A sleeping beast: Obstructive sleep apnea and stroke

Article Type
Changed
Mon, 06/03/2019 - 08:03
Display Headline
A sleeping beast: Obstructive sleep apnea and stroke

Obstructive sleep apnea (OSA) is an independent risk factor for ischemic stroke and may also, infrequently, be a consequence of stroke. It is significantly underdiagnosed in the general population and is highly prevalent in patients who have had a stroke. Many patients likely had their stroke because of this chronic untreated condition.

This review focuses on OSA and its prevalence, consequences, and treatment in patients after a stroke.

DEFINING AND QUANTIFYING OSA

OSA is the most common type of sleep-disordered breathing.1,2 It involves repeated narrowing or complete collapse of the upper airway despite ongoing respiratory effort.3,4 Apneic episodes are terminated by arousals from hypoxemia or efforts to breathe.5 In contrast, central sleep apnea is characterized by a patent airway but lack of airflow due to absent respiratory effort.5

In OSA, the number of episodes of apnea (absent airflow) and hypopnea (reduced airflow) are added together and divided by hours of sleep to calculate the apnea-hypopnea index (AHI). OSA is diagnosed by either of the following3,4:

  • AHI of 5 or higher, with clinical symptoms related to OSA (described below)
  • AHI of 15 or higher, regardless of symptoms.

The AHI also defines OSA severity, as follows3:

  • Mild: AHI 5 to 15
  • Moderate: AHI 15 to 30
  • Severe: AHI greater than 30.

Diagnostic criteria (eg, definition of hypopnea, testing methods, and AHI thresholds) have varied over time, an important consideration when reviewing the literature.

OSA IS MORE COMMON THAN EXPECTED AFTER STROKE

In the most methodologically sound and generalizable study of this topic to date, the Wisconsin Sleep Cohort Study6 reported in 2013 that about 14% of men and 5% of women ages 30 to 70 have an AHI greater than 5 (using 4% desaturation to score hypopneic episodes) with daytime sleepiness. Other studies suggest that 80% to 90% of people with OSA are undiagnosed and untreated.1,7

The prevalence of OSA in patients who have had a stroke is much higher, ranging from 30% to 96% depending on the study methods and population.1,8–12 A 2010 meta-analysis11 of 29 studies reported that 72% of patients who had a stroke had an AHI greater than 5, and 29% had severe OSA. In this analysis, 7% of those with sleep-disordered breathing had central sleep apnea; still, these data indicate that the prevalence of OSA in these patients is about 5 times higher than in the general population.

RISK FACTORS MAY DIFFER IN STROKE POPULATION

Several risk factors for OSA have been identified.

Obesity is one of the strongest risk factors, with increasing body mass index (BMI) associated with increased OSA prevalence.4,6,13 However, obesity appears to be a less significant risk factor in patients who have had a stroke than in the general population. In the 2010 meta-analysis11 of OSA after stroke, the average BMI was only 26.4 kg/m2 (with obesity defined as a BMI > 30.0 kg/m2), and increasing BMI was not associated with increasing AHI.

Male sex and advanced age are also OSA risk factors.4,5 They remain significant in patients after a stroke; about 65% of poststroke patients who have OSA are men, and the older the patient, the more likely the AHI is greater than 10.11

Ethnicity and genetics may also play important roles in OSA risk, with roughly 25% of OSA prevalence estimated to have a genetic basis.14,15 Some risk factors for OSA such as craniofacial shape, upper airway anatomy, upper airway muscle dysfunction, increased respiratory chemosensitivity, and poor arousal threshold during sleep are likely determined by genetics and ethnicity.14,15 Compared with people of European origin, Asians have a similar prevalence of OSA, but at a much lower average BMI, suggesting that other factors are significant.14 Possible genetically determined anatomic risk factors have not been specifically studied in the poststroke population, but it can be assumed they remain relevant.

Several studies have tried to find an association between OSA and type, location, etiology, or pattern of stroke.10,11,16–19 Although some suggest links between cardioembolic stroke and OSA,16,20 or thrombolysis and OSA,10 most have found no association between OSA and stroke features.11,12,21,22

HOW DOES OSA INCREASE STROKE RISK?

Untreated severe OSA is associated with increased cardiovascular mortality,21,22 and OSA is an independent risk factor for incident stroke.23 A number of mechanisms may explain these relationships.

Intermittent hypoxemia and recurrent sympathetic arousals resulting from OSA are thought to lead to many of the comorbid conditions with which it is associated: hypertension, coronary artery disease, heart failure, arrhythmias, pulmonary hypertension, and stroke. Repetitive decreases in ventilation lead to oxygen desaturations that result in cycles of increased sympathetic outflow and eventual sustained nocturnal hypertension and daytime chronic hypertension.1,5,9,13 Also implicated are various changes in vasodilator and vasoconstrictor substances due to endothelial dysfunction and inflammation, which are thought to play a role in the atherogenic and prothrombotic states induced by OSA.1,5,13

Cerebral circulation is altered primarily by the changes in partial pressure of carbon dioxide (Pco2). During apnea, the Pco2 rises, causing vasodilation and increased blood flow. After the apnea resolves, there is hyperpnea with resultant decreased Pco2, and vasoconstriction. In a patient who already has vascular disease, the enhanced vasoconstriction could lead to ischemia.1,5

Changes in intrathoracic pressure result in distortion of cardiac architecture. When the patient tries to breathe against an occluded airway, the intrathoracic pressure becomes more and more negative, increasing preload and afterload. When this happens repeatedly every night for years, it leads to remodeling of the heart such as left and right ventricular hypertrophy, with reduced stroke volume, myocardial ischemia, and increased risk of arrhythmia.1,5,13

Untreated OSA is believed to predispose patients to develop atrial fibrillation through sympathetic overactivity, vascular inflammation, heart rate variability, and cardiac remodeling.24 As atrial fibrillation is a major risk factor for stroke, particularly cardioembolic stroke, it may be another pathway of increased stroke risk in OSA.16,20,25

 

 

CLINICAL MANIFESTATIONS OF OSA NOT OBVIOUS AFTER STROKE

OSA typically causes both daytime symptoms (excessive sleepiness, poor concentration, morning headache, depressive symptoms) and nighttime signs and symptoms (snoring, choking, gasping, night sweats, insomnia, nocturia, witnessed episodes of apnea).3,4,26 Unfortunately, because these are nonspecific, OSA is often underdiagnosed.4,26

Identifying OSA after a stroke may be a particular challenge, as patients often do not report classic symptoms, and the typical picture of OSA may have less predictive validity in these patients.1,27,28 Within the first 24 hours after a stroke, hypersomnia, snoring history, and age are not predictive of OSA.1 Patients found to have OSA after a stroke frequently do not have the traditional symptoms (sleepiness, snoring) seen in usual OSA patients. And they have higher rates of OSA at a younger age than the usual OSA patients, so age is not a predictive risk factor. In addition, daytime sleepiness and obesity are often absent or less prominent.1,9,27,28  Finally, typical OSA signs and symptoms may be attributed to the stroke itself or to comorbidities affecting the patient, lowering suspicion for OSA.

OSA MAY HINDER STROKE RECOVERY, WORSEN OUTCOMES

OSA, particularly when moderate to severe, is linked to pathophysiologic changes that can hinder recovery from a stroke.

Intermittent hypoxemia during sleep can worsen vascular damage of at-risk tissue: nocturnal hypoxemia correlates with white matter hyperintensities on magnetic resonance imaging, a marker of ischemic demyelination.29 Oxidative stress and release of inflammatory mediators associated with intermittent hypoxemia may impair vascular blood flow to brain tissue attempting to repair itself.30 In addition, sympathetic overactivity and Pco2 fluctuations associated with OSA may impede cerebral circulation.

Taken together, such ongoing nocturnal insults can lead to clinical consequences during this vulnerable period.

A 1996 study31 of patients recovering from a stroke found that an oxygen desaturation index (number of times that the blood oxygen level drops below a certain threshold, as measured by overnight oximetry) of more than 10 per hour was associated with worse functional recovery at discharge and at 3 and 12 months after discharge. This study also noted an association between time spent with oxygen saturations below 90% and the rate of death at 1 year.

A 2003 study32 reported that patients with an AHI greater than 10 by polysomnography spent an average of 13 days longer on the rehabilitation service and had worse functional and cognitive status on discharge, even after controlling for multiple confounders. Several subsequent studies have confirmed these and similar findings.8,33,34

OSA has also been linked to depression,35 which is common after stroke and may worsen outcomes.36 The interaction between OSA, depression, and poststroke outcomes warrants further study.

In the general population, OSA has been independently associated with increased risk of stroke or death from any cause.21,22,37 These associations have also been reported in the poststroke population: a 2014 meta-analysis found that OSA increased the risk of a repeat stroke (relative risk [RR] 1.8, 95% confidence interval [CI] 1.2–2.6) and all-cause mortality (RR 1.69, 95% CI 1.4–2.1).38

TESTING FOR OSA AFTER STROKE

Because of the high prevalence of OSA in patients who have had a stroke and the potential for worse outcomes associated with untreated OSA, there should be a low threshold for evaluating for OSA soon after stroke. Objective testing is required to qualify for therapy,  and the gold standard for diagnosis of OSA is formal polysomnography conducted in a sleep laboratory.2–4 Unfortunately, polysomnography may be unacceptable to some patients, is costly, and is resource-intensive, particularly in an inpatient or rehabilitation setting.28 Ideally, to optimize testing efficiency, patients should be screened for the likelihood of OSA before polysomnography is ordered.

Questionnaires can help determine the need for further testing

Questionnaires developed to assess OSA risk39 include the following:

The Berlin questionnaire, developed in 1999, has 10 questions assessing daytime and nighttime signs and symptoms and presence of hypertension.

The STOP questionnaire, developed in 2008, assesses snoring, tiredness, observed apneic episodes, and elevated blood pressure.

The STOP-BANG questionnaire, published in 2010, includes the STOP questions plus BMI over 35 kg/m2, age over 50, neck circumference over 41 cm, and male gender.

A 2017 meta-analysis39 of 108 studies with nearly 50,000 people found that the STOP-BANG questionnaire performed best with regard to sensitivity and diagnostic odds ratio, but with poor specificity.

These screening tools and modified versions of them have also been evaluated in patients who have had a stroke.

In 2015, Boulos et al28 found that the STOP-BAG (a version of STOP-BANG that excludes neck circumference) and the 4-variable (4V) questionnaire (sex, BMI, blood pressure, snoring) had moderate predictive value for OSA within 6 months after sroke.

In 2016, Katzan et al40 found that the STOP-BAG2 (STOP-BAG criteria plus continuous variables for BMI and age) had a high sensitivity for polysomnographically diagnosed OSA within the first year after a stroke. The specificity was significantly better than the STOP-BANG or the STOP-BAG questionnaire, although it remained suboptimal at 60.5%.

In 2017, Sico et al41 developed and assessed the SLEEP Inventory (sex, left heart failure, Epworth Sleepiness Scale, enlarged neck, weight in pounds, insulin resistance or diabetes, and National Institutes of Health Stroke Scale) and found that it outperformed the Berlin and STOP-BANG questionnaires in the poststroke setting. The SLEEP Inventory had the best specificity and negative predictive value, and a slightly better ability to correctly classify patients as having OSA or not, classifying 80% of patients correctly.

These newer screening tools (eg, STOP-BAG, STOP-BAG2, SLEEP) can be used to identify with reasonable accuracy which patients need definitive testing after stroke.

Pulse oximetry is another possible screening tool          

Overnight pulse oximetry may also help screen for sleep apnea and stratify risk after a stroke. A 2012 study42 of overnight oximetry to screen patients before surgery found that the oxygen desaturation index was significantly associated with the AHI measured by polysomnography. However, oximetry testing cannot distinguish between OSA and central sleep apnea, so it is insufficient to diagnose OSA or qualify patients for therapy. Further study is needed to examine the ability of overnight pulse oximetry to screen or to stratify risk for OSA after stroke.

Polysomnography vs home testing

Polysomnography is the gold standard for diagnosing OSA. Benefits include technical support and trouble-shooting, determining relationships between OSA, body position, and sleep stage, and the ability to intervene with treatment.2 However, polysomnography can be cumbersome, costly, and resource-intensive.

A home sleep apnea test, ie, an unattended, limited-channel sleep study, may be an acceptable alternative.2–4,43,44 Home testing does not require a sleep technologist to be present during testing, uses fewer sensors, and is less expensive than overnight polysomnography, but its utility can be limited: it fails to accurately discriminate between episodes of OSA and central sleep apnea, there is potential for false-negative results, and it can underestimate sleep apnea burden because it does not measure sleep.2

Institutional resources and logistics may influence the choice of diagnostic modality. No data exist on outcomes from different diagnostic testing methods in poststroke patients. Further research is needed.

 

 

POSITIVE AIRWAY PRESSURE THERAPY: BENEFITS, CHALLENGES, ALTERNATIVES

The first-line treatment for OSA is positive airway pressure (PAP).3 For most patients, this is continuous PAP (CPAP) or autoadjusting PAP (APAP). In some instances, particularly for those who cannot tolerate CPAP or who have comorbid hypoventilation, bilevel PAP (BPAP) may be indicated. More advanced PAP therapies are unlikely to be used after stroke.

PAP therapy is associated with reduced daytime sleepiness, improved mood, normalization of sleep architecture, improved systemic and pulmonary artery blood pressure, reduced rates of atrial fibrillation after ablation, and improved insulin sensitivity.45–49 Whether it reduces the risk of cardiovascular events, including stroke, remains controversial; most data suggest that it does not.50,51 However, when adherence to PAP therapy is considered rather than intention to treat, treatment has been found to lead to improved cardiovascular outcomes.52

Mixed evidence of benefits after stroke

Observational studies provide evidence that CPAP may help patients with OSA after stroke, although results are mixed.53–58 The studies ranged in size from 14 to 105 patients, enrolled patients with mostly moderate to severe OSA, and followed patients from 10 days to 7 years. Adherence to therapy was generally good in the short term (50%–70%), but only  15% to 30% of patients remained adherent at 5 to 7 years. Variable outcomes were reported, with some studies finding improved symptoms in the near term and mixed evidence of cardiovascular benefit in the longer ones. However, as these studies lacked randomization, drawing definitive conclusions on CPAP efficacy is difficult.

mckee_obstructivesleepapnea_t1.jpg
Several short-term randomized controlled trials of CPAP have been performed in patients after stroke. A 2018 meta-analysis59 included 10 such trials with a total of 564 patients (range 30–140 patients), with most having 1 to 3 months of follow-up (range 1 week to over 5 years). Eight of the 10 studies are summarized in Table 1 (1 study was omitted because many of the patients had central sleep apnea, and 1 was primarily a feasibility study).60–67

Patients were enrolled in the index admission or when starting a rehabilitation service—generally 2 to 3 weeks after their stroke. No clear association was found between the timing of initiating PAP therapy and outcomes. All patients had ischemic strokes, but few details were provided regarding stroke location, size, and severity. Exclusion criteria included severe underlying cardiopulmonary disease, confusion, severe stroke with marked impairment, and inability to cooperate. Almost all patients had moderate to severe OSA, and patients with central sleep apnea were excluded.

The major outcomes examined were drop-out rates, PAP adherence, and neurologic improvement based on neurologic functional scales (National Institutes of Health Stroke Scale and Canadian Neurologic Scale). As expected, dropout rates were higher in patients randomized to CPAP (OR 1.83, 95% CI 1.05–3.21, P = .03), although overall adherence was better than anticipated, with mean CPAP use across trials of 4.5 hours per night (95% CI 3.97–5.08) and with about 50% to 60% of patients adhering to therapy for at least 4 hours nightly.

Improvement in neurologic outcomes favored CPAP (standard mean difference 0.54, 95% CI 0.026–1.05), although considerable heterogeneity was seen. Improved sleepiness outcomes were inconsistent. Major cardiovascular outcomes were reported in only 2 studies (using the same data set) and showed delayed time to the next cardiovascular event for those treated with CPAP but no difference in cardiovascular event-free survival.

PAP poses more challenges after stroke

The primary limitation to PAP therapy is poor acceptance and adherence to therapy.59 High rates of refusal of therapy and difficulty complying with treatment have been noted in the poststroke population, although recent studies have reported better adherence rates. How rates of adherence play out in real-world settings, outside of the controlled environment of a research study, has yet to be determined.

In general, CPAP adherence is affected by claustrophobia, difficulty tolerating a mask, problems with pressure intolerance, irritating air leaks, nasal congestion, and naso-oral dryness. Many such barriers can be overcome with use of a properly fitted mask, an appropriate pressure setting, heated humidification, nasal sprays (eg, saline, inhaled steroids), and education, encouragement, and reassurance.

After a stroke, additional obstacles may impede the ability to use PAP therapy.68 Facial paresis (hemi- or bifacial) may make fitting of the mask problematic. Paralysis or weakness of the extremities may limit the ability to adjust or remove a mask. Aphasia can impair communication and understanding of the need to use PAP therapy, and upper-airway problems related to stroke, including dysphagia, may lead to pressure intolerance or risk of aspiration. Finally, a lack of perceived benefit, particularly if the patient does not have daytime sleepiness, may limit motivation.

Consider alternatives

For patients unlikely to succeed with PAP therapy, there are alternatives. Surgery and oral appliances are not usually realistic options in the setting of recent stroke, but positional therapy, including the use of body positioners to prevent supine sleep, as well as elevating the head of the bed, may be of some benefit.69,70 A nasopharyngeal airway stenting device (nasal trumpet) may also be tolerated by some patients.

mckee_obstructivesleepapnea_f1.jpg
%3Cp%3EFigure%201.%20Managing%20obstructive%20sleep%20apnea%20after%20stroke.%3C%2Fp%3E
Avoiding or minimizing sedating medications that may worsen OSA, such as benzodiazepines and opioids, should be considered.3 Oxygen therapy, while helping to maintain oxygen saturation during sleep, does not prevent airway collapse, and its role for treating OSA in patients after stroke is unclear.

A proposed algorithm for screening, diagnosing, and treating OSA in patients after stroke is presented in Figure 1.

References
  1. Selim B, Roux FJ. Stroke and sleep disorders. Sleep Med Clin 2012; 7(4):597–607. doi:10.1016/j.jsmc.2012.08.007
  2. Kapur VK, Auckley DH, Chowdhuri S, et al. Clinical practice guideline for diagnostic testing for adult obstructive sleep apnea: an American Academy of Sleep Medicine Clinical Practice Guideline. J Clin Sleep Med 2017; 13(3):479–504. doi:10.5664/jcsm.6506
  3. Epstein LJ, Kristo D, Strollo PJ Jr, et al; Adult Obstructive Sleep Apnea Task Force of the American Academy of Sleep Medicine. Clinical guideline for the evaluation, management, and long-term care of obstructive sleep apnea in adults. J Clin Sleep Med 2009; 5(3):263–276. pmid:19960649
  4. Patil SP, Schneider H, Schwartz AR, Smith PL. Adult obstructive sleep apnea: pathophysiology and diagnosis. Chest 2007; 132(1):325–337. doi:10.1378/chest.07-0040
  5. Dempsey JA, Veasey SC, Morgan BJ, O’Donnell CP. Pathophysiology of sleep apnea. Physiol Rev 2010; 90(1):47–112. doi:10.1152/physrev.00043.2008
  6. Peppard PE, Young T, Barnet JH, Palta M, Hagen EW, Hla KM. Increased prevalence of sleep-disordered breathing in adults. Am J Epidemiol 2013; 177(9):1006–1014. doi:10.1093/aje/kws342
  7. Redline S, Sotres-Alvarez D, Loredo J, et al. Sleep-disordered breathing in Hispanic/Latino individuals of diverse backgrounds. The Hispanic Community Health Study/Study of Latinos. Am J Resp Crit Care Med 2014; 189(3):335–344. doi:10.1164/rccm.201309-1735OC
  8. Aaronson JA, van Bennekom CA, Hofman WF, et al. Obstructive sleep apnea is related to impaired cognitive and functional status after stroke. Sleep 2015; 38(9):1431–1437. doi:10.5665/sleep.4984
  9. Sharma S, Culebras A. Sleep apnoea and stroke. Stroke Vasc Neurol 2016; 1(4):185–191. doi:10.1136/svn-2016-000038
  10. Huhtakangas JK, Huhtakangas J, Bloigu R, Saaresranta T. Prevalence of sleep apnea at the acute phase of ischemic stroke with or without thrombolysis. Sleep Med 2017; 40:40–46. doi:10.1016/j.sleep.2017.08.018
  11. Johnson KG, Johnson DC. Frequency of sleep apnea in stroke and TIA patients: a meta-analysis. J Clin Sleep Med 2010; 6(2):131–137. pmid:20411688
  12. Iranzo A, Santamaria J, Berenguer J, Sanchez M, Chamorro A. Prevalence and clinical importance of sleep apnea in the first night after cerebral infarction. Neurology 2002; 58:911–916. pmid:11914407
  13. Javaheri S, Barbe F, Campos-Rodriguez F, et al. Sleep apnea: types, mechanisms, and clinical cardiovascular consequences. J Am Coll Cardiol 2017; 69(7):841–858. doi:10.1016/j.jacc.2016.11.069
  14. Dudley KA, Patel SR. Disparities and genetic risk factors in obstructive sleep apnea. Sleep Med 2016; 18:96–102. doi:10.1016/j.sleep.2015.01.015
  15. Redline S, Tishler PV. The genetics of sleep apnea. Sleep Med Rev 2000; 4(6):583–602. doi:10.1053/smrv.2000.0120
  16. Lipford MC, Flemming KD, Calvin AD, et al. Associations between cardioembolic stroke and obstructive sleep apnea. Sleep 2015; 38(11):1699–1705. doi:10.5665/sleep.5146
  17. Wang Y, Wang Y, Chen J, Yi X, Dong S, Cao L. Stroke patterns, topography, and etiology in patients with obstructive sleep apnea hypopnea syndrome. Int J Clin Exp Med 2017; 10(4):7137–7143.
  18. Fisse AL, Kemmling A, Teuber A, et al. The association of lesion location and sleep related breathing disorder in patients with acute ischemic stroke. PLoS One 2017; 12(1):e0171243. doi:10.1371/journal.pone.0171243
  19. Brown DL, Mowla A, McDermott M, et al. Ischemic stroke subtype and presence of sleep-disordered breathing: the BASIC sleep apnea study. J Stroke Cerebrovasc Dis 2015; 24(2):388–393. doi:10.1016/j.jstrokecerebrovasdis.2014.09.007
  20. Poli M, Philip P, Taillard J, et al. Atrial fibrillation as a major cause of stroke in apneic patients: a prospective study. Sleep Med 2017; 30:251–254. doi:10.1016/j.sleep.2015.07.031
  21. Young T, Finn L, Peppard PE, et al. Sleep disordered breathing and mortality: eighteen-year follow-up of the Wisconsin Sleep Cohort. Sleep 2008; 31(8):1071–1078. pmid:18714778
  22. Molnar MZ, Mucsi I, Novak M, et al. Association of incident obstructive sleep apnoea with outcomes in a large cohort of US veterans. Thorax 2015; 70(9):888–895. doi:10.1136/thoraxjnl-2015-206970
  23. Redline S, Yenokyan G, Gottlieb DJ, et al. Obstructive sleep apnea-hypopnea and incident stroke: the sleep heart health study. Am J Respir Crit Care Med 2010; 182(2):269–277. doi:10.1164/rccm.200911-1746OC
  24. Marulanda-Londono E, Chaturvedi S. The interplay between obstructive sleep apnea and atrial fibrillation. Fron Neurol 2017; 8:668. doi:10.3389/fneur.2017.00668
  25. Szymanski FM, Filipiak KJ, Platek AE, Hrynkiewicz-Szymanska A, Karpinski G, Opolski G. Assessment of CHADS2 and CHA 2DS 2-VASc scores in obstructive sleep apnea patients with atrial fibrillation. Sleep Breath 2015; 19(2):531–537. doi:10.1007/s11325-014-1042-5
  26. Stansbury RC, Strollo PJ. Clinical manifestations of sleep apnea. J Thoracic Dis 2015; 7(9):E298–E310. doi:10.3978/j.issn.2072-1439.2015.09.13
  27. Chan W, Coutts SB, Hanly P. Sleep apnea in patients with transient ischemic attack and minor stroke: opportunity for risk reduction of recurrent stroke? Stroke 2010; 41(12):2973–2975. doi:10.1161/STROKEAHA.110.596759
  28. Boulos MI, Wan A, Im J, et al. Identifying obstructive sleep apnea after stroke/TIA: evaluating four simple screening tools. Sleep Med 2016; 21:133–139. doi:10.1016/j.sleep.2015.12.013
  29. Patel SK, Hanly PJ, Smith EE, Chan W, Coutts SB. Nocturnal hypoxemia is associated with white matter hyperintensities in patients with a minor stroke or transient ischemic attack. J Clin Sleep Med 2015; 11(12):1417–1424. doi:10.5664/jcsm.5278
  30. McCarty MF, DiNicolantonio JJ, O’Keefe JH. NADPH oxidase, uncoupled endothelial nitric oxide synthase, and NF-KappaB are key mediators of the pathogenic impact of obstructive sleep apnea—therapeutic implications. J Integr Cardiol 2016; 2(5):367–374. doi:10.15761/JIC.1000177
  31. Good DC, Henkle JQ, Gelber D, Welsh J, Verhulst S. Sleep-disordered breathing and poor functional outcome after stroke. Stroke 1996; 27(2):252–259. pmid:8571419
  32. Kaneko Y, Hajek VE, Zivanovic V, Raboud J, Bradley TD. Relationship of sleep apnea to functional capacity and length of hospitalization following stroke. Sleep 2003; 26(3):293–297. pmid:12749548
  33. Yan-fang S, Yu-ping W. Sleep-disordered breathing: impact on functional outcome of ischemic stroke patients. Sleep Med 2009; 10(7):717–719. doi:10.1016/j.sleep.2008.08.006
  34. Kumar R, Suri JC, Manocha R. Study of association of severity of sleep disordered breathing and functional outcome in stroke patients. Sleep Med 2017; 34:50–56. doi:10.1016/j.sleep.2017.02.025
  35. Kerner NA, Roose SP. Obstructive sleep apnea is linked to depression and cognitive impairment: evidence and potential mechanisms. Am J Geriatr Psychiatry 2016; 24(6):496–508. doi:10.1016/j.jagp.2016.01.134
  36. Bartoli F, Lillia N, Lax A, et al. Depression after stroke and risk of mortality: a systematic review and meta-analysis. Stroke Res Treat 2013; 2013:862978. doi:10.1155/2013/862978
  37. Yaggi HK, Concato J, Kernan WN, Lichtman JH, Brass LM, Mohsenin V. Obstructive sleep apnea as a risk factor for stroke and death. N Engl J Med 2005; 353(19):2034–2041. doi:10.1056/NEJMoa043104
  38. Xie W, Zheng F, Song X. Obstructive sleep apnea and serious adverse outcomes in patients with cardiovascular or cerebrovascular disease: a PRISMA-compliant systematic review and meta-analysis. Medicine (Baltimore) 2014; 93(29):e336. doi:10.1097/MD.0000000000000336
  39. Chiu HY, Chen PY, Chuang LP, et al. Diagnostic accuracy of the Berlin questionnaire, STOP-BANG, STOP, and Epworth sleepiness scale in detecting obstructive sleep apnea: a bivariate meta-analysis. Sleep Med Rev 2017; 36:57–70. doi:10.1016/j.smrv.2016.10.004
  40. Katzan IL, Thompson NR, Uchino K, Foldvary-Schaefer N. A screening tool for obstructive sleep apnea in cerebrovascular patients. Sleep Med 2016; 21:70–76. doi:10.1016/j.sleep.2016.02.001
  41. Sico JJ, Yaggi HK, Ofner S, et al. Development, validation, and assessment of an ischemic stroke or transient ischemic attack-specific prediction tool for obstructive sleep apnea. J Stroke Cerebrovasc Dis 2017; 26(8):1745–1754. doi:10.1016/j.jstrokecerebrovasdis.2017.03.042
  42. Chung F, Liao P, Elsaid H, Islam S, Shapiro CM, Sun Y. Oxygen desaturation index from nocturnal oximetry: a sensitive and specific tool to detect sleep-disordered breathing in surgical patients. Anesth Analg 2012; 114(5):993–1000. doi:10.1213/ANE.0b013e318248f4f5
  43. Boulos MI, Elias S, Wan A, et al. Unattended hospital and home sleep apnea testing following cerebrovascular events. J Stroke Cerebrovasc Dis 2017; 26(1):143–149. doi:10.1016/j.jstrokecerebrovasdis.2016.09.001
  44. Saletu MT, Kotzian ST, Schwarzinger A, Haider S, Spatt J, Saletu B. Home sleep apnea testing is a feasible and accurate method to diagnose obstructive sleep apnea in stroke patients during in-hospital rehabilitation. J Clin Sleep Med 2018; 14(9):1495–1501. doi:10.5664/jcsm.7322
  45. Giles TL, Lasserson TJ, Smith BH, White J, Wright J, Cates CJ. Continuous positive airways pressure for obstructive sleep apnoea in adults. Cochrane Database Syst Rev 2006; (3):CD001106. doi:10.1002/14651858.CD001106.pub3
  46. Fatureto-Borges F, Lorenzi-Filho G, Drager LF. Effectiveness of continuous positive airway pressure in lowering blood pressure in patients with obstructive sleep apnea: a critical review of the literature. Integr Blood Press Control 2016; 9:43–47. doi:10.2147/IBPC.S70402
  47. Imran TF, Gharzipura M, Liu S, et al. Effect of continuous positive airway pressure treatment on pulmonary artery pressure in patients with isolated obstructive sleep apnea: a meta-analysis. Heart Fail Rev 2016; 21(5):591–598. doi:10.1007/s10741-016-9548-5
  48. Deng F, Raza A, Guo J. Treating obstructive sleep apnea with continuous positive airway pressure reduces risk of recurrent atrial fibrillation after catheter ablation: a meta-analysis. Sleep Med 2018; 46:5–11. doi:10.1016/j.sleep.2018.02.013
  49. Seetho IW, Wilding JPH. Sleep-disordered breathing, type 2 diabetes, and the metabolic syndrome. Chronic Resp Dis 2014; 11(4):257–275. doi:10.1177/1479972314552806
  50. Kim Y, Koo YS, Lee HY, Lee SY. Can continuous positive airway pressure reduce the risk of stroke in obstructive sleep apnea patients? A systematic review and meta-analysis. PloS ONE 2016; 11(1):e0146317. doi:10.1371/journal.pone.0146317
  51. Yu J, Zhou Z, McEvoy RD, et al. Association of positive airway pressure with cardiovascular events and death in adults with sleep apnea: a systematic review and meta-analysis. JAMA 2017; 318(2):156–166. doi:10.1001/jama.2017.7967
  52. Peker Y, Glantz H, Eulenburg C, Wegscheider K, Herlitz J, Thunström E. Effect of positive airway pressure on cardiovascular outcomes in coronary artery disease patients with nonsleepy obstructive sleep apnea. The RICCADSA randomized controlled trial. Am J Respir Crit Care Med 2016; 194(5):613–620. doi:10.1164/rccm.201601-0088OC
  53. Martinez-Garcia MA, Soler-Cataluna JJ, Ejarque-Martinez L, et al. Continuous positive airway pressure treatment reduces mortality in patients with ischemic stroke and obstructive sleep apnea: a 5-year follow-up study. Am J Respir Crit Care Med 2009; 180(1):36–41. doi:10.1164/rccm.200808-1341OC
  54. Broadley SA, Jorgensen L, Cheek A, et al. Early investigation and treatment of obstructive sleep apnoea after acute stroke. J Clin Neurosci 2007; 14(4):328–333. doi:10.1016/j.jocn.2006.01.017
  55. Wessendorf TE, Wang YM, Thilmann AF, Sorgenfrei U, Konietzko N, Teschler H. Treatment of obstructive sleep apnoea with nasal continuous positive airway pressure in stroke. Eur Respir J 2001; 18(4):623–629. pmid:11716165
  56. Bassetti CL, Milanova M, Gugger M. Sleep-disordered breathing and acute ischemic stroke: diagnosis, risk factors, treatment, evolution, and long-term clinical outcome. Stroke 2006; 37(4):967–972. doi:10.1161/01.STR.0000208215.49243.c3
  57. Palombini L, Guilleminault C. Stroke and treatment with nasal CPAP. Eur J Neurol 2006; 13(2):198–200. doi:10.1111/j.1468-1331.2006.01169.x
  58. Martínez-García MA, Campos-Rodríguez F, Soler-Cataluña JJ, Catalán-Serra P, Román-Sánchez P, Montserrat JM. Increased incidence of nonfatal cardiovascular events in stroke patients with sleep apnoea: effect of CPAP treatment. Eur Respir J 2012; 39(4):906–912. doi:10.1183/09031936.00011311
  59. Brill AK, Horvath T, Seiler A, et al. CPAP as treatment of sleep apnea after stroke: a meta-analysis of randomized trials. Neurology 2018; 90(14):e1222–e1230. doi:10.1212/WNL.0000000000005262
  60. Hsu C, Vennelle M, Li H, Engleman HM, Dennis MS, Douglas NJ. Sleep-disordered breathing after stroke: a randomised controlled trial of continuous positive airway pressure. J Neurol Neurosurg Psychiatry 2006; 77(10):1143–1149. doi:10.1136/jnnp.2005.086686
  61. Parra O, Sanchez-Armengol A, Bonnin M, et al. Early treatment of obstructive apnoea and stroke outcome: a randomised controlled trial. Eur Resp J 2011; 37(5):1128–1136. doi:10.1183/09031936.00034410
  62. Ryan CM, Bayley M, Green R, Murray BJ, Bradley TD. Influence of continuous positive airway pressure on outcomes of rehabilitation in stroke patients with obstructive sleep apnea. Stroke 2011; 42(4):1062–1067. doi:10.1161/STROKEAHA.110.597468
  63. Bravata DM, Concato J, Fried T, et al. Continuous positive airway pressure: evaluation of a novel therapy for patients with acute ischemic stroke. Sleep 2011; 34(9):1271–1277. doi:10.5665/SLEEP.1254
  64. Parra O, Sanchez-Armengol A, Capote F, et al. Efficacy of continuous positive airway pressure treatment on 5-year survival in patients with ischaemic stroke and obstructive sleep apnea: a randomized controlled trial. J Sleep Res 2015; 24(1):47–53. doi:10.1111/jsr.12181
  65. Khot SP, Davis AP, Crane DA, et al. Effect of continuous positive airway pressure on stroke rehabilitation: a pilot randomized sham-controlled trial. J Clin Sleep Med 2016; 12(7):1019–1026. doi:10.5664/jcsm.5940
  66. Aaronson JA, Hofman WF, van Bennekom CA, et al. Effects of continuous positive airway pressure on cognitive and functional outcome of stroke patients with obstructive sleep apnea: a randomized controlled trial. J Clin Sleep Med 2016; 12(4):533–541. doi:10.5664/jcsm.5684
  67. Gupta A, Shukla G, Afsar M, et al. Role of positive airway pressure therapy for obstructive sleep apnea in patients with stroke: a randomized controlled trial. J Clin Sleep Med 2018; 14(4):511–521. doi:10.5664/jcsm.7034
  68. Mello-Fujita L, Kim LJ, Palombini Lde O, et al. Treatment of obstructive sleep apnea syndrome associated with stroke. Sleep Med 2015; 16(6):691–696. doi:10.1016/j.sleep.2014.12.017
  69. Svatikova A, Chervin RD, Wing JJ, Sanchez BN, Migda EM, Brown DL. Positional therapy in ischemic stroke patients with obstructive sleep apnea. Sleep Med 2011; 12(3):262–266. doi:10.1016/j.sleep.2010.12.008
  70. Souza FJ, Genta PR, de Souza Filho AJ, Wellman A, Lorenzi-Filho G. The influence of head-of-bed elevation in patients with obstructive sleep apnea. Sleep Breath 2017; 21(4):815–820. doi:10.1007/s11325-017-1524-3
Article PDF
Author and Disclosure Information

Zachary McKee, BS
Medical student, Case Western Reserve University School of Medicine, Cleveland OH

Dennis H. Auckley, MD
Professor of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH; Pulmonary, Sleep, and Critical Care, MetroHealth Medical Center, Cleveland, OH

Address: Dennis H. Auckley MD, Pulmonary, Sleep, and Critical Care, MetroHealth Medical Center, BG 3-90, 2500 MetroHealth Drive, Cleveland, OH 44109; dauckley@metrohealth.org

Issue
Cleveland Clinic Journal of Medicine - 86(6)
Publications
Topics
Page Number
407-415
Legacy Keywords
obstructive sleep apnea, OSA, snoring, stroke, cerebrovascular accident, CVA, transient ischemic attack, TIA, continuous positive airway pressure, CPAP, polysomnography, sleep study, apnea-hypoponea index, AHI, Zachary McKee, Dennis Auckley
Sections
Author and Disclosure Information

Zachary McKee, BS
Medical student, Case Western Reserve University School of Medicine, Cleveland OH

Dennis H. Auckley, MD
Professor of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH; Pulmonary, Sleep, and Critical Care, MetroHealth Medical Center, Cleveland, OH

Address: Dennis H. Auckley MD, Pulmonary, Sleep, and Critical Care, MetroHealth Medical Center, BG 3-90, 2500 MetroHealth Drive, Cleveland, OH 44109; dauckley@metrohealth.org

Author and Disclosure Information

Zachary McKee, BS
Medical student, Case Western Reserve University School of Medicine, Cleveland OH

Dennis H. Auckley, MD
Professor of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH; Pulmonary, Sleep, and Critical Care, MetroHealth Medical Center, Cleveland, OH

Address: Dennis H. Auckley MD, Pulmonary, Sleep, and Critical Care, MetroHealth Medical Center, BG 3-90, 2500 MetroHealth Drive, Cleveland, OH 44109; dauckley@metrohealth.org

Article PDF
Article PDF
Related Articles

Obstructive sleep apnea (OSA) is an independent risk factor for ischemic stroke and may also, infrequently, be a consequence of stroke. It is significantly underdiagnosed in the general population and is highly prevalent in patients who have had a stroke. Many patients likely had their stroke because of this chronic untreated condition.

This review focuses on OSA and its prevalence, consequences, and treatment in patients after a stroke.

DEFINING AND QUANTIFYING OSA

OSA is the most common type of sleep-disordered breathing.1,2 It involves repeated narrowing or complete collapse of the upper airway despite ongoing respiratory effort.3,4 Apneic episodes are terminated by arousals from hypoxemia or efforts to breathe.5 In contrast, central sleep apnea is characterized by a patent airway but lack of airflow due to absent respiratory effort.5

In OSA, the number of episodes of apnea (absent airflow) and hypopnea (reduced airflow) are added together and divided by hours of sleep to calculate the apnea-hypopnea index (AHI). OSA is diagnosed by either of the following3,4:

  • AHI of 5 or higher, with clinical symptoms related to OSA (described below)
  • AHI of 15 or higher, regardless of symptoms.

The AHI also defines OSA severity, as follows3:

  • Mild: AHI 5 to 15
  • Moderate: AHI 15 to 30
  • Severe: AHI greater than 30.

Diagnostic criteria (eg, definition of hypopnea, testing methods, and AHI thresholds) have varied over time, an important consideration when reviewing the literature.

OSA IS MORE COMMON THAN EXPECTED AFTER STROKE

In the most methodologically sound and generalizable study of this topic to date, the Wisconsin Sleep Cohort Study6 reported in 2013 that about 14% of men and 5% of women ages 30 to 70 have an AHI greater than 5 (using 4% desaturation to score hypopneic episodes) with daytime sleepiness. Other studies suggest that 80% to 90% of people with OSA are undiagnosed and untreated.1,7

The prevalence of OSA in patients who have had a stroke is much higher, ranging from 30% to 96% depending on the study methods and population.1,8–12 A 2010 meta-analysis11 of 29 studies reported that 72% of patients who had a stroke had an AHI greater than 5, and 29% had severe OSA. In this analysis, 7% of those with sleep-disordered breathing had central sleep apnea; still, these data indicate that the prevalence of OSA in these patients is about 5 times higher than in the general population.

RISK FACTORS MAY DIFFER IN STROKE POPULATION

Several risk factors for OSA have been identified.

Obesity is one of the strongest risk factors, with increasing body mass index (BMI) associated with increased OSA prevalence.4,6,13 However, obesity appears to be a less significant risk factor in patients who have had a stroke than in the general population. In the 2010 meta-analysis11 of OSA after stroke, the average BMI was only 26.4 kg/m2 (with obesity defined as a BMI > 30.0 kg/m2), and increasing BMI was not associated with increasing AHI.

Male sex and advanced age are also OSA risk factors.4,5 They remain significant in patients after a stroke; about 65% of poststroke patients who have OSA are men, and the older the patient, the more likely the AHI is greater than 10.11

Ethnicity and genetics may also play important roles in OSA risk, with roughly 25% of OSA prevalence estimated to have a genetic basis.14,15 Some risk factors for OSA such as craniofacial shape, upper airway anatomy, upper airway muscle dysfunction, increased respiratory chemosensitivity, and poor arousal threshold during sleep are likely determined by genetics and ethnicity.14,15 Compared with people of European origin, Asians have a similar prevalence of OSA, but at a much lower average BMI, suggesting that other factors are significant.14 Possible genetically determined anatomic risk factors have not been specifically studied in the poststroke population, but it can be assumed they remain relevant.

Several studies have tried to find an association between OSA and type, location, etiology, or pattern of stroke.10,11,16–19 Although some suggest links between cardioembolic stroke and OSA,16,20 or thrombolysis and OSA,10 most have found no association between OSA and stroke features.11,12,21,22

HOW DOES OSA INCREASE STROKE RISK?

Untreated severe OSA is associated with increased cardiovascular mortality,21,22 and OSA is an independent risk factor for incident stroke.23 A number of mechanisms may explain these relationships.

Intermittent hypoxemia and recurrent sympathetic arousals resulting from OSA are thought to lead to many of the comorbid conditions with which it is associated: hypertension, coronary artery disease, heart failure, arrhythmias, pulmonary hypertension, and stroke. Repetitive decreases in ventilation lead to oxygen desaturations that result in cycles of increased sympathetic outflow and eventual sustained nocturnal hypertension and daytime chronic hypertension.1,5,9,13 Also implicated are various changes in vasodilator and vasoconstrictor substances due to endothelial dysfunction and inflammation, which are thought to play a role in the atherogenic and prothrombotic states induced by OSA.1,5,13

Cerebral circulation is altered primarily by the changes in partial pressure of carbon dioxide (Pco2). During apnea, the Pco2 rises, causing vasodilation and increased blood flow. After the apnea resolves, there is hyperpnea with resultant decreased Pco2, and vasoconstriction. In a patient who already has vascular disease, the enhanced vasoconstriction could lead to ischemia.1,5

Changes in intrathoracic pressure result in distortion of cardiac architecture. When the patient tries to breathe against an occluded airway, the intrathoracic pressure becomes more and more negative, increasing preload and afterload. When this happens repeatedly every night for years, it leads to remodeling of the heart such as left and right ventricular hypertrophy, with reduced stroke volume, myocardial ischemia, and increased risk of arrhythmia.1,5,13

Untreated OSA is believed to predispose patients to develop atrial fibrillation through sympathetic overactivity, vascular inflammation, heart rate variability, and cardiac remodeling.24 As atrial fibrillation is a major risk factor for stroke, particularly cardioembolic stroke, it may be another pathway of increased stroke risk in OSA.16,20,25

 

 

CLINICAL MANIFESTATIONS OF OSA NOT OBVIOUS AFTER STROKE

OSA typically causes both daytime symptoms (excessive sleepiness, poor concentration, morning headache, depressive symptoms) and nighttime signs and symptoms (snoring, choking, gasping, night sweats, insomnia, nocturia, witnessed episodes of apnea).3,4,26 Unfortunately, because these are nonspecific, OSA is often underdiagnosed.4,26

Identifying OSA after a stroke may be a particular challenge, as patients often do not report classic symptoms, and the typical picture of OSA may have less predictive validity in these patients.1,27,28 Within the first 24 hours after a stroke, hypersomnia, snoring history, and age are not predictive of OSA.1 Patients found to have OSA after a stroke frequently do not have the traditional symptoms (sleepiness, snoring) seen in usual OSA patients. And they have higher rates of OSA at a younger age than the usual OSA patients, so age is not a predictive risk factor. In addition, daytime sleepiness and obesity are often absent or less prominent.1,9,27,28  Finally, typical OSA signs and symptoms may be attributed to the stroke itself or to comorbidities affecting the patient, lowering suspicion for OSA.

OSA MAY HINDER STROKE RECOVERY, WORSEN OUTCOMES

OSA, particularly when moderate to severe, is linked to pathophysiologic changes that can hinder recovery from a stroke.

Intermittent hypoxemia during sleep can worsen vascular damage of at-risk tissue: nocturnal hypoxemia correlates with white matter hyperintensities on magnetic resonance imaging, a marker of ischemic demyelination.29 Oxidative stress and release of inflammatory mediators associated with intermittent hypoxemia may impair vascular blood flow to brain tissue attempting to repair itself.30 In addition, sympathetic overactivity and Pco2 fluctuations associated with OSA may impede cerebral circulation.

Taken together, such ongoing nocturnal insults can lead to clinical consequences during this vulnerable period.

A 1996 study31 of patients recovering from a stroke found that an oxygen desaturation index (number of times that the blood oxygen level drops below a certain threshold, as measured by overnight oximetry) of more than 10 per hour was associated with worse functional recovery at discharge and at 3 and 12 months after discharge. This study also noted an association between time spent with oxygen saturations below 90% and the rate of death at 1 year.

A 2003 study32 reported that patients with an AHI greater than 10 by polysomnography spent an average of 13 days longer on the rehabilitation service and had worse functional and cognitive status on discharge, even after controlling for multiple confounders. Several subsequent studies have confirmed these and similar findings.8,33,34

OSA has also been linked to depression,35 which is common after stroke and may worsen outcomes.36 The interaction between OSA, depression, and poststroke outcomes warrants further study.

In the general population, OSA has been independently associated with increased risk of stroke or death from any cause.21,22,37 These associations have also been reported in the poststroke population: a 2014 meta-analysis found that OSA increased the risk of a repeat stroke (relative risk [RR] 1.8, 95% confidence interval [CI] 1.2–2.6) and all-cause mortality (RR 1.69, 95% CI 1.4–2.1).38

TESTING FOR OSA AFTER STROKE

Because of the high prevalence of OSA in patients who have had a stroke and the potential for worse outcomes associated with untreated OSA, there should be a low threshold for evaluating for OSA soon after stroke. Objective testing is required to qualify for therapy,  and the gold standard for diagnosis of OSA is formal polysomnography conducted in a sleep laboratory.2–4 Unfortunately, polysomnography may be unacceptable to some patients, is costly, and is resource-intensive, particularly in an inpatient or rehabilitation setting.28 Ideally, to optimize testing efficiency, patients should be screened for the likelihood of OSA before polysomnography is ordered.

Questionnaires can help determine the need for further testing

Questionnaires developed to assess OSA risk39 include the following:

The Berlin questionnaire, developed in 1999, has 10 questions assessing daytime and nighttime signs and symptoms and presence of hypertension.

The STOP questionnaire, developed in 2008, assesses snoring, tiredness, observed apneic episodes, and elevated blood pressure.

The STOP-BANG questionnaire, published in 2010, includes the STOP questions plus BMI over 35 kg/m2, age over 50, neck circumference over 41 cm, and male gender.

A 2017 meta-analysis39 of 108 studies with nearly 50,000 people found that the STOP-BANG questionnaire performed best with regard to sensitivity and diagnostic odds ratio, but with poor specificity.

These screening tools and modified versions of them have also been evaluated in patients who have had a stroke.

In 2015, Boulos et al28 found that the STOP-BAG (a version of STOP-BANG that excludes neck circumference) and the 4-variable (4V) questionnaire (sex, BMI, blood pressure, snoring) had moderate predictive value for OSA within 6 months after sroke.

In 2016, Katzan et al40 found that the STOP-BAG2 (STOP-BAG criteria plus continuous variables for BMI and age) had a high sensitivity for polysomnographically diagnosed OSA within the first year after a stroke. The specificity was significantly better than the STOP-BANG or the STOP-BAG questionnaire, although it remained suboptimal at 60.5%.

In 2017, Sico et al41 developed and assessed the SLEEP Inventory (sex, left heart failure, Epworth Sleepiness Scale, enlarged neck, weight in pounds, insulin resistance or diabetes, and National Institutes of Health Stroke Scale) and found that it outperformed the Berlin and STOP-BANG questionnaires in the poststroke setting. The SLEEP Inventory had the best specificity and negative predictive value, and a slightly better ability to correctly classify patients as having OSA or not, classifying 80% of patients correctly.

These newer screening tools (eg, STOP-BAG, STOP-BAG2, SLEEP) can be used to identify with reasonable accuracy which patients need definitive testing after stroke.

Pulse oximetry is another possible screening tool          

Overnight pulse oximetry may also help screen for sleep apnea and stratify risk after a stroke. A 2012 study42 of overnight oximetry to screen patients before surgery found that the oxygen desaturation index was significantly associated with the AHI measured by polysomnography. However, oximetry testing cannot distinguish between OSA and central sleep apnea, so it is insufficient to diagnose OSA or qualify patients for therapy. Further study is needed to examine the ability of overnight pulse oximetry to screen or to stratify risk for OSA after stroke.

Polysomnography vs home testing

Polysomnography is the gold standard for diagnosing OSA. Benefits include technical support and trouble-shooting, determining relationships between OSA, body position, and sleep stage, and the ability to intervene with treatment.2 However, polysomnography can be cumbersome, costly, and resource-intensive.

A home sleep apnea test, ie, an unattended, limited-channel sleep study, may be an acceptable alternative.2–4,43,44 Home testing does not require a sleep technologist to be present during testing, uses fewer sensors, and is less expensive than overnight polysomnography, but its utility can be limited: it fails to accurately discriminate between episodes of OSA and central sleep apnea, there is potential for false-negative results, and it can underestimate sleep apnea burden because it does not measure sleep.2

Institutional resources and logistics may influence the choice of diagnostic modality. No data exist on outcomes from different diagnostic testing methods in poststroke patients. Further research is needed.

 

 

POSITIVE AIRWAY PRESSURE THERAPY: BENEFITS, CHALLENGES, ALTERNATIVES

The first-line treatment for OSA is positive airway pressure (PAP).3 For most patients, this is continuous PAP (CPAP) or autoadjusting PAP (APAP). In some instances, particularly for those who cannot tolerate CPAP or who have comorbid hypoventilation, bilevel PAP (BPAP) may be indicated. More advanced PAP therapies are unlikely to be used after stroke.

