy, the continuation of warfarin is associated with a lower incidence of hematoma compared with bridging with heparin in patients undergoing CIED procedures.46 Therefore, if anticoagulation can be withheld, it is better to stop the anticoagulant before the procedure. When this is not possible or when it carries significant risk (eg, a patient with a mechanical mitral valve who needs a CIED implantation), it is better to maintain the patient on warfarin therapy with a therapeutic international normalized ratio rather than bridging with heparin products.

Antibacterial envelop and new devices

TYRX
Figure 3. The TYRX absorbable antibacterial envelope is a mesh coated with the antibiotics rifampin and minocycline, which elute off the mesh within approximately 7 days. The mesh is completely absorbed into the body in about 9 weeks.
A new development in the prevention of CIED infection is the TYRX absorbable antibacterial envelope (Medtronic Inc.) (Figure 3), a multifilament knitted mesh coated with the antibiotics rifampin and minocycline, which are released in the device pocket over 7 days. The first-generation envelope was nonabsorbable; the new product uses a fully bioabsorbable polymer that dissolves within 9 weeks. Data from nonrandomized studies using mainly the nonabsorbable version showed favorable outcomes in reducing the rate of CIED infections.47,48 The World-wide Randomized Antibiotic Envelope Infection Prevention Trial (WRAP-IT) is a large randomized clinical trial assessing the efficacy of the absorbable envelope in reducing CIED infection rates in patients undergoing CIED replacement or upgrade.49

A leadless pacemaker in the right ventricle.
Figure 4. A leadless pacemaker in the right ventricle. The left atrial appendage exclusion clip is present.
The development of new cardiac devices carries the potential of reducing certain types of infection. The subcutaneous ICD is an entirely subcutaneous system with no endovascular component, and therefore it can prevent endovascular infection, especially in patients at high risk of infection (eg, patients on hemodialysis).50 On the other hand, the leadless pacemaker is a single-chamber pacemaker deployed percutaneously in the right ventricle without the need for a pocket, thereby eliminating the risk of pocket infection (Figure 4).51,52 Whether the risk of endovascular infection will be reduced is not yet known.

 

 

CONCLUSION

CIED infection is a major complication that carries significant risk of morbidity and death. Early diagnosis and referral to a multidisciplinary treatment team is crucial to increasing the possibility of a cure. While device extraction has risks, it is nevertheless typically required for complete resolution of the infection. Large clinical trials are under way to address current knowledge gaps about CIED infection, including our understanding of the true incidence rate, risk factors, and efficacy of various implantation techniques. Future trends to minimize the risk of CIED infection include better screening, better diagnostic tools, new devices with fewer or no leads, longer battery life to minimize the need for additional procedures, and the use of supportive tools and products to minimize the risk of infection.

Cardiac implantable electronic devices (CIEDs) have become common tools to improve the quality of life and longevity of patients with cardiac disease over the last few decades.1–4 CIEDs include implantable cardioverter defibrillators (ICDs), permanent pacemakers, biventricular pacemakers providing cardiac resynchronization therapy with or without a defibrillator, subcutaneous ICDs, and implantable loop recorders. With increasing approved indications, the number of CIEDs implanted each year continues to grow. This, paired with the aging population of patients receiving devices and their medical complexity, has led to a corresponding increase in device-related complications.2,3 One of the most serious complications is CIED infection, which leads to significant morbidity and death. These infections also represent a significant cost burden to the healthcare system, with treatment costs for a CIED infection estimated at over $146,000 in 2008.5

SCOPE OF THE PROBLEM

More than half a million permanent pacemakers and ICDs are implanted each year in the United States, with more than 4 million implanted between 1993 and 2008.5 The risk of infection is 0.5% to 1%, for a first-time implantation and 1% to 5% for a device replacement or upgrade.1,2,5–9 These infections can involve the generator pocket, bloodstream, or cardiac structures, leading to infective endocarditis.10 The timing of CIED infection appears to be bimodal in distribution: early infections usually occur as a result of the implantation procedure itself, whereas late infections occur in patients who are generally unwell or because of an insidious process that eventually crosses a threshold of clinical significance.3,11,12

Incidence and risk factors

Klug et al13 investigated the incidence rate and risk factors of CIED infection prospectively in a large cohort of patients from 44 centers who underwent CIED implantation. Of 6,319 procedures, 4,465 were first implants and the other 1,854 were a replacement or revision; 42 patients (0.68%) developed CIED infection by 12 months after the procedure, and the incidence of infection in replacement or revision cases was nearly twice the rate found in first implants.13 Risk factors for CIED infection included renal failure, heart failure, diabetes, and fever within last 24 hours before CIED implantation.14 The Implantable Cardiac Pulse Generator Replacement (REPLACE) registry found the 6-month incidence rate of CIED infection to be 1.4% after CIED replacement.6

Recently, there has been concern that the rate of newly infected CIEDs has outpaced the rate of newly implanted ones.5,15 Voigt et al15 reported a 12% increase in the rate of CIED implantation from 2004 to 2006 and an out-of-proportion 57% increase in the rate of CIED infection. A review from 2011 confirmed these findings, showing the annual CIED implantation incidence increased an average of 4.7% per year between 1993 and 2008.5 This was probably driven by clinical trials that broadened the indications for ICD implantation for primary prevention.16–19 Between 1993 and 2008, the rate of newly implanted devices increased by 96%, while the rate for newly infected CIEDs increased by 210%; the majority of this increase occurred after 2004.5 The study showed that comorbidities in patients receiving CIEDs increased sharply starting in 2004—alluding to the contribution of comorbid medical conditions such as renal failure, respiratory failure, heart failure, and diabetes to infection risk.5

However, a major obstacle to defining the true incidence rate of CIED infection is the lack of a clear denominator. CIED infection is not limited to the first few months after implantation. In fact, over half of these patients present more than 1 year after the last CIED intervention.12 Therefore, the number of patients at risk continues to grow each year and includes patients who underwent implantation that year or before, making it very difficult to compare infection rates. Additionally, the lack of a clear definition of CIED infection and the variations in duration of follow-up in different studies make it difficult to accurately assess the incidence of CIED infection.

PATHOGENESIS

Pathogens identified in 816 patients with lead extraction or device removal for CIED infection
A CIED can become infected at the time of implantation or pocket revision. The infection can then track along the endovascular portion of the leads resulting in endovascular infection and possibly endocarditis. A CIED can also become infected as a result of the hematogenous seeding of the leads or pocket during an episode of bacteremia. Most of these infections (70%) are caused by staphylococcal species, and many are becoming resistant to methicillin.12 Other species include gram-negative organisms (9%), enterococci (4.2%), streptococci (2.5%), and fungi (1%) (Table 1). Despite clear evidence of clinical CIED infection, the cultures remain negative in about 13% of cases, perhaps because of the unfortunately common practice of starting antibiotic therapy before obtaining cultures or because of the need to incubate culture samples for a longer duration.12 A longer incubation time is particularly important for infections involving Proprionibacterium acnes, an aerobic gram-positive rod commonly associated with acne vulgaris.20

 

 

DIAGNOSIS

Prompt and accurate diagnosis of CIED infection is critical as it allows for early management with antibiotic therapy and device removal. As the number of CIED implantations increases, providers on the front lines—emergency, family practice, and internal medicine physicians—will play an increasing role in recognizing and diagnosing CIED infection. Patients with CIED infection present with a range of signs and symptoms including fever, chills, erythema, swelling, drainage, tenderness, malaise, erosion, and warmth of the skin overlying the generator pocket.2 In 55% of cases, patients present with localized pocket infection, while the remaining patients have signs of an endovascular infection without obvious pocket involvement.12 Localized pocket infection is more common during the first year after device implantation. CIED-associated endovascular infections occur more commonly in patients with multiple comorbidities including diabetes, renal failure, prior heart valve operation, rheumatic heart disease, and prior bloodstream infection.2 Despite the theoretical divide in CIED infections (endovascular vs pocket), overlap is common: many patients with pocket infection show evidence of bacteremia and vegetations on the leads.

Pocket infection after placement of a cardiac implantable electronic device can present as erythema and drainage (A); swelling, skin necrosis, and eschar formation (B); and erythema, swelling, and bullae formation (C).
Figure 1. Pocket infection after placement of a cardiac implantable electronic device can present as erythema and drainage (A); swelling, skin necrosis, and eschar formation (B); and erythema, swelling, and bullae formation (C).
Physical examination of the pocket is critical as it may reveal visible signs of infection and support the diagnosis of localized pocket infection (Figure 1). Blood cultures are essential and should be collected before starting antibiotic therapy. Culture results assist in the diagnosis of CIED infection and also help identify the microorganism involved, and this information helps tailor the choice and duration of antibiotic therapy. Echocardiography (transthoracic and transesophageal) can assist the clinician in the diagnosis of CIED infection but requires careful interpretation because some patients with no signs or symptoms of infection can have small fibrinous strands or thrombi attached to the CIED leads.14 These findings should only be interpreted in correlation to the clinical presentation.

Diagnosing pocket infection from the physical examination can be difficult due to the often subtle manifestations of the underlying pathophysiology and because visible changes to the pocket can occur over weeks and months. Furthermore, differentiating superficial infection, hematoma, seroma, and allergic reactions from deep pocket infection can be challenging. In cases when the diagnosis is not clear and there are no systemic findings of infection, conservative management with close follow-up is reasonable. Similarly, the diagnosis of endovascular infection is sometimes delayed because the symptoms are not very specific or because of a lack of awareness of the presence of a CIED and its role in endovascular infection.

MANAGEMENT

A multidisciplinary approach involving cardiology, infectious disease, electrophysiology, and cardiothoracic surgery teams is required to optimize outcomes in patients with CIED infection. CIED infection is particularly difficult to treat with antibiotic therapy alone because it involves infection of an implanted device and an associated biofilm that is resistant to the effects of antibiotics. Once infection is confirmed, antibiotic therapy serves as an adjunct to the complete removal of the hardware. Most patients receive 2 weeks of intravenous antibiotics after removal of an infected CIED, with longer courses for patients with Staphylococcus aureus infection or documented endocarditis.21

Infectious disease consultation is paramount in order to choose the appropriate type and duration of antibiotic therapy. Conservative approaches that involve using antibiotics alone or incomplete system removal have high failure rates with high rates of morbidity and mortality.13,21–28 However, chronic antibiotic suppressive therapy may be considered as a palliative measure for patients who are not candidates for lead extraction.

DEVICE REMOVAL

Confirmation of CIED infection is a class I indication for device removal and the patient should be referred to an electrophysiologist. Transvenous lead extraction (TLE) is a percutaneous procedure performed by the electrophysiologist in the electrophysiology laboratory or hybrid operating room with cardiothoracic surgery support, and it is generally performed under general anesthesia with invasive hemodynamic monitoring. After opening and debriding the infected pocket, the generator is disconnected from the leads. After the lead tips are unscrewed from the myocardium, gentle traction is applied to determine if the leads can easily be removed. If traction is unsuccessful, additional tools (both powered or mechanical sheaths) are used to complete the lead extraction29; the goal is to lyse and free the fibrotic attachments between parallel leads and between the leads and vessel wall or the myocardium. Once the lead is freed from the adhesions it can be removed safely.

In a patient with endocarditis after cardiac implantable electronic device placement, transthoracic echocardiography shows a large vegetation near the right atrium, right ventricle, and across the tricuspid valve.
Figure 2. In a patient with endocarditis after cardiac implantable electronic device placement, transthoracic echocardiography shows a large vegetation (V) near the right atrium (RA), right ventricle (RV), and across the tricuspid valve (TV). This required surgical extraction of the organized vegetation along with the device and leads.
The incidence of major complications with lead extraction is low (1.8%), but the procedure can be life-threatening.30 Major complications include cardiac avulsion, vascular laceration, pericardial effusion, tamponade, hemothorax, valve injury, and death during the procedure.30 Risk factors for major complications with TLE include renal failure, low body mass index, and the presence of a defibrillator coil on the lead.30,31 In a large cohort of more than 3,000 patients requiring 6,000 TLE procedures at our tertiary care center, the incidence of catastrophic complications that required emergency cardiac surgery or vascular intervention was 0.8%.32 Many of these patients were rescued through emergency surgical repair of a venous laceration or cardiac perforation but still had an in-hospital mortality rate of 36%. Surgical lead extraction is usually performed if percutaneous lead extraction has failed, if epicardial leads are present, if large vegetations are attached to the leads, or if surgery is warranted for valvular involvement with endocarditis (Figure 2).14

 

 

REIMPLANTATION

The need for reimplantation after removal of an infected CIED should be thought about before the extraction. In general, extracting an infected CIED should be viewed as an opportunity to reassess the need for the device. Almost one-third of patients who undergo extraction of infected CIED do not require immediate reimplantation.2 This could be due to reversal of the initial indication, emergence of new clinical conditions, patient preference, or the lack of an absolute indication. If reimplantation is necessary, the new device is typically placed on the opposite side of the chest from the previously infected pocket site after blood cultures are negative for at least 72 hours.21

CIED INFECTION MORTALITY

Despite proper management with CIED removal supported by antibiotic therapy, CIED infection carries a high risk of death. The 30-day mortality is estimated to be between 5% and 6%.33 In a large case series of 412 CIED extractions, there were 19 in-hospital deaths. Of these 19 deaths, 2 were related to the extraction itself with the other 17 related to sepsis, multiorgan failure, stroke, renal failure, or heart failure.2 The 1-year mortality rate is also increased for this population; recent data show 1-year mortality rates of 8% to 17% despite device removal and antibiotic therapy.2,34,35 This increased mortality rate was also demonstrated in a large cohort of Medicare patients undergoing CIED procedures.36 Medicare patients with CIED infection had double the risk of death at 1 year compared with patients without infection.36

Risk factors for death at 1 year include worse baseline functional status, renal failure, and type of infection; eg, endovascular infection carries a risk of death 2 times higher than pocket infection.37

PREVENTION

Because CIED infection carries significant short-term and long-term mortality rates despite optimal management, the best strategy is prevention. Preventing CIED infection begins with the decision to implant a device with careful assessment of the indication, the timing of the procedure, and the patient’s clinical status. CIED procedures are performed under strict sterile surgical techniques with great attention to the incision and proper closure. Surgical data favor the use of chlorhexidine-alcohol solutions for skin preparation compared with povidone-iodine solutions to prevent both superficial and deep surgical wound infections.38 However, recent studies showed no significant difference between the 2 preparation methods in reducing rates of CIED infection.39,40 In individuals colonized with S aureus, the risk of CIED infection can be reduced using a body wash containing chlorhexidine and a nasal spray containing mupirocin.41,42

Preoperative antibiotics

The use of preoperative antibiotics has been shown to reduce the risk of infection.43 In a large prospective cohort of patients undergoing a de novo or secondary CIED procedure, the use of perioperative antibiotics was negatively associated with the risk of CIED infection.13 This was later confirmed by a double-blind randomized trial of 1,000 patients undergoing permanent pacemaker or ICD initial implantation or generator replacement. This study was stopped prematurely as the use of antibiotics was clearly associated with a lower risk of CIED infection.44 Therefore, prophylaxis with an antibiotic active against staphylococci before the incision is made is a class I indication to prevent infection.1

Currently, no data support giving prophylactic antibiotics after the procedure; however, the Prevention of Arrhythmia Device Infection Trial (PADIT) is currently comparing the risk of infection with conventional preoperative antibiotics vs a regimen of pre- and post-procedure antibiotics (clinicaltrial.gov: NCT01628666).

Hemostasis

Adequate hemostasis is critical, since the risk of CIED infection is 7 times greater with formation of a hematoma.45 Heparin products, especially low-molecular-weight heparin, should be avoided at the time of CIED implantation. In patients at high risk for thromboembolism who are on warfarin therapy, the continuation of warfarin is associated with a lower incidence of hematoma compared with bridging with heparin in patients undergoing CIED procedures.46 Therefore, if anticoagulation can be withheld, it is better to stop the anticoagulant before the procedure. When this is not possible or when it carries significant risk (eg, a patient with a mechanical mitral valve who needs a CIED implantation), it is better to maintain the patient on warfarin therapy with a therapeutic international normalized ratio rather than bridging with heparin products.

Antibacterial envelop and new devices

TYRX
Figure 3. The TYRX absorbable antibacterial envelope is a mesh coated with the antibiotics rifampin and minocycline, which elute off the mesh within approximately 7 days. The mesh is completely absorbed into the body in about 9 weeks.
A new development in the prevention of CIED infection is the TYRX absorbable antibacterial envelope (Medtronic Inc.) (Figure 3), a multifilament knitted mesh coated with the antibiotics rifampin and minocycline, which are released in the device pocket over 7 days. The first-generation envelope was nonabsorbable; the new product uses a fully bioabsorbable polymer that dissolves within 9 weeks. Data from nonrandomized studies using mainly the nonabsorbable version showed favorable outcomes in reducing the rate of CIED infections.47,48 The World-wide Randomized Antibiotic Envelope Infection Prevention Trial (WRAP-IT) is a large randomized clinical trial assessing the efficacy of the absorbable envelope in reducing CIED infection rates in patients undergoing CIED replacement or upgrade.49

A leadless pacemaker in the right ventricle.
Figure 4. A leadless pacemaker in the right ventricle. The left atrial appendage exclusion clip is present.
The development of new cardiac devices carries the potential of reducing certain types of infection. The subcutaneous ICD is an entirely subcutaneous system with no endovascular component, and therefore it can prevent endovascular infection, especially in patients at high risk of infection (eg, patients on hemodialysis).50 On the other hand, the leadless pacemaker is a single-chamber pacemaker deployed percutaneously in the right ventricle without the need for a pocket, thereby eliminating the risk of pocket infection (Figure 4).51,52 Whether the risk of endovascular infection will be reduced is not yet known.

 

 

CONCLUSION

CIED infection is a major complication that carries significant risk of morbidity and death. Early diagnosis and referral to a multidisciplinary treatment team is crucial to increasing the possibility of a cure. While device extraction has risks, it is nevertheless typically required for complete resolution of the infection. Large clinical trials are under way to address current knowledge gaps about CIED infection, including our understanding of the true incidence rate, risk factors, and efficacy of various implantation techniques. Future trends to minimize the risk of CIED infection include better screening, better diagnostic tools, new devices with fewer or no leads, longer battery life to minimize the need for additional procedures, and the use of supportive tools and products to minimize the risk of infection.

References
  1. Baddour LM, Epstein AE, Erickson CC, et al; American Heart Association Rheumatic Fever, Endocarditis, and Kawasaki Disease Committee of the Council on Cardiovascular Disease in the Young; Council on Cardiovascular Surgery and Anesthesia; Council on Cardiovascular Nursing; Council on Clinical Cardiology; and the Interdisciplinary Council on Quality of Care and Outcomes Research. Update on cardiovascular implantable electronic device infections and their management: a scientific statement from the American Heart Association. Circulation 2010; 121:458–477.
  2. Tarakji KG, Chan EJ, Cantillon DJ, et al. Cardiac implantable electronic device infections: presentation, management, and patient outcomes. Heart Rhythm 2010; 7:1043–1047.
  3. Baddour LM. Cardiac device infection—or not. Circulation 2010; 121:1686–1687.
  4. Kusumoto FM, Schoenfeld MH, Wilkoff BL, et al. 2017 HRS expert consensus statement on cardiovascular implantable electronic device lead management and extraction. Heart Rhythm 2017; Sept 15. pii: S1547-5271(17)31080-9. doi:10.1016/j.hrthm.2017.09.001. [Epub ahead of print]
  5. Greenspon AJ, Patel JD, Lau E, et al. 16-Year trends in the infection burden for pacemakers and implantable cardioverter-defibrillators in the United States: 1993 to 2008. J Am Coll Cardiol 2011; 58:1001–1006.
  6. Poole JE, Gleva MJ, Mela T, et al; REPLACE Registry Investigators. Complication rates associated with pacemaker or implantable cardioverter-defibrillator generator replacements and upgrade procedures: results from the REPLACE registry. Circulation 2010; 122:1553–1561.
  7. Mela T, McGovern BA, Garan H, et al. Long-term infection rates associated with the pectoral versus abdominal approach to cardioverter-defibrillator implants. Am J Cardiol 2001; 88:750–753.
  8. de Bie MK, van Rees JB, Thijssen J, et al. Cardiac device infections are associated with a significant mortality risk. Heart Rhythm 2012; 9:494–498.
  9. Polyzos KA, Konstantelias AA, Falagas ME. Risk factors for cardiac implantable electronic device infection: a systematic review and meta-analysis. Europace 2015; 17:767–777.
  10. Deharo J-C, Quatre A, Mancini J, et al. Long-term outcomes following infection of cardiac implantable electronic devices: a prospective matched cohort study. Heart 2012; 98:724–731.
  11. Sohail MR, Hussain S, Le KY, et al; Mayo Cardiovascular Infections Study Group. Risk factors associated with early- versus late-onset implantable cardioverter-defibrillator infections. J Interv Card Electrophysiol 2011; 31:171–183.
  12. Hussein AA, Baghdy Y, Wazni OM, et al. Microbiology of cardiac implantable electronic device infections. JACC Clin Electrophysiol 2016; 2:498–505.
  13. Klug D, Balde M, Pavin D, et al; PEOPLE Study Group. Risk factors related to infections of implanted pacemakers and cardioverter-defibrillators: results of a large prospective study. Circulation 2007; 116:1349–1355.
  14. Tarakji KG, Wilkoff BL. Management of cardiac implantable electronic device infections: the challenges of understanding the scope of the problem and its associated mortality. Expert Rev Cardiovasc Ther 2013; 11:607–616.
  15. Voigt A, Shalaby A, Saba S. Continued rise in rates of cardiovascular implantable electronic device infections in the United States: temporal trends and causative insights. Pacing Clin Electrophysiol 2010; 33:414–419.
  16. Bardy GH, Lee KL, Mark DB, et al; Sudden Cardiac Death in Heart Failure Trial (SCD-HeFT) Investigators. Amiodarone or an implantable cardioverter-defibrillator for congestive heart failure. N Engl J Med 2005; 352:225–237.
  17. Moss AJ, Zareba W, Hall WJ, et al; Multicenter Automatic Defibrillator Implantation Trial II Investigators. Prophylactic implantation of a defibrillator in patients with myocardial infarction and reduced ejection fraction. N Engl J Med 2002; 346:877–883.
  18. Kadish A, Dyer A, Daubert JP, et al; Defibrillators in Non-Ischemic Cardiomyopathy Treatment Evaluation (DEFINITE) Investigators. Prophylactic defibrillator implantation in patients with nonischemic dilated cardiomyopathy. N Engl J Med 2004; 350:2151–2158.
  19. Buxton AE, Lee KL, Fisher JD, Josephson ME, Prystowsky EN, Hafley G; Multicenter Unsustained Tachycardia Trial Investigators. A randomized study of the prevention of sudden death in patients with coronary artery disease. N Engl J Med 1999; 341:1882–1890.
  20. Abdulmassih R, Makadia J, Como J, Paulson M, Min Z, Bhanot N. Propionibacterium acnes: Time-to-positivity in standard bacterial culture from different anatomical sites. J Clin Med Res 2016; 8:916–918.
  21. Sohail MR, Uslan DZ, Khan AH, et al. Management and outcome of permanent pacemaker and implantable cardioverter-defibrillator infections. J Am Coll Cardiol 2007; 49:1851–1859.
  22. Cacoub P, Leprince P, Nataf P, et al. Pacemaker infective endocarditis. Am J Cardiol 1998; 82:480–484.
  23. Chua JD, Wilkoff BL, Lee I, Juratli N, Longworth DL, Gordon SM. Diagnosis and management of infections involving implantable electrophysiologic cardiac devices. Ann Intern Med 2000; 133:604–608.
  24. Bracke FA, Meijer A, van Gelder LM. Pacemaker lead complications: when is extraction appropriate and what can we learn from published data? Heart 2001; 85:254–259.
  25. Camus C, Leport C, Raffi F, Michelet C, Cartier F, Vilde JL. Sustained bacteremia in 26 patients with a permanent endocardial pacemaker: assessment of wire removal. Clin Infect Dis 1993; 17:46–55.
  26. Molina JE. Undertreatment and overtreatment of patients with infected antiarrhythmic implantable devices. Ann Thorac Surg 1997; 63:504–509.
  27. Viganego F, O’Donoghue S, Eldadah Z, et al. Effect of early diagnosis and treatment with percutaneous lead extraction on survival in patients with cardiac device infections. Am J Cardiol 2012; 109:1466–1471.
  28. Le KY, Sohail MR, Friedman PA, et al; Mayo Cardiovascular Infections Study Group. Impact of timing of device removal on mortality in patients with cardiovascular implantable electronic device infections. Heart Rhythm 2011; 8:1678–1685.
  29. Wazni O, Wilkoff BL. Considerations for cardiac device lead extraction. Nat Rev Cardiol 2016; 13:221–229.
  30. Brunner MP, Cronin EM, Duarte VE, et al. Clinical predictors of adverse patient outcomes in an experience of more than 5000 chronic endovascular pacemaker and defibrillator lead extractions. Heart Rhythm 2014; 11:799–805.
  31. Wazni O, Epstein LM, Carrillo RG, et al. Lead extraction in the contemporary setting: the LExICon study: an observational retrospective study of consecutive laser lead extractions. J Am Coll Cardiol 2010; 55:579–586.
  32. Brunner MP, Cronin EM, Wazni O, et al. Outcomes of patients requiring emergent surgical or endovascular intervention for catastrophic complications during transvenous lead extraction. Heart Rhythm 2014; 11:419–425.
  33. Habib A, Le KY, Baddour LM, et al; for the Mayo Cardiovascular Infections Study Group. Predictors of mortality in patients with cardiovascular implantable electronic device infections. Am J Cardiol 2013; 111:874–879.
  34. Baman TS, Gupta SK, Valle JA, Yamada E. Risk factors for mortality in patients with cardiac device-related infection. Circ Arrhythm Electrophysiol 2009; 2:129–134.
  35. Deckx S, Marynissen T, Rega F, et al. Predictors of 30-day and 1-year mortality after transvenous lead extraction: a single-centre experience. Europace 2014; 16:1218–1225.
  36. Sohail MR, Henrikson CA, Braid-Forbes MJ, Forbes KF, Lerner DJ. Increased long-term mortality in patients with cardiovascular implantable electronic device infections. Pacing Clin Electrophysiol 2015; 38:231–239.
  37. Tarakji KG, Wazni OM, Harb S, Hsu A, Saliba W, Wilkoff BL. Risk factors for 1-year mortality among patients with cardiac implantable electronic device infection undergoing transvenous lead extraction: the impact of the infection type and the presence of vegetation on survival. Europace 2014; 16:1490–1495.
  38. Darouiche RO, Wall MJ Jr., Itani KM, et al. Chlorhexidine—alcohol versus povidone—iodine for surgical-site antisepsis. N Engl J Med 2010; 362:18–26.
  39. Qintar M, Zardkoohi O, Hammadah M, et al. The impact of changing antiseptic skin preparation agent used for cardiac implantable electronic device (CIED) procedures on the risk of infection. Pacing Clin Electrophysiol 2015; 38:240–246.
  40. Da Costa A, Tulane C, Dauphinot V, et al. Preoperative skin antiseptics for prevention of cardiac implantable electronic device infections: a historical-controlled interventional trial comparing aqueous against alcoholic povidone-iodine solutions. Europace 2015; 17:1092–1098.
  41. Padfield GJ, Steinberg C, Bennett MT, et al. Preventing cardiac implantable electronic device infections. Heart Rhythm 2015; 12:2344–2356.
  42. Bode LGM, Kluytmans JAJW, Wertheim HFL, et al. Preventing surgical-site infections in nasal carriers of Staphylococcus aureus. N Engl J Med 2010; 362:9–17.
  43. Da Costa A, Kirkorian G, Cucherat M, et al. Antibiotic prophylaxis for permanent pacemaker implantation: a meta-analysis. Circulation 1998; 97:1796–1801.
  44. de Oliveira JC, Martinelli M, Nishioka SADO, et al. Efficacy of antibiotic prophylaxis before the implantation of pacemakers and cardioverter-defibrillators: results of a large, prospective, randomized, double-blinded, placebo-controlled trial. Circ Arrhythm Electrophysiol 2009; 2:29–34.
  45. Essebag V, Verma A, Healey JS, et al; BRUISE CONTROL Investigators. Clinically significant pocket hematoma increases long-term risk of device infection: BRUISE CONTROL INFECTION study. J Am Coll Cardiol 2016; 67:1300–1308.
  46. Birnie DH, Healey JS, Wells GA, et al; BRUISE CONTROL Investigators. Pacemaker or defibrillator surgery without interruption of anticoagulation. N Engl J Med 2013; 368:2084–2093.
  47. Henrikson CA, Sohail MR, Acosta H, et al. Antibacterial envelope is associated with low infection rates after implantable cardioverter-defibrillator and cardiac resynchronization therapy device replacement: results of the Citadel and Centurion studies. 2017 http://dx.doi.org/10.1016/j.jacep.2017.02.016
  48. Mittal S, Shaw RE, Michel K, et al. Cardiac implantable electronic device infections: incidence, risk factors, and the effect of the AigisRx antibacterial envelope. Heart Rhythm 2014; 11:595–601.
  49. Tarakji KG, Mittal S, Kennergren C, et al. Worldwide Randomized Antibiotic EnveloPe Infection PrevenTion Trial (WRAP-IT). Am Heart J 2016; 180:12–B21.
  50. Burke MC, Gold MR, Knight BP, et al. Safety and efficacy of the totally subcutaneous implantable defibrillator: 2-year results from a pooled analysis of the IDE study and EFFORTLESS registry. J Am Coll Cardiol 2015; 65:1605–1615.
  51. Reddy VY, Exner DV, Cantillon DJ, et al; LEADLESS II Study Investigators. Percutaneous implantation of an entirely intracardiac leadless pacemaker. N Engl J Med 2015; 373:1125–1135.
  52. Reynolds D, Duray GZ, Omar R, et al; Micra Transcatheter Pacing Study Group. A leadless intracardiac transcatheter pacing system. N Engl J Med 2016; 374:533–541.
References
  1. Baddour LM, Epstein AE, Erickson CC, et al; American Heart Association Rheumatic Fever, Endocarditis, and Kawasaki Disease Committee of the Council on Cardiovascular Disease in the Young; Council on Cardiovascular Surgery and Anesthesia; Council on Cardiovascular Nursing; Council on Clinical Cardiology; and the Interdisciplinary Council on Quality of Care and Outcomes Research. Update on cardiovascular implantable electronic device infections and their management: a scientific statement from the American Heart Association. Circulation 2010; 121:458–477.
  2. Tarakji KG, Chan EJ, Cantillon DJ, et al. Cardiac implantable electronic device infections: presentation, management, and patient outcomes. Heart Rhythm 2010; 7:1043–1047.
  3. Baddour LM. Cardiac device infection—or not. Circulation 2010; 121:1686–1687.
  4. Kusumoto FM, Schoenfeld MH, Wilkoff BL, et al. 2017 HRS expert consensus statement on cardiovascular implantable electronic device lead management and extraction. Heart Rhythm 2017; Sept 15. pii: S1547-5271(17)31080-9. doi:10.1016/j.hrthm.2017.09.001. [Epub ahead of print]
  5. Greenspon AJ, Patel JD, Lau E, et al. 16-Year trends in the infection burden for pacemakers and implantable cardioverter-defibrillators in the United States: 1993 to 2008. J Am Coll Cardiol 2011; 58:1001–1006.
  6. Poole JE, Gleva MJ, Mela T, et al; REPLACE Registry Investigators. Complication rates associated with pacemaker or implantable cardioverter-defibrillator generator replacements and upgrade procedures: results from the REPLACE registry. Circulation 2010; 122:1553–1561.
  7. Mela T, McGovern BA, Garan H, et al. Long-term infection rates associated with the pectoral versus abdominal approach to cardioverter-defibrillator implants. Am J Cardiol 2001; 88:750–753.
  8. de Bie MK, van Rees JB, Thijssen J, et al. Cardiac device infections are associated with a significant mortality risk. Heart Rhythm 2012; 9:494–498.
  9. Polyzos KA, Konstantelias AA, Falagas ME. Risk factors for cardiac implantable electronic device infection: a systematic review and meta-analysis. Europace 2015; 17:767–777.
  10. Deharo J-C, Quatre A, Mancini J, et al. Long-term outcomes following infection of cardiac implantable electronic devices: a prospective matched cohort study. Heart 2012; 98:724–731.
  11. Sohail MR, Hussain S, Le KY, et al; Mayo Cardiovascular Infections Study Group. Risk factors associated with early- versus late-onset implantable cardioverter-defibrillator infections. J Interv Card Electrophysiol 2011; 31:171–183.
  12. Hussein AA, Baghdy Y, Wazni OM, et al. Microbiology of cardiac implantable electronic device infections. JACC Clin Electrophysiol 2016; 2:498–505.
  13. Klug D, Balde M, Pavin D, et al; PEOPLE Study Group. Risk factors related to infections of implanted pacemakers and cardioverter-defibrillators: results of a large prospective study. Circulation 2007; 116:1349–1355.
  14. Tarakji KG, Wilkoff BL. Management of cardiac implantable electronic device infections: the challenges of understanding the scope of the problem and its associated mortality. Expert Rev Cardiovasc Ther 2013; 11:607–616.
  15. Voigt A, Shalaby A, Saba S. Continued rise in rates of cardiovascular implantable electronic device infections in the United States: temporal trends and causative insights. Pacing Clin Electrophysiol 2010; 33:414–419.
  16. Bardy GH, Lee KL, Mark DB, et al; Sudden Cardiac Death in Heart Failure Trial (SCD-HeFT) Investigators. Amiodarone or an implantable cardioverter-defibrillator for congestive heart failure. N Engl J Med 2005; 352:225–237.
  17. Moss AJ, Zareba W, Hall WJ, et al; Multicenter Automatic Defibrillator Implantation Trial II Investigators. Prophylactic implantation of a defibrillator in patients with myocardial infarction and reduced ejection fraction. N Engl J Med 2002; 346:877–883.
  18. Kadish A, Dyer A, Daubert JP, et al; Defibrillators in Non-Ischemic Cardiomyopathy Treatment Evaluation (DEFINITE) Investigators. Prophylactic defibrillator implantation in patients with nonischemic dilated cardiomyopathy. N Engl J Med 2004; 350:2151–2158.
  19. Buxton AE, Lee KL, Fisher JD, Josephson ME, Prystowsky EN, Hafley G; Multicenter Unsustained Tachycardia Trial Investigators. A randomized study of the prevention of sudden death in patients with coronary artery disease. N Engl J Med 1999; 341:1882–1890.
  20. Abdulmassih R, Makadia J, Como J, Paulson M, Min Z, Bhanot N. Propionibacterium acnes: Time-to-positivity in standard bacterial culture from different anatomical sites. J Clin Med Res 2016; 8:916–918.
  21. Sohail MR, Uslan DZ, Khan AH, et al. Management and outcome of permanent pacemaker and implantable cardioverter-defibrillator infections. J Am Coll Cardiol 2007; 49:1851–1859.
  22. Cacoub P, Leprince P, Nataf P, et al. Pacemaker infective endocarditis. Am J Cardiol 1998; 82:480–484.
  23. Chua JD, Wilkoff BL, Lee I, Juratli N, Longworth DL, Gordon SM. Diagnosis and management of infections involving implantable electrophysiologic cardiac devices. Ann Intern Med 2000; 133:604–608.
  24. Bracke FA, Meijer A, van Gelder LM. Pacemaker lead complications: when is extraction appropriate and what can we learn from published data? Heart 2001; 85:254–259.
  25. Camus C, Leport C, Raffi F, Michelet C, Cartier F, Vilde JL. Sustained bacteremia in 26 patients with a permanent endocardial pacemaker: assessment of wire removal. Clin Infect Dis 1993; 17:46–55.
  26. Molina JE. Undertreatment and overtreatment of patients with infected antiarrhythmic implantable devices. Ann Thorac Surg 1997; 63:504–509.
  27. Viganego F, O’Donoghue S, Eldadah Z, et al. Effect of early diagnosis and treatment with percutaneous lead extraction on survival in patients with cardiac device infections. Am J Cardiol 2012; 109:1466–1471.
  28. Le KY, Sohail MR, Friedman PA, et al; Mayo Cardiovascular Infections Study Group. Impact of timing of device removal on mortality in patients with cardiovascular implantable electronic device infections. Heart Rhythm 2011; 8:1678–1685.
  29. Wazni O, Wilkoff BL. Considerations for cardiac device lead extraction. Nat Rev Cardiol 2016; 13:221–229.
  30. Brunner MP, Cronin EM, Duarte VE, et al. Clinical predictors of adverse patient outcomes in an experience of more than 5000 chronic endovascular pacemaker and defibrillator lead extractions. Heart Rhythm 2014; 11:799–805.
  31. Wazni O, Epstein LM, Carrillo RG, et al. Lead extraction in the contemporary setting: the LExICon study: an observational retrospective study of consecutive laser lead extractions. J Am Coll Cardiol 2010; 55:579–586.
  32. Brunner MP, Cronin EM, Wazni O, et al. Outcomes of patients requiring emergent surgical or endovascular intervention for catastrophic complications during transvenous lead extraction. Heart Rhythm 2014; 11:419–425.
  33. Habib A, Le KY, Baddour LM, et al; for the Mayo Cardiovascular Infections Study Group. Predictors of mortality in patients with cardiovascular implantable electronic device infections. Am J Cardiol 2013; 111:874–879.
  34. Baman TS, Gupta SK, Valle JA, Yamada E. Risk factors for mortality in patients with cardiac device-related infection. Circ Arrhythm Electrophysiol 2009; 2:129–134.
  35. Deckx S, Marynissen T, Rega F, et al. Predictors of 30-day and 1-year mortality after transvenous lead extraction: a single-centre experience. Europace 2014; 16:1218–1225.
  36. Sohail MR, Henrikson CA, Braid-Forbes MJ, Forbes KF, Lerner DJ. Increased long-term mortality in patients with cardiovascular implantable electronic device infections. Pacing Clin Electrophysiol 2015; 38:231–239.
  37. Tarakji KG, Wazni OM, Harb S, Hsu A, Saliba W, Wilkoff BL. Risk factors for 1-year mortality among patients with cardiac implantable electronic device infection undergoing transvenous lead extraction: the impact of the infection type and the presence of vegetation on survival. Europace 2014; 16:1490–1495.
  38. Darouiche RO, Wall MJ Jr., Itani KM, et al. Chlorhexidine—alcohol versus povidone—iodine for surgical-site antisepsis. N Engl J Med 2010; 362:18–26.
  39. Qintar M, Zardkoohi O, Hammadah M, et al. The impact of changing antiseptic skin preparation agent used for cardiac implantable electronic device (CIED) procedures on the risk of infection. Pacing Clin Electrophysiol 2015; 38:240–246.
  40. Da Costa A, Tulane C, Dauphinot V, et al. Preoperative skin antiseptics for prevention of cardiac implantable electronic device infections: a historical-controlled interventional trial comparing aqueous against alcoholic povidone-iodine solutions. Europace 2015; 17:1092–1098.
  41. Padfield GJ, Steinberg C, Bennett MT, et al. Preventing cardiac implantable electronic device infections. Heart Rhythm 2015; 12:2344–2356.
  42. Bode LGM, Kluytmans JAJW, Wertheim HFL, et al. Preventing surgical-site infections in nasal carriers of Staphylococcus aureus. N Engl J Med 2010; 362:9–17.
  43. Da Costa A, Kirkorian G, Cucherat M, et al. Antibiotic prophylaxis for permanent pacemaker implantation: a meta-analysis. Circulation 1998; 97:1796–1801.
  44. de Oliveira JC, Martinelli M, Nishioka SADO, et al. Efficacy of antibiotic prophylaxis before the implantation of pacemakers and cardioverter-defibrillators: results of a large, prospective, randomized, double-blinded, placebo-controlled trial. Circ Arrhythm Electrophysiol 2009; 2:29–34.
  45. Essebag V, Verma A, Healey JS, et al; BRUISE CONTROL Investigators. Clinically significant pocket hematoma increases long-term risk of device infection: BRUISE CONTROL INFECTION study. J Am Coll Cardiol 2016; 67:1300–1308.
  46. Birnie DH, Healey JS, Wells GA, et al; BRUISE CONTROL Investigators. Pacemaker or defibrillator surgery without interruption of anticoagulation. N Engl J Med 2013; 368:2084–2093.
  47. Henrikson CA, Sohail MR, Acosta H, et al. Antibacterial envelope is associated with low infection rates after implantable cardioverter-defibrillator and cardiac resynchronization therapy device replacement: results of the Citadel and Centurion studies. 2017 http://dx.doi.org/10.1016/j.jacep.2017.02.016
  48. Mittal S, Shaw RE, Michel K, et al. Cardiac implantable electronic device infections: incidence, risk factors, and the effect of the AigisRx antibacterial envelope. Heart Rhythm 2014; 11:595–601.
  49. Tarakji KG, Mittal S, Kennergren C, et al. Worldwide Randomized Antibiotic EnveloPe Infection PrevenTion Trial (WRAP-IT). Am Heart J 2016; 180:12–B21.
  50. Burke MC, Gold MR, Knight BP, et al. Safety and efficacy of the totally subcutaneous implantable defibrillator: 2-year results from a pooled analysis of the IDE study and EFFORTLESS registry. J Am Coll Cardiol 2015; 65:1605–1615.
  51. Reddy VY, Exner DV, Cantillon DJ, et al; LEADLESS II Study Investigators. Percutaneous implantation of an entirely intracardiac leadless pacemaker. N Engl J Med 2015; 373:1125–1135.
  52. Reynolds D, Duray GZ, Omar R, et al; Micra Transcatheter Pacing Study Group. A leadless intracardiac transcatheter pacing system. N Engl J Med 2016; 374:533–541.
Page Number
47-53
Page Number
47-53
Publications
Publications
Article Type
Display Headline
Cardiac implantable electronic device infection
Display Headline
Cardiac implantable electronic device infection
Legacy Keywords
cardiac implantable electronic device infection, CIED infection, pacemaker, implantable cardioverter-defibrillator, MRSA, methicillin-resistant Staphylococcus aureus, VRE, vancomycin-resistant Enterococcus species, antibiotics, vegetation, Cameron Lamberg, Khaldoun Tarakji
Legacy Keywords
cardiac implantable electronic device infection, CIED infection, pacemaker, implantable cardioverter-defibrillator, MRSA, methicillin-resistant Staphylococcus aureus, VRE, vancomycin-resistant Enterococcus species, antibiotics, vegetation, Cameron Lamberg, Khaldoun Tarakji
Citation Override
Cleveland Clinic Journal of Medicine 2017 December;84(suppl 3):47-53
Inside the Article

KEY POINTS

  • CIED use is increasing, as are the number of CIED infections, which are associated with significant morbidity and mortality.
  • Prompt diagnosis of CIED infection allows for early management with antibiotics and device removal, which is typically needed for resolution of the infection.
  • Prevention of CIED infection is an important strategy, and more research is needed to inform the incidence of CIED infection, risk factors, and devices and techniques to minimize the risk of infection.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Article PDF Media

Lung transplant: Candidates for referral and the waiting list

Article Type
Changed
Mon, 10/01/2018 - 14:33
Display Headline
Lung transplant: Candidates for referral and the waiting list

Lung transplant is the therapy of choice for a growing number of patients with end-stage lung diseases. Patients receiving a lung transplant are faced with many challenges including drug toxicities, infections, and the risk of rejection.1 Despite these challenges, lung transplant may significantly prolong survival and improve quality of life for many patients.

CANDIDATES FOR LUNG TRANSPLANT

Identifying patients who are appropriate candidates for lung transplant is important to achieving favorable transplant outcomes and to maximizing life expectancy for each patient. The most recent edition of International Society for Heart and Lung Transplant (ISHLT) Guidelines for the Selection of Lung Transplant Candidates is an excellent guide to help physicians identify when to refer potential patients and to how to identify patients who are the most likely to benefit from lung transplant.2

Adults with end-stage lung disease are generally candidates for lung transplant if they meet the following criteria:

  • A greater than 50% risk of death from lung disease within 2 years if a lung transplant is not performed
  • A greater than 80% likelihood of surviving at least 90 days after the lung transplant procedure
  • A greater than 80% likelihood of a 5-year survival posttransplant if graft function is preserved.2

These can only be estimated by transplant programs and not by the referring team in most cases.

Once a patient is identified as a candidate for lung transplant, early referral of patients to a lung transplant program has several advantages and is essential for positive outcomes. Early patient referral allows for timely completion of the formal evaluation of candidacy, patient and family education, as well as the opportunity for the patient and family to raise funds or use other resources to overcome financial hurdles. Listing a patient on the transplant waitlist implies that the patient has a limited life expectancy without a lung transplant and that the risk-benefit ratio favors lung transplant since all other medical options have been exhausted.1

New candidates age 12 years and older on the lung transplant waiting list by year added.
Adapted from reference 4.
Figure 1. New candidates age 12 years and older on the lung transplant waiting list by year added.
Each year, the number of new candidates added to the lung transplant waitlist grows (Figure 1). Since 2005, the allocation of organs for transplant has shifted from a time-based system to a risk of mortality-based system. The Lung Allocation System prioritizes candidates with the highest risk of mortality. Thus, the number of sicker and older patients on the wait list has increased since the implementation of the Lung Allocation System.3 Because lung transplant is associated with significant perioperative morbidity and mortality, and older and sicker patients are being considered for listing, the contraindications and comorbidities should be vetted thoroughly prior to listing.

NONCANDIDATES FOR LUNG TRANSPLANT

There are very few absolute contraindications to lung transplant. Generally, most transplant centers in the United States agree that contraindications to lung transplant include conditions associated with increased risk of mortality, including:

  • A recent history of a major malignancy. Patients with a 2-year, disease-free interval combined with a low predicted risk of recurrence may be considered in certain cases of localized, non-melanoma skin cancer. A 5-year, disease-free survival is strongly suggested in patients with a history of breast, bladder, or kidney cancer as well as in cases of sarcoma, melanoma, lymphoma and certain hematologic disorders.
  • The presence of significant dysfunction of another major organ systems including the heart, liver, kidney, or brain unless a combined organ transplant can be considered and performed.
  • Significant coronary heart disease not amenable to revascularization or intervention prior to or at the time of lung transplant.
  • The presence of an acute medical condition including but not limited to sepsis and acute liver failure.
  • Active Mycobacterium tuberculosis and other highly virulent or highly resistant microbes that are poorly controlled pretransplant.
  • Severe obesity with a body mass index greater than 35.
  • A history of nonadherence to medical therapy, psychiatric or psychological conditions that might lead to nonadherence, poor or limited social support system, and limited functional status not amenable to rehabilitation.
  • Current substance abuse or dependence, including illicit substances, alcohol, and tobacco (nicotine-containing substances). Most centers require at least 6 months’ abstinence from illicit substances prior to being added to the lung transplant waitlist.2

CANDIDATE COMORBIDITIES

Age

Many transplant centers in the US define the age cutoff for lung transplant at 65; however, some centers may consider candidates older than 65. Advanced age by itself should not be considered a contraindication to lung transplant. However, increased age is usually associated with other comorbid conditions that may increase perioperative and long-term morbidity and mortality. As mentioned previously, the number of older candidates for lung transplant has increased. In the US, 29% of the patients on the national waiting list in 2015 were over age 65.4

Past chest surgery

It is not uncommon for lung transplant candidates to have a history of chest surgery such as lung resection, pleurodesis, or coronary artery bypass grafting. The limited literature regarding the outcomes for these patients suggests they may experience higher rates of bleeding, re-exploration, and renal dysfunction.2 However, these patients should not be excluded from lung transplant and successful transplant outcomes have been achieved in this population by experienced centers.5 In candidates with a history of chronic obstructive pulmonary disease (COPD) and lung-volume reduction surgery (LVRS), early case series indicate that these patients did well after lung transplant.6 However, more recent data demonstrate that patients with prior LVRS who undergo lung transplant experience higher rates of bleeding, worse early graft dysfunction, and worse outcomes overall.7 As with lung transplant candidates with previous chest surgery, lung transplant candidates with previous LVRS are best served by experienced transplant centers.

Hepatitis and HIV

Patients with a history of infection with hepatitis B, hepatitis C, or human immunodeficiency virus (HIV) are candidates for lung transplant at centers experienced with lung transplant in patients with these infections. Most centers advocate that patients with a history of hepatitis B or C have viral infection levels that are controlled or reduced as low as possible and that there is no evidence of portal hypertension or severe cirrhosis.8,9 In the case of HIV, patients should have controlled disease with a negative or undetectable viral load and have no current acquired immunodeficiency defining illness.10 Patients colonized with particular species of Burkholderia cepacia or Mycobacterium abscessus subspecies can be considered for lung transplant only at centers with established preoperative and postoperative protocols for these infections due to the increased risk of perioperative mortality associated with these organisms.11,12

 

 

DISEASE-SPECIFIC INDICATIONS

Chronic obstructive pulmonary disease

COPD (both non- and alpha-1 antitrypsin deficiency) is the most common indication for lung transplant and accounts for almost 32% of lung transplants worldwide.13 Patients should be referred for lung transplant when medical therapies, surgical interventions (ie, LVRS) and pulmonary rehabilitation have been maximized. In COPD, the loss of lung function occurs over a long period of time but patients are often more limited by diminished quality of life as lung function slowly declines.