PAP therapy is associated with reduced daytime sleepiness, improved mood, normalization of sleep architecture, improved systemic and pulmonary artery blood pressure, reduced rates of atrial fibrillation after ablation, and improved insulin sensitivity.45–49 Whether it reduces the risk of cardiovascular events, including stroke, remains controversial; most data suggest that it does not.50,51 However, when adherence to PAP therapy is considered rather than intention to treat, treatment has been found to lead to improved cardiovascular outcomes.52

Mixed evidence of benefits after stroke

Observational studies provide evidence that CPAP may help patients with OSA after stroke, although results are mixed.53–58 The studies ranged in size from 14 to 105 patients, enrolled patients with mostly moderate to severe OSA, and followed patients from 10 days to 7 years. Adherence to therapy was generally good in the short term (50%–70%), but only  15% to 30% of patients remained adherent at 5 to 7 years. Variable outcomes were reported, with some studies finding improved symptoms in the near term and mixed evidence of cardiovascular benefit in the longer ones. However, as these studies lacked randomization, drawing definitive conclusions on CPAP efficacy is difficult.

mckee_obstructivesleepapnea_t1.jpg
Several short-term randomized controlled trials of CPAP have been performed in patients after stroke. A 2018 meta-analysis59 included 10 such trials with a total of 564 patients (range 30–140 patients), with most having 1 to 3 months of follow-up (range 1 week to over 5 years). Eight of the 10 studies are summarized in Table 1 (1 study was omitted because many of the patients had central sleep apnea, and 1 was primarily a feasibility study).60–67

Patients were enrolled in the index admission or when starting a rehabilitation service—generally 2 to 3 weeks after their stroke. No clear association was found between the timing of initiating PAP therapy and outcomes. All patients had ischemic strokes, but few details were provided regarding stroke location, size, and severity. Exclusion criteria included severe underlying cardiopulmonary disease, confusion, severe stroke with marked impairment, and inability to cooperate. Almost all patients had moderate to severe OSA, and patients with central sleep apnea were excluded.

The major outcomes examined were drop-out rates, PAP adherence, and neurologic improvement based on neurologic functional scales (National Institutes of Health Stroke Scale and Canadian Neurologic Scale). As expected, dropout rates were higher in patients randomized to CPAP (OR 1.83, 95% CI 1.05–3.21, P = .03), although overall adherence was better than anticipated, with mean CPAP use across trials of 4.5 hours per night (95% CI 3.97–5.08) and with about 50% to 60% of patients adhering to therapy for at least 4 hours nightly.

Improvement in neurologic outcomes favored CPAP (standard mean difference 0.54, 95% CI 0.026–1.05), although considerable heterogeneity was seen. Improved sleepiness outcomes were inconsistent. Major cardiovascular outcomes were reported in only 2 studies (using the same data set) and showed delayed time to the next cardiovascular event for those treated with CPAP but no difference in cardiovascular event-free survival.

PAP poses more challenges after stroke

The primary limitation to PAP therapy is poor acceptance and adherence to therapy.59 High rates of refusal of therapy and difficulty complying with treatment have been noted in the poststroke population, although recent studies have reported better adherence rates. How rates of adherence play out in real-world settings, outside of the controlled environment of a research study, has yet to be determined.

In general, CPAP adherence is affected by claustrophobia, difficulty tolerating a mask, problems with pressure intolerance, irritating air leaks, nasal congestion, and naso-oral dryness. Many such barriers can be overcome with use of a properly fitted mask, an appropriate pressure setting, heated humidification, nasal sprays (eg, saline, inhaled steroids), and education, encouragement, and reassurance.

After a stroke, additional obstacles may impede the ability to use PAP therapy.68 Facial paresis (hemi- or bifacial) may make fitting of the mask problematic. Paralysis or weakness of the extremities may limit the ability to adjust or remove a mask. Aphasia can impair communication and understanding of the need to use PAP therapy, and upper-airway problems related to stroke, including dysphagia, may lead to pressure intolerance or risk of aspiration. Finally, a lack of perceived benefit, particularly if the patient does not have daytime sleepiness, may limit motivation.

Consider alternatives

For patients unlikely to succeed with PAP therapy, there are alternatives. Surgery and oral appliances are not usually realistic options in the setting of recent stroke, but positional therapy, including the use of body positioners to prevent supine sleep, as well as elevating the head of the bed, may be of some benefit.69,70 A nasopharyngeal airway stenting device (nasal trumpet) may also be tolerated by some patients.

mckee_obstructivesleepapnea_f1.jpg
%3Cp%3EFigure%201.%20Managing%20obstructive%20sleep%20apnea%20after%20stroke.%3C%2Fp%3E
Avoiding or minimizing sedating medications that may worsen OSA, such as benzodiazepines and opioids, should be considered.3 Oxygen therapy, while helping to maintain oxygen saturation during sleep, does not prevent airway collapse, and its role for treating OSA in patients after stroke is unclear.

A proposed algorithm for screening, diagnosing, and treating OSA in patients after stroke is presented in Figure 1.

Obstructive sleep apnea (OSA) is an independent risk factor for ischemic stroke and may also, infrequently, be a consequence of stroke. It is significantly underdiagnosed in the general population and is highly prevalent in patients who have had a stroke. Many patients likely had their stroke because of this chronic untreated condition.

This review focuses on OSA and its prevalence, consequences, and treatment in patients after a stroke.

DEFINING AND QUANTIFYING OSA

OSA is the most common type of sleep-disordered breathing.1,2 It involves repeated narrowing or complete collapse of the upper airway despite ongoing respiratory effort.3,4 Apneic episodes are terminated by arousals from hypoxemia or efforts to breathe.5 In contrast, central sleep apnea is characterized by a patent airway but lack of airflow due to absent respiratory effort.5

In OSA, the number of episodes of apnea (absent airflow) and hypopnea (reduced airflow) are added together and divided by hours of sleep to calculate the apnea-hypopnea index (AHI). OSA is diagnosed by either of the following3,4:

  • AHI of 5 or higher, with clinical symptoms related to OSA (described below)
  • AHI of 15 or higher, regardless of symptoms.

The AHI also defines OSA severity, as follows3:

  • Mild: AHI 5 to 15
  • Moderate: AHI 15 to 30
  • Severe: AHI greater than 30.

Diagnostic criteria (eg, definition of hypopnea, testing methods, and AHI thresholds) have varied over time, an important consideration when reviewing the literature.

OSA IS MORE COMMON THAN EXPECTED AFTER STROKE

In the most methodologically sound and generalizable study of this topic to date, the Wisconsin Sleep Cohort Study6 reported in 2013 that about 14% of men and 5% of women ages 30 to 70 have an AHI greater than 5 (using 4% desaturation to score hypopneic episodes) with daytime sleepiness. Other studies suggest that 80% to 90% of people with OSA are undiagnosed and untreated.1,7

The prevalence of OSA in patients who have had a stroke is much higher, ranging from 30% to 96% depending on the study methods and population.1,8–12 A 2010 meta-analysis11 of 29 studies reported that 72% of patients who had a stroke had an AHI greater than 5, and 29% had severe OSA. In this analysis, 7% of those with sleep-disordered breathing had central sleep apnea; still, these data indicate that the prevalence of OSA in these patients is about 5 times higher than in the general population.

RISK FACTORS MAY DIFFER IN STROKE POPULATION

Several risk factors for OSA have been identified.

Obesity is one of the strongest risk factors, with increasing body mass index (BMI) associated with increased OSA prevalence.4,6,13 However, obesity appears to be a less significant risk factor in patients who have had a stroke than in the general population. In the 2010 meta-analysis11 of OSA after stroke, the average BMI was only 26.4 kg/m2 (with obesity defined as a BMI > 30.0 kg/m2), and increasing BMI was not associated with increasing AHI.

Male sex and advanced age are also OSA risk factors.4,5 They remain significant in patients after a stroke; about 65% of poststroke patients who have OSA are men, and the older the patient, the more likely the AHI is greater than 10.11

Ethnicity and genetics may also play important roles in OSA risk, with roughly 25% of OSA prevalence estimated to have a genetic basis.14,15 Some risk factors for OSA such as craniofacial shape, upper airway anatomy, upper airway muscle dysfunction, increased respiratory chemosensitivity, and poor arousal threshold during sleep are likely determined by genetics and ethnicity.14,15 Compared with people of European origin, Asians have a similar prevalence of OSA, but at a much lower average BMI, suggesting that other factors are significant.14 Possible genetically determined anatomic risk factors have not been specifically studied in the poststroke population, but it can be assumed they remain relevant.

Several studies have tried to find an association between OSA and type, location, etiology, or pattern of stroke.10,11,16–19 Although some suggest links between cardioembolic stroke and OSA,16,20 or thrombolysis and OSA,10 most have found no association between OSA and stroke features.11,12,21,22

HOW DOES OSA INCREASE STROKE RISK?

Untreated severe OSA is associated with increased cardiovascular mortality,21,22 and OSA is an independent risk factor for incident stroke.23 A number of mechanisms may explain these relationships.

Intermittent hypoxemia and recurrent sympathetic arousals resulting from OSA are thought to lead to many of the comorbid conditions with which it is associated: hypertension, coronary artery disease, heart failure, arrhythmias, pulmonary hypertension, and stroke. Repetitive decreases in ventilation lead to oxygen desaturations that result in cycles of increased sympathetic outflow and eventual sustained nocturnal hypertension and daytime chronic hypertension.1,5,9,13 Also implicated are various changes in vasodilator and vasoconstrictor substances due to endothelial dysfunction and inflammation, which are thought to play a role in the atherogenic and prothrombotic states induced by OSA.1,5,13

Cerebral circulation is altered primarily by the changes in partial pressure of carbon dioxide (Pco2). During apnea, the Pco2 rises, causing vasodilation and increased blood flow. After the apnea resolves, there is hyperpnea with resultant decreased Pco2, and vasoconstriction. In a patient who already has vascular disease, the enhanced vasoconstriction could lead to ischemia.1,5

Changes in intrathoracic pressure result in distortion of cardiac architecture. When the patient tries to breathe against an occluded airway, the intrathoracic pressure becomes more and more negative, increasing preload and afterload. When this happens repeatedly every night for years, it leads to remodeling of the heart such as left and right ventricular hypertrophy, with reduced stroke volume, myocardial ischemia, and increased risk of arrhythmia.1,5,13

Untreated OSA is believed to predispose patients to develop atrial fibrillation through sympathetic overactivity, vascular inflammation, heart rate variability, and cardiac remodeling.24 As atrial fibrillation is a major risk factor for stroke, particularly cardioembolic stroke, it may be another pathway of increased stroke risk in OSA.16,20,25

 

 

CLINICAL MANIFESTATIONS OF OSA NOT OBVIOUS AFTER STROKE

OSA typically causes both daytime symptoms (excessive sleepiness, poor concentration, morning headache, depressive symptoms) and nighttime signs and symptoms (snoring, choking, gasping, night sweats, insomnia, nocturia, witnessed episodes of apnea).3,4,26 Unfortunately, because these are nonspecific, OSA is often underdiagnosed.4,26

Identifying OSA after a stroke may be a particular challenge, as patients often do not report classic symptoms, and the typical picture of OSA may have less predictive validity in these patients.1,27,28 Within the first 24 hours after a stroke, hypersomnia, snoring history, and age are not predictive of OSA.1 Patients found to have OSA after a stroke frequently do not have the traditional symptoms (sleepiness, snoring) seen in usual OSA patients. And they have higher rates of OSA at a younger age than the usual OSA patients, so age is not a predictive risk factor. In addition, daytime sleepiness and obesity are often absent or less prominent.1,9,27,28  Finally, typical OSA signs and symptoms may be attributed to the stroke itself or to comorbidities affecting the patient, lowering suspicion for OSA.

OSA MAY HINDER STROKE RECOVERY, WORSEN OUTCOMES

OSA, particularly when moderate to severe, is linked to pathophysiologic changes that can hinder recovery from a stroke.

Intermittent hypoxemia during sleep can worsen vascular damage of at-risk tissue: nocturnal hypoxemia correlates with white matter hyperintensities on magnetic resonance imaging, a marker of ischemic demyelination.29 Oxidative stress and release of inflammatory mediators associated with intermittent hypoxemia may impair vascular blood flow to brain tissue attempting to repair itself.30 In addition, sympathetic overactivity and Pco2 fluctuations associated with OSA may impede cerebral circulation.

Taken together, such ongoing nocturnal insults can lead to clinical consequences during this vulnerable period.

A 1996 study31 of patients recovering from a stroke found that an oxygen desaturation index (number of times that the blood oxygen level drops below a certain threshold, as measured by overnight oximetry) of more than 10 per hour was associated with worse functional recovery at discharge and at 3 and 12 months after discharge. This study also noted an association between time spent with oxygen saturations below 90% and the rate of death at 1 year.

A 2003 study32 reported that patients with an AHI greater than 10 by polysomnography spent an average of 13 days longer on the rehabilitation service and had worse functional and cognitive status on discharge, even after controlling for multiple confounders. Several subsequent studies have confirmed these and similar findings.8,33,34

OSA has also been linked to depression,35 which is common after stroke and may worsen outcomes.36 The interaction between OSA, depression, and poststroke outcomes warrants further study.

In the general population, OSA has been independently associated with increased risk of stroke or death from any cause.21,22,37 These associations have also been reported in the poststroke population: a 2014 meta-analysis found that OSA increased the risk of a repeat stroke (relative risk [RR] 1.8, 95% confidence interval [CI] 1.2–2.6) and all-cause mortality (RR 1.69, 95% CI 1.4–2.1).38

TESTING FOR OSA AFTER STROKE

Because of the high prevalence of OSA in patients who have had a stroke and the potential for worse outcomes associated with untreated OSA, there should be a low threshold for evaluating for OSA soon after stroke. Objective testing is required to qualify for therapy,  and the gold standard for diagnosis of OSA is formal polysomnography conducted in a sleep laboratory.2–4 Unfortunately, polysomnography may be unacceptable to some patients, is costly, and is resource-intensive, particularly in an inpatient or rehabilitation setting.28 Ideally, to optimize testing efficiency, patients should be screened for the likelihood of OSA before polysomnography is ordered.

Questionnaires can help determine the need for further testing

Questionnaires developed to assess OSA risk39 include the following:

The Berlin questionnaire, developed in 1999, has 10 questions assessing daytime and nighttime signs and symptoms and presence of hypertension.

The STOP questionnaire, developed in 2008, assesses snoring, tiredness, observed apneic episodes, and elevated blood pressure.

The STOP-BANG questionnaire, published in 2010, includes the STOP questions plus BMI over 35 kg/m2, age over 50, neck circumference over 41 cm, and male gender.

A 2017 meta-analysis39 of 108 studies with nearly 50,000 people found that the STOP-BANG questionnaire performed best with regard to sensitivity and diagnostic odds ratio, but with poor specificity.

These screening tools and modified versions of them have also been evaluated in patients who have had a stroke.

In 2015, Boulos et al28 found that the STOP-BAG (a version of STOP-BANG that excludes neck circumference) and the 4-variable (4V) questionnaire (sex, BMI, blood pressure, snoring) had moderate predictive value for OSA within 6 months after sroke.

In 2016, Katzan et al40 found that the STOP-BAG2 (STOP-BAG criteria plus continuous variables for BMI and age) had a high sensitivity for polysomnographically diagnosed OSA within the first year after a stroke. The specificity was significantly better than the STOP-BANG or the STOP-BAG questionnaire, although it remained suboptimal at 60.5%.

In 2017, Sico et al41 developed and assessed the SLEEP Inventory (sex, left heart failure, Epworth Sleepiness Scale, enlarged neck, weight in pounds, insulin resistance or diabetes, and National Institutes of Health Stroke Scale) and found that it outperformed the Berlin and STOP-BANG questionnaires in the poststroke setting. The SLEEP Inventory had the best specificity and negative predictive value, and a slightly better ability to correctly classify patients as having OSA or not, classifying 80% of patients correctly.

These newer screening tools (eg, STOP-BAG, STOP-BAG2, SLEEP) can be used to identify with reasonable accuracy which patients need definitive testing after stroke.

Pulse oximetry is another possible screening tool          

Overnight pulse oximetry may also help screen for sleep apnea and stratify risk after a stroke. A 2012 study42 of overnight oximetry to screen patients before surgery found that the oxygen desaturation index was significantly associated with the AHI measured by polysomnography. However, oximetry testing cannot distinguish between OSA and central sleep apnea, so it is insufficient to diagnose OSA or qualify patients for therapy. Further study is needed to examine the ability of overnight pulse oximetry to screen or to stratify risk for OSA after stroke.

Polysomnography vs home testing

Polysomnography is the gold standard for diagnosing OSA. Benefits include technical support and trouble-shooting, determining relationships between OSA, body position, and sleep stage, and the ability to intervene with treatment.2 However, polysomnography can be cumbersome, costly, and resource-intensive.

A home sleep apnea test, ie, an unattended, limited-channel sleep study, may be an acceptable alternative.2–4,43,44 Home testing does not require a sleep technologist to be present during testing, uses fewer sensors, and is less expensive than overnight polysomnography, but its utility can be limited: it fails to accurately discriminate between episodes of OSA and central sleep apnea, there is potential for false-negative results, and it can underestimate sleep apnea burden because it does not measure sleep.2

Institutional resources and logistics may influence the choice of diagnostic modality. No data exist on outcomes from different diagnostic testing methods in poststroke patients. Further research is needed.

 

 

POSITIVE AIRWAY PRESSURE THERAPY: BENEFITS, CHALLENGES, ALTERNATIVES

The first-line treatment for OSA is positive airway pressure (PAP).3 For most patients, this is continuous PAP (CPAP) or autoadjusting PAP (APAP). In some instances, particularly for those who cannot tolerate CPAP or who have comorbid hypoventilation, bilevel PAP (BPAP) may be indicated. More advanced PAP therapies are unlikely to be used after stroke.

PAP therapy is associated with reduced daytime sleepiness, improved mood, normalization of sleep architecture, improved systemic and pulmonary artery blood pressure, reduced rates of atrial fibrillation after ablation, and improved insulin sensitivity.45–49 Whether it reduces the risk of cardiovascular events, including stroke, remains controversial; most data suggest that it does not.50,51 However, when adherence to PAP therapy is considered rather than intention to treat, treatment has been found to lead to improved cardiovascular outcomes.52

Mixed evidence of benefits after stroke

Observational studies provide evidence that CPAP may help patients with OSA after stroke, although results are mixed.53–58 The studies ranged in size from 14 to 105 patients, enrolled patients with mostly moderate to severe OSA, and followed patients from 10 days to 7 years. Adherence to therapy was generally good in the short term (50%–70%), but only  15% to 30% of patients remained adherent at 5 to 7 years. Variable outcomes were reported, with some studies finding improved symptoms in the near term and mixed evidence of cardiovascular benefit in the longer ones. However, as these studies lacked randomization, drawing definitive conclusions on CPAP efficacy is difficult.

mckee_obstructivesleepapnea_t1.jpg
Several short-term randomized controlled trials of CPAP have been performed in patients after stroke. A 2018 meta-analysis59 included 10 such trials with a total of 564 patients (range 30–140 patients), with most having 1 to 3 months of follow-up (range 1 week to over 5 years). Eight of the 10 studies are summarized in Table 1 (1 study was omitted because many of the patients had central sleep apnea, and 1 was primarily a feasibility study).60–67

Patients were enrolled in the index admission or when starting a rehabilitation service—generally 2 to 3 weeks after their stroke. No clear association was found between the timing of initiating PAP therapy and outcomes. All patients had ischemic strokes, but few details were provided regarding stroke location, size, and severity. Exclusion criteria included severe underlying cardiopulmonary disease, confusion, severe stroke with marked impairment, and inability to cooperate. Almost all patients had moderate to severe OSA, and patients with central sleep apnea were excluded.

The major outcomes examined were drop-out rates, PAP adherence, and neurologic improvement based on neurologic functional scales (National Institutes of Health Stroke Scale and Canadian Neurologic Scale). As expected, dropout rates were higher in patients randomized to CPAP (OR 1.83, 95% CI 1.05–3.21, P = .03), although overall adherence was better than anticipated, with mean CPAP use across trials of 4.5 hours per night (95% CI 3.97–5.08) and with about 50% to 60% of patients adhering to therapy for at least 4 hours nightly.

Improvement in neurologic outcomes favored CPAP (standard mean difference 0.54, 95% CI 0.026–1.05), although considerable heterogeneity was seen. Improved sleepiness outcomes were inconsistent. Major cardiovascular outcomes were reported in only 2 studies (using the same data set) and showed delayed time to the next cardiovascular event for those treated with CPAP but no difference in cardiovascular event-free survival.

PAP poses more challenges after stroke

The primary limitation to PAP therapy is poor acceptance and adherence to therapy.59 High rates of refusal of therapy and difficulty complying with treatment have been noted in the poststroke population, although recent studies have reported better adherence rates. How rates of adherence play out in real-world settings, outside of the controlled environment of a research study, has yet to be determined.

In general, CPAP adherence is affected by claustrophobia, difficulty tolerating a mask, problems with pressure intolerance, irritating air leaks, nasal congestion, and naso-oral dryness. Many such barriers can be overcome with use of a properly fitted mask, an appropriate pressure setting, heated humidification, nasal sprays (eg, saline, inhaled steroids), and education, encouragement, and reassurance.

After a stroke, additional obstacles may impede the ability to use PAP therapy.68 Facial paresis (hemi- or bifacial) may make fitting of the mask problematic. Paralysis or weakness of the extremities may limit the ability to adjust or remove a mask. Aphasia can impair communication and understanding of the need to use PAP therapy, and upper-airway problems related to stroke, including dysphagia, may lead to pressure intolerance or risk of aspiration. Finally, a lack of perceived benefit, particularly if the patient does not have daytime sleepiness, may limit motivation.

Consider alternatives

For patients unlikely to succeed with PAP therapy, there are alternatives. Surgery and oral appliances are not usually realistic options in the setting of recent stroke, but positional therapy, including the use of body positioners to prevent supine sleep, as well as elevating the head of the bed, may be of some benefit.69,70 A nasopharyngeal airway stenting device (nasal trumpet) may also be tolerated by some patients.

mckee_obstructivesleepapnea_f1.jpg
%3Cp%3EFigure%201.%20Managing%20obstructive%20sleep%20apnea%20after%20stroke.%3C%2Fp%3E
Avoiding or minimizing sedating medications that may worsen OSA, such as benzodiazepines and opioids, should be considered.3 Oxygen therapy, while helping to maintain oxygen saturation during sleep, does not prevent airway collapse, and its role for treating OSA in patients after stroke is unclear.

A proposed algorithm for screening, diagnosing, and treating OSA in patients after stroke is presented in Figure 1.

References
  1. Selim B, Roux FJ. Stroke and sleep disorders. Sleep Med Clin 2012; 7(4):597–607. doi:10.1016/j.jsmc.2012.08.007
  2. Kapur VK, Auckley DH, Chowdhuri S, et al. Clinical practice guideline for diagnostic testing for adult obstructive sleep apnea: an American Academy of Sleep Medicine Clinical Practice Guideline. J Clin Sleep Med 2017; 13(3):479–504. doi:10.5664/jcsm.6506
  3. Epstein LJ, Kristo D, Strollo PJ Jr, et al; Adult Obstructive Sleep Apnea Task Force of the American Academy of Sleep Medicine. Clinical guideline for the evaluation, management, and long-term care of obstructive sleep apnea in adults. J Clin Sleep Med 2009; 5(3):263–276. pmid:19960649
  4. Patil SP, Schneider H, Schwartz AR, Smith PL. Adult obstructive sleep apnea: pathophysiology and diagnosis. Chest 2007; 132(1):325–337. doi:10.1378/chest.07-0040
  5. Dempsey JA, Veasey SC, Morgan BJ, O’Donnell CP. Pathophysiology of sleep apnea. Physiol Rev 2010; 90(1):47–112. doi:10.1152/physrev.00043.2008
  6. Peppard PE, Young T, Barnet JH, Palta M, Hagen EW, Hla KM. Increased prevalence of sleep-disordered breathing in adults. Am J Epidemiol 2013; 177(9):1006–1014. doi:10.1093/aje/kws342
  7. Redline S, Sotres-Alvarez D, Loredo J, et al. Sleep-disordered breathing in Hispanic/Latino individuals of diverse backgrounds. The Hispanic Community Health Study/Study of Latinos. Am J Resp Crit Care Med 2014; 189(3):335–344. doi:10.1164/rccm.201309-1735OC
  8. Aaronson JA, van Bennekom CA, Hofman WF, et al. Obstructive sleep apnea is related to impaired cognitive and functional status after stroke. Sleep 2015; 38(9):1431–1437. doi:10.5665/sleep.4984
  9. Sharma S, Culebras A. Sleep apnoea and stroke. Stroke Vasc Neurol 2016; 1(4):185–191. doi:10.1136/svn-2016-000038
  10. Huhtakangas JK, Huhtakangas J, Bloigu R, Saaresranta T. Prevalence of sleep apnea at the acute phase of ischemic stroke with or without thrombolysis. Sleep Med 2017; 40:40–46. doi:10.1016/j.sleep.2017.08.018
  11. Johnson KG, Johnson DC. Frequency of sleep apnea in stroke and TIA patients: a meta-analysis. J Clin Sleep Med 2010; 6(2):131–137. pmid:20411688
  12. Iranzo A, Santamaria J, Berenguer J, Sanchez M, Chamorro A. Prevalence and clinical importance of sleep apnea in the first night after cerebral infarction. Neurology 2002; 58:911–916. pmid:11914407
  13. Javaheri S, Barbe F, Campos-Rodriguez F, et al. Sleep apnea: types, mechanisms, and clinical cardiovascular consequences. J Am Coll Cardiol 2017; 69(7):841–858. doi:10.1016/j.jacc.2016.11.069
  14. Dudley KA, Patel SR. Disparities and genetic risk factors in obstructive sleep apnea. Sleep Med 2016; 18:96–102. doi:10.1016/j.sleep.2015.01.015
  15. Redline S, Tishler PV. The genetics of sleep apnea. Sleep Med Rev 2000; 4(6):583–602. doi:10.1053/smrv.2000.0120
  16. Lipford MC, Flemming KD, Calvin AD, et al. Associations between cardioembolic stroke and obstructive sleep apnea. Sleep 2015; 38(11):1699–1705. doi:10.5665/sleep.5146
  17. Wang Y, Wang Y, Chen J, Yi X, Dong S, Cao L. Stroke patterns, topography, and etiology in patients with obstructive sleep apnea hypopnea syndrome. Int J Clin Exp Med 2017; 10(4):7137–7143.
  18. Fisse AL, Kemmling A, Teuber A, et al. The association of lesion location and sleep related breathing disorder in patients with acute ischemic stroke. PLoS One 2017; 12(1):e0171243. doi:10.1371/journal.pone.0171243
  19. Brown DL, Mowla A, McDermott M, et al. Ischemic stroke subtype and presence of sleep-disordered breathing: the BASIC sleep apnea study. J Stroke Cerebrovasc Dis 2015; 24(2):388–393. doi:10.1016/j.jstrokecerebrovasdis.2014.09.007
  20. Poli M, Philip P, Taillard J, et al. Atrial fibrillation as a major cause of stroke in apneic patients: a prospective study. Sleep Med 2017; 30:251–254. doi:10.1016/j.sleep.2015.07.031
  21. Young T, Finn L, Peppard PE, et al. Sleep disordered breathing and mortality: eighteen-year follow-up of the Wisconsin Sleep Cohort. Sleep 2008; 31(8):1071–1078. pmid:18714778
  22. Molnar MZ, Mucsi I, Novak M, et al. Association of incident obstructive sleep apnoea with outcomes in a large cohort of US veterans. Thorax 2015; 70(9):888–895. doi:10.1136/thoraxjnl-2015-206970
  23. Redline S, Yenokyan G, Gottlieb DJ, et al. Obstructive sleep apnea-hypopnea and incident stroke: the sleep heart health study. Am J Respir Crit Care Med 2010; 182(2):269–277. doi:10.1164/rccm.200911-1746OC
  24. Marulanda-Londono E, Chaturvedi S. The interplay between obstructive sleep apnea and atrial fibrillation. Fron Neurol 2017; 8:668. doi:10.3389/fneur.2017.00668
  25. Szymanski FM, Filipiak KJ, Platek AE, Hrynkiewicz-Szymanska A, Karpinski G, Opolski G. Assessment of CHADS2 and CHA 2DS 2-VASc scores in obstructive sleep apnea patients with atrial fibrillation. Sleep Breath 2015; 19(2):531–537. doi:10.1007/s11325-014-1042-5
  26. Stansbury RC, Strollo PJ. Clinical manifestations of sleep apnea. J Thoracic Dis 2015; 7(9):E298–E310. doi:10.3978/j.issn.2072-1439.2015.09.13
  27. Chan W, Coutts SB, Hanly P. Sleep apnea in patients with transient ischemic attack and minor stroke: opportunity for risk reduction of recurrent stroke? Stroke 2010; 41(12):2973–2975. doi:10.1161/STROKEAHA.110.596759
  28. Boulos MI, Wan A, Im J, et al. Identifying obstructive sleep apnea after stroke/TIA: evaluating four simple screening tools. Sleep Med 2016; 21:133–139. doi:10.1016/j.sleep.2015.12.013
  29. Patel SK, Hanly PJ, Smith EE, Chan W, Coutts SB. Nocturnal hypoxemia is associated with white matter hyperintensities in patients with a minor stroke or transient ischemic attack. J Clin Sleep Med 2015; 11(12):1417–1424. doi:10.5664/jcsm.5278
  30. McCarty MF, DiNicolantonio JJ, O’Keefe JH. NADPH oxidase, uncoupled endothelial nitric oxide synthase, and NF-KappaB are key mediators of the pathogenic impact of obstructive sleep apnea—therapeutic implications. J Integr Cardiol 2016; 2(5):367–374. doi:10.15761/JIC.1000177
  31. Good DC, Henkle JQ, Gelber D, Welsh J, Verhulst S. Sleep-disordered breathing and poor functional outcome after stroke. Stroke 1996; 27(2):252–259. pmid:8571419
  32. Kaneko Y, Hajek VE, Zivanovic V, Raboud J, Bradley TD. Relationship of sleep apnea to functional capacity and length of hospitalization following stroke. Sleep 2003; 26(3):293–297. pmid:12749548
  33. Yan-fang S, Yu-ping W. Sleep-disordered breathing: impact on functional outcome of ischemic stroke patients. Sleep Med 2009; 10(7):717–719. doi:10.1016/j.sleep.2008.08.006
  34. Kumar R, Suri JC, Manocha R. Study of association of severity of sleep disordered breathing and functional outcome in stroke patients. Sleep Med 2017; 34:50–56. doi:10.1016/j.sleep.2017.02.025
  35. Kerner NA, Roose SP. Obstructive sleep apnea is linked to depression and cognitive impairment: evidence and potential mechanisms. Am J Geriatr Psychiatry 2016; 24(6):496–508. doi:10.1016/j.jagp.2016.01.134
  36. Bartoli F, Lillia N, Lax A, et al. Depression after stroke and risk of mortality: a systematic review and meta-analysis. Stroke Res Treat 2013; 2013:862978. doi:10.1155/2013/862978
  37. Yaggi HK, Concato J, Kernan WN, Lichtman JH, Brass LM, Mohsenin V. Obstructive sleep apnea as a risk factor for stroke and death. N Engl J Med 2005; 353(19):2034–2041. doi:10.1056/NEJMoa043104
  38. Xie W, Zheng F, Song X. Obstructive sleep apnea and serious adverse outcomes in patients with cardiovascular or cerebrovascular disease: a PRISMA-compliant systematic review and meta-analysis. Medicine (Baltimore) 2014; 93(29):e336. doi:10.1097/MD.0000000000000336
  39. Chiu HY, Chen PY, Chuang LP, et al. Diagnostic accuracy of the Berlin questionnaire, STOP-BANG, STOP, and Epworth sleepiness scale in detecting obstructive sleep apnea: a bivariate meta-analysis. Sleep Med Rev 2017; 36:57–70. doi:10.1016/j.smrv.2016.10.004
  40. Katzan IL, Thompson NR, Uchino K, Foldvary-Schaefer N. A screening tool for obstructive sleep apnea in cerebrovascular patients. Sleep Med 2016; 21:70–76. doi:10.1016/j.sleep.2016.02.001
  41. Sico JJ, Yaggi HK, Ofner S, et al. Development, validation, and assessment of an ischemic stroke or transient ischemic attack-specific prediction tool for obstructive sleep apnea. J Stroke Cerebrovasc Dis 2017; 26(8):1745–1754. doi:10.1016/j.jstrokecerebrovasdis.2017.03.042
  42. Chung F, Liao P, Elsaid H, Islam S, Shapiro CM, Sun Y. Oxygen desaturation index from nocturnal oximetry: a sensitive and specific tool to detect sleep-disordered breathing in surgical patients. Anesth Analg 2012; 114(5):993–1000. doi:10.1213/ANE.0b013e318248f4f5
  43. Boulos MI, Elias S, Wan A, et al. Unattended hospital and home sleep apnea testing following cerebrovascular events. J Stroke Cerebrovasc Dis 2017; 26(1):143–149. doi:10.1016/j.jstrokecerebrovasdis.2016.09.001
  44. Saletu MT, Kotzian ST, Schwarzinger A, Haider S, Spatt J, Saletu B. Home sleep apnea testing is a feasible and accurate method to diagnose obstructive sleep apnea in stroke patients during in-hospital rehabilitation. J Clin Sleep Med 2018; 14(9):1495–1501. doi:10.5664/jcsm.7322
  45. Giles TL, Lasserson TJ, Smith BH, White J, Wright J, Cates CJ. Continuous positive airways pressure for obstructive sleep apnoea in adults. Cochrane Database Syst Rev 2006; (3):CD001106. doi:10.1002/14651858.CD001106.pub3
  46. Fatureto-Borges F, Lorenzi-Filho G, Drager LF. Effectiveness of continuous positive airway pressure in lowering blood pressure in patients with obstructive sleep apnea: a critical review of the literature. Integr Blood Press Control 2016; 9:43–47. doi:10.2147/IBPC.S70402
  47. Imran TF, Gharzipura M, Liu S, et al. Effect of continuous positive airway pressure treatment on pulmonary artery pressure in patients with isolated obstructive sleep apnea: a meta-analysis. Heart Fail Rev 2016; 21(5):591–598. doi:10.1007/s10741-016-9548-5
  48. Deng F, Raza A, Guo J. Treating obstructive sleep apnea with continuous positive airway pressure reduces risk of recurrent atrial fibrillation after catheter ablation: a meta-analysis. Sleep Med 2018; 46:5–11. doi:10.1016/j.sleep.2018.02.013
  49. Seetho IW, Wilding JPH. Sleep-disordered breathing, type 2 diabetes, and the metabolic syndrome. Chronic Resp Dis 2014; 11(4):257–275. doi:10.1177/1479972314552806
  50. Kim Y, Koo YS, Lee HY, Lee SY. Can continuous positive airway pressure reduce the risk of stroke in obstructive sleep apnea patients? A systematic review and meta-analysis. PloS ONE 2016; 11(1):e0146317. doi:10.1371/journal.pone.0146317
  51. Yu J, Zhou Z, McEvoy RD, et al. Association of positive airway pressure with cardiovascular events and death in adults with sleep apnea: a systematic review and meta-analysis. JAMA 2017; 318(2):156–166. doi:10.1001/jama.2017.7967
  52. Peker Y, Glantz H, Eulenburg C, Wegscheider K, Herlitz J, Thunström E. Effect of positive airway pressure on cardiovascular outcomes in coronary artery disease patients with nonsleepy obstructive sleep apnea. The RICCADSA randomized controlled trial. Am J Respir Crit Care Med 2016; 194(5):613–620. doi:10.1164/rccm.201601-0088OC
  53. Martinez-Garcia MA, Soler-Cataluna JJ, Ejarque-Martinez L, et al. Continuous positive airway pressure treatment reduces mortality in patients with ischemic stroke and obstructive sleep apnea: a 5-year follow-up study. Am J Respir Crit Care Med 2009; 180(1):36–41. doi:10.1164/rccm.200808-1341OC
  54. Broadley SA, Jorgensen L, Cheek A, et al. Early investigation and treatment of obstructive sleep apnoea after acute stroke. J Clin Neurosci 2007; 14(4):328–333. doi:10.1016/j.jocn.2006.01.017
  55. Wessendorf TE, Wang YM, Thilmann AF, Sorgenfrei U, Konietzko N, Teschler H. Treatment of obstructive sleep apnoea with nasal continuous positive airway pressure in stroke. Eur Respir J 2001; 18(4):623–629. pmid:11716165
  56. Bassetti CL, Milanova M, Gugger M. Sleep-disordered breathing and acute ischemic stroke: diagnosis, risk factors, treatment, evolution, and long-term clinical outcome. Stroke 2006; 37(4):967–972. doi:10.1161/01.STR.0000208215.49243.c3
  57. Palombini L, Guilleminault C. Stroke and treatment with nasal CPAP. Eur J Neurol 2006; 13(2):198–200. doi:10.1111/j.1468-1331.2006.01169.x
  58. Martínez-García MA, Campos-Rodríguez F, Soler-Cataluña JJ, Catalán-Serra P, Román-Sánchez P, Montserrat JM. Increased incidence of nonfatal cardiovascular events in stroke patients with sleep apnoea: effect of CPAP treatment. Eur Respir J 2012; 39(4):906–912. doi:10.1183/09031936.00011311
  59. Brill AK, Horvath T, Seiler A, et al. CPAP as treatment of sleep apnea after stroke: a meta-analysis of randomized trials. Neurology 2018; 90(14):e1222–e1230. doi:10.1212/WNL.0000000000005262
  60. Hsu C, Vennelle M, Li H, Engleman HM, Dennis MS, Douglas NJ. Sleep-disordered breathing after stroke: a randomised controlled trial of continuous positive airway pressure. J Neurol Neurosurg Psychiatry 2006; 77(10):1143–1149. doi:10.1136/jnnp.2005.086686
  61. Parra O, Sanchez-Armengol A, Bonnin M, et al. Early treatment of obstructive apnoea and stroke outcome: a randomised controlled trial. Eur Resp J 2011; 37(5):1128–1136. doi:10.1183/09031936.00034410
  62. Ryan CM, Bayley M, Green R, Murray BJ, Bradley TD. Influence of continuous positive airway pressure on outcomes of rehabilitation in stroke patients with obstructive sleep apnea. Stroke 2011; 42(4):1062–1067. doi:10.1161/STROKEAHA.110.597468
  63. Bravata DM, Concato J, Fried T, et al. Continuous positive airway pressure: evaluation of a novel therapy for patients with acute ischemic stroke. Sleep 2011; 34(9):1271–1277. doi:10.5665/SLEEP.1254
  64. Parra O, Sanchez-Armengol A, Capote F, et al. Efficacy of continuous positive airway pressure treatment on 5-year survival in patients with ischaemic stroke and obstructive sleep apnea: a randomized controlled trial. J Sleep Res 2015; 24(1):47–53. doi:10.1111/jsr.12181
  65. Khot SP, Davis AP, Crane DA, et al. Effect of continuous positive airway pressure on stroke rehabilitation: a pilot randomized sham-controlled trial. J Clin Sleep Med 2016; 12(7):1019–1026. doi:10.5664/jcsm.5940
  66. Aaronson JA, Hofman WF, van Bennekom CA, et al. Effects of continuous positive airway pressure on cognitive and functional outcome of stroke patients with obstructive sleep apnea: a randomized controlled trial. J Clin Sleep Med 2016; 12(4):533–541. doi:10.5664/jcsm.5684
  67. Gupta A, Shukla G, Afsar M, et al. Role of positive airway pressure therapy for obstructive sleep apnea in patients with stroke: a randomized controlled trial. J Clin Sleep Med 2018; 14(4):511–521. doi:10.5664/jcsm.7034
  68. Mello-Fujita L, Kim LJ, Palombini Lde O, et al. Treatment of obstructive sleep apnea syndrome associated with stroke. Sleep Med 2015; 16(6):691–696. doi:10.1016/j.sleep.2014.12.017
  69. Svatikova A, Chervin RD, Wing JJ, Sanchez BN, Migda EM, Brown DL. Positional therapy in ischemic stroke patients with obstructive sleep apnea. Sleep Med 2011; 12(3):262–266. doi:10.1016/j.sleep.2010.12.008
  70. Souza FJ, Genta PR, de Souza Filho AJ, Wellman A, Lorenzi-Filho G. The influence of head-of-bed elevation in patients with obstructive sleep apnea. Sleep Breath 2017; 21(4):815–820. doi:10.1007/s11325-017-1524-3
References
  1. Selim B, Roux FJ. Stroke and sleep disorders. Sleep Med Clin 2012; 7(4):597–607. doi:10.1016/j.jsmc.2012.08.007
  2. Kapur VK, Auckley DH, Chowdhuri S, et al. Clinical practice guideline for diagnostic testing for adult obstructive sleep apnea: an American Academy of Sleep Medicine Clinical Practice Guideline. J Clin Sleep Med 2017; 13(3):479–504. doi:10.5664/jcsm.6506
  3. Epstein LJ, Kristo D, Strollo PJ Jr, et al; Adult Obstructive Sleep Apnea Task Force of the American Academy of Sleep Medicine. Clinical guideline for the evaluation, management, and long-term care of obstructive sleep apnea in adults. J Clin Sleep Med 2009; 5(3):263–276. pmid:19960649
  4. Patil SP, Schneider H, Schwartz AR, Smith PL. Adult obstructive sleep apnea: pathophysiology and diagnosis. Chest 2007; 132(1):325–337. doi:10.1378/chest.07-0040
  5. Dempsey JA, Veasey SC, Morgan BJ, O’Donnell CP. Pathophysiology of sleep apnea. Physiol Rev 2010; 90(1):47–112. doi:10.1152/physrev.00043.2008
  6. Peppard PE, Young T, Barnet JH, Palta M, Hagen EW, Hla KM. Increased prevalence of sleep-disordered breathing in adults. Am J Epidemiol 2013; 177(9):1006–1014. doi:10.1093/aje/kws342
  7. Redline S, Sotres-Alvarez D, Loredo J, et al. Sleep-disordered breathing in Hispanic/Latino individuals of diverse backgrounds. The Hispanic Community Health Study/Study of Latinos. Am J Resp Crit Care Med 2014; 189(3):335–344. doi:10.1164/rccm.201309-1735OC
  8. Aaronson JA, van Bennekom CA, Hofman WF, et al. Obstructive sleep apnea is related to impaired cognitive and functional status after stroke. Sleep 2015; 38(9):1431–1437. doi:10.5665/sleep.4984
  9. Sharma S, Culebras A. Sleep apnoea and stroke. Stroke Vasc Neurol 2016; 1(4):185–191. doi:10.1136/svn-2016-000038
  10. Huhtakangas JK, Huhtakangas J, Bloigu R, Saaresranta T. Prevalence of sleep apnea at the acute phase of ischemic stroke with or without thrombolysis. Sleep Med 2017; 40:40–46. doi:10.1016/j.sleep.2017.08.018
  11. Johnson KG, Johnson DC. Frequency of sleep apnea in stroke and TIA patients: a meta-analysis. J Clin Sleep Med 2010; 6(2):131–137. pmid:20411688
  12. Iranzo A, Santamaria J, Berenguer J, Sanchez M, Chamorro A. Prevalence and clinical importance of sleep apnea in the first night after cerebral infarction. Neurology 2002; 58:911–916. pmid:11914407
  13. Javaheri S, Barbe F, Campos-Rodriguez F, et al. Sleep apnea: types, mechanisms, and clinical cardiovascular consequences. J Am Coll Cardiol 2017; 69(7):841–858. doi:10.1016/j.jacc.2016.11.069
  14. Dudley KA, Patel SR. Disparities and genetic risk factors in obstructive sleep apnea. Sleep Med 2016; 18:96–102. doi:10.1016/j.sleep.2015.01.015
  15. Redline S, Tishler PV. The genetics of sleep apnea. Sleep Med Rev 2000; 4(6):583–602. doi:10.1053/smrv.2000.0120
  16. Lipford MC, Flemming KD, Calvin AD, et al. Associations between cardioembolic stroke and obstructive sleep apnea. Sleep 2015; 38(11):1699–1705. doi:10.5665/sleep.5146
  17. Wang Y, Wang Y, Chen J, Yi X, Dong S, Cao L. Stroke patterns, topography, and etiology in patients with obstructive sleep apnea hypopnea syndrome. Int J Clin Exp Med 2017; 10(4):7137–7143.
  18. Fisse AL, Kemmling A, Teuber A, et al. The association of lesion location and sleep related breathing disorder in patients with acute ischemic stroke. PLoS One 2017; 12(1):e0171243. doi:10.1371/journal.pone.0171243
  19. Brown DL, Mowla A, McDermott M, et al. Ischemic stroke subtype and presence of sleep-disordered breathing: the BASIC sleep apnea study. J Stroke Cerebrovasc Dis 2015; 24(2):388–393. doi:10.1016/j.jstrokecerebrovasdis.2014.09.007
  20. Poli M, Philip P, Taillard J, et al. Atrial fibrillation as a major cause of stroke in apneic patients: a prospective study. Sleep Med 2017; 30:251–254. doi:10.1016/j.sleep.2015.07.031
  21. Young T, Finn L, Peppard PE, et al. Sleep disordered breathing and mortality: eighteen-year follow-up of the Wisconsin Sleep Cohort. Sleep 2008; 31(8):1071–1078. pmid:18714778
  22. Molnar MZ, Mucsi I, Novak M, et al. Association of incident obstructive sleep apnoea with outcomes in a large cohort of US veterans. Thorax 2015; 70(9):888–895. doi:10.1136/thoraxjnl-2015-206970
  23. Redline S, Yenokyan G, Gottlieb DJ, et al. Obstructive sleep apnea-hypopnea and incident stroke: the sleep heart health study. Am J Respir Crit Care Med 2010; 182(2):269–277. doi:10.1164/rccm.200911-1746OC
  24. Marulanda-Londono E, Chaturvedi S. The interplay between obstructive sleep apnea and atrial fibrillation. Fron Neurol 2017; 8:668. doi:10.3389/fneur.2017.00668
  25. Szymanski FM, Filipiak KJ, Platek AE, Hrynkiewicz-Szymanska A, Karpinski G, Opolski G. Assessment of CHADS2 and CHA 2DS 2-VASc scores in obstructive sleep apnea patients with atrial fibrillation. Sleep Breath 2015; 19(2):531–537. doi:10.1007/s11325-014-1042-5
  26. Stansbury RC, Strollo PJ. Clinical manifestations of sleep apnea. J Thoracic Dis 2015; 7(9):E298–E310. doi:10.3978/j.issn.2072-1439.2015.09.13
  27. Chan W, Coutts SB, Hanly P. Sleep apnea in patients with transient ischemic attack and minor stroke: opportunity for risk reduction of recurrent stroke? Stroke 2010; 41(12):2973–2975. doi:10.1161/STROKEAHA.110.596759
  28. Boulos MI, Wan A, Im J, et al. Identifying obstructive sleep apnea after stroke/TIA: evaluating four simple screening tools. Sleep Med 2016; 21:133–139. doi:10.1016/j.sleep.2015.12.013
  29. Patel SK, Hanly PJ, Smith EE, Chan W, Coutts SB. Nocturnal hypoxemia is associated with white matter hyperintensities in patients with a minor stroke or transient ischemic attack. J Clin Sleep Med 2015; 11(12):1417–1424. doi:10.5664/jcsm.5278
  30. McCarty MF, DiNicolantonio JJ, O’Keefe JH. NADPH oxidase, uncoupled endothelial nitric oxide synthase, and NF-KappaB are key mediators of the pathogenic impact of obstructive sleep apnea—therapeutic implications. J Integr Cardiol 2016; 2(5):367–374. doi:10.15761/JIC.1000177
  31. Good DC, Henkle JQ, Gelber D, Welsh J, Verhulst S. Sleep-disordered breathing and poor functional outcome after stroke. Stroke 1996; 27(2):252–259. pmid:8571419
  32. Kaneko Y, Hajek VE, Zivanovic V, Raboud J, Bradley TD. Relationship of sleep apnea to functional capacity and length of hospitalization following stroke. Sleep 2003; 26(3):293–297. pmid:12749548
  33. Yan-fang S, Yu-ping W. Sleep-disordered breathing: impact on functional outcome of ischemic stroke patients. Sleep Med 2009; 10(7):717–719. doi:10.1016/j.sleep.2008.08.006
  34. Kumar R, Suri JC, Manocha R. Study of association of severity of sleep disordered breathing and functional outcome in stroke patients. Sleep Med 2017; 34:50–56. doi:10.1016/j.sleep.2017.02.025
  35. Kerner NA, Roose SP. Obstructive sleep apnea is linked to depression and cognitive impairment: evidence and potential mechanisms. Am J Geriatr Psychiatry 2016; 24(6):496–508. doi:10.1016/j.jagp.2016.01.134
  36. Bartoli F, Lillia N, Lax A, et al. Depression after stroke and risk of mortality: a systematic review and meta-analysis. Stroke Res Treat 2013; 2013:862978. doi:10.1155/2013/862978
  37. Yaggi HK, Concato J, Kernan WN, Lichtman JH, Brass LM, Mohsenin V. Obstructive sleep apnea as a risk factor for stroke and death. N Engl J Med 2005; 353(19):2034–2041. doi:10.1056/NEJMoa043104
  38. Xie W, Zheng F, Song X. Obstructive sleep apnea and serious adverse outcomes in patients with cardiovascular or cerebrovascular disease: a PRISMA-compliant systematic review and meta-analysis. Medicine (Baltimore) 2014; 93(29):e336. doi:10.1097/MD.0000000000000336
  39. Chiu HY, Chen PY, Chuang LP, et al. Diagnostic accuracy of the Berlin questionnaire, STOP-BANG, STOP, and Epworth sleepiness scale in detecting obstructive sleep apnea: a bivariate meta-analysis. Sleep Med Rev 2017; 36:57–70. doi:10.1016/j.smrv.2016.10.004
  40. Katzan IL, Thompson NR, Uchino K, Foldvary-Schaefer N. A screening tool for obstructive sleep apnea in cerebrovascular patients. Sleep Med 2016; 21:70–76. doi:10.1016/j.sleep.2016.02.001
  41. Sico JJ, Yaggi HK, Ofner S, et al. Development, validation, and assessment of an ischemic stroke or transient ischemic attack-specific prediction tool for obstructive sleep apnea. J Stroke Cerebrovasc Dis 2017; 26(8):1745–1754. doi:10.1016/j.jstrokecerebrovasdis.2017.03.042
  42. Chung F, Liao P, Elsaid H, Islam S, Shapiro CM, Sun Y. Oxygen desaturation index from nocturnal oximetry: a sensitive and specific tool to detect sleep-disordered breathing in surgical patients. Anesth Analg 2012; 114(5):993–1000. doi:10.1213/ANE.0b013e318248f4f5
  43. Boulos MI, Elias S, Wan A, et al. Unattended hospital and home sleep apnea testing following cerebrovascular events. J Stroke Cerebrovasc Dis 2017; 26(1):143–149. doi:10.1016/j.jstrokecerebrovasdis.2016.09.001
  44. Saletu MT, Kotzian ST, Schwarzinger A, Haider S, Spatt J, Saletu B. Home sleep apnea testing is a feasible and accurate method to diagnose obstructive sleep apnea in stroke patients during in-hospital rehabilitation. J Clin Sleep Med 2018; 14(9):1495–1501. doi:10.5664/jcsm.7322
  45. Giles TL, Lasserson TJ, Smith BH, White J, Wright J, Cates CJ. Continuous positive airways pressure for obstructive sleep apnoea in adults. Cochrane Database Syst Rev 2006; (3):CD001106. doi:10.1002/14651858.CD001106.pub3
  46. Fatureto-Borges F, Lorenzi-Filho G, Drager LF. Effectiveness of continuous positive airway pressure in lowering blood pressure in patients with obstructive sleep apnea: a critical review of the literature. Integr Blood Press Control 2016; 9:43–47. doi:10.2147/IBPC.S70402
  47. Imran TF, Gharzipura M, Liu S, et al. Effect of continuous positive airway pressure treatment on pulmonary artery pressure in patients with isolated obstructive sleep apnea: a meta-analysis. Heart Fail Rev 2016; 21(5):591–598. doi:10.1007/s10741-016-9548-5
  48. Deng F, Raza A, Guo J. Treating obstructive sleep apnea with continuous positive airway pressure reduces risk of recurrent atrial fibrillation after catheter ablation: a meta-analysis. Sleep Med 2018; 46:5–11. doi:10.1016/j.sleep.2018.02.013
  49. Seetho IW, Wilding JPH. Sleep-disordered breathing, type 2 diabetes, and the metabolic syndrome. Chronic Resp Dis 2014; 11(4):257–275. doi:10.1177/1479972314552806
  50. Kim Y, Koo YS, Lee HY, Lee SY. Can continuous positive airway pressure reduce the risk of stroke in obstructive sleep apnea patients? A systematic review and meta-analysis. PloS ONE 2016; 11(1):e0146317. doi:10.1371/journal.pone.0146317
  51. Yu J, Zhou Z, McEvoy RD, et al. Association of positive airway pressure with cardiovascular events and death in adults with sleep apnea: a systematic review and meta-analysis. JAMA 2017; 318(2):156–166. doi:10.1001/jama.2017.7967
  52. Peker Y, Glantz H, Eulenburg C, Wegscheider K, Herlitz J, Thunström E. Effect of positive airway pressure on cardiovascular outcomes in coronary artery disease patients with nonsleepy obstructive sleep apnea. The RICCADSA randomized controlled trial. Am J Respir Crit Care Med 2016; 194(5):613–620. doi:10.1164/rccm.201601-0088OC
  53. Martinez-Garcia MA, Soler-Cataluna JJ, Ejarque-Martinez L, et al. Continuous positive airway pressure treatment reduces mortality in patients with ischemic stroke and obstructive sleep apnea: a 5-year follow-up study. Am J Respir Crit Care Med 2009; 180(1):36–41. doi:10.1164/rccm.200808-1341OC
  54. Broadley SA, Jorgensen L, Cheek A, et al. Early investigation and treatment of obstructive sleep apnoea after acute stroke. J Clin Neurosci 2007; 14(4):328–333. doi:10.1016/j.jocn.2006.01.017
  55. Wessendorf TE, Wang YM, Thilmann AF, Sorgenfrei U, Konietzko N, Teschler H. Treatment of obstructive sleep apnoea with nasal continuous positive airway pressure in stroke. Eur Respir J 2001; 18(4):623–629. pmid:11716165
  56. Bassetti CL, Milanova M, Gugger M. Sleep-disordered breathing and acute ischemic stroke: diagnosis, risk factors, treatment, evolution, and long-term clinical outcome. Stroke 2006; 37(4):967–972. doi:10.1161/01.STR.0000208215.49243.c3
  57. Palombini L, Guilleminault C. Stroke and treatment with nasal CPAP. Eur J Neurol 2006; 13(2):198–200. doi:10.1111/j.1468-1331.2006.01169.x
  58. Martínez-García MA, Campos-Rodríguez F, Soler-Cataluña JJ, Catalán-Serra P, Román-Sánchez P, Montserrat JM. Increased incidence of nonfatal cardiovascular events in stroke patients with sleep apnoea: effect of CPAP treatment. Eur Respir J 2012; 39(4):906–912. doi:10.1183/09031936.00011311
  59. Brill AK, Horvath T, Seiler A, et al. CPAP as treatment of sleep apnea after stroke: a meta-analysis of randomized trials. Neurology 2018; 90(14):e1222–e1230. doi:10.1212/WNL.0000000000005262
  60. Hsu C, Vennelle M, Li H, Engleman HM, Dennis MS, Douglas NJ. Sleep-disordered breathing after stroke: a randomised controlled trial of continuous positive airway pressure. J Neurol Neurosurg Psychiatry 2006; 77(10):1143–1149. doi:10.1136/jnnp.2005.086686
  61. Parra O, Sanchez-Armengol A, Bonnin M, et al. Early treatment of obstructive apnoea and stroke outcome: a randomised controlled trial. Eur Resp J 2011; 37(5):1128–1136. doi:10.1183/09031936.00034410
  62. Ryan CM, Bayley M, Green R, Murray BJ, Bradley TD. Influence of continuous positive airway pressure on outcomes of rehabilitation in stroke patients with obstructive sleep apnea. Stroke 2011; 42(4):1062–1067. doi:10.1161/STROKEAHA.110.597468
  63. Bravata DM, Concato J, Fried T, et al. Continuous positive airway pressure: evaluation of a novel therapy for patients with acute ischemic stroke. Sleep 2011; 34(9):1271–1277. doi:10.5665/SLEEP.1254
  64. Parra O, Sanchez-Armengol A, Capote F, et al. Efficacy of continuous positive airway pressure treatment on 5-year survival in patients with ischaemic stroke and obstructive sleep apnea: a randomized controlled trial. J Sleep Res 2015; 24(1):47–53. doi:10.1111/jsr.12181
  65. Khot SP, Davis AP, Crane DA, et al. Effect of continuous positive airway pressure on stroke rehabilitation: a pilot randomized sham-controlled trial. J Clin Sleep Med 2016; 12(7):1019–1026. doi:10.5664/jcsm.5940
  66. Aaronson JA, Hofman WF, van Bennekom CA, et al. Effects of continuous positive airway pressure on cognitive and functional outcome of stroke patients with obstructive sleep apnea: a randomized controlled trial. J Clin Sleep Med 2016; 12(4):533–541. doi:10.5664/jcsm.5684
  67. Gupta A, Shukla G, Afsar M, et al. Role of positive airway pressure therapy for obstructive sleep apnea in patients with stroke: a randomized controlled trial. J Clin Sleep Med 2018; 14(4):511–521. doi:10.5664/jcsm.7034
  68. Mello-Fujita L, Kim LJ, Palombini Lde O, et al. Treatment of obstructive sleep apnea syndrome associated with stroke. Sleep Med 2015; 16(6):691–696. doi:10.1016/j.sleep.2014.12.017
  69. Svatikova A, Chervin RD, Wing JJ, Sanchez BN, Migda EM, Brown DL. Positional therapy in ischemic stroke patients with obstructive sleep apnea. Sleep Med 2011; 12(3):262–266. doi:10.1016/j.sleep.2010.12.008
  70. Souza FJ, Genta PR, de Souza Filho AJ, Wellman A, Lorenzi-Filho G. The influence of head-of-bed elevation in patients with obstructive sleep apnea. Sleep Breath 2017; 21(4):815–820. doi:10.1007/s11325-017-1524-3
Issue
Cleveland Clinic Journal of Medicine - 86(6)
Issue
Cleveland Clinic Journal of Medicine - 86(6)
Page Number
407-415
Page Number
407-415
Publications
Publications
Topics
Article Type
Display Headline
A sleeping beast: Obstructive sleep apnea and stroke
Display Headline
A sleeping beast: Obstructive sleep apnea and stroke
Legacy Keywords
obstructive sleep apnea, OSA, snoring, stroke, cerebrovascular accident, CVA, transient ischemic attack, TIA, continuous positive airway pressure, CPAP, polysomnography, sleep study, apnea-hypoponea index, AHI, Zachary McKee, Dennis Auckley
Legacy Keywords
obstructive sleep apnea, OSA, snoring, stroke, cerebrovascular accident, CVA, transient ischemic attack, TIA, continuous positive airway pressure, CPAP, polysomnography, sleep study, apnea-hypoponea index, AHI, Zachary McKee, Dennis Auckley
Sections
Inside the Article