Patients with COPD should be referred for lung transplant if the body mass index, airflow obstruction, dyspnea, and exercise capacity (BODE) index is 5 to 6.2 The original BODE index developed by Celli et al,14 is a scoring system from 0 to 10 with a higher score indicating more severe disease and worse survival. A score of 5 to 6 indicates an estimated mortality of 60% at 4 yrs.2,14,15 Other considerations for referral for lung transplant include the presence of hypercapnia with partial pressure of carbon dioxide greater than 50 mm Hg or higher or hypoxemia with partial pressure of oxygen less than 60 mm Hg or a forced expiratory volume at 1 sec (FEV1) less than 25% predicted.

Patients with COPD should considered for listing for lung transplant if any one of the following criteria is met: BODE index of 7 or greater; FEV1 less than 15% to 20%; 3 of more severe exacerbations during the preceding year; 1 severe exacerbation with acute hypercapnic respiratory failure; or presence of moderate to severe pulmonary hypertension.2,16

Cystic fibrosis

In patients with cystic fibrosis, lung transplant should be considered in patients with an estimated 2-year survival of less than 50% and with a New York Heart Association (NYHA) Functional Classification III or IV. Referral for lung transplant is recommended for patients with a rapid decrease in FEV1 despite optimal therapy, female patients with declining weight and lung function, colonization or infection with nontuberculous mycobacterial disease, or cystic fibrosis-related diabetes. The development of pulmonary hypertension, reduction in walk distance, increasing antibiotic resistance, acute respiratory failure requiring noninvasive ventilation, worsening nutritional status, pneumothorax, and life-threatening hemoptysis despite embolization are all indications for referral for lung transplant.

Patients with cystic fibrosis with hypoxia or hypercapnia with declining lung function, needing long-term noninvasive ventilation, having more frequent exacerbations or exhibiting a decline in functional status should be listed for lung transplant.2,17–19

Restrictive lung disease

Patients with restrictive lung diseases, including interstitial pulmonary fibrosis (usual interstitial pneumonitis, nonspecific interstitial pneumonia), or interstitial lung disease, and hypersensitivity pneumonitis, should be referred for transplant evaluation at the time of diagnosis irrespective of lung function due to the unpredictable nature of these diseases.20 Some clinicians may advocate for a trial of medical therapy with antifibrotics, but this should be done in conjunction with transplant referral.

Patients should be listed for transplant if a 10% or greater decrease in FEV1 occurred in the past 6 months (of note, even a 5% decrease in FEV1 is associated with an overall poorer prognosis and warrants consideration of listing for transplant), if the diffusing capacity of the lung for carbon monoxide decreases 15% or greater during the 6-month follow-up, or if a decline of more than 50 meters is noted on the 6-minute walking test. A documented desaturation of less than 88% or a distance of less than 250 meters on the 6-minute walking test is another indication for listing. Any evidence of secondary pulmonary hypertension on right heart catheterization or on echocardiography or hospitalization for respiratory decline are also indications for listing.21 In cases of scleroderma-associated interstitial lung disease or mixed connective tissue interstitial lung disease, similar guidelines for referral and listing should be followed.2

Pulmonary arterial hypertension

Patients with pulmonary arterial hypertension should be referred for lung transplant if any 1 of the following conditions is present: rapidly progressive disease; NYHA Functional Classification III or IV symptoms during escalating therapy; use of parenteral pulmonary arterial hypertension therapy; or known or suspected pulmonary veno-occlusive disease or pulmonary capillary hemangiomatosis.2,22

Patients with pulmonary arterial hypertension should be listed for lung transplant if any of the following are present: NYHA Functional Classification III or IV symptoms despite combination therapy; right heart catheterization demonstrating a cardiac index less than 2 L/min/m2; mean right atrial pressure greater than 15 mm Hg; 6-minute walking test less than 350 meters; or development of pericardial effusion, hemoptysis, or signs of worsening right heart failure, including renal insufficiency, rising bilirubin or evidence of ascites.2,22

BRIDGE TO TRANSPLANT

Acute respiratory decompensation may occur in some candidates for lung transplant prior to listing for transplant or while on the transplant waitlist. In patients with failure of a single lung, a bridge to transplant may be necessary until a suitable organ is available. Mechanical ventilation and extracorporeal life support (ECLS) are 2 bridge strategies for lung transplant candidates. Mechanical ventilation is the most common lung transplant bridge strategy but it is less than ideal because it can lead to deconditioning and ventilator-associated infections that can negatively impact a patient’s suitability for transplant.

ECLS techniques that allow spontaneous breathing and potentially ambulation, known as awake or ambulatory ECLS, is a popular bridge therapy. Ambulatory ECLS is used as an alternative to mechanical ventilation to avoid the complications of mechanical ventilation and allow patients to avoid sedation and participate in rehabilitation.23 Irrespective of the therapy used as a bridge to transplant, patients considered for a bridge are optimally evaluated from a medical and psychosocial perspective prior to bridge therapy.

Both bridge therapies increase the risk of infection, bleeding, and neurologic events; thus, patients need to be assessed repeatedly for these risks to determine ongoing suitability for lung transplant. It is important to note that delayed referral of patients with advanced disease or patients in an acute exacerbation negatively impacts the evaluation for lung transplant, placement on the lung transplant waitlist, outcomes, and suitability for bridge transplant strategies.

CONCLUSION

To ensure good patient outcomes, the evaluation and selection of candidates for lung transplant requires communication between referring physicians and lung transplant centers. Physicians need basic knowledge of patient conditions appropriate for lung transplant and direct communication with lung transplant centers. The workup, required testing, and timing of listing for lung transplant varies among transplant centers across the country, making communication between the referring providers and transplant centers crucial to good patient care. An open, 2-way dialogue between referring providers and transplant centers facilitates listing patients for transplant in a timely manner, reduces delays, and improves outcomes.

References
  1. Kreider M, Hadjiliadis D, Kotloff R. Candidate selection, timing of listing, and choice of procedure for lung transplantation. Clin Chest Med 2011; 32:199–211.
  2. Weill D, Benden C, Corris P, et al. A consensus document for the selection of lung transplant candidates: 2014—An update from the Pulmonary Transplant Council of the International Society of Heart and Lung Transplantation. J Heart Lung Transplant 2015; 34:1–15.
  3. Tsuang WM. Contemporary issues in lung transplant allocation practices. Curr Transplant Rep 2017; 4:238–242.
  4. Valapour M, Skeans MA, Smith JM, et al. OPTN/SRTR 2015 annual data report: lung. Am J Transplant 2017; 17(suppl 1):357–424.
  5. Omara M, Okamoto T, Arafat A, Thuita L, Blackstone EH, McCurry KR. Lung transplantation in patients who have undergone prior cardiothoracic procedures. J Heart Lung Transplant 2016; 35:1462–1470.
  6. Senbaklavaci O, Wisser W, Ozpeker C, et al. Successful lung volume reduction surgery brings patients into better condition for later lung transplantation. Eur J Cardiothorac Surg 2002; 22:363–367.
  7. Shigemura N, Gilbert S, Bhama JK et al. Lung transplantation after lung volume reduction surgery. Transplantation 2013; 96:421–425.
  8. Sahi H, Zein NN, Mehta AC, Blazey HC, Meyer KH, Budev M. Outcomes after lung transplantation in patients with chronic hepatitis C virus infection. J Heart Lung Transplant 2007; 26:466–471.
  9. Kim EY, Ko HH, Yoshida EM. A concise review of hepatitis C in heart and lung transplantation. Can J Gastroenterol 2011; 25:445–448.
  10. Kern RM, Seethamraju H, Blanc PD, et al. The feasibility of lung transplantation in HIV-seropositive patients. Ann Am Thorac Soc 2014; 11:882–889.
  11. De Soyza A, Corris A, McDowell A, Archer L, et al. Burkholderia cepacia complex genomovars and pulmonary transplant outcomes in patients with cystic fibrosis. Lancet 2001; 358:1780–1781.
  12. De Soyza A, Meachery G, Hester HL, et al. Lung transplant for patients with cystic fibrosis and Burkholderia cepacia complex infection: a single center experience. J Heart Lung Transplant 2010; 29:1395–1404.
  13. Yusen RD, Edwards LB, Kucheryavaya AY, et al. The registry of the International Society for Heart and Lung Transplantation: thirty-second official adult and heart-lung transplantation report—2015; focus theme: early graft failure. J Heart Lung Transplant 2015; 34:1264–1277.
  14. Celli BR, Cote CG, Marin JM, et al. The body–mass index, airflow obstruction, dyspna and exercise capacity index in chronic obstructive pulmonary disease. N Engl J Med 2004; 350:1005–1012.
  15. Marchand E. The BODE index as a tool to predict survival in COPD lung transplant candidates. Eur Respir J 2010; 36:1494–1495.
  16. Lahzami S, Bridevaux PO, Soccal PM, et al. Survival impact of lung transplant for COPD. Eur Respir J 2010; 36:74–80.
  17. Rosenbluth DB, Wilson K, Ferkol T, Schuster DP. Lung function decline in cystic fibrosis patients and timing for lung transplantation referral. Chest 2004; 126:412–419.
  18. Mayer-Hamblett N, Rosenfield M, Emerson J, Goss CH, Aitken ML. Developing cystic fibrosis lung transplant referral criteria using predictors of 2-year mortality. Am J Respir Crit Care Med 2002; 166:1550–1556.
  19. Liou TG, Adler FR, Cahill BC, et al. Survival effect of lung transplantation among patients with cystic fibrosis. JAMA 2001; 286:2683–2689.
  20. Raghu G, Collard HR, Egan JJ, et al; ATS/ERS/JRS/ALAT Committee on Idiopathic Pulmonary Fibrosis. An official ATS/ERS/JRS/ALAT statement: idiopathic pulmonary fibrosis: evidenced-based guidelines for diagnosis and management. Am J Respir Crit Care Med 2011; 183:788–824.
  21. Collard HR, King TE Jr, Bartelson BB, Vourlekis JS, Schwarz MI, Brown KK. Changes in clinical and physiologic variables predict survival in idiopathic pulmonary fibrosis. Am J Respir Crit Care Med 2003; 168:538–542.
  22. Edelman, JD. Navigating the road to transplantation for pulmonary arterial hypertension. Advances in Pulmonary Hypertension 2016; 15:14–18.
  23. Strueber M. Bridges to lung transplant. Curr Opin Organ Transplant 2011; 16:458–461.
Article PDF
Author and Disclosure Information

Kenneth R. McCurry, MD
Department of Thoracic and Cardiovascular Surgery, Heart and Vascular Institute, Cleveland Clinic

Marie M. Budev, DO, MPH
Department of Pulmonary Medicine, Respiratory Institute, Cleveland Clinic

Correspondence: Marie M. Budev, DO, MPH, Department of Pulmonary Medicine, Respiratory Institute, A90, Cleveland Clinic, Cleveland, OH 44195; [email protected]

Both authors reported no financial interests or relationships that pose a potential conflict of interest with this article.

Publications
Page Number
54-58
Legacy Keywords
lung transplant, pulmonary transplant, end-stage lung disease, waiting list, chronic obstructive pulmonary disease, COPD, cystic fibrosis, CF, restrictive lung disease, pulmonary fibrosis, pulmonary arterial hypertension, Marie Budev, Kenneth McCurry
Author and Disclosure Information

Kenneth R. McCurry, MD
Department of Thoracic and Cardiovascular Surgery, Heart and Vascular Institute, Cleveland Clinic

Marie M. Budev, DO, MPH
Department of Pulmonary Medicine, Respiratory Institute, Cleveland Clinic

Correspondence: Marie M. Budev, DO, MPH, Department of Pulmonary Medicine, Respiratory Institute, A90, Cleveland Clinic, Cleveland, OH 44195; [email protected]

Both authors reported no financial interests or relationships that pose a potential conflict of interest with this article.

Author and Disclosure Information

Kenneth R. McCurry, MD
Department of Thoracic and Cardiovascular Surgery, Heart and Vascular Institute, Cleveland Clinic

Marie M. Budev, DO, MPH
Department of Pulmonary Medicine, Respiratory Institute, Cleveland Clinic

Correspondence: Marie M. Budev, DO, MPH, Department of Pulmonary Medicine, Respiratory Institute, A90, Cleveland Clinic, Cleveland, OH 44195; [email protected]

Both authors reported no financial interests or relationships that pose a potential conflict of interest with this article.

Article PDF
Article PDF
Related Articles

Lung transplant is the therapy of choice for a growing number of patients with end-stage lung diseases. Patients receiving a lung transplant are faced with many challenges including drug toxicities, infections, and the risk of rejection.1 Despite these challenges, lung transplant may significantly prolong survival and improve quality of life for many patients.

CANDIDATES FOR LUNG TRANSPLANT

Identifying patients who are appropriate candidates for lung transplant is important to achieving favorable transplant outcomes and to maximizing life expectancy for each patient. The most recent edition of International Society for Heart and Lung Transplant (ISHLT) Guidelines for the Selection of Lung Transplant Candidates is an excellent guide to help physicians identify when to refer potential patients and to how to identify patients who are the most likely to benefit from lung transplant.2

Adults with end-stage lung disease are generally candidates for lung transplant if they meet the following criteria:

  • A greater than 50% risk of death from lung disease within 2 years if a lung transplant is not performed
  • A greater than 80% likelihood of surviving at least 90 days after the lung transplant procedure
  • A greater than 80% likelihood of a 5-year survival posttransplant if graft function is preserved.2

These can only be estimated by transplant programs and not by the referring team in most cases.

Once a patient is identified as a candidate for lung transplant, early referral of patients to a lung transplant program has several advantages and is essential for positive outcomes. Early patient referral allows for timely completion of the formal evaluation of candidacy, patient and family education, as well as the opportunity for the patient and family to raise funds or use other resources to overcome financial hurdles. Listing a patient on the transplant waitlist implies that the patient has a limited life expectancy without a lung transplant and that the risk-benefit ratio favors lung transplant since all other medical options have been exhausted.1

New candidates age 12 years and older on the lung transplant waiting list by year added.
Adapted from reference 4.
Figure 1. New candidates age 12 years and older on the lung transplant waiting list by year added.
Each year, the number of new candidates added to the lung transplant waitlist grows (Figure 1). Since 2005, the allocation of organs for transplant has shifted from a time-based system to a risk of mortality-based system. The Lung Allocation System prioritizes candidates with the highest risk of mortality. Thus, the number of sicker and older patients on the wait list has increased since the implementation of the Lung Allocation System.3 Because lung transplant is associated with significant perioperative morbidity and mortality, and older and sicker patients are being considered for listing, the contraindications and comorbidities should be vetted thoroughly prior to listing.

NONCANDIDATES FOR LUNG TRANSPLANT

There are very few absolute contraindications to lung transplant. Generally, most transplant centers in the United States agree that contraindications to lung transplant include conditions associated with increased risk of mortality, including:

  • A recent history of a major malignancy. Patients with a 2-year, disease-free interval combined with a low predicted risk of recurrence may be considered in certain cases of localized, non-melanoma skin cancer. A 5-year, disease-free survival is strongly suggested in patients with a history of breast, bladder, or kidney cancer as well as in cases of sarcoma, melanoma, lymphoma and certain hematologic disorders.
  • The presence of significant dysfunction of another major organ systems including the heart, liver, kidney, or brain unless a combined organ transplant can be considered and performed.
  • Significant coronary heart disease not amenable to revascularization or intervention prior to or at the time of lung transplant.
  • The presence of an acute medical condition including but not limited to sepsis and acute liver failure.
  • Active Mycobacterium tuberculosis and other highly virulent or highly resistant microbes that are poorly controlled pretransplant.
  • Severe obesity with a body mass index greater than 35.
  • A history of nonadherence to medical therapy, psychiatric or psychological conditions that might lead to nonadherence, poor or limited social support system, and limited functional status not amenable to rehabilitation.
  • Current substance abuse or dependence, including illicit substances, alcohol, and tobacco (nicotine-containing substances). Most centers require at least 6 months’ abstinence from illicit substances prior to being added to the lung transplant waitlist.2

CANDIDATE COMORBIDITIES

Age

Many transplant centers in the US define the age cutoff for lung transplant at 65; however, some centers may consider candidates older than 65. Advanced age by itself should not be considered a contraindication to lung transplant. However, increased age is usually associated with other comorbid conditions that may increase perioperative and long-term morbidity and mortality. As mentioned previously, the number of older candidates for lung transplant has increased. In the US, 29% of the patients on the national waiting list in 2015 were over age 65.4

Past chest surgery

It is not uncommon for lung transplant candidates to have a history of chest surgery such as lung resection, pleurodesis, or coronary artery bypass grafting. The limited literature regarding the outcomes for these patients suggests they may experience higher rates of bleeding, re-exploration, and renal dysfunction.2 However, these patients should not be excluded from lung transplant and successful transplant outcomes have been achieved in this population by experienced centers.5 In candidates with a history of chronic obstructive pulmonary disease (COPD) and lung-volume reduction surgery (LVRS), early case series indicate that these patients did well after lung transplant.6 However, more recent data demonstrate that patients with prior LVRS who undergo lung transplant experience higher rates of bleeding, worse early graft dysfunction, and worse outcomes overall.7 As with lung transplant candidates with previous chest surgery, lung transplant candidates with previous LVRS are best served by experienced transplant centers.

Hepatitis and HIV

Patients with a history of infection with hepatitis B, hepatitis C, or human immunodeficiency virus (HIV) are candidates for lung transplant at centers experienced with lung transplant in patients with these infections. Most centers advocate that patients with a history of hepatitis B or C have viral infection levels that are controlled or reduced as low as possible and that there is no evidence of portal hypertension or severe cirrhosis.8,9 In the case of HIV, patients should have controlled disease with a negative or undetectable viral load and have no current acquired immunodeficiency defining illness.10 Patients colonized with particular species of Burkholderia cepacia or Mycobacterium abscessus subspecies can be considered for lung transplant only at centers with established preoperative and postoperative protocols for these infections due to the increased risk of perioperative mortality associated with these organisms.11,12

 

 

DISEASE-SPECIFIC INDICATIONS

Chronic obstructive pulmonary disease

COPD (both non- and alpha-1 antitrypsin deficiency) is the most common indication for lung transplant and accounts for almost 32% of lung transplants worldwide.13 Patients should be referred for lung transplant when medical therapies, surgical interventions (ie, LVRS) and pulmonary rehabilitation have been maximized. In COPD, the loss of lung function occurs over a long period of time but patients are often more limited by diminished quality of life as lung function slowly declines.

Patients with COPD should be referred for lung transplant if the body mass index, airflow obstruction, dyspnea, and exercise capacity (BODE) index is 5 to 6.2 The original BODE index developed by Celli et al,14 is a scoring system from 0 to 10 with a higher score indicating more severe disease and worse survival. A score of 5 to 6 indicates an estimated mortality of 60% at 4 yrs.2,14,15 Other considerations for referral for lung transplant include the presence of hypercapnia with partial pressure of carbon dioxide greater than 50 mm Hg or higher or hypoxemia with partial pressure of oxygen less than 60 mm Hg or a forced expiratory volume at 1 sec (FEV1) less than 25% predicted.

Patients with COPD should considered for listing for lung transplant if any one of the following criteria is met: BODE index of 7 or greater; FEV1 less than 15% to 20%; 3 of more severe exacerbations during the preceding year; 1 severe exacerbation with acute hypercapnic respiratory failure; or presence of moderate to severe pulmonary hypertension.2,16

Cystic fibrosis

In patients with cystic fibrosis, lung transplant should be considered in patients with an estimated 2-year survival of less than 50% and with a New York Heart Association (NYHA) Functional Classification III or IV. Referral for lung transplant is recommended for patients with a rapid decrease in FEV1 despite optimal therapy, female patients with declining weight and lung function, colonization or infection with nontuberculous mycobacterial disease, or cystic fibrosis-related diabetes. The development of pulmonary hypertension, reduction in walk distance, increasing antibiotic resistance, acute respiratory failure requiring noninvasive ventilation, worsening nutritional status, pneumothorax, and life-threatening hemoptysis despite embolization are all indications for referral for lung transplant.

Patients with cystic fibrosis with hypoxia or hypercapnia with declining lung function, needing long-term noninvasive ventilation, having more frequent exacerbations or exhibiting a decline in functional status should be listed for lung transplant.2,17–19

Restrictive lung disease

Patients with restrictive lung diseases, including interstitial pulmonary fibrosis (usual interstitial pneumonitis, nonspecific interstitial pneumonia), or interstitial lung disease, and hypersensitivity pneumonitis, should be referred for transplant evaluation at the time of diagnosis irrespective of lung function due to the unpredictable nature of these diseases.20 Some clinicians may advocate for a trial of medical therapy with antifibrotics, but this should be done in conjunction with transplant referral.

Patients should be listed for transplant if a 10% or greater decrease in FEV1 occurred in the past 6 months (of note, even a 5% decrease in FEV1 is associated with an overall poorer prognosis and warrants consideration of listing for transplant), if the diffusing capacity of the lung for carbon monoxide decreases 15% or greater during the 6-month follow-up, or if a decline of more than 50 meters is noted on the 6-minute walking test. A documented desaturation of less than 88% or a distance of less than 250 meters on the 6-minute walking test is another indication for listing. Any evidence of secondary pulmonary hypertension on right heart catheterization or on echocardiography or hospitalization for respiratory decline are also indications for listing.21 In cases of scleroderma-associated interstitial lung disease or mixed connective tissue interstitial lung disease, similar guidelines for referral and listing should be followed.2

Pulmonary arterial hypertension

Patients with pulmonary arterial hypertension should be referred for lung transplant if any 1 of the following conditions is present: rapidly progressive disease; NYHA Functional Classification III or IV symptoms during escalating therapy; use of parenteral pulmonary arterial hypertension therapy; or known or suspected pulmonary veno-occlusive disease or pulmonary capillary hemangiomatosis.2,22

Patients with pulmonary arterial hypertension should be listed for lung transplant if any of the following are present: NYHA Functional Classification III or IV symptoms despite combination therapy; right heart catheterization demonstrating a cardiac index less than 2 L/min/m2; mean right atrial pressure greater than 15 mm Hg; 6-minute walking test less than 350 meters; or development of pericardial effusion, hemoptysis, or signs of worsening right heart failure, including renal insufficiency, rising bilirubin or evidence of ascites.2,22

BRIDGE TO TRANSPLANT

Acute respiratory decompensation may occur in some candidates for lung transplant prior to listing for transplant or while on the transplant waitlist. In patients with failure of a single lung, a bridge to transplant may be necessary until a suitable organ is available. Mechanical ventilation and extracorporeal life support (ECLS) are 2 bridge strategies for lung transplant candidates. Mechanical ventilation is the most common lung transplant bridge strategy but it is less than ideal because it can lead to deconditioning and ventilator-associated infections that can negatively impact a patient’s suitability for transplant.

ECLS techniques that allow spontaneous breathing and potentially ambulation, known as awake or ambulatory ECLS, is a popular bridge therapy. Ambulatory ECLS is used as an alternative to mechanical ventilation to avoid the complications of mechanical ventilation and allow patients to avoid sedation and participate in rehabilitation.23 Irrespective of the therapy used as a bridge to transplant, patients considered for a bridge are optimally evaluated from a medical and psychosocial perspective prior to bridge therapy.

Both bridge therapies increase the risk of infection, bleeding, and neurologic events; thus, patients need to be assessed repeatedly for these risks to determine ongoing suitability for lung transplant. It is important to note that delayed referral of patients with advanced disease or patients in an acute exacerbation negatively impacts the evaluation for lung transplant, placement on the lung transplant waitlist, outcomes, and suitability for bridge transplant strategies.

CONCLUSION

To ensure good patient outcomes, the evaluation and selection of candidates for lung transplant requires communication between referring physicians and lung transplant centers. Physicians need basic knowledge of patient conditions appropriate for lung transplant and direct communication with lung transplant centers. The workup, required testing, and timing of listing for lung transplant varies among transplant centers across the country, making communication between the referring providers and transplant centers crucial to good patient care. An open, 2-way dialogue between referring providers and transplant centers facilitates listing patients for transplant in a timely manner, reduces delays, and improves outcomes.

Lung transplant is the therapy of choice for a growing number of patients with end-stage lung diseases. Patients receiving a lung transplant are faced with many challenges including drug toxicities, infections, and the risk of rejection.1 Despite these challenges, lung transplant may significantly prolong survival and improve quality of life for many patients.

CANDIDATES FOR LUNG TRANSPLANT

Identifying patients who are appropriate candidates for lung transplant is important to achieving favorable transplant outcomes and to maximizing life expectancy for each patient. The most recent edition of International Society for Heart and Lung Transplant (ISHLT) Guidelines for the Selection of Lung Transplant Candidates is an excellent guide to help physicians identify when to refer potential patients and to how to identify patients who are the most likely to benefit from lung transplant.2

Adults with end-stage lung disease are generally candidates for lung transplant if they meet the following criteria:

  • A greater than 50% risk of death from lung disease within 2 years if a lung transplant is not performed
  • A greater than 80% likelihood of surviving at least 90 days after the lung transplant procedure
  • A greater than 80% likelihood of a 5-year survival posttransplant if graft function is preserved.2

These can only be estimated by transplant programs and not by the referring team in most cases.

Once a patient is identified as a candidate for lung transplant, early referral of patients to a lung transplant program has several advantages and is essential for positive outcomes. Early patient referral allows for timely completion of the formal evaluation of candidacy, patient and family education, as well as the opportunity for the patient and family to raise funds or use other resources to overcome financial hurdles. Listing a patient on the transplant waitlist implies that the patient has a limited life expectancy without a lung transplant and that the risk-benefit ratio favors lung transplant since all other medical options have been exhausted.1

New candidates age 12 years and older on the lung transplant waiting list by year added.
Adapted from reference 4.
Figure 1. New candidates age 12 years and older on the lung transplant waiting list by year added.
Each year, the number of new candidates added to the lung transplant waitlist grows (Figure 1). Since 2005, the allocation of organs for transplant has shifted from a time-based system to a risk of mortality-based system. The Lung Allocation System prioritizes candidates with the highest risk of mortality. Thus, the number of sicker and older patients on the wait list has increased since the implementation of the Lung Allocation System.3 Because lung transplant is associated with significant perioperative morbidity and mortality, and older and sicker patients are being considered for listing, the contraindications and comorbidities should be vetted thoroughly prior to listing.

NONCANDIDATES FOR LUNG TRANSPLANT

There are very few absolute contraindications to lung transplant. Generally, most transplant centers in the United States agree that contraindications to lung transplant include conditions associated with increased risk of mortality, including:

  • A recent history of a major malignancy. Patients with a 2-year, disease-free interval combined with a low predicted risk of recurrence may be considered in certain cases of localized, non-melanoma skin cancer. A 5-year, disease-free survival is strongly suggested in patients with a history of breast, bladder, or kidney cancer as well as in cases of sarcoma, melanoma, lymphoma and certain hematologic disorders.
  • The presence of significant dysfunction of another major organ systems including the heart, liver, kidney, or brain unless a combined organ transplant can be considered and performed.
  • Significant coronary heart disease not amenable to revascularization or intervention prior to or at the time of lung transplant.
  • The presence of an acute medical condition including but not limited to sepsis and acute liver failure.
  • Active Mycobacterium tuberculosis and other highly virulent or highly resistant microbes that are poorly controlled pretransplant.
  • Severe obesity with a body mass index greater than 35.
  • A history of nonadherence to medical therapy, psychiatric or psychological conditions that might lead to nonadherence, poor or limited social support system, and limited functional status not amenable to rehabilitation.
  • Current substance abuse or dependence, including illicit substances, alcohol, and tobacco (nicotine-containing substances). Most centers require at least 6 months’ abstinence from illicit substances prior to being added to the lung transplant waitlist.2

CANDIDATE COMORBIDITIES

Age

Many transplant centers in the US define the age cutoff for lung transplant at 65; however, some centers may consider candidates older than 65. Advanced age by itself should not be considered a contraindication to lung transplant. However, increased age is usually associated with other comorbid conditions that may increase perioperative and long-term morbidity and mortality. As mentioned previously, the number of older candidates for lung transplant has increased. In the US, 29% of the patients on the national waiting list in 2015 were over age 65.4

Past chest surgery

It is not uncommon for lung transplant candidates to have a history of chest surgery such as lung resection, pleurodesis, or coronary artery bypass grafting. The limited literature regarding the outcomes for these patients suggests they may experience higher rates of bleeding, re-exploration, and renal dysfunction.2 However, these patients should not be excluded from lung transplant and successful transplant outcomes have been achieved in this population by experienced centers.5 In candidates with a history of chronic obstructive pulmonary disease (COPD) and lung-volume reduction surgery (LVRS), early case series indicate that these patients did well after lung transplant.6 However, more recent data demonstrate that patients with prior LVRS who undergo lung transplant experience higher rates of bleeding, worse early graft dysfunction, and worse outcomes overall.7 As with lung transplant candidates with previous chest surgery, lung transplant candidates with previous LVRS are best served by experienced transplant centers.

Hepatitis and HIV

Patients with a history of infection with hepatitis B, hepatitis C, or human immunodeficiency virus (HIV) are candidates for lung transplant at centers experienced with lung transplant in patients with these infections. Most centers advocate that patients with a history of hepatitis B or C have viral infection levels that are controlled or reduced as low as possible and that there is no evidence of portal hypertension or severe cirrhosis.8,9 In the case of HIV, patients should have controlled disease with a negative or undetectable viral load and have no current acquired immunodeficiency defining illness.10 Patients colonized with particular species of Burkholderia cepacia or Mycobacterium abscessus subspecies can be considered for lung transplant only at centers with established preoperative and postoperative protocols for these infections due to the increased risk of perioperative mortality associated with these organisms.11,12

 

 

DISEASE-SPECIFIC INDICATIONS

Chronic obstructive pulmonary disease

COPD (both non- and alpha-1 antitrypsin deficiency) is the most common indication for lung transplant and accounts for almost 32% of lung transplants worldwide.13 Patients should be referred for lung transplant when medical therapies, surgical interventions (ie, LVRS) and pulmonary rehabilitation have been maximized. In COPD, the loss of lung function occurs over a long period of time but patients are often more limited by diminished quality of life as lung function slowly declines.

Patients with COPD should be referred for lung transplant if the body mass index, airflow obstruction, dyspnea, and exercise capacity (BODE) index is 5 to 6.2 The original BODE index developed by Celli et al,14 is a scoring system from 0 to 10 with a higher score indicating more severe disease and worse survival. A score of 5 to 6 indicates an estimated mortality of 60% at 4 yrs.2,14,15 Other considerations for referral for lung transplant include the presence of hypercapnia with partial pressure of carbon dioxide greater than 50 mm Hg or higher or hypoxemia with partial pressure of oxygen less than 60 mm Hg or a forced expiratory volume at 1 sec (FEV1) less than 25% predicted.

Patients with COPD should considered for listing for lung transplant if any one of the following criteria is met: BODE index of 7 or greater; FEV1 less than 15% to 20%; 3 of more severe exacerbations during the preceding year; 1 severe exacerbation with acute hypercapnic respiratory failure; or presence of moderate to severe pulmonary hypertension.2,16

Cystic fibrosis

In patients with cystic fibrosis, lung transplant should be considered in patients with an estimated 2-year survival of less than 50% and with a New York Heart Association (NYHA) Functional Classification III or IV. Referral for lung transplant is recommended for patients with a rapid decrease in FEV1 despite optimal therapy, female patients with declining weight and lung function, colonization or infection with nontuberculous mycobacterial disease, or cystic fibrosis-related diabetes. The development of pulmonary hypertension, reduction in walk distance, increasing antibiotic resistance, acute respiratory failure requiring noninvasive ventilation, worsening nutritional status, pneumothorax, and life-threatening hemoptysis despite embolization are all indications for referral for lung transplant.

Patients with cystic fibrosis with hypoxia or hypercapnia with declining lung function, needing long-term noninvasive ventilation, having more frequent exacerbations or exhibiting a decline in functional status should be listed for lung transplant.2,17–19

Restrictive lung disease

Patients with restrictive lung diseases, including interstitial pulmonary fibrosis (usual interstitial pneumonitis, nonspecific interstitial pneumonia), or interstitial lung disease, and hypersensitivity pneumonitis, should be referred for transplant evaluation at the time of diagnosis irrespective of lung function due to the unpredictable nature of these diseases.20 Some clinicians may advocate for a trial of medical therapy with antifibrotics, but this should be done in conjunction with transplant referral.

Patients should be listed for transplant if a 10% or greater decrease in FEV1 occurred in the past 6 months (of note, even a 5% decrease in FEV1 is associated with an overall poorer prognosis and warrants consideration of listing for transplant), if the diffusing capacity of the lung for carbon monoxide decreases 15% or greater during the 6-month follow-up, or if a decline of more than 50 meters is noted on the 6-minute walking test. A documented desaturation of less than 88% or a distance of less than 250 meters on the 6-minute walking test is another indication for listing. Any evidence of secondary pulmonary hypertension on right heart catheterization or on echocardiography or hospitalization for respiratory decline are also indications for listing.21 In cases of scleroderma-associated interstitial lung disease or mixed connective tissue interstitial lung disease, similar guidelines for referral and listing should be followed.2

Pulmonary arterial hypertension

Patients with pulmonary arterial hypertension should be referred for lung transplant if any 1 of the following conditions is present: rapidly progressive disease; NYHA Functional Classification III or IV symptoms during escalating therapy; use of parenteral pulmonary arterial hypertension therapy; or known or suspected pulmonary veno-occlusive disease or pulmonary capillary hemangiomatosis.2,22

Patients with pulmonary arterial hypertension should be listed for lung transplant if any of the following are present: NYHA Functional Classification III or IV symptoms despite combination therapy; right heart catheterization demonstrating a cardiac index less than 2 L/min/m2; mean right atrial pressure greater than 15 mm Hg; 6-minute walking test less than 350 meters; or development of pericardial effusion, hemoptysis, or signs of worsening right heart failure, including renal insufficiency, rising bilirubin or evidence of ascites.2,22

BRIDGE TO TRANSPLANT

Acute respiratory decompensation may occur in some candidates for lung transplant prior to listing for transplant or while on the transplant waitlist. In patients with failure of a single lung, a bridge to transplant may be necessary until a suitable organ is available. Mechanical ventilation and extracorporeal life support (ECLS) are 2 bridge strategies for lung transplant candidates. Mechanical ventilation is the most common lung transplant bridge strategy but it is less than ideal because it can lead to deconditioning and ventilator-associated infections that can negatively impact a patient’s suitability for transplant.

ECLS techniques that allow spontaneous breathing and potentially ambulation, known as awake or ambulatory ECLS, is a popular bridge therapy. Ambulatory ECLS is used as an alternative to mechanical ventilation to avoid the complications of mechanical ventilation and allow patients to avoid sedation and participate in rehabilitation.23 Irrespective of the therapy used as a bridge to transplant, patients considered for a bridge are optimally evaluated from a medical and psychosocial perspective prior to bridge therapy.

Both bridge therapies increase the risk of infection, bleeding, and neurologic events; thus, patients need to be assessed repeatedly for these risks to determine ongoing suitability for lung transplant. It is important to note that delayed referral of patients with advanced disease or patients in an acute exacerbation negatively impacts the evaluation for lung transplant, placement on the lung transplant waitlist, outcomes, and suitability for bridge transplant strategies.

CONCLUSION

To ensure good patient outcomes, the evaluation and selection of candidates for lung transplant requires communication between referring physicians and lung transplant centers. Physicians need basic knowledge of patient conditions appropriate for lung transplant and direct communication with lung transplant centers. The workup, required testing, and timing of listing for lung transplant varies among transplant centers across the country, making communication between the referring providers and transplant centers crucial to good patient care. An open, 2-way dialogue between referring providers and transplant centers facilitates listing patients for transplant in a timely manner, reduces delays, and improves outcomes.

References
  1. Kreider M, Hadjiliadis D, Kotloff R. Candidate selection, timing of listing, and choice of procedure for lung transplantation. Clin Chest Med 2011; 32:199–211.
  2. Weill D, Benden C, Corris P, et al. A consensus document for the selection of lung transplant candidates: 2014—An update from the Pulmonary Transplant Council of the International Society of Heart and Lung Transplantation. J Heart Lung Transplant 2015; 34:1–15.
  3. Tsuang WM. Contemporary issues in lung transplant allocation practices. Curr Transplant Rep 2017; 4:238–242.
  4. Valapour M, Skeans MA, Smith JM, et al. OPTN/SRTR 2015 annual data report: lung. Am J Transplant 2017; 17(suppl 1):357–424.
  5. Omara M, Okamoto T, Arafat A, Thuita L, Blackstone EH, McCurry KR. Lung transplantation in patients who have undergone prior cardiothoracic procedures. J Heart Lung Transplant 2016; 35:1462–1470.
  6. Senbaklavaci O, Wisser W, Ozpeker C, et al. Successful lung volume reduction surgery brings patients into better condition for later lung transplantation. Eur J Cardiothorac Surg 2002; 22:363–367.
  7. Shigemura N, Gilbert S, Bhama JK et al. Lung transplantation after lung volume reduction surgery. Transplantation 2013; 96:421–425.
  8. Sahi H, Zein NN, Mehta AC, Blazey HC, Meyer KH, Budev M. Outcomes after lung transplantation in patients with chronic hepatitis C virus infection. J Heart Lung Transplant 2007; 26:466–471.
  9. Kim EY, Ko HH, Yoshida EM. A concise review of hepatitis C in heart and lung transplantation. Can J Gastroenterol 2011; 25:445–448.
  10. Kern RM, Seethamraju H, Blanc PD, et al. The feasibility of lung transplantation in HIV-seropositive patients. Ann Am Thorac Soc 2014; 11:882–889.
  11. De Soyza A, Corris A, McDowell A, Archer L, et al. Burkholderia cepacia complex genomovars and pulmonary transplant outcomes in patients with cystic fibrosis. Lancet 2001; 358:1780–1781.
  12. De Soyza A, Meachery G, Hester HL, et al. Lung transplant for patients with cystic fibrosis and Burkholderia cepacia complex infection: a single center experience. J Heart Lung Transplant 2010; 29:1395–1404.
  13. Yusen RD, Edwards LB, Kucheryavaya AY, et al. The registry of the International Society for Heart and Lung Transplantation: thirty-second official adult and heart-lung transplantation report—2015; focus theme: early graft failure. J Heart Lung Transplant 2015; 34:1264–1277.
  14. Celli BR, Cote CG, Marin JM, et al. The body–mass index, airflow obstruction, dyspna and exercise capacity index in chronic obstructive pulmonary disease. N Engl J Med 2004; 350:1005–1012.
  15. Marchand E. The BODE index as a tool to predict survival in COPD lung transplant candidates. Eur Respir J 2010; 36:1494–1495.
  16. Lahzami S, Bridevaux PO, Soccal PM, et al. Survival impact of lung transplant for COPD. Eur Respir J 2010; 36:74–80.
  17. Rosenbluth DB, Wilson K, Ferkol T, Schuster DP. Lung function decline in cystic fibrosis patients and timing for lung transplantation referral. Chest 2004; 126:412–419.
  18. Mayer-Hamblett N, Rosenfield M, Emerson J, Goss CH, Aitken ML. Developing cystic fibrosis lung transplant referral criteria using predictors of 2-year mortality. Am J Respir Crit Care Med 2002; 166:1550–1556.
  19. Liou TG, Adler FR, Cahill BC, et al. Survival effect of lung transplantation among patients with cystic fibrosis. JAMA 2001; 286:2683–2689.
  20. Raghu G, Collard HR, Egan JJ, et al; ATS/ERS/JRS/ALAT Committee on Idiopathic Pulmonary Fibrosis. An official ATS/ERS/JRS/ALAT statement: idiopathic pulmonary fibrosis: evidenced-based guidelines for diagnosis and management. Am J Respir Crit Care Med 2011; 183:788–824.
  21. Collard HR, King TE Jr, Bartelson BB, Vourlekis JS, Schwarz MI, Brown KK. Changes in clinical and physiologic variables predict survival in idiopathic pulmonary fibrosis. Am J Respir Crit Care Med 2003; 168:538–542.
  22. Edelman, JD. Navigating the road to transplantation for pulmonary arterial hypertension. Advances in Pulmonary Hypertension 2016; 15:14–18.
  23. Strueber M. Bridges to lung transplant. Curr Opin Organ Transplant 2011; 16:458–461.
References
  1. Kreider M, Hadjiliadis D, Kotloff R. Candidate selection, timing of listing, and choice of procedure for lung transplantation. Clin Chest Med 2011; 32:199–211.
  2. Weill D, Benden C, Corris P, et al. A consensus document for the selection of lung transplant candidates: 2014—An update from the Pulmonary Transplant Council of the International Society of Heart and Lung Transplantation. J Heart Lung Transplant 2015; 34:1–15.
  3. Tsuang WM. Contemporary issues in lung transplant allocation practices. Curr Transplant Rep 2017; 4:238–242.
  4. Valapour M, Skeans MA, Smith JM, et al. OPTN/SRTR 2015 annual data report: lung. Am J Transplant 2017; 17(suppl 1):357–424.
  5. Omara M, Okamoto T, Arafat A, Thuita L, Blackstone EH, McCurry KR. Lung transplantation in patients who have undergone prior cardiothoracic procedures. J Heart Lung Transplant 2016; 35:1462–1470.
  6. Senbaklavaci O, Wisser W, Ozpeker C, et al. Successful lung volume reduction surgery brings patients into better condition for later lung transplantation. Eur J Cardiothorac Surg 2002; 22:363–367.
  7. Shigemura N, Gilbert S, Bhama JK et al. Lung transplantation after lung volume reduction surgery. Transplantation 2013; 96:421–425.
  8. Sahi H, Zein NN, Mehta AC, Blazey HC, Meyer KH, Budev M. Outcomes after lung transplantation in patients with chronic hepatitis C virus infection. J Heart Lung Transplant 2007; 26:466–471.
  9. Kim EY, Ko HH, Yoshida EM. A concise review of hepatitis C in heart and lung transplantation. Can J Gastroenterol 2011; 25:445–448.
  10. Kern RM, Seethamraju H, Blanc PD, et al. The feasibility of lung transplantation in HIV-seropositive patients. Ann Am Thorac Soc 2014; 11:882–889.
  11. De Soyza A, Corris A, McDowell A, Archer L, et al. Burkholderia cepacia complex genomovars and pulmonary transplant outcomes in patients with cystic fibrosis. Lancet 2001; 358:1780–1781.
  12. De Soyza A, Meachery G, Hester HL, et al. Lung transplant for patients with cystic fibrosis and Burkholderia cepacia complex infection: a single center experience. J Heart Lung Transplant 2010; 29:1395–1404.
  13. Yusen RD, Edwards LB, Kucheryavaya AY, et al. The registry of the International Society for Heart and Lung Transplantation: thirty-second official adult and heart-lung transplantation report—2015; focus theme: early graft failure. J Heart Lung Transplant 2015; 34:1264–1277.
  14. Celli BR, Cote CG, Marin JM, et al. The body–mass index, airflow obstruction, dyspna and exercise capacity index in chronic obstructive pulmonary disease. N Engl J Med 2004; 350:1005–1012.
  15. Marchand E. The BODE index as a tool to predict survival in COPD lung transplant candidates. Eur Respir J 2010; 36:1494–1495.
  16. Lahzami S, Bridevaux PO, Soccal PM, et al. Survival impact of lung transplant for COPD. Eur Respir J 2010; 36:74–80.
  17. Rosenbluth DB, Wilson K, Ferkol T, Schuster DP. Lung function decline in cystic fibrosis patients and timing for lung transplantation referral. Chest 2004; 126:412–419.
  18. Mayer-Hamblett N, Rosenfield M, Emerson J, Goss CH, Aitken ML. Developing cystic fibrosis lung transplant referral criteria using predictors of 2-year mortality. Am J Respir Crit Care Med 2002; 166:1550–1556.
  19. Liou TG, Adler FR, Cahill BC, et al. Survival effect of lung transplantation among patients with cystic fibrosis. JAMA 2001; 286:2683–2689.
  20. Raghu G, Collard HR, Egan JJ, et al; ATS/ERS/JRS/ALAT Committee on Idiopathic Pulmonary Fibrosis. An official ATS/ERS/JRS/ALAT statement: idiopathic pulmonary fibrosis: evidenced-based guidelines for diagnosis and management. Am J Respir Crit Care Med 2011; 183:788–824.
  21. Collard HR, King TE Jr, Bartelson BB, Vourlekis JS, Schwarz MI, Brown KK. Changes in clinical and physiologic variables predict survival in idiopathic pulmonary fibrosis. Am J Respir Crit Care Med 2003; 168:538–542.
  22. Edelman, JD. Navigating the road to transplantation for pulmonary arterial hypertension. Advances in Pulmonary Hypertension 2016; 15:14–18.
  23. Strueber M. Bridges to lung transplant. Curr Opin Organ Transplant 2011; 16:458–461.
Page Number
54-58
Page Number
54-58
Publications
Publications
Article Type
Display Headline
Lung transplant: Candidates for referral and the waiting list
Display Headline
Lung transplant: Candidates for referral and the waiting list
Legacy Keywords
lung transplant, pulmonary transplant, end-stage lung disease, waiting list, chronic obstructive pulmonary disease, COPD, cystic fibrosis, CF, restrictive lung disease, pulmonary fibrosis, pulmonary arterial hypertension, Marie Budev, Kenneth McCurry
Legacy Keywords
lung transplant, pulmonary transplant, end-stage lung disease, waiting list, chronic obstructive pulmonary disease, COPD, cystic fibrosis, CF, restrictive lung disease, pulmonary fibrosis, pulmonary arterial hypertension, Marie Budev, Kenneth McCurry
Citation Override
Cleveland Clinic Journal of Medicine 2017 December;84(suppl 3):54-58
Inside the Article

KEY POINTS

  • Lung transplant is the therapy of choice for a growing number of patients with end-stage lung disease.
  • There are very few absolute contraindications to lung transplant. Potential contraindications and comorbidities can be discussed with the transplant center and vetted prior to listing for lung transplant.
  • The workup for a lung transplant varies among transplant centers across the country, thus good communication between referring providers and transplant centers is crucial to quality care.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Article PDF Media

Familial essential thrombocythemia associated with JAK2 V617F mutation in siblings

Article Type
Changed
Fri, 01/04/2019 - 11:16

Three myeloproliferative neoplasms (MPN), polycythemia vera (PV), essential thrombocythemia (ET), and primary myelofibrosis (PMF), are associated with an abnormal somatic mutation of the JAK2 gene. Essential thrombocythemia is considered when there is a persistent increase in the peripheral blood platelet count, associated with a proliferation of atypical megakaryocytes in the bone marrow. The manifestations of PV, ET, and PMF all typically occur within the sixth or seventh decade of life. A patient may present with an abnormal blood count but be asymptomatic at the time. Over the course and progression of the disease, increases in hematocrit or platelet counts along with symptoms such as headaches, blurred vision, and plethora may occur.1 The JAK2 V617F mutation is responsible for the production of the JAK2 protein, which is continuously activated, promoting the growth and division of cells such as erythrocytes, granulocytes, and platelets. It has been reported that there is a nearly 100% incidence of the JAK2 mutation in patients with polycythemia vera, and a 50% incidence in patients with essential thrombocythemia and primary myelofibrosis.2

The discovery of the JAK2 mutation in PV, ET, and PMF was an important advancement in helping distinguish these disorders from other MPNs, including chronic myelogenous leukemia, but its presence does not explain why some individuals develop ET and others, PV or PMF.3 Although there have been familial cases proven of ET, the somatic JAK2 mutation is acquired and not inherited. In this report, we describe the unusual circumstance of JAK2 V617F mutation in a brother and a sister who were both diagnosed with essential thrombocythemia.

Case presentations and summaries

RS, a 69-year-old white man, was referred to our service in 2006 for continued care of previously diagnosed essential thrombocythemia. At the time of his initial visit to our clinic, his complete blood count was normal, the platelet count being adequately controlled by anagrelide at a daily dose of 4.0 mg. He complained of palpitations and peripheral neuropathy. A bone marrow biopsy was performed, revealing moderate hypercellularity, atypical megakaryocytosis, and a negative BCR-ABL mutation but a positive JAK2 V617F mutation. The patient is now treated with hydroxyurea 1,000 mg daily in divided doses, which better controls his counts and does not have the side effects of anagrelide.

SW, a 73-year-old woman, and brother of RS (they share the same biological mother and father), was noted to have a mild thrombocytosis in 2008. In 2013, her platelet count rose to 865,000 cells/uL (normal, 150,000-450,000 cells/uL, age and sex adjusted) and she was referred to our clinic. A bone marrow biopsy was performed, revealing borderline hypercellularity with atypical megakaryocytosis and the presence of a JAK2 V617F mutation. As with her brother, the BCR-ABL mutation was not present. She has also responded to treatment with hydroxyurea, but at a reduced dosage of 500 mg daily.

A third sibling, AS, again of the same biological mother and father, had died of multiple veno-occlusive cerebral vascular events long before the diagnoses on his younger siblings had been made. The suggestion of any underling hematologic pathology would be interesting, but speculative. Nothing is known about the parents’ medical history. None of the three siblings had children.

Discussion

Much research has been done to understand the pathogenesis of and find a cure for myeloproliferative disorders, but despite some progress, a cure remains elusive. However, there have been some advances that have contributed to partial cures for MPNs. One of the major breakthroughs in MPN research, about 50 years ago, was related to the “sporadic vs familial debate” around the Philadelphia chromosome.4 It led to the discovery of the reciprocal translocation between chromosomes 9 and 22, known as the BCR-ABL mutation, which is found in many CML patients. This discovery allowed researchers to focus their attention on other tyrosine kinase domains, such as the JAK2 V617F mutation, which is presented in the three other MPNs; PV, ET, and PMF. Both the JAK2 V617F and BCR-ABL mutations are active in signaling transcription, more commonly growth of cells.4

Since the discovery of the JAK2 V617F mutation in early 2005, it has become a leading diagnostic criteria for myeloproliferative diseases. The presence of the JAK2 V617F mutation and the measurement of its allele burden can be assessed by examination of either peripheral blood or bone marrow samples.5

The JAK2 V617F mutation is a result of a single change in the DNA nucleotide base pair that causes a substitution of a valine amino acid for a phenylalanine amino acid at the 617 position on exon 14 within the JAK2 kinase regulatory domain. This point mutation disrupts the regular control of the JAK2 by removing its ability to turn off, leading to uncontrolled blood cell growth.6 When the JAK2 V617F mutation cannot be demonstrated in a patient with the hallmarks of an MPN, the detection of other JAK2 and MPL proto-oncogene, thrombopoietin receptor mutations may be used as a diagnostic procedure for other MPNs.7

Other mutations incorporated in JAK2 domain can be detected in the coding portions of the DNA known as exons. One such mutation is the JAK2 exon 12, which is involved in JAK2 V617F-negative PV patients. This mutation is not detected in patients with ET or PMF and is 2%-5% present in patients with PV. There are other somatic mutations in the thrombopoietin receptors that work in accordance with thrombopoietin: MPL W515L and MPL W515K, which are found at chromosome 1p34, are identified in about 5% of PMF and 1% of ET patients, but are not present in PV patients.8.9

Pikman and colleagues reported in 2009 that the JAK2 V617F mutation is not acquired randomly.9 Their findings showed that, only in white populations, does the JAK2 V617F mutation arise preferentially on a specific constitutional JAK2 46/1 haplotype. According to the authors, the preconceived notion a of randomly acquired JAK2 V617F mutation does not account for familial MPN’s. Familial MPNs are thought to be produced by sporadic and extremely penetrant substitutions in genes that still are not identified and the 46/1 haplotype does not explain for the phenotypic diversity correlated with the JAK2 V617F gene. The 46/1 haplotype, however, correlates more frequently with different MPN subtypes. There are two hypotheses that try to explain how an acquired mutation as prevailing as the JAK2 V617F mutation can be associated with certain inherited backgrounds. The first hypothesis asserts the V617F accumulates at a faster rate than other genes because of the fundamentally unstable genetics of the 46/1 haplotype. The second theory is that all the mutated genes, including the V617F, arise at equal rates, but 46/1 may grant a selective advantage to the V617F-positive clone or interacts in some way to increase the likelihood of abnormal blood counts. A study that examined both these hypotheses concluded that the 46/1 haplotype was present more frequently in patients with myeloproliferative disorders than in their control groups and even more so in cases that were proven to be V617F-positive.10

There are very few cases that have reported familial MPN’s, especially as the pedigrees of the familial MPN’s illustrate that inheritance patterns are notably heterogeneous, indicating that there may be a range of different germline mutations driving the susceptibility. With recent data, the JAK2 V617F mutation in tandem with MPL W515L/K and inactivating TET2 mutations still continue to be the most frequently acquired mutations involved in both familial and sporadic MPN. As far as we know, there have been no cases to prove that JAK2 V617F and MPL W515L/K mutations are inherited through the germline, but there are other alleles that may pass through the germline that can be associated with hereditary thrombocytosis.11 Further cytogenetic studies will clarify the pathogenesis of these disorders and possibly lead to effective targeted therapies.

References

1. Murphy S, Peterson P, Iland H, Laszio J. Experience of the Polycythemia Vera Study Group with essential thrombocythemia: a final report on diagnostic criteria, survival, and leukemic transition by treatment. Semin Hematol. 1997;34:29-39.

2. Zhan H, Spivak JL. The diagnosis and management of polycythemia vera, essential thrombocythemia, and primary myelofibrosis in the JAK2 V617F era. Clin Adv Hematol Oncol. 2009;7:334-342.

3. Higgs JR, Sadek I, Neumann PE, et al. Familial essential thrombocythemia with spontaneous megakaryocyte colony formation and acquired JAK2 mutations. Leukemia. 2008;22:1551-1556.

4. Senyak Z. Eileen Wiggins – out of the blue. http://www.mpnresearchfoundation.org/White-Paper-3A-Nature-2C-Nurture-2C-or-Both-3F. Published October 2010. Accessed May 23, 2017.

5. Cankovic M, Whiteley L, Hawley RC, Zarbo RJ, Chitale D. Clinical performance of JAK2 V617F mutation detection assays in a molecular diagnostics laboratory: evaluation of screening and quantitation methods. Am J Clin Pathol. 2009;132:713-721.

6. Kralovics R, Teo SS, Li S, et al. Acquisition of the V617F mutation of JAK2 is a late genetic event in a subset of patients with myeloproliferative disorders. Blood. 2006;108:1377-1380.

7. James C. The JAK2V617F mutation in polycythemia vera and other myeloproliferative disorders: one mutation for three diseases? Hematology Am Soc Hematol Educ Program. 2008:69-75.

8. Pancrazzi A, Guglielmelli P, Ponziani V, et al. A sensitive detection method for MPLW515L or MPLW515K mutation in chronic myeloproliferative disorders with locked nucleic acid-modified probes and real-time polymerase chain reaction. J Mol Diagn. 2008;10:435-441.

9. Pikman Y, Lee BH, Mercher T, et al. MPLW515L is a novel somatic activating mutation in myelofibrosis with myeloid metaplasia. PLoS Med. 2006;3:1140-1151.

10. Jones AV, Campbell PJ, Beer PA, et al. The JAK2 46/1 haplotype predisposes to MPL-mutated myeloproliferative neoplasms. Blood. 2010;115:4517-4523.

11. Jones AV, Cross NCP. Inherited predisposition to myeloproliferative neoplasms. Ther Adv Hematol. 2013;4:237-253.

Article PDF
Author and Disclosure Information

Lexi Leigh Sirota, BA, and Hal Gerstein, MD

Cancer Institute of Long Island, Great Neck, New York

Issue
The Journal of Community and Supportive Oncology - 15(5)
Publications
Topics
Sections
Author and Disclosure Information

Lexi Leigh Sirota, BA, and Hal Gerstein, MD

Cancer Institute of Long Island, Great Neck, New York

Author and Disclosure Information

Lexi Leigh Sirota, BA, and Hal Gerstein, MD

Cancer Institute of Long Island, Great Neck, New York

Article PDF
Article PDF

Three myeloproliferative neoplasms (MPN), polycythemia vera (PV), essential thrombocythemia (ET), and primary myelofibrosis (PMF), are associated with an abnormal somatic mutation of the JAK2 gene. Essential thrombocythemia is considered when there is a persistent increase in the peripheral blood platelet count, associated with a proliferation of atypical megakaryocytes in the bone marrow. The manifestations of PV, ET, and PMF all typically occur within the sixth or seventh decade of life. A patient may present with an abnormal blood count but be asymptomatic at the time. Over the course and progression of the disease, increases in hematocrit or platelet counts along with symptoms such as headaches, blurred vision, and plethora may occur.1 The JAK2 V617F mutation is responsible for the production of the JAK2 protein, which is continuously activated, promoting the growth and division of cells such as erythrocytes, granulocytes, and platelets. It has been reported that there is a nearly 100% incidence of the JAK2 mutation in patients with polycythemia vera, and a 50% incidence in patients with essential thrombocythemia and primary myelofibrosis.2

The discovery of the JAK2 mutation in PV, ET, and PMF was an important advancement in helping distinguish these disorders from other MPNs, including chronic myelogenous leukemia, but its presence does not explain why some individuals develop ET and others, PV or PMF.3 Although there have been familial cases proven of ET, the somatic JAK2 mutation is acquired and not inherited. In this report, we describe the unusual circumstance of JAK2 V617F mutation in a brother and a sister who were both diagnosed with essential thrombocythemia.

Case presentations and summaries

RS, a 69-year-old white man, was referred to our service in 2006 for continued care of previously diagnosed essential thrombocythemia. At the time of his initial visit to our clinic, his complete blood count was normal, the platelet count being adequately controlled by anagrelide at a daily dose of 4.0 mg. He complained of palpitations and peripheral neuropathy. A bone marrow biopsy was performed, revealing moderate hypercellularity, atypical megakaryocytosis, and a negative BCR-ABL mutation but a positive JAK2 V617F mutation. The patient is now treated with hydroxyurea 1,000 mg daily in divided doses, which better controls his counts and does not have the side effects of anagrelide.

SW, a 73-year-old woman, and brother of RS (they share the same biological mother and father), was noted to have a mild thrombocytosis in 2008. In 2013, her platelet count rose to 865,000 cells/uL (normal, 150,000-450,000 cells/uL, age and sex adjusted) and she was referred to our clinic. A bone marrow biopsy was performed, revealing borderline hypercellularity with atypical megakaryocytosis and the presence of a JAK2 V617F mutation. As with her brother, the BCR-ABL mutation was not present. She has also responded to treatment with hydroxyurea, but at a reduced dosage of 500 mg daily.

A third sibling, AS, again of the same biological mother and father, had died of multiple veno-occlusive cerebral vascular events long before the diagnoses on his younger siblings had been made. The suggestion of any underling hematologic pathology would be interesting, but speculative. Nothing is known about the parents’ medical history. None of the three siblings had children.

Discussion

Much research has been done to understand the pathogenesis of and find a cure for myeloproliferative disorders, but despite some progress, a cure remains elusive. However, there have been some advances that have contributed to partial cures for MPNs. One of the major breakthroughs in MPN research, about 50 years ago, was related to the “sporadic vs familial debate” around the Philadelphia chromosome.4 It led to the discovery of the reciprocal translocation between chromosomes 9 and 22, known as the BCR-ABL mutation, which is found in many CML patients. This discovery allowed researchers to focus their attention on other tyrosine kinase domains, such as the JAK2 V617F mutation, which is presented in the three other MPNs; PV, ET, and PMF. Both the JAK2 V617F and BCR-ABL mutations are active in signaling transcription, more commonly growth of cells.4

Since the discovery of the JAK2 V617F mutation in early 2005, it has become a leading diagnostic criteria for myeloproliferative diseases. The presence of the JAK2 V617F mutation and the measurement of its allele burden can be assessed by examination of either peripheral blood or bone marrow samples.5

The JAK2 V617F mutation is a result of a single change in the DNA nucleotide base pair that causes a substitution of a valine amino acid for a phenylalanine amino acid at the 617 position on exon 14 within the JAK2 kinase regulatory domain. This point mutation disrupts the regular control of the JAK2 by removing its ability to turn off, leading to uncontrolled blood cell growth.6 When the JAK2 V617F mutation cannot be demonstrated in a patient with the hallmarks of an MPN, the detection of other JAK2 and MPL proto-oncogene, thrombopoietin receptor mutations may be used as a diagnostic procedure for other MPNs.7

Other mutations incorporated in JAK2 domain can be detected in the coding portions of the DNA known as exons. One such mutation is the JAK2 exon 12, which is involved in JAK2 V617F-negative PV patients. This mutation is not detected in patients with ET or PMF and is 2%-5% present in patients with PV. There are other somatic mutations in the thrombopoietin receptors that work in accordance with thrombopoietin: MPL W515L and MPL W515K, which are found at chromosome 1p34, are identified in about 5% of PMF and 1% of ET patients, but are not present in PV patients.8.9

Pikman and colleagues reported in 2009 that the JAK2 V617F mutation is not acquired randomly.9 Their findings showed that, only in white populations, does the JAK2 V617F mutation arise preferentially on a specific constitutional JAK2 46/1 haplotype. According to the authors, the preconceived notion a of randomly acquired JAK2 V617F mutation does not account for familial MPN’s. Familial MPNs are thought to be produced by sporadic and extremely penetrant substitutions in genes that still are not identified and the 46/1 haplotype does not explain for the phenotypic diversity correlated with the JAK2 V617F gene. The 46/1 haplotype, however, correlates more frequently with different MPN subtypes. There are two hypotheses that try to explain how an acquired mutation as prevailing as the JAK2 V617F mutation can be associated with certain inherited backgrounds. The first hypothesis asserts the V617F accumulates at a faster rate than other genes because of the fundamentally unstable genetics of the 46/1 haplotype. The second theory is that all the mutated genes, including the V617F, arise at equal rates, but 46/1 may grant a selective advantage to the V617F-positive clone or interacts in some way to increase the likelihood of abnormal blood counts. A study that examined both these hypotheses concluded that the 46/1 haplotype was present more frequently in patients with myeloproliferative disorders than in their control groups and even more so in cases that were proven to be V617F-positive.10

There are very few cases that have reported familial MPN’s, especially as the pedigrees of the familial MPN’s illustrate that inheritance patterns are notably heterogeneous, indicating that there may be a range of different germline mutations driving the susceptibility. With recent data, the JAK2 V617F mutation in tandem with MPL W515L/K and inactivating TET2 mutations still continue to be the most frequently acquired mutations involved in both familial and sporadic MPN. As far as we know, there have been no cases to prove that JAK2 V617F and MPL W515L/K mutations are inherited through the germline, but there are other alleles that may pass through the germline that can be associated with hereditary thrombocytosis.11 Further cytogenetic studies will clarify the pathogenesis of these disorders and possibly lead to effective targeted therapies.

Three myeloproliferative neoplasms (MPN), polycythemia vera (PV), essential thrombocythemia (ET), and primary myelofibrosis (PMF), are associated with an abnormal somatic mutation of the JAK2 gene. Essential thrombocythemia is considered when there is a persistent increase in the peripheral blood platelet count, associated with a proliferation of atypical megakaryocytes in the bone marrow. The manifestations of PV, ET, and PMF all typically occur within the sixth or seventh decade of life. A patient may present with an abnormal blood count but be asymptomatic at the time. Over the course and progression of the disease, increases in hematocrit or platelet counts along with symptoms such as headaches, blurred vision, and plethora may occur.1 The JAK2 V617F mutation is responsible for the production of the JAK2 protein, which is continuously activated, promoting the growth and division of cells such as erythrocytes, granulocytes, and platelets. It has been reported that there is a nearly 100% incidence of the JAK2 mutation in patients with polycythemia vera, and a 50% incidence in patients with essential thrombocythemia and primary myelofibrosis.2

The discovery of the JAK2 mutation in PV, ET, and PMF was an important advancement in helping distinguish these disorders from other MPNs, including chronic myelogenous leukemia, but its presence does not explain why some individuals develop ET and others, PV or PMF.3 Although there have been familial cases proven of ET, the somatic JAK2 mutation is acquired and not inherited. In this report, we describe the unusual circumstance of JAK2 V617F mutation in a brother and a sister who were both diagnosed with essential thrombocythemia.

Case presentations and summaries

RS, a 69-year-old white man, was referred to our service in 2006 for continued care of previously diagnosed essential thrombocythemia. At the time of his initial visit to our clinic, his complete blood count was normal, the platelet count being adequately controlled by anagrelide at a daily dose of 4.0 mg. He complained of palpitations and peripheral neuropathy. A bone marrow biopsy was performed, revealing moderate hypercellularity, atypical megakaryocytosis, and a negative BCR-ABL mutation but a positive JAK2 V617F mutation. The patient is now treated with hydroxyurea 1,000 mg daily in divided doses, which better controls his counts and does not have the side effects of anagrelide.

SW, a 73-year-old woman, and brother of RS (they share the same biological mother and father), was noted to have a mild thrombocytosis in 2008. In 2013, her platelet count rose to 865,000 cells/uL (normal, 150,000-450,000 cells/uL, age and sex adjusted) and she was referred to our clinic. A bone marrow biopsy was performed, revealing borderline hypercellularity with atypical megakaryocytosis and the presence of a JAK2 V617F mutation. As with her brother, the BCR-ABL mutation was not present. She has also responded to treatment with hydroxyurea, but at a reduced dosage of 500 mg daily.

A third sibling, AS, again of the same biological mother and father, had died of multiple veno-occlusive cerebral vascular events long before the diagnoses on his younger siblings had been made. The suggestion of any underling hematologic pathology would be interesting, but speculative. Nothing is known about the parents’ medical history. None of the three siblings had children.

Discussion

Much research has been done to understand the pathogenesis of and find a cure for myeloproliferative disorders, but despite some progress, a cure remains elusive. However, there have been some advances that have contributed to partial cures for MPNs. One of the major breakthroughs in MPN research, about 50 years ago, was related to the “sporadic vs familial debate” around the Philadelphia chromosome.4 It led to the discovery of the reciprocal translocation between chromosomes 9 and 22, known as the BCR-ABL mutation, which is found in many CML patients. This discovery allowed researchers to focus their attention on other tyrosine kinase domains, such as the JAK2 V617F mutation, which is presented in the three other MPNs; PV, ET, and PMF. Both the JAK2 V617F and BCR-ABL mutations are active in signaling transcription, more commonly growth of cells.4

Since the discovery of the JAK2 V617F mutation in early 2005, it has become a leading diagnostic criteria for myeloproliferative diseases. The presence of the JAK2 V617F mutation and the measurement of its allele burden can be assessed by examination of either peripheral blood or bone marrow samples.5

The JAK2 V617F mutation is a result of a single change in the DNA nucleotide base pair that causes a substitution of a valine amino acid for a phenylalanine amino acid at the 617 position on exon 14 within the JAK2 kinase regulatory domain. This point mutation disrupts the regular control of the JAK2 by removing its ability to turn off, leading to uncontrolled blood cell growth.6 When the JAK2 V617F mutation cannot be demonstrated in a patient with the hallmarks of an MPN, the detection of other JAK2 and MPL proto-oncogene, thrombopoietin receptor mutations may be used as a diagnostic procedure for other MPNs.7

Other mutations incorporated in JAK2 domain can be detected in the coding portions of the DNA known as exons. One such mutation is the JAK2 exon 12, which is involved in JAK2 V617F-negative PV patients. This mutation is not detected in patients with ET or PMF and is 2%-5% present in patients with PV. There are other somatic mutations in the thrombopoietin receptors that work in accordance with thrombopoietin: MPL W515L and MPL W515K, which are found at chromosome 1p34, are identified in about 5% of PMF and 1% of ET patients, but are not present in PV patients.8.9

Pikman and colleagues reported in 2009 that the JAK2 V617F mutation is not acquired randomly.9 Their findings showed that, only in white populations, does the JAK2 V617F mutation arise preferentially on a specific constitutional JAK2 46/1 haplotype. According to the authors, the preconceived notion a of randomly acquired JAK2 V617F mutation does not account for familial MPN’s. Familial MPNs are thought to be produced by sporadic and extremely penetrant substitutions in genes that still are not identified and the 46/1 haplotype does not explain for the phenotypic diversity correlated with the JAK2 V617F gene. The 46/1 haplotype, however, correlates more frequently with different MPN subtypes. There are two hypotheses that try to explain how an acquired mutation as prevailing as the JAK2 V617F mutation can be associated with certain inherited backgrounds. The first hypothesis asserts the V617F accumulates at a faster rate than other genes because of the fundamentally unstable genetics of the 46/1 haplotype. The second theory is that all the mutated genes, including the V617F, arise at equal rates, but 46/1 may grant a selective advantage to the V617F-positive clone or interacts in some way to increase the likelihood of abnormal blood counts. A study that examined both these hypotheses concluded that the 46/1 haplotype was present more frequently in patients with myeloproliferative disorders than in their control groups and even more so in cases that were proven to be V617F-positive.10

There are very few cases that have reported familial MPN’s, especially as the pedigrees of the familial MPN’s illustrate that inheritance patterns are notably heterogeneous, indicating that there may be a range of different germline mutations driving the susceptibility. With recent data, the JAK2 V617F mutation in tandem with MPL W515L/K and inactivating TET2 mutations still continue to be the most frequently acquired mutations involved in both familial and sporadic MPN. As far as we know, there have been no cases to prove that JAK2 V617F and MPL W515L/K mutations are inherited through the germline, but there are other alleles that may pass through the germline that can be associated with hereditary thrombocytosis.11 Further cytogenetic studies will clarify the pathogenesis of these disorders and possibly lead to effective targeted therapies.

References

1. Murphy S, Peterson P, Iland H, Laszio J. Experience of the Polycythemia Vera Study Group with essential thrombocythemia: a final report on diagnostic criteria, survival, and leukemic transition by treatment. Semin Hematol. 1997;34:29-39.

2. Zhan H, Spivak JL. The diagnosis and management of polycythemia vera, essential thrombocythemia, and primary myelofibrosis in the JAK2 V617F era. Clin Adv Hematol Oncol. 2009;7:334-342.

3. Higgs JR, Sadek I, Neumann PE, et al. Familial essential thrombocythemia with spontaneous megakaryocyte colony formation and acquired JAK2 mutations. Leukemia. 2008;22:1551-1556.

4. Senyak Z. Eileen Wiggins – out of the blue. http://www.mpnresearchfoundation.org/White-Paper-3A-Nature-2C-Nurture-2C-or-Both-3F. Published October 2010. Accessed May 23, 2017.

5. Cankovic M, Whiteley L, Hawley RC, Zarbo RJ, Chitale D. Clinical performance of JAK2 V617F mutation detection assays in a molecular diagnostics laboratory: evaluation of screening and quantitation methods. Am J Clin Pathol. 2009;132:713-721.

6. Kralovics R, Teo SS, Li S, et al. Acquisition of the V617F mutation of JAK2 is a late genetic event in a subset of patients with myeloproliferative disorders. Blood. 2006;108:1377-1380.

7. James C. The JAK2V617F mutation in polycythemia vera and other myeloproliferative disorders: one mutation for three diseases? Hematology Am Soc Hematol Educ Program. 2008:69-75.

8. Pancrazzi A, Guglielmelli P, Ponziani V, et al. A sensitive detection method for MPLW515L or MPLW515K mutation in chronic myeloproliferative disorders with locked nucleic acid-modified probes and real-time polymerase chain reaction. J Mol Diagn. 2008;10:435-441.

9. Pikman Y, Lee BH, Mercher T, et al. MPLW515L is a novel somatic activating mutation in myelofibrosis with myeloid metaplasia. PLoS Med. 2006;3:1140-1151.

10. Jones AV, Campbell PJ, Beer PA, et al. The JAK2 46/1 haplotype predisposes to MPL-mutated myeloproliferative neoplasms. Blood. 2010;115:4517-4523.

11. Jones AV, Cross NCP. Inherited predisposition to myeloproliferative neoplasms. Ther Adv Hematol. 2013;4:237-253.

References

1. Murphy S, Peterson P, Iland H, Laszio J. Experience of the Polycythemia Vera Study Group with essential thrombocythemia: a final report on diagnostic criteria, survival, and leukemic transition by treatment. Semin Hematol. 1997;34:29-39.

2. Zhan H, Spivak JL. The diagnosis and management of polycythemia vera, essential thrombocythemia, and primary myelofibrosis in the JAK2 V617F era. Clin Adv Hematol Oncol. 2009;7:334-342.

3. Higgs JR, Sadek I, Neumann PE, et al. Familial essential thrombocythemia with spontaneous megakaryocyte colony formation and acquired JAK2 mutations. Leukemia. 2008;22:1551-1556.

4. Senyak Z. Eileen Wiggins – out of the blue. http://www.mpnresearchfoundation.org/White-Paper-3A-Nature-2C-Nurture-2C-or-Both-3F. Published October 2010. Accessed May 23, 2017.

5. Cankovic M, Whiteley L, Hawley RC, Zarbo RJ, Chitale D. Clinical performance of JAK2 V617F mutation detection assays in a molecular diagnostics laboratory: evaluation of screening and quantitation methods. Am J Clin Pathol. 2009;132:713-721.

6. Kralovics R, Teo SS, Li S, et al. Acquisition of the V617F mutation of JAK2 is a late genetic event in a subset of patients with myeloproliferative disorders. Blood. 2006;108:1377-1380.

7. James C. The JAK2V617F mutation in polycythemia vera and other myeloproliferative disorders: one mutation for three diseases? Hematology Am Soc Hematol Educ Program. 2008:69-75.

8. Pancrazzi A, Guglielmelli P, Ponziani V, et al. A sensitive detection method for MPLW515L or MPLW515K mutation in chronic myeloproliferative disorders with locked nucleic acid-modified probes and real-time polymerase chain reaction. J Mol Diagn. 2008;10:435-441.

9. Pikman Y, Lee BH, Mercher T, et al. MPLW515L is a novel somatic activating mutation in myelofibrosis with myeloid metaplasia. PLoS Med. 2006;3:1140-1151.

10. Jones AV, Campbell PJ, Beer PA, et al. The JAK2 46/1 haplotype predisposes to MPL-mutated myeloproliferative neoplasms. Blood. 2010;115:4517-4523.

11. Jones AV, Cross NCP. Inherited predisposition to myeloproliferative neoplasms. Ther Adv Hematol. 2013;4:237-253.

Issue
The Journal of Community and Supportive Oncology - 15(5)
Issue
The Journal of Community and Supportive Oncology - 15(5)
Publications
Publications
Topics
Article Type
Sections
Citation Override
JCSO 2017;1(5):e274-e276
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Article PDF Media

Management of tonsillar carcinoma with advanced radiation therapy and chemotherapy techniques

Article Type
Changed
Fri, 01/04/2019 - 11:16

Tonsillar carcinoma is the most common of the oropharyngeal malignancies of the head and neck region after thyroid and laryngeal carcinoma. Squamous cell carcinoma is the most frequent histologic type of these tumors.1 Tonsillar tumors may originate in the oral cavity, oropharynx, hypopharynx, or larynx. In the United States, more than 5,000 new cases of oropharynx cancer are diagnosed annually.2 Men are affected three to four times more often than are women, and the rate of incidence increases after the 4th decade of life.3 Surveillance, Epidemiology, and End Results data from 1975-2004 show that tonsillar squamous cell carcinoma has had one of the largest increases in the male-to-female incidence rate ratios.4 The overall incidence of tonsillar carcinoma is increasing, especially in the younger population, and this may be attributed to increasing rates of human papilloma virus.5,6

Squamous cell carcinoma in the head and neck originate from subsites within the oral cavity, oropharynx, hypopharynx, larynx, and nasopharynx.7 Traditionally, alcohol consumption and tobacco use were considered the most significant risk factors for the development of tonsillar cancer.8 More recently, however, the high-risk oncogenic human papilloma virus has emerged as a clinical entity in the pathogenesis of squamous cell carcinoma in the head and neck. Other risk factors include poor oral hygiene, mechanical irritation, chewing of betel quid preparations, and a lack of vegetables and fruits in the diet.9-11 Squamous cell carcinoma of the oropharynx often presents late with lymph node involvement at the time of diagnosis. Nonspecific symptoms such as a sore throat and dysphagia can allow head and neck cancer to evade early detection. Many patients with tonsillar carcinoma present with advanced disease because early lesions are generally asymptomatic when small. This absence of symptoms is responsible for 67%-77% of patients presenting with tumors larger than 2.0 cm and often with regional nodal metastasis. At presentation, 45% of anterior tonsillar pillar lesions and 76% of tonsillar fossa lesions have clinically positive necks.12

Despite significant treatment advances, the management of advanced squamous cell carcinoma of the tonsil remains challenging. Historically, surgery was considered the standard of care for patients with tonsillar carcinoma with or without postoperative adjuvant radiotherapy. In locally advanced tonsillar carcinoma, extensive surgery with major tissue reconstruction was necessary, leading to speech dysfunction, cosmetic deformities, and difficulties in swallowing, all of which are detrimental to patient quality of life.13 Given the critical role of the oropharynx in speech and swallowing, nonsurgical therapy with organ-preserving chemoradiation has gained a greater role in the treatment of tonsil carcinoma.13 Over the past decade, innovations in radiation therapy techniques have led to the introduction of intensity-modulated radiation therapy (IMRT) and image-guided radiation therapy (IGRT) for the treatment of various cancers including tonsillar carcinoma.14,15 IMRT is an advanced mode of conformal high-precision radiotherapy that uses computer-controlled multiple small radiation beams of varying intensities to deliver precise radiation doses to the target tissues while sparing adjacent healthy tissues.14 By incorporating three-dimensional computed-tomography (CT) or positron-emission–tomography (PET) imaging technology, IMRT allows the radiation dose to conform more precisely to the three-dimensional shape of the tumor while modulating the intensity of the radiation beam and minimizing its dose to those adjacent sensitive and unaffected organs. IGRT uses a range of two-, three-, and four-dimensional imaging techniques that improve the precision and accuracy of the delivery of the radiation dose to the targeted tumor tissue while minimizing the dose to the surrounding normal tissue during the course of radiation therapy (Figure 1). In this report, we present challenging cases of advanced tonsillar carcinoma and describe our experience in managing the disease using a hyperfractionated IMRT-IGRT based three-dimensional conformal radiation therapy protocol with concurrent chemotherapy.


 

Case presentations and summaries

Case 1

A 52-year-old white, nonsmoking man who worked in a research chemical laboratory, presented with complaints of throat pain and difficulty in swallowing. The patient had a history of asthma and allergies and had been seen by an ear, nose, and throat (ENT) specialist prior to his visit to our oncology center. A biopsy was performed on a right tonsillar mass measuring 2.7 x 3.6 cm. A computed-tomography (CT) scan showed 2 enlarged inhomogeneous lymph nodes measuring 2.9 cm and 1.7 cm. The nodes were well defined with no soft tissue edema. Neoplasm was favored as a diagnosis and biopsy of the mass was carried out. A biopsy specimen measuring 1.0 x 0.4 x 0.3 cm revealed a moderately differentiated infiltrating squamous cell carcinoma, which extended to the edge of the biopsy specimen. The patient’s Karnofsky performance status was 90% (ie, able to carry on normal activity; minor signs or symptoms of disease).

 

 

A CT scan of the chest was clear with no evidence of malignant involvement. A subsequent CT scan of the neck revealed a primary neoplasm of the right faucial tonsil measuring 3.3 x 3.0 cm and associated with right level II, level III, and level IV pathological lymphadenopathy. Positron-emission tomography (PET) imaging of the neck revealed a right tonsillar lesion of 2.7 x 3.0 cm involving the right parapharyngeal space (Figure 2, Case 1). The standardized uptake value (SUV) of the PET scan of the primary lesion was measured at 7.3. A cluster of right level II cervical nodes measuring 3.2 x 2.5 cm had an SUV of 3.5. A 1.0-cm right level III jugular node was also seen with an SUV of 1.6, and a right level IV lymph node measuring 1.5 x 1.0 cm was seen with an SUV of 1.8. No other lesions were noted. The tumor stage was T2N2bM0, a stage IVa disease.



The patient had a percutaneous endoscopic gastrostomy (PEG) tube placement before starting radiation. He underwent a course of hyperfractionated intensity-modulated radiation therapy with image guidance (IMRT-IGRT) in 67 fractions of 120 cGy twice a day to a final tumor dose of 8,040 cGy.16 Concurrently, the patient received systemic chemotherapy with carboplatin at a dose of 240 mg weekly. To optimize the treatment, molecular profiling was performed to identify the sensitive genetic targets to systemic chemotherapy drugs.17, 18 Targets sensitive to paclitaxel and docetaxel were identified by molecular profiling of the tumor tissue, then chemotherapy with paclitaxel or docetaxel (25 mg/m2 weekly for 3 weeks and 1 week off) was also administered to the patient.

The follow-up after 41 months indicated that the patient had no evidence of recurrent disease (Figure 2, Case 1). Posttreatment magnetic-resonance imaging (MRI) of the neck also indicated no evidence of residual tonsillar cancer. The patient’s demographics, tumor characteristics, and the treatment details are summarized in the Table.

Case 2

A 49-year-old black male presented with throat pain and a mass seen initially by his family physician. The patient had a history of tobacco use (at least 1 cigar a day) periodically for about 10 years and had quit cigar smoking 15 years prior to developing his disease. An initial evaluation indicated that the patient had a hypopharyngeal mass in the left inferior pole of his tonsil with near occlusion of the hypopharyngeal airway. His larynx could not be visualized because of the obstructive mass. A neck lymph node measuring 3.0 cm in the left jugulodigastric region was also noted. The patient’s Karnofsky performance status was 90%. Subsequently, the patient underwent excision of the right tonsil and left tonsillar region.

The pathology of the right tonsil was found to be benign. Histology of the left tonsil revealed invasive squamous cell carcinoma. The resected tumor size measured 3.7 x 2.7 x 2.5 cm. The tumor was moderately differentiated involving the deep surgical margins. No lymphovascular invasion was seen. A PET scan revealed a mass arising from the left tonsillar pillar measuring 3.6 x 2.6 x 3.3 cm with deviation of the epiglottis posteriorly nearing the left vallecula. In addition, multiple large cervical nodal lesions in the left level II nodal chain were seen, with the largest measuring 3.1 x 3.0 x 4.5 cm with an SUV of 3.4. Displacement of the left submandibular gland with several further enlarged level II lymph nodes was observed. In the region of left vallecula, there was soft tissue thickening with increased activity measuring 2.7 x 1.5 cm, likely crossing the midline with an SUV of 5.5. The rest of the neck was negative for metastatic involvement (Figure 2, Case 2). The tumor stage was T3N2Mx, a stage IVa disease.

The patient had a Port-A-Cath placed, which caused a hemothorax after placement of the port and delayed initiating his treatment. A pretreatment MRI scan of the neck revealed multiple conglomerate hypodense peripherally enhancing nodular areas in the left neck posterior to the left submandibular gland deep to the parotid tail worrisome for necrotic lymphadenopathy. The patient underwent a course of hyperfractionated IMRT-IGRT in 67 fractions of 120 cGy twice daily for a total dose of 8,040 cGy to the primary tumor site.16 The patient had a port and PEG tube prior to initiating his radiation therapy. He received IMRT-IGRT with concurrent chemotherapy that was selected based on the recommendation of his genomic testing.17,18 The chemotherapy regimen used included carboplatin (300 mg weekly) and docetaxel (400 mg weekly). The patient had a treatment break because he was hospitalized for anemia and pancytopenia from his chemotherapy and he received supportive cancer care with epoetin alfa.A post therapy PET scan was negative for evidence of hypermetabolic malignancy; however, a 3.3 x 2.7 cm calcified lesion representing likely level III jugular lymph node exhibited no measurable activity at that time. The follow-up after 40 months indicated that the patient had no reported recurrence of the disease (Figure 2, Case 2). The patient’s demographics, tumor characteristics, and the treatment details are summarized in the Table.
 

 

 

Case 3

A 53-year-old white man, who had no smoking or tobacco history but who was exposed to chemicals including sulfuric acid, hydrogen chloride gas, and glycols at work, presented initially with a sore throat that became more painful over time. His ENT specialist referred him for a CT scan of the neck, which revealed a left-sided neck mass measuring 2.5 cm in diameter posterior to the submandibular gland and lateral to carotid sheath and anterior to the triangle (Figure 2, Case 3). The mass appeared to be encapsulated. There was a lobulated spherical mass in the left supraglottic area with formation of the airway of the pyriform sinus and additional anterior vascular involvement was noted. The mass measured 3.6 cm in transverse diameter.

A left tonsillar biopsy specimen measuring 1.4 x 0.6 x 0.2 cm was obtained, and its pathology revealed that the patient had a metastatic squamous cell carcinoma. The left neck lymph node mass aspiration also revealed the presence of squamous cell carcinoma. A PET-CT scan staging showed a dominant tonsillar fossa mass extending from the soft palate down to the pyriform sinus measuring 4.2 x 3.8 cm, with an SUV uptake of 7.3. There was a dominant left level II necrotic lymph node presence measuring 5.0 x 3.7 cm, with an SUV of 3.0. The patient’s Karnofsky performance status was 90%. The tumor stage was T4N2M0, a stage IVa disease. The patient received a course of conformal hyperfractionated IMRT-IGRT delivered to the primary tumor in 67 fractions at 120 cGy twice daily for a total dose of 8,040 cGy16 and concurrent carboplatin chemotherapy at a weekly dose of 200 mg.

After completion of his radiation therapy, chemotherapy was changed based on genomic testing from single agent to doublet with carboplatin (area under the curve (AUC) dose of 2 or 200 mg, weekly) plus docetaxel (25 mg/m2 weekly for 3 weeks and 1 week off ).17,18 A PET scan after chemoradiation therapy revealed a marked anatomical improvement in the primary neoplastic disease seen in the faucial tonsil. The tonsillar mass noted previously had almost completely resolved over the interval, with only a mild persistent asymmetrical thickening of around 1.5 cm, with a peak SUV of 2.0. A lymph node of 2.8 x 2.0 cm was present anterior to the left sternocleidomastoid muscle exhibiting SUV of only 1.8. No other abnormal lesions were noted (Figure 2, Case 3). The patient continues to do extremely well without local recurrence of the disease 46 months after radiation therapy (see Table for patient demographics, tumor characteristics, and therapy details.)
 

Discussion

The management of patients with primary squamous cell carcinoma of the oropharyngeal remains controversial. Traditionally, early-stage tonsillar squamous cell carcinoma was managed by a single modality treatment, either by surgery or radiation therapy, each showing similar efficacy and outcomes.19 For late-stage disease, a combined approach using surgery and radiation therapy was found to be superior to single modality treatment. However, surgery in conjugation with radiation therapy has been associated with significant toxicities compared with the radiation therapy alone.13Therefore, the use of radiation therapy without surgery is becoming more common with increasingly sophisticated radiation therapy techniques and organ preservation approach in patients with squamous cell carcinoma of the tonsil.

Findings from several studies have shown that in stage I or II oropharyngeal cancer, single modality treatment with radiation therapy achieves 80%-90% of local control of the disease, but poorer outcomes are reported for locally advanced stages III/IV with a local control rate of 63%-74%.20 These findings and others have led to a shift to evaluate the clinical benefits of radiation therapy given with concurrent chemotherapy for the primary treatment of advanced stage oropharyngeal squamous cell carcinoma.20,21 Findings from a number of studies have since reported comparable efficacy and toxicity outcomes using this regimen with concurrent chemotherapy in patients with locally advanced head and neck squamous cell cancer.22-24 Synchronous carboplatin chemotherapy was used effectively as an alternative to cisplatin with fewer potential adverse effects in the good prognosis group of patients with oropharyngeal squamous cell carcinoma.25,26 For our 3 patients, we used carboplatin-based chemotherapy with concurrent advanced hyperfractionated radiation therapy techniques to successfully manage tonsillar squamous cell carcinoma and reduce renal toxicity and neuropathy.

Advanced radiation therapy techniques such as IMRT-IGRT are used routinely at the University Cancer and Diagnostic Centers in Houston, Texas, to manage a range of malignant cancers.27 These innovative techniques have the potential to deliver highly conformal dose-intense radiation to targeted regions of disease, while sparing adjacent critical nonmalignant tissue. The improved shaping of high-dose distributions with IMRT-IGRT could mitigate treatment-related toxicities. For example, the use of advanced radiation therapy techniques has been associated with increased preservation of parotid salivary flow.28-30 The use of advanced radiation therapy techniques in head and neck squamous cell carcinoma is growing, and early evidence confirms its ability to secure excellent local and regional disease control.31,32 In this study, we have demonstrated that by using hyperfractionated conformal three-dimensional IMRT-IGRT we were able not only to manage advanced tonsillar squamous cell carcinoma and treat the malignant metastasis, but also spare adjacent critical organs that were not involved in the disease, thus reducing many of the detrimental side effects associated with hyperfractionated chemoradiation.

All 3 patients were followed for between 40 and 46 months. They continue to do extremely well without local recurrence of their disease, indicating a 100% disease control and overall survival rate. The disease control and survival outcomes for our patients with stage IVA disease compare favorably to other published reports in the literature.33,34 Findings from a study by Prestwich and colleagues33 of 41 patients with stage IV tonsillar carcinoma showed that the radiation therapy with concurrent chemotherapy achieved local and regional disease control in 91% of complete responders and an overall survival rate of 66% at 3 years. Similarly, Setton and colleagues34 reported on 442 patients – 50% with tonsillar cancer, 46% with base-of-tongue cancer – who underwent IMRT and concurrent chemotherapy and who achieved a 3-year overall survival of 84.9%. Our study findings demonstrate that hyperfractionated conformal three-dimensional IMRT-IGRT with concurrent chemotherapy can be delivered safely and effectively to patients with advanced tonsillar squamous cell carcinoma.
 

 

 

Acknowledgment

The authors thank Ms June Lyliston, LVN, for editing and proofreading the manuscript.

References

1. Stambuk HE, Karimi S, Lee N, Patel SG. Oral cavity and oropharynx tumors. Radiol Clin North Am. 2007;45(1):1-20.

2. Lin DT, Cohen SM, Coppit GL, Burkey BB. Squamous cell carcinoma of the oropharynx and hypopharynx. Otolaryngol Clin North Am. 2005;38(1):59-74, viii.

3. Golas SM. Trends in palatine tonsillar cancer incidence and mortality rates in the United States. Community Dent Oral Epidemiol. 2007;35(2):98-108.

4. Cook MB, Dawsey SM, Freedman ND, et al. Sex disparities in cancer incidence by period and age. Cancer Epidemiol Biomarkers Prev. 2009;18(4):1174-1182.

5. Enomoto LM, Bann DV, Hollenbeak CS, Goldenberg D. Trends in the Incidence of oropharyngeal cancers in the United States. Otolaryngol Head Neck Surg. 2016.

6. Shiboski CH, Schmidt BL, Jordan RC. Tongue and tonsil carcinoma: increasing trends in the U.S. population ages 20-44 years. Cancer. 2005;103(9):1843-1849.

7. Marur S, Forastiere AA. Head and neck cancer: changing epidemiology, diagnosis, and treatment. Mayo Clin Proc. 2008;83(4):489-501.

8. Hong AM, Martin A, Chatfield M, et al. Human papillomavirus, smoking status and outcomes in tonsillar squamous cell carcinoma. Int J Cancer. 2013;132(12):2748-2754.

9. Velly AM, Franco EL, Schlecht N, et al. Relationship between dental factors and risk of upper aerodigestive tract cancer. Oral Oncol. 1998;34(4):284-291.

10. Farrow DC, Vaughan TL, Berwick M, et al. Diet and nasopharyngeal cancer in a low-risk population. Int J Cancer. 1998;78(6):675-679.

11. Freedman ND, Park Y, Subar AF, et al. Fruit and vegetable intake and head and neck cancer risk in a large United States prospective cohort study. Int J Cancer. 2008;122(10):2330-2336.

12. Guay ME, Lavertu P. Tonsillar carcinoma. Eur Arch Otorhinolaryngol. 1995;252(5):259-264.

13. Parsons JT, Mendenhall WM, Stringer SP, et al. Squamous cell carcinoma of the oropharynx: surgery, radiation therapy, or both. Cancer. 2002;94(11):2967-2980.

14. Yao M, Dornfeld KJ, Buatti JM, et al. Intensity-modulated radiation treatment for head-and-neck squamous cell carcinoma--the University of Iowa experience. Int J Radiat Oncol Biol Phys. 2005;63(2):410-421.

15. Yang ES, Murphy BM, Chung CH, et al. Evolution of clinical trials in head and neck cancer. Crit Rev Oncol Hematol. 2009;71(1):29-42.

16. Beitler JJ, Zhang Q, Fu KK, et al. Final results of local-regional control and late toxicity of RTOG 9003: a randomized trial of altered fractionation radiation for locally advanced head and neck cancer. Int J Radiat Oncol Biol Phys. 2014;89(1):13-20.

17. Tomkiewicz C, Hans S, Mucchielli MH, et al. A head and neck cancer tumor response-specific gene signature for cisplatin, 5-fluorouracil induction chemotherapy fails with added taxanes. PLoS One. 2012;7(10):e47170.

18. Feldman R, Gatalica Z, Knezetic J, et al. Molecular profiling of head and neck squamous cell carcinoma. Head Neck. 2016;38 Suppl 1:E1625-1638.

19. Moose BD, Kelly MD, Levine PA, et al. Definitive radiotherapy for T1 and T2 squamous cell carcinoma of the tonsil. Head Neck. 1995;17(4):334-338.

20. Chen AY, Schrag N, Hao Y, Stewart A, Ward E. Changes in treatment of advanced oropharyngeal cancer, 1985-2001. Laryngoscope. 2007;117(1):16-21.

21. Machtay M, Rosenthal DI, Hershock D, et al. Organ preservation therapy using induction plus concurrent chemoradiation for advanced resectable oropharyngeal carcinoma: a University of Pennsylvania Phase II Trial. J Clin Oncol. 2002;20(19):3964-3971.

22. Jegannathen A, Swindell R, Yap B, et al. Can synchronous chemotherapy be added to accelerated hypofractionated radiotherapy in patients with base of tongue cancer? Clin Oncol (R Coll Radiol). 2010;22(3):185-191.

23. Budach V, Becker ET, Boehmer D, et al. Concurrent hyperfractionated accelerated radiotherapy with 5-FU and once weekly cisplatin in locally advanced head and neck cancer. The 10-year results of a prospective phase II trial. Strahlenther Onkol. 2014;190(3):250-255.

24. Tobias JS, Monson K, Gupta N, et al. Chemoradiotherapy for locally advanced head and neck cancer: 10-year follow-up of the UK Head and Neck (UKHAN1) trial. Lancet Oncol. 2010;11(1):66-74.

25. Wilkins AC, Rosenfelder N, Schick U, et al. Equivalence of cisplatin and carboplatin-based chemoradiation for locally advanced squamous cell carcinoma of the head and neck: a matched-pair analysis. Oral Oncol. 2013;49(6):615-619.

26. Benghiat H, Sanghera P, Cashmore1 J, et al. Four week hypofractionated accelerated intensity modulated radiotherapy and synchronous carboplatin or cetuximab in biologically staged oropharyngeal carcinoma. Cancer and Clinical Oncology. 2014;3:1-9.

27. D’Andrea MA, Reddy GK. Management of metastatic malignant thymoma with advanced radiation and chemotherapy techniques: report of a rare case. World J Surg Oncol. 2015;13:77.

28. Little M, Schipper M, Feng FY, et al. Reducing xerostomia after chemo-IMRT for head-and-neck cancer: beyond sparing the parotid glands. Int J Radiat Oncol Biol Phys. 2012;83(3):1007-1014.

29. Eisbruch A. Reducing xerostomia by IMRT: what may, and may not, be achieved. J Clin Oncol. 2007;25(31):4863-4864.

30. Pow EH, Kwong DL, McMillan AS, et al. Xerostomia and quality of life after intensity-modulated radiotherapy vs. conventional radiotherapy for early-stage nasopharyngeal carcinoma: initial report on a randomized controlled clinical trial. Int J Radiat Oncol Biol Phys. 2006;66(4):981-991.

31. Lee NY, de Arruda FF, Puri DR, et al. A comparison of intensity-modulated radiation therapy and concomitant boost radiotherapy in the setting of concurrent chemotherapy for locally advanced oropharyngeal carcinoma. Int J Radiat Oncol Biol Phys. 2006;66(4):966-974.

32. Daly ME, Lieskovsky Y, Pawlicki T, et al. Evaluation of patterns of failure and subjective salivary function in patients treated with intensity modulated radiotherapy for head and neck squamous cell carcinoma. Head Neck. 2007;29(3):211-220.

33. Prestwich RJ, Kancherla K, Oksuz DC, et al. A single centre experience with sequential and concomitant chemoradiotherapy in locally advanced stage IV tonsillar cancer. Radiat Oncol. 2010;5:121.

34. Setton J, Caria N, Romanyshyn J, et al. Intensity-modulated radiotherapy in the treatment of oropharyngeal cancer: an update of the Memorial Sloan-Kettering Cancer Center experience. Int J Radiat Oncol Biol Phys. 2012;82(1):291-298.