KEY POINTS

  • A low threshold for evaluating for OSA after a stroke is warranted: the prevalence is high in this population, and risk factors for OSA and its typical clinical picture may not be present.
  • Questionnaires can help screen for the likelihood of OSA and the need for more definitive assessment with polysomnography or home sleep apnea testing, tests that pose additional challenges after stroke.
  • Positive airway pressure (PAP) therapy remains the first-line treatment for OSA after stroke; it may improve recovery and reduce long-term sequelae of untreated OSA.
  • Acceptance of and adherence to PAP therapy can be especially problematic in this population, and alternatives should be considered if needed.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Fri, 05/24/2019 - 11:00
Un-Gate On Date
Fri, 05/24/2019 - 11:00
Use ProPublica
CFC Schedule Remove Status
Fri, 05/24/2019 - 11:00
Hide sidebar & use full width
render the right sidebar.
Article PDF Media

Anti-Xa assays: What is their role today in antithrombotic therapy?

Article Type
Changed
Mon, 06/03/2019 - 08:05
Display Headline
Anti-Xa assays: What is their role today in antithrombotic therapy?

Should clinicians abandon the activated partial thromboplastin time (aPTT) for monitoring heparin therapy in favor of tests that measure the activity of the patient’s plasma against activated factor X (anti-Xa assays)?

Although other anticoagulants are now available for preventing and treating arterial and venous thromboembolism, unfractionated heparin—which requires laboratory monitoring of therapy—is still widely used. And this monitoring can be challenging. Despite its wide use, the aPTT lacks standardization, and the role of alternative monitoring assays such as the anti-Xa assay is not well defined.

This article reviews the advantages, limitations, and clinical applicability of anti-Xa assays for monitoring therapy with unfractionated heparin and other anticoagulants.

UNFRACTIONATED HEPARIN AND WARFARIN ARE STILL WIDELY USED

Until the mid-1990s, unfractionated heparin and oral vitamin K antagonists (eg, warfarin) were the only anticoagulants widely available for clinical use. These agents have complex pharmacokinetic and pharmacodynamic properties, resulting in highly variable dosing requirements (both between patients and in individual patients) and narrow therapeutic windows, making frequent laboratory monitoring and dose adjustments mandatory.

Over the past 3 decades, other anticoagulants have been approved, including low-molecular-weight heparins, fondaparinux, parenteral direct thrombin inhibitors, and direct oral anticoagulants. While these agents have expanded the options for preventing and treating thromboembolism, unfractionated heparin and warfarin are still the most appropriate choices for many patients, eg, those with stage 4 chronic kidney disease and end-stage renal disease on dialysis, and those with mechanical heart valves.

In addition, unfractionated heparin remains the anticoagulant of choice during procedures such as hemodialysis, percutaneous transluminal angioplasty, and cardiopulmonary bypass, as well as in hospitalized and critically ill patients, who often have acute kidney injury or require frequent interruptions of therapy for invasive procedures. In these scenarios, unfractionated heparin is typically preferred because of its short plasma half-life, complete reversibility by protamine, safety regardless of renal function, and low cost compared with parenteral direct thrombin inhibitors.

As long as unfractionated heparin and warfarin remain important therapies, the need for their laboratory monitoring continues. For warfarin monitoring, the prothrombin time and international normalized ratio are validated and widely reproducible methods. But monitoring unfractionated heparin therapy remains a challenge.

UNFRACTIONATED HEPARIN’S EFFECT IS UNPREDICTABLE

Unfractionated heparin, a negatively charged mucopolysaccharide, inhibits coagulation by binding to antithrombin through the high-affinity pentasaccharide sequence.1–6 Such binding induces a conformational change in the antithrombin molecule, converting it to a rapid inhibitor of several coagulation proteins, especially factors IIa and Xa.2–4

Unfractionated heparin inhibits factors IIa and Xa in a 1:1 ratio, but low-molecular-weight heparins inhibit factor Xa more than factor IIa, with IIa-Xa inhibition ratios ranging from 1:2 to 1:4, owing to their smaller molecular size.7

One of the most important reasons for the unpredictable and highly variable individual responses to unfractionated heparin is that, infused into the blood, the large and negatively charged unfractionated heparin molecules bind nonspecifically to positively charged plasma proteins.7 In patients who are critically ill, have acute infections or inflammatory states, or have undergone major surgery, unfractionated heparin binds to acute-phase proteins that are elevated, particularly factor VIII. This results in fewer free heparin molecules and a variable anticoagulant effect.8

In contrast, low-molecular-weight heparins have longer half-lives and bind less to plasma proteins, resulting in more predictable plasma levels following subcutaneous injection.9

 

 

MONITORING UNFRACTIONATED HEPARIN IMPROVES OUTCOMES

In 1960, Barritt and Jordan10 conducted a small but landmark trial that established the clinical importance of unfractionated heparin for treating venous thromboembolism. None of the patients who received unfractionated heparin for acute pulmonary embolism developed a recurrence during the subsequent 2 weeks, while 50% of those who did not receive it had recurrent pulmonary embolism, fatal in half of the cases.

The importance of achieving a specific aPTT therapeutic target was not demonstrated until a 1972 study by Basu et al,11 in which 162 patients with venous thromboembolism were treated with heparin with a target aPTT of 1.5 to 2.5 times the control value. Patients who suffered recurrent events had subtherapeutic aPTT values on 71% of treatment days, while the rest of the patients, with no recurrences, had subtherapeutic aPTT values only 28% of treatment days. The different outcomes could not be explained by the average daily dose of unfractionated heparin, which was similar in the patients regardless of recurrence.

Subsequent studies showed that the best outcomes occur when unfractionated heparin is given in doses high enough to rapidly achieve a therapeutic prolongation of the aPTT,12–14 and that the total daily dose is also important in preventing recurrences.15,16 Failure to achieve a target aPTT within 24 hours of starting unfractionated heparin is associated with increased risk of recurrent venous thromboembolism.13,17

Raschke et al17 found that patients prospectively randomized to weight-based doses of intravenous unfractionated heparin (bolus plus infusion) achieved significantly higher rates of therapeutic aPTT within 6 hours and 24 hours after starting the infusion, and had significantly lower rates of recurrent venous thromboembolism than those randomized to a fixed unfractionated heparin protocol, without an increase in major bleeding.

Smith et al,18 in a study of 400 consecutive patients with acute pulmonary embolism treated with unfractionated heparin, found that patients who achieved a therapeutic aPTT within 24 hours had lower in-hospital and 30-day mortality rates than those who did not achieve the first therapeutic aPTT until more than 24 hours after starting unfractionated heparin infusion.

Such data lend support to the widely accepted practice and current guideline recommendation8 of using laboratory assays to adjust the dose of unfractionated heparin to achieve and maintain a therapeutic target. The use of dosing nomograms significantly reduces the time to achieve a therapeutic aPTT while minimizing subtherapeutic and supratherapeutic unfractionated heparin levels.19,20

THE aPTT REFLECTS THROMBIN INHIBITION

The aPTT has a log-linear relationship with plasma concentrations of unfractionated heparin,21 but it was not developed specifically for monitoring unfractionated heparin therapy. Originally described in 1953 as a screening tool for hemophilia,22–24 the aPTT is prolonged in the setting of factor deficiencies (typically with levels < 45%, except for factors VII and XIII), as well as lupus anticoagulants and therapy with parenteral direct thrombin inhibitors.8,25,26

Because thrombin (factor IIa) is 10 times more sensitive than factor Xa to inhibition by the heparin-antithrombin complex,4,7 thrombin inhibition appears to be the most likely mechanism by which unfractionated heparin prolongs the aPTT. In contrast, aPTT is minimally or not at all prolonged by low-molecular-weight heparins, which are predominantly factor Xa inhibitors.7

HEPARIN ASSAYS MEASURE UNFRACTIONATED HEPARIN ACTIVITY

While the aPTT is a surrogate marker of unfractionated heparin activity in plasma, unfractionated heparin activity can be measured more precisely by so-called heparin assays, which are typically not direct measures of the plasma concentration of heparins, but rather functional assays that provide indirect estimates. They include protamine sulfate titration assays and anti-Xa assays.

Protamine sulfate titration assays measure the amount of protamine sulfate required to neutralize heparin: the more protamine required, the greater the estimated concentration of unfractionated heparin in plasma.8,27–29 Protamine titration assays are technically demanding, so they are rarely used clinically.

Anti-Xa assays provide a measure of the functional level of heparins in plasma.29–33 Chromogenic anti-Xa assays are available on automated analyzers with standardized kits29,33,34 and may be faster to perform than the aPTT.35

Experiments in rabbits show that unfractionated heparin inhibits thrombus formation and extension at concentrations of 0.2 to 0.4 U/mL as measured by the protamine titration assay,27 which correlated with an anti-Xa activity of 0.35 to 0.67 U/mL in a randomized controlled trial.32

Assays that directly measure the plasma concentration of heparin exist but are not clinically relevant because they also measure heparin molecules lacking the pentasaccharide sequence, which have no anticoagulant activity.36

 

 

ANTI-Xa ASSAY VS THE aPTT

Anti-Xa assays are more expensive than the aPTT and are not available in all hospitals. For these reasons, the aPTT remains the most commonly used laboratory assay for monitoring unfractionated heparin therapy.

However, the aPTT correlates poorly with the activity level of unfractionated heparin in plasma. In one study, an anti-Xa level of 0.3 U/mL corresponded to aPTT results ranging from 47 to 108 seconds.31 Furthermore, in studies that used a heparin therapeutic target based on an aPTT ratio 1.5 to 2.5 times the control aPTT value, the lower end of that target range was often associated with subtherapeutic plasma unfractionated heparin activity measured by anti-Xa and protamine titration assays.28,31

Because of these limitations, individual laboratories should determine their own aPTT therapeutic target ranges for unfractionated heparin based on the response curves obtained with the reagent and coagulometer used. The optimal therapeutic aPTT range for treating acute venous thromboembolism should be defined as the aPTT range (in seconds) that correlates with a plasma activity level of unfractionated heparin of 0.3 to 0.7 U/mL based on a chromogenic anti-Xa assay, or 0.2 to 0.4 U/mL based on a protamine titration assay.32,34–36

Nevertheless, the anticoagulant effect of unfractionated heparin as measured by the aPTT can be unpredictable and can vary widely among individuals and in the same patient.7 This wide variability can be explained by a number of technical and biologic variables. Different commercial aPTT reagents, different lots of the same reagent, and different reagent and instrument combinations have different sensitivities to unfractionated heparin, which can lead to variable aPTT results.37 Moreover, high plasma levels of acute-phase proteins, low plasma antithrombin levels, consumptive coagulopathies, liver failure, and lupus anticoagulants may also affect the aPTT.7,25,32,36–41 These variables account for the poor correlation—ranging from 25% to 66%—reported between aPTT and anti-Xa assays.32,42–48

Such discrepancies may have serious clinical implications: if a patient’s aPTT is low (subtherapeutic) or high (supratherapeutic) but the anti-Xa assay result is within the therapeutic range (0.3–0.7 units/mL), changing the dose of unfractionated heparin (guided by an aPTT nomogram) may increase the risk of bleeding or of recurrent thromboembolism.

CLINICAL APPLICABILITY OF THE ANTI-Xa ASSAY

Neither anti-Xa nor protamine titration assays are standardized across reference laboratories, but chromogenic anti-Xa assays have better interlaboratory correlation than the aPTT49,50 and can be calibrated specifically for unfractionated or low-molecular-weight heparins.29,33

Although reagent costs are higher for chromogenic anti-Xa assays than for the aPTT, some technical variables (described below) may partially offset the cost difference.29,33,41 In addition, unlike the aPTT, anti-Xa assays do not need local calibration; the therapeutic range for unfractionated heparin is the same (0.3–0.7 U/mL) regardless of instrument or reagent.33,41

Most important, studies have found that patients monitored by anti-Xa assay achieve significantly higher rates of therapeutic anticoagulation within 24 and 48 hours after starting unfractionated heparin infusion than those monitored by the aPTT. Fewer dose adjustments and repeat tests are required, which may also result in lower cost.32,51–55

While these studies found chromogenic anti-Xa assays better for achieving laboratory end points, data regarding relevant clinical outcomes are more limited. In a retrospective, observational cohort study,51 the rate of venous thromboembolism or bleeding-related death was 2% in patients receiving unfractionated heparin therapy monitored by anti-Xa assay and 6% in patients monitored by aPTT (P = .62). Rates of major hemorrhage were also not significantly different.

In a randomized controlled trial32 in 131 patients with acute venous thromboembolism and heparin resistance, rates of recurrent venous thromboembolism were 4.6% and 6.1% in the groups randomized to anti-Xa and aPTT monitoring, respectively, whereas overall bleeding rates were 1.5% and 6.1%, respectively. Again, the differences were not statistically significant.

huttcenteno_anti-xaassay_t1.jpg
Though some have suggested that the anti-Xa should be the preferred monitoring assay for intravenous unfractionated heparin therapy,29,41 the ideal assay has not been established by large-scale randomized controlled trials correlating different assays with meaningful clinical outcomes.8,33 Nevertheless, anti-Xa assays are considered the most accurate method of monitoring unfractionated heparin in cases of heparin resistance or lupus anticoagulant, and in other clinical circumstances (Table 1).56–58

Heparin resistance. Some patients require unusually high doses of unfractionated heparin to achieve a therapeutic aPTT: typically, more than 35,000 U over 24 hours,7,8,32 or total daily doses that exceed their estimated weight-based requirements. Heparin resistance has been observed in various clinical settings.7,8,32,37–40,59–61 Patients with heparin resistance monitored by anti-Xa had similar rates of recurrent venous thromboembolism while receiving significantly lower doses of unfractionated heparin than those monitored by the aPTT.32

Lupus anticoagulant. Patients with the specific antiphospholipid antibody known as lupus anticoagulant frequently have a prolonged baseline aPTT,25 making it an unreliable marker of anticoagulant effect for intravenous unfractionated heparin therapy.

Critically ill infants and children. Arachchillage et al35 found that infants (< 1 year old) treated with intravenous unfractionated heparin in an intensive care department had only a 32.4% correlation between aPTT and anti-Xa levels, which was lower than that found in children ages 1 to 15 (66%) and adults (52%). In two-thirds of cases of discordant aPTT and anti-Xa levels, the aPTT was elevated (supratherapeutic) while the anti-Xa assay was within the therapeutic range (0.3–0.7 U/mL). Despite the lack of data on clinical outcomes (eg, rates of thrombosis and bleeding) with the use of an anti-Xa assay, it has been considered the method of choice for unfractionated heparin monitoring in critically ill children, and especially in those under age 1.41,44,62–64

While anti-Xa assays may also be better for unfractionated heparin monitoring in critically ill adults, the lack of clinical outcome data from large-scale randomized trials has precluded evidence-based recommendations favoring them over the aPTT.8,34

 

 

LIMITATIONS OF ANTI-Xa ASSAYS

Anti-Xa assays are hampered by some technical limitations:

Samples must be processed within 1 hour to avoid heparin neutralization.34

Samples must be clear. Hemolyzed or opaque samples (eg, due to bilirubin levels > 6.6 mg/dL or triglyceride levels > 360 mg/dL) cannot be processed, as they can cause falsely low levels.

Exposure to other anticoagulants can interfere with the results. The anti-Xa assay may be unreliable for unfractionated heparin monitoring in patients who are transitioned from low-molecular-weight heparins, fondaparinux, or an oral factor Xa inhibitor (apixaban, betrixaban, edoxaban, rivaroxaban) to intravenous unfractionated heparin, eg, due to hospitalization or acute kidney injury.65,66 Different reports have found that anti-Xa assays may be elevated for as long as 63 to 96 hours after the last dose of oral Xa inhibitors,67–69 potentially resulting in underdosing of unfractionated heparin. In such settings, unfractionated heparin therapy should be monitored by the aPTT.

ANTI-Xa ASSAYS AND LOW-MOLECULAR-WEIGHT HEPARINS

Most patients receiving low-molecular-weight heparins do not need laboratory monitoring.8 Alhenc-Gelas et al70 randomized patients to receive dalteparin in doses either based on weight or guided by anti-Xa assay results, and found that dose adjustments were rare and lacked clinical benefit.

huttcenteno_anti-xaassay_t2.jpg
However, the use of low-molecular-weight heparin-specific anti-Xa assays should be considered for certain patients (Table 2).8

The suggested therapeutic anti-Xa levels for low-molecular-weight heparins are:

  • 0.5–1.2 U/mL for twice-daily enoxaparin
  • 1.0–2.0 U/mL for once-daily enoxaparin or dalteparin.

Levels should be measured at peak plasma level (ie, 3–4 hours after subcutaneous injection, except during pregnancy, when it is 4–6 hours), and only after at least 3 doses of low-molecular-weight heparin.8,71 Unlike the anti-Xa therapeutic range recommended for unfractionated heparin therapy, these ranges are not based on prospective data, and if the assay result is outside the suggested therapeutic target range, current guidelines offer no advice on safely adjusting the dose.8,71

Measuring anti-Xa activity is particularly important for pregnant women with a mechanical prosthetic heart valve who are treated with low-molecular-weight heparins. In this setting, valve thrombosis and cardioembolic events have been reported in patients with peak low-molecular-weight heparin anti-Xa assay levels below or even at the lower end of the therapeutic range, and increased bleeding risk has been reported with elevated anti-Xa levels.71–74 Measuring trough low-molecular-weight heparin anti-Xa levels has been suggested to guide dose adjustments during pregnancy.75

Clearance of low-molecular-weight heparins as measured by the anti-Xa assay is highly correlated with creatinine clearance.76,77 A strong linear correlation has been demonstrated between creatine clearance and anti-Xa levels of enoxaparin after multiple therapeutic doses, and low-molecular-weight heparins accumulate in the plasma, especially in patients with creatine clearance less than 30 mL/min.78 The risk of major bleeding is significantly increased in patients with severe renal insufficiency (creatinine clearance < 30 mL/min) not on dialysis who are treated with either prophylactic or therapeutic doses of low-molecular-weight heparin.79–81 In a meta-analysis, the risk of bleeding with therapeutic-intensity doses of enoxaparin was 4 times higher than with prophylactic-intensity doses.79 Although bleeding risk appears to be reduced when the enoxaparin dose is reduced by 50%,8 the efficacy and safety of this strategy has not been determined by prospective trials.

ANTI-Xa ASSAYS IN PATIENTS RECEIVING DIRECT ORAL ANTICOAGULANTS

Direct oral factor Xa inhibitors cannot be measured accurately by heparin anti-Xa assays. Nevertheless, such assays may be useful to assess whether clinically relevant plasma levels are present in cases of major bleeding, suspected anticoagulant failure, or patient noncompliance.82

Intense research has focused on developing drug-specific chromogenic anti-Xa assays using calibrators and standards for apixaban, edoxaban, and rivaroxaban,82,83 and good linear correlation has been shown with some assays.82,84 In patients treated with oral factor Xa inhibitors who need to undergo an urgent invasive procedure associated with high bleeding risk, use of a specific reversal agent may be considered with drug concentrations more than 30 ng/mL measured by a drug-specific anti-Xa assay. A similar suggestion has been made for drug concentrations more than 50 ng/mL in the setting of major bleeding.85 Unfortunately, such assays are not widely available at this time.82,86

While drug-specific anti-Xa assays could become clinically important to guide reversal strategies, their relevance for drug monitoring remains uncertain. This is because no therapeutic target ranges have been established for any of the direct oral anticoagulants, which were approved on the basis of favorable clinical trial outcomes that neither measured nor were correlated with specific drug levels in plasma. Therefore, a specific anti-Xa level cannot yet be used as a marker of clinical efficacy for any specific oral direct Xa inhibitor.

References
  1. Abildgaard U. Highly purified antithrombin 3 with heparin cofactor activity prepared by disc electrophoresis. Scand J Clin Lab Invest 1968; 21(1):89–91. pmid:5637480
  2. Rosenberg RD, Lam L. Correlation between structure and function of heparin. Proc Natl Acad Sci USA 1979; 76(3):1218–1222. pmid:286307
  3. Lindahl U, Bäckström G, Höök M, Thunberg L, Fransson LA, Linker A. Structure of the antithrombin-binding site of heparin. Proc Natl Acad Sci USA 1979; 76(7):3198–3202. pmid:226960
  4. Rosenberg RD, Rosenberg JS. Natural anticoagulant mechanisms. J Clin Invest 1984; 74(1):1–6. doi:10.1172/JCI111389
  5. Casu B, Oreste P, Torri G, et al. The structure of heparin oligosaccharide fragments with high anti-(factor Xa) activity containing the minimal antithrombin III-binding sequence. Chemical and 13C nuclear-magnetic-resonance studies. Biochem J 1981; 197(3):599–609. pmid:7325974
  6. Choay J, Lormeau JC, Petitou M, Sinaÿ P, Fareed J. Structural studies on a biologically active hexasaccharide obtained from heparin. Ann NY Acad Sci 1981; 370: 644–649. pmid:6943974
  7. Hirsh J, Warkentin TE, Shaughnessy SG, et al. Heparin and low-molecular-weight heparin: mechanisms of action, pharmacokinetics, dosing, monitoring, efficacy, and safety. Chest 2001; 119(suppl 1):64S–94S. pmid:11157643
  8. Garcia DA, Baglin TP, Weitz JI, Samama MM. Parenteral anticoagulants: antithrombotic therapy and prevention of thrombosis, 9th ed: American College of Chest Physicians Evidence-Based Clinical Practice Guidelines. Chest 2012; 141(suppl 2):e24S–e43S. doi:10.1378/chest.11-2291
  9. Hirsh J, Levine M. Low-molecular weight heparin. Blood 1992; 79(1):1–17. pmid:1309422
  10. Barritt DW, Jordan SC. Anticoagulant drugs in the treatment of pulmonary embolism. A controlled trial. Lancet 1960; 1(7138):1309–1312. pmid:13797091
  11. Basu D, Gallus A, Hirsh J, Cade J. A prospective study of the value of monitoring heparin treatment with the activated partial thromboplastin time. N Engl J Med 1972; 287(7):324–327. doi:10.1056/NEJM197208172870703
  12. Hull RD, Raskob GE, Hirsh J, et al. Continuous intravenous heparin compared with intermittent subcutaneous heparin in the initial treatment of proximal-vein thrombosis. N Engl J Med 1986; 315(18):1109–1114. doi:10.1056/NEJM198610303151801
  13. Hull RD, Raskob GE, Brant RF, Pineo GF, Valentine KA. Relation between the time to achieve the lower limit of the APTT therapeutic range and recurrent venous thromboembolism during heparin treatment for deep vein thrombosis. Arch Intern Med 1997; 157(22):2562–2568. pmid:9531224
  14. Hull RD, Raskob GE, Brant RF, Pineo GF, Valentine KA. The importance of initial heparin treatment on long-term clinical outcomes of antithrombotic therapy. The emerging theme of delayed recurrence. Arch Intern Med 1997; 157(20):2317–2321. pmid:9361572
  15. Anand S, Ginsberg JS, Kearon C, Gent M, Hirsh J. The relation between the activated partial thromboplastin time response and recurrence in patients with venous thrombosis treated with continuous intravenous heparin. Arch Intern Med 1996; 156(15):1677–1681. pmid:8694666
  16. Anand SS, Bates S, Ginsberg JS, et al. Recurrent venous thrombosis and heparin therapy: an evaluation of the importance of early activated partial thromboplastin times. Arch Intern Med 1999; 159(17):2029–2032. pmid:10510988
  17. Raschke RA, Reilly BM, Guidry JR, Fontana JR, Srinivas S. The weight-based heparin dosing nomogram compared with a “standard care” nomogram. A randomized controlled trial. Ann Intern Med 1993; 119(9):874–881. pmid:8214998
  18. Smith SB, Geske JB, Maguire JM, Zane NA, Carter RE, Morgenthaler TI. Early anticoagulation is associated with reduced mortality for acute pulmonary embolism. Chest 2010; 137(6):1382–1390. doi:10.1378/chest.09-0959
  19. Cruickshank MK, Levine MN, Hirsh J, Roberts R, Siguenza M. A standard heparin nomogram for the management of heparin therapy. Arch Intern Med 1991; 151(2):333–337. pmid:1789820
  20. Raschke RA, Gollihare B, Peirce J. The effectiveness of implementing the weight-based heparin nomogram as a practice guideline. Arch Intern Med 1996; 156(15):1645–1649. pmid:8694662
  21. Simko RJ, Tsung FF, Stanek EJ. Activated clotting time versus activated partial thromboplastin time for therapeutic monitoring of heparin. Ann Pharmacother 1995; 29(10):1015–1021. doi:10.1177/106002809502901012
  22. Langdell RD, Wagner RH, Brinkhous KM. Effect of antihemophilic factor on one-stage clotting tests; a presumptive test for hemophilia and a simple one-stage antihemophilic factor assy procedure. J Lab Clin Med 1953; 41(4):637–647.
  23. White GC 2nd. The partial thromboplastin time: defining an era in coagulation. J Thromb Haemost 2003; 1(11):2267–2270. pmid:14629454
  24. Proctor RR, Rapaport SI. The partial thromboplastin time with kaolin. A simple screening test for first stage plasma clotting factor deficiencies. Am J Clin Pathol 1961; 36:212–219. pmid:13738153
  25. Brandt JT, Triplett DA, Rock WA, Bovill EG, Arkin CF. Effect of lupus anticoagulants on the activated partial thromboplastin time. Results of the College of American Pathologists survey program. Arch Pathol Lab Med 1991; 115(2):109–114. pmid:1899555
  26. Tripodi A, Mannucci PM. Activated partial thromboplastin time (APTT). New indications for an old test? J Thromb Haemost 2006; 4(4):750–751. doi:10.1111/j.1538-7836.2006.01857.x
  27. Chiu HM, Hirsh J, Yung WL, Regoeczi E, Gent M. Relationship between the anticoagulant and antithrombotic effects of heparin in experimental venous thrombosis. Blood 1977; 49(2):171–184. pmid:831872
  28. Brill-Edwards P, Ginsberg JS, Johnston M, Hirsh J. Establishing a therapeutic range for heparin therapy. Ann Intern Med 1993; 119(2):104–109. pmid:8512158
  29. Vandiver JW, Vondracek TG. Antifactor Xa levels versus activated partial thromboplastin time for monitoring unfractionated heparin. Pharmacotherapy 2012; 32(6):546–558. doi:10.1002/j.1875-9114.2011.01049.x
  30. Newall F. Anti-factor Xa (anti-Xa). In: Monagle P, ed. Haemostasis: Methods and Protocols. New York, NY: Springer-Verlag; 2013.
  31. Bates SM, Weitz JI, Johnston M, Hirsh J, Ginsberg JS. Use of a fixed activated partial thromboplastin time ratio to establish a therapeutic range for unfractionated heparin. Arch Intern Med 2001; 161(3):385–391. pmid:11176764
  32. Levine MN, Hirsh J, Gent M, et al. A randomized trial comparing activated thromboplastin time with heparin assay in patients with acute venous thromboembolism requiring large doses of heparin. Arch Intern Med 1994; 154(1):49–56. pmid:8267489
  33. Wool GD, Lu CM; Education Committee of the Academy of Clinical Laboratory Physicians and Scientists. Pathology consultation on anticoagulation monitoring: factor X-related assays. Am J Clin Pathol 2013; 140(5):623–634. doi:10.1309/AJCPR3JTOK7NKDBJ
  34. Lehman CM, Frank EL. Laboratory monitoring of heparin therapy: partial thromboplastin time or anti-Xa assay? Lab Med 2009; 40(1):47–51. doi:10.1309/LM9NJGW2ZIOLPHY6
  35. Arachchillage DR, Kamani F, Deplano S, Banya W, Laffan M. Should we abandon the aPTT for monitoring unfractionated heparin? Thromb Res 2017; 157:157–161. doi:10.1016/j.thromres.2017.07.006
  36. Olson JD, Arkin CA, Brandt JT, et al. College of American Pathologists Conference XXXI on Laboratory Monitoring of Anticoagulant Therapy: laboratory monitoring of unfractionated heparin therapy. Arch Pathol Lab Med 1998; 122(9):782–798. pmid:9740136
  37. Eikelboom JW, Hirsh J. Monitoring unfractionated heparin with the aPTT: time for a fresh look. Thromb Haemost 2006; 96(5):547–552. pmid:17080209
  38. Young E, Prins M, Levine MN, Hirsh J. Heparin binding to plasma proteins, an important mechanism of heparin resistance. Thromb Haemost 1992; 67(6):639–643. pmid:1509402
  39. Edson JR, Krivit W, White JG. Kaolin partial thromboplastin time: high levels of procoagulants producing short clotting times or masking deficiencies of other procoagulants or low concentrations of anticoagulants. J Lab Clin Med 1967; 70(3):463–470. pmid:6072020
  40. Whitfield LR, Lele AS, Levy G. Effect of pregnancy on the relationship between concentration and anticoagulant action of heparin. Clin Pharmacol Ther 1983; 34(1):23–28. pmid:6861435
  41. Marci CD, Prager D. A review of the clinical indications for the plasma heparin assay. Am J Clin Pathol 1993; 99(5):546–550.
  42. Takemoto CM, Streiff MB, Shermock KM, et al. Activated partial thromboplastin time and anti-Xa measurements in heparin monitoring: biochemical basis of discordance. Am J Clin Pathol 2013; 139(4):450–456. doi:10.1309/AJCPS6OW6DYNOGNH
  43. Adatya S, Uriel N, Yarmohammadi H, et al. Anti-factor Xa and activated partial thromboplastin time measurements for heparin monitoring in mechanical circulatory support. JACC Heart Fail 2015; 3(4):314–322. doi:10.1016/j.jchf.2014.11.009
  44. Kuhle S, Eulmesekian P, Kavanagh B, et al. Lack of correlation between heparin dose and standard clinical monitoring tests in treatment with unfractionated heparin in critically ill children. Haematologica 2007; 92(4):554–557. pmid:17488668
  45. Price EA, Jin J, Nguyen HM, Krishnan G, Bowen R, Zehnder JL. Discordant aPTT and anti-Xa values and outcomes in hospitalized patients treated with intravenous unfractionated heparin. Ann Pharmacother 2013; 47(2):151–158. doi:10.1345/aph.1R635
  46. Baker BA, Adelman MD, Smith PA, Osborn JC. Inability of the activated partial thromboplastin time to predict heparin levels. Arch Intern Med 1997; 157(21):2475–2479. pmid:9385299
  47. Koerber JM, Smythe MA, Begle RL, Mattson JC, Kershaw BP, Westley SJ. Correlation of activated clotting time and activated partial thromboplastin time to plasma heparin concentration. Pharmacotherapy 1999; 19(8):922–931. pmid:10453963
  48. Smythe MA, Mattson JC, Koerber JM. The heparin anti-Xa therapeutic range: are we there yet? Chest 2002; 121(1):303–304. pmid:11796474
  49. Cuker A, Ptashkin B, Konkle A, et al. Interlaboratory agreement in the monitoring of unfractionated heparin using the anti-factor Xa-correlated activated partial thromboplastin time. J Thromb Haemost 2009; 7(1):80–86. doi:10.1111/j.1538-7836.2008.03224.x
  50. Taylor CT, Petros WP, Ortel TL. Two instruments to determine activated partial thromboplastin time: implications for heparin monitoring. Pharmacotherapy 1999; 19(4):383–387. pmid:10212007
  51. Guervil DJ, Rosenberg AF, Winterstein AG, Harris NS, Johns TE, Zumberg MS. Activated partial thromboplastin time versus antifactor Xa heparin assay in monitoring unfractionated heparin by continuous intravenous infusion. Ann Pharmacother 2011; 45(7–8):861–868. doi:10.1345/aph.1Q161
  52. Fruge KS, Lee YR. Comparison of unfractionated heparin protocols using antifactor Xa monitoring or activated partial thrombin time monitoring. Am J Health Syst Pharm 2015; 72(17 suppl 2):S90–S97. doi:10.2146/sp150016
  53. Rosborough TK. Monitoring unfractionated heparin therapy with antifactor Xa activity results in fewer monitoring tests and dosage changes than monitoring with activated partial thromboplastin time. Pharmacotherapy 1999; 19(6):760–766. pmid:10391423
  54. Rosborough TK, Shepherd MF. Achieving target antifactor Xa activity with a heparin protocol based on sex, age, height, and weight. Pharmacotherapy 2004; 24(6):713–719. doi:10.1592/phco.24.8.713.36067
  55. Smith ML, Wheeler KE. Weight-based heparin protocol using antifactor Xa monitoring. Am J Health Syst Pharm 2010; 67(5):371–374. doi:10.2146/ajhp090123
  56. Bartholomew JR, Kottke-Marchant K. Monitoring anticoagulation therapy in patients with the lupus anticoagulant. J Clin Rheumatol 1998; 4(6):307–312. pmid:19078327
  57. Wool GD, Lu CM; Education Committee of the Academy of Clinical Laboratory Physicians and Scientists. Pathology consultation on anticoagulation monitoring: factor X-related assays. Am J Clin Pathol 2013; 140(5):623–634. doi:10.1309/AJCPR3JTOK7NKDBJ
  58. Mehta TP, Smythe MA, Mattson JC. Strategies for managing heparin therapy in patients with antiphospholipid antibody syndrome. Pharmacotherapy 2011; 31(12):1221–1231. doi:10.1592/phco.31.12.1221
  59. Levine SP, Sorenson RR, Harris MA, Knieriem LK. The effect of platelet factor 4 (PF4) on assays of plasma heparin. Br J Haematol 1984; 57(4):585–596. pmid:6743573
  60. Fisher AR, Bailey CR, Shannon CN, Wielogorski AK. Heparin resistance after aprotinin. Lancet 1992; 340(8829):1230–1231. pmid:1279335
  61. Becker RC, Corrao JM, Bovill EG, et al. Intravenous nitroglycerin-induced heparin resistance: a qualitative antithrombin III abnormality. Am Heart J 1990; 119(6):1254–1261. pmid:2112878
  62. Monagle P, Chan AK, Goldenberg NA, et al. Antithrombotic therapy in neonates and children: Antithrombotic Therapy and Prevention of Thrombosis, 9th ed: American College of Chest Physicians Evidence-Based Clinical Practice Guidelines. Chest 2012; 141(suppl 2):e737S–e801S. doi:10.1378/chest.11-2308
  63. Long E, Pitfield AF, Kissoon N. Anticoagulation therapy: indications, monitoring, and complications. Pediatr Emerg Care 2011; 27(1):55–61. doi:10.1097/PEC.0b013e31820461b1
  64. Andrew M, Schmidt B. Use of heparin in newborn infants. Semin Thromb Hemost 1988; 14(1):28–32. doi:10.1055/s-2007-1002752
  65. Teien AN, Lie M, Abildgaard U. Assay of heparin in plasma using a chromogenic substrate for activated factor X. Thromb Res 1976; 8(3):413–416. pmid:1265712
  66. Vera-Aguillera J, Yousef H, Beltran-Melgarejo D, et al. Clinical scenarios for discordant anti-Xa. Adv Hematol 2016; 2016:4054806. doi:10.1155/2016/4054806
  67. Macedo KA, Tatarian P, Eugenio KR. Influence of direct oral anticoagulants on anti-factor Xa measurements utilized for monitoring heparin. Ann Pharmacother 2018; 52(2):154–159. doi:10.1177/1060028017729481
  68. Wendte J, Voss G, Van Overschelde B. Influence of apixaban on antifactor Xa levels in a patient with acute kidney injury. Am J Health Syst Pharm 2016; 73(8):563–567. doi:10.2146/ajhp150360
  69. Faust AC, Kanyer D, Wittkowsky AK. Managing transitions from oral factor Xa inhibitors to unfractionated heparin infusions. Am J Health Syst Pharm 2016; 73(24):2037–2041. doi:10.2146/ajhp150596
  70. Alhenc-Gelas M, Jestin-Le Guernic C, Vitoux JF, Kher A, Aiach M, Fiessinger JN. Adjusted versus fixed doses of the low-molecular-weight heparin fragmin in the treatment of deep vein thrombosis. Fragmin-Study Group. Thromb Haemost 1994; 71(6):698–702. pmid:7974334
  71. Bates SM, Greer IA, Middeldorp S, Veenstra DL, Prabulos AM, Vandvik PO. VTE, thrombophilia, antithrombotic therapy, and pregnancy: Antithrombotic Therapy and Prevention of Thrombosis, 9th ed: American College of Chest Physicians Evidence-Based Clinical Practice Guidelines. Chest 2012; 141(suppl 2):e691S–e736S. doi:10.1378/chest.11-2300
  72. Bara L, Leizorovicz A, Picolet H, Samama M. Correlation between anti-Xa and occurrence of thrombosis and haemorrhage in post-surgical patients treated with either Logiparin (LMWH) or unfractionated heparin. Post-surgery Logiparin Study Group. Thromb Res 1992; 65(4–5):641–650. pmid:1319619
  73. Prandoni P, Lensing AW, Büller HR, et al. Comparison of subcutaneous low-molecular-weight heparin with intravenous standard heparin in proximal deep-vein thrombosis. Lancet 1992; 339(8791):441–445. pmid:1346817
  74. Walenga JM, Hoppensteadt D, Fareed J. Laboratory monitoring of the clinical effects of low molecular weight heparins. Thromb Res Suppl 1991;14:49–62. pmid:1658970
  75. Elkayam U. Anticoagulation therapy for pregnant women with mechanical prosthetic heart valves: how to improve safety? J Am Coll Cardiol 2017; 69(22):2692–2695. doi:10.1016/j.jacc.2017.04.034
  76. Brophy DF, Wazny LD, Gehr TW, Comstock TJ, Venitz J. The pharmacokinetics of subcutaneous enoxaparin in end-stage renal disease. Pharmacotherapy 2001; 21(2):169–174. pmid:11213853
  77. Becker RC, Spencer FA, Gibson M, et al; TIMI 11A Investigators. Influence of patient characteristics and renal function on factor Xa inhibition pharmacokinetics and pharmacodynamics after enoxaparin administration in non-ST-segment elevation acute coronary syndromes. Am Heart J 2002; 143(5):753–759. pmid:12040334
  78. Chow SL, Zammit K, West K, Dannenhoffer M, Lopez-Candales A. Correlation of antifactor Xa concentrations with renal function in patients on enoxaparin. J Clin Pharmacol 2003; 43(6):586–590. pmid:12817521
  79. Lim W, Dentali F, Eikelboom JW, Crowther MA. Meta-analysis: low-molecular-weight heparin and bleeding in patients with severe renal insufficiency. Ann Intern Med 2006; 144(9):673–684. pmid:16670137
  80. Spinler SA, Inverso SM, Cohen M, Goodman SG, Stringer KA, Antman EM; ESSENCE and TIMI 11B Investigators. Safety and efficacy of unfractionated heparin versus enoxaparin in patients who are obese and patients with severe renal impairment: analysis from the ESSENCE and TIMI 11B studies. Am Heart J 2003; 146(1):33–41. doi:10.1016/S0002-8703(03)00121-2
  81. Cestac P, Bagheri H, Lapeyre-Mestre M, et al. Utilisation and safety of low molecular weight heparins: prospective observational study in medical inpatients. Drug Saf 2003; 26(3):197–207. doi:10.2165/00002018-200326030-00005
  82. Douxfils J, Ageno W, Samama CM, et al. Laboratory testing in patients treated with direct oral anticoagulants: a practical guide for clinicians. J Thromb Haemost 2018; 16(2):209–219. doi:10.1111/jth.13912
  83. Samuelson BT, Cuker A, Siegal DM, Crowther M, Garcia DA. Laboratory assessment of the anticoagulant activity of direct oral anticoagulants: a systematic review. Chest 2017; 151(1):127–138. doi:10.1016/j.chest.2016.08.1462
  84. Gosselin RC, Francart SJ, Hawes EM, Moll S, Dager WE, Adcock DM. Heparin-calibrated chromogenic anti-Xa activity measurements in patients receiving rivaroxaban: can this test be used to quantify drug level? Ann Pharmacother 2015; 49(7):777–783. doi:10.1177/1060028015578451
  85. Levy JH, Ageno W, Chan NC, Crowther M, Verhamme P, Weitz JI; Subcommittee on Control of Anticoagulation. When and how to use antidotes for the reversal of direct oral anticoagulants: guidance from the SSC of the ISTH. J Thromb Haemost 2016; 14(3):623–627. doi:10.1111/jth.13227
  86. Cuker A, Siegal D. Monitoring and reversal of direct oral anticoagulants. Hematology Am Soc Hematol Educ Program 2015; 2015:117–124. doi:10.1182/asheducation-2015.1.117
Article PDF
Author and Disclosure Information

Erika Hutt Centeno, MD
Department of Internal Medicine, Cleveland Clinic; Clinical Instructor, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH

Michael Militello, PharmD, RPh, BCPS
Medical Operations, Inpatient Pharmacy, Cleveland Clinic

Marcelo P. Gomes, MD
Department of Vascular Medicine, Cleveland Clinic; Clinical Assistant Professor, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH

Address: Erika Hutt Centeno, MD, Department of Internal Medicine, G10, Cleveland Clinic; 9500 Euclid Avenue, Cleveland, OH, 44195; huttcee@ccf.org

Issue
Cleveland Clinic Journal of Medicine - 86(6)
Publications
Topics
Page Number
417-425
Legacy Keywords
Activated factor X, factor Xa, anti-factor Xa assays, anti-Xa assays, heparin, activated partial thromboplastin time, aPTT, anticoagulation, monitoring, antithrombotic therapy, venous thromboembolism, VTE, pulmonary embolism, PE, deep vein thrombosis, DVT, Erika hutt Centeno, Michael militello, marcelo gomes
Sections
Author and Disclosure Information

Erika Hutt Centeno, MD
Department of Internal Medicine, Cleveland Clinic; Clinical Instructor, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH

Michael Militello, PharmD, RPh, BCPS
Medical Operations, Inpatient Pharmacy, Cleveland Clinic

Marcelo P. Gomes, MD
Department of Vascular Medicine, Cleveland Clinic; Clinical Assistant Professor, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH

Address: Erika Hutt Centeno, MD, Department of Internal Medicine, G10, Cleveland Clinic; 9500 Euclid Avenue, Cleveland, OH, 44195; huttcee@ccf.org

Author and Disclosure Information

Erika Hutt Centeno, MD
Department of Internal Medicine, Cleveland Clinic; Clinical Instructor, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH

Michael Militello, PharmD, RPh, BCPS
Medical Operations, Inpatient Pharmacy, Cleveland Clinic

Marcelo P. Gomes, MD
Department of Vascular Medicine, Cleveland Clinic; Clinical Assistant Professor, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH

Address: Erika Hutt Centeno, MD, Department of Internal Medicine, G10, Cleveland Clinic; 9500 Euclid Avenue, Cleveland, OH, 44195; huttcee@ccf.org

Article PDF
Article PDF
Related Articles

Should clinicians abandon the activated partial thromboplastin time (aPTT) for monitoring heparin therapy in favor of tests that measure the activity of the patient’s plasma against activated factor X (anti-Xa assays)?

Although other anticoagulants are now available for preventing and treating arterial and venous thromboembolism, unfractionated heparin—which requires laboratory monitoring of therapy—is still widely used. And this monitoring can be challenging. Despite its wide use, the aPTT lacks standardization, and the role of alternative monitoring assays such as the anti-Xa assay is not well defined.

This article reviews the advantages, limitations, and clinical applicability of anti-Xa assays for monitoring therapy with unfractionated heparin and other anticoagulants.

UNFRACTIONATED HEPARIN AND WARFARIN ARE STILL WIDELY USED

Until the mid-1990s, unfractionated heparin and oral vitamin K antagonists (eg, warfarin) were the only anticoagulants widely available for clinical use. These agents have complex pharmacokinetic and pharmacodynamic properties, resulting in highly variable dosing requirements (both between patients and in individual patients) and narrow therapeutic windows, making frequent laboratory monitoring and dose adjustments mandatory.

Over the past 3 decades, other anticoagulants have been approved, including low-molecular-weight heparins, fondaparinux, parenteral direct thrombin inhibitors, and direct oral anticoagulants. While these agents have expanded the options for preventing and treating thromboembolism, unfractionated heparin and warfarin are still the most appropriate choices for many patients, eg, those with stage 4 chronic kidney disease and end-stage renal disease on dialysis, and those with mechanical heart valves.

In addition, unfractionated heparin remains the anticoagulant of choice during procedures such as hemodialysis, percutaneous transluminal angioplasty, and cardiopulmonary bypass, as well as in hospitalized and critically ill patients, who often have acute kidney injury or require frequent interruptions of therapy for invasive procedures. In these scenarios, unfractionated heparin is typically preferred because of its short plasma half-life, complete reversibility by protamine, safety regardless of renal function, and low cost compared with parenteral direct thrombin inhibitors.

As long as unfractionated heparin and warfarin remain important therapies, the need for their laboratory monitoring continues. For warfarin monitoring, the prothrombin time and international normalized ratio are validated and widely reproducible methods. But monitoring unfractionated heparin therapy remains a challenge.

UNFRACTIONATED HEPARIN’S EFFECT IS UNPREDICTABLE

Unfractionated heparin, a negatively charged mucopolysaccharide, inhibits coagulation by binding to antithrombin through the high-affinity pentasaccharide sequence.1–6 Such binding induces a conformational change in the antithrombin molecule, converting it to a rapid inhibitor of several coagulation proteins, especially factors IIa and Xa.2–4

Unfractionated heparin inhibits factors IIa and Xa in a 1:1 ratio, but low-molecular-weight heparins inhibit factor Xa more than factor IIa, with IIa-Xa inhibition ratios ranging from 1:2 to 1:4, owing to their smaller molecular size.7

One of the most important reasons for the unpredictable and highly variable individual responses to unfractionated heparin is that, infused into the blood, the large and negatively charged unfractionated heparin molecules bind nonspecifically to positively charged plasma proteins.7 In patients who are critically ill, have acute infections or inflammatory states, or have undergone major surgery, unfractionated heparin binds to acute-phase proteins that are elevated, particularly factor VIII. This results in fewer free heparin molecules and a variable anticoagulant effect.8

In contrast, low-molecular-weight heparins have longer half-lives and bind less to plasma proteins, resulting in more predictable plasma levels following subcutaneous injection.9

 

 

MONITORING UNFRACTIONATED HEPARIN IMPROVES OUTCOMES

In 1960, Barritt and Jordan10 conducted a small but landmark trial that established the clinical importance of unfractionated heparin for treating venous thromboembolism. None of the patients who received unfractionated heparin for acute pulmonary embolism developed a recurrence during the subsequent 2 weeks, while 50% of those who did not receive it had recurrent pulmonary embolism, fatal in half of the cases.

The importance of achieving a specific aPTT therapeutic target was not demonstrated until a 1972 study by Basu et al,11 in which 162 patients with venous thromboembolism were treated with heparin with a target aPTT of 1.5 to 2.5 times the control value. Patients who suffered recurrent events had subtherapeutic aPTT values on 71% of treatment days, while the rest of the patients, with no recurrences, had subtherapeutic aPTT values only 28% of treatment days. The different outcomes could not be explained by the average daily dose of unfractionated heparin, which was similar in the patients regardless of recurrence.

Subsequent studies showed that the best outcomes occur when unfractionated heparin is given in doses high enough to rapidly achieve a therapeutic prolongation of the aPTT,12–14 and that the total daily dose is also important in preventing recurrences.15,16 Failure to achieve a target aPTT within 24 hours of starting unfractionated heparin is associated with increased risk of recurrent venous thromboembolism.13,17

Raschke et al17 found that patients prospectively randomized to weight-based doses of intravenous unfractionated heparin (bolus plus infusion) achieved significantly higher rates of therapeutic aPTT within 6 hours and 24 hours after starting the infusion, and had significantly lower rates of recurrent venous thromboembolism than those randomized to a fixed unfractionated heparin protocol, without an increase in major bleeding.

Smith et al,18 in a study of 400 consecutive patients with acute pulmonary embolism treated with unfractionated heparin, found that patients who achieved a therapeutic aPTT within 24 hours had lower in-hospital and 30-day mortality rates than those who did not achieve the first therapeutic aPTT until more than 24 hours after starting unfractionated heparin infusion.

Such data lend support to the widely accepted practice and current guideline recommendation8 of using laboratory assays to adjust the dose of unfractionated heparin to achieve and maintain a therapeutic target. The use of dosing nomograms significantly reduces the time to achieve a therapeutic aPTT while minimizing subtherapeutic and supratherapeutic unfractionated heparin levels.19,20

THE aPTT REFLECTS THROMBIN INHIBITION

The aPTT has a log-linear relationship with plasma concentrations of unfractionated heparin,21 but it was not developed specifically for monitoring unfractionated heparin therapy. Originally described in 1953 as a screening tool for hemophilia,22–24 the aPTT is prolonged in the setting of factor deficiencies (typically with levels < 45%, except for factors VII and XIII), as well as lupus anticoagulants and therapy with parenteral direct thrombin inhibitors.8,25,26

Because thrombin (factor IIa) is 10 times more sensitive than factor Xa to inhibition by the heparin-antithrombin complex,4,7 thrombin inhibition appears to be the most likely mechanism by which unfractionated heparin prolongs the aPTT. In contrast, aPTT is minimally or not at all prolonged by low-molecular-weight heparins, which are predominantly factor Xa inhibitors.7

HEPARIN ASSAYS MEASURE UNFRACTIONATED HEPARIN ACTIVITY

While the aPTT is a surrogate marker of unfractionated heparin activity in plasma, unfractionated heparin activity can be measured more precisely by so-called heparin assays, which are typically not direct measures of the plasma concentration of heparins, but rather functional assays that provide indirect estimates. They include protamine sulfate titration assays and anti-Xa assays.

Protamine sulfate titration assays measure the amount of protamine sulfate required to neutralize heparin: the more protamine required, the greater the estimated concentration of unfractionated heparin in plasma.8,27–29 Protamine titration assays are technically demanding, so they are rarely used clinically.

Anti-Xa assays provide a measure of the functional level of heparins in plasma.29–33 Chromogenic anti-Xa assays are available on automated analyzers with standardized kits29,33,34 and may be faster to perform than the aPTT.35

Experiments in rabbits show that unfractionated heparin inhibits thrombus formation and extension at concentrations of 0.2 to 0.4 U/mL as measured by the protamine titration assay,27 which correlated with an anti-Xa activity of 0.35 to 0.67 U/mL in a randomized controlled trial.32

Assays that directly measure the plasma concentration of heparin exist but are not clinically relevant because they also measure heparin molecules lacking the pentasaccharide sequence, which have no anticoagulant activity.36

 

 

ANTI-Xa ASSAY VS THE aPTT

Anti-Xa assays are more expensive than the aPTT and are not available in all hospitals. For these reasons, the aPTT remains the most commonly used laboratory assay for monitoring unfractionated heparin therapy.

However, the aPTT correlates poorly with the activity level of unfractionated heparin in plasma. In one study, an anti-Xa level of 0.3 U/mL corresponded to aPTT results ranging from 47 to 108 seconds.31 Furthermore, in studies that used a heparin therapeutic target based on an aPTT ratio 1.5 to 2.5 times the control aPTT value, the lower end of that target range was often associated with subtherapeutic plasma unfractionated heparin activity measured by anti-Xa and protamine titration assays.28,31

Because of these limitations, individual laboratories should determine their own aPTT therapeutic target ranges for unfractionated heparin based on the response curves obtained with the reagent and coagulometer used. The optimal therapeutic aPTT range for treating acute venous thromboembolism should be defined as the aPTT range (in seconds) that correlates with a plasma activity level of unfractionated heparin of 0.3 to 0.7 U/mL based on a chromogenic anti-Xa assay, or 0.2 to 0.4 U/mL based on a protamine titration assay.32,34–36

Nevertheless, the anticoagulant effect of unfractionated heparin as measured by the aPTT can be unpredictable and can vary widely among individuals and in the same patient.7 This wide variability can be explained by a number of technical and biologic variables. Different commercial aPTT reagents, different lots of the same reagent, and different reagent and instrument combinations have different sensitivities to unfractionated heparin, which can lead to variable aPTT results.37 Moreover, high plasma levels of acute-phase proteins, low plasma antithrombin levels, consumptive coagulopathies, liver failure, and lupus anticoagulants may also affect the aPTT.7,25,32,36–41 These variables account for the poor correlation—ranging from 25% to 66%—reported between aPTT and anti-Xa assays.32,42–48

Such discrepancies may have serious clinical implications: if a patient’s aPTT is low (subtherapeutic) or high (supratherapeutic) but the anti-Xa assay result is within the therapeutic range (0.3–0.7 units/mL), changing the dose of unfractionated heparin (guided by an aPTT nomogram) may increase the risk of bleeding or of recurrent thromboembolism.

CLINICAL APPLICABILITY OF THE ANTI-Xa ASSAY

Neither anti-Xa nor protamine titration assays are standardized across reference laboratories, but chromogenic anti-Xa assays have better interlaboratory correlation than the aPTT49,50 and can be calibrated specifically for unfractionated or low-molecular-weight heparins.29,33

Although reagent costs are higher for chromogenic anti-Xa assays than for the aPTT, some technical variables (described below) may partially offset the cost difference.29,33,41 In addition, unlike the aPTT, anti-Xa assays do not need local calibration; the therapeutic range for unfractionated heparin is the same (0.3–0.7 U/mL) regardless of instrument or reagent.33,41

Most important, studies have found that patients monitored by anti-Xa assay achieve significantly higher rates of therapeutic anticoagulation within 24 and 48 hours after starting unfractionated heparin infusion than those monitored by the aPTT. Fewer dose adjustments and repeat tests are required, which may also result in lower cost.32,51–55

While these studies found chromogenic anti-Xa assays better for achieving laboratory end points, data regarding relevant clinical outcomes are more limited. In a retrospective, observational cohort study,51 the rate of venous thromboembolism or bleeding-related death was 2% in patients receiving unfractionated heparin therapy monitored by anti-Xa assay and 6% in patients monitored by aPTT (P = .62). Rates of major hemorrhage were also not significantly different.

In a randomized controlled trial32 in 131 patients with acute venous thromboembolism and heparin resistance, rates of recurrent venous thromboembolism were 4.6% and 6.1% in the groups randomized to anti-Xa and aPTT monitoring, respectively, whereas overall bleeding rates were 1.5% and 6.1%, respectively. Again, the differences were not statistically significant.

huttcenteno_anti-xaassay_t1.jpg
Though some have suggested that the anti-Xa should be the preferred monitoring assay for intravenous unfractionated heparin therapy,29,41 the ideal assay has not been established by large-scale randomized controlled trials correlating different assays with meaningful clinical outcomes.8,33 Nevertheless, anti-Xa assays are considered the most accurate method of monitoring unfractionated heparin in cases of heparin resistance or lupus anticoagulant, and in other clinical circumstances (Table 1).56–58

Heparin resistance. Some patients require unusually high doses of unfractionated heparin to achieve a therapeutic aPTT: typically, more than 35,000 U over 24 hours,7,8,32 or total daily doses that exceed their estimated weight-based requirements. Heparin resistance has been observed in various clinical settings.7,8,32,37–40,59–61 Patients with heparin resistance monitored by anti-Xa had similar rates of recurrent venous thromboembolism while receiving significantly lower doses of unfractionated heparin than those monitored by the aPTT.32

Lupus anticoagulant. Patients with the specific antiphospholipid antibody known as lupus anticoagulant frequently have a prolonged baseline aPTT,25 making it an unreliable marker of anticoagulant effect for intravenous unfractionated heparin therapy.

Critically ill infants and children. Arachchillage et al35 found that infants (< 1 year old) treated with intravenous unfractionated heparin in an intensive care department had only a 32.4% correlation between aPTT and anti-Xa levels, which was lower than that found in children ages 1 to 15 (66%) and adults (52%). In two-thirds of cases of discordant aPTT and anti-Xa levels, the aPTT was elevated (supratherapeutic) while the anti-Xa assay was within the therapeutic range (0.3–0.7 U/mL). Despite the lack of data on clinical outcomes (eg, rates of thrombosis and bleeding) with the use of an anti-Xa assay, it has been considered the method of choice for unfractionated heparin monitoring in critically ill children, and especially in those under age 1.41,44,62–64

While anti-Xa assays may also be better for unfractionated heparin monitoring in critically ill adults, the lack of clinical outcome data from large-scale randomized trials has precluded evidence-based recommendations favoring them over the aPTT.8,34

 

 

LIMITATIONS OF ANTI-Xa ASSAYS

Anti-Xa assays are hampered by some technical limitations:

Samples must be processed within 1 hour to avoid heparin neutralization.34

Samples must be clear. Hemolyzed or opaque samples (eg, due to bilirubin levels > 6.6 mg/dL or triglyceride levels > 360 mg/dL) cannot be processed, as they can cause falsely low levels.

Exposure to other anticoagulants can interfere with the results. The anti-Xa assay may be unreliable for unfractionated heparin monitoring in patients who are transitioned from low-molecular-weight heparins, fondaparinux, or an oral factor Xa inhibitor (apixaban, betrixaban, edoxaban, rivaroxaban) to intravenous unfractionated heparin, eg, due to hospitalization or acute kidney injury.65,66 Different reports have found that anti-Xa assays may be elevated for as long as 63 to 96 hours after the last dose of oral Xa inhibitors,67–69 potentially resulting in underdosing of unfractionated heparin. In such settings, unfractionated heparin therapy should be monitored by the aPTT.

ANTI-Xa ASSAYS AND LOW-MOLECULAR-WEIGHT HEPARINS

Most patients receiving low-molecular-weight heparins do not need laboratory monitoring.8 Alhenc-Gelas et al70 randomized patients to receive dalteparin in doses either based on weight or guided by anti-Xa assay results, and found that dose adjustments were rare and lacked clinical benefit.

huttcenteno_anti-xaassay_t2.jpg
However, the use of low-molecular-weight heparin-specific anti-Xa assays should be considered for certain patients (Table 2).8

The suggested therapeutic anti-Xa levels for low-molecular-weight heparins are:

  • 0.5–1.2 U/mL for twice-daily enoxaparin
  • 1.0–2.0 U/mL for once-daily enoxaparin or dalteparin.

Levels should be measured at peak plasma level (ie, 3–4 hours after subcutaneous injection, except during pregnancy, when it is 4–6 hours), and only after at least 3 doses of low-molecular-weight heparin.8,71 Unlike the anti-Xa therapeutic range recommended for unfractionated heparin therapy, these ranges are not based on prospective data, and if the assay result is outside the suggested therapeutic target range, current guidelines offer no advice on safely adjusting the dose.8,71

Measuring anti-Xa activity is particularly important for pregnant women with a mechanical prosthetic heart valve who are treated with low-molecular-weight heparins. In this setting, valve thrombosis and cardioembolic events have been reported in patients with peak low-molecular-weight heparin anti-Xa assay levels below or even at the lower end of the therapeutic range, and increased bleeding risk has been reported with elevated anti-Xa levels.71–74 Measuring trough low-molecular-weight heparin anti-Xa levels has been suggested to guide dose adjustments during pregnancy.75

Clearance of low-molecular-weight heparins as measured by the anti-Xa assay is highly correlated with creatinine clearance.76,77 A strong linear correlation has been demonstrated between creatine clearance and anti-Xa levels of enoxaparin after multiple therapeutic doses, and low-molecular-weight heparins accumulate in the plasma, especially in patients with creatine clearance less than 30 mL/min.78 The risk of major bleeding is significantly increased in patients with severe renal insufficiency (creatinine clearance < 30 mL/min) not on dialysis who are treated with either prophylactic or therapeutic doses of low-molecular-weight heparin.79–81 In a meta-analysis, the risk of bleeding with therapeutic-intensity doses of enoxaparin was 4 times higher than with prophylactic-intensity doses.79 Although bleeding risk appears to be reduced when the enoxaparin dose is reduced by 50%,8 the efficacy and safety of this strategy has not been determined by prospective trials.

ANTI-Xa ASSAYS IN PATIENTS RECEIVING DIRECT ORAL ANTICOAGULANTS

Direct oral factor Xa inhibitors cannot be measured accurately by heparin anti-Xa assays. Nevertheless, such assays may be useful to assess whether clinically relevant plasma levels are present in cases of major bleeding, suspected anticoagulant failure, or patient noncompliance.82

Intense research has focused on developing drug-specific chromogenic anti-Xa assays using calibrators and standards for apixaban, edoxaban, and rivaroxaban,82,83 and good linear correlation has been shown with some assays.82,84 In patients treated with oral factor Xa inhibitors who need to undergo an urgent invasive procedure associated with high bleeding risk, use of a specific reversal agent may be considered with drug concentrations more than 30 ng/mL measured by a drug-specific anti-Xa assay. A similar suggestion has been made for drug concentrations more than 50 ng/mL in the setting of major bleeding.85 Unfortunately, such assays are not widely available at this time.82,86

While drug-specific anti-Xa assays could become clinically important to guide reversal strategies, their relevance for drug monitoring remains uncertain. This is because no therapeutic target ranges have been established for any of the direct oral anticoagulants, which were approved on the basis of favorable clinical trial outcomes that neither measured nor were correlated with specific drug levels in plasma. Therefore, a specific anti-Xa level cannot yet be used as a marker of clinical efficacy for any specific oral direct Xa inhibitor.

Should clinicians abandon the activated partial thromboplastin time (aPTT) for monitoring heparin therapy in favor of tests that measure the activity of the patient’s plasma against activated factor X (anti-Xa assays)?

Although other anticoagulants are now available for preventing and treating arterial and venous thromboembolism, unfractionated heparin—which requires laboratory monitoring of therapy—is still widely used. And this monitoring can be challenging. Despite its wide use, the aPTT lacks standardization, and the role of alternative monitoring assays such as the anti-Xa assay is not well defined.

This article reviews the advantages, limitations, and clinical applicability of anti-Xa assays for monitoring therapy with unfractionated heparin and other anticoagulants.

UNFRACTIONATED HEPARIN AND WARFARIN ARE STILL WIDELY USED

Until the mid-1990s, unfractionated heparin and oral vitamin K antagonists (eg, warfarin) were the only anticoagulants widely available for clinical use. These agents have complex pharmacokinetic and pharmacodynamic properties, resulting in highly variable dosing requirements (both between patients and in individual patients) and narrow therapeutic windows, making frequent laboratory monitoring and dose adjustments mandatory.

Over the past 3 decades, other anticoagulants have been approved, including low-molecular-weight heparins, fondaparinux, parenteral direct thrombin inhibitors, and direct oral anticoagulants. While these agents have expanded the options for preventing and treating thromboembolism, unfractionated heparin and warfarin are still the most appropriate choices for many patients, eg, those with stage 4 chronic kidney disease and end-stage renal disease on dialysis, and those with mechanical heart valves.

In addition, unfractionated heparin remains the anticoagulant of choice during procedures such as hemodialysis, percutaneous transluminal angioplasty, and cardiopulmonary bypass, as well as in hospitalized and critically ill patients, who often have acute kidney injury or require frequent interruptions of therapy for invasive procedures. In these scenarios, unfractionated heparin is typically preferred because of its short plasma half-life, complete reversibility by protamine, safety regardless of renal function, and low cost compared with parenteral direct thrombin inhibitors.

As long as unfractionated heparin and warfarin remain important therapies, the need for their laboratory monitoring continues. For warfarin monitoring, the prothrombin time and international normalized ratio are validated and widely reproducible methods. But monitoring unfractionated heparin therapy remains a challenge.

UNFRACTIONATED HEPARIN’S EFFECT IS UNPREDICTABLE

Unfractionated heparin, a negatively charged mucopolysaccharide, inhibits coagulation by binding to antithrombin through the high-affinity pentasaccharide sequence.1–6 Such binding induces a conformational change in the antithrombin molecule, converting it to a rapid inhibitor of several coagulation proteins, especially factors IIa and Xa.2–4

Unfractionated heparin inhibits factors IIa and Xa in a 1:1 ratio, but low-molecular-weight heparins inhibit factor Xa more than factor IIa, with IIa-Xa inhibition ratios ranging from 1:2 to 1:4, owing to their smaller molecular size.7

One of the most important reasons for the unpredictable and highly variable individual responses to unfractionated heparin is that, infused into the blood, the large and negatively charged unfractionated heparin molecules bind nonspecifically to positively charged plasma proteins.7 In patients who are critically ill, have acute infections or inflammatory states, or have undergone major surgery, unfractionated heparin binds to acute-phase proteins that are elevated, particularly factor VIII. This results in fewer free heparin molecules and a variable anticoagulant effect.8

In contrast, low-molecular-weight heparins have longer half-lives and bind less to plasma proteins, resulting in more predictable plasma levels following subcutaneous injection.9

 

 

MONITORING UNFRACTIONATED HEPARIN IMPROVES OUTCOMES

In 1960, Barritt and Jordan10 conducted a small but landmark trial that established the clinical importance of unfractionated heparin for treating venous thromboembolism. None of the patients who received unfractionated heparin for acute pulmonary embolism developed a recurrence during the subsequent 2 weeks, while 50% of those who did not receive it had recurrent pulmonary embolism, fatal in half of the cases.

The importance of achieving a specific aPTT therapeutic target was not demonstrated until a 1972 study by Basu et al,11 in which 162 patients with venous thromboembolism were treated with heparin with a target aPTT of 1.5 to 2.5 times the control value. Patients who suffered recurrent events had subtherapeutic aPTT values on 71% of treatment days, while the rest of the patients, with no recurrences, had subtherapeutic aPTT values only 28% of treatment days. The different outcomes could not be explained by the average daily dose of unfractionated heparin, which was similar in the patients regardless of recurrence.

Subsequent studies showed that the best outcomes occur when unfractionated heparin is given in doses high enough to rapidly achieve a therapeutic prolongation of the aPTT,12–14 and that the total daily dose is also important in preventing recurrences.15,16 Failure to achieve a target aPTT within 24 hours of starting unfractionated heparin is associated with increased risk of recurrent venous thromboembolism.13,17

Raschke et al17 found that patients prospectively randomized to weight-based doses of intravenous unfractionated heparin (bolus plus infusion) achieved significantly higher rates of therapeutic aPTT within 6 hours and 24 hours after starting the infusion, and had significantly lower rates of recurrent venous thromboembolism than those randomized to a fixed unfractionated heparin protocol, without an increase in major bleeding.

Smith et al,18 in a study of 400 consecutive patients with acute pulmonary embolism treated with unfractionated heparin, found that patients who achieved a therapeutic aPTT within 24 hours had lower in-hospital and 30-day mortality rates than those who did not achieve the first therapeutic aPTT until more than 24 hours after starting unfractionated heparin infusion.

Such data lend support to the widely accepted practice and current guideline recommendation8 of using laboratory assays to adjust the dose of unfractionated heparin to achieve and maintain a therapeutic target. The use of dosing nomograms significantly reduces the time to achieve a therapeutic aPTT while minimizing subtherapeutic and supratherapeutic unfractionated heparin levels.19,20

THE aPTT REFLECTS THROMBIN INHIBITION

The aPTT has a log-linear relationship with plasma concentrations of unfractionated heparin,21 but it was not developed specifically for monitoring unfractionated heparin therapy. Originally described in 1953 as a screening tool for hemophilia,22–24 the aPTT is prolonged in the setting of factor deficiencies (typically with levels < 45%, except for factors VII and XIII), as well as lupus anticoagulants and therapy with parenteral direct thrombin inhibitors.8,25,26

Because thrombin (factor IIa) is 10 times more sensitive than factor Xa to inhibition by the heparin-antithrombin complex,4,7 thrombin inhibition appears to be the most likely mechanism by which unfractionated heparin prolongs the aPTT. In contrast, aPTT is minimally or not at all prolonged by low-molecular-weight heparins, which are predominantly factor Xa inhibitors.7

HEPARIN ASSAYS MEASURE UNFRACTIONATED HEPARIN ACTIVITY

While the aPTT is a surrogate marker of unfractionated heparin activity in plasma, unfractionated heparin activity can be measured more precisely by so-called heparin assays, which are typically not direct measures of the plasma concentration of heparins, but rather functional assays that provide indirect estimates. They include protamine sulfate titration assays and anti-Xa assays.

Protamine sulfate titration assays measure the amount of protamine sulfate required to neutralize heparin: the more protamine required, the greater the estimated concentration of unfractionated heparin in plasma.8,27–29 Protamine titration assays are technically demanding, so they are rarely used clinically.

Anti-Xa assays provide a measure of the functional level of heparins in plasma.29–33 Chromogenic anti-Xa assays are available on automated analyzers with standardized kits29,33,34 and may be faster to perform than the aPTT.35

Experiments in rabbits show that unfractionated heparin inhibits thrombus formation and extension at concentrations of 0.2 to 0.4 U/mL as measured by the protamine titration assay,27 which correlated with an anti-Xa activity of 0.35 to 0.67 U/mL in a randomized controlled trial.32

Assays that directly measure the plasma concentration of heparin exist but are not clinically relevant because they also measure heparin molecules lacking the pentasaccharide sequence, which have no anticoagulant activity.36

 

 

ANTI-Xa ASSAY VS THE aPTT

Anti-Xa assays are more expensive than the aPTT and are not available in all hospitals. For these reasons, the aPTT remains the most commonly used laboratory assay for monitoring unfractionated heparin therapy.

However, the aPTT correlates poorly with the activity level of unfractionated heparin in plasma. In one study, an anti-Xa level of 0.3 U/mL corresponded to aPTT results ranging from 47 to 108 seconds.31 Furthermore, in studies that used a heparin therapeutic target based on an aPTT ratio 1.5 to 2.5 times the control aPTT value, the lower end of that target range was often associated with subtherapeutic plasma unfractionated heparin activity measured by anti-Xa and protamine titration assays.28,31

Because of these limitations, individual laboratories should determine their own aPTT therapeutic target ranges for unfractionated heparin based on the response curves obtained with the reagent and coagulometer used. The optimal therapeutic aPTT range for treating acute venous thromboembolism should be defined as the aPTT range (in seconds) that correlates with a plasma activity level of unfractionated heparin of 0.3 to 0.7 U/mL based on a chromogenic anti-Xa assay, or 0.2 to 0.4 U/mL based on a protamine titration assay.32,34–36

Nevertheless, the anticoagulant effect of unfractionated heparin as measured by the aPTT can be unpredictable and can vary widely among individuals and in the same patient.7 This wide variability can be explained by a number of technical and biologic variables. Different commercial aPTT reagents, different lots of the same reagent, and different reagent and instrument combinations have different sensitivities to unfractionated heparin, which can lead to variable aPTT results.37 Moreover, high plasma levels of acute-phase proteins, low plasma antithrombin levels, consumptive coagulopathies, liver failure, and lupus anticoagulants may also affect the aPTT.7,25,32,36–41 These variables account for the poor correlation—ranging from 25% to 66%—reported between aPTT and anti-Xa assays.32,42–48

Such discrepancies may have serious clinical implications: if a patient’s aPTT is low (subtherapeutic) or high (supratherapeutic) but the anti-Xa assay result is within the therapeutic range (0.3–0.7 units/mL), changing the dose of unfractionated heparin (guided by an aPTT nomogram) may increase the risk of bleeding or of recurrent thromboembolism.

CLINICAL APPLICABILITY OF THE ANTI-Xa ASSAY

Neither anti-Xa nor protamine titration assays are standardized across reference laboratories, but chromogenic anti-Xa assays have better interlaboratory correlation than the aPTT49,50 and can be calibrated specifically for unfractionated or low-molecular-weight heparins.29,33

Although reagent costs are higher for chromogenic anti-Xa assays than for the aPTT, some technical variables (described below) may partially offset the cost difference.29,33,41 In addition, unlike the aPTT, anti-Xa assays do not need local calibration; the therapeutic range for unfractionated heparin is the same (0.3–0.7 U/mL) regardless of instrument or reagent.33,41

Most important, studies have found that patients monitored by anti-Xa assay achieve significantly higher rates of therapeutic anticoagulation within 24 and 48 hours after starting unfractionated heparin infusion than those monitored by the aPTT. Fewer dose adjustments and repeat tests are required, which may also result in lower cost.32,51–55

While these studies found chromogenic anti-Xa assays better for achieving laboratory end points, data regarding relevant clinical outcomes are more limited. In a retrospective, observational cohort study,51 the rate of venous thromboembolism or bleeding-related death was 2% in patients receiving unfractionated heparin therapy monitored by anti-Xa assay and 6% in patients monitored by aPTT (P = .62). Rates of major hemorrhage were also not significantly different.

In a randomized controlled trial32 in 131 patients with acute venous thromboembolism and heparin resistance, rates of recurrent venous thromboembolism were 4.6% and 6.1% in the groups randomized to anti-Xa and aPTT monitoring, respectively, whereas overall bleeding rates were 1.5% and 6.1%, respectively. Again, the differences were not statistically significant.

huttcenteno_anti-xaassay_t1.jpg
Though some have suggested that the anti-Xa should be the preferred monitoring assay for intravenous unfractionated heparin therapy,29,41 the ideal assay has not been established by large-scale randomized controlled trials correlating different assays with meaningful clinical outcomes.8,33 Nevertheless, anti-Xa assays are considered the most accurate method of monitoring unfractionated heparin in cases of heparin resistance or lupus anticoagulant, and in other clinical circumstances (Table 1).56–58

Heparin resistance. Some patients require unusually high doses of unfractionated heparin to achieve a therapeutic aPTT: typically, more than 35,000 U over 24 hours,7,8,32 or total daily doses that exceed their estimated weight-based requirements. Heparin resistance has been observed in various clinical settings.7,8,32,37–40,59–61 Patients with heparin resistance monitored by anti-Xa had similar rates of recurrent venous thromboembolism while receiving significantly lower doses of unfractionated heparin than those monitored by the aPTT.32

Lupus anticoagulant. Patients with the specific antiphospholipid antibody known as lupus anticoagulant frequently have a prolonged baseline aPTT,25 making it an unreliable marker of anticoagulant effect for intravenous unfractionated heparin therapy.

Critically ill infants and children. Arachchillage et al35 found that infants (< 1 year old) treated with intravenous unfractionated heparin in an intensive care department had only a 32.4% correlation between aPTT and anti-Xa levels, which was lower than that found in children ages 1 to 15 (66%) and adults (52%). In two-thirds of cases of discordant aPTT and anti-Xa levels, the aPTT was elevated (supratherapeutic) while the anti-Xa assay was within the therapeutic range (0.3–0.7 U/mL). Despite the lack of data on clinical outcomes (eg, rates of thrombosis and bleeding) with the use of an anti-Xa assay, it has been considered the method of choice for unfractionated heparin monitoring in critically ill children, and especially in those under age 1.41,44,62–64

While anti-Xa assays may also be better for unfractionated heparin monitoring in critically ill adults, the lack of clinical outcome data from large-scale randomized trials has precluded evidence-based recommendations favoring them over the aPTT.8,34

 

 

LIMITATIONS OF ANTI-Xa ASSAYS

Anti-Xa assays are hampered by some technical limitations:

Samples must be processed within 1 hour to avoid heparin neutralization.34

Samples must be clear. Hemolyzed or opaque samples (eg, due to bilirubin levels > 6.6 mg/dL or triglyceride levels > 360 mg/dL) cannot be processed, as they can cause falsely low levels.

Exposure to other anticoagulants can interfere with the results. The anti-Xa assay may be unreliable for unfractionated heparin monitoring in patients who are transitioned from low-molecular-weight heparins, fondaparinux, or an oral factor Xa inhibitor (apixaban, betrixaban, edoxaban, rivaroxaban) to intravenous unfractionated heparin, eg, due to hospitalization or acute kidney injury.65,66 Different reports have found that anti-Xa assays may be elevated for as long as 63 to 96 hours after the last dose of oral Xa inhibitors,67–69 potentially resulting in underdosing of unfractionated heparin. In such settings, unfractionated heparin therapy should be monitored by the aPTT.

ANTI-Xa ASSAYS AND LOW-MOLECULAR-WEIGHT HEPARINS

Most patients receiving low-molecular-weight heparins do not need laboratory monitoring.8 Alhenc-Gelas et al70 randomized patients to receive dalteparin in doses either based on weight or guided by anti-Xa assay results, and found that dose adjustments were rare and lacked clinical benefit.

huttcenteno_anti-xaassay_t2.jpg
However, the use of low-molecular-weight heparin-specific anti-Xa assays should be considered for certain patients (Table 2).8

The suggested therapeutic anti-Xa levels for low-molecular-weight heparins are:

  • 0.5–1.2 U/mL for twice-daily enoxaparin
  • 1.0–2.0 U/mL for once-daily enoxaparin or dalteparin.

Levels should be measured at peak plasma level (ie, 3–4 hours after subcutaneous injection, except during pregnancy, when it is 4–6 hours), and only after at least 3 doses of low-molecular-weight heparin.8,71 Unlike the anti-Xa therapeutic range recommended for unfractionated heparin therapy, these ranges are not based on prospective data, and if the assay result is outside the suggested therapeutic target range, current guidelines offer no advice on safely adjusting the dose.8,71

Measuring anti-Xa activity is particularly important for pregnant women with a mechanical prosthetic heart valve who are treated with low-molecular-weight heparins. In this setting, valve thrombosis and cardioembolic events have been reported in patients with peak low-molecular-weight heparin anti-Xa assay levels below or even at the lower end of the therapeutic range, and increased bleeding risk has been reported with elevated anti-Xa levels.71–74 Measuring trough low-molecular-weight heparin anti-Xa levels has been suggested to guide dose adjustments during pregnancy.75

Clearance of low-molecular-weight heparins as measured by the anti-Xa assay is highly correlated with creatinine clearance.76,77 A strong linear correlation has been demonstrated between creatine clearance and anti-Xa levels of enoxaparin after multiple therapeutic doses, and low-molecular-weight heparins accumulate in the plasma, especially in patients with creatine clearance less than 30 mL/min.78 The risk of major bleeding is significantly increased in patients with severe renal insufficiency (creatinine clearance < 30 mL/min) not on dialysis who are treated with either prophylactic or therapeutic doses of low-molecular-weight heparin.79–81 In a meta-analysis, the risk of bleeding with therapeutic-intensity doses of enoxaparin was 4 times higher than with prophylactic-intensity doses.79 Although bleeding risk appears to be reduced when the enoxaparin dose is reduced by 50%,8 the efficacy and safety of this strategy has not been determined by prospective trials.

ANTI-Xa ASSAYS IN PATIENTS RECEIVING DIRECT ORAL ANTICOAGULANTS

Direct oral factor Xa inhibitors cannot be measured accurately by heparin anti-Xa assays. Nevertheless, such assays may be useful to assess whether clinically relevant plasma levels are present in cases of major bleeding, suspected anticoagulant failure, or patient noncompliance.82

Intense research has focused on developing drug-specific chromogenic anti-Xa assays using calibrators and standards for apixaban, edoxaban, and rivaroxaban,82,83 and good linear correlation has been shown with some assays.82,84 In patients treated with oral factor Xa inhibitors who need to undergo an urgent invasive procedure associated with high bleeding risk, use of a specific reversal agent may be considered with drug concentrations more than 30 ng/mL measured by a drug-specific anti-Xa assay. A similar suggestion has been made for drug concentrations more than 50 ng/mL in the setting of major bleeding.85 Unfortunately, such assays are not widely available at this time.82,86

While drug-specific anti-Xa assays could become clinically important to guide reversal strategies, their relevance for drug monitoring remains uncertain. This is because no therapeutic target ranges have been established for any of the direct oral anticoagulants, which were approved on the basis of favorable clinical trial outcomes that neither measured nor were correlated with specific drug levels in plasma. Therefore, a specific anti-Xa level cannot yet be used as a marker of clinical efficacy for any specific oral direct Xa inhibitor.