Article PDF
Author and Disclosure Information

Mark A D’Andrea, MD, FACRO; Marshall Clarke; and G Kesava Reddy, PhD, MHA

University Cancer and Diagnostic Centers, Houston, Texas

Publications
Topics
Sections
Author and Disclosure Information

Mark A D’Andrea, MD, FACRO; Marshall Clarke; and G Kesava Reddy, PhD, MHA

University Cancer and Diagnostic Centers, Houston, Texas

Author and Disclosure Information

Mark A D’Andrea, MD, FACRO; Marshall Clarke; and G Kesava Reddy, PhD, MHA

University Cancer and Diagnostic Centers, Houston, Texas

Article PDF
Article PDF

Tonsillar carcinoma is the most common of the oropharyngeal malignancies of the head and neck region after thyroid and laryngeal carcinoma. Squamous cell carcinoma is the most frequent histologic type of these tumors.1 Tonsillar tumors may originate in the oral cavity, oropharynx, hypopharynx, or larynx. In the United States, more than 5,000 new cases of oropharynx cancer are diagnosed annually.2 Men are affected three to four times more often than are women, and the rate of incidence increases after the 4th decade of life.3 Surveillance, Epidemiology, and End Results data from 1975-2004 show that tonsillar squamous cell carcinoma has had one of the largest increases in the male-to-female incidence rate ratios.4 The overall incidence of tonsillar carcinoma is increasing, especially in the younger population, and this may be attributed to increasing rates of human papilloma virus.5,6

Squamous cell carcinoma in the head and neck originate from subsites within the oral cavity, oropharynx, hypopharynx, larynx, and nasopharynx.7 Traditionally, alcohol consumption and tobacco use were considered the most significant risk factors for the development of tonsillar cancer.8 More recently, however, the high-risk oncogenic human papilloma virus has emerged as a clinical entity in the pathogenesis of squamous cell carcinoma in the head and neck. Other risk factors include poor oral hygiene, mechanical irritation, chewing of betel quid preparations, and a lack of vegetables and fruits in the diet.9-11 Squamous cell carcinoma of the oropharynx often presents late with lymph node involvement at the time of diagnosis. Nonspecific symptoms such as a sore throat and dysphagia can allow head and neck cancer to evade early detection. Many patients with tonsillar carcinoma present with advanced disease because early lesions are generally asymptomatic when small. This absence of symptoms is responsible for 67%-77% of patients presenting with tumors larger than 2.0 cm and often with regional nodal metastasis. At presentation, 45% of anterior tonsillar pillar lesions and 76% of tonsillar fossa lesions have clinically positive necks.12

Despite significant treatment advances, the management of advanced squamous cell carcinoma of the tonsil remains challenging. Historically, surgery was considered the standard of care for patients with tonsillar carcinoma with or without postoperative adjuvant radiotherapy. In locally advanced tonsillar carcinoma, extensive surgery with major tissue reconstruction was necessary, leading to speech dysfunction, cosmetic deformities, and difficulties in swallowing, all of which are detrimental to patient quality of life.13 Given the critical role of the oropharynx in speech and swallowing, nonsurgical therapy with organ-preserving chemoradiation has gained a greater role in the treatment of tonsil carcinoma.13 Over the past decade, innovations in radiation therapy techniques have led to the introduction of intensity-modulated radiation therapy (IMRT) and image-guided radiation therapy (IGRT) for the treatment of various cancers including tonsillar carcinoma.14,15 IMRT is an advanced mode of conformal high-precision radiotherapy that uses computer-controlled multiple small radiation beams of varying intensities to deliver precise radiation doses to the target tissues while sparing adjacent healthy tissues.14 By incorporating three-dimensional computed-tomography (CT) or positron-emission–tomography (PET) imaging technology, IMRT allows the radiation dose to conform more precisely to the three-dimensional shape of the tumor while modulating the intensity of the radiation beam and minimizing its dose to those adjacent sensitive and unaffected organs. IGRT uses a range of two-, three-, and four-dimensional imaging techniques that improve the precision and accuracy of the delivery of the radiation dose to the targeted tumor tissue while minimizing the dose to the surrounding normal tissue during the course of radiation therapy (Figure 1). In this report, we present challenging cases of advanced tonsillar carcinoma and describe our experience in managing the disease using a hyperfractionated IMRT-IGRT based three-dimensional conformal radiation therapy protocol with concurrent chemotherapy.


 

Case presentations and summaries

Case 1

A 52-year-old white, nonsmoking man who worked in a research chemical laboratory, presented with complaints of throat pain and difficulty in swallowing. The patient had a history of asthma and allergies and had been seen by an ear, nose, and throat (ENT) specialist prior to his visit to our oncology center. A biopsy was performed on a right tonsillar mass measuring 2.7 x 3.6 cm. A computed-tomography (CT) scan showed 2 enlarged inhomogeneous lymph nodes measuring 2.9 cm and 1.7 cm. The nodes were well defined with no soft tissue edema. Neoplasm was favored as a diagnosis and biopsy of the mass was carried out. A biopsy specimen measuring 1.0 x 0.4 x 0.3 cm revealed a moderately differentiated infiltrating squamous cell carcinoma, which extended to the edge of the biopsy specimen. The patient’s Karnofsky performance status was 90% (ie, able to carry on normal activity; minor signs or symptoms of disease).

 

 

A CT scan of the chest was clear with no evidence of malignant involvement. A subsequent CT scan of the neck revealed a primary neoplasm of the right faucial tonsil measuring 3.3 x 3.0 cm and associated with right level II, level III, and level IV pathological lymphadenopathy. Positron-emission tomography (PET) imaging of the neck revealed a right tonsillar lesion of 2.7 x 3.0 cm involving the right parapharyngeal space (Figure 2, Case 1). The standardized uptake value (SUV) of the PET scan of the primary lesion was measured at 7.3. A cluster of right level II cervical nodes measuring 3.2 x 2.5 cm had an SUV of 3.5. A 1.0-cm right level III jugular node was also seen with an SUV of 1.6, and a right level IV lymph node measuring 1.5 x 1.0 cm was seen with an SUV of 1.8. No other lesions were noted. The tumor stage was T2N2bM0, a stage IVa disease.



The patient had a percutaneous endoscopic gastrostomy (PEG) tube placement before starting radiation. He underwent a course of hyperfractionated intensity-modulated radiation therapy with image guidance (IMRT-IGRT) in 67 fractions of 120 cGy twice a day to a final tumor dose of 8,040 cGy.16 Concurrently, the patient received systemic chemotherapy with carboplatin at a dose of 240 mg weekly. To optimize the treatment, molecular profiling was performed to identify the sensitive genetic targets to systemic chemotherapy drugs.17, 18 Targets sensitive to paclitaxel and docetaxel were identified by molecular profiling of the tumor tissue, then chemotherapy with paclitaxel or docetaxel (25 mg/m2 weekly for 3 weeks and 1 week off) was also administered to the patient.

The follow-up after 41 months indicated that the patient had no evidence of recurrent disease (Figure 2, Case 1). Posttreatment magnetic-resonance imaging (MRI) of the neck also indicated no evidence of residual tonsillar cancer. The patient’s demographics, tumor characteristics, and the treatment details are summarized in the Table.

Case 2

A 49-year-old black male presented with throat pain and a mass seen initially by his family physician. The patient had a history of tobacco use (at least 1 cigar a day) periodically for about 10 years and had quit cigar smoking 15 years prior to developing his disease. An initial evaluation indicated that the patient had a hypopharyngeal mass in the left inferior pole of his tonsil with near occlusion of the hypopharyngeal airway. His larynx could not be visualized because of the obstructive mass. A neck lymph node measuring 3.0 cm in the left jugulodigastric region was also noted. The patient’s Karnofsky performance status was 90%. Subsequently, the patient underwent excision of the right tonsil and left tonsillar region.

The pathology of the right tonsil was found to be benign. Histology of the left tonsil revealed invasive squamous cell carcinoma. The resected tumor size measured 3.7 x 2.7 x 2.5 cm. The tumor was moderately differentiated involving the deep surgical margins. No lymphovascular invasion was seen. A PET scan revealed a mass arising from the left tonsillar pillar measuring 3.6 x 2.6 x 3.3 cm with deviation of the epiglottis posteriorly nearing the left vallecula. In addition, multiple large cervical nodal lesions in the left level II nodal chain were seen, with the largest measuring 3.1 x 3.0 x 4.5 cm with an SUV of 3.4. Displacement of the left submandibular gland with several further enlarged level II lymph nodes was observed. In the region of left vallecula, there was soft tissue thickening with increased activity measuring 2.7 x 1.5 cm, likely crossing the midline with an SUV of 5.5. The rest of the neck was negative for metastatic involvement (Figure 2, Case 2). The tumor stage was T3N2Mx, a stage IVa disease.

The patient had a Port-A-Cath placed, which caused a hemothorax after placement of the port and delayed initiating his treatment. A pretreatment MRI scan of the neck revealed multiple conglomerate hypodense peripherally enhancing nodular areas in the left neck posterior to the left submandibular gland deep to the parotid tail worrisome for necrotic lymphadenopathy. The patient underwent a course of hyperfractionated IMRT-IGRT in 67 fractions of 120 cGy twice daily for a total dose of 8,040 cGy to the primary tumor site.16 The patient had a port and PEG tube prior to initiating his radiation therapy. He received IMRT-IGRT with concurrent chemotherapy that was selected based on the recommendation of his genomic testing.17,18 The chemotherapy regimen used included carboplatin (300 mg weekly) and docetaxel (400 mg weekly). The patient had a treatment break because he was hospitalized for anemia and pancytopenia from his chemotherapy and he received supportive cancer care with epoetin alfa.A post therapy PET scan was negative for evidence of hypermetabolic malignancy; however, a 3.3 x 2.7 cm calcified lesion representing likely level III jugular lymph node exhibited no measurable activity at that time. The follow-up after 40 months indicated that the patient had no reported recurrence of the disease (Figure 2, Case 2). The patient’s demographics, tumor characteristics, and the treatment details are summarized in the Table.
 

 

 

Case 3

A 53-year-old white man, who had no smoking or tobacco history but who was exposed to chemicals including sulfuric acid, hydrogen chloride gas, and glycols at work, presented initially with a sore throat that became more painful over time. His ENT specialist referred him for a CT scan of the neck, which revealed a left-sided neck mass measuring 2.5 cm in diameter posterior to the submandibular gland and lateral to carotid sheath and anterior to the triangle (Figure 2, Case 3). The mass appeared to be encapsulated. There was a lobulated spherical mass in the left supraglottic area with formation of the airway of the pyriform sinus and additional anterior vascular involvement was noted. The mass measured 3.6 cm in transverse diameter.

A left tonsillar biopsy specimen measuring 1.4 x 0.6 x 0.2 cm was obtained, and its pathology revealed that the patient had a metastatic squamous cell carcinoma. The left neck lymph node mass aspiration also revealed the presence of squamous cell carcinoma. A PET-CT scan staging showed a dominant tonsillar fossa mass extending from the soft palate down to the pyriform sinus measuring 4.2 x 3.8 cm, with an SUV uptake of 7.3. There was a dominant left level II necrotic lymph node presence measuring 5.0 x 3.7 cm, with an SUV of 3.0. The patient’s Karnofsky performance status was 90%. The tumor stage was T4N2M0, a stage IVa disease. The patient received a course of conformal hyperfractionated IMRT-IGRT delivered to the primary tumor in 67 fractions at 120 cGy twice daily for a total dose of 8,040 cGy16 and concurrent carboplatin chemotherapy at a weekly dose of 200 mg.

After completion of his radiation therapy, chemotherapy was changed based on genomic testing from single agent to doublet with carboplatin (area under the curve (AUC) dose of 2 or 200 mg, weekly) plus docetaxel (25 mg/m2 weekly for 3 weeks and 1 week off ).17,18 A PET scan after chemoradiation therapy revealed a marked anatomical improvement in the primary neoplastic disease seen in the faucial tonsil. The tonsillar mass noted previously had almost completely resolved over the interval, with only a mild persistent asymmetrical thickening of around 1.5 cm, with a peak SUV of 2.0. A lymph node of 2.8 x 2.0 cm was present anterior to the left sternocleidomastoid muscle exhibiting SUV of only 1.8. No other abnormal lesions were noted (Figure 2, Case 3). The patient continues to do extremely well without local recurrence of the disease 46 months after radiation therapy (see Table for patient demographics, tumor characteristics, and therapy details.)
 

Discussion

The management of patients with primary squamous cell carcinoma of the oropharyngeal remains controversial. Traditionally, early-stage tonsillar squamous cell carcinoma was managed by a single modality treatment, either by surgery or radiation therapy, each showing similar efficacy and outcomes.19 For late-stage disease, a combined approach using surgery and radiation therapy was found to be superior to single modality treatment. However, surgery in conjugation with radiation therapy has been associated with significant toxicities compared with the radiation therapy alone.13Therefore, the use of radiation therapy without surgery is becoming more common with increasingly sophisticated radiation therapy techniques and organ preservation approach in patients with squamous cell carcinoma of the tonsil.

Findings from several studies have shown that in stage I or II oropharyngeal cancer, single modality treatment with radiation therapy achieves 80%-90% of local control of the disease, but poorer outcomes are reported for locally advanced stages III/IV with a local control rate of 63%-74%.20 These findings and others have led to a shift to evaluate the clinical benefits of radiation therapy given with concurrent chemotherapy for the primary treatment of advanced stage oropharyngeal squamous cell carcinoma.20,21 Findings from a number of studies have since reported comparable efficacy and toxicity outcomes using this regimen with concurrent chemotherapy in patients with locally advanced head and neck squamous cell cancer.22-24 Synchronous carboplatin chemotherapy was used effectively as an alternative to cisplatin with fewer potential adverse effects in the good prognosis group of patients with oropharyngeal squamous cell carcinoma.25,26 For our 3 patients, we used carboplatin-based chemotherapy with concurrent advanced hyperfractionated radiation therapy techniques to successfully manage tonsillar squamous cell carcinoma and reduce renal toxicity and neuropathy.

Advanced radiation therapy techniques such as IMRT-IGRT are used routinely at the University Cancer and Diagnostic Centers in Houston, Texas, to manage a range of malignant cancers.27 These innovative techniques have the potential to deliver highly conformal dose-intense radiation to targeted regions of disease, while sparing adjacent critical nonmalignant tissue. The improved shaping of high-dose distributions with IMRT-IGRT could mitigate treatment-related toxicities. For example, the use of advanced radiation therapy techniques has been associated with increased preservation of parotid salivary flow.28-30 The use of advanced radiation therapy techniques in head and neck squamous cell carcinoma is growing, and early evidence confirms its ability to secure excellent local and regional disease control.31,32 In this study, we have demonstrated that by using hyperfractionated conformal three-dimensional IMRT-IGRT we were able not only to manage advanced tonsillar squamous cell carcinoma and treat the malignant metastasis, but also spare adjacent critical organs that were not involved in the disease, thus reducing many of the detrimental side effects associated with hyperfractionated chemoradiation.

All 3 patients were followed for between 40 and 46 months. They continue to do extremely well without local recurrence of their disease, indicating a 100% disease control and overall survival rate. The disease control and survival outcomes for our patients with stage IVA disease compare favorably to other published reports in the literature.33,34 Findings from a study by Prestwich and colleagues33 of 41 patients with stage IV tonsillar carcinoma showed that the radiation therapy with concurrent chemotherapy achieved local and regional disease control in 91% of complete responders and an overall survival rate of 66% at 3 years. Similarly, Setton and colleagues34 reported on 442 patients – 50% with tonsillar cancer, 46% with base-of-tongue cancer – who underwent IMRT and concurrent chemotherapy and who achieved a 3-year overall survival of 84.9%. Our study findings demonstrate that hyperfractionated conformal three-dimensional IMRT-IGRT with concurrent chemotherapy can be delivered safely and effectively to patients with advanced tonsillar squamous cell carcinoma.
 

 

 

Acknowledgment

The authors thank Ms June Lyliston, LVN, for editing and proofreading the manuscript.

Tonsillar carcinoma is the most common of the oropharyngeal malignancies of the head and neck region after thyroid and laryngeal carcinoma. Squamous cell carcinoma is the most frequent histologic type of these tumors.1 Tonsillar tumors may originate in the oral cavity, oropharynx, hypopharynx, or larynx. In the United States, more than 5,000 new cases of oropharynx cancer are diagnosed annually.2 Men are affected three to four times more often than are women, and the rate of incidence increases after the 4th decade of life.3 Surveillance, Epidemiology, and End Results data from 1975-2004 show that tonsillar squamous cell carcinoma has had one of the largest increases in the male-to-female incidence rate ratios.4 The overall incidence of tonsillar carcinoma is increasing, especially in the younger population, and this may be attributed to increasing rates of human papilloma virus.5,6

Squamous cell carcinoma in the head and neck originate from subsites within the oral cavity, oropharynx, hypopharynx, larynx, and nasopharynx.7 Traditionally, alcohol consumption and tobacco use were considered the most significant risk factors for the development of tonsillar cancer.8 More recently, however, the high-risk oncogenic human papilloma virus has emerged as a clinical entity in the pathogenesis of squamous cell carcinoma in the head and neck. Other risk factors include poor oral hygiene, mechanical irritation, chewing of betel quid preparations, and a lack of vegetables and fruits in the diet.9-11 Squamous cell carcinoma of the oropharynx often presents late with lymph node involvement at the time of diagnosis. Nonspecific symptoms such as a sore throat and dysphagia can allow head and neck cancer to evade early detection. Many patients with tonsillar carcinoma present with advanced disease because early lesions are generally asymptomatic when small. This absence of symptoms is responsible for 67%-77% of patients presenting with tumors larger than 2.0 cm and often with regional nodal metastasis. At presentation, 45% of anterior tonsillar pillar lesions and 76% of tonsillar fossa lesions have clinically positive necks.12

Despite significant treatment advances, the management of advanced squamous cell carcinoma of the tonsil remains challenging. Historically, surgery was considered the standard of care for patients with tonsillar carcinoma with or without postoperative adjuvant radiotherapy. In locally advanced tonsillar carcinoma, extensive surgery with major tissue reconstruction was necessary, leading to speech dysfunction, cosmetic deformities, and difficulties in swallowing, all of which are detrimental to patient quality of life.13 Given the critical role of the oropharynx in speech and swallowing, nonsurgical therapy with organ-preserving chemoradiation has gained a greater role in the treatment of tonsil carcinoma.13 Over the past decade, innovations in radiation therapy techniques have led to the introduction of intensity-modulated radiation therapy (IMRT) and image-guided radiation therapy (IGRT) for the treatment of various cancers including tonsillar carcinoma.14,15 IMRT is an advanced mode of conformal high-precision radiotherapy that uses computer-controlled multiple small radiation beams of varying intensities to deliver precise radiation doses to the target tissues while sparing adjacent healthy tissues.14 By incorporating three-dimensional computed-tomography (CT) or positron-emission–tomography (PET) imaging technology, IMRT allows the radiation dose to conform more precisely to the three-dimensional shape of the tumor while modulating the intensity of the radiation beam and minimizing its dose to those adjacent sensitive and unaffected organs. IGRT uses a range of two-, three-, and four-dimensional imaging techniques that improve the precision and accuracy of the delivery of the radiation dose to the targeted tumor tissue while minimizing the dose to the surrounding normal tissue during the course of radiation therapy (Figure 1). In this report, we present challenging cases of advanced tonsillar carcinoma and describe our experience in managing the disease using a hyperfractionated IMRT-IGRT based three-dimensional conformal radiation therapy protocol with concurrent chemotherapy.


 

Case presentations and summaries

Case 1

A 52-year-old white, nonsmoking man who worked in a research chemical laboratory, presented with complaints of throat pain and difficulty in swallowing. The patient had a history of asthma and allergies and had been seen by an ear, nose, and throat (ENT) specialist prior to his visit to our oncology center. A biopsy was performed on a right tonsillar mass measuring 2.7 x 3.6 cm. A computed-tomography (CT) scan showed 2 enlarged inhomogeneous lymph nodes measuring 2.9 cm and 1.7 cm. The nodes were well defined with no soft tissue edema. Neoplasm was favored as a diagnosis and biopsy of the mass was carried out. A biopsy specimen measuring 1.0 x 0.4 x 0.3 cm revealed a moderately differentiated infiltrating squamous cell carcinoma, which extended to the edge of the biopsy specimen. The patient’s Karnofsky performance status was 90% (ie, able to carry on normal activity; minor signs or symptoms of disease).

 

 

A CT scan of the chest was clear with no evidence of malignant involvement. A subsequent CT scan of the neck revealed a primary neoplasm of the right faucial tonsil measuring 3.3 x 3.0 cm and associated with right level II, level III, and level IV pathological lymphadenopathy. Positron-emission tomography (PET) imaging of the neck revealed a right tonsillar lesion of 2.7 x 3.0 cm involving the right parapharyngeal space (Figure 2, Case 1). The standardized uptake value (SUV) of the PET scan of the primary lesion was measured at 7.3. A cluster of right level II cervical nodes measuring 3.2 x 2.5 cm had an SUV of 3.5. A 1.0-cm right level III jugular node was also seen with an SUV of 1.6, and a right level IV lymph node measuring 1.5 x 1.0 cm was seen with an SUV of 1.8. No other lesions were noted. The tumor stage was T2N2bM0, a stage IVa disease.



The patient had a percutaneous endoscopic gastrostomy (PEG) tube placement before starting radiation. He underwent a course of hyperfractionated intensity-modulated radiation therapy with image guidance (IMRT-IGRT) in 67 fractions of 120 cGy twice a day to a final tumor dose of 8,040 cGy.16 Concurrently, the patient received systemic chemotherapy with carboplatin at a dose of 240 mg weekly. To optimize the treatment, molecular profiling was performed to identify the sensitive genetic targets to systemic chemotherapy drugs.17, 18 Targets sensitive to paclitaxel and docetaxel were identified by molecular profiling of the tumor tissue, then chemotherapy with paclitaxel or docetaxel (25 mg/m2 weekly for 3 weeks and 1 week off) was also administered to the patient.

The follow-up after 41 months indicated that the patient had no evidence of recurrent disease (Figure 2, Case 1). Posttreatment magnetic-resonance imaging (MRI) of the neck also indicated no evidence of residual tonsillar cancer. The patient’s demographics, tumor characteristics, and the treatment details are summarized in the Table.

Case 2

A 49-year-old black male presented with throat pain and a mass seen initially by his family physician. The patient had a history of tobacco use (at least 1 cigar a day) periodically for about 10 years and had quit cigar smoking 15 years prior to developing his disease. An initial evaluation indicated that the patient had a hypopharyngeal mass in the left inferior pole of his tonsil with near occlusion of the hypopharyngeal airway. His larynx could not be visualized because of the obstructive mass. A neck lymph node measuring 3.0 cm in the left jugulodigastric region was also noted. The patient’s Karnofsky performance status was 90%. Subsequently, the patient underwent excision of the right tonsil and left tonsillar region.

The pathology of the right tonsil was found to be benign. Histology of the left tonsil revealed invasive squamous cell carcinoma. The resected tumor size measured 3.7 x 2.7 x 2.5 cm. The tumor was moderately differentiated involving the deep surgical margins. No lymphovascular invasion was seen. A PET scan revealed a mass arising from the left tonsillar pillar measuring 3.6 x 2.6 x 3.3 cm with deviation of the epiglottis posteriorly nearing the left vallecula. In addition, multiple large cervical nodal lesions in the left level II nodal chain were seen, with the largest measuring 3.1 x 3.0 x 4.5 cm with an SUV of 3.4. Displacement of the left submandibular gland with several further enlarged level II lymph nodes was observed. In the region of left vallecula, there was soft tissue thickening with increased activity measuring 2.7 x 1.5 cm, likely crossing the midline with an SUV of 5.5. The rest of the neck was negative for metastatic involvement (Figure 2, Case 2). The tumor stage was T3N2Mx, a stage IVa disease.

The patient had a Port-A-Cath placed, which caused a hemothorax after placement of the port and delayed initiating his treatment. A pretreatment MRI scan of the neck revealed multiple conglomerate hypodense peripherally enhancing nodular areas in the left neck posterior to the left submandibular gland deep to the parotid tail worrisome for necrotic lymphadenopathy. The patient underwent a course of hyperfractionated IMRT-IGRT in 67 fractions of 120 cGy twice daily for a total dose of 8,040 cGy to the primary tumor site.16 The patient had a port and PEG tube prior to initiating his radiation therapy. He received IMRT-IGRT with concurrent chemotherapy that was selected based on the recommendation of his genomic testing.17,18 The chemotherapy regimen used included carboplatin (300 mg weekly) and docetaxel (400 mg weekly). The patient had a treatment break because he was hospitalized for anemia and pancytopenia from his chemotherapy and he received supportive cancer care with epoetin alfa.A post therapy PET scan was negative for evidence of hypermetabolic malignancy; however, a 3.3 x 2.7 cm calcified lesion representing likely level III jugular lymph node exhibited no measurable activity at that time. The follow-up after 40 months indicated that the patient had no reported recurrence of the disease (Figure 2, Case 2). The patient’s demographics, tumor characteristics, and the treatment details are summarized in the Table.
 

 

 

Case 3

A 53-year-old white man, who had no smoking or tobacco history but who was exposed to chemicals including sulfuric acid, hydrogen chloride gas, and glycols at work, presented initially with a sore throat that became more painful over time. His ENT specialist referred him for a CT scan of the neck, which revealed a left-sided neck mass measuring 2.5 cm in diameter posterior to the submandibular gland and lateral to carotid sheath and anterior to the triangle (Figure 2, Case 3). The mass appeared to be encapsulated. There was a lobulated spherical mass in the left supraglottic area with formation of the airway of the pyriform sinus and additional anterior vascular involvement was noted. The mass measured 3.6 cm in transverse diameter.

A left tonsillar biopsy specimen measuring 1.4 x 0.6 x 0.2 cm was obtained, and its pathology revealed that the patient had a metastatic squamous cell carcinoma. The left neck lymph node mass aspiration also revealed the presence of squamous cell carcinoma. A PET-CT scan staging showed a dominant tonsillar fossa mass extending from the soft palate down to the pyriform sinus measuring 4.2 x 3.8 cm, with an SUV uptake of 7.3. There was a dominant left level II necrotic lymph node presence measuring 5.0 x 3.7 cm, with an SUV of 3.0. The patient’s Karnofsky performance status was 90%. The tumor stage was T4N2M0, a stage IVa disease. The patient received a course of conformal hyperfractionated IMRT-IGRT delivered to the primary tumor in 67 fractions at 120 cGy twice daily for a total dose of 8,040 cGy16 and concurrent carboplatin chemotherapy at a weekly dose of 200 mg.

After completion of his radiation therapy, chemotherapy was changed based on genomic testing from single agent to doublet with carboplatin (area under the curve (AUC) dose of 2 or 200 mg, weekly) plus docetaxel (25 mg/m2 weekly for 3 weeks and 1 week off ).17,18 A PET scan after chemoradiation therapy revealed a marked anatomical improvement in the primary neoplastic disease seen in the faucial tonsil. The tonsillar mass noted previously had almost completely resolved over the interval, with only a mild persistent asymmetrical thickening of around 1.5 cm, with a peak SUV of 2.0. A lymph node of 2.8 x 2.0 cm was present anterior to the left sternocleidomastoid muscle exhibiting SUV of only 1.8. No other abnormal lesions were noted (Figure 2, Case 3). The patient continues to do extremely well without local recurrence of the disease 46 months after radiation therapy (see Table for patient demographics, tumor characteristics, and therapy details.)
 

Discussion

The management of patients with primary squamous cell carcinoma of the oropharyngeal remains controversial. Traditionally, early-stage tonsillar squamous cell carcinoma was managed by a single modality treatment, either by surgery or radiation therapy, each showing similar efficacy and outcomes.19 For late-stage disease, a combined approach using surgery and radiation therapy was found to be superior to single modality treatment. However, surgery in conjugation with radiation therapy has been associated with significant toxicities compared with the radiation therapy alone.13Therefore, the use of radiation therapy without surgery is becoming more common with increasingly sophisticated radiation therapy techniques and organ preservation approach in patients with squamous cell carcinoma of the tonsil.

Findings from several studies have shown that in stage I or II oropharyngeal cancer, single modality treatment with radiation therapy achieves 80%-90% of local control of the disease, but poorer outcomes are reported for locally advanced stages III/IV with a local control rate of 63%-74%.20 These findings and others have led to a shift to evaluate the clinical benefits of radiation therapy given with concurrent chemotherapy for the primary treatment of advanced stage oropharyngeal squamous cell carcinoma.20,21 Findings from a number of studies have since reported comparable efficacy and toxicity outcomes using this regimen with concurrent chemotherapy in patients with locally advanced head and neck squamous cell cancer.22-24 Synchronous carboplatin chemotherapy was used effectively as an alternative to cisplatin with fewer potential adverse effects in the good prognosis group of patients with oropharyngeal squamous cell carcinoma.25,26 For our 3 patients, we used carboplatin-based chemotherapy with concurrent advanced hyperfractionated radiation therapy techniques to successfully manage tonsillar squamous cell carcinoma and reduce renal toxicity and neuropathy.

Advanced radiation therapy techniques such as IMRT-IGRT are used routinely at the University Cancer and Diagnostic Centers in Houston, Texas, to manage a range of malignant cancers.27 These innovative techniques have the potential to deliver highly conformal dose-intense radiation to targeted regions of disease, while sparing adjacent critical nonmalignant tissue. The improved shaping of high-dose distributions with IMRT-IGRT could mitigate treatment-related toxicities. For example, the use of advanced radiation therapy techniques has been associated with increased preservation of parotid salivary flow.28-30 The use of advanced radiation therapy techniques in head and neck squamous cell carcinoma is growing, and early evidence confirms its ability to secure excellent local and regional disease control.31,32 In this study, we have demonstrated that by using hyperfractionated conformal three-dimensional IMRT-IGRT we were able not only to manage advanced tonsillar squamous cell carcinoma and treat the malignant metastasis, but also spare adjacent critical organs that were not involved in the disease, thus reducing many of the detrimental side effects associated with hyperfractionated chemoradiation.

All 3 patients were followed for between 40 and 46 months. They continue to do extremely well without local recurrence of their disease, indicating a 100% disease control and overall survival rate. The disease control and survival outcomes for our patients with stage IVA disease compare favorably to other published reports in the literature.33,34 Findings from a study by Prestwich and colleagues33 of 41 patients with stage IV tonsillar carcinoma showed that the radiation therapy with concurrent chemotherapy achieved local and regional disease control in 91% of complete responders and an overall survival rate of 66% at 3 years. Similarly, Setton and colleagues34 reported on 442 patients – 50% with tonsillar cancer, 46% with base-of-tongue cancer – who underwent IMRT and concurrent chemotherapy and who achieved a 3-year overall survival of 84.9%. Our study findings demonstrate that hyperfractionated conformal three-dimensional IMRT-IGRT with concurrent chemotherapy can be delivered safely and effectively to patients with advanced tonsillar squamous cell carcinoma.
 

 

 

Acknowledgment

The authors thank Ms June Lyliston, LVN, for editing and proofreading the manuscript.

References

1. Stambuk HE, Karimi S, Lee N, Patel SG. Oral cavity and oropharynx tumors. Radiol Clin North Am. 2007;45(1):1-20.

2. Lin DT, Cohen SM, Coppit GL, Burkey BB. Squamous cell carcinoma of the oropharynx and hypopharynx. Otolaryngol Clin North Am. 2005;38(1):59-74, viii.

3. Golas SM. Trends in palatine tonsillar cancer incidence and mortality rates in the United States. Community Dent Oral Epidemiol. 2007;35(2):98-108.

4. Cook MB, Dawsey SM, Freedman ND, et al. Sex disparities in cancer incidence by period and age. Cancer Epidemiol Biomarkers Prev. 2009;18(4):1174-1182.

5. Enomoto LM, Bann DV, Hollenbeak CS, Goldenberg D. Trends in the Incidence of oropharyngeal cancers in the United States. Otolaryngol Head Neck Surg. 2016.

6. Shiboski CH, Schmidt BL, Jordan RC. Tongue and tonsil carcinoma: increasing trends in the U.S. population ages 20-44 years. Cancer. 2005;103(9):1843-1849.

7. Marur S, Forastiere AA. Head and neck cancer: changing epidemiology, diagnosis, and treatment. Mayo Clin Proc. 2008;83(4):489-501.

8. Hong AM, Martin A, Chatfield M, et al. Human papillomavirus, smoking status and outcomes in tonsillar squamous cell carcinoma. Int J Cancer. 2013;132(12):2748-2754.

9. Velly AM, Franco EL, Schlecht N, et al. Relationship between dental factors and risk of upper aerodigestive tract cancer. Oral Oncol. 1998;34(4):284-291.

10. Farrow DC, Vaughan TL, Berwick M, et al. Diet and nasopharyngeal cancer in a low-risk population. Int J Cancer. 1998;78(6):675-679.

11. Freedman ND, Park Y, Subar AF, et al. Fruit and vegetable intake and head and neck cancer risk in a large United States prospective cohort study. Int J Cancer. 2008;122(10):2330-2336.

12. Guay ME, Lavertu P. Tonsillar carcinoma. Eur Arch Otorhinolaryngol. 1995;252(5):259-264.

13. Parsons JT, Mendenhall WM, Stringer SP, et al. Squamous cell carcinoma of the oropharynx: surgery, radiation therapy, or both. Cancer. 2002;94(11):2967-2980.

14. Yao M, Dornfeld KJ, Buatti JM, et al. Intensity-modulated radiation treatment for head-and-neck squamous cell carcinoma--the University of Iowa experience. Int J Radiat Oncol Biol Phys. 2005;63(2):410-421.

15. Yang ES, Murphy BM, Chung CH, et al. Evolution of clinical trials in head and neck cancer. Crit Rev Oncol Hematol. 2009;71(1):29-42.

16. Beitler JJ, Zhang Q, Fu KK, et al. Final results of local-regional control and late toxicity of RTOG 9003: a randomized trial of altered fractionation radiation for locally advanced head and neck cancer. Int J Radiat Oncol Biol Phys. 2014;89(1):13-20.

17. Tomkiewicz C, Hans S, Mucchielli MH, et al. A head and neck cancer tumor response-specific gene signature for cisplatin, 5-fluorouracil induction chemotherapy fails with added taxanes. PLoS One. 2012;7(10):e47170.

18. Feldman R, Gatalica Z, Knezetic J, et al. Molecular profiling of head and neck squamous cell carcinoma. Head Neck. 2016;38 Suppl 1:E1625-1638.

19. Moose BD, Kelly MD, Levine PA, et al. Definitive radiotherapy for T1 and T2 squamous cell carcinoma of the tonsil. Head Neck. 1995;17(4):334-338.

20. Chen AY, Schrag N, Hao Y, Stewart A, Ward E. Changes in treatment of advanced oropharyngeal cancer, 1985-2001. Laryngoscope. 2007;117(1):16-21.

21. Machtay M, Rosenthal DI, Hershock D, et al. Organ preservation therapy using induction plus concurrent chemoradiation for advanced resectable oropharyngeal carcinoma: a University of Pennsylvania Phase II Trial. J Clin Oncol. 2002;20(19):3964-3971.

22. Jegannathen A, Swindell R, Yap B, et al. Can synchronous chemotherapy be added to accelerated hypofractionated radiotherapy in patients with base of tongue cancer? Clin Oncol (R Coll Radiol). 2010;22(3):185-191.

23. Budach V, Becker ET, Boehmer D, et al. Concurrent hyperfractionated accelerated radiotherapy with 5-FU and once weekly cisplatin in locally advanced head and neck cancer. The 10-year results of a prospective phase II trial. Strahlenther Onkol. 2014;190(3):250-255.

24. Tobias JS, Monson K, Gupta N, et al. Chemoradiotherapy for locally advanced head and neck cancer: 10-year follow-up of the UK Head and Neck (UKHAN1) trial. Lancet Oncol. 2010;11(1):66-74.

25. Wilkins AC, Rosenfelder N, Schick U, et al. Equivalence of cisplatin and carboplatin-based chemoradiation for locally advanced squamous cell carcinoma of the head and neck: a matched-pair analysis. Oral Oncol. 2013;49(6):615-619.

26. Benghiat H, Sanghera P, Cashmore1 J, et al. Four week hypofractionated accelerated intensity modulated radiotherapy and synchronous carboplatin or cetuximab in biologically staged oropharyngeal carcinoma. Cancer and Clinical Oncology. 2014;3:1-9.

27. D’Andrea MA, Reddy GK. Management of metastatic malignant thymoma with advanced radiation and chemotherapy techniques: report of a rare case. World J Surg Oncol. 2015;13:77.

28. Little M, Schipper M, Feng FY, et al. Reducing xerostomia after chemo-IMRT for head-and-neck cancer: beyond sparing the parotid glands. Int J Radiat Oncol Biol Phys. 2012;83(3):1007-1014.

29. Eisbruch A. Reducing xerostomia by IMRT: what may, and may not, be achieved. J Clin Oncol. 2007;25(31):4863-4864.

30. Pow EH, Kwong DL, McMillan AS, et al. Xerostomia and quality of life after intensity-modulated radiotherapy vs. conventional radiotherapy for early-stage nasopharyngeal carcinoma: initial report on a randomized controlled clinical trial. Int J Radiat Oncol Biol Phys. 2006;66(4):981-991.

31. Lee NY, de Arruda FF, Puri DR, et al. A comparison of intensity-modulated radiation therapy and concomitant boost radiotherapy in the setting of concurrent chemotherapy for locally advanced oropharyngeal carcinoma. Int J Radiat Oncol Biol Phys. 2006;66(4):966-974.

32. Daly ME, Lieskovsky Y, Pawlicki T, et al. Evaluation of patterns of failure and subjective salivary function in patients treated with intensity modulated radiotherapy for head and neck squamous cell carcinoma. Head Neck. 2007;29(3):211-220.

33. Prestwich RJ, Kancherla K, Oksuz DC, et al. A single centre experience with sequential and concomitant chemoradiotherapy in locally advanced stage IV tonsillar cancer. Radiat Oncol. 2010;5:121.

34. Setton J, Caria N, Romanyshyn J, et al. Intensity-modulated radiotherapy in the treatment of oropharyngeal cancer: an update of the Memorial Sloan-Kettering Cancer Center experience. Int J Radiat Oncol Biol Phys. 2012;82(1):291-298.

References

1. Stambuk HE, Karimi S, Lee N, Patel SG. Oral cavity and oropharynx tumors. Radiol Clin North Am. 2007;45(1):1-20.

2. Lin DT, Cohen SM, Coppit GL, Burkey BB. Squamous cell carcinoma of the oropharynx and hypopharynx. Otolaryngol Clin North Am. 2005;38(1):59-74, viii.

3. Golas SM. Trends in palatine tonsillar cancer incidence and mortality rates in the United States. Community Dent Oral Epidemiol. 2007;35(2):98-108.

4. Cook MB, Dawsey SM, Freedman ND, et al. Sex disparities in cancer incidence by period and age. Cancer Epidemiol Biomarkers Prev. 2009;18(4):1174-1182.

5. Enomoto LM, Bann DV, Hollenbeak CS, Goldenberg D. Trends in the Incidence of oropharyngeal cancers in the United States. Otolaryngol Head Neck Surg. 2016.

6. Shiboski CH, Schmidt BL, Jordan RC. Tongue and tonsil carcinoma: increasing trends in the U.S. population ages 20-44 years. Cancer. 2005;103(9):1843-1849.

7. Marur S, Forastiere AA. Head and neck cancer: changing epidemiology, diagnosis, and treatment. Mayo Clin Proc. 2008;83(4):489-501.

8. Hong AM, Martin A, Chatfield M, et al. Human papillomavirus, smoking status and outcomes in tonsillar squamous cell carcinoma. Int J Cancer. 2013;132(12):2748-2754.

9. Velly AM, Franco EL, Schlecht N, et al. Relationship between dental factors and risk of upper aerodigestive tract cancer. Oral Oncol. 1998;34(4):284-291.

10. Farrow DC, Vaughan TL, Berwick M, et al. Diet and nasopharyngeal cancer in a low-risk population. Int J Cancer. 1998;78(6):675-679.

11. Freedman ND, Park Y, Subar AF, et al. Fruit and vegetable intake and head and neck cancer risk in a large United States prospective cohort study. Int J Cancer. 2008;122(10):2330-2336.

12. Guay ME, Lavertu P. Tonsillar carcinoma. Eur Arch Otorhinolaryngol. 1995;252(5):259-264.

13. Parsons JT, Mendenhall WM, Stringer SP, et al. Squamous cell carcinoma of the oropharynx: surgery, radiation therapy, or both. Cancer. 2002;94(11):2967-2980.

14. Yao M, Dornfeld KJ, Buatti JM, et al. Intensity-modulated radiation treatment for head-and-neck squamous cell carcinoma--the University of Iowa experience. Int J Radiat Oncol Biol Phys. 2005;63(2):410-421.

15. Yang ES, Murphy BM, Chung CH, et al. Evolution of clinical trials in head and neck cancer. Crit Rev Oncol Hematol. 2009;71(1):29-42.

16. Beitler JJ, Zhang Q, Fu KK, et al. Final results of local-regional control and late toxicity of RTOG 9003: a randomized trial of altered fractionation radiation for locally advanced head and neck cancer. Int J Radiat Oncol Biol Phys. 2014;89(1):13-20.

17. Tomkiewicz C, Hans S, Mucchielli MH, et al. A head and neck cancer tumor response-specific gene signature for cisplatin, 5-fluorouracil induction chemotherapy fails with added taxanes. PLoS One. 2012;7(10):e47170.

18. Feldman R, Gatalica Z, Knezetic J, et al. Molecular profiling of head and neck squamous cell carcinoma. Head Neck. 2016;38 Suppl 1:E1625-1638.

19. Moose BD, Kelly MD, Levine PA, et al. Definitive radiotherapy for T1 and T2 squamous cell carcinoma of the tonsil. Head Neck. 1995;17(4):334-338.

20. Chen AY, Schrag N, Hao Y, Stewart A, Ward E. Changes in treatment of advanced oropharyngeal cancer, 1985-2001. Laryngoscope. 2007;117(1):16-21.

21. Machtay M, Rosenthal DI, Hershock D, et al. Organ preservation therapy using induction plus concurrent chemoradiation for advanced resectable oropharyngeal carcinoma: a University of Pennsylvania Phase II Trial. J Clin Oncol. 2002;20(19):3964-3971.

22. Jegannathen A, Swindell R, Yap B, et al. Can synchronous chemotherapy be added to accelerated hypofractionated radiotherapy in patients with base of tongue cancer? Clin Oncol (R Coll Radiol). 2010;22(3):185-191.

23. Budach V, Becker ET, Boehmer D, et al. Concurrent hyperfractionated accelerated radiotherapy with 5-FU and once weekly cisplatin in locally advanced head and neck cancer. The 10-year results of a prospective phase II trial. Strahlenther Onkol. 2014;190(3):250-255.

24. Tobias JS, Monson K, Gupta N, et al. Chemoradiotherapy for locally advanced head and neck cancer: 10-year follow-up of the UK Head and Neck (UKHAN1) trial. Lancet Oncol. 2010;11(1):66-74.

25. Wilkins AC, Rosenfelder N, Schick U, et al. Equivalence of cisplatin and carboplatin-based chemoradiation for locally advanced squamous cell carcinoma of the head and neck: a matched-pair analysis. Oral Oncol. 2013;49(6):615-619.

26. Benghiat H, Sanghera P, Cashmore1 J, et al. Four week hypofractionated accelerated intensity modulated radiotherapy and synchronous carboplatin or cetuximab in biologically staged oropharyngeal carcinoma. Cancer and Clinical Oncology. 2014;3:1-9.

27. D’Andrea MA, Reddy GK. Management of metastatic malignant thymoma with advanced radiation and chemotherapy techniques: report of a rare case. World J Surg Oncol. 2015;13:77.

28. Little M, Schipper M, Feng FY, et al. Reducing xerostomia after chemo-IMRT for head-and-neck cancer: beyond sparing the parotid glands. Int J Radiat Oncol Biol Phys. 2012;83(3):1007-1014.

29. Eisbruch A. Reducing xerostomia by IMRT: what may, and may not, be achieved. J Clin Oncol. 2007;25(31):4863-4864.

30. Pow EH, Kwong DL, McMillan AS, et al. Xerostomia and quality of life after intensity-modulated radiotherapy vs. conventional radiotherapy for early-stage nasopharyngeal carcinoma: initial report on a randomized controlled clinical trial. Int J Radiat Oncol Biol Phys. 2006;66(4):981-991.

31. Lee NY, de Arruda FF, Puri DR, et al. A comparison of intensity-modulated radiation therapy and concomitant boost radiotherapy in the setting of concurrent chemotherapy for locally advanced oropharyngeal carcinoma. Int J Radiat Oncol Biol Phys. 2006;66(4):966-974.

32. Daly ME, Lieskovsky Y, Pawlicki T, et al. Evaluation of patterns of failure and subjective salivary function in patients treated with intensity modulated radiotherapy for head and neck squamous cell carcinoma. Head Neck. 2007;29(3):211-220.

33. Prestwich RJ, Kancherla K, Oksuz DC, et al. A single centre experience with sequential and concomitant chemoradiotherapy in locally advanced stage IV tonsillar cancer. Radiat Oncol. 2010;5:121.

34. Setton J, Caria N, Romanyshyn J, et al. Intensity-modulated radiotherapy in the treatment of oropharyngeal cancer: an update of the Memorial Sloan-Kettering Cancer Center experience. Int J Radiat Oncol Biol Phys. 2012;82(1):291-298.

Publications
Publications
Topics
Article Type
Sections
Citation Override
JCSO 2017;15(5):e268-e273
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Article PDF Media

AGA Clinical Practice Update: Treatment of fecal incontinence and defecatory disorders

Article Type
Changed
Fri, 01/18/2019 - 17:09

 

About 25% of patients with fecal incontinence benefit from conservative treatments, which merit a “rigorous trial” before considering surgery, experts write in a Clinical Practice Update in the December issue of Clinical Gastroenterology and Hepatology (doi: 10.1016/j.cgh.2017.08.023).

“A stepwise approach should be followed for management of fecal incontinence. In our experience, many incontinent patients who are considered refractory to conservative therapy have not received an optimal trial of conservative therapy,” states Adil E. Bharucha, MBBS, MD, of the Mayo Clinic and the Mayo Foundation in Rochester, Minn., and his associates.

Fecal incontinence affects 7%-15% of individuals and has potentially “devastating” implications for quality of life, the experts note. They recommend starting treatment by meticulously documenting bowel habits, triggers of incontinence, and treatment history. For fecal incontinence with diarrhea, they suggest eliminating caffeine and poorly absorbed dietary sugars, such as sorbitol and fructose, and adding loperamide, starting with one 2-mg tablet taken 30 minutes before breakfast and titrating up to a maximum of 16 mg per day. Other conservative therapeutic options for diarrhea include fiber supplementation, scheduled toileting, a bowel retraining program, anticholinergic agents, clonidine, and cholestyramine or colesevelam to correct bile salt malabsorption. Patients whose fecal incontinence involves constipation should start with laxatives and anorectal testing for evacuation disorders. Rectal cleansing with a small enema or tap water can help prevent stool leakage, the experts write.

If these conservative measures fail to improve fecal incontinence, they recommend anorectal manometry to test for anal weakness, reduced or increased rectal sensation, and impaired rectal balloon expulsion, all of which can improve with biofeedback therapy to retrain the pelvic floor. If biofeedback fails, consider perianal bulking agents, such as intra-anal injection of dextranomer, the experts suggest. Sacral nerve stimulation might be indicated if moderate or severe fecal incontinence does not respond to at least 3 months of conservative treatment. However, the experts do not recommend percutaneous tibial nerve stimulation, which failed to outperform sham stimulation in a 12-week, double-blind, multicenter trial (Lancet. 2015;386:1640-8). Surgery is indicated for fecal incontinence associated with major anatomic defects, such as rectovaginal fistula, full-thickness rectal prolapse, fistula in ano, or cloaca-like deformity. Additionally, sphincteroplasty is an option for postpartum women with fecal incontinence, patients with recent sphincter injuries, and patients with sphincter damage and fecal incontinence fecal incontinence that fails to improve with conservative and biofeedback therapy, perianal bulking injection, and sacral nerve stimulation, according to the clinical practice update.

Barrier devices should be offered if fecal incontinence fails conservative treatments and surgery, or if surgery is not an option. Most anal plugs are “poorly tolerated,” with two exceptions – a Food and Drug Administration–approved device from Renew Medical and a vaginal insert and pressure-regulated pump from Pelvalon. Colostomy might be indicated if patients with severe fecal incontinence fail conservative treatment and or are not candidates for barrier devices, minimally invasive surgeries, and sphincteroplasty.

If severe fecal incontinence that is refractory to or contraindicated for all these interventions, the experts suggest considering artificial anal sphincter repair by dynamic graciloplasty. Surgery also is indicated to repair major anatomic defects such as rectovaginal fistula, full-thickness rectal prolapse, fistula in ano, or cloaca-like deformity, they noted. A magnetic anal sphincter device is a possibility for patients with medically refractory severe fecal incontinence who have failed or are not candidates for barrier devices, perianal bulking injection, sacral nerve stimulation, sphincteroplasty, or a colostomy. However, the study that led to FDA approval of a magnetic anal sphincter device included only 35 patients, and 7 (20%) had the device removed because of infection, erosion, or inefficacy. Another patient required a stoma in order to be able to defecate, and a total of 40% had moderate or severe complications when pain and bleeding were also considered, the experts noted.

Biofeedback is the preferred treatment for defecatory disorders – that is, chronic constipation or constipation-predominant irritable bowel syndrome with impaired rectal evacuation, according to the clinical practice update. The experts recommend against sacral nerve stimulation, anteretrograde colonic enemas, and stapled transanal rectal resection for patients with defecatory disorders. Surgical treatment typically is reserved for the small minority of patients with considerable pelvic organ or rectal prolapse, they note.

The National Institutes of Health Sciences provided funding. The authors reported having no conflicts of interest.

Publications
Topics
Sections

 

About 25% of patients with fecal incontinence benefit from conservative treatments, which merit a “rigorous trial” before considering surgery, experts write in a Clinical Practice Update in the December issue of Clinical Gastroenterology and Hepatology (doi: 10.1016/j.cgh.2017.08.023).

“A stepwise approach should be followed for management of fecal incontinence. In our experience, many incontinent patients who are considered refractory to conservative therapy have not received an optimal trial of conservative therapy,” states Adil E. Bharucha, MBBS, MD, of the Mayo Clinic and the Mayo Foundation in Rochester, Minn., and his associates.

Fecal incontinence affects 7%-15% of individuals and has potentially “devastating” implications for quality of life, the experts note. They recommend starting treatment by meticulously documenting bowel habits, triggers of incontinence, and treatment history. For fecal incontinence with diarrhea, they suggest eliminating caffeine and poorly absorbed dietary sugars, such as sorbitol and fructose, and adding loperamide, starting with one 2-mg tablet taken 30 minutes before breakfast and titrating up to a maximum of 16 mg per day. Other conservative therapeutic options for diarrhea include fiber supplementation, scheduled toileting, a bowel retraining program, anticholinergic agents, clonidine, and cholestyramine or colesevelam to correct bile salt malabsorption. Patients whose fecal incontinence involves constipation should start with laxatives and anorectal testing for evacuation disorders. Rectal cleansing with a small enema or tap water can help prevent stool leakage, the experts write.

If these conservative measures fail to improve fecal incontinence, they recommend anorectal manometry to test for anal weakness, reduced or increased rectal sensation, and impaired rectal balloon expulsion, all of which can improve with biofeedback therapy to retrain the pelvic floor. If biofeedback fails, consider perianal bulking agents, such as intra-anal injection of dextranomer, the experts suggest. Sacral nerve stimulation might be indicated if moderate or severe fecal incontinence does not respond to at least 3 months of conservative treatment. However, the experts do not recommend percutaneous tibial nerve stimulation, which failed to outperform sham stimulation in a 12-week, double-blind, multicenter trial (Lancet. 2015;386:1640-8). Surgery is indicated for fecal incontinence associated with major anatomic defects, such as rectovaginal fistula, full-thickness rectal prolapse, fistula in ano, or cloaca-like deformity. Additionally, sphincteroplasty is an option for postpartum women with fecal incontinence, patients with recent sphincter injuries, and patients with sphincter damage and fecal incontinence fecal incontinence that fails to improve with conservative and biofeedback therapy, perianal bulking injection, and sacral nerve stimulation, according to the clinical practice update.

Barrier devices should be offered if fecal incontinence fails conservative treatments and surgery, or if surgery is not an option. Most anal plugs are “poorly tolerated,” with two exceptions – a Food and Drug Administration–approved device from Renew Medical and a vaginal insert and pressure-regulated pump from Pelvalon. Colostomy might be indicated if patients with severe fecal incontinence fail conservative treatment and or are not candidates for barrier devices, minimally invasive surgeries, and sphincteroplasty.

If severe fecal incontinence that is refractory to or contraindicated for all these interventions, the experts suggest considering artificial anal sphincter repair by dynamic graciloplasty. Surgery also is indicated to repair major anatomic defects such as rectovaginal fistula, full-thickness rectal prolapse, fistula in ano, or cloaca-like deformity, they noted. A magnetic anal sphincter device is a possibility for patients with medically refractory severe fecal incontinence who have failed or are not candidates for barrier devices, perianal bulking injection, sacral nerve stimulation, sphincteroplasty, or a colostomy. However, the study that led to FDA approval of a magnetic anal sphincter device included only 35 patients, and 7 (20%) had the device removed because of infection, erosion, or inefficacy. Another patient required a stoma in order to be able to defecate, and a total of 40% had moderate or severe complications when pain and bleeding were also considered, the experts noted.

Biofeedback is the preferred treatment for defecatory disorders – that is, chronic constipation or constipation-predominant irritable bowel syndrome with impaired rectal evacuation, according to the clinical practice update. The experts recommend against sacral nerve stimulation, anteretrograde colonic enemas, and stapled transanal rectal resection for patients with defecatory disorders. Surgical treatment typically is reserved for the small minority of patients with considerable pelvic organ or rectal prolapse, they note.

The National Institutes of Health Sciences provided funding. The authors reported having no conflicts of interest.

 

About 25% of patients with fecal incontinence benefit from conservative treatments, which merit a “rigorous trial” before considering surgery, experts write in a Clinical Practice Update in the December issue of Clinical Gastroenterology and Hepatology (doi: 10.1016/j.cgh.2017.08.023).

“A stepwise approach should be followed for management of fecal incontinence. In our experience, many incontinent patients who are considered refractory to conservative therapy have not received an optimal trial of conservative therapy,” states Adil E. Bharucha, MBBS, MD, of the Mayo Clinic and the Mayo Foundation in Rochester, Minn., and his associates.

Fecal incontinence affects 7%-15% of individuals and has potentially “devastating” implications for quality of life, the experts note. They recommend starting treatment by meticulously documenting bowel habits, triggers of incontinence, and treatment history. For fecal incontinence with diarrhea, they suggest eliminating caffeine and poorly absorbed dietary sugars, such as sorbitol and fructose, and adding loperamide, starting with one 2-mg tablet taken 30 minutes before breakfast and titrating up to a maximum of 16 mg per day. Other conservative therapeutic options for diarrhea include fiber supplementation, scheduled toileting, a bowel retraining program, anticholinergic agents, clonidine, and cholestyramine or colesevelam to correct bile salt malabsorption. Patients whose fecal incontinence involves constipation should start with laxatives and anorectal testing for evacuation disorders. Rectal cleansing with a small enema or tap water can help prevent stool leakage, the experts write.

If these conservative measures fail to improve fecal incontinence, they recommend anorectal manometry to test for anal weakness, reduced or increased rectal sensation, and impaired rectal balloon expulsion, all of which can improve with biofeedback therapy to retrain the pelvic floor. If biofeedback fails, consider perianal bulking agents, such as intra-anal injection of dextranomer, the experts suggest. Sacral nerve stimulation might be indicated if moderate or severe fecal incontinence does not respond to at least 3 months of conservative treatment. However, the experts do not recommend percutaneous tibial nerve stimulation, which failed to outperform sham stimulation in a 12-week, double-blind, multicenter trial (Lancet. 2015;386:1640-8). Surgery is indicated for fecal incontinence associated with major anatomic defects, such as rectovaginal fistula, full-thickness rectal prolapse, fistula in ano, or cloaca-like deformity. Additionally, sphincteroplasty is an option for postpartum women with fecal incontinence, patients with recent sphincter injuries, and patients with sphincter damage and fecal incontinence fecal incontinence that fails to improve with conservative and biofeedback therapy, perianal bulking injection, and sacral nerve stimulation, according to the clinical practice update.

Barrier devices should be offered if fecal incontinence fails conservative treatments and surgery, or if surgery is not an option. Most anal plugs are “poorly tolerated,” with two exceptions – a Food and Drug Administration–approved device from Renew Medical and a vaginal insert and pressure-regulated pump from Pelvalon. Colostomy might be indicated if patients with severe fecal incontinence fail conservative treatment and or are not candidates for barrier devices, minimally invasive surgeries, and sphincteroplasty.

If severe fecal incontinence that is refractory to or contraindicated for all these interventions, the experts suggest considering artificial anal sphincter repair by dynamic graciloplasty. Surgery also is indicated to repair major anatomic defects such as rectovaginal fistula, full-thickness rectal prolapse, fistula in ano, or cloaca-like deformity, they noted. A magnetic anal sphincter device is a possibility for patients with medically refractory severe fecal incontinence who have failed or are not candidates for barrier devices, perianal bulking injection, sacral nerve stimulation, sphincteroplasty, or a colostomy. However, the study that led to FDA approval of a magnetic anal sphincter device included only 35 patients, and 7 (20%) had the device removed because of infection, erosion, or inefficacy. Another patient required a stoma in order to be able to defecate, and a total of 40% had moderate or severe complications when pain and bleeding were also considered, the experts noted.

Biofeedback is the preferred treatment for defecatory disorders – that is, chronic constipation or constipation-predominant irritable bowel syndrome with impaired rectal evacuation, according to the clinical practice update. The experts recommend against sacral nerve stimulation, anteretrograde colonic enemas, and stapled transanal rectal resection for patients with defecatory disorders. Surgical treatment typically is reserved for the small minority of patients with considerable pelvic organ or rectal prolapse, they note.

The National Institutes of Health Sciences provided funding. The authors reported having no conflicts of interest.

Publications
Publications
Topics
Article Type
Click for Credit Status
Ready
Sections
Article Source

FROM CLINICAL GASTROENTEROLOGY AND HEPATOLOGY

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default

A concise guide to monoamine oxidase inhibitors

Article Type
Changed
Tue, 12/11/2018 - 15:01
Display Headline
A concise guide to monoamine oxidase inhibitors
 

Despite an abundance of evidenced-based literature supporting monoamine oxidase inhibitors (MAOIs) as an effective treatment for depression, use of these agents has decreased drastically in the past 3 decades. A lack of industry support and the ease of use of other agents are contributing factors, but the biggest impediments to routine use of MAOIs are unfamiliarity with their efficacy advantages and concerns about adverse effects, particularly the risk of hypertensive crises and serotonin syndrome. Many misconceptions regarding these medications are based on outdated data and studies that are no longer reliable.

The goal of this 2-part review is to provide clinicians with updated information regarding MAOIs. Part 1 provides a brief description of:

  • the pharmacology of nonselective irreversible MAOIs
  • the mechanism by which tyramine induces hypertension
  • sources of clinically significant tyramine exposure
  • what to tell patients about dietary restrictions and MAOIs.

Part 2 of this guide will cover the risk of serotonin syndrome when MAOIs are combined with inhibitors of serotonin reuptake, how to initiate MAOI therapy, and augmenting MAOIs with other agents.

The pharmacology of MAOIs

First used clinically in the 1950s to treat tuberculosis, MAOIs have a long and interesting history (see the Box “A brief history of monoamine oxidase inhibitors”). Table 11 lists MAOIs currently available in the United States, including the MAO-B–specific agent rasagiline, which is used for Parkinson’s disease.

Manipulation of the monoamines serotonin, norepinephrine, and dopamine is fundamental to managing major depressive disorder (MDD), yet only nonselective MAOIs directly promote neurotransmission of all 3 by inhibiting MAO-A and MAO-B.2 The Sequenced Treatment Alternatives to Relieve Depression (STAR*D) study demonstrated that <50% of MDD patients achieve remission in monotherapy trials of selective serotonin reuptake inhibitors, serotonin norepinephrine reuptake inhibitors, mirtazapine, or bupropion, necessitating consideration of antidepressant combinations, augmentation options, and eventually irreversible, nonselective MAOIs such as phenelzine, tranylcypromine, or isocarboxazid.3,4 Nonselective MAOIs thus offer a therapeutic opportunity for patients who do not respond to single or dual-mechanism strategies; moreover, nonselective MAOIs have compelling effectiveness data for other conditions, including panic disorder and social phobia.5 Although MAOIs are among the most effective pharmacologic agents for MDD,6 they are underutilized because of an inadequate understanding of risk mechanisms and resultant fear of catastrophic outcomes. Because of the difficulties encountered in achieving clinical remission for MDD, the nonselective MAOIs deserve a second look.
 

Differentiation of MAO-A from MAO-B. It is essential to understand the mechanism of action of MAOIs, specifically the impact of MAO-A inhibition. Although the enzyme MAO was known in the 1950s, it wasn’t until 1968 that Johnston7 postulated the existence of >1 form. In 1971, Goridis and Neff8 used clorgyline to examine the deamination rate by MAO of tyramine and norepinephrine. They found that tyramine appeared to be a substrate of both MAO isoforms, but only 1 of the MAO types was sensitive to the inhibitory effects of clorgyline. They also discerned that norepinephrine was only a substrate for MAO-A, and that this form of MAO was sensitive to clorgyline inhibition. Thus, the forms of MAO were characterized by their preferred substrates (Table 29,10), and then later by their tissue distribution. Phenylethylamine is a naturally occurring compound found in foods, such as chocolate, and has an in vitro pharmacology similar to amphetamine but with 1 important difference: it has a short half-life of 5 to 10 minutes after oral ingestion, and therefore no appreciable CNS impact.

 

 

 

Within the CNS, norepinephrine and dopamine neurons possess both MAO forms, with the MAO-A content greater than MAO-B. Serotonergic neurons only contain MAO-B.11 Outside of the CNS, MAO-A predominates, with only platelets and lymphocytes possessing MAO-B activity.11 The overall relative tissue proportions of MAO-A to MAO-B activity are: brain, 25% MAO-A, 75% MAO-B; liver, 50% MAO-A, 50% MAO-B; intestine, 80% MAO-A, 20% MAO-B; and peripheral adrenergic neurons, 90% MAO-A, 10% MAO-B.

Because of its specificity for serotonin and norepinephrine, CNS MAO-A inhibition is necessary for antidepressant effects. MAO-B inhibition by itself does not appear to raise CNS dopamine levels unless exogenous dopamine is supplied.11 All MAOIs used in the United States to treat depression are irreversible, nonselective inhibitors of MAO-A and MAO-B.

Selegiline in oral form generates low plasma levels and primarily inhibits MAO-B. The transdermal form of selegiline achieves significantly greater systemic exposure, and at these higher plasma levels selegiline is a nonselective, irreversible MAOI effective for MDD (Figure 112). Administering selegiline systemically via a transdermal patch avoids clinically significant MAOI effects in the gut, so no dietary warnings exist for the lowest dose (6 mg/24 hours), although there are warnings for the higher dosages (9 mg/24 hours and 12 mg/24 hours).


Differentiation of MAOIs by chemical class. The earliest MAOI, iproniazid, was a hydrazine derivative and exhibited hepatotoxicity,13 as did certain other hydrazine MAOIs. This lead to a search for safer hydrazine and nonhydrazine alternatives. Isocarboxazid and phenelzine are the 2 hydrazine MAOIs available in the United States, while tranylcypromine and selegiline transdermal are nonhydrazines (Figure 2).


What distinguishes the nonhydrazine medication selegiline is that its metabolism generates L-amphetamine metabolites (Figure 314). This property was thought to be shared by other non­hydrazines, but recent studies indicate than neither tranylcypromine15 nor the MAO-B–selective rasagiline possess amphetamine metabolites.16 Unlike the dextro isomers, L-amphetamine structures do not inhibit dopamine reuptake or cause euphoria, but can cause stimulation (eg, sleep disturbance) by inhibiting norepinephrine reuptake, and also by interacting with the trace amine-associated receptor 1 (TAAR1), an intracellular receptor expressed within the presynaptic terminal of monoamine neurons. Activation of TAAR1 by tyramine is an important part of the hypertensive effects related to excessive tyramine exposure.17 (The importance of TAAR1 and the interaction with tyramine is discussed in the next section.) Importantly, patients taking selegiline must be warned that certain drug screens may not discriminate between levo and dextro isomers of amphetamines, and that the use of selegiline should be disclosed prior to drug testing procedures.

MAOIs and tyramine: Dietary requirements

Clinicians who are familiar with MAOIs recognize that there are dietary restrictions to minimize patients’ exposure to tyramine. As most clinicians know, significant tyramine ingestion may cause an increase in blood pressure (BP) in patients taking an MAOI, but many overestimate the prevalence of foods high in tyramine content since the original reports emerged in the early 1960s.18 In a recent monograph, one of the leading experts on MAOIs, Professor Ken Gillman, stated:

Very few foods now contain problematically high tyramine levels, that is a result of great changes in international food production methods and hygiene regulations. Cheese is the only food that, in the past, has been associated with documented fatalities resulting from hypertension. Nowadays most cheeses are quite safe, and even ‘matured’ cheeses are usually safe in healthy-sized portions. The variability of sensitivity to tyramine between individuals, and the sometimes unpredictable amount of tyramine content in foods, means a little knowledge and care are still required.19

 

 

 

What is tyramine? Tyramine is a biogenic amine that is virtually absent in fresh animal protein sources but is enriched after decay or fermentation.20 Modern food processing and handling methods have significantly limited the tyramine content in processed foods, with the exception of certain cheeses and sauces, as discussed below. Moreover, modern assaying techniques using high-performance liquid chromatography have generated extremely accurate assessments of the tyramine content of specific foods.21 Data published prior to 2000 are not reliable, because many of these publications employed outdated methods.17

When ingested, tyramine is metabolized by gut MAO-A, with doses up to 400 mg causing no known effects, although most people rarely ingest >25 mg during a meal.22 In addition to being a substrate for MAO-A, tyramine is also a substrate for the dopamine transporter, norepinephrine transporter (NET), the vesicular monoamine transporter 2, and TAAR1.23 Tyramine enters the cell via NET, where it interacts with TAAR1, a G protein-coupled receptor that is responsive to trace amines, such as tyramine, as well as amphetamines.20 The agonist properties at TAAR1 are the presumed site of action for the BP effects of tyramine, because binding results in potent release of norepinephrine.20,24 When tyramine is supplied to an animal in which MAO-A is inhibited, the decreased peripheral catabolism of tyramine results in markedly increased norepinephrine release by peripheral adrenergic neurons. Moreover, the absence of MAO-A activity in those neurons prevents any norepinephrine breakdown, resulting in robust synaptic norepinephrine delivery and peripheral effects.

All orally administered irreversible MAOIs potently inhibit gut and systemic MAO-A, and are susceptible to the impact of significant tyramine ingestion. The exception is selegiline transdermal (Figure 112), as appreciable gut MAO-A inhibition does not occur until doses >6 mg/24 hours are reached.22 No significant pressor response was seen in participants taking selegiline transdermal, 6 mg/24 hours for 13 days, who consumed a meal that provided 400 mg of tyramine.22 Conversely, for oral agents that produce gut MAO-A inhibition, tyramine doses as low as 8 to 10 mg (when administered as tyramine capsules) may increase systolic pressure by 30 mm Hg.25 The dietary warnings do not apply to rasagiline, which is a selective MAO-B inhibitor, although rasagiline may have an impact on resting BP; the prescribing information for rasagiline includes warnings about hypotension and hypertension.26

What to tell patients about tyramine. Although administering pure tyramine capsules can induce a measurable change in systolic BP, when ingested as food, tyramine doses <50 mg are unlikely to cause an increase in BP sufficient to warrant clinical intervention, although some individuals can be sensitive to 10 to 25 mg.19 When discussing with patients safety issues related to diet, there are a few important concepts to remember19:

  • In an era when the tyramine content of foods was much higher (1960 to 1964) and MAOI users received no dietary guidance, only 14 deaths were reported among an estimated 1.5 million patients who took MAOIs.
  • MAOIs do not raise BP, and their use is associated with orthostasis in some patients.
  • Routine exercise or other vigorous activities (eg, weightlifting) can raise systolic pressure well above 200 mm Hg, and routine baseline systolic pressures, ranging from 180 to 220 mm Hg, do not increase the risk of subarachnoid hemorrhage.
  • Hospital evaluation is needed only if a substantial amount of tyramine is ingested (eg, estimated ≥100 mg), and self-monitoring shows a systolic BP ≥220 mm Hg over a prolonged period (eg, 2 hours). Ingestion of 100 mg of tyramine would almost certainly have to be intentional, as it would require one to consume 3.5 oz of the most highly tyramine-laden cheeses.
     

Emphasize to patients that only a small number of highly aged cheeses, foods, and sauces contain high quantities of tyramine, and that even these foods can be enjoyed in small amounts. All patients who are prescribed an MAOI also should purchase a portable BP cuff for those rare instances when a dietary indiscretion may have occurred and the person experiences a headache within 1 to 2 hours after tyramine ingestion. Most reactions are self-limited and resolve over 2 to 4 hours.

Patients who ingest ≥100 mg of tyramine should be evaluated by a physician. Under no circumstances should a patient be given a prescription for nifedipine or other medications that can abruptly lower BP, because this may result in complications, including myocardial infarction.27,28 Counsel patients to remain calm. Some clinicians endorse the use of low doses of benzodiazepines (the equivalent of alprazolam 0.5 mg) to facilitate this, because anxiety elevates BP. A recent emergency room study of patients with an initial systolic BP ≥160 mm Hg or diastolic BP ≥100 mm Hg without end organ damage demonstrated that alprazolam, 0.5 mg, was as effective as captopril, 25 mg, in lowering BP.29

Also, tell patients that if a food is unfamiliar and highly aged or fermented, they should avoid it until they can further inquire about it. In a review, Gillman19 provides the tyramine content of an exhaustive list of cheeses, aged meats, and sauces (see Related Resources). For other products, patients often can obtain information directly from the manufacturer. In many parts of the world, assays for tyramine content are required as a demonstration of adequate product safety procedures. Even the most highly aged cheeses with a tyramine content of 1,000 g/kg can be enjoyed in small amounts (<1 oz), and most products would require heroic intake to achieve clinically significant tyramine ingestion (Table 319).

Improved education can clarify the risks

Medications such as lithium, clozapine, and MAOIs have a proven record of efficacy, yet often are underused due to fears engendered by lack of systematic training. A recent initiative in New York thus aimed to increase rates of clozapine prescribing by providing clinicians with an education consultation center.30 Similarly, enhanced education regarding MAOIs could increase the use of these highly effective medications. With a better understanding of MAOIs, clinicians can become adept at using these medications, and therefore expand the armamentarium of agents available to patients with MDD, as well as to those with panic disorder and social phobia.
 

Bottom Line

Monoamine oxidase inhibitors (MAOIs) are among the most effective medications for treating depression but are underutilized because of misunderstanding of risk mechanisms and fear of catastrophic outcomes. Through education, astute clinicians can master the proper use of MAOIs and add these agents to their treatment armamentarium.

Related Resource

  • Gillman PK. Monoamine oxidase inhibitors: a review concerning dietary tyramine and drug interactions. PsychoTropical Commentaries. 2016;16(6):1-97.

Drug Brand Names

Alprazolam • Xanax
Bupropion • Wellbutrin, Zyban
Captopril • Capoten
Clozapine • Clozaril
Imipramine • Tofranil
Iproniazid • Marsilid
Isocarboxazid • Marplan
Lithium • Eskalith, Lithobid
Meperidine • Demerol
Methadone • Dolophine, Methadose
Mirtazapine • Remeron
Nifedipine • Adalat, Procardia
Norepinephrine • Levophed
Phenelzine • Nardil
Rasagiline • Azilect
Selegiline oral • Eldepryl
Selegiline transdermal • Emsam
Tramadol • Ultram
Tranylcypromine • Parnate

References

1. Panisset M, Chen JJ, Rhyee SH, et al. Serotonin toxicity association with concomitant antidepressants and rasagiline treatment: retrospective study (STACCATO). Pharmacotherapy. 2014;34(12):1250-1258.
2. López-Muñoz F, Alamo C. Monoaminergic neuro­transmission: the history of the discovery of antidepressants from 1950s until today. Curr Pharm Des. 2009;15(14):1563-1586.
3. Nierenberg AA, Fava M, Trivedi MH, et al. A comparison of lithium and T(3) augmentation following two failed medication treatments for depression: a STAR*D report. Am J Psychiatry. 2006;163(9):1519-1530; quiz 1665.
4. Trivedi MH, Fava M, Wisniewski SR, et al; STAR*D Study Team. Medication augmentation after the failure of SSRIs for depression. New Engl J Med. 2006;354(12):1243-1252.
5. Bandelow B, Zohar J, Hollander E, et al; World Federation of Societies of Biological Psychiatry Task Force on Treatment Guidelines for Anxiety, Obsessive-Compulsive and Posttraumatic Stress Disorders. World Federation of Societies of Biological Psychiatry (WFSBP) guidelines for the pharmacological treatment of anxiety, obsessive-compulsive and posttraumatic stress disorders. World J Biol Psychiatry. 2002;3(4):171-199.
6. Shulman KI, Herrmann N, Walker SE. Current place of monoamine oxidase inhibitors in the treatment of depression. CNS Drugs. 2013;27(10):789-797.
7. Johnston JP. Some observations upon a new inhibitor of monoamine oxidase in brain tissue. Biochem Pharmacol. 1968;17(7):1285-1297.
8. Goridis C, Neff NH. Monoamine oxidase in sympathetic nerves: a transmitter specific enzyme type. Br J Pharmacol. 1971;43(4):814-818.
9. Geha RM, Rebrin I, Chen K, et al. Substrate and inhibitor specificities for human monoamine oxidase A and B are influenced by a single amino acid. J Biol Chem. 2001;276(13):9877-9882.
10. O’Carroll AM, Fowler CJ, Phillips JP, et al. The deamination of dopamine by human brain monoamine oxidase. Specificity for the two enzyme forms in seven brain regions. Naunyn Schmiedebergs Arch Pharmacol. 1983;322(3):198-202.
11. Stahl SM, Felker A. Monoamine oxidase inhibitors: a modern guide to an unrequited class of antidepressants. CNS Spectr. 2008;13(10):855-780.
12. Mawhinney M, Cole D, Azzaro AJ. Daily transdermal administration of selegiline to guinea-pigs preferentially inhibits monoamine oxidase activity in brain when compared with intestinal and hepatic tissues. J Pharm Pharmacol. 2003;55(1):27-34.
13. Maille F, Duvoux C, Cherqui D, et al. Auxiliary hepatic transplantation in iproniazid-induced subfulminant hepatitis. Should iproniazid still be sold in France? [in French]. Gastroenterol Clin Biol. 1999;23(10):1083-1085.
14. Salonen JS, Nyman L, Boobis AR, et al. Comparative studies on the cytochrome p450-associated metabolism and interaction potential of selegiline between human liver-derived in vitro systems. Drug Metab Dispos. 2003;31(9):1093-1102.
15. Iwersen S, Schmoldt A. One fatal and one nonfatal intoxication with tranylcypromine. Absence of amphetamines as metabolites. J Anal Toxicol. 1996;20(5):301-304.
16. Müller T, Hoffmann JA, Dimpfel W, et al. Switch from selegiline to rasagiline is beneficial in patients with Parkinson’s disease. J Neural Transm (Vienna). 2013;120(5):761-765.
17. Lewin AH, Miller GM, Gilmour B. Trace amine-associated receptor 1 is a stereoselective binding site for compounds in the amphetamine class. Bioorg Med Chem. 2011;19(23):7044-7048.
18. Blackwell B. Hypertensive crisis due to monoamine-oxidase inhibitors. Lancet. 1963;2(7313):849-850.
19. Gillman PK. Monoamine oxidase inhibitors: a review concerning dietary tyramine and drug interactions. PsychoTropical Commentaries. 2016;16(6):1-97.
20. Pei Y, Asif-Malik A, Canales JJ. Trace amines and the trace amine-associated receptor 1: pharmacology, neurochemistry, and clinical implications. Front Neurosci. 2016;10:148.
21. Fiechter G, Sivec G, Mayer HK. Application of UHPLC for the simultaneous analysis of free amino acids and biogenic amines in ripened acid-curd cheeses. J Chromatogr B Analyt Technol Biomed Life Sci. 2013;927:191-200.
22. Blob LF, Sharoky M, Campbell BJ, et al. Effects of a tyramine-enriched meal on blood pressure response in healthy male volunteers treated with selegiline transdermal system 6 mg/24 hour. CNS Spectr. 2007;12(1):25-34.
23. Partilla JS, Dempsey AG, Nagpal AS, et al. Interaction of amphetamines and related compounds at the vesicular monoamine transporter. J Pharmacol Exp Ther. 2006;319(1):237-246.
24. Borowsky B, Adham N, Jones KA, et al. Trace amines: identification of a family of mammalian G protein-coupled receptors. Proc Natl Acad Sci U S A. 2001;98(16):8966-8971.
25. Azzaro AJ, Vandenberg CM, Blob LF, et al. Tyramine pressor sensitivity during treatment with the selegiline transdermal system 6 mg/24 h in healthy subjects. J Clin Pharmacol. 2006;46(8):933-944.
26. Azilect [package insert]. Overland Park, KS: Teva Neuroscience, Inc.; 2014.
27. Marik PE, Varon J. Hypertensive crises: challenges and management. Chest. 2007;131(6):1949-1962.
28. Burton TJ, Wilkinson IB. The dangers of immediate-release nifedipine in the emergency treatment of hypertension. J Hum Hypertens. 2008;22(4):301-302.
29. Yilmaz S, Pekdemir M, Tural U, et al. Comparison of alprazolam versus captopril in high blood pressure: a randomized controlled trial. Blood Press. 2011;20(4):239-243.
30. Carruthers J, Radigan M, Erlich MD, et al. An initiative to improve clozapine prescribing in New York State. Psychiatr Serv. 2016;67(4):369-371.

Article PDF
Author and Disclosure Information

Jonathan M. Meyer, MD
Psychopharmacology Consultant
California Department of State Hospitals
Sacramento, California
Assistant Clinical Professor of Psychiatry
University of California, San Diego
San Diego, California
Deputy Editor, Current Psychiatry

Disclosure
Dr. Meyer is a consultant to Acadia Pharmaceuticals, Neurocrine Biosciences, Inc., and Teva Pharmaceutical Industries; and is a speaker for Acadia Pharmaceuticals, Alkermes, Allergan, Merck, Neurocrine Biosciences, Inc., Otsuka America, Inc., Sunovion Pharmaceuticals, and Teva Pharmaceutical Industries.

Issue
December 2017
Publications
Topics
Page Number
14-16,18-23,47,A
Sections
Author and Disclosure Information

Jonathan M. Meyer, MD
Psychopharmacology Consultant
California Department of State Hospitals
Sacramento, California
Assistant Clinical Professor of Psychiatry
University of California, San Diego
San Diego, California
Deputy Editor, Current Psychiatry

Disclosure
Dr. Meyer is a consultant to Acadia Pharmaceuticals, Neurocrine Biosciences, Inc., and Teva Pharmaceutical Industries; and is a speaker for Acadia Pharmaceuticals, Alkermes, Allergan, Merck, Neurocrine Biosciences, Inc., Otsuka America, Inc., Sunovion Pharmaceuticals, and Teva Pharmaceutical Industries.

Author and Disclosure Information

Jonathan M. Meyer, MD
Psychopharmacology Consultant
California Department of State Hospitals
Sacramento, California
Assistant Clinical Professor of Psychiatry
University of California, San Diego
San Diego, California
Deputy Editor, Current Psychiatry

Disclosure
Dr. Meyer is a consultant to Acadia Pharmaceuticals, Neurocrine Biosciences, Inc., and Teva Pharmaceutical Industries; and is a speaker for Acadia Pharmaceuticals, Alkermes, Allergan, Merck, Neurocrine Biosciences, Inc., Otsuka America, Inc., Sunovion Pharmaceuticals, and Teva Pharmaceutical Industries.

Article PDF
Article PDF
 

Despite an abundance of evidenced-based literature supporting monoamine oxidase inhibitors (MAOIs) as an effective treatment for depression, use of these agents has decreased drastically in the past 3 decades. A lack of industry support and the ease of use of other agents are contributing factors, but the biggest impediments to routine use of MAOIs are unfamiliarity with their efficacy advantages and concerns about adverse effects, particularly the risk of hypertensive crises and serotonin syndrome. Many misconceptions regarding these medications are based on outdated data and studies that are no longer reliable.

The goal of this 2-part review is to provide clinicians with updated information regarding MAOIs. Part 1 provides a brief description of:

  • the pharmacology of nonselective irreversible MAOIs
  • the mechanism by which tyramine induces hypertension
  • sources of clinically significant tyramine exposure
  • what to tell patients about dietary restrictions and MAOIs.

Part 2 of this guide will cover the risk of serotonin syndrome when MAOIs are combined with inhibitors of serotonin reuptake, how to initiate MAOI therapy, and augmenting MAOIs with other agents.

The pharmacology of MAOIs

First used clinically in the 1950s to treat tuberculosis, MAOIs have a long and interesting history (see the Box “A brief history of monoamine oxidase inhibitors”). Table 11 lists MAOIs currently available in the United States, including the MAO-B–specific agent rasagiline, which is used for Parkinson’s disease.

Manipulation of the monoamines serotonin, norepinephrine, and dopamine is fundamental to managing major depressive disorder (MDD), yet only nonselective MAOIs directly promote neurotransmission of all 3 by inhibiting MAO-A and MAO-B.2 The Sequenced Treatment Alternatives to Relieve Depression (STAR*D) study demonstrated that <50% of MDD patients achieve remission in monotherapy trials of selective serotonin reuptake inhibitors, serotonin norepinephrine reuptake inhibitors, mirtazapine, or bupropion, necessitating consideration of antidepressant combinations, augmentation options, and eventually irreversible, nonselective MAOIs such as phenelzine, tranylcypromine, or isocarboxazid.3,4 Nonselective MAOIs thus offer a therapeutic opportunity for patients who do not respond to single or dual-mechanism strategies; moreover, nonselective MAOIs have compelling effectiveness data for other conditions, including panic disorder and social phobia.5 Although MAOIs are among the most effective pharmacologic agents for MDD,6 they are underutilized because of an inadequate understanding of risk mechanisms and resultant fear of catastrophic outcomes. Because of the difficulties encountered in achieving clinical remission for MDD, the nonselective MAOIs deserve a second look.
 

Differentiation of MAO-A from MAO-B. It is essential to understand the mechanism of action of MAOIs, specifically the impact of MAO-A inhibition. Although the enzyme MAO was known in the 1950s, it wasn’t until 1968 that Johnston7 postulated the existence of >1 form. In 1971, Goridis and Neff8 used clorgyline to examine the deamination rate by MAO of tyramine and norepinephrine. They found that tyramine appeared to be a substrate of both MAO isoforms, but only 1 of the MAO types was sensitive to the inhibitory effects of clorgyline. They also discerned that norepinephrine was only a substrate for MAO-A, and that this form of MAO was sensitive to clorgyline inhibition. Thus, the forms of MAO were characterized by their preferred substrates (Table 29,10), and then later by their tissue distribution. Phenylethylamine is a naturally occurring compound found in foods, such as chocolate, and has an in vitro pharmacology similar to amphetamine but with 1 important difference: it has a short half-life of 5 to 10 minutes after oral ingestion, and therefore no appreciable CNS impact.

 

 

 

Within the CNS, norepinephrine and dopamine neurons possess both MAO forms, with the MAO-A content greater than MAO-B. Serotonergic neurons only contain MAO-B.11 Outside of the CNS, MAO-A predominates, with only platelets and lymphocytes possessing MAO-B activity.11 The overall relative tissue proportions of MAO-A to MAO-B activity are: brain, 25% MAO-A, 75% MAO-B; liver, 50% MAO-A, 50% MAO-B; intestine, 80% MAO-A, 20% MAO-B; and peripheral adrenergic neurons, 90% MAO-A, 10% MAO-B.

Because of its specificity for serotonin and norepinephrine, CNS MAO-A inhibition is necessary for antidepressant effects. MAO-B inhibition by itself does not appear to raise CNS dopamine levels unless exogenous dopamine is supplied.11 All MAOIs used in the United States to treat depression are irreversible, nonselective inhibitors of MAO-A and MAO-B.

Selegiline in oral form generates low plasma levels and primarily inhibits MAO-B. The transdermal form of selegiline achieves significantly greater systemic exposure, and at these higher plasma levels selegiline is a nonselective, irreversible MAOI effective for MDD (Figure 112). Administering selegiline systemically via a transdermal patch avoids clinically significant MAOI effects in the gut, so no dietary warnings exist for the lowest dose (6 mg/24 hours), although there are warnings for the higher dosages (9 mg/24 hours and 12 mg/24 hours).


Differentiation of MAOIs by chemical class. The earliest MAOI, iproniazid, was a hydrazine derivative and exhibited hepatotoxicity,13 as did certain other hydrazine MAOIs. This lead to a search for safer hydrazine and nonhydrazine alternatives. Isocarboxazid and phenelzine are the 2 hydrazine MAOIs available in the United States, while tranylcypromine and selegiline transdermal are nonhydrazines (Figure 2).


What distinguishes the nonhydrazine medication selegiline is that its metabolism generates L-amphetamine metabolites (Figure 314). This property was thought to be shared by other non­hydrazines, but recent studies indicate than neither tranylcypromine15 nor the MAO-B–selective rasagiline possess amphetamine metabolites.16 Unlike the dextro isomers, L-amphetamine structures do not inhibit dopamine reuptake or cause euphoria, but can cause stimulation (eg, sleep disturbance) by inhibiting norepinephrine reuptake, and also by interacting with the trace amine-associated receptor 1 (TAAR1), an intracellular receptor expressed within the presynaptic terminal of monoamine neurons. Activation of TAAR1 by tyramine is an important part of the hypertensive effects related to excessive tyramine exposure.17 (The importance of TAAR1 and the interaction with tyramine is discussed in the next section.) Importantly, patients taking selegiline must be warned that certain drug screens may not discriminate between levo and dextro isomers of amphetamines, and that the use of selegiline should be disclosed prior to drug testing procedures.

MAOIs and tyramine: Dietary requirements

Clinicians who are familiar with MAOIs recognize that there are dietary restrictions to minimize patients’ exposure to tyramine. As most clinicians know, significant tyramine ingestion may cause an increase in blood pressure (BP) in patients taking an MAOI, but many overestimate the prevalence of foods high in tyramine content since the original reports emerged in the early 1960s.18 In a recent monograph, one of the leading experts on MAOIs, Professor Ken Gillman, stated:

Very few foods now contain problematically high tyramine levels, that is a result of great changes in international food production methods and hygiene regulations. Cheese is the only food that, in the past, has been associated with documented fatalities resulting from hypertension. Nowadays most cheeses are quite safe, and even ‘matured’ cheeses are usually safe in healthy-sized portions. The variability of sensitivity to tyramine between individuals, and the sometimes unpredictable amount of tyramine content in foods, means a little knowledge and care are still required.19

 

 

 

What is tyramine? Tyramine is a biogenic amine that is virtually absent in fresh animal protein sources but is enriched after decay or fermentation.20 Modern food processing and handling methods have significantly limited the tyramine content in processed foods, with the exception of certain cheeses and sauces, as discussed below. Moreover, modern assaying techniques using high-performance liquid chromatography have generated extremely accurate assessments of the tyramine content of specific foods.21 Data published prior to 2000 are not reliable, because many of these publications employed outdated methods.17

When ingested, tyramine is metabolized by gut MAO-A, with doses up to 400 mg causing no known effects, although most people rarely ingest >25 mg during a meal.22 In addition to being a substrate for MAO-A, tyramine is also a substrate for the dopamine transporter, norepinephrine transporter (NET), the vesicular monoamine transporter 2, and TAAR1.23 Tyramine enters the cell via NET, where it interacts with TAAR1, a G protein-coupled receptor that is responsive to trace amines, such as tyramine, as well as amphetamines.20 The agonist properties at TAAR1 are the presumed site of action for the BP effects of tyramine, because binding results in potent release of norepinephrine.20,24 When tyramine is supplied to an animal in which MAO-A is inhibited, the decreased peripheral catabolism of tyramine results in markedly increased norepinephrine release by peripheral adrenergic neurons. Moreover, the absence of MAO-A activity in those neurons prevents any norepinephrine breakdown, resulting in robust synaptic norepinephrine delivery and peripheral effects.

All orally administered irreversible MAOIs potently inhibit gut and systemic MAO-A, and are susceptible to the impact of significant tyramine ingestion. The exception is selegiline transdermal (Figure 112), as appreciable gut MAO-A inhibition does not occur until doses >6 mg/24 hours are reached.22 No significant pressor response was seen in participants taking selegiline transdermal, 6 mg/24 hours for 13 days, who consumed a meal that provided 400 mg of tyramine.22 Conversely, for oral agents that produce gut MAO-A inhibition, tyramine doses as low as 8 to 10 mg (when administered as tyramine capsules) may increase systolic pressure by 30 mm Hg.25 The dietary warnings do not apply to rasagiline, which is a selective MAO-B inhibitor, although rasagiline may have an impact on resting BP; the prescribing information for rasagiline includes warnings about hypotension and hypertension.26

What to tell patients about tyramine. Although administering pure tyramine capsules can induce a measurable change in systolic BP, when ingested as food, tyramine doses <50 mg are unlikely to cause an increase in BP sufficient to warrant clinical intervention, although some individuals can be sensitive to 10 to 25 mg.19 When discussing with patients safety issues related to diet, there are a few important concepts to remember19:

  • In an era when the tyramine content of foods was much higher (1960 to 1964) and MAOI users received no dietary guidance, only 14 deaths were reported among an estimated 1.5 million patients who took MAOIs.
  • MAOIs do not raise BP, and their use is associated with orthostasis in some patients.
  • Routine exercise or other vigorous activities (eg, weightlifting) can raise systolic pressure well above 200 mm Hg, and routine baseline systolic pressures, ranging from 180 to 220 mm Hg, do not increase the risk of subarachnoid hemorrhage.
  • Hospital evaluation is needed only if a substantial amount of tyramine is ingested (eg, estimated ≥100 mg), and self-monitoring shows a systolic BP ≥220 mm Hg over a prolonged period (eg, 2 hours). Ingestion of 100 mg of tyramine would almost certainly have to be intentional, as it would require one to consume 3.5 oz of the most highly tyramine-laden cheeses.
     

Emphasize to patients that only a small number of highly aged cheeses, foods, and sauces contain high quantities of tyramine, and that even these foods can be enjoyed in small amounts. All patients who are prescribed an MAOI also should purchase a portable BP cuff for those rare instances when a dietary indiscretion may have occurred and the person experiences a headache within 1 to 2 hours after tyramine ingestion. Most reactions are self-limited and resolve over 2 to 4 hours.

Patients who ingest ≥100 mg of tyramine should be evaluated by a physician. Under no circumstances should a patient be given a prescription for nifedipine or other medications that can abruptly lower BP, because this may result in complications, including myocardial infarction.27,28 Counsel patients to remain calm. Some clinicians endorse the use of low doses of benzodiazepines (the equivalent of alprazolam 0.5 mg) to facilitate this, because anxiety elevates BP. A recent emergency room study of patients with an initial systolic BP ≥160 mm Hg or diastolic BP ≥100 mm Hg without end organ damage demonstrated that alprazolam, 0.5 mg, was as effective as captopril, 25 mg, in lowering BP.29

Also, tell patients that if a food is unfamiliar and highly aged or fermented, they should avoid it until they can further inquire about it. In a review, Gillman19 provides the tyramine content of an exhaustive list of cheeses, aged meats, and sauces (see Related Resources). For other products, patients often can obtain information directly from the manufacturer. In many parts of the world, assays for tyramine content are required as a demonstration of adequate product safety procedures. Even the most highly aged cheeses with a tyramine content of 1,000 g/kg can be enjoyed in small amounts (<1 oz), and most products would require heroic intake to achieve clinically significant tyramine ingestion (Table 319).

Improved education can clarify the risks

Medications such as lithium, clozapine, and MAOIs have a proven record of efficacy, yet often are underused due to fears engendered by lack of systematic training. A recent initiative in New York thus aimed to increase rates of clozapine prescribing by providing clinicians with an education consultation center.30 Similarly, enhanced education regarding MAOIs could increase the use of these highly effective medications. With a better understanding of MAOIs, clinicians can become adept at using these medications, and therefore expand the armamentarium of agents available to patients with MDD, as well as to those with panic disorder and social phobia.
 

Bottom Line

Monoamine oxidase inhibitors (MAOIs) are among the most effective medications for treating depression but are underutilized because of misunderstanding of risk mechanisms and fear of catastrophic outcomes. Through education, astute clinicians can master the proper use of MAOIs and add these agents to their treatment armamentarium.

Related Resource

  • Gillman PK. Monoamine oxidase inhibitors: a review concerning dietary tyramine and drug interactions. PsychoTropical Commentaries. 2016;16(6):1-97.

Drug Brand Names

Alprazolam • Xanax
Bupropion • Wellbutrin, Zyban
Captopril • Capoten
Clozapine • Clozaril
Imipramine • Tofranil
Iproniazid • Marsilid
Isocarboxazid • Marplan
Lithium • Eskalith, Lithobid
Meperidine • Demerol
Methadone • Dolophine, Methadose
Mirtazapine • Remeron
Nifedipine • Adalat, Procardia
Norepinephrine • Levophed
Phenelzine • Nardil
Rasagiline • Azilect
Selegiline oral • Eldepryl
Selegiline transdermal • Emsam
Tramadol • Ultram
Tranylcypromine • Parnate

 

Despite an abundance of evidenced-based literature supporting monoamine oxidase inhibitors (MAOIs) as an effective treatment for depression, use of these agents has decreased drastically in the past 3 decades. A lack of industry support and the ease of use of other agents are contributing factors, but the biggest impediments to routine use of MAOIs are unfamiliarity with their efficacy advantages and concerns about adverse effects, particularly the risk of hypertensive crises and serotonin syndrome. Many misconceptions regarding these medications are based on outdated data and studies that are no longer reliable.

The goal of this 2-part review is to provide clinicians with updated information regarding MAOIs. Part 1 provides a brief description of:

  • the pharmacology of nonselective irreversible MAOIs
  • the mechanism by which tyramine induces hypertension
  • sources of clinically significant tyramine exposure
  • what to tell patients about dietary restrictions and MAOIs.

Part 2 of this guide will cover the risk of serotonin syndrome when MAOIs are combined with inhibitors of serotonin reuptake, how to initiate MAOI therapy, and augmenting MAOIs with other agents.

The pharmacology of MAOIs

First used clinically in the 1950s to treat tuberculosis, MAOIs have a long and interesting history (see the Box “A brief history of monoamine oxidase inhibitors”). Table 11 lists MAOIs currently available in the United States, including the MAO-B–specific agent rasagiline, which is used for Parkinson’s disease.

Manipulation of the monoamines serotonin, norepinephrine, and dopamine is fundamental to managing major depressive disorder (MDD), yet only nonselective MAOIs directly promote neurotransmission of all 3 by inhibiting MAO-A and MAO-B.2 The Sequenced Treatment Alternatives to Relieve Depression (STAR*D) study demonstrated that <50% of MDD patients achieve remission in monotherapy trials of selective serotonin reuptake inhibitors, serotonin norepinephrine reuptake inhibitors, mirtazapine, or bupropion, necessitating consideration of antidepressant combinations, augmentation options, and eventually irreversible, nonselective MAOIs such as phenelzine, tranylcypromine, or isocarboxazid.3,4 Nonselective MAOIs thus offer a therapeutic opportunity for patients who do not respond to single or dual-mechanism strategies; moreover, nonselective MAOIs have compelling effectiveness data for other conditions, including panic disorder and social phobia.5 Although MAOIs are among the most effective pharmacologic agents for MDD,6 they are underutilized because of an inadequate understanding of risk mechanisms and resultant fear of catastrophic outcomes. Because of the difficulties encountered in achieving clinical remission for MDD, the nonselective MAOIs deserve a second look.
 

Differentiation of MAO-A from MAO-B. It is essential to understand the mechanism of action of MAOIs, specifically the impact of MAO-A inhibition. Although the enzyme MAO was known in the 1950s, it wasn’t until 1968 that Johnston7 postulated the existence of >1 form. In 1971, Goridis and Neff8 used clorgyline to examine the deamination rate by MAO of tyramine and norepinephrine. They found that tyramine appeared to be a substrate of both MAO isoforms, but only 1 of the MAO types was sensitive to the inhibitory effects of clorgyline. They also discerned that norepinephrine was only a substrate for MAO-A, and that this form of MAO was sensitive to clorgyline inhibition. Thus, the forms of MAO were characterized by their preferred substrates (Table 29,10), and then later by their tissue distribution. Phenylethylamine is a naturally occurring compound found in foods, such as chocolate, and has an in vitro pharmacology similar to amphetamine but with 1 important difference: it has a short half-life of 5 to 10 minutes after oral ingestion, and therefore no appreciable CNS impact.

 

 

 

Within the CNS, norepinephrine and dopamine neurons possess both MAO forms, with the MAO-A content greater than MAO-B. Serotonergic neurons only contain MAO-B.11 Outside of the CNS, MAO-A predominates, with only platelets and lymphocytes possessing MAO-B activity.11 The overall relative tissue proportions of MAO-A to MAO-B activity are: brain, 25% MAO-A, 75% MAO-B; liver, 50% MAO-A, 50% MAO-B; intestine, 80% MAO-A, 20% MAO-B; and peripheral adrenergic neurons, 90% MAO-A, 10% MAO-B.

Because of its specificity for serotonin and norepinephrine, CNS MAO-A inhibition is necessary for antidepressant effects. MAO-B inhibition by itself does not appear to raise CNS dopamine levels unless exogenous dopamine is supplied.11 All MAOIs used in the United States to treat depression are irreversible, nonselective inhibitors of MAO-A and MAO-B.

Selegiline in oral form generates low plasma levels and primarily inhibits MAO-B. The transdermal form of selegiline achieves significantly greater systemic exposure, and at these higher plasma levels selegiline is a nonselective, irreversible MAOI effective for MDD (Figure 112). Administering selegiline systemically via a transdermal patch avoids clinically significant MAOI effects in the gut, so no dietary warnings exist for the lowest dose (6 mg/24 hours), although there are warnings for the higher dosages (9 mg/24 hours and 12 mg/24 hours).


Differentiation of MAOIs by chemical class. The earliest MAOI, iproniazid, was a hydrazine derivative and exhibited hepatotoxicity,13 as did certain other hydrazine MAOIs. This lead to a search for safer hydrazine and nonhydrazine alternatives. Isocarboxazid and phenelzine are the 2 hydrazine MAOIs available in the United States, while tranylcypromine and selegiline transdermal are nonhydrazines (Figure 2).


What distinguishes the nonhydrazine medication selegiline is that its metabolism generates L-amphetamine metabolites (Figure 314). This property was thought to be shared by other non­hydrazines, but recent studies indicate than neither tranylcypromine15 nor the MAO-B–selective rasagiline possess amphetamine metabolites.16 Unlike the dextro isomers, L-amphetamine structures do not inhibit dopamine reuptake or cause euphoria, but can cause stimulation (eg, sleep disturbance) by inhibiting norepinephrine reuptake, and also by interacting with the trace amine-associated receptor 1 (TAAR1), an intracellular receptor expressed within the presynaptic terminal of monoamine neurons. Activation of TAAR1 by tyramine is an important part of the hypertensive effects related to excessive tyramine exposure.17 (The importance of TAAR1 and the interaction with tyramine is discussed in the next section.) Importantly, patients taking selegiline must be warned that certain drug screens may not discriminate between levo and dextro isomers of amphetamines, and that the use of selegiline should be disclosed prior to drug testing procedures.