References
  1. Abildgaard U. Highly purified antithrombin 3 with heparin cofactor activity prepared by disc electrophoresis. Scand J Clin Lab Invest 1968; 21(1):89–91. pmid:5637480
  2. Rosenberg RD, Lam L. Correlation between structure and function of heparin. Proc Natl Acad Sci USA 1979; 76(3):1218–1222. pmid:286307
  3. Lindahl U, Bäckström G, Höök M, Thunberg L, Fransson LA, Linker A. Structure of the antithrombin-binding site of heparin. Proc Natl Acad Sci USA 1979; 76(7):3198–3202. pmid:226960
  4. Rosenberg RD, Rosenberg JS. Natural anticoagulant mechanisms. J Clin Invest 1984; 74(1):1–6. doi:10.1172/JCI111389
  5. Casu B, Oreste P, Torri G, et al. The structure of heparin oligosaccharide fragments with high anti-(factor Xa) activity containing the minimal antithrombin III-binding sequence. Chemical and 13C nuclear-magnetic-resonance studies. Biochem J 1981; 197(3):599–609. pmid:7325974
  6. Choay J, Lormeau JC, Petitou M, Sinaÿ P, Fareed J. Structural studies on a biologically active hexasaccharide obtained from heparin. Ann NY Acad Sci 1981; 370: 644–649. pmid:6943974
  7. Hirsh J, Warkentin TE, Shaughnessy SG, et al. Heparin and low-molecular-weight heparin: mechanisms of action, pharmacokinetics, dosing, monitoring, efficacy, and safety. Chest 2001; 119(suppl 1):64S–94S. pmid:11157643
  8. Garcia DA, Baglin TP, Weitz JI, Samama MM. Parenteral anticoagulants: antithrombotic therapy and prevention of thrombosis, 9th ed: American College of Chest Physicians Evidence-Based Clinical Practice Guidelines. Chest 2012; 141(suppl 2):e24S–e43S. doi:10.1378/chest.11-2291
  9. Hirsh J, Levine M. Low-molecular weight heparin. Blood 1992; 79(1):1–17. pmid:1309422
  10. Barritt DW, Jordan SC. Anticoagulant drugs in the treatment of pulmonary embolism. A controlled trial. Lancet 1960; 1(7138):1309–1312. pmid:13797091
  11. Basu D, Gallus A, Hirsh J, Cade J. A prospective study of the value of monitoring heparin treatment with the activated partial thromboplastin time. N Engl J Med 1972; 287(7):324–327. doi:10.1056/NEJM197208172870703
  12. Hull RD, Raskob GE, Hirsh J, et al. Continuous intravenous heparin compared with intermittent subcutaneous heparin in the initial treatment of proximal-vein thrombosis. N Engl J Med 1986; 315(18):1109–1114. doi:10.1056/NEJM198610303151801
  13. Hull RD, Raskob GE, Brant RF, Pineo GF, Valentine KA. Relation between the time to achieve the lower limit of the APTT therapeutic range and recurrent venous thromboembolism during heparin treatment for deep vein thrombosis. Arch Intern Med 1997; 157(22):2562–2568. pmid:9531224
  14. Hull RD, Raskob GE, Brant RF, Pineo GF, Valentine KA. The importance of initial heparin treatment on long-term clinical outcomes of antithrombotic therapy. The emerging theme of delayed recurrence. Arch Intern Med 1997; 157(20):2317–2321. pmid:9361572
  15. Anand S, Ginsberg JS, Kearon C, Gent M, Hirsh J. The relation between the activated partial thromboplastin time response and recurrence in patients with venous thrombosis treated with continuous intravenous heparin. Arch Intern Med 1996; 156(15):1677–1681. pmid:8694666
  16. Anand SS, Bates S, Ginsberg JS, et al. Recurrent venous thrombosis and heparin therapy: an evaluation of the importance of early activated partial thromboplastin times. Arch Intern Med 1999; 159(17):2029–2032. pmid:10510988
  17. Raschke RA, Reilly BM, Guidry JR, Fontana JR, Srinivas S. The weight-based heparin dosing nomogram compared with a “standard care” nomogram. A randomized controlled trial. Ann Intern Med 1993; 119(9):874–881. pmid:8214998
  18. Smith SB, Geske JB, Maguire JM, Zane NA, Carter RE, Morgenthaler TI. Early anticoagulation is associated with reduced mortality for acute pulmonary embolism. Chest 2010; 137(6):1382–1390. doi:10.1378/chest.09-0959
  19. Cruickshank MK, Levine MN, Hirsh J, Roberts R, Siguenza M. A standard heparin nomogram for the management of heparin therapy. Arch Intern Med 1991; 151(2):333–337. pmid:1789820
  20. Raschke RA, Gollihare B, Peirce J. The effectiveness of implementing the weight-based heparin nomogram as a practice guideline. Arch Intern Med 1996; 156(15):1645–1649. pmid:8694662
  21. Simko RJ, Tsung FF, Stanek EJ. Activated clotting time versus activated partial thromboplastin time for therapeutic monitoring of heparin. Ann Pharmacother 1995; 29(10):1015–1021. doi:10.1177/106002809502901012
  22. Langdell RD, Wagner RH, Brinkhous KM. Effect of antihemophilic factor on one-stage clotting tests; a presumptive test for hemophilia and a simple one-stage antihemophilic factor assy procedure. J Lab Clin Med 1953; 41(4):637–647.
  23. White GC 2nd. The partial thromboplastin time: defining an era in coagulation. J Thromb Haemost 2003; 1(11):2267–2270. pmid:14629454
  24. Proctor RR, Rapaport SI. The partial thromboplastin time with kaolin. A simple screening test for first stage plasma clotting factor deficiencies. Am J Clin Pathol 1961; 36:212–219. pmid:13738153
  25. Brandt JT, Triplett DA, Rock WA, Bovill EG, Arkin CF. Effect of lupus anticoagulants on the activated partial thromboplastin time. Results of the College of American Pathologists survey program. Arch Pathol Lab Med 1991; 115(2):109–114. pmid:1899555
  26. Tripodi A, Mannucci PM. Activated partial thromboplastin time (APTT). New indications for an old test? J Thromb Haemost 2006; 4(4):750–751. doi:10.1111/j.1538-7836.2006.01857.x
  27. Chiu HM, Hirsh J, Yung WL, Regoeczi E, Gent M. Relationship between the anticoagulant and antithrombotic effects of heparin in experimental venous thrombosis. Blood 1977; 49(2):171–184. pmid:831872
  28. Brill-Edwards P, Ginsberg JS, Johnston M, Hirsh J. Establishing a therapeutic range for heparin therapy. Ann Intern Med 1993; 119(2):104–109. pmid:8512158
  29. Vandiver JW, Vondracek TG. Antifactor Xa levels versus activated partial thromboplastin time for monitoring unfractionated heparin. Pharmacotherapy 2012; 32(6):546–558. doi:10.1002/j.1875-9114.2011.01049.x
  30. Newall F. Anti-factor Xa (anti-Xa). In: Monagle P, ed. Haemostasis: Methods and Protocols. New York, NY: Springer-Verlag; 2013.
  31. Bates SM, Weitz JI, Johnston M, Hirsh J, Ginsberg JS. Use of a fixed activated partial thromboplastin time ratio to establish a therapeutic range for unfractionated heparin. Arch Intern Med 2001; 161(3):385–391. pmid:11176764
  32. Levine MN, Hirsh J, Gent M, et al. A randomized trial comparing activated thromboplastin time with heparin assay in patients with acute venous thromboembolism requiring large doses of heparin. Arch Intern Med 1994; 154(1):49–56. pmid:8267489
  33. Wool GD, Lu CM; Education Committee of the Academy of Clinical Laboratory Physicians and Scientists. Pathology consultation on anticoagulation monitoring: factor X-related assays. Am J Clin Pathol 2013; 140(5):623–634. doi:10.1309/AJCPR3JTOK7NKDBJ
  34. Lehman CM, Frank EL. Laboratory monitoring of heparin therapy: partial thromboplastin time or anti-Xa assay? Lab Med 2009; 40(1):47–51. doi:10.1309/LM9NJGW2ZIOLPHY6
  35. Arachchillage DR, Kamani F, Deplano S, Banya W, Laffan M. Should we abandon the aPTT for monitoring unfractionated heparin? Thromb Res 2017; 157:157–161. doi:10.1016/j.thromres.2017.07.006
  36. Olson JD, Arkin CA, Brandt JT, et al. College of American Pathologists Conference XXXI on Laboratory Monitoring of Anticoagulant Therapy: laboratory monitoring of unfractionated heparin therapy. Arch Pathol Lab Med 1998; 122(9):782–798. pmid:9740136
  37. Eikelboom JW, Hirsh J. Monitoring unfractionated heparin with the aPTT: time for a fresh look. Thromb Haemost 2006; 96(5):547–552. pmid:17080209
  38. Young E, Prins M, Levine MN, Hirsh J. Heparin binding to plasma proteins, an important mechanism of heparin resistance. Thromb Haemost 1992; 67(6):639–643. pmid:1509402
  39. Edson JR, Krivit W, White JG. Kaolin partial thromboplastin time: high levels of procoagulants producing short clotting times or masking deficiencies of other procoagulants or low concentrations of anticoagulants. J Lab Clin Med 1967; 70(3):463–470. pmid:6072020
  40. Whitfield LR, Lele AS, Levy G. Effect of pregnancy on the relationship between concentration and anticoagulant action of heparin. Clin Pharmacol Ther 1983; 34(1):23–28. pmid:6861435
  41. Marci CD, Prager D. A review of the clinical indications for the plasma heparin assay. Am J Clin Pathol 1993; 99(5):546–550.
  42. Takemoto CM, Streiff MB, Shermock KM, et al. Activated partial thromboplastin time and anti-Xa measurements in heparin monitoring: biochemical basis of discordance. Am J Clin Pathol 2013; 139(4):450–456. doi:10.1309/AJCPS6OW6DYNOGNH
  43. Adatya S, Uriel N, Yarmohammadi H, et al. Anti-factor Xa and activated partial thromboplastin time measurements for heparin monitoring in mechanical circulatory support. JACC Heart Fail 2015; 3(4):314–322. doi:10.1016/j.jchf.2014.11.009
  44. Kuhle S, Eulmesekian P, Kavanagh B, et al. Lack of correlation between heparin dose and standard clinical monitoring tests in treatment with unfractionated heparin in critically ill children. Haematologica 2007; 92(4):554–557. pmid:17488668
  45. Price EA, Jin J, Nguyen HM, Krishnan G, Bowen R, Zehnder JL. Discordant aPTT and anti-Xa values and outcomes in hospitalized patients treated with intravenous unfractionated heparin. Ann Pharmacother 2013; 47(2):151–158. doi:10.1345/aph.1R635
  46. Baker BA, Adelman MD, Smith PA, Osborn JC. Inability of the activated partial thromboplastin time to predict heparin levels. Arch Intern Med 1997; 157(21):2475–2479. pmid:9385299
  47. Koerber JM, Smythe MA, Begle RL, Mattson JC, Kershaw BP, Westley SJ. Correlation of activated clotting time and activated partial thromboplastin time to plasma heparin concentration. Pharmacotherapy 1999; 19(8):922–931. pmid:10453963
  48. Smythe MA, Mattson JC, Koerber JM. The heparin anti-Xa therapeutic range: are we there yet? Chest 2002; 121(1):303–304. pmid:11796474
  49. Cuker A, Ptashkin B, Konkle A, et al. Interlaboratory agreement in the monitoring of unfractionated heparin using the anti-factor Xa-correlated activated partial thromboplastin time. J Thromb Haemost 2009; 7(1):80–86. doi:10.1111/j.1538-7836.2008.03224.x
  50. Taylor CT, Petros WP, Ortel TL. Two instruments to determine activated partial thromboplastin time: implications for heparin monitoring. Pharmacotherapy 1999; 19(4):383–387. pmid:10212007
  51. Guervil DJ, Rosenberg AF, Winterstein AG, Harris NS, Johns TE, Zumberg MS. Activated partial thromboplastin time versus antifactor Xa heparin assay in monitoring unfractionated heparin by continuous intravenous infusion. Ann Pharmacother 2011; 45(7–8):861–868. doi:10.1345/aph.1Q161
  52. Fruge KS, Lee YR. Comparison of unfractionated heparin protocols using antifactor Xa monitoring or activated partial thrombin time monitoring. Am J Health Syst Pharm 2015; 72(17 suppl 2):S90–S97. doi:10.2146/sp150016
  53. Rosborough TK. Monitoring unfractionated heparin therapy with antifactor Xa activity results in fewer monitoring tests and dosage changes than monitoring with activated partial thromboplastin time. Pharmacotherapy 1999; 19(6):760–766. pmid:10391423
  54. Rosborough TK, Shepherd MF. Achieving target antifactor Xa activity with a heparin protocol based on sex, age, height, and weight. Pharmacotherapy 2004; 24(6):713–719. doi:10.1592/phco.24.8.713.36067
  55. Smith ML, Wheeler KE. Weight-based heparin protocol using antifactor Xa monitoring. Am J Health Syst Pharm 2010; 67(5):371–374. doi:10.2146/ajhp090123
  56. Bartholomew JR, Kottke-Marchant K. Monitoring anticoagulation therapy in patients with the lupus anticoagulant. J Clin Rheumatol 1998; 4(6):307–312. pmid:19078327
  57. Wool GD, Lu CM; Education Committee of the Academy of Clinical Laboratory Physicians and Scientists. Pathology consultation on anticoagulation monitoring: factor X-related assays. Am J Clin Pathol 2013; 140(5):623–634. doi:10.1309/AJCPR3JTOK7NKDBJ
  58. Mehta TP, Smythe MA, Mattson JC. Strategies for managing heparin therapy in patients with antiphospholipid antibody syndrome. Pharmacotherapy 2011; 31(12):1221–1231. doi:10.1592/phco.31.12.1221
  59. Levine SP, Sorenson RR, Harris MA, Knieriem LK. The effect of platelet factor 4 (PF4) on assays of plasma heparin. Br J Haematol 1984; 57(4):585–596. pmid:6743573
  60. Fisher AR, Bailey CR, Shannon CN, Wielogorski AK. Heparin resistance after aprotinin. Lancet 1992; 340(8829):1230–1231. pmid:1279335
  61. Becker RC, Corrao JM, Bovill EG, et al. Intravenous nitroglycerin-induced heparin resistance: a qualitative antithrombin III abnormality. Am Heart J 1990; 119(6):1254–1261. pmid:2112878
  62. Monagle P, Chan AK, Goldenberg NA, et al. Antithrombotic therapy in neonates and children: Antithrombotic Therapy and Prevention of Thrombosis, 9th ed: American College of Chest Physicians Evidence-Based Clinical Practice Guidelines. Chest 2012; 141(suppl 2):e737S–e801S. doi:10.1378/chest.11-2308
  63. Long E, Pitfield AF, Kissoon N. Anticoagulation therapy: indications, monitoring, and complications. Pediatr Emerg Care 2011; 27(1):55–61. doi:10.1097/PEC.0b013e31820461b1
  64. Andrew M, Schmidt B. Use of heparin in newborn infants. Semin Thromb Hemost 1988; 14(1):28–32. doi:10.1055/s-2007-1002752
  65. Teien AN, Lie M, Abildgaard U. Assay of heparin in plasma using a chromogenic substrate for activated factor X. Thromb Res 1976; 8(3):413–416. pmid:1265712
  66. Vera-Aguillera J, Yousef H, Beltran-Melgarejo D, et al. Clinical scenarios for discordant anti-Xa. Adv Hematol 2016; 2016:4054806. doi:10.1155/2016/4054806
  67. Macedo KA, Tatarian P, Eugenio KR. Influence of direct oral anticoagulants on anti-factor Xa measurements utilized for monitoring heparin. Ann Pharmacother 2018; 52(2):154–159. doi:10.1177/1060028017729481
  68. Wendte J, Voss G, Van Overschelde B. Influence of apixaban on antifactor Xa levels in a patient with acute kidney injury. Am J Health Syst Pharm 2016; 73(8):563–567. doi:10.2146/ajhp150360
  69. Faust AC, Kanyer D, Wittkowsky AK. Managing transitions from oral factor Xa inhibitors to unfractionated heparin infusions. Am J Health Syst Pharm 2016; 73(24):2037–2041. doi:10.2146/ajhp150596
  70. Alhenc-Gelas M, Jestin-Le Guernic C, Vitoux JF, Kher A, Aiach M, Fiessinger JN. Adjusted versus fixed doses of the low-molecular-weight heparin fragmin in the treatment of deep vein thrombosis. Fragmin-Study Group. Thromb Haemost 1994; 71(6):698–702. pmid:7974334
  71. Bates SM, Greer IA, Middeldorp S, Veenstra DL, Prabulos AM, Vandvik PO. VTE, thrombophilia, antithrombotic therapy, and pregnancy: Antithrombotic Therapy and Prevention of Thrombosis, 9th ed: American College of Chest Physicians Evidence-Based Clinical Practice Guidelines. Chest 2012; 141(suppl 2):e691S–e736S. doi:10.1378/chest.11-2300
  72. Bara L, Leizorovicz A, Picolet H, Samama M. Correlation between anti-Xa and occurrence of thrombosis and haemorrhage in post-surgical patients treated with either Logiparin (LMWH) or unfractionated heparin. Post-surgery Logiparin Study Group. Thromb Res 1992; 65(4–5):641–650. pmid:1319619
  73. Prandoni P, Lensing AW, Büller HR, et al. Comparison of subcutaneous low-molecular-weight heparin with intravenous standard heparin in proximal deep-vein thrombosis. Lancet 1992; 339(8791):441–445. pmid:1346817
  74. Walenga JM, Hoppensteadt D, Fareed J. Laboratory monitoring of the clinical effects of low molecular weight heparins. Thromb Res Suppl 1991;14:49–62. pmid:1658970
  75. Elkayam U. Anticoagulation therapy for pregnant women with mechanical prosthetic heart valves: how to improve safety? J Am Coll Cardiol 2017; 69(22):2692–2695. doi:10.1016/j.jacc.2017.04.034
  76. Brophy DF, Wazny LD, Gehr TW, Comstock TJ, Venitz J. The pharmacokinetics of subcutaneous enoxaparin in end-stage renal disease. Pharmacotherapy 2001; 21(2):169–174. pmid:11213853
  77. Becker RC, Spencer FA, Gibson M, et al; TIMI 11A Investigators. Influence of patient characteristics and renal function on factor Xa inhibition pharmacokinetics and pharmacodynamics after enoxaparin administration in non-ST-segment elevation acute coronary syndromes. Am Heart J 2002; 143(5):753–759. pmid:12040334
  78. Chow SL, Zammit K, West K, Dannenhoffer M, Lopez-Candales A. Correlation of antifactor Xa concentrations with renal function in patients on enoxaparin. J Clin Pharmacol 2003; 43(6):586–590. pmid:12817521
  79. Lim W, Dentali F, Eikelboom JW, Crowther MA. Meta-analysis: low-molecular-weight heparin and bleeding in patients with severe renal insufficiency. Ann Intern Med 2006; 144(9):673–684. pmid:16670137
  80. Spinler SA, Inverso SM, Cohen M, Goodman SG, Stringer KA, Antman EM; ESSENCE and TIMI 11B Investigators. Safety and efficacy of unfractionated heparin versus enoxaparin in patients who are obese and patients with severe renal impairment: analysis from the ESSENCE and TIMI 11B studies. Am Heart J 2003; 146(1):33–41. doi:10.1016/S0002-8703(03)00121-2
  81. Cestac P, Bagheri H, Lapeyre-Mestre M, et al. Utilisation and safety of low molecular weight heparins: prospective observational study in medical inpatients. Drug Saf 2003; 26(3):197–207. doi:10.2165/00002018-200326030-00005
  82. Douxfils J, Ageno W, Samama CM, et al. Laboratory testing in patients treated with direct oral anticoagulants: a practical guide for clinicians. J Thromb Haemost 2018; 16(2):209–219. doi:10.1111/jth.13912
  83. Samuelson BT, Cuker A, Siegal DM, Crowther M, Garcia DA. Laboratory assessment of the anticoagulant activity of direct oral anticoagulants: a systematic review. Chest 2017; 151(1):127–138. doi:10.1016/j.chest.2016.08.1462
  84. Gosselin RC, Francart SJ, Hawes EM, Moll S, Dager WE, Adcock DM. Heparin-calibrated chromogenic anti-Xa activity measurements in patients receiving rivaroxaban: can this test be used to quantify drug level? Ann Pharmacother 2015; 49(7):777–783. doi:10.1177/1060028015578451
  85. Levy JH, Ageno W, Chan NC, Crowther M, Verhamme P, Weitz JI; Subcommittee on Control of Anticoagulation. When and how to use antidotes for the reversal of direct oral anticoagulants: guidance from the SSC of the ISTH. J Thromb Haemost 2016; 14(3):623–627. doi:10.1111/jth.13227
  86. Cuker A, Siegal D. Monitoring and reversal of direct oral anticoagulants. Hematology Am Soc Hematol Educ Program 2015; 2015:117–124. doi:10.1182/asheducation-2015.1.117
References
  1. Abildgaard U. Highly purified antithrombin 3 with heparin cofactor activity prepared by disc electrophoresis. Scand J Clin Lab Invest 1968; 21(1):89–91. pmid:5637480
  2. Rosenberg RD, Lam L. Correlation between structure and function of heparin. Proc Natl Acad Sci USA 1979; 76(3):1218–1222. pmid:286307
  3. Lindahl U, Bäckström G, Höök M, Thunberg L, Fransson LA, Linker A. Structure of the antithrombin-binding site of heparin. Proc Natl Acad Sci USA 1979; 76(7):3198–3202. pmid:226960
  4. Rosenberg RD, Rosenberg JS. Natural anticoagulant mechanisms. J Clin Invest 1984; 74(1):1–6. doi:10.1172/JCI111389
  5. Casu B, Oreste P, Torri G, et al. The structure of heparin oligosaccharide fragments with high anti-(factor Xa) activity containing the minimal antithrombin III-binding sequence. Chemical and 13C nuclear-magnetic-resonance studies. Biochem J 1981; 197(3):599–609. pmid:7325974
  6. Choay J, Lormeau JC, Petitou M, Sinaÿ P, Fareed J. Structural studies on a biologically active hexasaccharide obtained from heparin. Ann NY Acad Sci 1981; 370: 644–649. pmid:6943974
  7. Hirsh J, Warkentin TE, Shaughnessy SG, et al. Heparin and low-molecular-weight heparin: mechanisms of action, pharmacokinetics, dosing, monitoring, efficacy, and safety. Chest 2001; 119(suppl 1):64S–94S. pmid:11157643
  8. Garcia DA, Baglin TP, Weitz JI, Samama MM. Parenteral anticoagulants: antithrombotic therapy and prevention of thrombosis, 9th ed: American College of Chest Physicians Evidence-Based Clinical Practice Guidelines. Chest 2012; 141(suppl 2):e24S–e43S. doi:10.1378/chest.11-2291
  9. Hirsh J, Levine M. Low-molecular weight heparin. Blood 1992; 79(1):1–17. pmid:1309422
  10. Barritt DW, Jordan SC. Anticoagulant drugs in the treatment of pulmonary embolism. A controlled trial. Lancet 1960; 1(7138):1309–1312. pmid:13797091
  11. Basu D, Gallus A, Hirsh J, Cade J. A prospective study of the value of monitoring heparin treatment with the activated partial thromboplastin time. N Engl J Med 1972; 287(7):324–327. doi:10.1056/NEJM197208172870703
  12. Hull RD, Raskob GE, Hirsh J, et al. Continuous intravenous heparin compared with intermittent subcutaneous heparin in the initial treatment of proximal-vein thrombosis. N Engl J Med 1986; 315(18):1109–1114. doi:10.1056/NEJM198610303151801
  13. Hull RD, Raskob GE, Brant RF, Pineo GF, Valentine KA. Relation between the time to achieve the lower limit of the APTT therapeutic range and recurrent venous thromboembolism during heparin treatment for deep vein thrombosis. Arch Intern Med 1997; 157(22):2562–2568. pmid:9531224
  14. Hull RD, Raskob GE, Brant RF, Pineo GF, Valentine KA. The importance of initial heparin treatment on long-term clinical outcomes of antithrombotic therapy. The emerging theme of delayed recurrence. Arch Intern Med 1997; 157(20):2317–2321. pmid:9361572
  15. Anand S, Ginsberg JS, Kearon C, Gent M, Hirsh J. The relation between the activated partial thromboplastin time response and recurrence in patients with venous thrombosis treated with continuous intravenous heparin. Arch Intern Med 1996; 156(15):1677–1681. pmid:8694666
  16. Anand SS, Bates S, Ginsberg JS, et al. Recurrent venous thrombosis and heparin therapy: an evaluation of the importance of early activated partial thromboplastin times. Arch Intern Med 1999; 159(17):2029–2032. pmid:10510988
  17. Raschke RA, Reilly BM, Guidry JR, Fontana JR, Srinivas S. The weight-based heparin dosing nomogram compared with a “standard care” nomogram. A randomized controlled trial. Ann Intern Med 1993; 119(9):874–881. pmid:8214998
  18. Smith SB, Geske JB, Maguire JM, Zane NA, Carter RE, Morgenthaler TI. Early anticoagulation is associated with reduced mortality for acute pulmonary embolism. Chest 2010; 137(6):1382–1390. doi:10.1378/chest.09-0959
  19. Cruickshank MK, Levine MN, Hirsh J, Roberts R, Siguenza M. A standard heparin nomogram for the management of heparin therapy. Arch Intern Med 1991; 151(2):333–337. pmid:1789820
  20. Raschke RA, Gollihare B, Peirce J. The effectiveness of implementing the weight-based heparin nomogram as a practice guideline. Arch Intern Med 1996; 156(15):1645–1649. pmid:8694662
  21. Simko RJ, Tsung FF, Stanek EJ. Activated clotting time versus activated partial thromboplastin time for therapeutic monitoring of heparin. Ann Pharmacother 1995; 29(10):1015–1021. doi:10.1177/106002809502901012
  22. Langdell RD, Wagner RH, Brinkhous KM. Effect of antihemophilic factor on one-stage clotting tests; a presumptive test for hemophilia and a simple one-stage antihemophilic factor assy procedure. J Lab Clin Med 1953; 41(4):637–647.
  23. White GC 2nd. The partial thromboplastin time: defining an era in coagulation. J Thromb Haemost 2003; 1(11):2267–2270. pmid:14629454
  24. Proctor RR, Rapaport SI. The partial thromboplastin time with kaolin. A simple screening test for first stage plasma clotting factor deficiencies. Am J Clin Pathol 1961; 36:212–219. pmid:13738153
  25. Brandt JT, Triplett DA, Rock WA, Bovill EG, Arkin CF. Effect of lupus anticoagulants on the activated partial thromboplastin time. Results of the College of American Pathologists survey program. Arch Pathol Lab Med 1991; 115(2):109–114. pmid:1899555
  26. Tripodi A, Mannucci PM. Activated partial thromboplastin time (APTT). New indications for an old test? J Thromb Haemost 2006; 4(4):750–751. doi:10.1111/j.1538-7836.2006.01857.x
  27. Chiu HM, Hirsh J, Yung WL, Regoeczi E, Gent M. Relationship between the anticoagulant and antithrombotic effects of heparin in experimental venous thrombosis. Blood 1977; 49(2):171–184. pmid:831872
  28. Brill-Edwards P, Ginsberg JS, Johnston M, Hirsh J. Establishing a therapeutic range for heparin therapy. Ann Intern Med 1993; 119(2):104–109. pmid:8512158
  29. Vandiver JW, Vondracek TG. Antifactor Xa levels versus activated partial thromboplastin time for monitoring unfractionated heparin. Pharmacotherapy 2012; 32(6):546–558. doi:10.1002/j.1875-9114.2011.01049.x
  30. Newall F. Anti-factor Xa (anti-Xa). In: Monagle P, ed. Haemostasis: Methods and Protocols. New York, NY: Springer-Verlag; 2013.
  31. Bates SM, Weitz JI, Johnston M, Hirsh J, Ginsberg JS. Use of a fixed activated partial thromboplastin time ratio to establish a therapeutic range for unfractionated heparin. Arch Intern Med 2001; 161(3):385–391. pmid:11176764
  32. Levine MN, Hirsh J, Gent M, et al. A randomized trial comparing activated thromboplastin time with heparin assay in patients with acute venous thromboembolism requiring large doses of heparin. Arch Intern Med 1994; 154(1):49–56. pmid:8267489
  33. Wool GD, Lu CM; Education Committee of the Academy of Clinical Laboratory Physicians and Scientists. Pathology consultation on anticoagulation monitoring: factor X-related assays. Am J Clin Pathol 2013; 140(5):623–634. doi:10.1309/AJCPR3JTOK7NKDBJ
  34. Lehman CM, Frank EL. Laboratory monitoring of heparin therapy: partial thromboplastin time or anti-Xa assay? Lab Med 2009; 40(1):47–51. doi:10.1309/LM9NJGW2ZIOLPHY6
  35. Arachchillage DR, Kamani F, Deplano S, Banya W, Laffan M. Should we abandon the aPTT for monitoring unfractionated heparin? Thromb Res 2017; 157:157–161. doi:10.1016/j.thromres.2017.07.006
  36. Olson JD, Arkin CA, Brandt JT, et al. College of American Pathologists Conference XXXI on Laboratory Monitoring of Anticoagulant Therapy: laboratory monitoring of unfractionated heparin therapy. Arch Pathol Lab Med 1998; 122(9):782–798. pmid:9740136
  37. Eikelboom JW, Hirsh J. Monitoring unfractionated heparin with the aPTT: time for a fresh look. Thromb Haemost 2006; 96(5):547–552. pmid:17080209
  38. Young E, Prins M, Levine MN, Hirsh J. Heparin binding to plasma proteins, an important mechanism of heparin resistance. Thromb Haemost 1992; 67(6):639–643. pmid:1509402
  39. Edson JR, Krivit W, White JG. Kaolin partial thromboplastin time: high levels of procoagulants producing short clotting times or masking deficiencies of other procoagulants or low concentrations of anticoagulants. J Lab Clin Med 1967; 70(3):463–470. pmid:6072020
  40. Whitfield LR, Lele AS, Levy G. Effect of pregnancy on the relationship between concentration and anticoagulant action of heparin. Clin Pharmacol Ther 1983; 34(1):23–28. pmid:6861435
  41. Marci CD, Prager D. A review of the clinical indications for the plasma heparin assay. Am J Clin Pathol 1993; 99(5):546–550.
  42. Takemoto CM, Streiff MB, Shermock KM, et al. Activated partial thromboplastin time and anti-Xa measurements in heparin monitoring: biochemical basis of discordance. Am J Clin Pathol 2013; 139(4):450–456. doi:10.1309/AJCPS6OW6DYNOGNH
  43. Adatya S, Uriel N, Yarmohammadi H, et al. Anti-factor Xa and activated partial thromboplastin time measurements for heparin monitoring in mechanical circulatory support. JACC Heart Fail 2015; 3(4):314–322. doi:10.1016/j.jchf.2014.11.009
  44. Kuhle S, Eulmesekian P, Kavanagh B, et al. Lack of correlation between heparin dose and standard clinical monitoring tests in treatment with unfractionated heparin in critically ill children. Haematologica 2007; 92(4):554–557. pmid:17488668
  45. Price EA, Jin J, Nguyen HM, Krishnan G, Bowen R, Zehnder JL. Discordant aPTT and anti-Xa values and outcomes in hospitalized patients treated with intravenous unfractionated heparin. Ann Pharmacother 2013; 47(2):151–158. doi:10.1345/aph.1R635
  46. Baker BA, Adelman MD, Smith PA, Osborn JC. Inability of the activated partial thromboplastin time to predict heparin levels. Arch Intern Med 1997; 157(21):2475–2479. pmid:9385299
  47. Koerber JM, Smythe MA, Begle RL, Mattson JC, Kershaw BP, Westley SJ. Correlation of activated clotting time and activated partial thromboplastin time to plasma heparin concentration. Pharmacotherapy 1999; 19(8):922–931. pmid:10453963
  48. Smythe MA, Mattson JC, Koerber JM. The heparin anti-Xa therapeutic range: are we there yet? Chest 2002; 121(1):303–304. pmid:11796474
  49. Cuker A, Ptashkin B, Konkle A, et al. Interlaboratory agreement in the monitoring of unfractionated heparin using the anti-factor Xa-correlated activated partial thromboplastin time. J Thromb Haemost 2009; 7(1):80–86. doi:10.1111/j.1538-7836.2008.03224.x
  50. Taylor CT, Petros WP, Ortel TL. Two instruments to determine activated partial thromboplastin time: implications for heparin monitoring. Pharmacotherapy 1999; 19(4):383–387. pmid:10212007
  51. Guervil DJ, Rosenberg AF, Winterstein AG, Harris NS, Johns TE, Zumberg MS. Activated partial thromboplastin time versus antifactor Xa heparin assay in monitoring unfractionated heparin by continuous intravenous infusion. Ann Pharmacother 2011; 45(7–8):861–868. doi:10.1345/aph.1Q161
  52. Fruge KS, Lee YR. Comparison of unfractionated heparin protocols using antifactor Xa monitoring or activated partial thrombin time monitoring. Am J Health Syst Pharm 2015; 72(17 suppl 2):S90–S97. doi:10.2146/sp150016
  53. Rosborough TK. Monitoring unfractionated heparin therapy with antifactor Xa activity results in fewer monitoring tests and dosage changes than monitoring with activated partial thromboplastin time. Pharmacotherapy 1999; 19(6):760–766. pmid:10391423
  54. Rosborough TK, Shepherd MF. Achieving target antifactor Xa activity with a heparin protocol based on sex, age, height, and weight. Pharmacotherapy 2004; 24(6):713–719. doi:10.1592/phco.24.8.713.36067
  55. Smith ML, Wheeler KE. Weight-based heparin protocol using antifactor Xa monitoring. Am J Health Syst Pharm 2010; 67(5):371–374. doi:10.2146/ajhp090123
  56. Bartholomew JR, Kottke-Marchant K. Monitoring anticoagulation therapy in patients with the lupus anticoagulant. J Clin Rheumatol 1998; 4(6):307–312. pmid:19078327
  57. Wool GD, Lu CM; Education Committee of the Academy of Clinical Laboratory Physicians and Scientists. Pathology consultation on anticoagulation monitoring: factor X-related assays. Am J Clin Pathol 2013; 140(5):623–634. doi:10.1309/AJCPR3JTOK7NKDBJ
  58. Mehta TP, Smythe MA, Mattson JC. Strategies for managing heparin therapy in patients with antiphospholipid antibody syndrome. Pharmacotherapy 2011; 31(12):1221–1231. doi:10.1592/phco.31.12.1221
  59. Levine SP, Sorenson RR, Harris MA, Knieriem LK. The effect of platelet factor 4 (PF4) on assays of plasma heparin. Br J Haematol 1984; 57(4):585–596. pmid:6743573
  60. Fisher AR, Bailey CR, Shannon CN, Wielogorski AK. Heparin resistance after aprotinin. Lancet 1992; 340(8829):1230–1231. pmid:1279335
  61. Becker RC, Corrao JM, Bovill EG, et al. Intravenous nitroglycerin-induced heparin resistance: a qualitative antithrombin III abnormality. Am Heart J 1990; 119(6):1254–1261. pmid:2112878
  62. Monagle P, Chan AK, Goldenberg NA, et al. Antithrombotic therapy in neonates and children: Antithrombotic Therapy and Prevention of Thrombosis, 9th ed: American College of Chest Physicians Evidence-Based Clinical Practice Guidelines. Chest 2012; 141(suppl 2):e737S–e801S. doi:10.1378/chest.11-2308
  63. Long E, Pitfield AF, Kissoon N. Anticoagulation therapy: indications, monitoring, and complications. Pediatr Emerg Care 2011; 27(1):55–61. doi:10.1097/PEC.0b013e31820461b1
  64. Andrew M, Schmidt B. Use of heparin in newborn infants. Semin Thromb Hemost 1988; 14(1):28–32. doi:10.1055/s-2007-1002752
  65. Teien AN, Lie M, Abildgaard U. Assay of heparin in plasma using a chromogenic substrate for activated factor X. Thromb Res 1976; 8(3):413–416. pmid:1265712
  66. Vera-Aguillera J, Yousef H, Beltran-Melgarejo D, et al. Clinical scenarios for discordant anti-Xa. Adv Hematol 2016; 2016:4054806. doi:10.1155/2016/4054806
  67. Macedo KA, Tatarian P, Eugenio KR. Influence of direct oral anticoagulants on anti-factor Xa measurements utilized for monitoring heparin. Ann Pharmacother 2018; 52(2):154–159. doi:10.1177/1060028017729481
  68. Wendte J, Voss G, Van Overschelde B. Influence of apixaban on antifactor Xa levels in a patient with acute kidney injury. Am J Health Syst Pharm 2016; 73(8):563–567. doi:10.2146/ajhp150360
  69. Faust AC, Kanyer D, Wittkowsky AK. Managing transitions from oral factor Xa inhibitors to unfractionated heparin infusions. Am J Health Syst Pharm 2016; 73(24):2037–2041. doi:10.2146/ajhp150596
  70. Alhenc-Gelas M, Jestin-Le Guernic C, Vitoux JF, Kher A, Aiach M, Fiessinger JN. Adjusted versus fixed doses of the low-molecular-weight heparin fragmin in the treatment of deep vein thrombosis. Fragmin-Study Group. Thromb Haemost 1994; 71(6):698–702. pmid:7974334
  71. Bates SM, Greer IA, Middeldorp S, Veenstra DL, Prabulos AM, Vandvik PO. VTE, thrombophilia, antithrombotic therapy, and pregnancy: Antithrombotic Therapy and Prevention of Thrombosis, 9th ed: American College of Chest Physicians Evidence-Based Clinical Practice Guidelines. Chest 2012; 141(suppl 2):e691S–e736S. doi:10.1378/chest.11-2300
  72. Bara L, Leizorovicz A, Picolet H, Samama M. Correlation between anti-Xa and occurrence of thrombosis and haemorrhage in post-surgical patients treated with either Logiparin (LMWH) or unfractionated heparin. Post-surgery Logiparin Study Group. Thromb Res 1992; 65(4–5):641–650. pmid:1319619
  73. Prandoni P, Lensing AW, Büller HR, et al. Comparison of subcutaneous low-molecular-weight heparin with intravenous standard heparin in proximal deep-vein thrombosis. Lancet 1992; 339(8791):441–445. pmid:1346817
  74. Walenga JM, Hoppensteadt D, Fareed J. Laboratory monitoring of the clinical effects of low molecular weight heparins. Thromb Res Suppl 1991;14:49–62. pmid:1658970
  75. Elkayam U. Anticoagulation therapy for pregnant women with mechanical prosthetic heart valves: how to improve safety? J Am Coll Cardiol 2017; 69(22):2692–2695. doi:10.1016/j.jacc.2017.04.034
  76. Brophy DF, Wazny LD, Gehr TW, Comstock TJ, Venitz J. The pharmacokinetics of subcutaneous enoxaparin in end-stage renal disease. Pharmacotherapy 2001; 21(2):169–174. pmid:11213853
  77. Becker RC, Spencer FA, Gibson M, et al; TIMI 11A Investigators. Influence of patient characteristics and renal function on factor Xa inhibition pharmacokinetics and pharmacodynamics after enoxaparin administration in non-ST-segment elevation acute coronary syndromes. Am Heart J 2002; 143(5):753–759. pmid:12040334
  78. Chow SL, Zammit K, West K, Dannenhoffer M, Lopez-Candales A. Correlation of antifactor Xa concentrations with renal function in patients on enoxaparin. J Clin Pharmacol 2003; 43(6):586–590. pmid:12817521
  79. Lim W, Dentali F, Eikelboom JW, Crowther MA. Meta-analysis: low-molecular-weight heparin and bleeding in patients with severe renal insufficiency. Ann Intern Med 2006; 144(9):673–684. pmid:16670137
  80. Spinler SA, Inverso SM, Cohen M, Goodman SG, Stringer KA, Antman EM; ESSENCE and TIMI 11B Investigators. Safety and efficacy of unfractionated heparin versus enoxaparin in patients who are obese and patients with severe renal impairment: analysis from the ESSENCE and TIMI 11B studies. Am Heart J 2003; 146(1):33–41. doi:10.1016/S0002-8703(03)00121-2
  81. Cestac P, Bagheri H, Lapeyre-Mestre M, et al. Utilisation and safety of low molecular weight heparins: prospective observational study in medical inpatients. Drug Saf 2003; 26(3):197–207. doi:10.2165/00002018-200326030-00005
  82. Douxfils J, Ageno W, Samama CM, et al. Laboratory testing in patients treated with direct oral anticoagulants: a practical guide for clinicians. J Thromb Haemost 2018; 16(2):209–219. doi:10.1111/jth.13912
  83. Samuelson BT, Cuker A, Siegal DM, Crowther M, Garcia DA. Laboratory assessment of the anticoagulant activity of direct oral anticoagulants: a systematic review. Chest 2017; 151(1):127–138. doi:10.1016/j.chest.2016.08.1462
  84. Gosselin RC, Francart SJ, Hawes EM, Moll S, Dager WE, Adcock DM. Heparin-calibrated chromogenic anti-Xa activity measurements in patients receiving rivaroxaban: can this test be used to quantify drug level? Ann Pharmacother 2015; 49(7):777–783. doi:10.1177/1060028015578451
  85. Levy JH, Ageno W, Chan NC, Crowther M, Verhamme P, Weitz JI; Subcommittee on Control of Anticoagulation. When and how to use antidotes for the reversal of direct oral anticoagulants: guidance from the SSC of the ISTH. J Thromb Haemost 2016; 14(3):623–627. doi:10.1111/jth.13227
  86. Cuker A, Siegal D. Monitoring and reversal of direct oral anticoagulants. Hematology Am Soc Hematol Educ Program 2015; 2015:117–124. doi:10.1182/asheducation-2015.1.117
Issue
Cleveland Clinic Journal of Medicine - 86(6)
Issue
Cleveland Clinic Journal of Medicine - 86(6)
Page Number
417-425
Page Number
417-425
Publications
Publications
Topics
Article Type
Display Headline
Anti-Xa assays: What is their role today in antithrombotic therapy?
Display Headline
Anti-Xa assays: What is their role today in antithrombotic therapy?
Legacy Keywords
Activated factor X, factor Xa, anti-factor Xa assays, anti-Xa assays, heparin, activated partial thromboplastin time, aPTT, anticoagulation, monitoring, antithrombotic therapy, venous thromboembolism, VTE, pulmonary embolism, PE, deep vein thrombosis, DVT, Erika hutt Centeno, Michael militello, marcelo gomes
Legacy Keywords
Activated factor X, factor Xa, anti-factor Xa assays, anti-Xa assays, heparin, activated partial thromboplastin time, aPTT, anticoagulation, monitoring, antithrombotic therapy, venous thromboembolism, VTE, pulmonary embolism, PE, deep vein thrombosis, DVT, Erika hutt Centeno, Michael militello, marcelo gomes
Sections
Inside the Article

KEY POINTS

  • Intravenous unfractionated heparin treatment is typically monitored by the activated partial thromboplastin time (aPTT), with a therapeutic target defined as the range that corresponds to an anti-Xa level of 0.3 to 0.7 U/mL.
  • Monitoring unfractionated heparin is important to achieve a therapeutic target within the first 24 hours and to maintain therapeutic levels thereafter.
  • The heparin anti-Xa assay is unreliable for unfractionated heparin monitoring when switching from oral factor Xa inhibitor therapy to intravenous unfractionated heparin. In such cases, the aPTT is preferred.
  • Most patients receiving low-molecular-weight heparin do not need monitoring, but monitoring should be considered for pregnant women with prosthetic heart valves, using an anti-Xa assay specific for low-molecular-weight heparin.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Tue, 05/28/2019 - 12:00
Un-Gate On Date
Tue, 05/28/2019 - 12:00
Use ProPublica
CFC Schedule Remove Status
Tue, 05/28/2019 - 12:00
Hide sidebar & use full width
render the right sidebar.
Article PDF Media

An obese 48-year-old man with progressive fatigue and decreased libido

Article Type
Changed
Wed, 05/01/2019 - 08:04
Display Headline
An obese 48-year-old man with progressive fatigue and decreased libido

A 48-year-old man presents to his primary care physician because of progressively decreasing energy and gradual decline in both libido and erectile function for the past 18 months. He has noticed decreased morning erections as well. He rates his libido at 3 to 4 on a scale of 10 for the past 6 months. He also reports poor motivation, depressed mood, impaired concentration, and sleep disturbances. He reports no hair loss, headache, or dizziness, and no decrease in shaving frequency. Review of his systems is otherwise unremarkable.

He has had dyslipidemia for 3 years and is not known to have hypertension or diabetes. His medications include atorvastatin, vitamin E, and multivitamins.

He is married with 3 children and does not wish to have more. He works as a software engineer and leads a sedentary lifestyle. He is a nonsmoker and occasionally drinks alcohol on the weekends.

On physical examination, he is alert and oriented and appears well. His height is 5 feet 10 inches (178 cm), weight 230 lb (104 kg), and body mass index (BMI) 32.8 kg/m2. His blood pressure is 115/83 mm Hg and pulse rate is 82 beats per minute and regular. Findings on cardiovascular and pulmonary examination are normal. He has large fatty breasts but without palpable glandular tissue.

farkouh_progressivefatigue_t1.jpg
Abdominal examination reveals central obesity—waist circumference 48 inches (122 cm)—without tenderness or organomegaly. There are no striae.

Genitourinary examination reveals normal hair distribution, a normal-sized penis, and slightly soft testes with testicular volume of 18–20 mL bilaterally.

His primary care physician suspects that he has low testosterone and orders some basic laboratory tests; the results are normal except for a low total testosterone level (Table 1).

FURTHER TESTING

1. Which of the following tests should his physician order next?

  • Repeat total testosterone measurement
  • Free testosterone measurement by commercial assay
  • Calculated free testosterone
  • Bioavailable testosterone measurement
  • Serum inhibin B measurement

This patient presents with several nonspecific symptoms. But collectively they suggest testosterone deficiency (hypogonadism).

farkouh_progressivefatigue_t2.jpg
Symptoms and signs of low testosterone vary according to age of onset. Prepubertal onset is associated with incomplete or delayed puberty, no development of secondary sexual characteristics, eunuchoid features, and small penis and testes. Postpubertal onset is associated with a wide array of symptoms (Table 2). Most manifestations of low testosterone are nonspecific, such as fatigue, impaired concentration, and sleep disturbance.1

Together, erectile dysfunction, low libido, and decreased morning erections strongly suggest hypogonadism.2 Loss of body hair and decreased shaving frequency are specific symptoms of hypogonadism; however, they require years to develop.3 Gynecomastia can also occur due to loss of the inhibitory action of testosterone on breast growth and a relative increase in estradiol. This occurs more in primary hypogonadism, due to the increase in luteinizing hormone (LH), which stimulates the remaining Leydig cells to secrete estradiol rather than testosterone.4

farkouh_progressivefatigue_t3.jpg
Screening for hypogonadism in men may be warranted in several conditions, even without clinical manifestations of low testosterone (Table 3).5–10

To diagnose hypogonadism in men and to start treatment for it, current guidelines recommend that the patient should have clinical features as well as laboratory evidence of low testosterone.5,6

Measuring testosterone: Total, free, bound, and bioavailable

Testosterone, a steroid hormone, circulates in the serum either as free testosterone or bound to several plasma proteins, mainly sex-hormone binding globulin (SHBG) and albumin.

Total testosterone includes both the free and bound fractions, whereas bioavailable testosterone includes both free and the portion bound to albumin, which has low affinity and can dissociate and be used at the tissue level.11

Low levels of total testosterone do not necessarily reflect a hypogonadal state, as a man with altered SHBG levels or binding capabilities can have low total but normal free testosterone levels and no manifestations.12 Several conditions can alter the levels of SHBG, including obesity, diabetes, aging, thyroid dysfunction, and others.5,13

Because our patient is obese, his total testosterone level is not a reliable indicator of hypogonadism, and repeating its measurement will not add diagnostic value.

Therefore, an alternative measurement should be used to accurately reflect the testosterone levels. From a physiologic point of view, bioavailable testosterone is the active form of testosterone and is the most accurate to be measured in a patient with hypogonadism. Nevertheless, because of technical difficulties in its measurement and lack of evidence correlating bioavailable testosterone with the clinical picture of hypogonadism, it is recommended that the level of free testosterone be used.5

The gold standard for direct measurement of serum free testosterone is equilibrium dialysis, but this is expensive and time-consuming.14 Commercial assays for free testosterone exist but have been deemed unreliable.14,15 It is recommended that free testosterone be measured by equilibrium dialysis or calculated using equations based on total testosterone, SHBG, and albumin levels.5 These equations are reliable and give results very close to the values obtained by equilibrium dialysis.15 Therefore, in our patient, it would be suitable to calculate the free testosterone level next.

Serum levels of free testosterone vary according to several factors. Diurnal variation of testosterone has been established: levels are highest in the morning and decline throughout the day.16 Food decreases testosterone levels.17 In addition, there is considerable day-to-day variation.18 Therefore, at least 2 readings of fasting morning testosterone on 2 separate days are recommended for the diagnosis of hypogonadism.5

Inhibin B is a hormone produced by Sertoli cells in the testes in response to follicle-stimulating hormone (FSH) stimulation. In turn, it acts as negative feedback, together with testosterone, to inhibit FSH release from the pituitary. Inhibin B has been shown to reflect spermatogenesis in the testes and therefore fertility.19 Inhibin B levels were found to be low in patients with central hypogonadism, due to less FSH release; however, they did not correlate with testosterone levels.20

 

 

CASE RESUMED: CHARACTERIZING HIS HYPOGONADISM

The patient’s physician orders morning fasting total testosterone, SHBG, and albumin testing and calculates the free testosterone level, which yields a value of 3 ng/dL (reference range 4.5–17). This is confirmed by a repeat measurement, which yields a value of 2.9 ng/dL. Laboratory test results combined with his clinical presentation are consistent with hypogonadism.

2. What is the most appropriate next step?

  • Measurement of serum LH and FSH
  • Measurement of serum prolactin
  • Scrotal ultrasonography
  • Gonadotropin-releasing hormone (GnRH) stimulation test
  • Semen analysis

After hypogonadism is diagnosed, it is important to distinguish if it is primary or central. This is achieved by measuring serum LH and FSH.5 All biotin supplements should be stopped at least 72 hours before measuring LH and FSH, as biotin can interfere with the assays, yielding false values.21

Secretion of FSH and LH from the anterior pituitary is under the influence of pulsatile release of GnRH from the hypothalamus. LH acts on Leydig cells in the testes to produce testosterone, whereas FSH acts on Sertoli cells, together with testosterone, to bring about spermatogenesis in the seminiferous tubules. Testosterone acts centrally as negative feedback to decrease the release of LH and FSH.

Primary hypogonadism occurs due to testicular failure, ie, the testes themselves fail to produce testosterone, leading to hypogonadism. The decrease in testosterone levels, together with inhibin B if Sertoli cells are damaged, lead to loss of negative feedback on the hypothalamus and pituitary, and therefore increased levels of LH and FSH. This is termed hypergonadotropic hypogonadism. Testicular failure may also result in impaired spermatogenesis and infertility due to destruction of testicular structures, in which case fertility cannot be restored.

Central hypogonadism occurs when the pituitary fails to produce LH and FSH (secondary hypogonadism) or when the hypothalamus fails to produce GnRH and subsequently the lack of secretion of LH and FSH from the pituitary (tertiary hypogonadism). The lack of LH will result in no stimulation of Leydig cells to produce testosterone, and therefore its deficiency. Serum hormone levels in central hypogonadism will reveal low testosterone, with either low or inappropriately normal gonadotropins (LH and FSH). This is termed hypogonadotropic hypogonadism. The lack of FSH, together with testosterone deficiency will also result in decreased spermatogenesis and therefore infertility. Testicular structures are preserved, however, and fertility can be restored with appropriate therapy, as discussed below.

Prolactin should be measured only if the patient has central hypogonadism. Its measurement is not warranted at this point in the patient’s workup. The implications of prolactin and its relationship to hypogonadism will be discussed later.

Although, this stepwise approach is not convenient for many patients, some physicians follow it because it is cost-effective, especially in those who are not insured. However, other physicians order FSH, LH, and sometimes prolactin with the confirmatory low testosterone measurement. Laboratories can also be instructed to wait to measure the pituitary hormones and to do so only if low testosterone is confirmed.

Varicocele, a possible cause of male infertility, can also impair Leydig cell function and cause low testosterone. In fact, surgical repair of varicocele has been demonstrated to increase serum testosterone.22 Scrotal ultrasonography is used to diagnose varicocele, but this also should be ordered at a later stage in the workup if primary hypogonadism is diagnosed.

The GnRH stimulation test is important for the diagnosis and evaluation of precocious or delayed puberty in children. In boys with delayed puberty, a poorer response to GnRH stimulation indicates central hypogonadism rather than constitutional delay.23 It has no role in the evaluation of postpubertal or adult-onset hypogonadism.

Semen analysis is important to evaluate fertility if the patient is interested in further procreation.5 Low testosterone levels may result in impaired spermatogenesis and therefore infertility. On the other hand, treatment with exogenous testosterone will also result in infertility, by feedback inhibition of LH and FSH and therefore inhibition of spermatogenesis. If the patient wishes to preserve fertility, treatment options other than testosterone should be considered; examples include clomiphene citrate, human menopausal gonadotropin, and human chorionic gonadotropin.23,24

Our patient has no desire to expand his family; therefore, a semen analysis and attempts to preserve spermatogenesis are not indicated.

 

 

CASE RESUMED: SEARCHING FOR CAUSES

His physician orders testing of serum LH and FSH, yielding the following values:

  • LH 1.6 mIU/mL (reference range 1.8–12)
  • FSH 1.9 mIU/mL (reference range 1.5–12.5).

The diagnosis of central hypogonadism is established.

3. Which investigation is the least appropriate in the further evaluation of this patient?

  • farkouh_progressivefatigue_t4.jpg
    Serum free thyroxine (T4) and morning cortisol measurement
  • Serum prolactin measurement
  • Serum ferritin measurement
  • Pituitary magnetic resonance imaging (MRI)
  • Chromosomal karyotyping

The diagnosis of central hypogonadism warrants evaluation for possible causes. These are summarized in Table 4.

Serum free thyroxine and morning cortisol

Since this patient’s LH and FSH values are abnormal, it is important to evaluate the status of other anterior pituitary hormones. In patients with pituitary abnormalities, serum free T4 is a more reliable test for assessing thyroid function than thyroid-stimulating hormone (TSH), because of loss of the negative feedback of thyroid hormones on the diseased pituitary. In contrast, serum TSH is considered the best single thyroid test to assess primary thyroid dysfunction.

Other measurements include prolactin and morning cortisol (reflecting adrenocorticotropic hormone status).

Prolactin measurement

Prolactin measurement is important to evaluate for hyperprolactinemia, as this will lead to hypogonadism by inhibition of GnRH secretion.25 Different pathologic, pharmacologic, and physiologic conditions can result in hyperprolactinemia, including prolactinomas, other pituitary and hypothalamic lesions, primary hypothyroidism, and medications such as antipsychotics.25 Dopamine agonists are the mainstay treatment for hyperprolactinemia.

Ferritin measurement

Ferritin measurement is indicated to diagnose iron overload conditions such as hemochromatosis, which can result in primary hypogonadism via testicular damage or in secondary hypogonadism via pituitary damage.26

Pituitary MRI with contrast

Pituitary MRI with contrast is used to diagnose structural lesions of the pituitary or hypothalamus. This diagnostic modality is indicated for patients with pituitary dysfunction, including central hypogonadism, manifestations of a mass effect (headache, visual field defects), persistent hyperprolactinemia, and panhypopituitarism, among others. To improve the diagnostic yield of pituitary MRI, the Endocrine Society guidelines recommend it for men with serum total testosterone levels below 150 ng/dL.5 However, some clinicians have a lower threshold for ordering pituitary MRI for patients with central hypogonadism. Physician judgment and expertise should be exercised and the decision made on an individual basis.

Chromosomal karyotyping

Chromosomal karyotyping is not indicated in our patient. It is reserved for those with primary hypogonadism to diagnose Klinefelter syndrome, which has a karyotype of 47,XXY.

CASE RESUMED: MOSH SYNDROME

Our patient’s prolactin, free T4, morning cortisol, and ferritin levels are measured, yielding normal values. No abnormalities are seen on pituitary MRI. A clinical reevaluation is conducted, revealing no history of head trauma or head and neck radiation. The lack of an obvious cause in our patient’s clinical presentation and workup, together with his obesity (BMI 32.8 kg/m2) supports the diagnosis of obesity as the cause of his hypogonadism.

Obesity can be a cause of secondary hypogonadism, which has led to the term “MOSH” (male obesity-associated secondary hypogonadism) syndrome. In fact, a cross-sectional study has demonstrated that 40% of nondiabetic obese (BMI ≥ 30 kg/m2) men over age 45 have low serum free testosterone levels, compared with 26% for lean (BMI < 25 kg/m2) men.27 Moreover, obesity has been found to be a strong predictor of testosterone replacement therapy.28 Other studies have also found an inverse relationship between BMI and testosterone levels.29

Several mechanisms interact in the pathogenesis of MOSH syndrome. Adipose tissue possesses aromatase activity, which converts androgens into estrogens.30 Peripheral estrogen production can in turn exert feedback inhibition on pituitary gonadotropin secretion.31 In obese men, increased adipose tissue leads to increased aromatase activity and more estrogen, so more feedback inhibition on the pituitary and subsequently secondary hypogonadism. 


Leptin, a hormone produced by adipocytes, is also increased in obesity, and was found to be inversely correlated with serum testosterone.32 Studies have demonstrated that leptin has an inhibitory effect on the enzymatic pathway that synthesizes testosterone in Leydig cells.33

Proinflammatory cytokines have also been implicated, as central obesity is associated with an increase in these cytokines, which in turn act negatively on the hypothalamus and impair GnRH release leading to lower testosterone.34,35

Treating obesity-related hypogonadism

In a pilot study,36 lifestyle attempts to reduce obesity were shown to improve hormonal levels. Bariatric surgery has also been demonstrated to be successful.37

Clomiphene citrate, a selective estrogen receptor modulator, increases endogenous testosterone secretion by inhibiting the negative feedback of estrogen on the hypothalamus and pituitary and thus increasing LH and FSH. It also preserves endogenous testosterone production, since it does not suppress the hypothalamic-pituitary-testicular axis.38 This made clomiphene citrate a potential treatment for men with central hypogonadism including those with MOSH.39

Nevertheless, there are no randomized trials to prove its safety and efficacy in the management of central hypogonadism.5 Regarding its use in men wishing to preserve fertility, most studies did not show improvement. However, a meta-analysis demonstrated statistically significant increased pregnancy rates in partners of men with idiopathic infertility if the men used 50 mg of clomiphene citrate daily.40

Testosterone deficiency can be a marker of metabolic syndrome, which needs to be managed more urgently than hypogonadism. A cross-sectional study found not only an association between metabolic syndrome and low serum testosterone, but also with each individual component of metabolic syndrome on its own, all of which need to be addressed.10

 

 

CASE CONTINUED: BEGINNING TREATMENT

The physician counsels the patient regarding the implications, potential adverse outcomes, and available treatments for his obesity, including lifestyle modification and bariatric surgery. The patient declines surgery and wishes to adopt a weight-reducing diet and exercise program, for which he is referred to a dietitian.

In addition, in view of the patient’s clinically and biochemically proven hypogonadism, his physician offers testosterone replacement therapy. He orders a serum prostate-specific antigen (PSA) level, which is 1.3 ng/dL (reference range < 4 ng/dL). The patient is prescribed 5 g of 1% testosterone gel daily.

TESTOSTERONE REPLACEMENT THERAPY

4. Which is the most common adverse effect of testosterone replacement therapy?

  • Cardiovascular events
  • Erythrocytosis
  • Prostate cancer
  • Infertility
  • Obstructive sleep apnea

farkouh_progressivefatigue_t5.jpg
Testosterone is indicated for men with an established diagnosis of hypogonadism. The benefits of testosterone replacement are summarized in Table 5.5,6

Clinicians should be very cautious in initiating testosterone replacement therapy in any patient with an unstable medical condition.

There are several formulations of testosterone replacement therapy, including intramuscular injections, transdermal gels or patches, buccal tablets, an intranasal gel, and oral tablets. Of note, there are 2 different forms of oral testosterone preparations: testosterone undecanoate and 17-alpha alkylated testosterone. The former is unavailable in the United States and the latter is not recommended for use due to its proven hepatic toxicity.41

Testosterone and erythrocytosis

Meta-analyses have concluded that the most frequent adverse event of testosterone replacement therapy is a significant rise in hematocrit.42 This rise was found to be dose-dependent and was more marked in older men.43 Although all preparations can cause erythrocytosis, parenteral forms have been observed to raise it the most, particularly short-term injectables.44,45

The mechanism behind this increase is attributed to increased erythropoietin levels and improved usage of iron for red blood cell synthesis.46 In fact, testosterone replacement therapy has been shown to improve hemoglobin levels in patients with anemia.47 On the other hand, increasing hematocrit levels may lead to thrombotic and vasoocclusive events.44

farkouh_progressivefatigue_f1.jpg
%3Cp%3EFigure%201.%20Hematocrit%20monitoring%20for%20patients%20on%20testosterone%20replacement%20therapy.%3C%2Fp%3E
It is strongly recommended that baseline hematocrit levels be measured before initiating testosterone replacement therapy.5,6 The hematocrit level should also be monitored 3 to 6 months into treatment and yearly thereafter while on testosterone.5Figure 1 summarizes the appropriate steps to undertake regarding hematocrit levels, according to the American Urological Association.6

Testosterone and prostate cancer

The relationship between testosterone treatment and prostate cancer has long been studied. Historically, testosterone replacement therapy was believed to increase the risk of prostate cancer; however, recent studies and meta-analyses have shown that this is not the case.42,48 Nevertheless, clinical guidelines still recommend prostate monitoring for men on testosterone replacement therapy.5,6

farkouh_progressivefatigue_t6.jpg
Furthermore, the clinician should make sure the patient does not have prostate cancer before initiating testosterone replacement therapy. Since there is a significant incidence of prostate cancer in men with serum PSA of 2.5–4.0 ng/mL, a patient with hypogonadism and a serum PSA in that range or higher should have appropriate evaluation before initiating testosterone replacement therapy.49 The Endocrine Society recommendations for prostate monitoring are summarized in Table 6.5

Testosterone and cardiovascular risk

The evidence regarding this issue has been contradictory and inconsistent. Meta-analyses have demonstrated that low testosterone is associated with higher risk of major adverse cardiovascular events.50 These studies argue for the use of testosterone replacement therapy in hypogonadal men to decrease the risk. However, other studies and meta-analyses have found that testosterone replacement therapy is associated with increased cardiovascular risk and have concluded that major adverse cardiac events are in fact a risk of testosterone replacement therapy.51

Current recommendations advocate against the use of testosterone replacement therapy in men with uncontrolled heart failure or with cardiovascular events in the past 3 to 6 months.5,6 Cardiovascular risk factors should be addressed and corrected, and patients should be educated on cardiovascular symptoms and the need to report them if they occur.

Testosterone and infertility

As described earlier, testosterone replacement therapy increases negative feedback on the pituitary and decreases LH and FSH production, leading to less spermatogenesis. Other treatment options should be sought for hypogonadal men wishing to preserve fertility.

Other adverse effects

Other adverse effects of testosterone replacement therapy include acne, oily skin, obstructive sleep apnea, gynecomastia, and balding.

Given all the adverse events that can be associated with testosterone replacement therapy, the risks and benefits of treating hypogonadism in each patient should be taken into consideration, and an individualized approach is required.

 

 

CASE RESUMED: FOLLOW-UP

The patient presents 3 months later for follow-up. He reports significant improvement in his presenting symptoms including energy, libido, and erectile function. He also reports some improvement in his mood and concentration. He has lost 12 lb (5.4 kg) and is still trying to improve his diet and exercise program. He is compliant with his testosterone gel therapy.

His serum calculated free testosterone level is 7.8 ng/dL (4.5–17), and his hematocrit is 46%. The patient is instructed to continue his treatment and to return after 9 months for further follow-up.

TAKE-HOME POINTS

  • Men with hypogonadism usually present with nonspecific manifestations, so clinicians should keep a high index of suspicion.
  • Both clinical and biochemical evidence of hypogonadism should be present to diagnose and start treatment for it.
  • Low levels of serum total testosterone do not necessarily reflect hypogonadism.
  • The hormonal profile of central hypogonadism reveals low serum testosterone with low or inappropriately normal serum LH and FSH levels.

Obesity can cause central hypogonadism and should be suspected after pituitary and other systemic causes are excluded.

References
  1. Araujo AB, Esche GR, Kupelian V, et al. Prevalence of symptomatic androgen deficiency in men. J Clin Endocrinol Metab 2007; 92(11):4241–4247. doi:10.1210/jc.2007-1245
  2. Wu FCW, Tajar A, Beynon JM, et al; EMAS Group. Identification of late-onset hypogonadism in middle-aged and elderly men. N Engl J Med 2010; 363(2):123–135. doi:10.1056/NEJMoa0911101
  3. Arver S, Lehtihet M. Current guidelines for the diagnosis of testosterone deficiency. Front Horm Res 2009; 37:5–20. doi:10.1159/000175839
  4. Narula HS, Carlson HE. Gynaecomastia—pathophysiology, diagnosis and treatment. Nat Rev Endocrinol 2014; 10(11):684–698. doi:10.1038/nrendo.2014.139
  5. Bhasin S, Brito JP, Cunningham GR, et al. Testosterone therapy in men with hypogonadism: an Endocrine Society clinical practice guideline. J Clin Endocrinol Metab 2018; 103(5):1715–1744. doi:10.1210/jc.2018-00229
  6. Mulhall JP, Trost LW, Brannigan RE, et al. Evaluation and management of testosterone deficiency: AUA guideline. J Urol 2018; 200(2):423–432. doi:10.1016/j.juro.2018.03.115
  7. Balasubramanian V, Naing S. Hypogonadism in chronic obstructive pulmonary disease: incidence and effects. Curr Opin Pulm Med 2012; 18(2):112–117. doi:10.1097/MCP.0b013e32834feb37
  8. Atlantis E, Fahey P, Cochrane B, Wittert G, Smith S. Endogenous testosterone level and testosterone supplementation therapy in chronic obstructive pulmonary disease (COPD): a systematic review and meta-analysis. BMJ Open 2013; 3(8)pii:e003127. doi:10.1136/bmjopen-2013-003127
  9. Bawor M, Bami H, Dennis BB, et al. Testosterone suppression in opioid users: a systematic review and meta-analysis. Drug Alcohol Depend 2015; 149:1–9. doi:10.1016/j.drugalcdep.2015.01.038
  10. Tan WS, Ng CJ, Khoo EM, Low WY, Tan HM. The triad of erectile dysfunction, testosterone deficiency syndrome and metabolic syndrome: findings from a multi-ethnic Asian men study (The Subang Men's Health Study). Aging Male 2011; 14(4):231–236. doi:10.3109/13685538.2011.597463
  11. Goldman AL, Bhasin S, Wu FCW, Krishna M, Matsumoto AM, Jasuja R. A reappraisal of testosterone’s binding in circulation: physiological and clinical implications. Endocr Rev 2017; 38(4):302–324. doi:10.1210/er.2017-00025
  12. Antonio L, Wu FC, O’Neill TW, et al; European Male Ageing Study Study Group. Low free testosterone is associated with hypogonadal signs and symptoms in men with normal total testosterone. J Clin Endocrinol Metab 2016; 101(7):2647–2657. doi:10.1210/jc.2015-4106
  13. Liu F, Shen X, Wang R, et al. Association of central obesity with sex hormone binding globulin: a cross-sectional study of 1166 Chinese men. Open Med (Wars) 2018; 13:196–202. doi:10.1515/med-2018-0030
  14. Vermeulen A, Verdonck L, Kaufman JM. A critical evaluation of simple methods for the estimation of free testosterone in serum. J Clin Endocrinol Metab 1999; 84(10):3666–3672. doi:10.1210/jcem.84.10.6079
  15. Halmenschlager G, Rhoden EL, Riedner CE. Calculated free testosterone and radioimmunoassay free testosterone as a predictor of subnormal levels of total testosterone. Int Urol Nephrol 2012; 44(3):673–681. doi:10.1007/s11255-011-0066-z
  16. Brambilla DJ, Matsumoto AM, Araujo AB, McKinlay JB. The effect of diurnal variation on clinical measurement of serum testosterone and other sex hormone levels in men. J Clin Endocrinol Metab 2009; 94(3):907–913. doi:10.1210/jc.2008-1902
  17. Lehtihet M, Arver S, Bartuseviciene I, Pousette Å. S-testosterone decrease after a mixed meal in healthy men independent of SHBG and gonadotrophin levels. Andrologia 2012; 44(6):405–410. doi:10.1111/j.1439-0272.2012.01296.x
  18. Brambilla DJ, O’Donnell AB, Matsumoto AM, McKinlay JB. Intraindividual variation in levels of serum testosterone and other reproductive and adrenal hormones in men. Clin Endocrinol (Oxf) 2007; 67(6):853–862. doi:10.1111/j.1365-2265.2007.02976.x
  19. Manzoor SM, Sattar A, Hashim R, et al. Serum inhibin B as a diagnostic marker of male infertility. J Ayub Med Coll Abbottabad 2012; 24(3–4):113–116. pmid:24669628
  20. Kolb BA, Stanczyk FZ, Sokol RZ. Serum inhibin B levels in males with gonadal dysfunction. Fertil Steril 2000; 74(2):234–238. pmid:10927037
  21. Trambas CM, Sikaris KA, Lu ZX. More on biotin treatment mimicking Graves’ disease. N Engl J Med 2016; 375(17):1698. doi:10.1056/NEJMc1611875
  22. Li F, Yue H, Yamaguchi K, et al. Effect of surgical repair on testosterone production in infertile men with varicocele: a meta-analysis. Int J Urol 2012; 19(2):149–154. doi:10.1111/j.1442-2042.2011.02890.x
  23. Crosnoe-Shipley LE, Elkelany OO, Rahnema CD, Kim ED. Treatment of hypogonadotropic male hypogonadism: case-based scenarios. World J Nephrol 2015; 4(2):245–253. doi:10.5527/wjn.v4.i2.245
  24. Majzoub A, Sabanegh E Jr. Testosterone replacement in the infertile man. Transl Androl Urol 2016; 5(6):859–865. doi:10.21037/tau.2016.08.03
  25. Majumdar A, Mangal NS. Hyperprolactinemia. J Hum Reprod Sci 2013; 6(3):168–175. doi:10.4103/0974-1208.121400
  26. El Osta R, Grandpre N, Monnin N, Hubert J, Koscinski I. Hypogonadotropic hypogonadism in men with hereditary hemochromatosis. Basic Clin Androl 2017; 27:13. doi:10.1186/s12610-017-0057-8
  27. Dhindsa S, Miller MG, McWhirter CL, et al. Testosterone concentrations in diabetic and nondiabetic obese men. Diabetes Care 2010; 33(6):1186–1192. doi:10.2337/dc09-1649
  28. Jasuja GK, Bhasin S, Reisman JI, et al. Who gets testosterone? Patient characteristics associated with testosterone prescribing in the Veteran Affairs system: a cross-sectional study. J Gen Intern Med 2017; 32(3):304–311. doi:10.1007/s11606-016-3940-7
  29. Kaplan SA, Lee JY, O’Neill EA, Meehan AG, Kusek JW. Prevalence of low testosterone and its relationship to body mass index in older men with lower urinary tract symptoms associated with benign prostatic hyperplasia. Aging Male 2013; 16(4):169–172. doi:10.3109/13685538.2013.844786
  30. Lee HK, Lee JK, Cho B. The role of androgen in the adipose tissue of males. World J Mens Health 2013; 31(2):136–140. doi:10.5534/wjmh.2013.31.2.136
  31. Raven G, De Jong FH, Kaufman JM, De Ronde W. In men, peripheral estradiol levels directly reflect the action of estrogens at the hypothalamo-pituitary level to inhibit gonadotropin secretion. J Clin Endocrinol Metab 2006; 91(9):3324–3328. doi:10.1210/jc.2006-0462
  32. Hofny ER, Ali ME, Abdel-Hafez HZ, et al. Semen parameters and hormonal profile in obese fertile and infertile males. Fertil Steril 2010; 94(2):581–584. doi:10.1016/j.fertnstert.2009.03.085
  33. Isidori AM, Caprio M, Strollo F, et al. Leptin and androgens in male obesity: evidence for leptin contribution to reduced androgen levels. J Clin Endocrinol Metab 1999; 84(10):3673–3680. doi:10.1210/jcem.84.10.6082
  34. El-Wakkad A, Hassan NM, Sibaii H, El-Zayat SR. Proinflammatory, anti-inflammatory cytokines and adiponkines in students with central obesity. Cytokine 2013; 61(2):682–687. doi:10.1016/j.cyto.2012.11.010
  35. Maggio M, Basaria S, Ceda GP, et al. The relationship between testosterone and molecular markers of inflammation in older men. J Endocrinol Invest 2005; 28(suppl proceedings 11):116–119. pmid:16760639
  36. de Lorenzo A, Noce A, Moriconi E, et al. MOSH syndrome (male obesity secondary hypogonadism): clinical assessment and possible therapeutic approaches. Nutrients 2018; 10(4)pii:E474. doi:10.3390/nu10040474
  37. Escobar-Morreale HF, Santacruz E, Luque-Ramírez M, Botella Carretero JI. Prevalence of ‘obesity-associated gonadal dysfunction’ in severely obese men and women and its resolution after bariatric surgery: a systematic review and meta-analysis. Hum Reprod Update 2017; 23(4):390–408. doi:10.1093/humupd/dmx012
  38. Lo EM, Rodriguez KM, Pastuszak AW, Khera M. Alternatives to testosterone therapy: a review. Sex Med Rev 2018; 6(1):106–113. doi:10.1016/j.sxmr.2017.09.004
  39. Soares AH, Horie NC, Chiang LAP, et al. Effects of clomiphene citrate on male obesity-associated hypogonadism: a randomized, double-blind, placebo-controlled study. Int J Obes (Lond) 2018; 42(5):953–963. doi:10.1038/s41366-018-0105-2
  40. Chua ME, Escusa KG, Luna S, Tapia LC, Dofitas B, Morales M. Revisiting oestrogen antagonists (clomiphene or tamoxifen) as medical empiric therapy for idiopathic male infertility: a meta-analysis. Andrology 2013; 1(5):749–757. doi:10.1111/j.2047-2927.2013.00107.x
  41. Westaby D, Ogle SJ, Paradinas FJ, Randell JB, Murray-Lyon IM. Liver damage from long-term methyltestosterone. Lancet 1977; 2(8032):262–263. pmid:69876
  42. Fernández-Balsells MM, Murad MH, Lane M, et al. Clinical review 1: Adverse effects of testosterone therapy in adult men: a systematic review and meta-analysis. J Clin Endocrinol Metab 2010; 95(6):2560–2575. doi:10.1210/jc.2009-2575
  43. Coviello AD, Kaplan B, Lakshman KM, Chen T, Singh AB, Bhasin S. Effects of graded doses of testosterone on erythropoiesis in healthy young and older men. J Clin Endocrinol Metab 2008; 93(3):914–919. doi:10.1210/jc.2007-1692
  44. Ohlander SJ, Varghese B, Pastuszak AW. Erythrocytosis following testosterone therapy. Sex Med Rev 2018; 6(1):77–85. doi:10.1016/j.sxmr.2017.04.001
  45. Jones SD Jr, Dukovac T, Sangkum P, Yafi FA, Hellstrom WJ. Erythrocytosis and polycythemia secondary to testosterone replacement therapy in the aging male. Sex Med Rev 2015; 3(2):101–112. doi:10.1002/smrj.43
  46. Bachman E, Travison TG, Basaria S, et al. Testosterone induces erythrocytosis via increased erythropoietin and suppressed hepcidin: evidence for a new erythropoietin/hemoglobin set point. J Gerontol A Biol Sci Med Sci 2014; 69(6):725–735. doi:10.1093/gerona/glt154
  47. Roy CN, Snyder PJ, Stephens-Shields AJ, et al. Association of testosterone levels with anemia in older men: a controlled clinical trial. JAMA Intern Med 2017; 177(4):480–490. doi:10.1001/jamainternmed.2016.9540
  48. Klap J, Schmid M, Loughlin KR. The relationship between total testosterone levels and prostate cancer: a review of the continuing controversy. J Urol 2015; 193(2):403–413. doi:10.1016/j.juro.2014.07.123
  49. Gilbert SM, Cavallo CB, Kahane H, Lowe FC. Evidence suggesting PSA cutpoint of 2.5 ng/mL for prompting prostate biopsy: review of 36,316 biopsies. Urology 2005; 65(3):549–553. doi:10.1016/j.urology.2004.10.064
  50. Araujo AB, Dixon JM, Suarez EA, Murad MH, Guey LT, Wittert GA. Clinical review: Endogenous testosterone and mortality in men: a systematic review and meta-analysis. J Clin Endocrinol Metab 2011; 96(10):3007–3019. doi:10.1210/jc.2011-1137
  51. Xu L, Freeman G, Cowling BJ, Schooling CM. Testosterone therapy and cardiovascular events among men: a systematic review and meta-analysis of placebo-controlled randomized trials. BMC Med 2013; 11:108. doi:10.1186/1741-7015-11-108
Article PDF
Author and Disclosure Information

Ala’a N. Farkouh
School of Medicine, The University of Jordan, Amman, Jordan

Ayman A. Zayed, MD, MSc, FACE, FACP
Professor of Medicine and Chief, Division of Endocrinology, Diabetes, and Metabolism, Department of Internal Medicine, School of Medicine, The University of Jordan, Jordan University Hospital, Amman, Jordan

Address: Ayman A. Zayed, MD, MSc, FACE, FACP, Department of Internal Medicine, Jordan University Hospital, The University of Jordan, Queen Rania Street, Amman, Jordan, 11942; baraaayman@gmail.com

Issue
Cleveland Clinic Journal of Medicine - 86(5)
Publications
Topics
Page Number
321-331
Legacy Keywords
male hypogonadism, testosterone, low T, obesity, decreased libido, erectile dysfunction, ED, sex hormone binding globulin, SHBG, luteinizing hormone, LH, follicle-stimulating hormone, FSH, inhibin B, pituitary, thyroxine, T4, prolactin, ferritin, laboratory testing, cortisol, MOSH syndrome, male obesity-associated secondary hypogonadism, prostate-specific antigen, PSA, prostate cancer, Ala’A Farkouh, Ayman Zayed
Sections
Author and Disclosure Information

Ala’a N. Farkouh
School of Medicine, The University of Jordan, Amman, Jordan

Ayman A. Zayed, MD, MSc, FACE, FACP
Professor of Medicine and Chief, Division of Endocrinology, Diabetes, and Metabolism, Department of Internal Medicine, School of Medicine, The University of Jordan, Jordan University Hospital, Amman, Jordan

Address: Ayman A. Zayed, MD, MSc, FACE, FACP, Department of Internal Medicine, Jordan University Hospital, The University of Jordan, Queen Rania Street, Amman, Jordan, 11942; baraaayman@gmail.com

Author and Disclosure Information

Ala’a N. Farkouh
School of Medicine, The University of Jordan, Amman, Jordan

Ayman A. Zayed, MD, MSc, FACE, FACP
Professor of Medicine and Chief, Division of Endocrinology, Diabetes, and Metabolism, Department of Internal Medicine, School of Medicine, The University of Jordan, Jordan University Hospital, Amman, Jordan

Address: Ayman A. Zayed, MD, MSc, FACE, FACP, Department of Internal Medicine, Jordan University Hospital, The University of Jordan, Queen Rania Street, Amman, Jordan, 11942; baraaayman@gmail.com

Article PDF
Article PDF
Related Articles

A 48-year-old man presents to his primary care physician because of progressively decreasing energy and gradual decline in both libido and erectile function for the past 18 months. He has noticed decreased morning erections as well. He rates his libido at 3 to 4 on a scale of 10 for the past 6 months. He also reports poor motivation, depressed mood, impaired concentration, and sleep disturbances. He reports no hair loss, headache, or dizziness, and no decrease in shaving frequency. Review of his systems is otherwise unremarkable.

He has had dyslipidemia for 3 years and is not known to have hypertension or diabetes. His medications include atorvastatin, vitamin E, and multivitamins.

He is married with 3 children and does not wish to have more. He works as a software engineer and leads a sedentary lifestyle. He is a nonsmoker and occasionally drinks alcohol on the weekends.

On physical examination, he is alert and oriented and appears well. His height is 5 feet 10 inches (178 cm), weight 230 lb (104 kg), and body mass index (BMI) 32.8 kg/m2. His blood pressure is 115/83 mm Hg and pulse rate is 82 beats per minute and regular. Findings on cardiovascular and pulmonary examination are normal. He has large fatty breasts but without palpable glandular tissue.

farkouh_progressivefatigue_t1.jpg
Abdominal examination reveals central obesity—waist circumference 48 inches (122 cm)—without tenderness or organomegaly. There are no striae.

Genitourinary examination reveals normal hair distribution, a normal-sized penis, and slightly soft testes with testicular volume of 18–20 mL bilaterally.

His primary care physician suspects that he has low testosterone and orders some basic laboratory tests; the results are normal except for a low total testosterone level (Table 1).

FURTHER TESTING

1. Which of the following tests should his physician order next?

  • Repeat total testosterone measurement
  • Free testosterone measurement by commercial assay
  • Calculated free testosterone
  • Bioavailable testosterone measurement
  • Serum inhibin B measurement

This patient presents with several nonspecific symptoms. But collectively they suggest testosterone deficiency (hypogonadism).

farkouh_progressivefatigue_t2.jpg
Symptoms and signs of low testosterone vary according to age of onset. Prepubertal onset is associated with incomplete or delayed puberty, no development of secondary sexual characteristics, eunuchoid features, and small penis and testes. Postpubertal onset is associated with a wide array of symptoms (Table 2). Most manifestations of low testosterone are nonspecific, such as fatigue, impaired concentration, and sleep disturbance.1

Together, erectile dysfunction, low libido, and decreased morning erections strongly suggest hypogonadism.2 Loss of body hair and decreased shaving frequency are specific symptoms of hypogonadism; however, they require years to develop.3 Gynecomastia can also occur due to loss of the inhibitory action of testosterone on breast growth and a relative increase in estradiol. This occurs more in primary hypogonadism, due to the increase in luteinizing hormone (LH), which stimulates the remaining Leydig cells to secrete estradiol rather than testosterone.4

farkouh_progressivefatigue_t3.jpg
Screening for hypogonadism in men may be warranted in several conditions, even without clinical manifestations of low testosterone (Table 3).5–10

To diagnose hypogonadism in men and to start treatment for it, current guidelines recommend that the patient should have clinical features as well as laboratory evidence of low testosterone.5,6

Measuring testosterone: Total, free, bound, and bioavailable

Testosterone, a steroid hormone, circulates in the serum either as free testosterone or bound to several plasma proteins, mainly sex-hormone binding globulin (SHBG) and albumin.

Total testosterone includes both the free and bound fractions, whereas bioavailable testosterone includes both free and the portion bound to albumin, which has low affinity and can dissociate and be used at the tissue level.11

Low levels of total testosterone do not necessarily reflect a hypogonadal state, as a man with altered SHBG levels or binding capabilities can have low total but normal free testosterone levels and no manifestations.12 Several conditions can alter the levels of SHBG, including obesity, diabetes, aging, thyroid dysfunction, and others.5,13

Because our patient is obese, his total testosterone level is not a reliable indicator of hypogonadism, and repeating its measurement will not add diagnostic value.

Therefore, an alternative measurement should be used to accurately reflect the testosterone levels. From a physiologic point of view, bioavailable testosterone is the active form of testosterone and is the most accurate to be measured in a patient with hypogonadism. Nevertheless, because of technical difficulties in its measurement and lack of evidence correlating bioavailable testosterone with the clinical picture of hypogonadism, it is recommended that the level of free testosterone be used.5

The gold standard for direct measurement of serum free testosterone is equilibrium dialysis, but this is expensive and time-consuming.14 Commercial assays for free testosterone exist but have been deemed unreliable.14,15 It is recommended that free testosterone be measured by equilibrium dialysis or calculated using equations based on total testosterone, SHBG, and albumin levels.5 These equations are reliable and give results very close to the values obtained by equilibrium dialysis.15 Therefore, in our patient, it would be suitable to calculate the free testosterone level next.

Serum levels of free testosterone vary according to several factors. Diurnal variation of testosterone has been established: levels are highest in the morning and decline throughout the day.16 Food decreases testosterone levels.17 In addition, there is considerable day-to-day variation.18 Therefore, at least 2 readings of fasting morning testosterone on 2 separate days are recommended for the diagnosis of hypogonadism.5

Inhibin B is a hormone produced by Sertoli cells in the testes in response to follicle-stimulating hormone (FSH) stimulation. In turn, it acts as negative feedback, together with testosterone, to inhibit FSH release from the pituitary. Inhibin B has been shown to reflect spermatogenesis in the testes and therefore fertility.19 Inhibin B levels were found to be low in patients with central hypogonadism, due to less FSH release; however, they did not correlate with testosterone levels.20

 

 

CASE RESUMED: CHARACTERIZING HIS HYPOGONADISM

The patient’s physician orders morning fasting total testosterone, SHBG, and albumin testing and calculates the free testosterone level, which yields a value of 3 ng/dL (reference range 4.5–17). This is confirmed by a repeat measurement, which yields a value of 2.9 ng/dL. Laboratory test results combined with his clinical presentation are consistent with hypogonadism.

2. What is the most appropriate next step?

  • Measurement of serum LH and FSH
  • Measurement of serum prolactin
  • Scrotal ultrasonography
  • Gonadotropin-releasing hormone (GnRH) stimulation test
  • Semen analysis

After hypogonadism is diagnosed, it is important to distinguish if it is primary or central. This is achieved by measuring serum LH and FSH.5 All biotin supplements should be stopped at least 72 hours before measuring LH and FSH, as biotin can interfere with the assays, yielding false values.21

Secretion of FSH and LH from the anterior pituitary is under the influence of pulsatile release of GnRH from the hypothalamus. LH acts on Leydig cells in the testes to produce testosterone, whereas FSH acts on Sertoli cells, together with testosterone, to bring about spermatogenesis in the seminiferous tubules. Testosterone acts centrally as negative feedback to decrease the release of LH and FSH.

Primary hypogonadism occurs due to testicular failure, ie, the testes themselves fail to produce testosterone, leading to hypogonadism. The decrease in testosterone levels, together with inhibin B if Sertoli cells are damaged, lead to loss of negative feedback on the hypothalamus and pituitary, and therefore increased levels of LH and FSH. This is termed hypergonadotropic hypogonadism. Testicular failure may also result in impaired spermatogenesis and infertility due to destruction of testicular structures, in which case fertility cannot be restored.

Central hypogonadism occurs when the pituitary fails to produce LH and FSH (secondary hypogonadism) or when the hypothalamus fails to produce GnRH and subsequently the lack of secretion of LH and FSH from the pituitary (tertiary hypogonadism). The lack of LH will result in no stimulation of Leydig cells to produce testosterone, and therefore its deficiency. Serum hormone levels in central hypogonadism will reveal low testosterone, with either low or inappropriately normal gonadotropins (LH and FSH). This is termed hypogonadotropic hypogonadism. The lack of FSH, together with testosterone deficiency will also result in decreased spermatogenesis and therefore infertility. Testicular structures are preserved, however, and fertility can be restored with appropriate therapy, as discussed below.

Prolactin should be measured only if the patient has central hypogonadism. Its measurement is not warranted at this point in the patient’s workup. The implications of prolactin and its relationship to hypogonadism will be discussed later.

Although, this stepwise approach is not convenient for many patients, some physicians follow it because it is cost-effective, especially in those who are not insured. However, other physicians order FSH, LH, and sometimes prolactin with the confirmatory low testosterone measurement. Laboratories can also be instructed to wait to measure the pituitary hormones and to do so only if low testosterone is confirmed.

Varicocele, a possible cause of male infertility, can also impair Leydig cell function and cause low testosterone. In fact, surgical repair of varicocele has been demonstrated to increase serum testosterone.22 Scrotal ultrasonography is used to diagnose varicocele, but this also should be ordered at a later stage in the workup if primary hypogonadism is diagnosed.

The GnRH stimulation test is important for the diagnosis and evaluation of precocious or delayed puberty in children. In boys with delayed puberty, a poorer response to GnRH stimulation indicates central hypogonadism rather than constitutional delay.23 It has no role in the evaluation of postpubertal or adult-onset hypogonadism.

Semen analysis is important to evaluate fertility if the patient is interested in further procreation.5 Low testosterone levels may result in impaired spermatogenesis and therefore infertility. On the other hand, treatment with exogenous testosterone will also result in infertility, by feedback inhibition of LH and FSH and therefore inhibition of spermatogenesis. If the patient wishes to preserve fertility, treatment options other than testosterone should be considered; examples include clomiphene citrate, human menopausal gonadotropin, and human chorionic gonadotropin.23,24

Our patient has no desire to expand his family; therefore, a semen analysis and attempts to preserve spermatogenesis are not indicated.

 

 

CASE RESUMED: SEARCHING FOR CAUSES

His physician orders testing of serum LH and FSH, yielding the following values:

  • LH 1.6 mIU/mL (reference range 1.8–12)
  • FSH 1.9 mIU/mL (reference range 1.5–12.5).

The diagnosis of central hypogonadism is established.

3. Which investigation is the least appropriate in the further evaluation of this patient?

  • farkouh_progressivefatigue_t4.jpg
    Serum free thyroxine (T4) and morning cortisol measurement
  • Serum prolactin measurement
  • Serum ferritin measurement
  • Pituitary magnetic resonance imaging (MRI)
  • Chromosomal karyotyping

The diagnosis of central hypogonadism warrants evaluation for possible causes. These are summarized in Table 4.

Serum free thyroxine and morning cortisol

Since this patient’s LH and FSH values are abnormal, it is important to evaluate the status of other anterior pituitary hormones. In patients with pituitary abnormalities, serum free T4 is a more reliable test for assessing thyroid function than thyroid-stimulating hormone (TSH), because of loss of the negative feedback of thyroid hormones on the diseased pituitary. In contrast, serum TSH is considered the best single thyroid test to assess primary thyroid dysfunction.

Other measurements include prolactin and morning cortisol (reflecting adrenocorticotropic hormone status).

Prolactin measurement

Prolactin measurement is important to evaluate for hyperprolactinemia, as this will lead to hypogonadism by inhibition of GnRH secretion.25 Different pathologic, pharmacologic, and physiologic conditions can result in hyperprolactinemia, including prolactinomas, other pituitary and hypothalamic lesions, primary hypothyroidism, and medications such as antipsychotics.25 Dopamine agonists are the mainstay treatment for hyperprolactinemia.

Ferritin measurement

Ferritin measurement is indicated to diagnose iron overload conditions such as hemochromatosis, which can result in primary hypogonadism via testicular damage or in secondary hypogonadism via pituitary damage.26

Pituitary MRI with contrast

Pituitary MRI with contrast is used to diagnose structural lesions of the pituitary or hypothalamus. This diagnostic modality is indicated for patients with pituitary dysfunction, including central hypogonadism, manifestations of a mass effect (headache, visual field defects), persistent hyperprolactinemia, and panhypopituitarism, among others. To improve the diagnostic yield of pituitary MRI, the Endocrine Society guidelines recommend it for men with serum total testosterone levels below 150 ng/dL.5 However, some clinicians have a lower threshold for ordering pituitary MRI for patients with central hypogonadism. Physician judgment and expertise should be exercised and the decision made on an individual basis.

Chromosomal karyotyping

Chromosomal karyotyping is not indicated in our patient. It is reserved for those with primary hypogonadism to diagnose Klinefelter syndrome, which has a karyotype of 47,XXY.

CASE RESUMED: MOSH SYNDROME

Our patient’s prolactin, free T4, morning cortisol, and ferritin levels are measured, yielding normal values. No abnormalities are seen on pituitary MRI. A clinical reevaluation is conducted, revealing no history of head trauma or head and neck radiation. The lack of an obvious cause in our patient’s clinical presentation and workup, together with his obesity (BMI 32.8 kg/m2) supports the diagnosis of obesity as the cause of his hypogonadism.

Obesity can be a cause of secondary hypogonadism, which has led to the term “MOSH” (male obesity-associated secondary hypogonadism) syndrome. In fact, a cross-sectional study has demonstrated that 40% of nondiabetic obese (BMI ≥ 30 kg/m2) men over age 45 have low serum free testosterone levels, compared with 26% for lean (BMI < 25 kg/m2) men.27 Moreover, obesity has been found to be a strong predictor of testosterone replacement therapy.28 Other studies have also found an inverse relationship between BMI and testosterone levels.29

Several mechanisms interact in the pathogenesis of MOSH syndrome. Adipose tissue possesses aromatase activity, which converts androgens into estrogens.30 Peripheral estrogen production can in turn exert feedback inhibition on pituitary gonadotropin secretion.31 In obese men, increased adipose tissue leads to increased aromatase activity and more estrogen, so more feedback inhibition on the pituitary and subsequently secondary hypogonadism. 


Leptin, a hormone produced by adipocytes, is also increased in obesity, and was found to be inversely correlated with serum testosterone.32 Studies have demonstrated that leptin has an inhibitory effect on the enzymatic pathway that synthesizes testosterone in Leydig cells.33

Proinflammatory cytokines have also been implicated, as central obesity is associated with an increase in these cytokines, which in turn act negatively on the hypothalamus and impair GnRH release leading to lower testosterone.34,35

Treating obesity-related hypogonadism

In a pilot study,36 lifestyle attempts to reduce obesity were shown to improve hormonal levels. Bariatric surgery has also been demonstrated to be successful.37

Clomiphene citrate, a selective estrogen receptor modulator, increases endogenous testosterone secretion by inhibiting the negative feedback of estrogen on the hypothalamus and pituitary and thus increasing LH and FSH. It also preserves endogenous testosterone production, since it does not suppress the hypothalamic-pituitary-testicular axis.38 This made clomiphene citrate a potential treatment for men with central hypogonadism including those with MOSH.39

Nevertheless, there are no randomized trials to prove its safety and efficacy in the management of central hypogonadism.5 Regarding its use in men wishing to preserve fertility, most studies did not show improvement. However, a meta-analysis demonstrated statistically significant increased pregnancy rates in partners of men with idiopathic infertility if the men used 50 mg of clomiphene citrate daily.40

Testosterone deficiency can be a marker of metabolic syndrome, which needs to be managed more urgently than hypogonadism. A cross-sectional study found not only an association between metabolic syndrome and low serum testosterone, but also with each individual component of metabolic syndrome on its own, all of which need to be addressed.10

 

 

CASE CONTINUED: BEGINNING TREATMENT

The physician counsels the patient regarding the implications, potential adverse outcomes, and available treatments for his obesity, including lifestyle modification and bariatric surgery. The patient declines surgery and wishes to adopt a weight-reducing diet and exercise program, for which he is referred to a dietitian.

In addition, in view of the patient’s clinically and biochemically proven hypogonadism, his physician offers testosterone replacement therapy. He orders a serum prostate-specific antigen (PSA) level, which is 1.3 ng/dL (reference range < 4 ng/dL). The patient is prescribed 5 g of 1% testosterone gel daily.

TESTOSTERONE REPLACEMENT THERAPY

4. Which is the most common adverse effect of testosterone replacement therapy?

  • Cardiovascular events
  • Erythrocytosis
  • Prostate cancer
  • Infertility
  • Obstructive sleep apnea

farkouh_progressivefatigue_t5.jpg
Testosterone is indicated for men with an established diagnosis of hypogonadism. The benefits of testosterone replacement are summarized in Table 5.5,6

Clinicians should be very cautious in initiating testosterone replacement therapy in any patient with an unstable medical condition.

There are several formulations of testosterone replacement therapy, including intramuscular injections, transdermal gels or patches, buccal tablets, an intranasal gel, and oral tablets. Of note, there are 2 different forms of oral testosterone preparations: testosterone undecanoate and 17-alpha alkylated testosterone. The former is unavailable in the United States and the latter is not recommended for use due to its proven hepatic toxicity.41

Testosterone and erythrocytosis

Meta-analyses have concluded that the most frequent adverse event of testosterone replacement therapy is a significant rise in hematocrit.42 This rise was found to be dose-dependent and was more marked in older men.43 Although all preparations can cause erythrocytosis, parenteral forms have been observed to raise it the most, particularly short-term injectables.44,45

The mechanism behind this increase is attributed to increased erythropoietin levels and improved usage of iron for red blood cell synthesis.46 In fact, testosterone replacement therapy has been shown to improve hemoglobin levels in patients with anemia.47 On the other hand, increasing hematocrit levels may lead to thrombotic and vasoocclusive events.44

farkouh_progressivefatigue_f1.jpg
%3Cp%3EFigure%201.%20Hematocrit%20monitoring%20for%20patients%20on%20testosterone%20replacement%20therapy.%3C%2Fp%3E
It is strongly recommended that baseline hematocrit levels be measured before initiating testosterone replacement therapy.5,6 The hematocrit level should also be monitored 3 to 6 months into treatment and yearly thereafter while on testosterone.5Figure 1 summarizes the appropriate steps to undertake regarding hematocrit levels, according to the American Urological Association.6

Testosterone and prostate cancer

The relationship between testosterone treatment and prostate cancer has long been studied. Historically, testosterone replacement therapy was believed to increase the risk of prostate cancer; however, recent studies and meta-analyses have shown that this is not the case.42,48 Nevertheless, clinical guidelines still recommend prostate monitoring for men on testosterone replacement therapy.5,6

farkouh_progressivefatigue_t6.jpg
Furthermore, the clinician should make sure the patient does not have prostate cancer before initiating testosterone replacement therapy. Since there is a significant incidence of prostate cancer in men with serum PSA of 2.5–4.0 ng/mL, a patient with hypogonadism and a serum PSA in that range or higher should have appropriate evaluation before initiating testosterone replacement therapy.49 The Endocrine Society recommendations for prostate monitoring are summarized in Table 6.5

Testosterone and cardiovascular risk

The evidence regarding this issue has been contradictory and inconsistent. Meta-analyses have demonstrated that low testosterone is associated with higher risk of major adverse cardiovascular events.50 These studies argue for the use of testosterone replacement therapy in hypogonadal men to decrease the risk. However, other studies and meta-analyses have found that testosterone replacement therapy is associated with increased cardiovascular risk and have concluded that major adverse cardiac events are in fact a risk of testosterone replacement therapy.51

Current recommendations advocate against the use of testosterone replacement therapy in men with uncontrolled heart failure or with cardiovascular events in the past 3 to 6 months.5,6 Cardiovascular risk factors should be addressed and corrected, and patients should be educated on cardiovascular symptoms and the need to report them if they occur.

Testosterone and infertility

As described earlier, testosterone replacement therapy increases negative feedback on the pituitary and decreases LH and FSH production, leading to less spermatogenesis. Other treatment options should be sought for hypogonadal men wishing to preserve fertility.

Other adverse effects

Other adverse effects of testosterone replacement therapy include acne, oily skin, obstructive sleep apnea, gynecomastia, and balding.

Given all the adverse events that can be associated with testosterone replacement therapy, the risks and benefits of treating hypogonadism in each patient should be taken into consideration, and an individualized approach is required.

 

 

CASE RESUMED: FOLLOW-UP

The patient presents 3 months later for follow-up. He reports significant improvement in his presenting symptoms including energy, libido, and erectile function. He also reports some improvement in his mood and concentration. He has lost 12 lb (5.4 kg) and is still trying to improve his diet and exercise program. He is compliant with his testosterone gel therapy.

His serum calculated free testosterone level is 7.8 ng/dL (4.5–17), and his hematocrit is 46%. The patient is instructed to continue his treatment and to return after 9 months for further follow-up.

TAKE-HOME POINTS

  • Men with hypogonadism usually present with nonspecific manifestations, so clinicians should keep a high index of suspicion.
  • Both clinical and biochemical evidence of hypogonadism should be present to diagnose and start treatment for it.
  • Low levels of serum total testosterone do not necessarily reflect hypogonadism.
  • The hormonal profile of central hypogonadism reveals low serum testosterone with low or inappropriately normal serum LH and FSH levels.

Obesity can cause central hypogonadism and should be suspected after pituitary and other systemic causes are excluded.

A 48-year-old man presents to his primary care physician because of progressively decreasing energy and gradual decline in both libido and erectile function for the past 18 months. He has noticed decreased morning erections as well. He rates his libido at 3 to 4 on a scale of 10 for the past 6 months. He also reports poor motivation, depressed mood, impaired concentration, and sleep disturbances. He reports no hair loss, headache, or dizziness, and no decrease in shaving frequency. Review of his systems is otherwise unremarkable.

He has had dyslipidemia for 3 years and is not known to have hypertension or diabetes. His medications include atorvastatin, vitamin E, and multivitamins.

He is married with 3 children and does not wish to have more. He works as a software engineer and leads a sedentary lifestyle. He is a nonsmoker and occasionally drinks alcohol on the weekends.

On physical examination, he is alert and oriented and appears well. His height is 5 feet 10 inches (178 cm), weight 230 lb (104 kg), and body mass index (BMI) 32.8 kg/m2. His blood pressure is 115/83 mm Hg and pulse rate is 82 beats per minute and regular. Findings on cardiovascular and pulmonary examination are normal. He has large fatty breasts but without palpable glandular tissue.

farkouh_progressivefatigue_t1.jpg
Abdominal examination reveals central obesity—waist circumference 48 inches (122 cm)—without tenderness or organomegaly. There are no striae.

Genitourinary examination reveals normal hair distribution, a normal-sized penis, and slightly soft testes with testicular volume of 18–20 mL bilaterally.

His primary care physician suspects that he has low testosterone and orders some basic laboratory tests; the results are normal except for a low total testosterone level (Table 1).

FURTHER TESTING

1. Which of the following tests should his physician order next?

  • Repeat total testosterone measurement
  • Free testosterone measurement by commercial assay
  • Calculated free testosterone
  • Bioavailable testosterone measurement
  • Serum inhibin B measurement

This patient presents with several nonspecific symptoms. But collectively they suggest testosterone deficiency (hypogonadism).

farkouh_progressivefatigue_t2.jpg
Symptoms and signs of low testosterone vary according to age of onset. Prepubertal onset is associated with incomplete or delayed puberty, no development of secondary sexual characteristics, eunuchoid features, and small penis and testes. Postpubertal onset is associated with a wide array of symptoms (Table 2). Most manifestations of low testosterone are nonspecific, such as fatigue, impaired concentration, and sleep disturbance.1

Together, erectile dysfunction, low libido, and decreased morning erections strongly suggest hypogonadism.2 Loss of body hair and decreased shaving frequency are specific symptoms of hypogonadism; however, they require years to develop.3 Gynecomastia can also occur due to loss of the inhibitory action of testosterone on breast growth and a relative increase in estradiol. This occurs more in primary hypogonadism, due to the increase in luteinizing hormone (LH), which stimulates the remaining Leydig cells to secrete estradiol rather than testosterone.4

farkouh_progressivefatigue_t3.jpg
Screening for hypogonadism in men may be warranted in several conditions, even without clinical manifestations of low testosterone (Table 3).5–10

To diagnose hypogonadism in men and to start treatment for it, current guidelines recommend that the patient should have clinical features as well as laboratory evidence of low testosterone.5,6

Measuring testosterone: Total, free, bound, and bioavailable

Testosterone, a steroid hormone, circulates in the serum either as free testosterone or bound to several plasma proteins, mainly sex-hormone binding globulin (SHBG) and albumin.

Total testosterone includes both the free and bound fractions, whereas bioavailable testosterone includes both free and the portion bound to albumin, which has low affinity and can dissociate and be used at the tissue level.11

Low levels of total testosterone do not necessarily reflect a hypogonadal state, as a man with altered SHBG levels or binding capabilities can have low total but normal free testosterone levels and no manifestations.12 Several conditions can alter the levels of SHBG, including obesity, diabetes, aging, thyroid dysfunction, and others.5,13

Because our patient is obese, his total testosterone level is not a reliable indicator of hypogonadism, and repeating its measurement will not add diagnostic value.

Therefore, an alternative measurement should be used to accurately reflect the testosterone levels. From a physiologic point of view, bioavailable testosterone is the active form of testosterone and is the most accurate to be measured in a patient with hypogonadism. Nevertheless, because of technical difficulties in its measurement and lack of evidence correlating bioavailable testosterone with the clinical picture of hypogonadism, it is recommended that the level of free testosterone be used.5

The gold standard for direct measurement of serum free testosterone is equilibrium dialysis, but this is expensive and time-consuming.14 Commercial assays for free testosterone exist but have been deemed unreliable.14,15 It is recommended that free testosterone be measured by equilibrium dialysis or calculated using equations based on total testosterone, SHBG, and albumin levels.5 These equations are reliable and give results very close to the values obtained by equilibrium dialysis.15 Therefore, in our patient, it would be suitable to calculate the free testosterone level next.

Serum levels of free testosterone vary according to several factors. Diurnal variation of testosterone has been established: levels are highest in the morning and decline throughout the day.16 Food decreases testosterone levels.17 In addition, there is considerable day-to-day variation.18 Therefore, at least 2 readings of fasting morning testosterone on 2 separate days are recommended for the diagnosis of hypogonadism.5

Inhibin B is a hormone produced by Sertoli cells in the testes in response to follicle-stimulating hormone (FSH) stimulation. In turn, it acts as negative feedback, together with testosterone, to inhibit FSH release from the pituitary. Inhibin B has been shown to reflect spermatogenesis in the testes and therefore fertility.19 Inhibin B levels were found to be low in patients with central hypogonadism, due to less FSH release; however, they did not correlate with testosterone levels.20

 

 

CASE RESUMED: CHARACTERIZING HIS HYPOGONADISM

The patient’s physician orders morning fasting total testosterone, SHBG, and albumin testing and calculates the free testosterone level, which yields a value of 3 ng/dL (reference range 4.5–17). This is confirmed by a repeat measurement, which yields a value of 2.9 ng/dL. Laboratory test results combined with his clinical presentation are consistent with hypogonadism.

2. What is the most appropriate next step?

  • Measurement of serum LH and FSH
  • Measurement of serum prolactin
  • Scrotal ultrasonography
  • Gonadotropin-releasing hormone (GnRH) stimulation test
  • Semen analysis

After hypogonadism is diagnosed, it is important to distinguish if it is primary or central. This is achieved by measuring serum LH and FSH.5 All biotin supplements should be stopped at least 72 hours before measuring LH and FSH, as biotin can interfere with the assays, yielding false values.21

Secretion of FSH and LH from the anterior pituitary is under the influence of pulsatile release of GnRH from the hypothalamus. LH acts on Leydig cells in the testes to produce testosterone, whereas FSH acts on Sertoli cells, together with testosterone, to bring about spermatogenesis in the seminiferous tubules. Testosterone acts centrally as negative feedback to decrease the release of LH and FSH.

Primary hypogonadism occurs due to testicular failure, ie, the testes themselves fail to produce testosterone, leading to hypogonadism. The decrease in testosterone levels, together with inhibin B if Sertoli cells are damaged, lead to loss of negative feedback on the hypothalamus and pituitary, and therefore increased levels of LH and FSH. This is termed hypergonadotropic hypogonadism. Testicular failure may also result in impaired spermatogenesis and infertility due to destruction of testicular structures, in which case fertility cannot be restored.

Central hypogonadism occurs when the pituitary fails to produce LH and FSH (secondary hypogonadism) or when the hypothalamus fails to produce GnRH and subsequently the lack of secretion of LH and FSH from the pituitary (tertiary hypogonadism). The lack of LH will result in no stimulation of Leydig cells to produce testosterone, and therefore its deficiency. Serum hormone levels in central hypogonadism will reveal low testosterone, with either low or inappropriately normal gonadotropins (LH and FSH). This is termed hypogonadotropic hypogonadism. The lack of FSH, together with testosterone deficiency will also result in decreased spermatogenesis and therefore infertility. Testicular structures are preserved, however, and fertility can be restored with appropriate therapy, as discussed below.

Prolactin should be measured only if the patient has central hypogonadism. Its measurement is not warranted at this point in the patient’s workup. The implications of prolactin and its relationship to hypogonadism will be discussed later.

Although, this stepwise approach is not convenient for many patients, some physicians follow it because it is cost-effective, especially in those who are not insured. However, other physicians order FSH, LH, and sometimes prolactin with the confirmatory low testosterone measurement. Laboratories can also be instructed to wait to measure the pituitary hormones and to do so only if low testosterone is confirmed.

Varicocele, a possible cause of male infertility, can also impair Leydig cell function and cause low testosterone. In fact, surgical repair of varicocele has been demonstrated to increase serum testosterone.22 Scrotal ultrasonography is used to diagnose varicocele, but this also should be ordered at a later stage in the workup if primary hypogonadism is diagnosed.

The GnRH stimulation test is important for the diagnosis and evaluation of precocious or delayed puberty in children. In boys with delayed puberty, a poorer response to GnRH stimulation indicates central hypogonadism rather than constitutional delay.23 It has no role in the evaluation of postpubertal or adult-onset hypogonadism.

Semen analysis is important to evaluate fertility if the patient is interested in further procreation.5 Low testosterone levels may result in impaired spermatogenesis and therefore infertility. On the other hand, treatment with exogenous testosterone will also result in infertility, by feedback inhibition of LH and FSH and therefore inhibition of spermatogenesis. If the patient wishes to preserve fertility, treatment options other than testosterone should be considered; examples include clomiphene citrate, human menopausal gonadotropin, and human chorionic gonadotropin.23,24

Our patient has no desire to expand his family; therefore, a semen analysis and attempts to preserve spermatogenesis are not indicated.

 

 

CASE RESUMED: SEARCHING FOR CAUSES

His physician orders testing of serum LH and FSH, yielding the following values:

  • LH 1.6 mIU/mL (reference range 1.8–12)
  • FSH 1.9 mIU/mL (reference range 1.5–12.5).

The diagnosis of central hypogonadism is established.

3. Which investigation is the least appropriate in the further evaluation of this patient?

  • farkouh_progressivefatigue_t4.jpg
    Serum free thyroxine (T4) and morning cortisol measurement
  • Serum prolactin measurement
  • Serum ferritin measurement
  • Pituitary magnetic resonance imaging (MRI)
  • Chromosomal karyotyping

The diagnosis of central hypogonadism warrants evaluation for possible causes. These are summarized in Table 4.

Serum free thyroxine and morning cortisol

Since this patient’s LH and FSH values are abnormal, it is important to evaluate the status of other anterior pituitary hormones. In patients with pituitary abnormalities, serum free T4 is a more reliable test for assessing thyroid function than thyroid-stimulating hormone (TSH), because of loss of the negative feedback of thyroid hormones on the diseased pituitary. In contrast, serum TSH is considered the best single thyroid test to assess primary thyroid dysfunction.

Other measurements include prolactin and morning cortisol (reflecting adrenocorticotropic hormone status).

Prolactin measurement

Prolactin measurement is important to evaluate for hyperprolactinemia, as this will lead to hypogonadism by inhibition of GnRH secretion.25 Different pathologic, pharmacologic, and physiologic conditions can result in hyperprolactinemia, including prolactinomas, other pituitary and hypothalamic lesions, primary hypothyroidism, and medications such as antipsychotics.25 Dopamine agonists are the mainstay treatment for hyperprolactinemia.

Ferritin measurement

Ferritin measurement is indicated to diagnose iron overload conditions such as hemochromatosis, which can result in primary hypogonadism via testicular damage or in secondary hypogonadism via pituitary damage.26

Pituitary MRI with contrast

Pituitary MRI with contrast is used to diagnose structural lesions of the pituitary or hypothalamus. This diagnostic modality is indicated for patients with pituitary dysfunction, including central hypogonadism, manifestations of a mass effect (headache, visual field defects), persistent hyperprolactinemia, and panhypopituitarism, among others. To improve the diagnostic yield of pituitary MRI, the Endocrine Society guidelines recommend it for men with serum total testosterone levels below 150 ng/dL.5 However, some clinicians have a lower threshold for ordering pituitary MRI for patients with central hypogonadism. Physician judgment and expertise should be exercised and the decision made on an individual basis.

Chromosomal karyotyping

Chromosomal karyotyping is not indicated in our patient. It is reserved for those with primary hypogonadism to diagnose Klinefelter syndrome, which has a karyotype of 47,XXY.

CASE RESUMED: MOSH SYNDROME

Our patient’s prolactin, free T4, morning cortisol, and ferritin levels are measured, yielding normal values. No abnormalities are seen on pituitary MRI. A clinical reevaluation is conducted, revealing no history of head trauma or head and neck radiation. The lack of an obvious cause in our patient’s clinical presentation and workup, together with his obesity (BMI 32.8 kg/m2) supports the diagnosis of obesity as the cause of his hypogonadism.

Obesity can be a cause of secondary hypogonadism, which has led to the term “MOSH” (male obesity-associated secondary hypogonadism) syndrome. In fact, a cross-sectional study has demonstrated that 40% of nondiabetic obese (BMI ≥ 30 kg/m2) men over age 45 have low serum free testosterone levels, compared with 26% for lean (BMI < 25 kg/m2) men.27 Moreover, obesity has been found to be a strong predictor of testosterone replacement therapy.28 Other studies have also found an inverse relationship between BMI and testosterone levels.29

Several mechanisms interact in the pathogenesis of MOSH syndrome. Adipose tissue possesses aromatase activity, which converts androgens into estrogens.30 Peripheral estrogen production can in turn exert feedback inhibition on pituitary gonadotropin secretion.31 In obese men, increased adipose tissue leads to increased aromatase activity and more estrogen, so more feedback inhibition on the pituitary and subsequently secondary hypogonadism. 


Leptin, a hormone produced by adipocytes, is also increased in obesity, and was found to be inversely correlated with serum testosterone.32 Studies have demonstrated that leptin has an inhibitory effect on the enzymatic pathway that synthesizes testosterone in Leydig cells.33

Proinflammatory cytokines have also been implicated, as central obesity is associated with an increase in these cytokines, which in turn act negatively on the hypothalamus and impair GnRH release leading to lower testosterone.34,35

Treating obesity-related hypogonadism

In a pilot study,36 lifestyle attempts to reduce obesity were shown to improve hormonal levels. Bariatric surgery has also been demonstrated to be successful.37

Clomiphene citrate, a selective estrogen receptor modulator, increases endogenous testosterone secretion by inhibiting the negative feedback of estrogen on the hypothalamus and pituitary and thus increasing LH and FSH. It also preserves endogenous testosterone production, since it does not suppress the hypothalamic-pituitary-testicular axis.38 This made clomiphene citrate a potential treatment for men with central hypogonadism including those with MOSH.39

Nevertheless, there are no randomized trials to prove its safety and efficacy in the management of central hypogonadism.5 Regarding its use in men wishing to preserve fertility, most studies did not show improvement. However, a meta-analysis demonstrated statistically significant increased pregnancy rates in partners of men with idiopathic infertility if the men used 50 mg of clomiphene citrate daily.40

Testosterone deficiency can be a marker of metabolic syndrome, which needs to be managed more urgently than hypogonadism. A cross-sectional study found not only an association between metabolic syndrome and low serum testosterone, but also with each individual component of metabolic syndrome on its own, all of which need to be addressed.10

 

 

CASE CONTINUED: BEGINNING TREATMENT

The physician counsels the patient regarding the implications, potential adverse outcomes, and available treatments for his obesity, including lifestyle modification and bariatric surgery. The patient declines surgery and wishes to adopt a weight-reducing diet and exercise program, for which he is referred to a dietitian.

In addition, in view of the patient’s clinically and biochemically proven hypogonadism, his physician offers testosterone replacement therapy. He orders a serum prostate-specific antigen (PSA) level, which is 1.3 ng/dL (reference range < 4 ng/dL). The patient is prescribed 5 g of 1% testosterone gel daily.

TESTOSTERONE REPLACEMENT THERAPY

4. Which is the most common adverse effect of testosterone replacement therapy?

  • Cardiovascular events
  • Erythrocytosis
  • Prostate cancer
  • Infertility
  • Obstructive sleep apnea

farkouh_progressivefatigue_t5.jpg
Testosterone is indicated for men with an established diagnosis of hypogonadism. The benefits of testosterone replacement are summarized in Table 5.5,6

Clinicians should be very cautious in initiating testosterone replacement therapy in any patient with an unstable medical condition.