MAOIs and tyramine: Dietary requirements

Clinicians who are familiar with MAOIs recognize that there are dietary restrictions to minimize patients’ exposure to tyramine. As most clinicians know, significant tyramine ingestion may cause an increase in blood pressure (BP) in patients taking an MAOI, but many overestimate the prevalence of foods high in tyramine content since the original reports emerged in the early 1960s.18 In a recent monograph, one of the leading experts on MAOIs, Professor Ken Gillman, stated:

Very few foods now contain problematically high tyramine levels, that is a result of great changes in international food production methods and hygiene regulations. Cheese is the only food that, in the past, has been associated with documented fatalities resulting from hypertension. Nowadays most cheeses are quite safe, and even ‘matured’ cheeses are usually safe in healthy-sized portions. The variability of sensitivity to tyramine between individuals, and the sometimes unpredictable amount of tyramine content in foods, means a little knowledge and care are still required.19

 

 

 

What is tyramine? Tyramine is a biogenic amine that is virtually absent in fresh animal protein sources but is enriched after decay or fermentation.20 Modern food processing and handling methods have significantly limited the tyramine content in processed foods, with the exception of certain cheeses and sauces, as discussed below. Moreover, modern assaying techniques using high-performance liquid chromatography have generated extremely accurate assessments of the tyramine content of specific foods.21 Data published prior to 2000 are not reliable, because many of these publications employed outdated methods.17

When ingested, tyramine is metabolized by gut MAO-A, with doses up to 400 mg causing no known effects, although most people rarely ingest >25 mg during a meal.22 In addition to being a substrate for MAO-A, tyramine is also a substrate for the dopamine transporter, norepinephrine transporter (NET), the vesicular monoamine transporter 2, and TAAR1.23 Tyramine enters the cell via NET, where it interacts with TAAR1, a G protein-coupled receptor that is responsive to trace amines, such as tyramine, as well as amphetamines.20 The agonist properties at TAAR1 are the presumed site of action for the BP effects of tyramine, because binding results in potent release of norepinephrine.20,24 When tyramine is supplied to an animal in which MAO-A is inhibited, the decreased peripheral catabolism of tyramine results in markedly increased norepinephrine release by peripheral adrenergic neurons. Moreover, the absence of MAO-A activity in those neurons prevents any norepinephrine breakdown, resulting in robust synaptic norepinephrine delivery and peripheral effects.

All orally administered irreversible MAOIs potently inhibit gut and systemic MAO-A, and are susceptible to the impact of significant tyramine ingestion. The exception is selegiline transdermal (Figure 112), as appreciable gut MAO-A inhibition does not occur until doses >6 mg/24 hours are reached.22 No significant pressor response was seen in participants taking selegiline transdermal, 6 mg/24 hours for 13 days, who consumed a meal that provided 400 mg of tyramine.22 Conversely, for oral agents that produce gut MAO-A inhibition, tyramine doses as low as 8 to 10 mg (when administered as tyramine capsules) may increase systolic pressure by 30 mm Hg.25 The dietary warnings do not apply to rasagiline, which is a selective MAO-B inhibitor, although rasagiline may have an impact on resting BP; the prescribing information for rasagiline includes warnings about hypotension and hypertension.26

What to tell patients about tyramine. Although administering pure tyramine capsules can induce a measurable change in systolic BP, when ingested as food, tyramine doses <50 mg are unlikely to cause an increase in BP sufficient to warrant clinical intervention, although some individuals can be sensitive to 10 to 25 mg.19 When discussing with patients safety issues related to diet, there are a few important concepts to remember19:

  • In an era when the tyramine content of foods was much higher (1960 to 1964) and MAOI users received no dietary guidance, only 14 deaths were reported among an estimated 1.5 million patients who took MAOIs.
  • MAOIs do not raise BP, and their use is associated with orthostasis in some patients.
  • Routine exercise or other vigorous activities (eg, weightlifting) can raise systolic pressure well above 200 mm Hg, and routine baseline systolic pressures, ranging from 180 to 220 mm Hg, do not increase the risk of subarachnoid hemorrhage.
  • Hospital evaluation is needed only if a substantial amount of tyramine is ingested (eg, estimated ≥100 mg), and self-monitoring shows a systolic BP ≥220 mm Hg over a prolonged period (eg, 2 hours). Ingestion of 100 mg of tyramine would almost certainly have to be intentional, as it would require one to consume 3.5 oz of the most highly tyramine-laden cheeses.
     

Emphasize to patients that only a small number of highly aged cheeses, foods, and sauces contain high quantities of tyramine, and that even these foods can be enjoyed in small amounts. All patients who are prescribed an MAOI also should purchase a portable BP cuff for those rare instances when a dietary indiscretion may have occurred and the person experiences a headache within 1 to 2 hours after tyramine ingestion. Most reactions are self-limited and resolve over 2 to 4 hours.

Patients who ingest ≥100 mg of tyramine should be evaluated by a physician. Under no circumstances should a patient be given a prescription for nifedipine or other medications that can abruptly lower BP, because this may result in complications, including myocardial infarction.27,28 Counsel patients to remain calm. Some clinicians endorse the use of low doses of benzodiazepines (the equivalent of alprazolam 0.5 mg) to facilitate this, because anxiety elevates BP. A recent emergency room study of patients with an initial systolic BP ≥160 mm Hg or diastolic BP ≥100 mm Hg without end organ damage demonstrated that alprazolam, 0.5 mg, was as effective as captopril, 25 mg, in lowering BP.29

Also, tell patients that if a food is unfamiliar and highly aged or fermented, they should avoid it until they can further inquire about it. In a review, Gillman19 provides the tyramine content of an exhaustive list of cheeses, aged meats, and sauces (see Related Resources). For other products, patients often can obtain information directly from the manufacturer. In many parts of the world, assays for tyramine content are required as a demonstration of adequate product safety procedures. Even the most highly aged cheeses with a tyramine content of 1,000 g/kg can be enjoyed in small amounts (<1 oz), and most products would require heroic intake to achieve clinically significant tyramine ingestion (Table 319).

Improved education can clarify the risks

Medications such as lithium, clozapine, and MAOIs have a proven record of efficacy, yet often are underused due to fears engendered by lack of systematic training. A recent initiative in New York thus aimed to increase rates of clozapine prescribing by providing clinicians with an education consultation center.30 Similarly, enhanced education regarding MAOIs could increase the use of these highly effective medications. With a better understanding of MAOIs, clinicians can become adept at using these medications, and therefore expand the armamentarium of agents available to patients with MDD, as well as to those with panic disorder and social phobia.
 

Bottom Line

Monoamine oxidase inhibitors (MAOIs) are among the most effective medications for treating depression but are underutilized because of misunderstanding of risk mechanisms and fear of catastrophic outcomes. Through education, astute clinicians can master the proper use of MAOIs and add these agents to their treatment armamentarium.

Related Resource

  • Gillman PK. Monoamine oxidase inhibitors: a review concerning dietary tyramine and drug interactions. PsychoTropical Commentaries. 2016;16(6):1-97.

Drug Brand Names

Alprazolam • Xanax
Bupropion • Wellbutrin, Zyban
Captopril • Capoten
Clozapine • Clozaril
Imipramine • Tofranil
Iproniazid • Marsilid
Isocarboxazid • Marplan
Lithium • Eskalith, Lithobid
Meperidine • Demerol
Methadone • Dolophine, Methadose
Mirtazapine • Remeron
Nifedipine • Adalat, Procardia
Norepinephrine • Levophed
Phenelzine • Nardil
Rasagiline • Azilect
Selegiline oral • Eldepryl
Selegiline transdermal • Emsam
Tramadol • Ultram
Tranylcypromine • Parnate

References

1. Panisset M, Chen JJ, Rhyee SH, et al. Serotonin toxicity association with concomitant antidepressants and rasagiline treatment: retrospective study (STACCATO). Pharmacotherapy. 2014;34(12):1250-1258.
2. López-Muñoz F, Alamo C. Monoaminergic neuro­transmission: the history of the discovery of antidepressants from 1950s until today. Curr Pharm Des. 2009;15(14):1563-1586.
3. Nierenberg AA, Fava M, Trivedi MH, et al. A comparison of lithium and T(3) augmentation following two failed medication treatments for depression: a STAR*D report. Am J Psychiatry. 2006;163(9):1519-1530; quiz 1665.
4. Trivedi MH, Fava M, Wisniewski SR, et al; STAR*D Study Team. Medication augmentation after the failure of SSRIs for depression. New Engl J Med. 2006;354(12):1243-1252.
5. Bandelow B, Zohar J, Hollander E, et al; World Federation of Societies of Biological Psychiatry Task Force on Treatment Guidelines for Anxiety, Obsessive-Compulsive and Posttraumatic Stress Disorders. World Federation of Societies of Biological Psychiatry (WFSBP) guidelines for the pharmacological treatment of anxiety, obsessive-compulsive and posttraumatic stress disorders. World J Biol Psychiatry. 2002;3(4):171-199.
6. Shulman KI, Herrmann N, Walker SE. Current place of monoamine oxidase inhibitors in the treatment of depression. CNS Drugs. 2013;27(10):789-797.
7. Johnston JP. Some observations upon a new inhibitor of monoamine oxidase in brain tissue. Biochem Pharmacol. 1968;17(7):1285-1297.
8. Goridis C, Neff NH. Monoamine oxidase in sympathetic nerves: a transmitter specific enzyme type. Br J Pharmacol. 1971;43(4):814-818.
9. Geha RM, Rebrin I, Chen K, et al. Substrate and inhibitor specificities for human monoamine oxidase A and B are influenced by a single amino acid. J Biol Chem. 2001;276(13):9877-9882.
10. O’Carroll AM, Fowler CJ, Phillips JP, et al. The deamination of dopamine by human brain monoamine oxidase. Specificity for the two enzyme forms in seven brain regions. Naunyn Schmiedebergs Arch Pharmacol. 1983;322(3):198-202.
11. Stahl SM, Felker A. Monoamine oxidase inhibitors: a modern guide to an unrequited class of antidepressants. CNS Spectr. 2008;13(10):855-780.
12. Mawhinney M, Cole D, Azzaro AJ. Daily transdermal administration of selegiline to guinea-pigs preferentially inhibits monoamine oxidase activity in brain when compared with intestinal and hepatic tissues. J Pharm Pharmacol. 2003;55(1):27-34.
13. Maille F, Duvoux C, Cherqui D, et al. Auxiliary hepatic transplantation in iproniazid-induced subfulminant hepatitis. Should iproniazid still be sold in France? [in French]. Gastroenterol Clin Biol. 1999;23(10):1083-1085.
14. Salonen JS, Nyman L, Boobis AR, et al. Comparative studies on the cytochrome p450-associated metabolism and interaction potential of selegiline between human liver-derived in vitro systems. Drug Metab Dispos. 2003;31(9):1093-1102.
15. Iwersen S, Schmoldt A. One fatal and one nonfatal intoxication with tranylcypromine. Absence of amphetamines as metabolites. J Anal Toxicol. 1996;20(5):301-304.
16. Müller T, Hoffmann JA, Dimpfel W, et al. Switch from selegiline to rasagiline is beneficial in patients with Parkinson’s disease. J Neural Transm (Vienna). 2013;120(5):761-765.
17. Lewin AH, Miller GM, Gilmour B. Trace amine-associated receptor 1 is a stereoselective binding site for compounds in the amphetamine class. Bioorg Med Chem. 2011;19(23):7044-7048.
18. Blackwell B. Hypertensive crisis due to monoamine-oxidase inhibitors. Lancet. 1963;2(7313):849-850.
19. Gillman PK. Monoamine oxidase inhibitors: a review concerning dietary tyramine and drug interactions. PsychoTropical Commentaries. 2016;16(6):1-97.
20. Pei Y, Asif-Malik A, Canales JJ. Trace amines and the trace amine-associated receptor 1: pharmacology, neurochemistry, and clinical implications. Front Neurosci. 2016;10:148.
21. Fiechter G, Sivec G, Mayer HK. Application of UHPLC for the simultaneous analysis of free amino acids and biogenic amines in ripened acid-curd cheeses. J Chromatogr B Analyt Technol Biomed Life Sci. 2013;927:191-200.
22. Blob LF, Sharoky M, Campbell BJ, et al. Effects of a tyramine-enriched meal on blood pressure response in healthy male volunteers treated with selegiline transdermal system 6 mg/24 hour. CNS Spectr. 2007;12(1):25-34.
23. Partilla JS, Dempsey AG, Nagpal AS, et al. Interaction of amphetamines and related compounds at the vesicular monoamine transporter. J Pharmacol Exp Ther. 2006;319(1):237-246.
24. Borowsky B, Adham N, Jones KA, et al. Trace amines: identification of a family of mammalian G protein-coupled receptors. Proc Natl Acad Sci U S A. 2001;98(16):8966-8971.
25. Azzaro AJ, Vandenberg CM, Blob LF, et al. Tyramine pressor sensitivity during treatment with the selegiline transdermal system 6 mg/24 h in healthy subjects. J Clin Pharmacol. 2006;46(8):933-944.
26. Azilect [package insert]. Overland Park, KS: Teva Neuroscience, Inc.; 2014.
27. Marik PE, Varon J. Hypertensive crises: challenges and management. Chest. 2007;131(6):1949-1962.
28. Burton TJ, Wilkinson IB. The dangers of immediate-release nifedipine in the emergency treatment of hypertension. J Hum Hypertens. 2008;22(4):301-302.
29. Yilmaz S, Pekdemir M, Tural U, et al. Comparison of alprazolam versus captopril in high blood pressure: a randomized controlled trial. Blood Press. 2011;20(4):239-243.
30. Carruthers J, Radigan M, Erlich MD, et al. An initiative to improve clozapine prescribing in New York State. Psychiatr Serv. 2016;67(4):369-371.

References

1. Panisset M, Chen JJ, Rhyee SH, et al. Serotonin toxicity association with concomitant antidepressants and rasagiline treatment: retrospective study (STACCATO). Pharmacotherapy. 2014;34(12):1250-1258.
2. López-Muñoz F, Alamo C. Monoaminergic neuro­transmission: the history of the discovery of antidepressants from 1950s until today. Curr Pharm Des. 2009;15(14):1563-1586.
3. Nierenberg AA, Fava M, Trivedi MH, et al. A comparison of lithium and T(3) augmentation following two failed medication treatments for depression: a STAR*D report. Am J Psychiatry. 2006;163(9):1519-1530; quiz 1665.
4. Trivedi MH, Fava M, Wisniewski SR, et al; STAR*D Study Team. Medication augmentation after the failure of SSRIs for depression. New Engl J Med. 2006;354(12):1243-1252.
5. Bandelow B, Zohar J, Hollander E, et al; World Federation of Societies of Biological Psychiatry Task Force on Treatment Guidelines for Anxiety, Obsessive-Compulsive and Posttraumatic Stress Disorders. World Federation of Societies of Biological Psychiatry (WFSBP) guidelines for the pharmacological treatment of anxiety, obsessive-compulsive and posttraumatic stress disorders. World J Biol Psychiatry. 2002;3(4):171-199.
6. Shulman KI, Herrmann N, Walker SE. Current place of monoamine oxidase inhibitors in the treatment of depression. CNS Drugs. 2013;27(10):789-797.
7. Johnston JP. Some observations upon a new inhibitor of monoamine oxidase in brain tissue. Biochem Pharmacol. 1968;17(7):1285-1297.
8. Goridis C, Neff NH. Monoamine oxidase in sympathetic nerves: a transmitter specific enzyme type. Br J Pharmacol. 1971;43(4):814-818.
9. Geha RM, Rebrin I, Chen K, et al. Substrate and inhibitor specificities for human monoamine oxidase A and B are influenced by a single amino acid. J Biol Chem. 2001;276(13):9877-9882.
10. O’Carroll AM, Fowler CJ, Phillips JP, et al. The deamination of dopamine by human brain monoamine oxidase. Specificity for the two enzyme forms in seven brain regions. Naunyn Schmiedebergs Arch Pharmacol. 1983;322(3):198-202.
11. Stahl SM, Felker A. Monoamine oxidase inhibitors: a modern guide to an unrequited class of antidepressants. CNS Spectr. 2008;13(10):855-780.
12. Mawhinney M, Cole D, Azzaro AJ. Daily transdermal administration of selegiline to guinea-pigs preferentially inhibits monoamine oxidase activity in brain when compared with intestinal and hepatic tissues. J Pharm Pharmacol. 2003;55(1):27-34.
13. Maille F, Duvoux C, Cherqui D, et al. Auxiliary hepatic transplantation in iproniazid-induced subfulminant hepatitis. Should iproniazid still be sold in France? [in French]. Gastroenterol Clin Biol. 1999;23(10):1083-1085.
14. Salonen JS, Nyman L, Boobis AR, et al. Comparative studies on the cytochrome p450-associated metabolism and interaction potential of selegiline between human liver-derived in vitro systems. Drug Metab Dispos. 2003;31(9):1093-1102.
15. Iwersen S, Schmoldt A. One fatal and one nonfatal intoxication with tranylcypromine. Absence of amphetamines as metabolites. J Anal Toxicol. 1996;20(5):301-304.
16. Müller T, Hoffmann JA, Dimpfel W, et al. Switch from selegiline to rasagiline is beneficial in patients with Parkinson’s disease. J Neural Transm (Vienna). 2013;120(5):761-765.
17. Lewin AH, Miller GM, Gilmour B. Trace amine-associated receptor 1 is a stereoselective binding site for compounds in the amphetamine class. Bioorg Med Chem. 2011;19(23):7044-7048.
18. Blackwell B. Hypertensive crisis due to monoamine-oxidase inhibitors. Lancet. 1963;2(7313):849-850.
19. Gillman PK. Monoamine oxidase inhibitors: a review concerning dietary tyramine and drug interactions. PsychoTropical Commentaries. 2016;16(6):1-97.
20. Pei Y, Asif-Malik A, Canales JJ. Trace amines and the trace amine-associated receptor 1: pharmacology, neurochemistry, and clinical implications. Front Neurosci. 2016;10:148.
21. Fiechter G, Sivec G, Mayer HK. Application of UHPLC for the simultaneous analysis of free amino acids and biogenic amines in ripened acid-curd cheeses. J Chromatogr B Analyt Technol Biomed Life Sci. 2013;927:191-200.
22. Blob LF, Sharoky M, Campbell BJ, et al. Effects of a tyramine-enriched meal on blood pressure response in healthy male volunteers treated with selegiline transdermal system 6 mg/24 hour. CNS Spectr. 2007;12(1):25-34.
23. Partilla JS, Dempsey AG, Nagpal AS, et al. Interaction of amphetamines and related compounds at the vesicular monoamine transporter. J Pharmacol Exp Ther. 2006;319(1):237-246.
24. Borowsky B, Adham N, Jones KA, et al. Trace amines: identification of a family of mammalian G protein-coupled receptors. Proc Natl Acad Sci U S A. 2001;98(16):8966-8971.
25. Azzaro AJ, Vandenberg CM, Blob LF, et al. Tyramine pressor sensitivity during treatment with the selegiline transdermal system 6 mg/24 h in healthy subjects. J Clin Pharmacol. 2006;46(8):933-944.
26. Azilect [package insert]. Overland Park, KS: Teva Neuroscience, Inc.; 2014.
27. Marik PE, Varon J. Hypertensive crises: challenges and management. Chest. 2007;131(6):1949-1962.
28. Burton TJ, Wilkinson IB. The dangers of immediate-release nifedipine in the emergency treatment of hypertension. J Hum Hypertens. 2008;22(4):301-302.
29. Yilmaz S, Pekdemir M, Tural U, et al. Comparison of alprazolam versus captopril in high blood pressure: a randomized controlled trial. Blood Press. 2011;20(4):239-243.
30. Carruthers J, Radigan M, Erlich MD, et al. An initiative to improve clozapine prescribing in New York State. Psychiatr Serv. 2016;67(4):369-371.

Issue
December 2017
Issue
December 2017
Page Number
14-16,18-23,47,A
Page Number
14-16,18-23,47,A
Publications
Publications
Topics
Article Type
Display Headline
A concise guide to monoamine oxidase inhibitors
Display Headline
A concise guide to monoamine oxidase inhibitors
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Article PDF Media

Dietary restrictions with MAOIs

Article Type
Changed
Thu, 06/10/2021 - 17:03
Display Headline
Dietary restrictions with MAOIs

Author and Disclosure Information

Dr. Meyer is Psychopharmacology Consultant, California Department of State Hospitals, Sacramento, California, and Assistant Clinical Professor of Psychiatry, University of California, San Diego, San Diego, California. He also is Deputy Editor of Current Psychiatry.

Issue
December 2017
Publications
Topics
Sections
Author and Disclosure Information

Dr. Meyer is Psychopharmacology Consultant, California Department of State Hospitals, Sacramento, California, and Assistant Clinical Professor of Psychiatry, University of California, San Diego, San Diego, California. He also is Deputy Editor of Current Psychiatry.

Author and Disclosure Information

Dr. Meyer is Psychopharmacology Consultant, California Department of State Hospitals, Sacramento, California, and Assistant Clinical Professor of Psychiatry, University of California, San Diego, San Diego, California. He also is Deputy Editor of Current Psychiatry.

Issue
December 2017
Issue
December 2017
Publications
Publications
Topics
Article Type
Display Headline
Dietary restrictions with MAOIs
Display Headline
Dietary restrictions with MAOIs
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

Prescribing antipsychotics in geriatric patients: Focus on dementia

Article Type
Changed
Tue, 12/11/2018 - 15:01
Display Headline
Prescribing antipsychotics in geriatric patients: Focus on dementia
 

According to the U.S. Department of Health and Human Services, in 2007, 88% of 1.4 million Medicare claims for second-generation antipsychotics (SGAs) in older adult nursing home residents were associated with a dementia diagnosis. Similar trends have been observed in Canada and Europe.1-4 In a retrospective analysis of medication data from older residents with dementia in 6 care homes in England, long-term (ie, >1 month) use of antipsychotics was the most frequent potentially inappropriate prescribing practice.3 In another study in 7 European countries and Israel, the overall prevalence of antipsychotic use among long-term care residents with dementia was 33%.1 Similarly, a recent literature review5 found that 22% to 86% of antipsychotic prescriptions to older individuals were off-label; this practice was particularly common for individuals with agitation.

Because of the aging population and widespread prescription of antipsychotics to older patients, clinicians need information on the relative risks of using these medications in this population. In the United States, all antipsychotics carry a FDA “black-box” warning of the increased risk of death in older adults with dementia. In addition, the risk of death is increased when prescribing antipsychotics to older adults with other conditions, such as Parkinson’s disease,6 and other safety and tolerability concerns, including falls and fractures, sedation, metabolic abnormalities, and extrapyramidal effects, are highly relevant to geriatric patients.

This 3-part review summarizes findings and recommendations on prescribing antipsychotics to older individuals with schizophrenia, bipolar disorder, depression, and dementia. This third and final installment:

  • briefly summarizes the major studies and analyses relevant to prescribing antipsychotics to older patients with dementia
  • provides a summative opinion on safety and tolerability issues in these older patients
  • highlights the gaps in the evidence base and areas that need additional research.


Summary of benefits, place in treatment armamentarium

Behavioral and psychological symptoms of dementia (BPSD) include agitation, delusional beliefs, repetitive questioning, hallucinations, aggression, wandering, and various socially inappropriate behaviors.7 These occur almost universally in all types and stages of dementia.7 BPSD are among the most complex, stressful, and costly aspects of dementia care, and lead to a myriad of poor health outcomes, including excess morbidity, mortality, hospital stays, and early nursing home placement.8-11 Because BPSD usually occur across all types and stages of dementia,7,12-16 the prevalence of BPSD mirrors the overall prevalence of dementia.

Although all expert organizations, including the American Psychiatric Association,17 recommend nonpharmacologic strategies as first-line treatment for BPSD, for the most part, these recommendations have not been translated into standard clinical management or routine care.18 Because of a perceived lack of other options, the current mainstay of treatment is the off-label use of psychotropics such as antipsy­chotics. Of all the agents currently used for BPSD, SGAs have the strongest evidence base, although benefits are modest at best (standardized effect size 0.13 to 0.16).19,20 In terms of individual SGAs, only risperidone is indicated for aggression in Canada and in Europe (not in the United States); risperidone has the best evidence for efficacy, with a meta-analysis of 5 published randomized controlled trials (RCTs) reporting that risperidone is superior to other SGAs for aggression in dementia.21,22 As a class, first-generation antipsychotics (FGAs) have no clear evidence for BPSD as defined broadly; however, there may be slight benefit for haloperidol for aggression.23,24


Clinical Trials

Adverse effects. A meta-analysis of RCTs of SGAs found that, compared with placebo, SGAs have increased rates of several adverse effects. These include somnolence (17% drug vs 7% placebo; odds ratio [OR], 2.84; 95% confidence interval [CI], 2.25 to 3.58; P < .00001); extrapyramidal symptoms (13% drug vs 8% placebo; OR, 1.51; 95% CI, 1.20 to 1.91; P = .0005; primarily attributable to risperidone); abnormal gait (10% drug vs 2% placebo; OR, 3.42; 95% CI, 1.78 to 6.56; P = .0002; attributable to olanzapine and risperidone); edema (9% drug vs 4% placebo; OR, 1.99; 95% CI, 1.20 to 3.30; P = .008; attributable to olanzapine and risperidone); urinary tract infections/incontinence (16% drug vs 12% placebo; OR, 1.28; 95% CI, 1.02 to 1.61; P = .04); cognitive impairment measured as difference in Mini-Mental State Examination score (95% CI, 0.38 to 1.09; P < .0001)25; and stroke (1.9% drug vs 0.9% placebo, OR, 2.13; 95% CI, 1.20 to 3.75; P = .009).21,26

In the 42-site Clinical Antipsychotic Trials of Intervention Effectiveness Alzheimer’s disease RCT, 421 outpatients with Alzheimer’s disease and BPSD were randomized to an SGA (risperidone, olanzapine, or quetiapine) or placebo. Compared with placebo, SGAs had a higher rate of parkinsonism or extrapyramidal signs (olanzapine and risperidone groups); sedation; confusion/changes in mental status (olanzapine and risperidone); psychotic symptoms (olanzapine); and increase in body weight and body mass index.26

In the 2005 FDA black-box warning, pneumonia and cardiac adverse effects were cited as primary causes of death for patients with dementia taking SGAs. A subsequent observational study confirmed that use of either FGAs or SGAs in geriatric patients was associated with an increased risk of pneumonia, in a dose-dependent manner.27 Although there is limited data on cardiac adverse effects in older adults, especially those with dementia taking antipsychotics,28 1 observational study of nursing home residents29 found that those taking FGAs had a significantly higher risk of hospitalization for ventricular arrhythmia or cardiac arrest compared with those who were not taking FGAs. In contrast, there was no increased risk with SGAs.

Mortality.
 In 2005, the FDA announced that based on a reanalysis of 17 placebo-controlled trials (many of which were unpublished) that SGAs were associated with a 1.7-fold increase in mortality compared with placebo.30 As a result, the FDA issued a black-box warning for using SGAs in patients with dementia. The overall OR in a published meta-analysis of mortality with SGAs was 1.54 (1.06 to 2.23; z = 2.28; P = .02), with pooled events of 3.5% mortality vs 2.3% (drug vs placebo).21 This meta-analysis21 also included ad hoc analyses of haloperidol; using combined data from 2 contrasts of haloperidol (with risperidone and quetiapine; 243 patients receiving haloperidol and 239 receiving placebo) they also found 15 deaths (6.2%) with haloperidol and 9 (3.8%) with placebo, resulting in an OR of 1.68.

 

 

 

Other clinical data

Observational studies. Most observational studies have confirmed concerns regarding increased mortality in patients with BPSD who take antipsychotics, with FGAs having a higher risk than SGAs18,31 and SGAs having a higher risk compared with most other psychotropics.32 Three studies that found no increase in mortality with antipsychotics in patients with dementia had methodological issues, including examining prevalence as opposed to new users,33,34 not controlling for exposure,10,33,34 power issues,10,34 not controlling for other psychiatric medications,10 and varying lengths of follow-up.10 An FDA black-box warning for FGAs was announced in 200830 based on 2 observational studies that showed an increased risk of mortality in older adults taking FGAs vs SGAs.35,36

In terms of specific SGAs, Kales et al37 examined the mortality risk associated with individual antipsychotics using various methods to control for confounding. Among a national sample of >33,000 older veterans with dementia newly started on haloperidol, risperidone, olanzapine, quetiapine, or valproic acid and derivatives (as a nonantipsychotic comparator), the highest mortality across all analyses (intent to treat, exposure, propensity-adjusted) was associated with haloperidol, followed by risperidone and olanzapine, valproic acid, and quetiapine.

Most recently, a retrospective case-control study (90,786 patients age ≥65 with dementia) examined the number needed to harm (NNH; ie, number of patients needed to receive treatment that would result in 1 death) over 180 days following initiation of an FGA or SGA.38 This study found the following NNHs: haloperidol, 26 (95% CI, 15 to 99); risperidone, 27 (95% CI, 19 to 46); olanzapine, 40 (95% CI, 21 to 312); and quetiapine, 50 (95% CI, 30 to 150).38 These results are congruent with a review of observational studies that found the highest risk of mortality was associated with haloperidol and chlorpromazine, and the lowest risk with olanzapine, quetiapine, and ziprasidone.28

Patterns of antipsychotic use in older dementia patients

There are high rates of antipsychotic use in patients with dementia. Before the FDA issued the black-box warning, the Aging Demographics and Memory study found that the rate of antipsychotic use in community (outpatient) older adults with dementia was approximately 19% between 2002 and 2004 in a representative sample of 307 older adults.39 Another study examining trends in community antipsychotic use in the U.S. Department of Veterans Affairs (VA) found that in the 1990s, SGA use was increasing; approximately 18% of outpatients with dementia were taking these agents.40 Use of SGAs began to decline in 2003, ahead of the 2005 black-box warning, in tandem with other advisories (eg, diabetes, metabolic syndrome,41 and stroke risk).42,43 Olanzapine and risperidone showed declining rates between 2003 and 2005, whereas quetiapine use significantly increased during this period. All 3 SGAs declined after the black-box warning. However, by the end of 2007, the use of SGAs had leveled off to approximately 12% of VA patients with dementia. A recent U.S. Government Accountability Office (GAO) report found that in 2012, 14% of older adult Medicare Part D enrollees with dementia living in the community were prescribed an antipsychotic.44

Use in nursing home residents. Because BPSD are one of the main reasons people with dementia are placed in nursing homes, it is not surprising that rates of antipsychotic use are higher in these settings than in the community. Prior to the black-box warning, studies found that 24% to 32% of nursing home residents were treated with antipsychotics.45-47 A study examining VA nursing homes (n = 133 facilities, n = 3,692 veterans) found that approximately 26% of residents were prescribed antipsychotics in 2004 to 2005.48 The Center for Medicare and Medicaid Services (CMS) National Partnership to Improve Dementia Care in Nursing Homes has appeared to lower antipsychotic medication use in nursing homes; the rate decreased from 24% in long-stay nursing home residents nationwide in 2011 to 19% by the end of 2014. Specific to dementia, a 2010 CMS report49 indicated that approximately 40% of nursing home residents with cognitive impairment and behavioral issues, without psychosis, received antipsychotics. The GAO data indicated that approximately 33% of older Medicare Part D enrollees with dementia who spent >100 days in a nursing home were prescribed an antipsychotic in 2012.44 A recent Canadian study using drug claims data found that overall psychotropic use in patients with dementia remains high, finding that three-fourths of all patients with dementia in long-term care are given at least 1 psychotropic, and up to one-third are prescribed SGAs.50 European data similarly show that antipsychotics continue to be prescribed to up to one-third of long-term care residents with dementia, with 7 out of 10 receiving an SGA.1

 

 

Conclusions

The Table provides a summary of the evidence regarding the use of antipsychotics in patients with dementia. Expert consensus is that among BPSD, aggression and psychosis are the primary indications for using antipsychotics.51 Based on multiple RCTs and meta-analyses, the evidence for using SGAs to treat these symptoms is moderate at best. However, in real-world practice settings, SGAs are widely used for symptoms, such as wandering, inappropriate behaviors, resistance to care, etc., for which there is no evidence for efficacy other than sedation. Furthermore, even when there is a potential for benefit, this must be balanced against the risk of adverse effects, including somnolence, worsened cognition, extra­pyramidal symptoms, stroke, and mortality.
 

Clinicians who care for older adults with BPSD should strive to increase the use of first-line nonpharmacologic strategies, by using structured approaches such as DICE (Describe, Investigate, Create, Evaluate) described in the Box.51 Antipsychotics should be reserved for situations in which nonpharmacologic approaches are unsuccessful, or there is concern for serious or imminent risk to the patient or others.

 

In the future, observational studies using biomarkers, such as neuroimaging markers, of brain health in older patients taking antipsychotics for various durations may give us a better understanding of long-term antipsychotic safety and tolerability and the monitoring required to assess long-term burden of specific antipsychotics in real-world samples.52 However, because of various biases, observational data may not provide answers to all questions,53 and a major challenge is that the number of published RCTs specific to geriatric patients is not growing substantially. Pharmacotherapy evidence is not keeping up with demographic trends. Key developments in RCTs will be the inclusion of biomarkers via neuroimaging, drug serum or brain levels, and genetic profiling. Because of the modest findings of benefits of antipsychotics in dementia and safety concerns addressing brain health in preclinical or early stages, identification of effective non-drug interventions and identifying true disease-modifying agents will be the next challenges of dementia research.
 

Bottom Line

Second-generation antipsychotics should be prescribed for patients with behavioral and psychological symptoms of dementia only when nonpharmacological approaches are unsuccessful, or there is an imminent risk to the patient or others because of aggression or psychosis.

Related Resources

  • Steinberg M, Lyketsos CG. Atypical antipsychotic use in patients with dementia: managing safety concerns. Am J Psychiatry. 2012;169(9):900-906.
  • Gitlin LN, Kales HC, Lyketsos CG. Nonpharmacologic management of behavioral symptoms in dementia. JAMA. 2012;308(19):2020-2029.

Drug Brand Names

Chlorpromazine Ormazine, Thorazine
Haloperidol Haldol
Olanzapine Zyprexa
Quetiapine Seroquel
Risperidone Risperdal
Valproic acid Depakene
Ziprasidone Geodon

References

1. Foebel AD, Liperoti R, Onder G, et al; SHELTER Study Investigators. Use of antipsychotic drugs among residents with dementia in European long-term care facilities: results from the SHELTER study. J Am Med Dir Assoc. 2014;15(12):911-917.
2. Foebel A, Ballokova A, Wellens NI, et al. A retrospective, longitudinal study of factors associated with new antipsychotic medication use among recently admitted long-term care residents. BMC Geriatr. 2015;15:128.
3. Parsons C, Johnston S, Mathie E, et al. Potentially inappropriate prescribing in older people with dementia in care homes: a retrospective analysis. Drugs Aging. 2012;29(2):143-155.
4. Vidal X, Agustí A, Vallano A, et al; Potentially Inappropriate Prescription in Older Patients in Spain (PIPOPS) Investigators’ project. Elderly patients treated with psychotropic medicines admitted to hospital: associated characteristics and inappropriate use. Eur J Clin Pharmacol. 2016;72(6):755-764.
5. Caron L, Cottencin O, Lapeyre-Mestre M, et al. Off-label prescribing of antipsychotics in adults, children and elderly individuals: a systematic review of recent prescription trends. Curr Pharm Des. 2015;21(23):3280-3297.
6. Weintraub D, Chiang C, Kim HM, et al. Association of antipsychotic use with mortality risk in patients with parkinson disease. JAMA Neurol. 2016;73(5):535-541.
7. Lyketsos CG, Carrillo MC, Ryan JM, et al. Neuropsychiatric symptoms in Alzheimer’s disease. Alzheimers Dement. 2011;7(5):532-539.
8. Kales HC, Chen P, Blow FC, et al. Rates of clinical depression diagnosis, functional impairment, and nursing home placement in coexisting dementia and depression. Am J Geriatr Psychiatry. 2005;13(6):441-449.
9. Yaffe K, Fox P, Newcomer R, et al. Patient and caregiver characteristics and nursing home placement in patients with dementia. JAMA. 2002;287(16):2090-2097.
10. Lopez OL, Becker JT, Chang YF, et al. The long-term effects of conventional and atypical antipsychotics in patients with probable Alzheimer’s disease. Am J Psychiatry. 2013;170(9):1051-1058.
11. Vilalta-Franch J, López-Pousa S, Calvó-Perxas L, et al. Psychosis of Alzheimer disease: prevalence, incidence, persistence, risk factors, and mortality. Am J Geriatr Psychiatry. 2013;21(11):1135-1143.
12. Spalletta G, Musicco M, Padovani A, et al. Neuropsychiatric symptoms and syndromes in a large cohort of newly diagnosed, untreated patients with Alzheimer disease. Am J Geriatr Psychiatry. 2010;18(11):1026-1035.
13. Steinberg M, Shao H, Zandi P, et al; Cache County Investigators. Point and 5-year period prevalence of neuropsychiatric symptoms in dementia: the Cache County Study. Int J Geriatr Psychiatry. 2008;23(2):170-177.
14. Finkel SI, Burns A. Behavioral and psychological symptoms of dementia (BPSD): a clinical and research update-introduction. International Psychogeriatrics. 2000;12:9-12.
15. Lyketsos CG. Neuropsychiatric symptoms (behavioral and psychological symptoms of dementia) and the development of dementia treatments. Int Psychogeriatr. 2007;19(3):409-420.
16. Kunik ME, Snow AL, Davila JA, et al. Causes of aggressive behavior in patients with dementia. J Clin Psychiatry. 2010;71(9):1145-1152.
17. Reus VI, Fochtmann LJ, Eyler AE, et al. The American Psychiatric Association practice guideline on the use of antipsychotics to treat agitation or psychosis in patients with dementia. Am J Psychiatry. 2016;173(5):543-546.
18. Kales HC, Gitlin LN, Lyketsos CG. Assessment and management of behavioral and psychological symptoms of dementia. BMJ. 2015;350:h369. doi: 10.1136/bmj.h369.
19. Schneider LS, Pollock VE, Lyness SA. A metaanalysis of controlled trials of neuroleptic treatment in dementia. J Am Geriatr Soc. 1990;38(5):553-563.
20. Yury CA, Fisher JE. Meta-analysis of the effectiveness of atypical antipsychotics for the treatment of behavioural problems in persons with dementia. Psychother Psychosom. 2007;76(4):213-218.
21. Schneider LS, Dagerman K, Insel PS. Efficacy and adverse effects of atypical antipsychotics for dementia: meta-analysis of randomized, placebo-controlled trials. Am J Geriatr Psychiatry. 2006;14(3):191-210.
22. Ballard CG, Waite J. The effectiveness of atypical antipsychotics for aggression and psychosis in Alzheimer’s disease. Cochrane Database Syst Rev. 2006:1:CD003476.
23. Sink KM, Holden KF, Yaffe K. Pharmacological treatment of neuropsychiatric symptoms of dementia: a review of the evidence. JAMA. 2005;293(5):596-608.
24. Aisen PS, Cummings J, Schneider LS. Symptomatic and nonamyloid/tau based pharmacologic treatment for Alzheimer disease. Cold Spring Harb Perspect Med. 2012;2(3):a006395. doi: 10.1101/cshperspect.a006395.
25. Schneider LS, Tariot PN, Dagerman KS, et al; CATIE-AD Study Group. Effectiveness of atypical antipsychotic drugs in patients with Alzheimer’s disease. N Engl J Med. 2006;355(15):1525-1538.
26. Trifirò G, Sultana J, Spina E. Are the safety profiles of antipsychotic drugs used in dementia the same? An updated review of observational studies. Drug Saf. 2014;37(7):501-520.
27. Trifirò G, Gambassi G, Sen EF, et al. Association of community-acquired pneumonia with antipsychotic drug use in elderly patients: a nested case-control study. Ann Intern Med. 2010;152(7):418-425, W139-W140.
28. Sultana J, Trifirò G. Drug safety warnings: a message in a bottle. Analysis. 2008;179:438-446.
29. Liperoti R, Gambassi G, Lapane KL, et al. Cerebrovascular events among elderly nursing home patients treated with conventional or atypical antipsychotics. J Clin Psychiatry. 2005;66(9):1090-1096.
30. U.S. Food and Drug Administration. Public health advisory: deaths with antipsychotics in elderly patients with behavioral disturbances. https://www.fda.gov/drugs/drugsafety/postmarketdrugsafety information forpatientsandproviders/ucm053171. Updated August 16, 2013. Accessed October 20, 2017.
31. Wang PS, Schneeweiss S, Avorn J, et al. Risk of death in elderly users of conventional vs. atypical antipsychotic medications. N Engl J Med. 2005;353(22):2335-2341.
32. Kales HC, Valenstein M, Kim HM, et al. Mortality risk in patients with dementia treated with antipsychotics versus other psychiatric medications. Am J Psychiatry. 2007;164(10):1568-1576; quiz 1623.
33. Simoni-Wastila L, Ryder PT, Qian J, et al. Association of antipsychotic use with hospital events and mortality among medicare beneficiaries residing in long-term care facilities. Am J Geriatr Psychiatry. 2009;17(5):417-427.
34. Raivio MM, Laurila JV, Strandberg TE, et al. Neither atypical nor conventional antipsychotics increase mortality or hospital admissions among elderly patients with dementia: a two-year prospective study. Am J Geriatr Psychiatry. 2007;15(5):416-424.
35. Gill SS, Bronskill SE, Normand SL, et al. Antipsychotic drug use and mortality in older adults with dementia. Ann Intern Med. 2007;146(11):775-786.
36. Schneeweiss S, Setoguchi S, Brookhart A, et al. Risk of death associated with the use of conventional versus atypical antipsychotic drugs among elderly patients. CMAJ. 2007;176(5):627-632.
37. Kales HC, Kim HM, Zivin K, et al. Risk of mortality among individual antipsychotics in patients with dementia. Am J Psychiatry. 2012;169(1):71-79.
38. Maust DT, Kim HM, Seyfried LS, et al. Antipsychotics, other psychotropics, and the risk of death in patients with dementia: number needed to harm. JAMA Psychiatry. 2015;72(5):438-445.
39. Rhee Y, Csernansky JG, Emanuel LL, et al. Psychotropic medication burden and factors associated with antipsychotic use: an analysis of a population-based sample of community-dwelling older persons with dementia. J Am Geriatr Soc. 2011;59(11):2100-2107.
40. Kales HC, Zivin K, Kim HM, et al. Trends in antipsychotic use in dementia 1999-2007. Arch Gen Psychiatry. 2011;68(2):190-197.
41. American Diabetes Association; American Psychiatric Association; American Association of Clinical Endocrinologists; North American Association for the Study of Obesity. Consensus development conference on antipsychotic drugs and obesity and diabetes. Diabetes Care. 2004;27(2):596-601.
42. Brodaty H, Ames D, Snowdon J, et al. A randomized placebo-controlled trial of risperidone for the treatment of aggression, agitation, and psychosis of dementia. J Clin Psychiatry. 2003;64(2):134-143.
43. Wooltorton E. Risperidone (Risperdal): increased rate of cerebrovascular events in dementia trials. CMAJ. 2002;167(11):1269-1270.
44. United States Government Accountability Office. Antipsychotic drug use: HHS has initiatives to reduce use among older adults in nursing homes, but should expand efforts to other settings. http://www.gao.gov/assets/670/668221.pdf. Published January 2015. Accessed October 20, 2017.
45. Chen Y, Briesacher BA, Field TS, et al. Unexplained variation across US nursing homes in antipsychotic prescribing rates. Arch Intern Med. 2010;170(1):89-95.
46. Feng Z, Hirdes JP, Smith TF, et al. Use of physical restraints and antipsychotic medications in nursing homes: a cross-national study. Int J Geriatr Psychiatry. 2009;24(10):1110-1118.
47. Kamble P, Chen H, Sherer J, et al. Antipsychotic drug use among elderly nursing home residents in the United States. Am J Geriatr Pharmacother. 2008;6(4):187-197.
48. Gellad WF, Aspinall SL, Handler SM, et al. Use of antipsychotics among older residents in VA nursing homes. Med Care. 2012;50(11):954-960.
49. Bonner A. Improving dementia care and reducing unnecessary use of antipsychotic medications in nursing homes. Center for Medicare and Medicaid Services. http://ltcombudsman.org/uploads/files/support/alice-bonner-slides.pdf. Published April 28, 2013. Accessed October 20, 2017.
50. Vasudev A, Shariff SZ, Liu K, et al. Trends in psychotropic dispensing among older adults with dementia living in long-term care facilities: 2004-2013. Am J Geriatr Psychiatry. 2015;23(12):1259-1269.
51. Kales HC, Gitlin LN, Lyketsos CG, et al; Detroit Expert Panel on Assessment and Management of Neuropsychiatric Symptoms of Dementia. Management of neuropsychiatric symptoms of dementia in clinical settings: recommendations from a multidisciplinary expert panel. J Am Geriatr Soc. 2014;62(4):762-769.
52. Andreasen NC, Liu D, Ziebell S, et al. Relapse duration, treatment intensity, and brain tissue loss in schizophrenia: a prospective longitudinal MRI study. Am J Psychiatry. 2013;170(6):609-615.
53. Mulsant BH. Challenges of the treatment of neuropsychiatric symptoms associated with dementia. Am J Geriatr Psychiatry. 2014;22(4):317-320.

Article PDF
Author and Disclosure Information

Helen C. Kales, MD
Professor of Psychiatry
Program for Positive Aging
Department of Psychiatry
University of Michigan
VA Center for Clinical Management Research
Ann Arbor, Michigan

Benoit H. Mulsant, MD, MS
rofessor and Chair
Department of Psychiatry
Senior Scientist
Centre for Addiction and Mental Health
University of Toronto
Toronto, Ontario

Martha Sajatovic, MD
Professor of Psychiatry and Professor of Neurology
Department of Psychiatry and Department of Neurology
Case Western Reserve University
University Hospitals Cleveland Medical Center
Cleveland, Ohio

Disclosures
Dr. Kales has received research support from the National Institutes of Health (NIH), Department of Defense, and Veterans Affairs, and reports no financial relationships with any companies whose products are mentioned in this article or with manufacturers of competing products. Dr. Mulsant has received research support from Brain Canada, the Centre for Addiction and Mental Health, the Canadian Institutes of Health Research, the NIH, Bristol-Myers Squibb (medications for an NIH-funded clinical trial), Eli Lilly (medications for an NIH-funded clinical trial), and Pfizer (medications for an NIH-funded clinical trial). Within the past 5 years, he also has received travel support from Roche. Dr. Sajatovic has received research grants from Alkermes, Merck, Janssen, Reuter Foundation, Woodruff Foundation, Reinberger Foundation, NIH, and the Centers for Disease Control and Prevention; has been a consultant to Bracket, Prophase, Otsuka, Sunovion, Supernus and Neurocrine; and has received royalties from Springer Press, Johns Hopkins University Press, Oxford Press, UpToDate, and Lexicomp, and compensation for CME activities from American Physician’s Institute, MCM Education, and CMEology.

Issue
December 2017
Publications
Topics
Page Number
24-30
Sections
Author and Disclosure Information

Helen C. Kales, MD
Professor of Psychiatry
Program for Positive Aging
Department of Psychiatry
University of Michigan
VA Center for Clinical Management Research
Ann Arbor, Michigan

Benoit H. Mulsant, MD, MS
rofessor and Chair
Department of Psychiatry
Senior Scientist
Centre for Addiction and Mental Health
University of Toronto
Toronto, Ontario

Martha Sajatovic, MD
Professor of Psychiatry and Professor of Neurology
Department of Psychiatry and Department of Neurology
Case Western Reserve University
University Hospitals Cleveland Medical Center
Cleveland, Ohio

Disclosures
Dr. Kales has received research support from the National Institutes of Health (NIH), Department of Defense, and Veterans Affairs, and reports no financial relationships with any companies whose products are mentioned in this article or with manufacturers of competing products. Dr. Mulsant has received research support from Brain Canada, the Centre for Addiction and Mental Health, the Canadian Institutes of Health Research, the NIH, Bristol-Myers Squibb (medications for an NIH-funded clinical trial), Eli Lilly (medications for an NIH-funded clinical trial), and Pfizer (medications for an NIH-funded clinical trial). Within the past 5 years, he also has received travel support from Roche. Dr. Sajatovic has received research grants from Alkermes, Merck, Janssen, Reuter Foundation, Woodruff Foundation, Reinberger Foundation, NIH, and the Centers for Disease Control and Prevention; has been a consultant to Bracket, Prophase, Otsuka, Sunovion, Supernus and Neurocrine; and has received royalties from Springer Press, Johns Hopkins University Press, Oxford Press, UpToDate, and Lexicomp, and compensation for CME activities from American Physician’s Institute, MCM Education, and CMEology.

Author and Disclosure Information

Helen C. Kales, MD
Professor of Psychiatry
Program for Positive Aging
Department of Psychiatry
University of Michigan
VA Center for Clinical Management Research
Ann Arbor, Michigan

Benoit H. Mulsant, MD, MS
rofessor and Chair
Department of Psychiatry
Senior Scientist
Centre for Addiction and Mental Health
University of Toronto
Toronto, Ontario

Martha Sajatovic, MD
Professor of Psychiatry and Professor of Neurology
Department of Psychiatry and Department of Neurology
Case Western Reserve University
University Hospitals Cleveland Medical Center
Cleveland, Ohio

Disclosures
Dr. Kales has received research support from the National Institutes of Health (NIH), Department of Defense, and Veterans Affairs, and reports no financial relationships with any companies whose products are mentioned in this article or with manufacturers of competing products. Dr. Mulsant has received research support from Brain Canada, the Centre for Addiction and Mental Health, the Canadian Institutes of Health Research, the NIH, Bristol-Myers Squibb (medications for an NIH-funded clinical trial), Eli Lilly (medications for an NIH-funded clinical trial), and Pfizer (medications for an NIH-funded clinical trial). Within the past 5 years, he also has received travel support from Roche. Dr. Sajatovic has received research grants from Alkermes, Merck, Janssen, Reuter Foundation, Woodruff Foundation, Reinberger Foundation, NIH, and the Centers for Disease Control and Prevention; has been a consultant to Bracket, Prophase, Otsuka, Sunovion, Supernus and Neurocrine; and has received royalties from Springer Press, Johns Hopkins University Press, Oxford Press, UpToDate, and Lexicomp, and compensation for CME activities from American Physician’s Institute, MCM Education, and CMEology.