There are several formulations of testosterone replacement therapy, including intramuscular injections, transdermal gels or patches, buccal tablets, an intranasal gel, and oral tablets. Of note, there are 2 different forms of oral testosterone preparations: testosterone undecanoate and 17-alpha alkylated testosterone. The former is unavailable in the United States and the latter is not recommended for use due to its proven hepatic toxicity.41

Testosterone and erythrocytosis

Meta-analyses have concluded that the most frequent adverse event of testosterone replacement therapy is a significant rise in hematocrit.42 This rise was found to be dose-dependent and was more marked in older men.43 Although all preparations can cause erythrocytosis, parenteral forms have been observed to raise it the most, particularly short-term injectables.44,45

The mechanism behind this increase is attributed to increased erythropoietin levels and improved usage of iron for red blood cell synthesis.46 In fact, testosterone replacement therapy has been shown to improve hemoglobin levels in patients with anemia.47 On the other hand, increasing hematocrit levels may lead to thrombotic and vasoocclusive events.44

farkouh_progressivefatigue_f1.jpg
%3Cp%3EFigure%201.%20Hematocrit%20monitoring%20for%20patients%20on%20testosterone%20replacement%20therapy.%3C%2Fp%3E
It is strongly recommended that baseline hematocrit levels be measured before initiating testosterone replacement therapy.5,6 The hematocrit level should also be monitored 3 to 6 months into treatment and yearly thereafter while on testosterone.5Figure 1 summarizes the appropriate steps to undertake regarding hematocrit levels, according to the American Urological Association.6

Testosterone and prostate cancer

The relationship between testosterone treatment and prostate cancer has long been studied. Historically, testosterone replacement therapy was believed to increase the risk of prostate cancer; however, recent studies and meta-analyses have shown that this is not the case.42,48 Nevertheless, clinical guidelines still recommend prostate monitoring for men on testosterone replacement therapy.5,6

farkouh_progressivefatigue_t6.jpg
Furthermore, the clinician should make sure the patient does not have prostate cancer before initiating testosterone replacement therapy. Since there is a significant incidence of prostate cancer in men with serum PSA of 2.5–4.0 ng/mL, a patient with hypogonadism and a serum PSA in that range or higher should have appropriate evaluation before initiating testosterone replacement therapy.49 The Endocrine Society recommendations for prostate monitoring are summarized in Table 6.5

Testosterone and cardiovascular risk

The evidence regarding this issue has been contradictory and inconsistent. Meta-analyses have demonstrated that low testosterone is associated with higher risk of major adverse cardiovascular events.50 These studies argue for the use of testosterone replacement therapy in hypogonadal men to decrease the risk. However, other studies and meta-analyses have found that testosterone replacement therapy is associated with increased cardiovascular risk and have concluded that major adverse cardiac events are in fact a risk of testosterone replacement therapy.51

Current recommendations advocate against the use of testosterone replacement therapy in men with uncontrolled heart failure or with cardiovascular events in the past 3 to 6 months.5,6 Cardiovascular risk factors should be addressed and corrected, and patients should be educated on cardiovascular symptoms and the need to report them if they occur.

Testosterone and infertility

As described earlier, testosterone replacement therapy increases negative feedback on the pituitary and decreases LH and FSH production, leading to less spermatogenesis. Other treatment options should be sought for hypogonadal men wishing to preserve fertility.

Other adverse effects

Other adverse effects of testosterone replacement therapy include acne, oily skin, obstructive sleep apnea, gynecomastia, and balding.

Given all the adverse events that can be associated with testosterone replacement therapy, the risks and benefits of treating hypogonadism in each patient should be taken into consideration, and an individualized approach is required.

 

 

CASE RESUMED: FOLLOW-UP

The patient presents 3 months later for follow-up. He reports significant improvement in his presenting symptoms including energy, libido, and erectile function. He also reports some improvement in his mood and concentration. He has lost 12 lb (5.4 kg) and is still trying to improve his diet and exercise program. He is compliant with his testosterone gel therapy.

His serum calculated free testosterone level is 7.8 ng/dL (4.5–17), and his hematocrit is 46%. The patient is instructed to continue his treatment and to return after 9 months for further follow-up.

TAKE-HOME POINTS

  • Men with hypogonadism usually present with nonspecific manifestations, so clinicians should keep a high index of suspicion.
  • Both clinical and biochemical evidence of hypogonadism should be present to diagnose and start treatment for it.
  • Low levels of serum total testosterone do not necessarily reflect hypogonadism.
  • The hormonal profile of central hypogonadism reveals low serum testosterone with low or inappropriately normal serum LH and FSH levels.

Obesity can cause central hypogonadism and should be suspected after pituitary and other systemic causes are excluded.

References
  1. Araujo AB, Esche GR, Kupelian V, et al. Prevalence of symptomatic androgen deficiency in men. J Clin Endocrinol Metab 2007; 92(11):4241–4247. doi:10.1210/jc.2007-1245
  2. Wu FCW, Tajar A, Beynon JM, et al; EMAS Group. Identification of late-onset hypogonadism in middle-aged and elderly men. N Engl J Med 2010; 363(2):123–135. doi:10.1056/NEJMoa0911101
  3. Arver S, Lehtihet M. Current guidelines for the diagnosis of testosterone deficiency. Front Horm Res 2009; 37:5–20. doi:10.1159/000175839
  4. Narula HS, Carlson HE. Gynaecomastia—pathophysiology, diagnosis and treatment. Nat Rev Endocrinol 2014; 10(11):684–698. doi:10.1038/nrendo.2014.139
  5. Bhasin S, Brito JP, Cunningham GR, et al. Testosterone therapy in men with hypogonadism: an Endocrine Society clinical practice guideline. J Clin Endocrinol Metab 2018; 103(5):1715–1744. doi:10.1210/jc.2018-00229
  6. Mulhall JP, Trost LW, Brannigan RE, et al. Evaluation and management of testosterone deficiency: AUA guideline. J Urol 2018; 200(2):423–432. doi:10.1016/j.juro.2018.03.115
  7. Balasubramanian V, Naing S. Hypogonadism in chronic obstructive pulmonary disease: incidence and effects. Curr Opin Pulm Med 2012; 18(2):112–117. doi:10.1097/MCP.0b013e32834feb37
  8. Atlantis E, Fahey P, Cochrane B, Wittert G, Smith S. Endogenous testosterone level and testosterone supplementation therapy in chronic obstructive pulmonary disease (COPD): a systematic review and meta-analysis. BMJ Open 2013; 3(8)pii:e003127. doi:10.1136/bmjopen-2013-003127
  9. Bawor M, Bami H, Dennis BB, et al. Testosterone suppression in opioid users: a systematic review and meta-analysis. Drug Alcohol Depend 2015; 149:1–9. doi:10.1016/j.drugalcdep.2015.01.038
  10. Tan WS, Ng CJ, Khoo EM, Low WY, Tan HM. The triad of erectile dysfunction, testosterone deficiency syndrome and metabolic syndrome: findings from a multi-ethnic Asian men study (The Subang Men's Health Study). Aging Male 2011; 14(4):231–236. doi:10.3109/13685538.2011.597463
  11. Goldman AL, Bhasin S, Wu FCW, Krishna M, Matsumoto AM, Jasuja R. A reappraisal of testosterone’s binding in circulation: physiological and clinical implications. Endocr Rev 2017; 38(4):302–324. doi:10.1210/er.2017-00025
  12. Antonio L, Wu FC, O’Neill TW, et al; European Male Ageing Study Study Group. Low free testosterone is associated with hypogonadal signs and symptoms in men with normal total testosterone. J Clin Endocrinol Metab 2016; 101(7):2647–2657. doi:10.1210/jc.2015-4106
  13. Liu F, Shen X, Wang R, et al. Association of central obesity with sex hormone binding globulin: a cross-sectional study of 1166 Chinese men. Open Med (Wars) 2018; 13:196–202. doi:10.1515/med-2018-0030
  14. Vermeulen A, Verdonck L, Kaufman JM. A critical evaluation of simple methods for the estimation of free testosterone in serum. J Clin Endocrinol Metab 1999; 84(10):3666–3672. doi:10.1210/jcem.84.10.6079
  15. Halmenschlager G, Rhoden EL, Riedner CE. Calculated free testosterone and radioimmunoassay free testosterone as a predictor of subnormal levels of total testosterone. Int Urol Nephrol 2012; 44(3):673–681. doi:10.1007/s11255-011-0066-z
  16. Brambilla DJ, Matsumoto AM, Araujo AB, McKinlay JB. The effect of diurnal variation on clinical measurement of serum testosterone and other sex hormone levels in men. J Clin Endocrinol Metab 2009; 94(3):907–913. doi:10.1210/jc.2008-1902
  17. Lehtihet M, Arver S, Bartuseviciene I, Pousette Å. S-testosterone decrease after a mixed meal in healthy men independent of SHBG and gonadotrophin levels. Andrologia 2012; 44(6):405–410. doi:10.1111/j.1439-0272.2012.01296.x
  18. Brambilla DJ, O’Donnell AB, Matsumoto AM, McKinlay JB. Intraindividual variation in levels of serum testosterone and other reproductive and adrenal hormones in men. Clin Endocrinol (Oxf) 2007; 67(6):853–862. doi:10.1111/j.1365-2265.2007.02976.x
  19. Manzoor SM, Sattar A, Hashim R, et al. Serum inhibin B as a diagnostic marker of male infertility. J Ayub Med Coll Abbottabad 2012; 24(3–4):113–116. pmid:24669628
  20. Kolb BA, Stanczyk FZ, Sokol RZ. Serum inhibin B levels in males with gonadal dysfunction. Fertil Steril 2000; 74(2):234–238. pmid:10927037
  21. Trambas CM, Sikaris KA, Lu ZX. More on biotin treatment mimicking Graves’ disease. N Engl J Med 2016; 375(17):1698. doi:10.1056/NEJMc1611875
  22. Li F, Yue H, Yamaguchi K, et al. Effect of surgical repair on testosterone production in infertile men with varicocele: a meta-analysis. Int J Urol 2012; 19(2):149–154. doi:10.1111/j.1442-2042.2011.02890.x
  23. Crosnoe-Shipley LE, Elkelany OO, Rahnema CD, Kim ED. Treatment of hypogonadotropic male hypogonadism: case-based scenarios. World J Nephrol 2015; 4(2):245–253. doi:10.5527/wjn.v4.i2.245
  24. Majzoub A, Sabanegh E Jr. Testosterone replacement in the infertile man. Transl Androl Urol 2016; 5(6):859–865. doi:10.21037/tau.2016.08.03
  25. Majumdar A, Mangal NS. Hyperprolactinemia. J Hum Reprod Sci 2013; 6(3):168–175. doi:10.4103/0974-1208.121400
  26. El Osta R, Grandpre N, Monnin N, Hubert J, Koscinski I. Hypogonadotropic hypogonadism in men with hereditary hemochromatosis. Basic Clin Androl 2017; 27:13. doi:10.1186/s12610-017-0057-8
  27. Dhindsa S, Miller MG, McWhirter CL, et al. Testosterone concentrations in diabetic and nondiabetic obese men. Diabetes Care 2010; 33(6):1186–1192. doi:10.2337/dc09-1649
  28. Jasuja GK, Bhasin S, Reisman JI, et al. Who gets testosterone? Patient characteristics associated with testosterone prescribing in the Veteran Affairs system: a cross-sectional study. J Gen Intern Med 2017; 32(3):304–311. doi:10.1007/s11606-016-3940-7
  29. Kaplan SA, Lee JY, O’Neill EA, Meehan AG, Kusek JW. Prevalence of low testosterone and its relationship to body mass index in older men with lower urinary tract symptoms associated with benign prostatic hyperplasia. Aging Male 2013; 16(4):169–172. doi:10.3109/13685538.2013.844786
  30. Lee HK, Lee JK, Cho B. The role of androgen in the adipose tissue of males. World J Mens Health 2013; 31(2):136–140. doi:10.5534/wjmh.2013.31.2.136
  31. Raven G, De Jong FH, Kaufman JM, De Ronde W. In men, peripheral estradiol levels directly reflect the action of estrogens at the hypothalamo-pituitary level to inhibit gonadotropin secretion. J Clin Endocrinol Metab 2006; 91(9):3324–3328. doi:10.1210/jc.2006-0462
  32. Hofny ER, Ali ME, Abdel-Hafez HZ, et al. Semen parameters and hormonal profile in obese fertile and infertile males. Fertil Steril 2010; 94(2):581–584. doi:10.1016/j.fertnstert.2009.03.085
  33. Isidori AM, Caprio M, Strollo F, et al. Leptin and androgens in male obesity: evidence for leptin contribution to reduced androgen levels. J Clin Endocrinol Metab 1999; 84(10):3673–3680. doi:10.1210/jcem.84.10.6082
  34. El-Wakkad A, Hassan NM, Sibaii H, El-Zayat SR. Proinflammatory, anti-inflammatory cytokines and adiponkines in students with central obesity. Cytokine 2013; 61(2):682–687. doi:10.1016/j.cyto.2012.11.010
  35. Maggio M, Basaria S, Ceda GP, et al. The relationship between testosterone and molecular markers of inflammation in older men. J Endocrinol Invest 2005; 28(suppl proceedings 11):116–119. pmid:16760639
  36. de Lorenzo A, Noce A, Moriconi E, et al. MOSH syndrome (male obesity secondary hypogonadism): clinical assessment and possible therapeutic approaches. Nutrients 2018; 10(4)pii:E474. doi:10.3390/nu10040474
  37. Escobar-Morreale HF, Santacruz E, Luque-Ramírez M, Botella Carretero JI. Prevalence of ‘obesity-associated gonadal dysfunction’ in severely obese men and women and its resolution after bariatric surgery: a systematic review and meta-analysis. Hum Reprod Update 2017; 23(4):390–408. doi:10.1093/humupd/dmx012
  38. Lo EM, Rodriguez KM, Pastuszak AW, Khera M. Alternatives to testosterone therapy: a review. Sex Med Rev 2018; 6(1):106–113. doi:10.1016/j.sxmr.2017.09.004
  39. Soares AH, Horie NC, Chiang LAP, et al. Effects of clomiphene citrate on male obesity-associated hypogonadism: a randomized, double-blind, placebo-controlled study. Int J Obes (Lond) 2018; 42(5):953–963. doi:10.1038/s41366-018-0105-2
  40. Chua ME, Escusa KG, Luna S, Tapia LC, Dofitas B, Morales M. Revisiting oestrogen antagonists (clomiphene or tamoxifen) as medical empiric therapy for idiopathic male infertility: a meta-analysis. Andrology 2013; 1(5):749–757. doi:10.1111/j.2047-2927.2013.00107.x
  41. Westaby D, Ogle SJ, Paradinas FJ, Randell JB, Murray-Lyon IM. Liver damage from long-term methyltestosterone. Lancet 1977; 2(8032):262–263. pmid:69876
  42. Fernández-Balsells MM, Murad MH, Lane M, et al. Clinical review 1: Adverse effects of testosterone therapy in adult men: a systematic review and meta-analysis. J Clin Endocrinol Metab 2010; 95(6):2560–2575. doi:10.1210/jc.2009-2575
  43. Coviello AD, Kaplan B, Lakshman KM, Chen T, Singh AB, Bhasin S. Effects of graded doses of testosterone on erythropoiesis in healthy young and older men. J Clin Endocrinol Metab 2008; 93(3):914–919. doi:10.1210/jc.2007-1692
  44. Ohlander SJ, Varghese B, Pastuszak AW. Erythrocytosis following testosterone therapy. Sex Med Rev 2018; 6(1):77–85. doi:10.1016/j.sxmr.2017.04.001
  45. Jones SD Jr, Dukovac T, Sangkum P, Yafi FA, Hellstrom WJ. Erythrocytosis and polycythemia secondary to testosterone replacement therapy in the aging male. Sex Med Rev 2015; 3(2):101–112. doi:10.1002/smrj.43
  46. Bachman E, Travison TG, Basaria S, et al. Testosterone induces erythrocytosis via increased erythropoietin and suppressed hepcidin: evidence for a new erythropoietin/hemoglobin set point. J Gerontol A Biol Sci Med Sci 2014; 69(6):725–735. doi:10.1093/gerona/glt154
  47. Roy CN, Snyder PJ, Stephens-Shields AJ, et al. Association of testosterone levels with anemia in older men: a controlled clinical trial. JAMA Intern Med 2017; 177(4):480–490. doi:10.1001/jamainternmed.2016.9540
  48. Klap J, Schmid M, Loughlin KR. The relationship between total testosterone levels and prostate cancer: a review of the continuing controversy. J Urol 2015; 193(2):403–413. doi:10.1016/j.juro.2014.07.123
  49. Gilbert SM, Cavallo CB, Kahane H, Lowe FC. Evidence suggesting PSA cutpoint of 2.5 ng/mL for prompting prostate biopsy: review of 36,316 biopsies. Urology 2005; 65(3):549–553. doi:10.1016/j.urology.2004.10.064
  50. Araujo AB, Dixon JM, Suarez EA, Murad MH, Guey LT, Wittert GA. Clinical review: Endogenous testosterone and mortality in men: a systematic review and meta-analysis. J Clin Endocrinol Metab 2011; 96(10):3007–3019. doi:10.1210/jc.2011-1137
  51. Xu L, Freeman G, Cowling BJ, Schooling CM. Testosterone therapy and cardiovascular events among men: a systematic review and meta-analysis of placebo-controlled randomized trials. BMC Med 2013; 11:108. doi:10.1186/1741-7015-11-108
References
  1. Araujo AB, Esche GR, Kupelian V, et al. Prevalence of symptomatic androgen deficiency in men. J Clin Endocrinol Metab 2007; 92(11):4241–4247. doi:10.1210/jc.2007-1245
  2. Wu FCW, Tajar A, Beynon JM, et al; EMAS Group. Identification of late-onset hypogonadism in middle-aged and elderly men. N Engl J Med 2010; 363(2):123–135. doi:10.1056/NEJMoa0911101
  3. Arver S, Lehtihet M. Current guidelines for the diagnosis of testosterone deficiency. Front Horm Res 2009; 37:5–20. doi:10.1159/000175839
  4. Narula HS, Carlson HE. Gynaecomastia—pathophysiology, diagnosis and treatment. Nat Rev Endocrinol 2014; 10(11):684–698. doi:10.1038/nrendo.2014.139
  5. Bhasin S, Brito JP, Cunningham GR, et al. Testosterone therapy in men with hypogonadism: an Endocrine Society clinical practice guideline. J Clin Endocrinol Metab 2018; 103(5):1715–1744. doi:10.1210/jc.2018-00229
  6. Mulhall JP, Trost LW, Brannigan RE, et al. Evaluation and management of testosterone deficiency: AUA guideline. J Urol 2018; 200(2):423–432. doi:10.1016/j.juro.2018.03.115
  7. Balasubramanian V, Naing S. Hypogonadism in chronic obstructive pulmonary disease: incidence and effects. Curr Opin Pulm Med 2012; 18(2):112–117. doi:10.1097/MCP.0b013e32834feb37
  8. Atlantis E, Fahey P, Cochrane B, Wittert G, Smith S. Endogenous testosterone level and testosterone supplementation therapy in chronic obstructive pulmonary disease (COPD): a systematic review and meta-analysis. BMJ Open 2013; 3(8)pii:e003127. doi:10.1136/bmjopen-2013-003127
  9. Bawor M, Bami H, Dennis BB, et al. Testosterone suppression in opioid users: a systematic review and meta-analysis. Drug Alcohol Depend 2015; 149:1–9. doi:10.1016/j.drugalcdep.2015.01.038
  10. Tan WS, Ng CJ, Khoo EM, Low WY, Tan HM. The triad of erectile dysfunction, testosterone deficiency syndrome and metabolic syndrome: findings from a multi-ethnic Asian men study (The Subang Men's Health Study). Aging Male 2011; 14(4):231–236. doi:10.3109/13685538.2011.597463
  11. Goldman AL, Bhasin S, Wu FCW, Krishna M, Matsumoto AM, Jasuja R. A reappraisal of testosterone’s binding in circulation: physiological and clinical implications. Endocr Rev 2017; 38(4):302–324. doi:10.1210/er.2017-00025
  12. Antonio L, Wu FC, O’Neill TW, et al; European Male Ageing Study Study Group. Low free testosterone is associated with hypogonadal signs and symptoms in men with normal total testosterone. J Clin Endocrinol Metab 2016; 101(7):2647–2657. doi:10.1210/jc.2015-4106
  13. Liu F, Shen X, Wang R, et al. Association of central obesity with sex hormone binding globulin: a cross-sectional study of 1166 Chinese men. Open Med (Wars) 2018; 13:196–202. doi:10.1515/med-2018-0030
  14. Vermeulen A, Verdonck L, Kaufman JM. A critical evaluation of simple methods for the estimation of free testosterone in serum. J Clin Endocrinol Metab 1999; 84(10):3666–3672. doi:10.1210/jcem.84.10.6079
  15. Halmenschlager G, Rhoden EL, Riedner CE. Calculated free testosterone and radioimmunoassay free testosterone as a predictor of subnormal levels of total testosterone. Int Urol Nephrol 2012; 44(3):673–681. doi:10.1007/s11255-011-0066-z
  16. Brambilla DJ, Matsumoto AM, Araujo AB, McKinlay JB. The effect of diurnal variation on clinical measurement of serum testosterone and other sex hormone levels in men. J Clin Endocrinol Metab 2009; 94(3):907–913. doi:10.1210/jc.2008-1902
  17. Lehtihet M, Arver S, Bartuseviciene I, Pousette Å. S-testosterone decrease after a mixed meal in healthy men independent of SHBG and gonadotrophin levels. Andrologia 2012; 44(6):405–410. doi:10.1111/j.1439-0272.2012.01296.x
  18. Brambilla DJ, O’Donnell AB, Matsumoto AM, McKinlay JB. Intraindividual variation in levels of serum testosterone and other reproductive and adrenal hormones in men. Clin Endocrinol (Oxf) 2007; 67(6):853–862. doi:10.1111/j.1365-2265.2007.02976.x
  19. Manzoor SM, Sattar A, Hashim R, et al. Serum inhibin B as a diagnostic marker of male infertility. J Ayub Med Coll Abbottabad 2012; 24(3–4):113–116. pmid:24669628
  20. Kolb BA, Stanczyk FZ, Sokol RZ. Serum inhibin B levels in males with gonadal dysfunction. Fertil Steril 2000; 74(2):234–238. pmid:10927037
  21. Trambas CM, Sikaris KA, Lu ZX. More on biotin treatment mimicking Graves’ disease. N Engl J Med 2016; 375(17):1698. doi:10.1056/NEJMc1611875
  22. Li F, Yue H, Yamaguchi K, et al. Effect of surgical repair on testosterone production in infertile men with varicocele: a meta-analysis. Int J Urol 2012; 19(2):149–154. doi:10.1111/j.1442-2042.2011.02890.x
  23. Crosnoe-Shipley LE, Elkelany OO, Rahnema CD, Kim ED. Treatment of hypogonadotropic male hypogonadism: case-based scenarios. World J Nephrol 2015; 4(2):245–253. doi:10.5527/wjn.v4.i2.245
  24. Majzoub A, Sabanegh E Jr. Testosterone replacement in the infertile man. Transl Androl Urol 2016; 5(6):859–865. doi:10.21037/tau.2016.08.03
  25. Majumdar A, Mangal NS. Hyperprolactinemia. J Hum Reprod Sci 2013; 6(3):168–175. doi:10.4103/0974-1208.121400
  26. El Osta R, Grandpre N, Monnin N, Hubert J, Koscinski I. Hypogonadotropic hypogonadism in men with hereditary hemochromatosis. Basic Clin Androl 2017; 27:13. doi:10.1186/s12610-017-0057-8
  27. Dhindsa S, Miller MG, McWhirter CL, et al. Testosterone concentrations in diabetic and nondiabetic obese men. Diabetes Care 2010; 33(6):1186–1192. doi:10.2337/dc09-1649
  28. Jasuja GK, Bhasin S, Reisman JI, et al. Who gets testosterone? Patient characteristics associated with testosterone prescribing in the Veteran Affairs system: a cross-sectional study. J Gen Intern Med 2017; 32(3):304–311. doi:10.1007/s11606-016-3940-7
  29. Kaplan SA, Lee JY, O’Neill EA, Meehan AG, Kusek JW. Prevalence of low testosterone and its relationship to body mass index in older men with lower urinary tract symptoms associated with benign prostatic hyperplasia. Aging Male 2013; 16(4):169–172. doi:10.3109/13685538.2013.844786
  30. Lee HK, Lee JK, Cho B. The role of androgen in the adipose tissue of males. World J Mens Health 2013; 31(2):136–140. doi:10.5534/wjmh.2013.31.2.136
  31. Raven G, De Jong FH, Kaufman JM, De Ronde W. In men, peripheral estradiol levels directly reflect the action of estrogens at the hypothalamo-pituitary level to inhibit gonadotropin secretion. J Clin Endocrinol Metab 2006; 91(9):3324–3328. doi:10.1210/jc.2006-0462
  32. Hofny ER, Ali ME, Abdel-Hafez HZ, et al. Semen parameters and hormonal profile in obese fertile and infertile males. Fertil Steril 2010; 94(2):581–584. doi:10.1016/j.fertnstert.2009.03.085
  33. Isidori AM, Caprio M, Strollo F, et al. Leptin and androgens in male obesity: evidence for leptin contribution to reduced androgen levels. J Clin Endocrinol Metab 1999; 84(10):3673–3680. doi:10.1210/jcem.84.10.6082
  34. El-Wakkad A, Hassan NM, Sibaii H, El-Zayat SR. Proinflammatory, anti-inflammatory cytokines and adiponkines in students with central obesity. Cytokine 2013; 61(2):682–687. doi:10.1016/j.cyto.2012.11.010
  35. Maggio M, Basaria S, Ceda GP, et al. The relationship between testosterone and molecular markers of inflammation in older men. J Endocrinol Invest 2005; 28(suppl proceedings 11):116–119. pmid:16760639
  36. de Lorenzo A, Noce A, Moriconi E, et al. MOSH syndrome (male obesity secondary hypogonadism): clinical assessment and possible therapeutic approaches. Nutrients 2018; 10(4)pii:E474. doi:10.3390/nu10040474
  37. Escobar-Morreale HF, Santacruz E, Luque-Ramírez M, Botella Carretero JI. Prevalence of ‘obesity-associated gonadal dysfunction’ in severely obese men and women and its resolution after bariatric surgery: a systematic review and meta-analysis. Hum Reprod Update 2017; 23(4):390–408. doi:10.1093/humupd/dmx012
  38. Lo EM, Rodriguez KM, Pastuszak AW, Khera M. Alternatives to testosterone therapy: a review. Sex Med Rev 2018; 6(1):106–113. doi:10.1016/j.sxmr.2017.09.004
  39. Soares AH, Horie NC, Chiang LAP, et al. Effects of clomiphene citrate on male obesity-associated hypogonadism: a randomized, double-blind, placebo-controlled study. Int J Obes (Lond) 2018; 42(5):953–963. doi:10.1038/s41366-018-0105-2
  40. Chua ME, Escusa KG, Luna S, Tapia LC, Dofitas B, Morales M. Revisiting oestrogen antagonists (clomiphene or tamoxifen) as medical empiric therapy for idiopathic male infertility: a meta-analysis. Andrology 2013; 1(5):749–757. doi:10.1111/j.2047-2927.2013.00107.x
  41. Westaby D, Ogle SJ, Paradinas FJ, Randell JB, Murray-Lyon IM. Liver damage from long-term methyltestosterone. Lancet 1977; 2(8032):262–263. pmid:69876
  42. Fernández-Balsells MM, Murad MH, Lane M, et al. Clinical review 1: Adverse effects of testosterone therapy in adult men: a systematic review and meta-analysis. J Clin Endocrinol Metab 2010; 95(6):2560–2575. doi:10.1210/jc.2009-2575
  43. Coviello AD, Kaplan B, Lakshman KM, Chen T, Singh AB, Bhasin S. Effects of graded doses of testosterone on erythropoiesis in healthy young and older men. J Clin Endocrinol Metab 2008; 93(3):914–919. doi:10.1210/jc.2007-1692
  44. Ohlander SJ, Varghese B, Pastuszak AW. Erythrocytosis following testosterone therapy. Sex Med Rev 2018; 6(1):77–85. doi:10.1016/j.sxmr.2017.04.001
  45. Jones SD Jr, Dukovac T, Sangkum P, Yafi FA, Hellstrom WJ. Erythrocytosis and polycythemia secondary to testosterone replacement therapy in the aging male. Sex Med Rev 2015; 3(2):101–112. doi:10.1002/smrj.43
  46. Bachman E, Travison TG, Basaria S, et al. Testosterone induces erythrocytosis via increased erythropoietin and suppressed hepcidin: evidence for a new erythropoietin/hemoglobin set point. J Gerontol A Biol Sci Med Sci 2014; 69(6):725–735. doi:10.1093/gerona/glt154
  47. Roy CN, Snyder PJ, Stephens-Shields AJ, et al. Association of testosterone levels with anemia in older men: a controlled clinical trial. JAMA Intern Med 2017; 177(4):480–490. doi:10.1001/jamainternmed.2016.9540
  48. Klap J, Schmid M, Loughlin KR. The relationship between total testosterone levels and prostate cancer: a review of the continuing controversy. J Urol 2015; 193(2):403–413. doi:10.1016/j.juro.2014.07.123
  49. Gilbert SM, Cavallo CB, Kahane H, Lowe FC. Evidence suggesting PSA cutpoint of 2.5 ng/mL for prompting prostate biopsy: review of 36,316 biopsies. Urology 2005; 65(3):549–553. doi:10.1016/j.urology.2004.10.064
  50. Araujo AB, Dixon JM, Suarez EA, Murad MH, Guey LT, Wittert GA. Clinical review: Endogenous testosterone and mortality in men: a systematic review and meta-analysis. J Clin Endocrinol Metab 2011; 96(10):3007–3019. doi:10.1210/jc.2011-1137
  51. Xu L, Freeman G, Cowling BJ, Schooling CM. Testosterone therapy and cardiovascular events among men: a systematic review and meta-analysis of placebo-controlled randomized trials. BMC Med 2013; 11:108. doi:10.1186/1741-7015-11-108
Issue
Cleveland Clinic Journal of Medicine - 86(5)
Issue
Cleveland Clinic Journal of Medicine - 86(5)
Page Number
321-331
Page Number
321-331
Publications
Publications
Topics
Article Type
Display Headline
An obese 48-year-old man with progressive fatigue and decreased libido
Display Headline
An obese 48-year-old man with progressive fatigue and decreased libido
Legacy Keywords
male hypogonadism, testosterone, low T, obesity, decreased libido, erectile dysfunction, ED, sex hormone binding globulin, SHBG, luteinizing hormone, LH, follicle-stimulating hormone, FSH, inhibin B, pituitary, thyroxine, T4, prolactin, ferritin, laboratory testing, cortisol, MOSH syndrome, male obesity-associated secondary hypogonadism, prostate-specific antigen, PSA, prostate cancer, Ala’A Farkouh, Ayman Zayed
Legacy Keywords
male hypogonadism, testosterone, low T, obesity, decreased libido, erectile dysfunction, ED, sex hormone binding globulin, SHBG, luteinizing hormone, LH, follicle-stimulating hormone, FSH, inhibin B, pituitary, thyroxine, T4, prolactin, ferritin, laboratory testing, cortisol, MOSH syndrome, male obesity-associated secondary hypogonadism, prostate-specific antigen, PSA, prostate cancer, Ala’A Farkouh, Ayman Zayed
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Wed, 04/24/2019 - 09:15
Un-Gate On Date
Wed, 04/24/2019 - 09:15
Use ProPublica
CFC Schedule Remove Status
Wed, 04/24/2019 - 09:15
Hide sidebar & use full width
render the right sidebar.
Article PDF Media

Evaluating and managing postural tachycardia syndrome

Article Type
Changed
Wed, 05/01/2019 - 07:59
Display Headline
Evaluating and managing postural tachycardia syndrome

Some people, most of them relatively young women, experience lightheadedness, a racing heart, and other symptoms (but not hypotension) when they stand up, in a condition known as postural tachycardia syndrome (POTS).1 Although not known to shorten life,1 it can be physically and mentally debilitating.2,3 Therapy rarely cures it, but a multifaceted approach can substantially improve quality of life.

This review outlines the evaluation and diagnosis of POTS and provides guidance for a therapy regimen.

HOW IS POTS DEFINED?

POTS is a multifactorial syndrome rather than a specific disease. It is characterized by all of the following1,4–6:

  • An increase in heart rate of ≥ 30 bpm, or ≥ 40 bpm for those under age 19, within 10 minutes of standing from a supine position
  • Sustained tachycardia (> 30 seconds) 
  • Absence of orthostatic hypotension (a fall in blood pressure of ≥ 20/10 mm Hg)
  • Frequent and chronic duration (≥ 6 months).

These features are critical to diagnosis. Hemodynamic criteria in isolation may describe postural tachycardia but are not sufficient to diagnose POTS.

The prevalence of POTS is estimated to be between 0.2% and 1.0%,7 affecting up to 3 million people in the United States. Most cases arise between ages 13 and 50, with a female-to-male ratio of 5:1.8

MANY NAMES, SAME CONDITION

In 1871, Da Costa9 described a condition he called “irritable heart syndrome” that had characteristics similar to those of POTS, including extreme fatigue and exercise intolerance. Decades later, Lewis10 and Wood11 provided more detailed descriptions of the disorder, renaming it “soldier’s heart” or “Da Costa syndrome.” As other cases were documented, more terms arose, including “effort syndrome” and “mitral valve prolapse syndrome.”

In 1982, Rosen and Cryer12 were the first to use the term “postural tachycardia syndrome” for patients with disabling tachycardia upon standing without orthostatic hypotension. In 1986, Fouad et al13 described patients with postural tachycardia, orthostatic intolerance, and a small degree of hypotension as having “idiopathic hypovolemia.”

In 1993, Schondorf and Low14 established the current definition of POTS, leading to increased awareness and research efforts to understand its pathophysiology.

MULTIFACTORIAL PATHOPHYSIOLOGY

During the last 2 decades, several often-overlapping forms of POTS have been recognized, all of which share a final common pathway of sustained orthostatic tachycardia.15–19 In addition, a number of common comorbidities were identified through review of large clinic populations of POTS.20,21

Hypovolemic POTS

Up to 70% of patients with POTS have hypovolemia. The average plasma volume deficit is about 13%, which typically causes only insignificant changes in heart rate and norepinephrine levels while a patient is supine. However, blood pooling associated with upright posture further compromises cardiac output and consequently increases sympathetic nerve activity. Abnormalities in the renin-angiotensin-aldosterone volume regulation system are also suspected to impair sodium retention, contributing to hypovolemia.1,22

Neuropathic POTS

About half of patients with POTS have partial sympathetic denervation (particularly in the lower limbs) and inadequate vasoconstriction upon standing, leading to reduced venous return and stroke volume.17,23 A compensatory increase in sympathetic tone results in tachycardia to maintain cardiac output and blood pressure.

Hyperadrenergic POTS

Up to 50% of patients with POTS have high norepinephrine levels (≥ 600 pg/mL) when upright. This subtype, hyperadrenergic POTS, is characterized by an increase in systolic blood pressure of at least 10 mm Hg within 10 minutes of standing, with concomitant tachycardia that can be similar to or greater than that seen in nonhyperadrenergic POTS. Patients with hyperadrenergic POTS tend to report more prominent symptoms of sympathetic activation, such as palpitations, anxiety, and tremulousness.24,25

Norepinephrine transporter deficiency

The norepinephrine transporter (NET) is on the presynaptic cleft of sympathetic neurons and serves to clear synaptic norepinephrine. NET deficiency leads to a hyperadrenergic state and elevated sympathetic nerve activation.18 NET deficiency may be induced by common antidepressants (eg, tricyclic antidepressants and serotonin-norepinephrine reuptake inhibitors) and attention-deficit disorder medications.4

Mast cell activation syndrome

The relationship between mast cell activation syndrome and POTS is poorly understood.4,26 Mast cell activation syndrome has been described in a subset of patients with POTS who have sinus tachycardia accompanied by severe episodic flushing. Patients with this subtype have a hyperadrenergic response to postural change and elevated urine methylhistamine during flushing episodes.

Patients with mast cell activation syndrome tend to have strong allergic symptoms and may also have severe gastrointestinal problems, food sensitivities, dermatographism, and neuropathy. Diagnosis can be difficult, as the condition is associated with numerous markers with varying sensitivity and specificity.

Autoimmune origin

A significant minority of patients report a viral-like illness before the onset of POTS symptoms, suggesting a possible autoimmune-mediated or inflammatory cause. Also, some autoimmune disorders (eg, Sjögren syndrome) can present with a POTS-like manifestation.

Research into the role of autoantibodies in the pathophysiology of POTS offers the potential to develop novel therapeutic targets. Auto­antibodies that have been reported in POTS include those against M1 to M3 muscarinic receptors (present in over 87% of patients with POTS),27 cardiac lipid raft-associated proteins,28 adrenergic G-protein coupled receptors, alpha-1-adrenergic receptors, and beta-1- and beta-2-adrenergic receptors.29 Although commercial enzyme-linked immunosorbent assays can assess for these antibody fragments, it is not known whether targeting the antibodies improves outcomes. At this time, antibody testing for POTS should be confined to the research setting.

LINKS TO OTHER SYNDROMES

POTS is often associated with other conditions whose symptoms cannot be explained by postural intolerance or tachycardia.

Ehlers-Danlos syndromes are a group of inherited heterogeneous disorders involving joint hypermobility, skin hyperextensibility, and tissue fragility.30 The hypermobile subtype is most commonly associated with POTS, with patients often having symptoms of autonomic dysregulation and autonomic test abnormalities.31–33 Patients with POTS may have a history of joint subluxations, joint pain, cervical instability, and spontaneous epidural leaks. The reason for the overlap between the two syndromes is not clear.

Chronic fatigue syndrome is characterized by persistent fatigue that does not resolve with rest and is not necessarily associated with orthostatic changes. More than 75% of patients with POTS report general fatigue as a major complaint, and up to 23% meet the full criteria for chronic fatigue syndrome.34

 

 

DIAGNOSTIC STRATEGY

A patient presenting with symptoms suggestive of POTS should first undergo a detailed history and physical examination. Other causes of sinus tachycardia should be considered. 

Detailed history, symptom review

The history should focus on determining symptom burden, including tachycardia onset, frequency, severity, and triggers; the presence of syncope; and the impact of symptoms on daily function and quality of life.

lei_posturaltachycardiasyndrome_t1.jpg
POTS-associated orthostatic intolerance manifests with cardiac and noncardiac symptoms (Table 1).

Presyncope and its associated symptoms occur in less than one-third of patients with POTS, and syncope is not a principal feature.4 If syncope is the predominant complaint, alternative causes should be investigated. The usual cause of syncope in the general population is thought to be vasovagal.

In addition to orthostatic intolerance, gastrointestinal disturbances are common in POTS, presenting as abdominal pain, heartburn, irregular bowel movements, diarrhea, or constipation. Symptoms of gastroparesis are less common. Gastrointestinal symptoms tend to be prolonged, lasting hours and occurring multiple times a week. They tend not to improve in the supine position.35 

POTS-associated symptoms may develop insidiously, but patients often report onset after an acute stressor such as pregnancy, major surgery, or a presumed viral illness.4 Whether these putative triggers are causative or coincidental is unknown. Symptoms of orthostatic intolerance tend to be exacerbated by dehydration, heat, alcohol, exercise, and menstruation.36,37

Consider the family history: 1 in 8 patients with POTS reports familial orthostatic intolerance,38 suggesting a genetic role in some patients. Inquire about symptoms or a previous diagnosis of Ehlers-Danlos syndrome and mast cell activation syndrome.

Consider other conditions

lei_posturaltachycardiasyndrome_t2.jpg
Other causes of orthostatic tachycardia are listed in Table 2.39–41 Most can be diagnosed with a careful history, physical examination, and laboratory tests. Two of the more challenging diagnoses are described below. 

Pheochromocytoma causes hyperadrenergic symptoms (eg, palpitations, lightheadedness) like those in POTS, but patients with pheochromocytoma typically have these symptoms while supine. Pheochromocytoma is also characterized by plasma norepinephrine levels much higher than in POTS.4 Plasma metanephrine testing helps diagnose or rule out pheochromocytoma.5

Inappropriate sinus tachycardia, like pheochromocytoma, also has clinical features similar to those of POTS, as well as tachycardia present when supine. It involves higher sympathetic tone and lower parasympathetic tone compared with POTS; patients commonly have a daytime resting heart rate of at least 100 bpm or a 24-hour mean heart rate of at least 90 bpm.1,42 While the intrinsic heart rate is heightened in inappropriate sinus tachycardia, it is not different between POTS patients and healthy individuals.42,43 Distinguishing POTS from inappropriate sinus tachycardia is further complicated by the broad inclusion criteria of most studies of inappropriate sinus tachycardia, which failed to exclude patients with POTS.44 The Heart Rhythm Society recently adopted distinct definitions for the 2 conditions.1

Physical examination: Focus on vital signs

lei_posturaltachycardiasyndrome_f1.jpg
%3Cp%3EFigure%201.%20Results%20of%20head-up%20tilt-table%20(HUT)%20testing%20in%20a%20healthy%20person%20(top)%20and%20in%20a%20patient%20with%20postural%20tachycardia%20syndrome%20(POTS)%20(bottom).%20Upon%20passive%20head-up%20tilting%2C%20the%20heart%20rate%20increases%20in%20POTS%20by%20at%20least%2030%20bpm%20but%20remains%20largely%20stable%20in%20healthy%20individuals.%20Orthostatic%20hypotension%20(a%20fall%20in%20blood%20pressure%20of%20%E2%89%A5%2020%2F10%20mm%20Hg)%20does%20not%20occur%20in%20either%20patient.%3C%2Fp%3E
The most critical component of the physical examination is thorough measurement of orthostatic vital signs (Figure 1). Blood pressure and heart rate should be measured while the patient has been supine for at least 5 minutes, and again after being upright for 1, 3, 5, and 10 minutes. These measurements determine if orthostatic hypotension is present and whether the patient meets the heart rate criteria for POTS. Patients with POTS tend to experience greater orthostatic tachycardia in the morning, so evaluation early in the day optimizes diagnostic sensitivity.5

Dependent acrocyanosis—dark red-blue discoloration of the lower legs that is cold to the touch—occurs in about half of patients with POTS upon standing.4 Dependent acrocyanosis is associated with joint hypermobility and Ehlers-Danlos syndrome, so these conditions should also be considered if findings are positive.

Laboratory testing for other causes

Laboratory testing is used mainly to detect primary causes of sinus tachycardia. Tests should include:

  • Complete blood cell count with hematocrit (for severe anemia)
  • Thyroid-stimulating hormone level (for hyperthyroidism)
  • Electrolyte panel (for significant electrolyte disturbances).

Evidence is insufficient to support routinely measuring the vitamin B12 level, iron indices, and serum markers for celiac disease, although these may be done if the history or physical examination suggests related problems.4 Sicca symptoms (severe dry eye or dry mouth) should trigger evaluation for Sjögren syndrome.

Electrocardiography needed

Electrocardiography should be performed to investigate for cardiac conduction abnormalities as well as for resting markers of a supraventricular tachyarrhythmia. Extended ambulatory (Holter) monitoring may be useful to evaluate for a transient reentrant tachyarrhythmia4; however, it does not record body position, so it can be difficult to determine if detected episodes of tachycardia are related to posture.

Additional testing for select cases

Further investigation is usually not needed to diagnose POTS but should be considered in some cases. Advanced tests are typically performed at a tertiary care referral center and include: 


  • Quantitative sensory testing to evaluate for small-fiber neuropathy (ie, Quantitative Sudomotor Axon Reflex Test, or QSART), which occurs in the neuropathic POTS subtype
  • Formal autonomic function testing to characterize neurovascular responsiveness  
  • Supine and standing plasma norepinephrine levels (fractionated catecholamines) to characterize the net activation of the sympathetic nervous system
  • Blood volume assessments to assess hypovolemia 
  • Formal exercise testing to objectively quantify exercise capacity.

 

 

GRADED MANAGEMENT

No single universal gold-standard therapy exists for POTS, and management should be individually determined with the primary goals of treating symptoms and restoring function. A graded approach should be used, starting with conservative nonpharmacologic therapies and adding medications as needed.

While the disease course varies substantially from patient to patient, proper management is strongly associated with eventual symptom improvement.1

NONPHARMACOLOGIC STEPS FIRST

lei_posturaltachycardiasyndrome_t3.jpg
A multipronged nonpharmacologic approach should be used for all patients before resorting to medications (Table 3). In an observational study, most patients reported that such interventions were more helpful than medications.45 The following elements are recommended:

Education

Patients should be informed of the nature of their condition and referred to appropriate healthcare personnel. POTS is a chronic illness requiring individualized coping strategies, intensive physician interaction, and support of a multidisciplinary team. Patients and family members can be reassured that most symptoms improve over time with appropriate diagnosis and treatment.1 Patients should be advised to avoid aggravating triggers and activities.

Exercise

Exercise programs are encouraged but should be introduced gradually, as physical activity can exacerbate symptoms, especially at the outset. Several studies have reported benefits from a short-term (3-month) program, in which the patient gradually progresses from non-upright exercise (eg, rowing machine, recumbent cycle, swimming) to upright endurance exercises. At the end of these programs, significant cardiac remodeling, improved quality of life, and reduced heart rate responses to standing have been reported, and benefits have been reported to persist in patients who continued exercising after the 3-month study period.46,47

Despite the benefits of exercise interventions, compliance is low.46,47 To prevent early discouragement, patients should be advised that it can take 4 to 6 weeks of continued exercise before benefits appear. Patients are encouraged to exercise every other day for 30 minutes or more. Regimens should primarily focus on aerobic conditioning, but resistance training, concentrating on thigh muscles, can also help. Exercise is a treatment and not a cure, and benefits can rapidly disappear if regular activity (at least 3 times per week) is stopped.48

Compression stockings

Compression stockings help reduce peripheral venous pooling and enhance venous return to the heart. Waist-high stockings with compression of at least 30 to 40 mm Hg offer the best results. 

Diet

Increased fluid and salt intake is advisable for patients with suspected hypovolemia. At least 2 to 3 L of water accompanied by 10 to 12 g of daily sodium intake is recommended.1 This can usually be accomplished with diet and salt added to food, but salt tablets can be used if the patient prefers. The resultant plasma volume expansion may help reduce the reflex tachycardia upon standing.49

Check medications

lei_posturaltachycardiasyndrome_t4.jpg
The clinician should review—and perhaps discontinue—medications the patient is already taking that may exacerbate tachycardia or related symptoms (Table 4).50 Venodilators decrease preload, thereby reducing cardiac output and blood pressure, which triggers compensatory tachycardia. Diuretics can reduce effective blood volume and lower preload, leading to worsened symptoms mediated by hypovolemia.

Rescue therapy with saline infusion

Intravenous saline infusion can augment blood volume in patients who are clinically decompensated and present with severe symptoms.1 Intermittent infusion of 1 L of normal saline has been found to significantly reduce orthostatic tachycardia and related symptoms in patients with POTS, contributing to improved quality of life.51,52

Chronic saline infusions are not recommended for long-term care because of the risk of access complications and infection.1 Moak et al53 reported a high rate of bacteremia in a cohort of children with POTS with regular saline infusions, most of whom had a central line. On the other hand, Ruzieh et al54 reported significantly improved symptoms with regular saline infusions without a high rate of complications, but patients in this study received infusions for only a few months and through a peripheral intravenous catheter.

 

 

DRUG THERAPY

lei_posturaltachycardiasyndrome_t5.jpg
Drug therapy for POTS should be used only if nonpharmacologic interventions do not adequately relieve symptoms. Given the heterogeneity of POTS, treatment should be tailored to the patient’s underlying pathophysiology, key clinical features, and comorbidities. These considerations should guide the initial selection of medications, with adjustments as needed to alleviate adverse effects (Table 5).

No medications are approved by the US Food and Drug Administration (FDA) or Health Canada specifically for treating POTS, making all pharmacologic recommendations off-label. Although the drugs discussed below have been evaluated for POTS in controlled laboratory settings, they have yet to be tested in robust clinical trials.

Blood volume expansion

Several drugs expand blood volume, which may reduce orthostatic tachycardia.

Fludrocortisone is a synthetic aldosterone analogue that enhances sodium and water retention. Although one observational study found that it normalizes hemodynamic changes in response to orthostatic stress, no high-level evidence exists for its effectiveness for POTS.55 It is generally well tolerated, although possible adverse effects include hyperkalemia, hypertension, fatigue, nausea, headache, and edema.5,56

Desmopressin is a synthetic version of a natural antidiuretic hormone that increases kidney-mediated free-water reabsorption without sodium retention. It significantly reduces upright heart rate in patients with POTS and improves symptom burden. Although potential adverse effects include edema and headache, hyponatremia is the primary concern with daily use, especially with the increased water intake advised for POTS.57 Patients should be advised to use desmopressin no more than once a week for the acute improvement of symptoms. Intermittent monitoring of serum sodium levels is recommended for safety.

Erythropoietin replacement has been suggested for treating POTS to address the significant deficit in red blood cell volume. Although erythropoietin therapy has a direct vasoconstrictive effect and largely improves red blood cell volume in patients with POTS, it does not expand plasma volume, so orthostatic tachycardia is not itself reduced.22 Nevertheless, it may significantly improve POTS symptoms refractory to more common methods of treatment, and it should be reserved for such cases. In addition to the lack of effect on orthostatic tachycardia, drawbacks to using erythropoietin include its high cost, the need for subcutaneous administration, and the risk of life-threatening complications such as myocardial infarction and stroke.58,59

Heart rate-lowering agents

Propranolol, a nonselective beta-adrenergic antagonist, can significantly reduce standing heart rate and improve symptoms at low dosages (10–20 mg). Higher dosages can further restrain orthostatic tachycardia but are not as well tolerated, mainly due to hypotension and worsening of existing symptoms such as fatigue.60 Regular-acting propranolol works for about 4 to 5 hours per dose, so full-day coverage often requires dosing 4 times per day.

Ivabradine is a selective blocker of the  “funny” (If) channel that reduces the sinus node firing rate without affecting blood pressure, so it slows heart rate without causing supine hypertension or orthostatic hypotension.

A retrospective case series found that 60% of patients with POTS treated with ivabradine reported symptomatic improvement, and all patients experienced reduced tachycardia with continued use.61 Ivabradine has not been compared with placebo or propranolol in a randomized controlled trial, and it has not been well studied in pregnancy and so should be avoided because of potential teratogenic effects.

When prescribing ivabradine for women of childbearing age, a negative pregnancy test may be documented prior to initiation of therapy, and the use of highly effective methods of contraception is recommended. Ivabradine should be avoided in women contemplating pregnancy. Insurance coverage can limit access to ivabradine in the United States.

Central nervous system sympatholytics

Patients with prominent hyperadrenergic features may benefit from central sympatholytic agents. However, these drugs may not be well tolerated in patients with neuropathic POTS because of the effects of reduced systemic vascular resistance5 and the possible exacerbation of drowsiness, fatigue, and mental clouding.4 Patients can be extremely sensitive to these medications, so they should initially be prescribed at the lowest dose, then gradually increased as tolerated.

Clonidine, an alpha-2-adrenergic agonist, decreases central sympathetic tone. In hyperadrenergic patients, clonidine can stabilize heart rate and blood pressure, thereby reducing orthostatic symptoms.62

Methyldopa has effects similar to those of clonidine but is easier to titrate owing to its longer half-life.63 Methyldopa is typically started at 125 mg at bedtime and increased to 125 mg twice daily, if tolerated.             