Article PDF
Article PDF
 

According to the U.S. Department of Health and Human Services, in 2007, 88% of 1.4 million Medicare claims for second-generation antipsychotics (SGAs) in older adult nursing home residents were associated with a dementia diagnosis. Similar trends have been observed in Canada and Europe.1-4 In a retrospective analysis of medication data from older residents with dementia in 6 care homes in England, long-term (ie, >1 month) use of antipsychotics was the most frequent potentially inappropriate prescribing practice.3 In another study in 7 European countries and Israel, the overall prevalence of antipsychotic use among long-term care residents with dementia was 33%.1 Similarly, a recent literature review5 found that 22% to 86% of antipsychotic prescriptions to older individuals were off-label; this practice was particularly common for individuals with agitation.

Because of the aging population and widespread prescription of antipsychotics to older patients, clinicians need information on the relative risks of using these medications in this population. In the United States, all antipsychotics carry a FDA “black-box” warning of the increased risk of death in older adults with dementia. In addition, the risk of death is increased when prescribing antipsychotics to older adults with other conditions, such as Parkinson’s disease,6 and other safety and tolerability concerns, including falls and fractures, sedation, metabolic abnormalities, and extrapyramidal effects, are highly relevant to geriatric patients.

This 3-part review summarizes findings and recommendations on prescribing antipsychotics to older individuals with schizophrenia, bipolar disorder, depression, and dementia. This third and final installment:

  • briefly summarizes the major studies and analyses relevant to prescribing antipsychotics to older patients with dementia
  • provides a summative opinion on safety and tolerability issues in these older patients
  • highlights the gaps in the evidence base and areas that need additional research.


Summary of benefits, place in treatment armamentarium

Behavioral and psychological symptoms of dementia (BPSD) include agitation, delusional beliefs, repetitive questioning, hallucinations, aggression, wandering, and various socially inappropriate behaviors.7 These occur almost universally in all types and stages of dementia.7 BPSD are among the most complex, stressful, and costly aspects of dementia care, and lead to a myriad of poor health outcomes, including excess morbidity, mortality, hospital stays, and early nursing home placement.8-11 Because BPSD usually occur across all types and stages of dementia,7,12-16 the prevalence of BPSD mirrors the overall prevalence of dementia.

Although all expert organizations, including the American Psychiatric Association,17 recommend nonpharmacologic strategies as first-line treatment for BPSD, for the most part, these recommendations have not been translated into standard clinical management or routine care.18 Because of a perceived lack of other options, the current mainstay of treatment is the off-label use of psychotropics such as antipsy­chotics. Of all the agents currently used for BPSD, SGAs have the strongest evidence base, although benefits are modest at best (standardized effect size 0.13 to 0.16).19,20 In terms of individual SGAs, only risperidone is indicated for aggression in Canada and in Europe (not in the United States); risperidone has the best evidence for efficacy, with a meta-analysis of 5 published randomized controlled trials (RCTs) reporting that risperidone is superior to other SGAs for aggression in dementia.21,22 As a class, first-generation antipsychotics (FGAs) have no clear evidence for BPSD as defined broadly; however, there may be slight benefit for haloperidol for aggression.23,24


Clinical Trials

Adverse effects. A meta-analysis of RCTs of SGAs found that, compared with placebo, SGAs have increased rates of several adverse effects. These include somnolence (17% drug vs 7% placebo; odds ratio [OR], 2.84; 95% confidence interval [CI], 2.25 to 3.58; P < .00001); extrapyramidal symptoms (13% drug vs 8% placebo; OR, 1.51; 95% CI, 1.20 to 1.91; P = .0005; primarily attributable to risperidone); abnormal gait (10% drug vs 2% placebo; OR, 3.42; 95% CI, 1.78 to 6.56; P = .0002; attributable to olanzapine and risperidone); edema (9% drug vs 4% placebo; OR, 1.99; 95% CI, 1.20 to 3.30; P = .008; attributable to olanzapine and risperidone); urinary tract infections/incontinence (16% drug vs 12% placebo; OR, 1.28; 95% CI, 1.02 to 1.61; P = .04); cognitive impairment measured as difference in Mini-Mental State Examination score (95% CI, 0.38 to 1.09; P < .0001)25; and stroke (1.9% drug vs 0.9% placebo, OR, 2.13; 95% CI, 1.20 to 3.75; P = .009).21,26

In the 42-site Clinical Antipsychotic Trials of Intervention Effectiveness Alzheimer’s disease RCT, 421 outpatients with Alzheimer’s disease and BPSD were randomized to an SGA (risperidone, olanzapine, or quetiapine) or placebo. Compared with placebo, SGAs had a higher rate of parkinsonism or extrapyramidal signs (olanzapine and risperidone groups); sedation; confusion/changes in mental status (olanzapine and risperidone); psychotic symptoms (olanzapine); and increase in body weight and body mass index.26

In the 2005 FDA black-box warning, pneumonia and cardiac adverse effects were cited as primary causes of death for patients with dementia taking SGAs. A subsequent observational study confirmed that use of either FGAs or SGAs in geriatric patients was associated with an increased risk of pneumonia, in a dose-dependent manner.27 Although there is limited data on cardiac adverse effects in older adults, especially those with dementia taking antipsychotics,28 1 observational study of nursing home residents29 found that those taking FGAs had a significantly higher risk of hospitalization for ventricular arrhythmia or cardiac arrest compared with those who were not taking FGAs. In contrast, there was no increased risk with SGAs.

Mortality.
 In 2005, the FDA announced that based on a reanalysis of 17 placebo-controlled trials (many of which were unpublished) that SGAs were associated with a 1.7-fold increase in mortality compared with placebo.30 As a result, the FDA issued a black-box warning for using SGAs in patients with dementia. The overall OR in a published meta-analysis of mortality with SGAs was 1.54 (1.06 to 2.23; z = 2.28; P = .02), with pooled events of 3.5% mortality vs 2.3% (drug vs placebo).21 This meta-analysis21 also included ad hoc analyses of haloperidol; using combined data from 2 contrasts of haloperidol (with risperidone and quetiapine; 243 patients receiving haloperidol and 239 receiving placebo) they also found 15 deaths (6.2%) with haloperidol and 9 (3.8%) with placebo, resulting in an OR of 1.68.

 

 

 

Other clinical data

Observational studies. Most observational studies have confirmed concerns regarding increased mortality in patients with BPSD who take antipsychotics, with FGAs having a higher risk than SGAs18,31 and SGAs having a higher risk compared with most other psychotropics.32 Three studies that found no increase in mortality with antipsychotics in patients with dementia had methodological issues, including examining prevalence as opposed to new users,33,34 not controlling for exposure,10,33,34 power issues,10,34 not controlling for other psychiatric medications,10 and varying lengths of follow-up.10 An FDA black-box warning for FGAs was announced in 200830 based on 2 observational studies that showed an increased risk of mortality in older adults taking FGAs vs SGAs.35,36

In terms of specific SGAs, Kales et al37 examined the mortality risk associated with individual antipsychotics using various methods to control for confounding. Among a national sample of >33,000 older veterans with dementia newly started on haloperidol, risperidone, olanzapine, quetiapine, or valproic acid and derivatives (as a nonantipsychotic comparator), the highest mortality across all analyses (intent to treat, exposure, propensity-adjusted) was associated with haloperidol, followed by risperidone and olanzapine, valproic acid, and quetiapine.

Most recently, a retrospective case-control study (90,786 patients age ≥65 with dementia) examined the number needed to harm (NNH; ie, number of patients needed to receive treatment that would result in 1 death) over 180 days following initiation of an FGA or SGA.38 This study found the following NNHs: haloperidol, 26 (95% CI, 15 to 99); risperidone, 27 (95% CI, 19 to 46); olanzapine, 40 (95% CI, 21 to 312); and quetiapine, 50 (95% CI, 30 to 150).38 These results are congruent with a review of observational studies that found the highest risk of mortality was associated with haloperidol and chlorpromazine, and the lowest risk with olanzapine, quetiapine, and ziprasidone.28

Patterns of antipsychotic use in older dementia patients

There are high rates of antipsychotic use in patients with dementia. Before the FDA issued the black-box warning, the Aging Demographics and Memory study found that the rate of antipsychotic use in community (outpatient) older adults with dementia was approximately 19% between 2002 and 2004 in a representative sample of 307 older adults.39 Another study examining trends in community antipsychotic use in the U.S. Department of Veterans Affairs (VA) found that in the 1990s, SGA use was increasing; approximately 18% of outpatients with dementia were taking these agents.40 Use of SGAs began to decline in 2003, ahead of the 2005 black-box warning, in tandem with other advisories (eg, diabetes, metabolic syndrome,41 and stroke risk).42,43 Olanzapine and risperidone showed declining rates between 2003 and 2005, whereas quetiapine use significantly increased during this period. All 3 SGAs declined after the black-box warning. However, by the end of 2007, the use of SGAs had leveled off to approximately 12% of VA patients with dementia. A recent U.S. Government Accountability Office (GAO) report found that in 2012, 14% of older adult Medicare Part D enrollees with dementia living in the community were prescribed an antipsychotic.44

Use in nursing home residents. Because BPSD are one of the main reasons people with dementia are placed in nursing homes, it is not surprising that rates of antipsychotic use are higher in these settings than in the community. Prior to the black-box warning, studies found that 24% to 32% of nursing home residents were treated with antipsychotics.45-47 A study examining VA nursing homes (n = 133 facilities, n = 3,692 veterans) found that approximately 26% of residents were prescribed antipsychotics in 2004 to 2005.48 The Center for Medicare and Medicaid Services (CMS) National Partnership to Improve Dementia Care in Nursing Homes has appeared to lower antipsychotic medication use in nursing homes; the rate decreased from 24% in long-stay nursing home residents nationwide in 2011 to 19% by the end of 2014. Specific to dementia, a 2010 CMS report49 indicated that approximately 40% of nursing home residents with cognitive impairment and behavioral issues, without psychosis, received antipsychotics. The GAO data indicated that approximately 33% of older Medicare Part D enrollees with dementia who spent >100 days in a nursing home were prescribed an antipsychotic in 2012.44 A recent Canadian study using drug claims data found that overall psychotropic use in patients with dementia remains high, finding that three-fourths of all patients with dementia in long-term care are given at least 1 psychotropic, and up to one-third are prescribed SGAs.50 European data similarly show that antipsychotics continue to be prescribed to up to one-third of long-term care residents with dementia, with 7 out of 10 receiving an SGA.1

 

 

Conclusions

The Table provides a summary of the evidence regarding the use of antipsychotics in patients with dementia. Expert consensus is that among BPSD, aggression and psychosis are the primary indications for using antipsychotics.51 Based on multiple RCTs and meta-analyses, the evidence for using SGAs to treat these symptoms is moderate at best. However, in real-world practice settings, SGAs are widely used for symptoms, such as wandering, inappropriate behaviors, resistance to care, etc., for which there is no evidence for efficacy other than sedation. Furthermore, even when there is a potential for benefit, this must be balanced against the risk of adverse effects, including somnolence, worsened cognition, extra­pyramidal symptoms, stroke, and mortality.
 

Clinicians who care for older adults with BPSD should strive to increase the use of first-line nonpharmacologic strategies, by using structured approaches such as DICE (Describe, Investigate, Create, Evaluate) described in the Box.51 Antipsychotics should be reserved for situations in which nonpharmacologic approaches are unsuccessful, or there is concern for serious or imminent risk to the patient or others.

 

In the future, observational studies using biomarkers, such as neuroimaging markers, of brain health in older patients taking antipsychotics for various durations may give us a better understanding of long-term antipsychotic safety and tolerability and the monitoring required to assess long-term burden of specific antipsychotics in real-world samples.52 However, because of various biases, observational data may not provide answers to all questions,53 and a major challenge is that the number of published RCTs specific to geriatric patients is not growing substantially. Pharmacotherapy evidence is not keeping up with demographic trends. Key developments in RCTs will be the inclusion of biomarkers via neuroimaging, drug serum or brain levels, and genetic profiling. Because of the modest findings of benefits of antipsychotics in dementia and safety concerns addressing brain health in preclinical or early stages, identification of effective non-drug interventions and identifying true disease-modifying agents will be the next challenges of dementia research.
 

Bottom Line

Second-generation antipsychotics should be prescribed for patients with behavioral and psychological symptoms of dementia only when nonpharmacological approaches are unsuccessful, or there is an imminent risk to the patient or others because of aggression or psychosis.

Related Resources

  • Steinberg M, Lyketsos CG. Atypical antipsychotic use in patients with dementia: managing safety concerns. Am J Psychiatry. 2012;169(9):900-906.
  • Gitlin LN, Kales HC, Lyketsos CG. Nonpharmacologic management of behavioral symptoms in dementia. JAMA. 2012;308(19):2020-2029.

Drug Brand Names

Chlorpromazine Ormazine, Thorazine
Haloperidol Haldol
Olanzapine Zyprexa
Quetiapine Seroquel
Risperidone Risperdal
Valproic acid Depakene
Ziprasidone Geodon

 

According to the U.S. Department of Health and Human Services, in 2007, 88% of 1.4 million Medicare claims for second-generation antipsychotics (SGAs) in older adult nursing home residents were associated with a dementia diagnosis. Similar trends have been observed in Canada and Europe.1-4 In a retrospective analysis of medication data from older residents with dementia in 6 care homes in England, long-term (ie, >1 month) use of antipsychotics was the most frequent potentially inappropriate prescribing practice.3 In another study in 7 European countries and Israel, the overall prevalence of antipsychotic use among long-term care residents with dementia was 33%.1 Similarly, a recent literature review5 found that 22% to 86% of antipsychotic prescriptions to older individuals were off-label; this practice was particularly common for individuals with agitation.

Because of the aging population and widespread prescription of antipsychotics to older patients, clinicians need information on the relative risks of using these medications in this population. In the United States, all antipsychotics carry a FDA “black-box” warning of the increased risk of death in older adults with dementia. In addition, the risk of death is increased when prescribing antipsychotics to older adults with other conditions, such as Parkinson’s disease,6 and other safety and tolerability concerns, including falls and fractures, sedation, metabolic abnormalities, and extrapyramidal effects, are highly relevant to geriatric patients.

This 3-part review summarizes findings and recommendations on prescribing antipsychotics to older individuals with schizophrenia, bipolar disorder, depression, and dementia. This third and final installment:

  • briefly summarizes the major studies and analyses relevant to prescribing antipsychotics to older patients with dementia
  • provides a summative opinion on safety and tolerability issues in these older patients
  • highlights the gaps in the evidence base and areas that need additional research.


Summary of benefits, place in treatment armamentarium

Behavioral and psychological symptoms of dementia (BPSD) include agitation, delusional beliefs, repetitive questioning, hallucinations, aggression, wandering, and various socially inappropriate behaviors.7 These occur almost universally in all types and stages of dementia.7 BPSD are among the most complex, stressful, and costly aspects of dementia care, and lead to a myriad of poor health outcomes, including excess morbidity, mortality, hospital stays, and early nursing home placement.8-11 Because BPSD usually occur across all types and stages of dementia,7,12-16 the prevalence of BPSD mirrors the overall prevalence of dementia.

Although all expert organizations, including the American Psychiatric Association,17 recommend nonpharmacologic strategies as first-line treatment for BPSD, for the most part, these recommendations have not been translated into standard clinical management or routine care.18 Because of a perceived lack of other options, the current mainstay of treatment is the off-label use of psychotropics such as antipsy­chotics. Of all the agents currently used for BPSD, SGAs have the strongest evidence base, although benefits are modest at best (standardized effect size 0.13 to 0.16).19,20 In terms of individual SGAs, only risperidone is indicated for aggression in Canada and in Europe (not in the United States); risperidone has the best evidence for efficacy, with a meta-analysis of 5 published randomized controlled trials (RCTs) reporting that risperidone is superior to other SGAs for aggression in dementia.21,22 As a class, first-generation antipsychotics (FGAs) have no clear evidence for BPSD as defined broadly; however, there may be slight benefit for haloperidol for aggression.23,24


Clinical Trials

Adverse effects. A meta-analysis of RCTs of SGAs found that, compared with placebo, SGAs have increased rates of several adverse effects. These include somnolence (17% drug vs 7% placebo; odds ratio [OR], 2.84; 95% confidence interval [CI], 2.25 to 3.58; P < .00001); extrapyramidal symptoms (13% drug vs 8% placebo; OR, 1.51; 95% CI, 1.20 to 1.91; P = .0005; primarily attributable to risperidone); abnormal gait (10% drug vs 2% placebo; OR, 3.42; 95% CI, 1.78 to 6.56; P = .0002; attributable to olanzapine and risperidone); edema (9% drug vs 4% placebo; OR, 1.99; 95% CI, 1.20 to 3.30; P = .008; attributable to olanzapine and risperidone); urinary tract infections/incontinence (16% drug vs 12% placebo; OR, 1.28; 95% CI, 1.02 to 1.61; P = .04); cognitive impairment measured as difference in Mini-Mental State Examination score (95% CI, 0.38 to 1.09; P < .0001)25; and stroke (1.9% drug vs 0.9% placebo, OR, 2.13; 95% CI, 1.20 to 3.75; P = .009).21,26

In the 42-site Clinical Antipsychotic Trials of Intervention Effectiveness Alzheimer’s disease RCT, 421 outpatients with Alzheimer’s disease and BPSD were randomized to an SGA (risperidone, olanzapine, or quetiapine) or placebo. Compared with placebo, SGAs had a higher rate of parkinsonism or extrapyramidal signs (olanzapine and risperidone groups); sedation; confusion/changes in mental status (olanzapine and risperidone); psychotic symptoms (olanzapine); and increase in body weight and body mass index.26

In the 2005 FDA black-box warning, pneumonia and cardiac adverse effects were cited as primary causes of death for patients with dementia taking SGAs. A subsequent observational study confirmed that use of either FGAs or SGAs in geriatric patients was associated with an increased risk of pneumonia, in a dose-dependent manner.27 Although there is limited data on cardiac adverse effects in older adults, especially those with dementia taking antipsychotics,28 1 observational study of nursing home residents29 found that those taking FGAs had a significantly higher risk of hospitalization for ventricular arrhythmia or cardiac arrest compared with those who were not taking FGAs. In contrast, there was no increased risk with SGAs.

Mortality.
 In 2005, the FDA announced that based on a reanalysis of 17 placebo-controlled trials (many of which were unpublished) that SGAs were associated with a 1.7-fold increase in mortality compared with placebo.30 As a result, the FDA issued a black-box warning for using SGAs in patients with dementia. The overall OR in a published meta-analysis of mortality with SGAs was 1.54 (1.06 to 2.23; z = 2.28; P = .02), with pooled events of 3.5% mortality vs 2.3% (drug vs placebo).21 This meta-analysis21 also included ad hoc analyses of haloperidol; using combined data from 2 contrasts of haloperidol (with risperidone and quetiapine; 243 patients receiving haloperidol and 239 receiving placebo) they also found 15 deaths (6.2%) with haloperidol and 9 (3.8%) with placebo, resulting in an OR of 1.68.

 

 

 

Other clinical data

Observational studies. Most observational studies have confirmed concerns regarding increased mortality in patients with BPSD who take antipsychotics, with FGAs having a higher risk than SGAs18,31 and SGAs having a higher risk compared with most other psychotropics.32 Three studies that found no increase in mortality with antipsychotics in patients with dementia had methodological issues, including examining prevalence as opposed to new users,33,34 not controlling for exposure,10,33,34 power issues,10,34 not controlling for other psychiatric medications,10 and varying lengths of follow-up.10 An FDA black-box warning for FGAs was announced in 200830 based on 2 observational studies that showed an increased risk of mortality in older adults taking FGAs vs SGAs.35,36

In terms of specific SGAs, Kales et al37 examined the mortality risk associated with individual antipsychotics using various methods to control for confounding. Among a national sample of >33,000 older veterans with dementia newly started on haloperidol, risperidone, olanzapine, quetiapine, or valproic acid and derivatives (as a nonantipsychotic comparator), the highest mortality across all analyses (intent to treat, exposure, propensity-adjusted) was associated with haloperidol, followed by risperidone and olanzapine, valproic acid, and quetiapine.

Most recently, a retrospective case-control study (90,786 patients age ≥65 with dementia) examined the number needed to harm (NNH; ie, number of patients needed to receive treatment that would result in 1 death) over 180 days following initiation of an FGA or SGA.38 This study found the following NNHs: haloperidol, 26 (95% CI, 15 to 99); risperidone, 27 (95% CI, 19 to 46); olanzapine, 40 (95% CI, 21 to 312); and quetiapine, 50 (95% CI, 30 to 150).38 These results are congruent with a review of observational studies that found the highest risk of mortality was associated with haloperidol and chlorpromazine, and the lowest risk with olanzapine, quetiapine, and ziprasidone.28

Patterns of antipsychotic use in older dementia patients

There are high rates of antipsychotic use in patients with dementia. Before the FDA issued the black-box warning, the Aging Demographics and Memory study found that the rate of antipsychotic use in community (outpatient) older adults with dementia was approximately 19% between 2002 and 2004 in a representative sample of 307 older adults.39 Another study examining trends in community antipsychotic use in the U.S. Department of Veterans Affairs (VA) found that in the 1990s, SGA use was increasing; approximately 18% of outpatients with dementia were taking these agents.40 Use of SGAs began to decline in 2003, ahead of the 2005 black-box warning, in tandem with other advisories (eg, diabetes, metabolic syndrome,41 and stroke risk).42,43 Olanzapine and risperidone showed declining rates between 2003 and 2005, whereas quetiapine use significantly increased during this period. All 3 SGAs declined after the black-box warning. However, by the end of 2007, the use of SGAs had leveled off to approximately 12% of VA patients with dementia. A recent U.S. Government Accountability Office (GAO) report found that in 2012, 14% of older adult Medicare Part D enrollees with dementia living in the community were prescribed an antipsychotic.44

Use in nursing home residents. Because BPSD are one of the main reasons people with dementia are placed in nursing homes, it is not surprising that rates of antipsychotic use are higher in these settings than in the community. Prior to the black-box warning, studies found that 24% to 32% of nursing home residents were treated with antipsychotics.45-47 A study examining VA nursing homes (n = 133 facilities, n = 3,692 veterans) found that approximately 26% of residents were prescribed antipsychotics in 2004 to 2005.48 The Center for Medicare and Medicaid Services (CMS) National Partnership to Improve Dementia Care in Nursing Homes has appeared to lower antipsychotic medication use in nursing homes; the rate decreased from 24% in long-stay nursing home residents nationwide in 2011 to 19% by the end of 2014. Specific to dementia, a 2010 CMS report49 indicated that approximately 40% of nursing home residents with cognitive impairment and behavioral issues, without psychosis, received antipsychotics. The GAO data indicated that approximately 33% of older Medicare Part D enrollees with dementia who spent >100 days in a nursing home were prescribed an antipsychotic in 2012.44 A recent Canadian study using drug claims data found that overall psychotropic use in patients with dementia remains high, finding that three-fourths of all patients with dementia in long-term care are given at least 1 psychotropic, and up to one-third are prescribed SGAs.50 European data similarly show that antipsychotics continue to be prescribed to up to one-third of long-term care residents with dementia, with 7 out of 10 receiving an SGA.1

 

 

Conclusions

The Table provides a summary of the evidence regarding the use of antipsychotics in patients with dementia. Expert consensus is that among BPSD, aggression and psychosis are the primary indications for using antipsychotics.51 Based on multiple RCTs and meta-analyses, the evidence for using SGAs to treat these symptoms is moderate at best. However, in real-world practice settings, SGAs are widely used for symptoms, such as wandering, inappropriate behaviors, resistance to care, etc., for which there is no evidence for efficacy other than sedation. Furthermore, even when there is a potential for benefit, this must be balanced against the risk of adverse effects, including somnolence, worsened cognition, extra­pyramidal symptoms, stroke, and mortality.
 

Clinicians who care for older adults with BPSD should strive to increase the use of first-line nonpharmacologic strategies, by using structured approaches such as DICE (Describe, Investigate, Create, Evaluate) described in the Box.51 Antipsychotics should be reserved for situations in which nonpharmacologic approaches are unsuccessful, or there is concern for serious or imminent risk to the patient or others.

 

In the future, observational studies using biomarkers, such as neuroimaging markers, of brain health in older patients taking antipsychotics for various durations may give us a better understanding of long-term antipsychotic safety and tolerability and the monitoring required to assess long-term burden of specific antipsychotics in real-world samples.52 However, because of various biases, observational data may not provide answers to all questions,53 and a major challenge is that the number of published RCTs specific to geriatric patients is not growing substantially. Pharmacotherapy evidence is not keeping up with demographic trends. Key developments in RCTs will be the inclusion of biomarkers via neuroimaging, drug serum or brain levels, and genetic profiling. Because of the modest findings of benefits of antipsychotics in dementia and safety concerns addressing brain health in preclinical or early stages, identification of effective non-drug interventions and identifying true disease-modifying agents will be the next challenges of dementia research.
 

Bottom Line

Second-generation antipsychotics should be prescribed for patients with behavioral and psychological symptoms of dementia only when nonpharmacological approaches are unsuccessful, or there is an imminent risk to the patient or others because of aggression or psychosis.

Related Resources

  • Steinberg M, Lyketsos CG. Atypical antipsychotic use in patients with dementia: managing safety concerns. Am J Psychiatry. 2012;169(9):900-906.
  • Gitlin LN, Kales HC, Lyketsos CG. Nonpharmacologic management of behavioral symptoms in dementia. JAMA. 2012;308(19):2020-2029.

Drug Brand Names

Chlorpromazine Ormazine, Thorazine
Haloperidol Haldol
Olanzapine Zyprexa
Quetiapine Seroquel
Risperidone Risperdal
Valproic acid Depakene
Ziprasidone Geodon

References

1. Foebel AD, Liperoti R, Onder G, et al; SHELTER Study Investigators. Use of antipsychotic drugs among residents with dementia in European long-term care facilities: results from the SHELTER study. J Am Med Dir Assoc. 2014;15(12):911-917.
2. Foebel A, Ballokova A, Wellens NI, et al. A retrospective, longitudinal study of factors associated with new antipsychotic medication use among recently admitted long-term care residents. BMC Geriatr. 2015;15:128.
3. Parsons C, Johnston S, Mathie E, et al. Potentially inappropriate prescribing in older people with dementia in care homes: a retrospective analysis. Drugs Aging. 2012;29(2):143-155.
4. Vidal X, Agustí A, Vallano A, et al; Potentially Inappropriate Prescription in Older Patients in Spain (PIPOPS) Investigators’ project. Elderly patients treated with psychotropic medicines admitted to hospital: associated characteristics and inappropriate use. Eur J Clin Pharmacol. 2016;72(6):755-764.
5. Caron L, Cottencin O, Lapeyre-Mestre M, et al. Off-label prescribing of antipsychotics in adults, children and elderly individuals: a systematic review of recent prescription trends. Curr Pharm Des. 2015;21(23):3280-3297.
6. Weintraub D, Chiang C, Kim HM, et al. Association of antipsychotic use with mortality risk in patients with parkinson disease. JAMA Neurol. 2016;73(5):535-541.
7. Lyketsos CG, Carrillo MC, Ryan JM, et al. Neuropsychiatric symptoms in Alzheimer’s disease. Alzheimers Dement. 2011;7(5):532-539.
8. Kales HC, Chen P, Blow FC, et al. Rates of clinical depression diagnosis, functional impairment, and nursing home placement in coexisting dementia and depression. Am J Geriatr Psychiatry. 2005;13(6):441-449.
9. Yaffe K, Fox P, Newcomer R, et al. Patient and caregiver characteristics and nursing home placement in patients with dementia. JAMA. 2002;287(16):2090-2097.
10. Lopez OL, Becker JT, Chang YF, et al. The long-term effects of conventional and atypical antipsychotics in patients with probable Alzheimer’s disease. Am J Psychiatry. 2013;170(9):1051-1058.
11. Vilalta-Franch J, López-Pousa S, Calvó-Perxas L, et al. Psychosis of Alzheimer disease: prevalence, incidence, persistence, risk factors, and mortality. Am J Geriatr Psychiatry. 2013;21(11):1135-1143.
12. Spalletta G, Musicco M, Padovani A, et al. Neuropsychiatric symptoms and syndromes in a large cohort of newly diagnosed, untreated patients with Alzheimer disease. Am J Geriatr Psychiatry. 2010;18(11):1026-1035.
13. Steinberg M, Shao H, Zandi P, et al; Cache County Investigators. Point and 5-year period prevalence of neuropsychiatric symptoms in dementia: the Cache County Study. Int J Geriatr Psychiatry. 2008;23(2):170-177.
14. Finkel SI, Burns A. Behavioral and psychological symptoms of dementia (BPSD): a clinical and research update-introduction. International Psychogeriatrics. 2000;12:9-12.
15. Lyketsos CG. Neuropsychiatric symptoms (behavioral and psychological symptoms of dementia) and the development of dementia treatments. Int Psychogeriatr. 2007;19(3):409-420.
16. Kunik ME, Snow AL, Davila JA, et al. Causes of aggressive behavior in patients with dementia. J Clin Psychiatry. 2010;71(9):1145-1152.
17. Reus VI, Fochtmann LJ, Eyler AE, et al. The American Psychiatric Association practice guideline on the use of antipsychotics to treat agitation or psychosis in patients with dementia. Am J Psychiatry. 2016;173(5):543-546.
18. Kales HC, Gitlin LN, Lyketsos CG. Assessment and management of behavioral and psychological symptoms of dementia. BMJ. 2015;350:h369. doi: 10.1136/bmj.h369.
19. Schneider LS, Pollock VE, Lyness SA. A metaanalysis of controlled trials of neuroleptic treatment in dementia. J Am Geriatr Soc. 1990;38(5):553-563.
20. Yury CA, Fisher JE. Meta-analysis of the effectiveness of atypical antipsychotics for the treatment of behavioural problems in persons with dementia. Psychother Psychosom. 2007;76(4):213-218.
21. Schneider LS, Dagerman K, Insel PS. Efficacy and adverse effects of atypical antipsychotics for dementia: meta-analysis of randomized, placebo-controlled trials. Am J Geriatr Psychiatry. 2006;14(3):191-210.
22. Ballard CG, Waite J. The effectiveness of atypical antipsychotics for aggression and psychosis in Alzheimer’s disease. Cochrane Database Syst Rev. 2006:1:CD003476.
23. Sink KM, Holden KF, Yaffe K. Pharmacological treatment of neuropsychiatric symptoms of dementia: a review of the evidence. JAMA. 2005;293(5):596-608.
24. Aisen PS, Cummings J, Schneider LS. Symptomatic and nonamyloid/tau based pharmacologic treatment for Alzheimer disease. Cold Spring Harb Perspect Med. 2012;2(3):a006395. doi: 10.1101/cshperspect.a006395.
25. Schneider LS, Tariot PN, Dagerman KS, et al; CATIE-AD Study Group. Effectiveness of atypical antipsychotic drugs in patients with Alzheimer’s disease. N Engl J Med. 2006;355(15):1525-1538.
26. Trifirò G, Sultana J, Spina E. Are the safety profiles of antipsychotic drugs used in dementia the same? An updated review of observational studies. Drug Saf. 2014;37(7):501-520.
27. Trifirò G, Gambassi G, Sen EF, et al. Association of community-acquired pneumonia with antipsychotic drug use in elderly patients: a nested case-control study. Ann Intern Med. 2010;152(7):418-425, W139-W140.
28. Sultana J, Trifirò G. Drug safety warnings: a message in a bottle. Analysis. 2008;179:438-446.
29. Liperoti R, Gambassi G, Lapane KL, et al. Cerebrovascular events among elderly nursing home patients treated with conventional or atypical antipsychotics. J Clin Psychiatry. 2005;66(9):1090-1096.
30. U.S. Food and Drug Administration. Public health advisory: deaths with antipsychotics in elderly patients with behavioral disturbances. https://www.fda.gov/drugs/drugsafety/postmarketdrugsafety information forpatientsandproviders/ucm053171. Updated August 16, 2013. Accessed October 20, 2017.
31. Wang PS, Schneeweiss S, Avorn J, et al. Risk of death in elderly users of conventional vs. atypical antipsychotic medications. N Engl J Med. 2005;353(22):2335-2341.
32. Kales HC, Valenstein M, Kim HM, et al. Mortality risk in patients with dementia treated with antipsychotics versus other psychiatric medications. Am J Psychiatry. 2007;164(10):1568-1576; quiz 1623.
33. Simoni-Wastila L, Ryder PT, Qian J, et al. Association of antipsychotic use with hospital events and mortality among medicare beneficiaries residing in long-term care facilities. Am J Geriatr Psychiatry. 2009;17(5):417-427.
34. Raivio MM, Laurila JV, Strandberg TE, et al. Neither atypical nor conventional antipsychotics increase mortality or hospital admissions among elderly patients with dementia: a two-year prospective study. Am J Geriatr Psychiatry. 2007;15(5):416-424.
35. Gill SS, Bronskill SE, Normand SL, et al. Antipsychotic drug use and mortality in older adults with dementia. Ann Intern Med. 2007;146(11):775-786.
36. Schneeweiss S, Setoguchi S, Brookhart A, et al. Risk of death associated with the use of conventional versus atypical antipsychotic drugs among elderly patients. CMAJ. 2007;176(5):627-632.
37. Kales HC, Kim HM, Zivin K, et al. Risk of mortality among individual antipsychotics in patients with dementia. Am J Psychiatry. 2012;169(1):71-79.
38. Maust DT, Kim HM, Seyfried LS, et al. Antipsychotics, other psychotropics, and the risk of death in patients with dementia: number needed to harm. JAMA Psychiatry. 2015;72(5):438-445.
39. Rhee Y, Csernansky JG, Emanuel LL, et al. Psychotropic medication burden and factors associated with antipsychotic use: an analysis of a population-based sample of community-dwelling older persons with dementia. J Am Geriatr Soc. 2011;59(11):2100-2107.
40. Kales HC, Zivin K, Kim HM, et al. Trends in antipsychotic use in dementia 1999-2007. Arch Gen Psychiatry. 2011;68(2):190-197.
41. American Diabetes Association; American Psychiatric Association; American Association of Clinical Endocrinologists; North American Association for the Study of Obesity. Consensus development conference on antipsychotic drugs and obesity and diabetes. Diabetes Care. 2004;27(2):596-601.
42. Brodaty H, Ames D, Snowdon J, et al. A randomized placebo-controlled trial of risperidone for the treatment of aggression, agitation, and psychosis of dementia. J Clin Psychiatry. 2003;64(2):134-143.
43. Wooltorton E. Risperidone (Risperdal): increased rate of cerebrovascular events in dementia trials. CMAJ. 2002;167(11):1269-1270.
44. United States Government Accountability Office. Antipsychotic drug use: HHS has initiatives to reduce use among older adults in nursing homes, but should expand efforts to other settings. http://www.gao.gov/assets/670/668221.pdf. Published January 2015. Accessed October 20, 2017.
45. Chen Y, Briesacher BA, Field TS, et al. Unexplained variation across US nursing homes in antipsychotic prescribing rates. Arch Intern Med. 2010;170(1):89-95.
46. Feng Z, Hirdes JP, Smith TF, et al. Use of physical restraints and antipsychotic medications in nursing homes: a cross-national study. Int J Geriatr Psychiatry. 2009;24(10):1110-1118.
47. Kamble P, Chen H, Sherer J, et al. Antipsychotic drug use among elderly nursing home residents in the United States. Am J Geriatr Pharmacother. 2008;6(4):187-197.
48. Gellad WF, Aspinall SL, Handler SM, et al. Use of antipsychotics among older residents in VA nursing homes. Med Care. 2012;50(11):954-960.
49. Bonner A. Improving dementia care and reducing unnecessary use of antipsychotic medications in nursing homes. Center for Medicare and Medicaid Services. http://ltcombudsman.org/uploads/files/support/alice-bonner-slides.pdf. Published April 28, 2013. Accessed October 20, 2017.
50. Vasudev A, Shariff SZ, Liu K, et al. Trends in psychotropic dispensing among older adults with dementia living in long-term care facilities: 2004-2013. Am J Geriatr Psychiatry. 2015;23(12):1259-1269.
51. Kales HC, Gitlin LN, Lyketsos CG, et al; Detroit Expert Panel on Assessment and Management of Neuropsychiatric Symptoms of Dementia. Management of neuropsychiatric symptoms of dementia in clinical settings: recommendations from a multidisciplinary expert panel. J Am Geriatr Soc. 2014;62(4):762-769.
52. Andreasen NC, Liu D, Ziebell S, et al. Relapse duration, treatment intensity, and brain tissue loss in schizophrenia: a prospective longitudinal MRI study. Am J Psychiatry. 2013;170(6):609-615.
53. Mulsant BH. Challenges of the treatment of neuropsychiatric symptoms associated with dementia. Am J Geriatr Psychiatry. 2014;22(4):317-320.

References

1. Foebel AD, Liperoti R, Onder G, et al; SHELTER Study Investigators. Use of antipsychotic drugs among residents with dementia in European long-term care facilities: results from the SHELTER study. J Am Med Dir Assoc. 2014;15(12):911-917.
2. Foebel A, Ballokova A, Wellens NI, et al. A retrospective, longitudinal study of factors associated with new antipsychotic medication use among recently admitted long-term care residents. BMC Geriatr. 2015;15:128.
3. Parsons C, Johnston S, Mathie E, et al. Potentially inappropriate prescribing in older people with dementia in care homes: a retrospective analysis. Drugs Aging. 2012;29(2):143-155.
4. Vidal X, Agustí A, Vallano A, et al; Potentially Inappropriate Prescription in Older Patients in Spain (PIPOPS) Investigators’ project. Elderly patients treated with psychotropic medicines admitted to hospital: associated characteristics and inappropriate use. Eur J Clin Pharmacol. 2016;72(6):755-764.
5. Caron L, Cottencin O, Lapeyre-Mestre M, et al. Off-label prescribing of antipsychotics in adults, children and elderly individuals: a systematic review of recent prescription trends. Curr Pharm Des. 2015;21(23):3280-3297.
6. Weintraub D, Chiang C, Kim HM, et al. Association of antipsychotic use with mortality risk in patients with parkinson disease. JAMA Neurol. 2016;73(5):535-541.
7. Lyketsos CG, Carrillo MC, Ryan JM, et al. Neuropsychiatric symptoms in Alzheimer’s disease. Alzheimers Dement. 2011;7(5):532-539.
8. Kales HC, Chen P, Blow FC, et al. Rates of clinical depression diagnosis, functional impairment, and nursing home placement in coexisting dementia and depression. Am J Geriatr Psychiatry. 2005;13(6):441-449.
9. Yaffe K, Fox P, Newcomer R, et al. Patient and caregiver characteristics and nursing home placement in patients with dementia. JAMA. 2002;287(16):2090-2097.
10. Lopez OL, Becker JT, Chang YF, et al. The long-term effects of conventional and atypical antipsychotics in patients with probable Alzheimer’s disease. Am J Psychiatry. 2013;170(9):1051-1058.
11. Vilalta-Franch J, López-Pousa S, Calvó-Perxas L, et al. Psychosis of Alzheimer disease: prevalence, incidence, persistence, risk factors, and mortality. Am J Geriatr Psychiatry. 2013;21(11):1135-1143.
12. Spalletta G, Musicco M, Padovani A, et al. Neuropsychiatric symptoms and syndromes in a large cohort of newly diagnosed, untreated patients with Alzheimer disease. Am J Geriatr Psychiatry. 2010;18(11):1026-1035.
13. Steinberg M, Shao H, Zandi P, et al; Cache County Investigators. Point and 5-year period prevalence of neuropsychiatric symptoms in dementia: the Cache County Study. Int J Geriatr Psychiatry. 2008;23(2):170-177.
14. Finkel SI, Burns A. Behavioral and psychological symptoms of dementia (BPSD): a clinical and research update-introduction. International Psychogeriatrics. 2000;12:9-12.
15. Lyketsos CG. Neuropsychiatric symptoms (behavioral and psychological symptoms of dementia) and the development of dementia treatments. Int Psychogeriatr. 2007;19(3):409-420.
16. Kunik ME, Snow AL, Davila JA, et al. Causes of aggressive behavior in patients with dementia. J Clin Psychiatry. 2010;71(9):1145-1152.
17. Reus VI, Fochtmann LJ, Eyler AE, et al. The American Psychiatric Association practice guideline on the use of antipsychotics to treat agitation or psychosis in patients with dementia. Am J Psychiatry. 2016;173(5):543-546.
18. Kales HC, Gitlin LN, Lyketsos CG. Assessment and management of behavioral and psychological symptoms of dementia. BMJ. 2015;350:h369. doi: 10.1136/bmj.h369.
19. Schneider LS, Pollock VE, Lyness SA. A metaanalysis of controlled trials of neuroleptic treatment in dementia. J Am Geriatr Soc. 1990;38(5):553-563.
20. Yury CA, Fisher JE. Meta-analysis of the effectiveness of atypical antipsychotics for the treatment of behavioural problems in persons with dementia. Psychother Psychosom. 2007;76(4):213-218.
21. Schneider LS, Dagerman K, Insel PS. Efficacy and adverse effects of atypical antipsychotics for dementia: meta-analysis of randomized, placebo-controlled trials. Am J Geriatr Psychiatry. 2006;14(3):191-210.
22. Ballard CG, Waite J. The effectiveness of atypical antipsychotics for aggression and psychosis in Alzheimer’s disease. Cochrane Database Syst Rev. 2006:1:CD003476.
23. Sink KM, Holden KF, Yaffe K. Pharmacological treatment of neuropsychiatric symptoms of dementia: a review of the evidence. JAMA. 2005;293(5):596-608.
24. Aisen PS, Cummings J, Schneider LS. Symptomatic and nonamyloid/tau based pharmacologic treatment for Alzheimer disease. Cold Spring Harb Perspect Med. 2012;2(3):a006395. doi: 10.1101/cshperspect.a006395.
25. Schneider LS, Tariot PN, Dagerman KS, et al; CATIE-AD Study Group. Effectiveness of atypical antipsychotic drugs in patients with Alzheimer’s disease. N Engl J Med. 2006;355(15):1525-1538.
26. Trifirò G, Sultana J, Spina E. Are the safety profiles of antipsychotic drugs used in dementia the same? An updated review of observational studies. Drug Saf. 2014;37(7):501-520.
27. Trifirò G, Gambassi G, Sen EF, et al. Association of community-acquired pneumonia with antipsychotic drug use in elderly patients: a nested case-control study. Ann Intern Med. 2010;152(7):418-425, W139-W140.
28. Sultana J, Trifirò G. Drug safety warnings: a message in a bottle. Analysis. 2008;179:438-446.
29. Liperoti R, Gambassi G, Lapane KL, et al. Cerebrovascular events among elderly nursing home patients treated with conventional or atypical antipsychotics. J Clin Psychiatry. 2005;66(9):1090-1096.
30. U.S. Food and Drug Administration. Public health advisory: deaths with antipsychotics in elderly patients with behavioral disturbances. https://www.fda.gov/drugs/drugsafety/postmarketdrugsafety information forpatientsandproviders/ucm053171. Updated August 16, 2013. Accessed October 20, 2017.
31. Wang PS, Schneeweiss S, Avorn J, et al. Risk of death in elderly users of conventional vs. atypical antipsychotic medications. N Engl J Med. 2005;353(22):2335-2341.
32. Kales HC, Valenstein M, Kim HM, et al. Mortality risk in patients with dementia treated with antipsychotics versus other psychiatric medications. Am J Psychiatry. 2007;164(10):1568-1576; quiz 1623.
33. Simoni-Wastila L, Ryder PT, Qian J, et al. Association of antipsychotic use with hospital events and mortality among medicare beneficiaries residing in long-term care facilities. Am J Geriatr Psychiatry. 2009;17(5):417-427.
34. Raivio MM, Laurila JV, Strandberg TE, et al. Neither atypical nor conventional antipsychotics increase mortality or hospital admissions among elderly patients with dementia: a two-year prospective study. Am J Geriatr Psychiatry. 2007;15(5):416-424.
35. Gill SS, Bronskill SE, Normand SL, et al. Antipsychotic drug use and mortality in older adults with dementia. Ann Intern Med. 2007;146(11):775-786.
36. Schneeweiss S, Setoguchi S, Brookhart A, et al. Risk of death associated with the use of conventional versus atypical antipsychotic drugs among elderly patients. CMAJ. 2007;176(5):627-632.
37. Kales HC, Kim HM, Zivin K, et al. Risk of mortality among individual antipsychotics in patients with dementia. Am J Psychiatry. 2012;169(1):71-79.
38. Maust DT, Kim HM, Seyfried LS, et al. Antipsychotics, other psychotropics, and the risk of death in patients with dementia: number needed to harm. JAMA Psychiatry. 2015;72(5):438-445.
39. Rhee Y, Csernansky JG, Emanuel LL, et al. Psychotropic medication burden and factors associated with antipsychotic use: an analysis of a population-based sample of community-dwelling older persons with dementia. J Am Geriatr Soc. 2011;59(11):2100-2107.
40. Kales HC, Zivin K, Kim HM, et al. Trends in antipsychotic use in dementia 1999-2007. Arch Gen Psychiatry. 2011;68(2):190-197.
41. American Diabetes Association; American Psychiatric Association; American Association of Clinical Endocrinologists; North American Association for the Study of Obesity. Consensus development conference on antipsychotic drugs and obesity and diabetes. Diabetes Care. 2004;27(2):596-601.
42. Brodaty H, Ames D, Snowdon J, et al. A randomized placebo-controlled trial of risperidone for the treatment of aggression, agitation, and psychosis of dementia. J Clin Psychiatry. 2003;64(2):134-143.
43. Wooltorton E. Risperidone (Risperdal): increased rate of cerebrovascular events in dementia trials. CMAJ. 2002;167(11):1269-1270.
44. United States Government Accountability Office. Antipsychotic drug use: HHS has initiatives to reduce use among older adults in nursing homes, but should expand efforts to other settings. http://www.gao.gov/assets/670/668221.pdf. Published January 2015. Accessed October 20, 2017.
45. Chen Y, Briesacher BA, Field TS, et al. Unexplained variation across US nursing homes in antipsychotic prescribing rates. Arch Intern Med. 2010;170(1):89-95.
46. Feng Z, Hirdes JP, Smith TF, et al. Use of physical restraints and antipsychotic medications in nursing homes: a cross-national study. Int J Geriatr Psychiatry. 2009;24(10):1110-1118.
47. Kamble P, Chen H, Sherer J, et al. Antipsychotic drug use among elderly nursing home residents in the United States. Am J Geriatr Pharmacother. 2008;6(4):187-197.
48. Gellad WF, Aspinall SL, Handler SM, et al. Use of antipsychotics among older residents in VA nursing homes. Med Care. 2012;50(11):954-960.
49. Bonner A. Improving dementia care and reducing unnecessary use of antipsychotic medications in nursing homes. Center for Medicare and Medicaid Services. http://ltcombudsman.org/uploads/files/support/alice-bonner-slides.pdf. Published April 28, 2013. Accessed October 20, 2017.
50. Vasudev A, Shariff SZ, Liu K, et al. Trends in psychotropic dispensing among older adults with dementia living in long-term care facilities: 2004-2013. Am J Geriatr Psychiatry. 2015;23(12):1259-1269.
51. Kales HC, Gitlin LN, Lyketsos CG, et al; Detroit Expert Panel on Assessment and Management of Neuropsychiatric Symptoms of Dementia. Management of neuropsychiatric symptoms of dementia in clinical settings: recommendations from a multidisciplinary expert panel. J Am Geriatr Soc. 2014;62(4):762-769.
52. Andreasen NC, Liu D, Ziebell S, et al. Relapse duration, treatment intensity, and brain tissue loss in schizophrenia: a prospective longitudinal MRI study. Am J Psychiatry. 2013;170(6):609-615.
53. Mulsant BH. Challenges of the treatment of neuropsychiatric symptoms associated with dementia. Am J Geriatr Psychiatry. 2014;22(4):317-320.

Issue
December 2017
Issue
December 2017
Page Number
24-30
Page Number
24-30
Publications
Publications
Topics
Article Type
Display Headline
Prescribing antipsychotics in geriatric patients: Focus on dementia
Display Headline
Prescribing antipsychotics in geriatric patients: Focus on dementia
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Article PDF Media

Using antipsychotics for dementia

Article Type
Changed
Fri, 09/28/2018 - 11:18
Display Headline
Using antipsychotics for dementia

Author and Disclosure Information

Dr. Kales is Professor of Psychiatry, Program for Positive Aging, Department of Psychiatry, University of Michigan, VA Center for Clinical Management Research, Ann Arbor, Michigan.

Issue
December 2017
Publications
Topics
Sections
Author and Disclosure Information

Dr. Kales is Professor of Psychiatry, Program for Positive Aging, Department of Psychiatry, University of Michigan, VA Center for Clinical Management Research, Ann Arbor, Michigan.

Author and Disclosure Information

Dr. Kales is Professor of Psychiatry, Program for Positive Aging, Department of Psychiatry, University of Michigan, VA Center for Clinical Management Research, Ann Arbor, Michigan.

Issue
December 2017
Issue
December 2017
Publications
Publications
Topics
Article Type
Display Headline
Using antipsychotics for dementia
Display Headline
Using antipsychotics for dementia
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica

Sexting: What are the clinical and legal implications?

Article Type
Changed
Tue, 12/11/2018 - 15:01
Display Headline
Sexting: What are the clinical and legal implications?
 

Sexting includes sending sexually explicit (or sexually suggestive) text messages and photos, usually by cell phone. This article focuses on sexts involving photos. Cell phones are almost ubiquitous among American teens, and with technological advances, sexts are getting easier to send. Sexting may occur to initiate a relationship or sustain one. Some teenagers are coerced into sexting. Many people do not realize the potential long-term consequences of sexting—particularly because of the impulsive nature of sexting and the belief that the behavior is harmless.

Media attention has recently focused on teens who face legal charges related to sexting. Sexting photos may be considered child pornography—even though the teens made it themselves. There are also social consequences to sexting. Photos meant to be private are sometimes forwarded to others. Cyberbullying is not uncommon with teen sexting, and suicides after experiencing this behavior have been reported.

Sexting may be a form of modern flirtation, but in some cases, it may be a marker of other risk behaviors, such as substance abuse. Psychiatrists must be aware of the frequency and meaning of this potentially dangerous behavior. Clinicians should feel comfortable asking their patients about it and provide education and counseling.

CASE

Private photos get shared

K, age 14, a freshman with no psychiatric history, is referred to you by her school psychologist for evaluation of suicidal ideation. K reports depressed mood, poor sleep, inattention, loss of appetite, anhedonia, and feelings of guilt for the past month. She recently ended a relationship with her boyfriend of 1 year after she learned that he had shared with his friends naked photos of her that she had sent him. The school administration learned of the photos when a student posted them on one of the school computers.

K’s boyfriend, age 16, was suspended after the school learned that he had shared the photos without K’s consent. K, who is a good student, missed several days of school, including cheerleading practice; previously she had never missed a day of school.

On evaluation, K is tearful, stating that her life is “over.” She says that her ex-boyfriend’s friends are harassing her, calling her “slut” and making sexual comments. She also feels guilty, because she learned that the police interviewed her ex-boyfriend in connection with posting her photos on the Internet. In a text, he said he “might get charged with child pornography.” On further questioning, K confides that she had naked photos of her ex-boyfriend on her phone. She admits to sharing the pictures with her best friend, because she was “angry and wanted to get back” at her ex-boyfriend. She also reports a several-month history of sexting with her ex-boyfriend. K deleted the photos and texts after learning that her ex-boyfriend “was in trouble with the police.”

K has no prior sexual experience. She dated 1 boy her age prior to her ex-boyfriend. She had never been evaluated by a mental health clinician. She is dysphoric and reports feeling “hopeless … Unless this can be erased, I can’t go back to school.”


Sexting: What is the extent of the problem?

The true prevalence of sexting is difficult to ascertain, because different studies have used different definitions and method­ologies. However, the rates are far from negligible. Sexting rates increase with age, over the teen years.1-3 Among American minors, 2.5% to 28% of middle school and high school students report that they have sent a sext (Table 11-9). Studies of American young adults (age ≥18) and university students have found 30% to 60% have sent sexts, and >40% have received a sext.4,5

Many people receive sexts—including individuals who are not the intended recipient. In 1 study, although most teens intended to share sexts only with their boyfriend/girlfriend, 25% to 33% had received sext photos intended for other people.6 In another recent study, 25% of teens had forwarded a sext that they received.7 Moreover, 12% of teenage boys and 5% of teenage girls had sent a sexually explicit photo that they took of another teen to a third person.7 Forwarding sexts can add exponentially to the psychosocial risks of the photographed teenager.

Who sexts? Current research indicates that the likelihood of sexting is related to age, personality, and social situation. Teens are approaching the peak age of their sex drive, and often are curious and feel invincible. Teens are more impulsive than adults. When it takes less than a minute to send a sext, irreversible poor choices can be made quickly. Teens who send sexts often engage in more text messaging than other teens.7

Teens may use sexting to initiate or sustain a relationship. Sexts also may be sent because of coercion. More than one-half of girls cited pressure to sext from a boy.6 Temple et al3 found that more than one-half of their study sample had been asked to send a sext. Girls were more likely than boys to be asked to send a sext; most were troubled by this.

One study that assessed knowledge of potential legal consequences of sexting found that many teens who sent sexts were aware of the potential consequences.7 Regarding personality traits, sexting among undergraduates was predicted by neuroticism and low agreeableness.10 Conversely, sending text messages with sexually suggestive writing was predicted by extraversion and problematic cell phone use.

Comorbidities. There are mixed findings about whether sexting is simply a modern dating strategy or a marker of other risk behaviors; age may play an important discriminating role. Sexual activity appears to be correlated with sexting. According to Temple and Choi,11 “Sexting fits within the context of adolescent sexual development and may be a viable indicator of adolescent sexual activity.”11

Some authors have suggested that sext­ing is a contemporary risk behavior that is likely to correlate with other risk behaviors. Among young teens—seventh graders who were referred to a risk prevention trial because of behavioral/emotional difficulties—those who sexted were more likely to engage in early sexual behaviors.8 These younger at-risk teens also had less understanding of their emotions and greater difficulty in regulating their emotions.

Among the general population of high school students, teens who sext are more likely to be sexually active.3 High school girls who engaged in sexting were noted to engage in other risk behaviors, including having multiple partners in the past year and using alcohol or drugs before sex.3 Teens who had sent a sext were more likely to be sexually active 1 year later than teens who had not.11Studies of sexting among university students also have had mixed findings. One study found that among undergraduates, sexting was associated with substance use and other risk behaviors.9 Another young adult study found sexting was not related to sexual risk or psychological well-being.4

 

 

 

Legal issues affect psychiatrists as well as patients

As a psychiatrist evaluating K, what are your duties as a mandated reporter? Psychiatrists are legally required to report suspected maltreatment or abuse of children.12 The circumstances under which psychiatrists may have a mandate to report include when a psychiatrist:

  • evaluates a child and suspects abuse
  • suspects child abuse based on an adult patient’s report
  • learns from a third party that a child may have been/is being abused.

Psychiatrists usually are not mandated to report other types of potentially criminal behavior. As such, reporting sexting might be considered a breach of confidentiality. Psychiatrists should be familiar with the specific reporting guidelines for the jurisdiction in which they practice. Psychiatrists who work with individuals who commit crimes should focus on changing the potentially dangerous behaviors rather than reporting them.

Does the transmission of naked photos of a minor in a sexual pose or act constitute child pornography or another criminal offense? The legal answer varies, but the role of the psychiatrist does not. Psychiatrists should educate their patients about potentially dangerous behaviors.

With regards to the legal consequences, some states classify underage sexting photos as child pornography. Others have less rigid definitions of child pornography and take into account the age of the participants and their intent. Such jurisdictions point out that sexting naked photos among adolescents is “age appropriate.” Some have enacted specific sexting laws to address the transmission of obscene material to a child through the Internet. In some jurisdictions, sexting laws are categorized to refer to behavior of individuals under or over age 18. The term “revenge porn” is used to refer to nonconsensual pornography with its dissemination motivated by spite.13 Some states have defined specific revenge porn laws to address the behavior. Currently, 20 states have sexting laws and 26 states have revenge porn laws.14 Twenty states address a minor age <18 sending the photo, while only 18 address the recipient. The law in this area can be complex and detailed, taking into account the age of the sender, the intentions of the sender, and the nature of the relationship between the sender and the recipient and the behavior of the recipient.

Laws regarding sexting vary greatly. Sexting may be a misdemeanor or a felony, depending on the state, the specific behavior, and the frequency. In the United States, 11 state laws include a diversion remedy—an option to pursue the case outside of the criminal juvenile system; 10 laws require counseling or another informal sanction; 11 states laws have the potential for misdemeanor punishment; and 4 state laws have the potential for felony punishment.14 Depending on the criminal charge, the perpetrator may have to register as a sex offender. For example, in some jurisdictions, a conviction for possession of child pornography requires sex offender registration. Thirty-eight states include juvenile sex offenders in their sex offender registries. Other states require juveniles to register if they are age ≥15 years or have been tried as an adult.15

The frequency of police involvement in sexting cases also greatly varies. A national study examining the characteristics of youth sexting cases revealed that law enforcement agencies handled approximately 3,477 cases of youth-produced sexual photos in 2008 and 2009.16 Situations that involved an adult or a minor engaged in malicious, nonconsensual, or abusive behavior comprised two-thirds of cases. Arrests occurred in 62% of the adult-involved cases and 36% of the aggravated youth-only cases. Arrests occurred in only 18% of investigated non-aggravated youth-only cases. Table 2 describes recent American sexting legal cases and their outcomes.

In K’s case, depending on the jurisdiction, K or her ex-boyfriend may be subject to arrest for child pornography, revenge pornography, or sexting.

 

 

 

Potential social and psychiatric consequences

What are the social and psychiatric ramifications for K? Aside from potential legal consequences of sexting, K is experiencing psychological and social consequences. She has developed depressive symptoms and suicidal ideation. Her ex-boyfriend’s dissemination of her nude photos on the school computer could be interpreted as cyberbullying. (The National Center for Missing and Exploited Children defines cyberbullying as “bullying through the use of technology or electronic devices such as telephones, cell phones, computers, and the Internet.”17 All 50 states have enacted laws against bullying; 48 states have electronic harassment in their bullying laws; and 22 states have laws specifically referencing “cyberbullying.”)

Her depressive symptoms developed in response to her feelings of guilt and shame related to sexting as well as the subsequent peer harassment. She is refusing to return to school because of her concerns about bullying. A careful inquiry into suicidality should be part of the evaluation when sexting has led to psychiatric symptoms. Several cases of sexting and cyberbullying have ended in suicide (Table 3).

How to ask patients about sexting

To screen patients for sexting, clinicians need to develop a new skill set, which at first may be uncomfortable. However, the questions to ask are not all that different from other questions about adolescent and young adult sexuality. The importance of patients seeing that we as physicians are comfortable with the topic and approachable about their sexual health cannot be overemphasized. When discussing sexting with patients, it is essential to:

  • explain that you are asking questions about their sexual health because they are important to overall health
  • engage patients in discussion in a nonthreatening and nonjudgmental way
  • develop rapport so patients feel comfortable disclosing behavior that may be embarrassing
  • listen to their stories and build a context for understanding their experiences. As you listen, ask questions when needed to help move the story along.

Sometimes when asking about topics that are uncomfortable, clinicians revert from open-ended to closed-ended questions, but when asking about a patient’s sexual life, it is especially important to be open-ended and ask questions in a nonjudgmental way. Contextualizing sexual questions by (for example) asking them while discussing the teen’s relationships will make them seem more natural.18 To best understand, inquire explicitly about specific behaviors, but do so without appearing voyeuristic.18

Sexting may precede sexual intercourse. Keep in mind that a patient may report that she (he) is not sexually active but still may be involved in sexting. Therefore, discuss sexting even if your patient reports not being sexually active. By understanding the prevalence of sexting among teens, you can ask questions in a normalizing way. Clinicians can inquire about sexting while discussing relationships and dating or online risk behaviors.

Also consider whether any of your patient’s sexual behaviors, including sext­ing, are the result of coercion: “Some of my patients tell me they feel pressured or coerced into having sex. Have you ever felt this way?”19 and “Have you ever been picked on or bullied? Is that still a problem?” are suggested safety screening questions about bullying,18 and one can also ask about specific cyberbullying behaviors.

Table 4 lists additional risk behaviors to ask about in a patient who has been sext­ing. Sexting is a widely accepted behavior among adolescents, yet many adolescents and parents are unaware of the legal implications of such a behavior. As physicians, we have a role in educating our patients about potentially harmful behaviors, and to do so in a manner that is not viewed as lecturing, which may result in receiving poor-quality information.

Bottom Line

Although it may seem harmless to adolescents, sexting has potentially serious legal, social, and psychological consequences. Sexting may co-occur with other risk behaviors, such as substance abuse. Clinicians should be prepared to ask patients about sexting, educate them about the risks, and consider whether patients also are engaging in other risky behaviors.

Related Resources

  • American Academy of Pediatrics. Talking to kids and teens about social media and sexting.
  • Connect Safely. Tips for dealing with teen sexting. http://www.connectsafely.org/tips-for-dealing-with-teen-sexting.
References

1. Mitchell KJ, Finkelhor D, Jones LM, et al. Prevalence and characteristics of youth sexting: a national study. Pediatrics. 2012;129(1):13-20.
2. Lenhart A. Teens and sexting. The Pew Research Center. http://www.pewinternet.org/2009/12/15/teens-and-sexting. Published December 15, 2009. Accessed October 31, 2017.
3. Temple JR, Paul JA, van den Berg P, et al. Teen sexting and its association with sexual behaviors. Arch Pediatr Adolesc Med. 2012;166(9):828-833.
4. Gordon-Messer D, Bauermeister JA, Grodzinski A, et al. Sexting among young adults. J Adolesc Health. 2013;52(3):301-306.
5. Henderson L. Sexting and sexual relationships among teens and young adults. McNair Scholars Research Journal. 2011;7(1):31-39.
6. The National Campaign to Prevent Teen and Unplanned Pregnancy. Sex and tech: results from a survey of teens and young adults. https://thenationalcampaign.org/sites/default/files/resource-primary-download/sex_and_tech_summary.pdf. Published December 2008. Accessed October 31, 2017.
7. Strassberg DS, McKinnon RK, Sustaíta MA, et al. Sexting by high school students: an exploratory and descriptive study. Arch Sex Behav. 2013;42(1):15-21.
8. Houck CD, Barker D, Rizzo C, et al. Sexting and sexual behavior in at-risk adolescents. Pediatrics. 2014;133(2):e276-e282.
9. Benotsch EG, Snipes DJ, Martin AM, et al. Sexting, substance use, and sexual risk behavior in young adults. J Adolesc Health. 2013;52(3):307-313.
10. Delevi R, Weisskirch RS. Personality factors as predictors of sexting. Comput Human Behav. 2013;29(6):2589-2594.
11. Temple JR, Choi H. Longitudinal association between teen sexting and sexual behavior. Pediatrics. 2014;134(5):1287-1292.
12. McEwan M, Friedman SH. Violence by parents against their children: reporting of maltreatment suspicions, child protection, and risk in mental illness. Psych Clin North Am. 2016;39(4):691-700.
13. Citron DK, Franks MA. Criminalizing revenge porn. Wake Forest Law Review. 2014;49:345-391.
14. Hinduja S, Patchin JW. State cyberbullying laws: a brief review of state cyberbullying laws and policies. Cyberbullying Research Center. https://cyberbullying.org/Bullying-and-Cyberbullying-Laws.pdf. Updated 2016. Accessed October 31, 2017.
15. Beitsch R. States slowly scale back juvenile sex offender registries. The Pew Charitable Trusts. http://www.pewtrusts.org/en/research-and-analysis/blogs/stateline/2015/11/19/states-slowly-scale-back-juvenile-sex-offender-registries. Published November 19, 2015. Accessed October 31, 2017.
16. Wolak J, Finkelhor D, Mitchell KJ. How often are teens arrested for sexting? Data from a national sample of police cases. Pediatrics. 2012;129(1):4-12.
17. The Campus School at Boston College. Bullying prevention policy. https://www.bc.edu/bc-web/schools/lsoe/sites/campus-school/who-we-are/policies-and-procedures/bullying-prevention-policy.html. Accessed October 31, 2017.
18. Goldenring JM, Rosen DS. Getting into adolescent heads: an essential update. Contemporary Pediatrics. 2004;21(1):64.
19. Klein DA, Goldenring JM, Adelman WP. HEEADSSS 3.0: the psychosocial interview for adolescents updated for a new century fueled by media. Contemporary Pediatrics. http://contemporarypediatrics.modernmedicine.com/contemporary-pediatrics/content/tags/adolescent-medicine/heeadsss-30-psychosocial-interview-adolesce?page=full. Published January 1, 2014. Accessed October 31, 2017.

Article PDF
Author and Disclosure Information

Susan Hatters Friedman, MD
Associate Professor of Psychological Medicine
University of Auckland
Auckland, New Zealand

Renee M. Sorrentino, MD
Director
The Institute for Sexual Wellness
Weymouth, Massachusetts
Assistant Professor
Department of Psychiatry
Harvard University Medical School
Boston, Massachusetts

Joshua B. Friedman, MD, PhD
Pediatrician
Director, Child Abuse Pediatrics
MetroHealth Medical Center
Co-Director, Child Protection Team
Cleveland Clinic Children’s Hospital
Cleveland, Ohio

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

Issue
December 2017
Publications
Page Number
35-41
Sections
Author and Disclosure Information

Susan Hatters Friedman, MD
Associate Professor of Psychological Medicine
University of Auckland
Auckland, New Zealand

Renee M. Sorrentino, MD
Director
The Institute for Sexual Wellness
Weymouth, Massachusetts
Assistant Professor
Department of Psychiatry
Harvard University Medical School
Boston, Massachusetts

Joshua B. Friedman, MD, PhD
Pediatrician
Director, Child Abuse Pediatrics
MetroHealth Medical Center
Co-Director, Child Protection Team
Cleveland Clinic Children’s Hospital
Cleveland, Ohio

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

Author and Disclosure Information

Susan Hatters Friedman, MD
Associate Professor of Psychological Medicine
University of Auckland
Auckland, New Zealand

Renee M. Sorrentino, MD
Director
The Institute for Sexual Wellness
Weymouth, Massachusetts
Assistant Professor
Department of Psychiatry
Harvard University Medical School
Boston, Massachusetts

Joshua B. Friedman, MD, PhD
Pediatrician
Director, Child Abuse Pediatrics
MetroHealth Medical Center
Co-Director, Child Protection Team
Cleveland Clinic Children’s Hospital
Cleveland, Ohio

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

Article PDF
Article PDF
 

Sexting includes sending sexually explicit (or sexually suggestive) text messages and photos, usually by cell phone. This article focuses on sexts involving photos. Cell phones are almost ubiquitous among American teens, and with technological advances, sexts are getting easier to send. Sexting may occur to initiate a relationship or sustain one. Some teenagers are coerced into sexting. Many people do not realize the potential long-term consequences of sexting—particularly because of the impulsive nature of sexting and the belief that the behavior is harmless.

Media attention has recently focused on teens who face legal charges related to sexting. Sexting photos may be considered child pornography—even though the teens made it themselves. There are also social consequences to sexting. Photos meant to be private are sometimes forwarded to others. Cyberbullying is not uncommon with teen sexting, and suicides after experiencing this behavior have been reported.

Sexting may be a form of modern flirtation, but in some cases, it may be a marker of other risk behaviors, such as substance abuse. Psychiatrists must be aware of the frequency and meaning of this potentially dangerous behavior. Clinicians should feel comfortable asking their patients about it and provide education and counseling.

CASE

Private photos get shared

K, age 14, a freshman with no psychiatric history, is referred to you by her school psychologist for evaluation of suicidal ideation. K reports depressed mood, poor sleep, inattention, loss of appetite, anhedonia, and feelings of guilt for the past month. She recently ended a relationship with her boyfriend of 1 year after she learned that he had shared with his friends naked photos of her that she had sent him. The school administration learned of the photos when a student posted them on one of the school computers.

K’s boyfriend, age 16, was suspended after the school learned that he had shared the photos without K’s consent. K, who is a good student, missed several days of school, including cheerleading practice; previously she had never missed a day of school.

On evaluation, K is tearful, stating that her life is “over.” She says that her ex-boyfriend’s friends are harassing her, calling her “slut” and making sexual comments. She also feels guilty, because she learned that the police interviewed her ex-boyfriend in connection with posting her photos on the Internet. In a text, he said he “might get charged with child pornography.” On further questioning, K confides that she had naked photos of her ex-boyfriend on her phone. She admits to sharing the pictures with her best friend, because she was “angry and wanted to get back” at her ex-boyfriend. She also reports a several-month history of sexting with her ex-boyfriend. K deleted the photos and texts after learning that her ex-boyfriend “was in trouble with the police.”

K has no prior sexual experience. She dated 1 boy her age prior to her ex-boyfriend. She had never been evaluated by a mental health clinician. She is dysphoric and reports feeling “hopeless … Unless this can be erased, I can’t go back to school.”


Sexting: What is the extent of the problem?

The true prevalence of sexting is difficult to ascertain, because different studies have used different definitions and method­ologies. However, the rates are far from negligible. Sexting rates increase with age, over the teen years.1-3 Among American minors, 2.5% to 28% of middle school and high school students report that they have sent a sext (Table 11-9). Studies of American young adults (age ≥18) and university students have found 30% to 60% have sent sexts, and >40% have received a sext.4,5

Many people receive sexts—including individuals who are not the intended recipient. In 1 study, although most teens intended to share sexts only with their boyfriend/girlfriend, 25% to 33% had received sext photos intended for other people.6 In another recent study, 25% of teens had forwarded a sext that they received.7 Moreover, 12% of teenage boys and 5% of teenage girls had sent a sexually explicit photo that they took of another teen to a third person.7 Forwarding sexts can add exponentially to the psychosocial risks of the photographed teenager.

Who sexts? Current research indicates that the likelihood of sexting is related to age, personality, and social situation. Teens are approaching the peak age of their sex drive, and often are curious and feel invincible. Teens are more impulsive than adults. When it takes less than a minute to send a sext, irreversible poor choices can be made quickly. Teens who send sexts often engage in more text messaging than other teens.7

Teens may use sexting to initiate or sustain a relationship. Sexts also may be sent because of coercion. More than one-half of girls cited pressure to sext from a boy.6 Temple et al3 found that more than one-half of their study sample had been asked to send a sext. Girls were more likely than boys to be asked to send a sext; most were troubled by this.

One study that assessed knowledge of potential legal consequences of sexting found that many teens who sent sexts were aware of the potential consequences.7 Regarding personality traits, sexting among undergraduates was predicted by neuroticism and low agreeableness.10 Conversely, sending text messages with sexually suggestive writing was predicted by extraversion and problematic cell phone use.

Comorbidities. There are mixed findings about whether sexting is simply a modern dating strategy or a marker of other risk behaviors; age may play an important discriminating role. Sexual activity appears to be correlated with sexting. According to Temple and Choi,11 “Sexting fits within the context of adolescent sexual development and may be a viable indicator of adolescent sexual activity.”11

Some authors have suggested that sext­ing is a contemporary risk behavior that is likely to correlate with other risk behaviors. Among young teens—seventh graders who were referred to a risk prevention trial because of behavioral/emotional difficulties—those who sexted were more likely to engage in early sexual behaviors.8 These younger at-risk teens also had less understanding of their emotions and greater difficulty in regulating their emotions.

Among the general population of high school students, teens who sext are more likely to be sexually active.3 High school girls who engaged in sexting were noted to engage in other risk behaviors, including having multiple partners in the past year and using alcohol or drugs before sex.3 Teens who had sent a sext were more likely to be sexually active 1 year later than teens who had not.11Studies of sexting among university students also have had mixed findings. One study found that among undergraduates, sexting was associated with substance use and other risk behaviors.9 Another young adult study found sexting was not related to sexual risk or psychological well-being.4

 

 

 

Legal issues affect psychiatrists as well as patients

As a psychiatrist evaluating K, what are your duties as a mandated reporter? Psychiatrists are legally required to report suspected maltreatment or abuse of children.12 The circumstances under which psychiatrists may have a mandate to report include when a psychiatrist:

  • evaluates a child and suspects abuse
  • suspects child abuse based on an adult patient’s report
  • learns from a third party that a child may have been/is being abused.

Psychiatrists usually are not mandated to report other types of potentially criminal behavior. As such, reporting sexting might be considered a breach of confidentiality. Psychiatrists should be familiar with the specific reporting guidelines for the jurisdiction in which they practice. Psychiatrists who work with individuals who commit crimes should focus on changing the potentially dangerous behaviors rather than reporting them.

Does the transmission of naked photos of a minor in a sexual pose or act constitute child pornography or another criminal offense? The legal answer varies, but the role of the psychiatrist does not. Psychiatrists should educate their patients about potentially dangerous behaviors.

With regards to the legal consequences, some states classify underage sexting photos as child pornography. Others have less rigid definitions of child pornography and take into account the age of the participants and their intent. Such jurisdictions point out that sexting naked photos among adolescents is “age appropriate.” Some have enacted specific sexting laws to address the transmission of obscene material to a child through the Internet. In some jurisdictions, sexting laws are categorized to refer to behavior of individuals under or over age 18. The term “revenge porn” is used to refer to nonconsensual pornography with its dissemination motivated by spite.13 Some states have defined specific revenge porn laws to address the behavior. Currently, 20 states have sexting laws and 26 states have revenge porn laws.14 Twenty states address a minor age <18 sending the photo, while only 18 address the recipient. The law in this area can be complex and detailed, taking into account the age of the sender, the intentions of the sender, and the nature of the relationship between the sender and the recipient and the behavior of the recipient.

Laws regarding sexting vary greatly. Sexting may be a misdemeanor or a felony, depending on the state, the specific behavior, and the frequency. In the United States, 11 state laws include a diversion remedy—an option to pursue the case outside of the criminal juvenile system; 10 laws require counseling or another informal sanction; 11 states laws have the potential for misdemeanor punishment; and 4 state laws have the potential for felony punishment.14 Depending on the criminal charge, the perpetrator may have to register as a sex offender. For example, in some jurisdictions, a conviction for possession of child pornography requires sex offender registration. Thirty-eight states include juvenile sex offenders in their sex offender registries. Other states require juveniles to register if they are age ≥15 years or have been tried as an adult.15

The frequency of police involvement in sexting cases also greatly varies. A national study examining the characteristics of youth sexting cases revealed that law enforcement agencies handled approximately 3,477 cases of youth-produced sexual photos in 2008 and 2009.16 Situations that involved an adult or a minor engaged in malicious, nonconsensual, or abusive behavior comprised two-thirds of cases. Arrests occurred in 62% of the adult-involved cases and 36% of the aggravated youth-only cases. Arrests occurred in only 18% of investigated non-aggravated youth-only cases. Table 2 describes recent American sexting legal cases and their outcomes.

In K’s case, depending on the jurisdiction, K or her ex-boyfriend may be subject to arrest for child pornography, revenge pornography, or sexting.

 

 

 

Potential social and psychiatric consequences

What are the social and psychiatric ramifications for K? Aside from potential legal consequences of sexting, K is experiencing psychological and social consequences. She has developed depressive symptoms and suicidal ideation. Her ex-boyfriend’s dissemination of her nude photos on the school computer could be interpreted as cyberbullying. (The National Center for Missing and Exploited Children defines cyberbullying as “bullying through the use of technology or electronic devices such as telephones, cell phones, computers, and the Internet.”17 All 50 states have enacted laws against bullying; 48 states have electronic harassment in their bullying laws; and 22 states have laws specifically referencing “cyberbullying.”)

Her depressive symptoms developed in response to her feelings of guilt and shame related to sexting as well as the subsequent peer harassment. She is refusing to return to school because of her concerns about bullying. A careful inquiry into suicidality should be part of the evaluation when sexting has led to psychiatric symptoms. Several cases of sexting and cyberbullying have ended in suicide (Table 3).

How to ask patients about sexting

To screen patients for sexting, clinicians need to develop a new skill set, which at first may be uncomfortable. However, the questions to ask are not all that different from other questions about adolescent and young adult sexuality. The importance of patients seeing that we as physicians are comfortable with the topic and approachable about their sexual health cannot be overemphasized. When discussing sexting with patients, it is essential to:

  • explain that you are asking questions about their sexual health because they are important to overall health
  • engage patients in discussion in a nonthreatening and nonjudgmental way
  • develop rapport so patients feel comfortable disclosing behavior that may be embarrassing
  • listen to their stories and build a context for understanding their experiences. As you listen, ask questions when needed to help move the story along.

Sometimes when asking about topics that are uncomfortable, clinicians revert from open-ended to closed-ended questions, but when asking about a patient’s sexual life, it is especially important to be open-ended and ask questions in a nonjudgmental way. Contextualizing sexual questions by (for example) asking them while discussing the teen’s relationships will make them seem more natural.18 To best understand, inquire explicitly about specific behaviors, but do so without appearing voyeuristic.18

Sexting may precede sexual intercourse. Keep in mind that a patient may report that she (he) is not sexually active but still may be involved in sexting. Therefore, discuss sexting even if your patient reports not being sexually active. By understanding the prevalence of sexting among teens, you can ask questions in a normalizing way. Clinicians can inquire about sexting while discussing relationships and dating or online risk behaviors.

Also consider whether any of your patient’s sexual behaviors, including sext­ing, are the result of coercion: “Some of my patients tell me they feel pressured or coerced into having sex. Have you ever felt this way?”19 and “Have you ever been picked on or bullied? Is that still a problem?” are suggested safety screening questions about bullying,18 and one can also ask about specific cyberbullying behaviors.

Table 4 lists additional risk behaviors to ask about in a patient who has been sext­ing. Sexting is a widely accepted behavior among adolescents, yet many adolescents and parents are unaware of the legal implications of such a behavior. As physicians, we have a role in educating our patients about potentially harmful behaviors, and to do so in a manner that is not viewed as lecturing, which may result in receiving poor-quality information.

Bottom Line

Although it may seem harmless to adolescents, sexting has potentially serious legal, social, and psychological consequences. Sexting may co-occur with other risk behaviors, such as substance abuse. Clinicians should be prepared to ask patients about sexting, educate them about the risks, and consider whether patients also are engaging in other risky behaviors.

Related Resources

  • American Academy of Pediatrics. Talking to kids and teens about social media and sexting.
  • Connect Safely. Tips for dealing with teen sexting. http://www.connectsafely.org/tips-for-dealing-with-teen-sexting.
 

Sexting includes sending sexually explicit (or sexually suggestive) text messages and photos, usually by cell phone. This article focuses on sexts involving photos. Cell phones are almost ubiquitous among American teens, and with technological advances, sexts are getting easier to send. Sexting may occur to initiate a relationship or sustain one. Some teenagers are coerced into sexting. Many people do not realize the potential long-term consequences of sexting—particularly because of the impulsive nature of sexting and the belief that the behavior is harmless.

Media attention has recently focused on teens who face legal charges related to sexting. Sexting photos may be considered child pornography—even though the teens made it themselves. There are also social consequences to sexting. Photos meant to be private are sometimes forwarded to others. Cyberbullying is not uncommon with teen sexting, and suicides after experiencing this behavior have been reported.

Sexting may be a form of modern flirtation, but in some cases, it may be a marker of other risk behaviors, such as substance abuse. Psychiatrists must be aware of the frequency and meaning of this potentially dangerous behavior. Clinicians should feel comfortable asking their patients about it and provide education and counseling.

CASE

Private photos get shared

K, age 14, a freshman with no psychiatric history, is referred to you by her school psychologist for evaluation of suicidal ideation. K reports depressed mood, poor sleep, inattention, loss of appetite, anhedonia, and feelings of guilt for the past month. She recently ended a relationship with her boyfriend of 1 year after she learned that he had shared with his friends naked photos of her that she had sent him. The school administration learned of the photos when a student posted them on one of the school computers.

K’s boyfriend, age 16, was suspended after the school learned that he had shared the photos without K’s consent. K, who is a good student, missed several days of school, including cheerleading practice; previously she had never missed a day of school.

On evaluation, K is tearful, stating that her life is “over.” She says that her ex-boyfriend’s friends are harassing her, calling her “slut” and making sexual comments. She also feels guilty, because she learned that the police interviewed her ex-boyfriend in connection with posting her photos on the Internet. In a text, he said he “might get charged with child pornography.” On further questioning, K confides that she had naked photos of her ex-boyfriend on her phone. She admits to sharing the pictures with her best friend, because she was “angry and wanted to get back” at her ex-boyfriend. She also reports a several-month history of sexting with her ex-boyfriend. K deleted the photos and texts after learning that her ex-boyfriend “was in trouble with the police.”

K has no prior sexual experience. She dated 1 boy her age prior to her ex-boyfriend. She had never been evaluated by a mental health clinician. She is dysphoric and reports feeling “hopeless … Unless this can be erased, I can’t go back to school.”


Sexting: What is the extent of the problem?

The true prevalence of sexting is difficult to ascertain, because different studies have used different definitions and method­ologies. However, the rates are far from negligible. Sexting rates increase with age, over the teen years.1-3 Among American minors, 2.5% to 28% of middle school and high school students report that they have sent a sext (Table 11-9). Studies of American young adults (age ≥18) and university students have found 30% to 60% have sent sexts, and >40% have received a sext.4,5

Many people receive sexts—including individuals who are not the intended recipient. In 1 study, although most teens intended to share sexts only with their boyfriend/girlfriend, 25% to 33% had received sext photos intended for other people.6 In another recent study, 25% of teens had forwarded a sext that they received.7 Moreover, 12% of teenage boys and 5% of teenage girls had sent a sexually explicit photo that they took of another teen to a third person.7 Forwarding sexts can add exponentially to the psychosocial risks of the photographed teenager.

Who sexts? Current research indicates that the likelihood of sexting is related to age, personality, and social situation. Teens are approaching the peak age of their sex drive, and often are curious and feel invincible. Teens are more impulsive than adults. When it takes less than a minute to send a sext, irreversible poor choices can be made quickly. Teens who send sexts often engage in more text messaging than other teens.7

Teens may use sexting to initiate or sustain a relationship. Sexts also may be sent because of coercion. More than one-half of girls cited pressure to sext from a boy.6 Temple et al3 found that more than one-half of their study sample had been asked to send a sext. Girls were more likely than boys to be asked to send a sext; most were troubled by this.

One study that assessed knowledge of potential legal consequences of sexting found that many teens who sent sexts were aware of the potential consequences.7 Regarding personality traits, sexting among undergraduates was predicted by neuroticism and low agreeableness.10 Conversely, sending text messages with sexually suggestive writing was predicted by extraversion and problematic cell phone use.

Comorbidities. There are mixed findings about whether sexting is simply a modern dating strategy or a marker of other risk behaviors; age may play an important discriminating role. Sexual activity appears to be correlated with sexting. According to Temple and Choi,11 “Sexting fits within the context of adolescent sexual development and may be a viable indicator of adolescent sexual activity.”11

Some authors have suggested that sext­ing is a contemporary risk behavior that is likely to correlate with other risk behaviors. Among young teens—seventh graders who were referred to a risk prevention trial because of behavioral/emotional difficulties—those who sexted were more likely to engage in early sexual behaviors.8 These younger at-risk teens also had less understanding of their emotions and greater difficulty in regulating their emotions.

Among the general population of high school students, teens who sext are more likely to be sexually active.3 High school girls who engaged in sexting were noted to engage in other risk behaviors, including having multiple partners in the past year and using alcohol or drugs before sex.3 Teens who had sent a sext were more likely to be sexually active 1 year later than teens who had not.11Studies of sexting among university students also have had mixed findings. One study found that among undergraduates, sexting was associated with substance use and other risk behaviors.9 Another young adult study found sexting was not related to sexual risk or psychological well-being.4

 

 

 

Legal issues affect psychiatrists as well as patients

As a psychiatrist evaluating K, what are your duties as a mandated reporter? Psychiatrists are legally required to report suspected maltreatment or abuse of children.12 The circumstances under which psychiatrists may have a mandate to report include when a psychiatrist:

  • evaluates a child and suspects abuse
  • suspects child abuse based on an adult patient’s report
  • learns from a third party that a child may have been/is being abused.

Psychiatrists usually are not mandated to report other types of potentially criminal behavior. As such, reporting sexting might be considered a breach of confidentiality. Psychiatrists should be familiar with the specific reporting guidelines for the jurisdiction in which they practice. Psychiatrists who work with individuals who commit crimes should focus on changing the potentially dangerous behaviors rather than reporting them.

Does the transmission of naked photos of a minor in a sexual pose or act constitute child pornography or another criminal offense? The legal answer varies, but the role of the psychiatrist does not. Psychiatrists should educate their patients about potentially dangerous behaviors.

With regards to the legal consequences, some states classify underage sexting photos as child pornography. Others have less rigid definitions of child pornography and take into account the age of the participants and their intent. Such jurisdictions point out that sexting naked photos among adolescents is “age appropriate.” Some have enacted specific sexting laws to address the transmission of obscene material to a child through the Internet. In some jurisdictions, sexting laws are categorized to refer to behavior of individuals under or over age 18. The term “revenge porn” is used to refer to nonconsensual pornography with its dissemination motivated by spite.13 Some states have defined specific revenge porn laws to address the behavior. Currently, 20 states have sexting laws and 26 states have revenge porn laws.14 Twenty states address a minor age <18 sending the photo, while only 18 address the recipient. The law in this area can be complex and detailed, taking into account the age of the sender, the intentions of the sender, and the nature of the relationship between the sender and the recipient and the behavior of the recipient.

Laws regarding sexting vary greatly. Sexting may be a misdemeanor or a felony, depending on the state, the specific behavior, and the frequency. In the United States, 11 state laws include a diversion remedy—an option to pursue the case outside of the criminal juvenile system; 10 laws require counseling or another informal sanction; 11 states laws have the potential for misdemeanor punishment; and 4 state laws have the potential for felony punishment.14 Depending on the criminal charge, the perpetrator may have to register as a sex offender. For example, in some jurisdictions, a conviction for possession of child pornography requires sex offender registration. Thirty-eight states include juvenile sex offenders in their sex offender registries. Other states require juveniles to register if they are age ≥15 years or have been tried as an adult.15

The frequency of police involvement in sexting cases also greatly varies. A national study examining the characteristics of youth sexting cases revealed that law enforcement agencies handled approximately 3,477 cases of youth-produced sexual photos in 2008 and 2009.16 Situations that involved an adult or a minor engaged in malicious, nonconsensual, or abusive behavior comprised two-thirds of cases. Arrests occurred in 62% of the adult-involved cases and 36% of the aggravated youth-only cases. Arrests occurred in only 18% of investigated non-aggravated youth-only cases. Table 2 describes recent American sexting legal cases and their outcomes.

In K’s case, depending on the jurisdiction, K or her ex-boyfriend may be subject to arrest for child pornography, revenge pornography, or sexting.

 

 

 

Potential social and psychiatric consequences

What are the social and psychiatric ramifications for K? Aside from potential legal consequences of sexting, K is experiencing psychological and social consequences. She has developed depressive symptoms and suicidal ideation. Her ex-boyfriend’s dissemination of her nude photos on the school computer could be interpreted as cyberbullying. (The National Center for Missing and Exploited Children defines cyberbullying as “bullying through the use of technology or electronic devices such as telephones, cell phones, computers, and the Internet.”17 All 50 states have enacted laws against bullying; 48 states have electronic harassment in their bullying laws; and 22 states have laws specifically referencing “cyberbullying.”)

Her depressive symptoms developed in response to her feelings of guilt and shame related to sexting as well as the subsequent peer harassment. She is refusing to return to school because of her concerns about bullying. A careful inquiry into suicidality should be part of the evaluation when sexting has led to psychiatric symptoms. Several cases of sexting and cyberbullying have ended in suicide (Table 3).

How to ask patients about sexting

To screen patients for sexting, clinicians need to develop a new skill set, which at first may be uncomfortable. However, the questions to ask are not all that different from other questions about adolescent and young adult sexuality. The importance of patients seeing that we as physicians are comfortable with the topic and approachable about their sexual health cannot be overemphasized. When discussing sexting with patients, it is essential to:

  • explain that you are asking questions about their sexual health because they are important to overall health
  • engage patients in discussion in a nonthreatening and nonjudgmental way
  • develop rapport so patients feel comfortable disclosing behavior that may be embarrassing
  • listen to their stories and build a context for understanding their experiences. As you listen, ask questions when needed to help move the story along.

Sometimes when asking about topics that are uncomfortable, clinicians revert from open-ended to closed-ended questions, but when asking about a patient’s sexual life, it is especially important to be open-ended and ask questions in a nonjudgmental way. Contextualizing sexual questions by (for example) asking them while discussing the teen’s relationships will make them seem more natural.18 To best understand, inquire explicitly about specific behaviors, but do so without appearing voyeuristic.18

Sexting may precede sexual intercourse. Keep in mind that a patient may report that she (he) is not sexually active but still may be involved in sexting. Therefore, discuss sexting even if your patient reports not being sexually active. By understanding the prevalence of sexting among teens, you can ask questions in a normalizing way. Clinicians can inquire about sexting while discussing relationships and dating or online risk behaviors.

Also consider whether any of your patient’s sexual behaviors, including sext­ing, are the result of coercion: “Some of my patients tell me they feel pressured or coerced into having sex. Have you ever felt this way?”19 and “Have you ever been picked on or bullied? Is that still a problem?” are suggested safety screening questions about bullying,18 and one can also ask about specific cyberbullying behaviors.

Table 4 lists additional risk behaviors to ask about in a patient who has been sext­ing. Sexting is a widely accepted behavior among adolescents, yet many adolescents and parents are unaware of the legal implications of such a behavior. As physicians, we have a role in educating our patients about potentially harmful behaviors, and to do so in a manner that is not viewed as lecturing, which may result in receiving poor-quality information.

Bottom Line

Although it may seem harmless to adolescents, sexting has potentially serious legal, social, and psychological consequences. Sexting may co-occur with other risk behaviors, such as substance abuse. Clinicians should be prepared to ask patients about sexting, educate them about the risks, and consider whether patients also are engaging in other risky behaviors.

Related Resources

  • American Academy of Pediatrics. Talking to kids and teens about social media and sexting.
  • Connect Safely. Tips for dealing with teen sexting. http://www.connectsafely.org/tips-for-dealing-with-teen-sexting.
References

1. Mitchell KJ, Finkelhor D, Jones LM, et al. Prevalence and characteristics of youth sexting: a national study. Pediatrics. 2012;129(1):13-20.
2. Lenhart A. Teens and sexting. The Pew Research Center. http://www.pewinternet.org/2009/12/15/teens-and-sexting. Published December 15, 2009. Accessed October 31, 2017.
3. Temple JR, Paul JA, van den Berg P, et al. Teen sexting and its association with sexual behaviors. Arch Pediatr Adolesc Med. 2012;166(9):828-833.
4. Gordon-Messer D, Bauermeister JA, Grodzinski A, et al. Sexting among young adults. J Adolesc Health. 2013;52(3):301-306.
5. Henderson L. Sexting and sexual relationships among teens and young adults. McNair Scholars Research Journal. 2011;7(1):31-39.
6. The National Campaign to Prevent Teen and Unplanned Pregnancy. Sex and tech: results from a survey of teens and young adults. https://thenationalcampaign.org/sites/default/files/resource-primary-download/sex_and_tech_summary.pdf. Published December 2008. Accessed October 31, 2017.
7. Strassberg DS, McKinnon RK, Sustaíta MA, et al. Sexting by high school students: an exploratory and descriptive study. Arch Sex Behav. 2013;42(1):15-21.
8. Houck CD, Barker D, Rizzo C, et al. Sexting and sexual behavior in at-risk adolescents. Pediatrics. 2014;133(2):e276-e282.
9. Benotsch EG, Snipes DJ, Martin AM, et al. Sexting, substance use, and sexual risk behavior in young adults. J Adolesc Health. 2013;52(3):307-313.
10. Delevi R, Weisskirch RS. Personality factors as predictors of sexting. Comput Human Behav. 2013;29(6):2589-2594.
11. Temple JR, Choi H. Longitudinal association between teen sexting and sexual behavior. Pediatrics. 2014;134(5):1287-1292.
12. McEwan M, Friedman SH. Violence by parents against their children: reporting of maltreatment suspicions, child protection, and risk in mental illness. Psych Clin North Am. 2016;39(4):691-700.
13. Citron DK, Franks MA. Criminalizing revenge porn. Wake Forest Law Review. 2014;49:345-391.
14. Hinduja S, Patchin JW. State cyberbullying laws: a brief review of state cyberbullying laws and policies. Cyberbullying Research Center. https://cyberbullying.org/Bullying-and-Cyberbullying-Laws.pdf. Updated 2016. Accessed October 31, 2017.
15. Beitsch R. States slowly scale back juvenile sex offender registries. The Pew Charitable Trusts. http://www.pewtrusts.org/en/research-and-analysis/blogs/stateline/2015/11/19/states-slowly-scale-back-juvenile-sex-offender-registries. Published November 19, 2015. Accessed October 31, 2017.
16. Wolak J, Finkelhor D, Mitchell KJ. How often are teens arrested for sexting? Data from a national sample of police cases. Pediatrics. 2012;129(1):4-12.
17. The Campus School at Boston College. Bullying prevention policy. https://www.bc.edu/bc-web/schools/lsoe/sites/campus-school/who-we-are/policies-and-procedures/bullying-prevention-policy.html. Accessed October 31, 2017.
18. Goldenring JM, Rosen DS. Getting into adolescent heads: an essential update. Contemporary Pediatrics. 2004;21(1):64.
19. Klein DA, Goldenring JM, Adelman WP. HEEADSSS 3.0: the psychosocial interview for adolescents updated for a new century fueled by media. Contemporary Pediatrics. http://contemporarypediatrics.modernmedicine.com/contemporary-pediatrics/content/tags/adolescent-medicine/heeadsss-30-psychosocial-interview-adolesce?page=full. Published January 1, 2014. Accessed October 31, 2017.

References

1. Mitchell KJ, Finkelhor D, Jones LM, et al. Prevalence and characteristics of youth sexting: a national study. Pediatrics. 2012;129(1):13-20.
2. Lenhart A. Teens and sexting. The Pew Research Center. http://www.pewinternet.org/2009/12/15/teens-and-sexting. Published December 15, 2009. Accessed October 31, 2017.
3. Temple JR, Paul JA, van den Berg P, et al. Teen sexting and its association with sexual behaviors. Arch Pediatr Adolesc Med. 2012;166(9):828-833.
4. Gordon-Messer D, Bauermeister JA, Grodzinski A, et al. Sexting among young adults. J Adolesc Health. 2013;52(3):301-306.
5. Henderson L. Sexting and sexual relationships among teens and young adults. McNair Scholars Research Journal. 2011;7(1):31-39.
6. The National Campaign to Prevent Teen and Unplanned Pregnancy. Sex and tech: results from a survey of teens and young adults. https://thenationalcampaign.org/sites/default/files/resource-primary-download/sex_and_tech_summary.pdf. Published December 2008. Accessed October 31, 2017.
7. Strassberg DS, McKinnon RK, Sustaíta MA, et al. Sexting by high school students: an exploratory and descriptive study. Arch Sex Behav. 2013;42(1):15-21.
8. Houck CD, Barker D, Rizzo C, et al. Sexting and sexual behavior in at-risk adolescents. Pediatrics. 2014;133(2):e276-e282.
9. Benotsch EG, Snipes DJ, Martin AM, et al. Sexting, substance use, and sexual risk behavior in young adults. J Adolesc Health. 2013;52(3):307-313.
10. Delevi R, Weisskirch RS. Personality factors as predictors of sexting. Comput Human Behav. 2013;29(6):2589-2594.
11. Temple JR, Choi H. Longitudinal association between teen sexting and sexual behavior. Pediatrics. 2014;134(5):1287-1292.
12. McEwan M, Friedman SH. Violence by parents against their children: reporting of maltreatment suspicions, child protection, and risk in mental illness. Psych Clin North Am. 2016;39(4):691-700.
13. Citron DK, Franks MA. Criminalizing revenge porn. Wake Forest Law Review. 2014;49:345-391.
14. Hinduja S, Patchin JW. State cyberbullying laws: a brief review of state cyberbullying laws and policies. Cyberbullying Research Center. https://cyberbullying.org/Bullying-and-Cyberbullying-Laws.pdf. Updated 2016. Accessed October 31, 2017.
15. Beitsch R. States slowly scale back juvenile sex offender registries. The Pew Charitable Trusts. http://www.pewtrusts.org/en/research-and-analysis/blogs/stateline/2015/11/19/states-slowly-scale-back-juvenile-sex-offender-registries. Published November 19, 2015. Accessed October 31, 2017.
16. Wolak J, Finkelhor D, Mitchell KJ. How often are teens arrested for sexting? Data from a national sample of police cases. Pediatrics. 2012;129(1):4-12.
17. The Campus School at Boston College. Bullying prevention policy. https://www.bc.edu/bc-web/schools/lsoe/sites/campus-school/who-we-are/policies-and-procedures/bullying-prevention-policy.html. Accessed October 31, 2017.
18. Goldenring JM, Rosen DS. Getting into adolescent heads: an essential update. Contemporary Pediatrics. 2004;21(1):64.
19. Klein DA, Goldenring JM, Adelman WP. HEEADSSS 3.0: the psychosocial interview for adolescents updated for a new century fueled by media. Contemporary Pediatrics. http://contemporarypediatrics.modernmedicine.com/contemporary-pediatrics/content/tags/adolescent-medicine/heeadsss-30-psychosocial-interview-adolesce?page=full. Published January 1, 2014. Accessed October 31, 2017.

Issue
December 2017
Issue
December 2017
Page Number
35-41
Page Number
35-41
Publications
Publications
Article Type
Display Headline
Sexting: What are the clinical and legal implications?
Display Headline
Sexting: What are the clinical and legal implications?
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Article PDF Media