 

 

Other agents

Midodrine is a prodrug. The active form, an alpha-1-adrenergic agonist, constricts peripheral veins and arteries to increase vascular resistance and venous return, thereby reducing orthostatic tachycardia.52 It is most useful in patients with impaired peripheral vasoconstriction (eg, neuropathic POTS) and may be less effective in those with hyperadrenergic POTS.64 Major limitations of midodrine include worsening supine hypertension and possible urinary retention.39

Because of midodrine’s short half-life, frequent dosing is required during daytime hours (eg, 8 AM, noon, and 4 PM), but it should not be taken within 4 to 5 hours of sleep because of the risk of supine hypertension. Midodrine is typically started at 2.5 to 5 mg per dose and can be titrated up to 15 mg per dose.

Midodrine is an FDA pregnancy category  C drug (adverse effects in pregnancy seen in animal models, but evidence lacking in humans). While ideally it should be avoided, we have used it safely in pregnant women with disabling POTS symptoms.

Pyridostigmine, an acetylcholinesterase inhibitor, increases cardiovagal tone and possibly sympathetic tone. It has been reported to significantly reduce standing heart rate and improve symptom burden in patients with POTS.65 However, pyridostigmine increases gastrointestinal mobility, leading to severe adverse effects in over 20% of patients, including abdominal cramps, nausea, and diarrhea.66

Droxidopa, a synthetic amino acid precursor of norepinephrine, improves dizziness and fatigue in POTS with minimal effects on blood pressure.67

Modafinil, a psychostimulant, may improve POTS-associated cognitive symptoms.4 It also raises upright blood pressure without significantly worsening standing heart rate or acute orthostatic symptoms.68

EFFECTS OF COMORBID DISORDERS ON MANAGEMENT

Ehlers-Danlos syndrome

Pharmacologic approaches to POTS should not be altered based on the presence of Ehlers-Danlos syndrome, but because many of these patients are prone to joint dislocation, exercise prescriptions may need adjusting.

A medical genetics consult is recommended for patients with Ehlers-Danlos syndrome. Although the hypermobile type (the form most commonly associated with POTS) is not associated with aortopathy, it can be confused with classical and vascular Ehlers-Danlos syndromes, which require serial aortic screening.30

Mast cell activation syndrome

Consultation with an allergist or immunologist may help patients with severe symptoms.

Autoantibodies and autoimmunity

Treatment of the underlying disorder is recommended and can result in significantly improved POTS symptoms.

SPECIALTY CARE REFERRAL

POTS can be challenging to manage. Given the range of physiologic, emotional, and functional distress patients experience, it often requires significant physician time and multidisciplinary care. Patients with continued severe or debilitating symptoms may benefit from referral to a tertiary-care center with experience in autonomic nervous system disorders.

PROGNOSIS

Limited data are available on the long-term prognosis of POTS, and more studies are needed in pediatric and adult populations. No deaths have been reported in the handful of published cases of POTS in patients older than 50.1 Some pediatric studies suggest that some teenagers “outgrow” their POTS. However, these data are not robust, and an alternative explanation is that as they get older, they see adult physicians for their POTS symptoms and so are lost to study follow-up.6,44,69 

We have not often seen POTS simply resolve without ongoing treatment. However, in our experience, most patients have improved symptoms and function with multimodal treatment (ie, exercise, salt, water, stockings, and some medications) and time.

References
  1. Sheldon RS, Grubb BP 2nd, Olshansky B, et al. 2015 Heart Rhythm Society expert consensus statement on the diagnosis and treatment of postural tachycardia syndrome, inappropriate sinus tachycardia, and vasovagal syncope. Heart Rhythm 2015; 12(6):e41–e63. doi:10.1016/j.hrthm.2015.03.029
  2. Bagai K, Song Y, Ling JF, et al. Sleep disturbances and diminished quality of life in postural tachycardia syndrome. J Clin Sleep Med 2011; 7(2):204–210. pmid:21509337
  3. Benrud-Larson LM, Dewar MS, Sandroni P, Rummans TA, Haythornthwaite JA, Low PA. Quality of life in patients with postural tachycardia syndrome. Mayo Clin Proc 2002; 77(6):531–537. doi:10.4065/77.6.531
  4. Raj SR. Postural tachycardia syndrome (POTS). Circulation 2013; 127(23):2336–2342. doi:10.1161/CIRCULATIONAHA.112.144501
  5. Raj SR. The postural tachycardia syndrome (POTS): pathophysiology, diagnosis & management. Indian Pacing Electrophysiol J 2006; 6(2):84–99. pmid:16943900
  6. Singer W, Sletten DM, Opfer-Gehrking TL, Brands CK, Fischer PR, Low PA. Postural tachycardia in children and adolescents: what is abnormal? J Pediatr 2012; 160(2):222–226. doi:10.1016/j.jpeds.2011.08.054
  7. Mar PL, Raj SR. Neuronal and hormonal perturbations in postural tachycardia syndrome. Front Physiol 2014; 5:220. doi:10.3389/fphys.2014.00220
  8. Garland EM, Raj SR, Black BK, Harris PA, Robertson D. The hemodynamic and neurohumoral phenotype of postural tachycardia syndrome. Neurology 2007; 69(8):790–798. doi:10.1212/01.wnl.0000267663.05398.40
  9. Da Costa JM. On irritable heart: a clinical study of a form of functional cardiac disorder and its consequences. Am J Med Sci 1871; 61(121):2–52.
  10. Lewis T. The tolerance of physical exertion, as shown by soldiers suffering from so-called “irritable heart.” Br Med J 1918; 1(2987):363–365. pmid:20768980
  11. Wood P. Da Costa’s syndrome (or effort syndrome): lecture I. Br Med J 1941; 1(4194):767–772. pmid:20783672
  12. Rosen SG, Cryer PE. Postural tachycardia syndrome. Reversal of sympathetic hyperresponsiveness and clinical improvement during sodium loading. Am J Med 1982; 72(5):847–850.
  13. Fouad FM, Tadena-Thome L, Bravo EL, Tarazi RC. Idiopathic hypovolemia. Ann Intern Med 1986; 104(3):298–303. pmid:3511818
  14. Schondorf R, Low PA. Idiopathic postural orthostatic tachycardia syndrome: an attenuated form of acute pandysautonomia? Neurology 1993; 43(1):132–137. pmid:8423877
  15. Vernino S, Low PA, Fealey RD, Stewart JD, Farrugia G, Lennon VA. Autoantibodies to ganglionic acetylcholine receptors in autoimmune autonomic neuropathies. N Engl J Med 2000; 343(12):847–855. doi:10.1056/NEJM200009213431204
  16. Raj SR, Robertson D. Blood volume perturbations in the postural tachycardia syndrome. Am J Med Sci 2007; 334(1):57–60. doi:10.1097/MAJ.0b013e318063c6c0
  17. Jacob G, Costa F, Shannon JR, et al. The neuropathic postural tachycardia syndrome. N Engl J Med 2000; 343(14):1008–1014. doi:10.1056/NEJM200010053431404
  18. Shannon JR, Flattem NL, Jordan J, et al. Orthostatic intolerance and tachycardia associated with norepinephrine-transporter deficiency. N Engl J Med 2000; 342(8):541–549. doi:10.1056/NEJM200002243420803
  19. Jones PK, Shaw BH, Raj SR. Clinical challenges in the diagnosis and management of postural tachycardia syndrome. Pract Neurol 2016; 16(6):431–438. doi:10.1136/practneurol-2016-001405
  20. Gunning WT, Karabin BL, Blomquist TM, Grubb BP. Postural orthostatic tachycardia syndrome is associated with platelet storage pool deficiency. Medicine (Baltimore) 2016; 95(37):e4849. doi:10.1097/MD.0000000000004849
  21. Kanjwal K, Sheikh M, Karabin B, Kanjwal Y, Grubb BP. Neurocardiogenic syncope coexisting with postural orthostatic tachycardia syndrome in patients suffering from orthostatic intolerance: a combined form of autonomic dysfunction. Pacing Clin Electrophysiol 2011; 34(5):549–554. doi:10.1111/j.1540-8159.2010.02994.x
  22. Raj SR, Biaggioni I, Yamhure PC, et al. Renin-aldosterone paradox and perturbed blood volume regulation underlying postural tachycardia syndrome. Circulation 2005; 111(13):1574–1582. doi:10.1161/01.CIR.0000160356.97313.5D
  23. Gibbons CH, Bonyhay I, Benson A, Wang N, Freeman R. Structural and functional small fiber abnormalities in the neuropathic postural tachycardia syndrome. PLoS One 2013; 8(12):e84716. doi:10.1371/journal.pone.0084716
  24. Low PA, Sandroni P, Joyner M, Shen WK. Postural tachycardia syndrome (POTS). J Cardiovasc Electrophysiol 2009; 20(3):352–358. doi:10.1111/j.1540-8167.2008.01407.x
  25. Kanjwal K, Saeed B, Karabin B, Kanjwal Y, Grubb BP. Clinical presentation and management of patients with hyperadrenergic postural orthostatic tachycardia syndrome. A single center experience. Cardiol J 2011; 18(5):527–531. pmid:21947988
  26. Shibao C, Arzubiaga C, Roberts J, et al. Hyperadrenergic postural tachycardia syndrome in mast cell activation disorders. Hypertension 2005; 45(3):385–390. doi:10.1161/01.HYP.0000158259.68614.40
  27. Dubey D, Hopkins S, Vernino S. M1 and M2 muscarinic receptor antibodies among patients with postural orthostatic tachycardia syndrome: potential disease biomarker [abstract]. J Clin Neuromuscul Dis 2016; 17(3):179S.
  28. Wang XL, Ling TY, Charlesworth MC, et al. Autoimmunoreactive IgGs against cardiac lipid raft-associated proteins in patients with postural orthostatic tachycardia syndrome. Transl Res 2013; 162(1):34–44. doi:10.1016/j.trsl.2013.03.002
  29. Li H, Yu X, Liles C, et al. Autoimmune basis for postural tachycardia syndrome. J Am Heart Assoc 2014; 3(1):e000755. doi:10.1161/JAHA.113.000755
  30. Malfait F, Francomano C, Byers P, et al. The 2017 international classification of the Ehlers-Danlos syndromes. Am J Med Genet C Semin Med Genet 2017; 175(1):8–26. doi:10.1002/ajmg.c.31552
  31. Wallman D, Weinberg J, Hohler AD. Ehlers-Danlos syndrome and postural tachycardia syndrome: a relationship study. J Neurol Sci 2014; 340(1-2):99–102. doi:10.1016/j.jns.2014.03.002
  32. De Wandele I, Calders P, Peersman W, et al. Autonomic symptom burden in the hypermobility type of Ehlers-Danlos syndrome: a comparative study with two other EDS types, fibromyalgia, and healthy controls. Semin Arthritis Rheum 2014; 44(3):353–361. doi:10.1016/j.semarthrit.2014.05.013
  33. Gazit Y, Nahir AM, Grahame R, Jacob G. Dysautonomia in the joint hypermobility syndrome. Am J Med 2003; 115(1):33–40. pmid:12867232
  34. Okamoto LE, Raj SR, Peltier A, et al. Neurohumoral and haemodynamic profile in postural tachycardia and chronic fatigue syndromes. Clin Sci (Lond) 2012; 122(4):183–192. doi:10.1042/CS20110200
  35. Wang LB, Culbertson CJ, Deb A, Morgenshtern K, Huang H, Hohler AD. Gastrointestinal dysfunction in postural tachycardia syndrome. J Neurol Sci 2015; 359(1-2):193–196. doi:10.1016/j.jns.2015.10.052
  36. Raj S, Sheldon R. Management of postural tachycardia syndrome, inappropriate sinus tachycardia and vasovagal syncope. Arrhythm Electrophysiol Rev 2016; 5(2):122–129. doi:10.15420/AER.2016.7.2
  37. Peggs KJ, Nguyen H, Enayat D, Keller NR, Al-Hendy A, Raj SR. Gynecologic disorders and menstrual cycle lightheadedness in postural tachycardia syndrome. Int J Gynaecol Obstet 2012; 118(3):242–246. doi:10.1016/j.ijgo.2012.04.014
  38. Thieben MJ, Sandroni P, Sletten DM, et al. Postural orthostatic tachycardia syndrome: the Mayo Clinic experience. Mayo Clin Proc 2007; 82(3):308–313. doi:10.4065/82.3.308
  39. Deb A, Morgenshtern K, Culbertson CJ, Wang LB, Hohler AD. A survey-based analysis of symptoms in patients with postural orthostatic tachycardia syndrome. J Am Coll Cardiol 2015; 28(7):157–159. pmid:25829642
  40. Ertek S, Cicero AF. Hyperthyroidism and cardiovascular complications: a narrative review on the basis of pathophysiology. Arch Med Sci 2013; 9(5):944–952. doi:10.5114/aoms.2013.38685
  41. Rangno RE, Langlois S. Comparison of withdrawal phenomena after propranolol, metoprolol and pindolol. Br J Clin Pharmacol 1982; 13(suppl 2):345S–351S. pmid:6125187
  42. Nwazue VC, Paranjape SY, Black BK, et al. Postural tachycardia syndrome and inappropriate sinus tachycardia: role of autonomic modulation and sinus node automaticity. J Am Heart Assoc 2014; 3(2):e000700. doi:10.1161/JAHA.113.000700
  43. Morillo CA, Klein GJ, Thakur RK, Li H, Zardini M, Yee R. Mechanism of “inappropriate” sinus tachycardia. Role of sympathovagal balance. Circulation 1994; 90(2):873–877. pmid:7913886
  44. Grubb BP. Postural tachycardia syndrome. Circulation 2008; 117(21):2814–2817. doi:10.1161/CIRCULATIONAHA.107.761643
  45. Bhatia R, Kizilbash SJ, Ahrens SP, et al. Outcomes of adolescent-onset postural orthostatic tachycardia syndrome. J Pediatr 2016; 173:149–153. doi:10.1016/j.jpeds.2016.02.035
  46. George SA, Bivens TB, Howden EJ, et al. The international POTS registry: evaluating the efficacy of an exercise training intervention in a community setting. Heart Rhythm 2016; 13(4):943–950. doi:10.1016/j.hrthm.2015.12.012
  47. Fu Q, VanGundy TB, Galbreath MM, et al. Cardiac origins of the postural orthostatic tachycardia syndrome. J Am Coll Cardiol 2010; 55(25):2858–2868. doi:10.1016/j.jacc.2010.02.043
  48. Raj SR. Row, row, row your way to treating postural tachycardia syndrome. Heart Rhythm 2016; 13(4):951–952. doi:10.1016/j.hrthm.2015.12.039
  49. Celedonio JE, Garland EM, Nwazue VC, et al. Effects of high sodium intake on blood volume and catecholamines in patients with postural tachycardia syndrome and healthy females [abstract]. Clin Auton Res 2014; 24:211.
  50. Garland EM, Celedonio JE, Raj SR. Postural tachycardia syndrome: beyond orthostatic intolerance. Curr Neurol Neurosci Rep 2015; 15(9):60. doi:10.1007/s11910-015-0583-8
  51. Gordon VM, Opfer-Gehrking TL, Novak V, Low PA. Hemodynamic and symptomatic effects of acute interventions on tilt in patients with postural tachycardia syndrome. Clin Auton Res 2000; 10:29–33. pmid:10750641
  52. Jacob G, Shannon JR, Black B, et al. Effects of volume loading and pressor agents in idiopathic orthostatic tachycardia. Circulation 1997; 96(2):575–580. pmid:9244228
  53. Moak JP, Leong D, Fabian R, et al. Intravenous hydration for management of medication-resistant orthostatic intolerance in the adolescent and young adult. Pediatr Cardiol 2016; 37(2):278–282. doi:10.1007/s00246-015-1274-6
  54. Ruzieh M, Baugh A, Dasa O, et al. Effects of intermittent intravenous saline infusions in patients with medication-refractory postural tachycardia syndrome. J Interv Card Electrophysiol 2017; 48(3):255–260. doi:10.1007/s10840-017-0225-y
  55. Freitas J, Santos R, Azevedo E, Costa O, Carvalho M, de Freitas AF. Clinical improvement in patients with orthostatic intolerance after treatment with bisoprolol and fludrocortisone. Clin Auton Res 2000; 10(5):293–299. pmid:11198485
  56. Lee AK, Krahn AD. Evaluation of syncope: focus on diagnosis and treatment of neurally mediated syncope. Expert Rev Cardiovasc Ther 2016; 14(6):725–736. doi:10.1586/14779072.2016.1164034
  57. Coffin ST, Black BK, Biaggioni I, et al. Desmopressin acutely decreases tachycardia and improves symptoms in the postural tachycardia syndrome. Heart Rhythm 2012; 9(9):1484–1490. doi:10.1016/j.hrthm.2012.05.002
  58. Kanjwal K, Saeed B, Karabin B, Kanjwal Y, Sheikh M, Grubb BP. Erythropoietin in the treatment of postural orthostatic tachycardia syndrome. Am J Ther 2012; 19(2):92–95. doi:10.1097/MJT.0b013e3181ef621a
  59. Hoeldtke RD, Horvath GG, Bryner KD. Treatment of orthostatic tachycardia with erythropoietin. Am J Med 1995; 99(5):525–529. pmid:7485211
  60. Raj SR, Black BK, Biaggioni I, et al. Propranolol decreases tachycardia and improves symptoms in the postural tachycardia syndrome: less is more. Circulation 2009; 120(9):725–734. doi:10.1161/CIRCULATIONAHA.108.846501
  61. McDonald C, Frith J, Newton JL. Single centre experience of ivabradine in postural orthostatic tachycardia syndrome. Europace 2011; 13(3):427–430. doi:10.1093/europace/euq390
  62. Gaffney FA, Lane LB, Pettinger W, Blomqvist G. Effects of long-term clonidine administration on the hemodynamic and neuroendocrine postural responses of patients with dysautonomia. Chest 1983; 83(suppl 2):436–438. pmid:6295714
  63. Jacob G, Biaggioni I. Idiopathic orthostatic intolerance and postural tachycardia syndromes. Am J Med Sci 1999; 317(2):88–101. pmid:10037112
  64. Ross AJ, Ocon AJ, Medow MS, Stewart JM. A double-blind placebo-controlled cross-over study of the vascular effects of midodrine in neuropathic compared with hyperadrenergic postural tachycardia syndrome. Clin Sci (Lond) 2014; 126(4):289–296. doi:10.1042/CS20130222
  65. Raj SR, Black BK, Biaggioni I, Harris PA, Robertson D. Acetylcholinesterase inhibition improves tachycardia in postural tachycardia syndrome. Circulation 2005; 111(21):2734–2340. doi:10.1161/CIRCULATIONAHA.104.497594
  66. Kanjwal K, Karabin B, Sheikh M, et al. Pyridostigmine in the treatment of postural orthostatic tachycardia: A single-center experience. Pacing Clin Electrophysiol 2011; 34(6):750–755. doi:10.1111/j.1540-8159.2011.03047.x
  67. Ruzieh M, Dasa O, Pacenta A, Karabin B, Grubb B. Droxidopa in the treatment of postural orthostatic tachycardia syndrome. Am J Ther 2017; 24(2):e157–e161. doi:10.1097/MJT.0000000000000468
  68. Kpaeyeh AG Jr, Mar PL, Raj V, et al. Hemodynamic profiles and tolerability of modafinil in the treatment of POTS: a randomized placebo-controlled trial. J Clin Psychopharmacol 2014; 34(6):738–741. doi:10.1097/JCP.0000000000000221
  69. Lai CC, Fischer PR, Brands CK, et al. Outcomes in adolescents with postural orthostatic tachycardia syndrome treated with midodrine and beta-blockers. Pacing Clin Electrophysiol 2009; 32(2):234–238. doi:10.1111/j.1540-8159.2008.02207.x
Article PDF
Author and Disclosure Information

Lucy Y. Lei
Department of Cardiac Sciences, Libin Cardiovascular Institute of Alberta, University of Calgary, Calgary, AB, Canada

Derek S. Chew, MD
Department of Cardiac Sciences, Libin Cardiovascular Institute of Alberta, University of Calgary, Calgary, AB, Canada

Robert S. Sheldon, MD, PhD
Department of Cardiac Sciences, Libin Cardiovascular Institute of Alberta, University of Calgary, Calgary, AB, Canada

Satish R. Raj, MD, MSCI, FRCPC
Department of Cardiac Sciences, Libin Cardiovascular Institute of Alberta, University of Calgary, Calgary, AB, Canada; Autonomic Dysfunction Center, Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 

Address: Satish R. Raj, MD, MSCI, FRCPC, Department of Cardiac Sciences, Libin Cardiovascular Institute of Alberta, University of Calgary, GAC70 HRIC Building, 3280 Hospital Drive NW, Calgary, AB T2N 4Z6, Canada; satish.raj@ucalgary.ca

Dr. Raj has disclosed consulting for Abbott Laboratories, Boston Scientific Corporation, GE Healthcare, and Lundbeck, and serving on the steering committee for the ADMIRE-ICD trial, funded by GE Healthcare.

Issue
Cleveland Clinic Journal of Medicine - 86(5)
Publications
Topics
Page Number
333-344
Legacy Keywords
postural tachycardia syndrome, POTS, autonomic nervous system, hypovolemia, hyperadrenergic, norepinephrine, mast cell activation syndrome, Ehlers-Danlos syndromes, tilt table, chronic fatigue syndrome, syncope, Lucy Lei, Derek Chew, Robert Sheldon, Satish Raj
Sections
Author and Disclosure Information

Lucy Y. Lei
Department of Cardiac Sciences, Libin Cardiovascular Institute of Alberta, University of Calgary, Calgary, AB, Canada

Derek S. Chew, MD
Department of Cardiac Sciences, Libin Cardiovascular Institute of Alberta, University of Calgary, Calgary, AB, Canada

Robert S. Sheldon, MD, PhD
Department of Cardiac Sciences, Libin Cardiovascular Institute of Alberta, University of Calgary, Calgary, AB, Canada

Satish R. Raj, MD, MSCI, FRCPC
Department of Cardiac Sciences, Libin Cardiovascular Institute of Alberta, University of Calgary, Calgary, AB, Canada; Autonomic Dysfunction Center, Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 

Address: Satish R. Raj, MD, MSCI, FRCPC, Department of Cardiac Sciences, Libin Cardiovascular Institute of Alberta, University of Calgary, GAC70 HRIC Building, 3280 Hospital Drive NW, Calgary, AB T2N 4Z6, Canada; satish.raj@ucalgary.ca

Dr. Raj has disclosed consulting for Abbott Laboratories, Boston Scientific Corporation, GE Healthcare, and Lundbeck, and serving on the steering committee for the ADMIRE-ICD trial, funded by GE Healthcare.

Author and Disclosure Information

Lucy Y. Lei
Department of Cardiac Sciences, Libin Cardiovascular Institute of Alberta, University of Calgary, Calgary, AB, Canada

Derek S. Chew, MD
Department of Cardiac Sciences, Libin Cardiovascular Institute of Alberta, University of Calgary, Calgary, AB, Canada

Robert S. Sheldon, MD, PhD
Department of Cardiac Sciences, Libin Cardiovascular Institute of Alberta, University of Calgary, Calgary, AB, Canada

Satish R. Raj, MD, MSCI, FRCPC
Department of Cardiac Sciences, Libin Cardiovascular Institute of Alberta, University of Calgary, Calgary, AB, Canada; Autonomic Dysfunction Center, Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 

Address: Satish R. Raj, MD, MSCI, FRCPC, Department of Cardiac Sciences, Libin Cardiovascular Institute of Alberta, University of Calgary, GAC70 HRIC Building, 3280 Hospital Drive NW, Calgary, AB T2N 4Z6, Canada; satish.raj@ucalgary.ca

Dr. Raj has disclosed consulting for Abbott Laboratories, Boston Scientific Corporation, GE Healthcare, and Lundbeck, and serving on the steering committee for the ADMIRE-ICD trial, funded by GE Healthcare.

Article PDF
Article PDF
Related Articles

Some people, most of them relatively young women, experience lightheadedness, a racing heart, and other symptoms (but not hypotension) when they stand up, in a condition known as postural tachycardia syndrome (POTS).1 Although not known to shorten life,1 it can be physically and mentally debilitating.2,3 Therapy rarely cures it, but a multifaceted approach can substantially improve quality of life.

This review outlines the evaluation and diagnosis of POTS and provides guidance for a therapy regimen.

HOW IS POTS DEFINED?

POTS is a multifactorial syndrome rather than a specific disease. It is characterized by all of the following1,4–6:

  • An increase in heart rate of ≥ 30 bpm, or ≥ 40 bpm for those under age 19, within 10 minutes of standing from a supine position
  • Sustained tachycardia (> 30 seconds) 
  • Absence of orthostatic hypotension (a fall in blood pressure of ≥ 20/10 mm Hg)
  • Frequent and chronic duration (≥ 6 months).

These features are critical to diagnosis. Hemodynamic criteria in isolation may describe postural tachycardia but are not sufficient to diagnose POTS.

The prevalence of POTS is estimated to be between 0.2% and 1.0%,7 affecting up to 3 million people in the United States. Most cases arise between ages 13 and 50, with a female-to-male ratio of 5:1.8

MANY NAMES, SAME CONDITION

In 1871, Da Costa9 described a condition he called “irritable heart syndrome” that had characteristics similar to those of POTS, including extreme fatigue and exercise intolerance. Decades later, Lewis10 and Wood11 provided more detailed descriptions of the disorder, renaming it “soldier’s heart” or “Da Costa syndrome.” As other cases were documented, more terms arose, including “effort syndrome” and “mitral valve prolapse syndrome.”

In 1982, Rosen and Cryer12 were the first to use the term “postural tachycardia syndrome” for patients with disabling tachycardia upon standing without orthostatic hypotension. In 1986, Fouad et al13 described patients with postural tachycardia, orthostatic intolerance, and a small degree of hypotension as having “idiopathic hypovolemia.”

In 1993, Schondorf and Low14 established the current definition of POTS, leading to increased awareness and research efforts to understand its pathophysiology.

MULTIFACTORIAL PATHOPHYSIOLOGY

During the last 2 decades, several often-overlapping forms of POTS have been recognized, all of which share a final common pathway of sustained orthostatic tachycardia.15–19 In addition, a number of common comorbidities were identified through review of large clinic populations of POTS.20,21

Hypovolemic POTS

Up to 70% of patients with POTS have hypovolemia. The average plasma volume deficit is about 13%, which typically causes only insignificant changes in heart rate and norepinephrine levels while a patient is supine. However, blood pooling associated with upright posture further compromises cardiac output and consequently increases sympathetic nerve activity. Abnormalities in the renin-angiotensin-aldosterone volume regulation system are also suspected to impair sodium retention, contributing to hypovolemia.1,22

Neuropathic POTS

About half of patients with POTS have partial sympathetic denervation (particularly in the lower limbs) and inadequate vasoconstriction upon standing, leading to reduced venous return and stroke volume.17,23 A compensatory increase in sympathetic tone results in tachycardia to maintain cardiac output and blood pressure.

Hyperadrenergic POTS

Up to 50% of patients with POTS have high norepinephrine levels (≥ 600 pg/mL) when upright. This subtype, hyperadrenergic POTS, is characterized by an increase in systolic blood pressure of at least 10 mm Hg within 10 minutes of standing, with concomitant tachycardia that can be similar to or greater than that seen in nonhyperadrenergic POTS. Patients with hyperadrenergic POTS tend to report more prominent symptoms of sympathetic activation, such as palpitations, anxiety, and tremulousness.24,25

Norepinephrine transporter deficiency

The norepinephrine transporter (NET) is on the presynaptic cleft of sympathetic neurons and serves to clear synaptic norepinephrine. NET deficiency leads to a hyperadrenergic state and elevated sympathetic nerve activation.18 NET deficiency may be induced by common antidepressants (eg, tricyclic antidepressants and serotonin-norepinephrine reuptake inhibitors) and attention-deficit disorder medications.4

Mast cell activation syndrome

The relationship between mast cell activation syndrome and POTS is poorly understood.4,26 Mast cell activation syndrome has been described in a subset of patients with POTS who have sinus tachycardia accompanied by severe episodic flushing. Patients with this subtype have a hyperadrenergic response to postural change and elevated urine methylhistamine during flushing episodes.

Patients with mast cell activation syndrome tend to have strong allergic symptoms and may also have severe gastrointestinal problems, food sensitivities, dermatographism, and neuropathy. Diagnosis can be difficult, as the condition is associated with numerous markers with varying sensitivity and specificity.

Autoimmune origin

A significant minority of patients report a viral-like illness before the onset of POTS symptoms, suggesting a possible autoimmune-mediated or inflammatory cause. Also, some autoimmune disorders (eg, Sjögren syndrome) can present with a POTS-like manifestation.

Research into the role of autoantibodies in the pathophysiology of POTS offers the potential to develop novel therapeutic targets. Auto­antibodies that have been reported in POTS include those against M1 to M3 muscarinic receptors (present in over 87% of patients with POTS),27 cardiac lipid raft-associated proteins,28 adrenergic G-protein coupled receptors, alpha-1-adrenergic receptors, and beta-1- and beta-2-adrenergic receptors.29 Although commercial enzyme-linked immunosorbent assays can assess for these antibody fragments, it is not known whether targeting the antibodies improves outcomes. At this time, antibody testing for POTS should be confined to the research setting.

LINKS TO OTHER SYNDROMES

POTS is often associated with other conditions whose symptoms cannot be explained by postural intolerance or tachycardia.

Ehlers-Danlos syndromes are a group of inherited heterogeneous disorders involving joint hypermobility, skin hyperextensibility, and tissue fragility.30 The hypermobile subtype is most commonly associated with POTS, with patients often having symptoms of autonomic dysregulation and autonomic test abnormalities.31–33 Patients with POTS may have a history of joint subluxations, joint pain, cervical instability, and spontaneous epidural leaks. The reason for the overlap between the two syndromes is not clear.

Chronic fatigue syndrome is characterized by persistent fatigue that does not resolve with rest and is not necessarily associated with orthostatic changes. More than 75% of patients with POTS report general fatigue as a major complaint, and up to 23% meet the full criteria for chronic fatigue syndrome.34

 

 

DIAGNOSTIC STRATEGY

A patient presenting with symptoms suggestive of POTS should first undergo a detailed history and physical examination. Other causes of sinus tachycardia should be considered. 

Detailed history, symptom review

The history should focus on determining symptom burden, including tachycardia onset, frequency, severity, and triggers; the presence of syncope; and the impact of symptoms on daily function and quality of life.

lei_posturaltachycardiasyndrome_t1.jpg
POTS-associated orthostatic intolerance manifests with cardiac and noncardiac symptoms (Table 1).

Presyncope and its associated symptoms occur in less than one-third of patients with POTS, and syncope is not a principal feature.4 If syncope is the predominant complaint, alternative causes should be investigated. The usual cause of syncope in the general population is thought to be vasovagal.

In addition to orthostatic intolerance, gastrointestinal disturbances are common in POTS, presenting as abdominal pain, heartburn, irregular bowel movements, diarrhea, or constipation. Symptoms of gastroparesis are less common. Gastrointestinal symptoms tend to be prolonged, lasting hours and occurring multiple times a week. They tend not to improve in the supine position.35 

POTS-associated symptoms may develop insidiously, but patients often report onset after an acute stressor such as pregnancy, major surgery, or a presumed viral illness.4 Whether these putative triggers are causative or coincidental is unknown. Symptoms of orthostatic intolerance tend to be exacerbated by dehydration, heat, alcohol, exercise, and menstruation.36,37

Consider the family history: 1 in 8 patients with POTS reports familial orthostatic intolerance,38 suggesting a genetic role in some patients. Inquire about symptoms or a previous diagnosis of Ehlers-Danlos syndrome and mast cell activation syndrome.

Consider other conditions

lei_posturaltachycardiasyndrome_t2.jpg
Other causes of orthostatic tachycardia are listed in Table 2.39–41 Most can be diagnosed with a careful history, physical examination, and laboratory tests. Two of the more challenging diagnoses are described below. 

Pheochromocytoma causes hyperadrenergic symptoms (eg, palpitations, lightheadedness) like those in POTS, but patients with pheochromocytoma typically have these symptoms while supine. Pheochromocytoma is also characterized by plasma norepinephrine levels much higher than in POTS.4 Plasma metanephrine testing helps diagnose or rule out pheochromocytoma.5

Inappropriate sinus tachycardia, like pheochromocytoma, also has clinical features similar to those of POTS, as well as tachycardia present when supine. It involves higher sympathetic tone and lower parasympathetic tone compared with POTS; patients commonly have a daytime resting heart rate of at least 100 bpm or a 24-hour mean heart rate of at least 90 bpm.1,42 While the intrinsic heart rate is heightened in inappropriate sinus tachycardia, it is not different between POTS patients and healthy individuals.42,43 Distinguishing POTS from inappropriate sinus tachycardia is further complicated by the broad inclusion criteria of most studies of inappropriate sinus tachycardia, which failed to exclude patients with POTS.44 The Heart Rhythm Society recently adopted distinct definitions for the 2 conditions.1

Physical examination: Focus on vital signs

lei_posturaltachycardiasyndrome_f1.jpg
%3Cp%3EFigure%201.%20Results%20of%20head-up%20tilt-table%20(HUT)%20testing%20in%20a%20healthy%20person%20(top)%20and%20in%20a%20patient%20with%20postural%20tachycardia%20syndrome%20(POTS)%20(bottom).%20Upon%20passive%20head-up%20tilting%2C%20the%20heart%20rate%20increases%20in%20POTS%20by%20at%20least%2030%20bpm%20but%20remains%20largely%20stable%20in%20healthy%20individuals.%20Orthostatic%20hypotension%20(a%20fall%20in%20blood%20pressure%20of%20%E2%89%A5%2020%2F10%20mm%20Hg)%20does%20not%20occur%20in%20either%20patient.%3C%2Fp%3E
The most critical component of the physical examination is thorough measurement of orthostatic vital signs (Figure 1). Blood pressure and heart rate should be measured while the patient has been supine for at least 5 minutes, and again after being upright for 1, 3, 5, and 10 minutes. These measurements determine if orthostatic hypotension is present and whether the patient meets the heart rate criteria for POTS. Patients with POTS tend to experience greater orthostatic tachycardia in the morning, so evaluation early in the day optimizes diagnostic sensitivity.5

Dependent acrocyanosis—dark red-blue discoloration of the lower legs that is cold to the touch—occurs in about half of patients with POTS upon standing.4 Dependent acrocyanosis is associated with joint hypermobility and Ehlers-Danlos syndrome, so these conditions should also be considered if findings are positive.

Laboratory testing for other causes

Laboratory testing is used mainly to detect primary causes of sinus tachycardia. Tests should include:

  • Complete blood cell count with hematocrit (for severe anemia)
  • Thyroid-stimulating hormone level (for hyperthyroidism)
  • Electrolyte panel (for significant electrolyte disturbances).

Evidence is insufficient to support routinely measuring the vitamin B12 level, iron indices, and serum markers for celiac disease, although these may be done if the history or physical examination suggests related problems.4 Sicca symptoms (severe dry eye or dry mouth) should trigger evaluation for Sjögren syndrome.

Electrocardiography needed

Electrocardiography should be performed to investigate for cardiac conduction abnormalities as well as for resting markers of a supraventricular tachyarrhythmia. Extended ambulatory (Holter) monitoring may be useful to evaluate for a transient reentrant tachyarrhythmia4; however, it does not record body position, so it can be difficult to determine if detected episodes of tachycardia are related to posture.

Additional testing for select cases

Further investigation is usually not needed to diagnose POTS but should be considered in some cases. Advanced tests are typically performed at a tertiary care referral center and include: 


  • Quantitative sensory testing to evaluate for small-fiber neuropathy (ie, Quantitative Sudomotor Axon Reflex Test, or QSART), which occurs in the neuropathic POTS subtype
  • Formal autonomic function testing to characterize neurovascular responsiveness  
  • Supine and standing plasma norepinephrine levels (fractionated catecholamines) to characterize the net activation of the sympathetic nervous system
  • Blood volume assessments to assess hypovolemia 
  • Formal exercise testing to objectively quantify exercise capacity.

 

 

GRADED MANAGEMENT

No single universal gold-standard therapy exists for POTS, and management should be individually determined with the primary goals of treating symptoms and restoring function. A graded approach should be used, starting with conservative nonpharmacologic therapies and adding medications as needed.

While the disease course varies substantially from patient to patient, proper management is strongly associated with eventual symptom improvement.1

NONPHARMACOLOGIC STEPS FIRST

lei_posturaltachycardiasyndrome_t3.jpg
A multipronged nonpharmacologic approach should be used for all patients before resorting to medications (Table 3). In an observational study, most patients reported that such interventions were more helpful than medications.45 The following elements are recommended:

Education

Patients should be informed of the nature of their condition and referred to appropriate healthcare personnel. POTS is a chronic illness requiring individualized coping strategies, intensive physician interaction, and support of a multidisciplinary team. Patients and family members can be reassured that most symptoms improve over time with appropriate diagnosis and treatment.1 Patients should be advised to avoid aggravating triggers and activities.

Exercise

Exercise programs are encouraged but should be introduced gradually, as physical activity can exacerbate symptoms, especially at the outset. Several studies have reported benefits from a short-term (3-month) program, in which the patient gradually progresses from non-upright exercise (eg, rowing machine, recumbent cycle, swimming) to upright endurance exercises. At the end of these programs, significant cardiac remodeling, improved quality of life, and reduced heart rate responses to standing have been reported, and benefits have been reported to persist in patients who continued exercising after the 3-month study period.46,47

Despite the benefits of exercise interventions, compliance is low.46,47 To prevent early discouragement, patients should be advised that it can take 4 to 6 weeks of continued exercise before benefits appear. Patients are encouraged to exercise every other day for 30 minutes or more. Regimens should primarily focus on aerobic conditioning, but resistance training, concentrating on thigh muscles, can also help. Exercise is a treatment and not a cure, and benefits can rapidly disappear if regular activity (at least 3 times per week) is stopped.48

Compression stockings

Compression stockings help reduce peripheral venous pooling and enhance venous return to the heart. Waist-high stockings with compression of at least 30 to 40 mm Hg offer the best results. 

Diet

Increased fluid and salt intake is advisable for patients with suspected hypovolemia. At least 2 to 3 L of water accompanied by 10 to 12 g of daily sodium intake is recommended.1 This can usually be accomplished with diet and salt added to food, but salt tablets can be used if the patient prefers. The resultant plasma volume expansion may help reduce the reflex tachycardia upon standing.49

Check medications

lei_posturaltachycardiasyndrome_t4.jpg
The clinician should review—and perhaps discontinue—medications the patient is already taking that may exacerbate tachycardia or related symptoms (Table 4).50 Venodilators decrease preload, thereby reducing cardiac output and blood pressure, which triggers compensatory tachycardia. Diuretics can reduce effective blood volume and lower preload, leading to worsened symptoms mediated by hypovolemia.

Rescue therapy with saline infusion

Intravenous saline infusion can augment blood volume in patients who are clinically decompensated and present with severe symptoms.1 Intermittent infusion of 1 L of normal saline has been found to significantly reduce orthostatic tachycardia and related symptoms in patients with POTS, contributing to improved quality of life.51,52

Chronic saline infusions are not recommended for long-term care because of the risk of access complications and infection.1 Moak et al53 reported a high rate of bacteremia in a cohort of children with POTS with regular saline infusions, most of whom had a central line. On the other hand, Ruzieh et al54 reported significantly improved symptoms with regular saline infusions without a high rate of complications, but patients in this study received infusions for only a few months and through a peripheral intravenous catheter.

 

 

DRUG THERAPY

lei_posturaltachycardiasyndrome_t5.jpg
Drug therapy for POTS should be used only if nonpharmacologic interventions do not adequately relieve symptoms. Given the heterogeneity of POTS, treatment should be tailored to the patient’s underlying pathophysiology, key clinical features, and comorbidities. These considerations should guide the initial selection of medications, with adjustments as needed to alleviate adverse effects (Table 5).

No medications are approved by the US Food and Drug Administration (FDA) or Health Canada specifically for treating POTS, making all pharmacologic recommendations off-label. Although the drugs discussed below have been evaluated for POTS in controlled laboratory settings, they have yet to be tested in robust clinical trials.

Blood volume expansion

Several drugs expand blood volume, which may reduce orthostatic tachycardia.

Fludrocortisone is a synthetic aldosterone analogue that enhances sodium and water retention. Although one observational study found that it normalizes hemodynamic changes in response to orthostatic stress, no high-level evidence exists for its effectiveness for POTS.55 It is generally well tolerated, although possible adverse effects include hyperkalemia, hypertension, fatigue, nausea, headache, and edema.5,56

Desmopressin is a synthetic version of a natural antidiuretic hormone that increases kidney-mediated free-water reabsorption without sodium retention. It significantly reduces upright heart rate in patients with POTS and improves symptom burden. Although potential adverse effects include edema and headache, hyponatremia is the primary concern with daily use, especially with the increased water intake advised for POTS.57 Patients should be advised to use desmopressin no more than once a week for the acute improvement of symptoms. Intermittent monitoring of serum sodium levels is recommended for safety.

Erythropoietin replacement has been suggested for treating POTS to address the significant deficit in red blood cell volume. Although erythropoietin therapy has a direct vasoconstrictive effect and largely improves red blood cell volume in patients with POTS, it does not expand plasma volume, so orthostatic tachycardia is not itself reduced.22 Nevertheless, it may significantly improve POTS symptoms refractory to more common methods of treatment, and it should be reserved for such cases. In addition to the lack of effect on orthostatic tachycardia, drawbacks to using erythropoietin include its high cost, the need for subcutaneous administration, and the risk of life-threatening complications such as myocardial infarction and stroke.58,59

Heart rate-lowering agents

Propranolol, a nonselective beta-adrenergic antagonist, can significantly reduce standing heart rate and improve symptoms at low dosages (10–20 mg). Higher dosages can further restrain orthostatic tachycardia but are not as well tolerated, mainly due to hypotension and worsening of existing symptoms such as fatigue.60 Regular-acting propranolol works for about 4 to 5 hours per dose, so full-day coverage often requires dosing 4 times per day.

Ivabradine is a selective blocker of the  “funny” (If) channel that reduces the sinus node firing rate without affecting blood pressure, so it slows heart rate without causing supine hypertension or orthostatic hypotension.

A retrospective case series found that 60% of patients with POTS treated with ivabradine reported symptomatic improvement, and all patients experienced reduced tachycardia with continued use.61 Ivabradine has not been compared with placebo or propranolol in a randomized controlled trial, and it has not been well studied in pregnancy and so should be avoided because of potential teratogenic effects.

When prescribing ivabradine for women of childbearing age, a negative pregnancy test may be documented prior to initiation of therapy, and the use of highly effective methods of contraception is recommended. Ivabradine should be avoided in women contemplating pregnancy. Insurance coverage can limit access to ivabradine in the United States.

Central nervous system sympatholytics

Patients with prominent hyperadrenergic features may benefit from central sympatholytic agents. However, these drugs may not be well tolerated in patients with neuropathic POTS because of the effects of reduced systemic vascular resistance5 and the possible exacerbation of drowsiness, fatigue, and mental clouding.4 Patients can be extremely sensitive to these medications, so they should initially be prescribed at the lowest dose, then gradually increased as tolerated.

Clonidine, an alpha-2-adrenergic agonist, decreases central sympathetic tone. In hyperadrenergic patients, clonidine can stabilize heart rate and blood pressure, thereby reducing orthostatic symptoms.62

Methyldopa has effects similar to those of clonidine but is easier to titrate owing to its longer half-life.63 Methyldopa is typically started at 125 mg at bedtime and increased to 125 mg twice daily, if tolerated.             

 

 

Other agents

Midodrine is a prodrug. The active form, an alpha-1-adrenergic agonist, constricts peripheral veins and arteries to increase vascular resistance and venous return, thereby reducing orthostatic tachycardia.52 It is most useful in patients with impaired peripheral vasoconstriction (eg, neuropathic POTS) and may be less effective in those with hyperadrenergic POTS.64 Major limitations of midodrine include worsening supine hypertension and possible urinary retention.39

Because of midodrine’s short half-life, frequent dosing is required during daytime hours (eg, 8 AM, noon, and 4 PM), but it should not be taken within 4 to 5 hours of sleep because of the risk of supine hypertension. Midodrine is typically started at 2.5 to 5 mg per dose and can be titrated up to 15 mg per dose.

Midodrine is an FDA pregnancy category  C drug (adverse effects in pregnancy seen in animal models, but evidence lacking in humans). While ideally it should be avoided, we have used it safely in pregnant women with disabling POTS symptoms.

Pyridostigmine, an acetylcholinesterase inhibitor, increases cardiovagal tone and possibly sympathetic tone. It has been reported to significantly reduce standing heart rate and improve symptom burden in patients with POTS.65 However, pyridostigmine increases gastrointestinal mobility, leading to severe adverse effects in over 20% of patients, including abdominal cramps, nausea, and diarrhea.66

Droxidopa, a synthetic amino acid precursor of norepinephrine, improves dizziness and fatigue in POTS with minimal effects on blood pressure.67

Modafinil, a psychostimulant, may improve POTS-associated cognitive symptoms.4 It also raises upright blood pressure without significantly worsening standing heart rate or acute orthostatic symptoms.68

EFFECTS OF COMORBID DISORDERS ON MANAGEMENT

Ehlers-Danlos syndrome

Pharmacologic approaches to POTS should not be altered based on the presence of Ehlers-Danlos syndrome, but because many of these patients are prone to joint dislocation, exercise prescriptions may need adjusting.

A medical genetics consult is recommended for patients with Ehlers-Danlos syndrome. Although the hypermobile type (the form most commonly associated with POTS) is not associated with aortopathy, it can be confused with classical and vascular Ehlers-Danlos syndromes, which require serial aortic screening.30

Mast cell activation syndrome

Consultation with an allergist or immunologist may help patients with severe symptoms.

Autoantibodies and autoimmunity

Treatment of the underlying disorder is recommended and can result in significantly improved POTS symptoms.

SPECIALTY CARE REFERRAL

POTS can be challenging to manage. Given the range of physiologic, emotional, and functional distress patients experience, it often requires significant physician time and multidisciplinary care. Patients with continued severe or debilitating symptoms may benefit from referral to a tertiary-care center with experience in autonomic nervous system disorders.

PROGNOSIS

Limited data are available on the long-term prognosis of POTS, and more studies are needed in pediatric and adult populations. No deaths have been reported in the handful of published cases of POTS in patients older than 50.1 Some pediatric studies suggest that some teenagers “outgrow” their POTS. However, these data are not robust, and an alternative explanation is that as they get older, they see adult physicians for their POTS symptoms and so are lost to study follow-up.6,44,69 

We have not often seen POTS simply resolve without ongoing treatment. However, in our experience, most patients have improved symptoms and function with multimodal treatment (ie, exercise, salt, water, stockings, and some medications) and time.

Some people, most of them relatively young women, experience lightheadedness, a racing heart, and other symptoms (but not hypotension) when they stand up, in a condition known as postural tachycardia syndrome (POTS).1 Although not known to shorten life,1 it can be physically and mentally debilitating.2,3 Therapy rarely cures it, but a multifaceted approach can substantially improve quality of life.

This review outlines the evaluation and diagnosis of POTS and provides guidance for a therapy regimen.

HOW IS POTS DEFINED?

POTS is a multifactorial syndrome rather than a specific disease. It is characterized by all of the following1,4–6:

  • An increase in heart rate of ≥ 30 bpm, or ≥ 40 bpm for those under age 19, within 10 minutes of standing from a supine position
  • Sustained tachycardia (> 30 seconds) 
  • Absence of orthostatic hypotension (a fall in blood pressure of ≥ 20/10 mm Hg)
  • Frequent and chronic duration (≥ 6 months).

These features are critical to diagnosis. Hemodynamic criteria in isolation may describe postural tachycardia but are not sufficient to diagnose POTS.

The prevalence of POTS is estimated to be between 0.2% and 1.0%,7 affecting up to 3 million people in the United States. Most cases arise between ages 13 and 50, with a female-to-male ratio of 5:1.8

MANY NAMES, SAME CONDITION

In 1871, Da Costa9 described a condition he called “irritable heart syndrome” that had characteristics similar to those of POTS, including extreme fatigue and exercise intolerance. Decades later, Lewis10 and Wood11 provided more detailed descriptions of the disorder, renaming it “soldier’s heart” or “Da Costa syndrome.” As other cases were documented, more terms arose, including “effort syndrome” and “mitral valve prolapse syndrome.”

In 1982, Rosen and Cryer12 were the first to use the term “postural tachycardia syndrome” for patients with disabling tachycardia upon standing without orthostatic hypotension. In 1986, Fouad et al13 described patients with postural tachycardia, orthostatic intolerance, and a small degree of hypotension as having “idiopathic hypovolemia.”

In 1993, Schondorf and Low14 established the current definition of POTS, leading to increased awareness and research efforts to understand its pathophysiology.

MULTIFACTORIAL PATHOPHYSIOLOGY

During the last 2 decades, several often-overlapping forms of POTS have been recognized, all of which share a final common pathway of sustained orthostatic tachycardia.15–19 In addition, a number of common comorbidities were identified through review of large clinic populations of POTS.20,21

Hypovolemic POTS

Up to 70% of patients with POTS have hypovolemia. The average plasma volume deficit is about 13%, which typically causes only insignificant changes in heart rate and norepinephrine levels while a patient is supine. However, blood pooling associated with upright posture further compromises cardiac output and consequently increases sympathetic nerve activity. Abnormalities in the renin-angiotensin-aldosterone volume regulation system are also suspected to impair sodium retention, contributing to hypovolemia.1,22

Neuropathic POTS

About half of patients with POTS have partial sympathetic denervation (particularly in the lower limbs) and inadequate vasoconstriction upon standing, leading to reduced venous return and stroke volume.17,23 A compensatory increase in sympathetic tone results in tachycardia to maintain cardiac output and blood pressure.

Hyperadrenergic POTS

Up to 50% of patients with POTS have high norepinephrine levels (≥ 600 pg/mL) when upright. This subtype, hyperadrenergic POTS, is characterized by an increase in systolic blood pressure of at least 10 mm Hg within 10 minutes of standing, with concomitant tachycardia that can be similar to or greater than that seen in nonhyperadrenergic POTS. Patients with hyperadrenergic POTS tend to report more prominent symptoms of sympathetic activation, such as palpitations, anxiety, and tremulousness.24,25

Norepinephrine transporter deficiency

The norepinephrine transporter (NET) is on the presynaptic cleft of sympathetic neurons and serves to clear synaptic norepinephrine. NET deficiency leads to a hyperadrenergic state and elevated sympathetic nerve activation.18 NET deficiency may be induced by common antidepressants (eg, tricyclic antidepressants and serotonin-norepinephrine reuptake inhibitors) and attention-deficit disorder medications.4

Mast cell activation syndrome

The relationship between mast cell activation syndrome and POTS is poorly understood.4,26 Mast cell activation syndrome has been described in a subset of patients with POTS who have sinus tachycardia accompanied by severe episodic flushing. Patients with this subtype have a hyperadrenergic response to postural change and elevated urine methylhistamine during flushing episodes.

Patients with mast cell activation syndrome tend to have strong allergic symptoms and may also have severe gastrointestinal problems, food sensitivities, dermatographism, and neuropathy. Diagnosis can be difficult, as the condition is associated with numerous markers with varying sensitivity and specificity.

Autoimmune origin

A significant minority of patients report a viral-like illness before the onset of POTS symptoms, suggesting a possible autoimmune-mediated or inflammatory cause. Also, some autoimmune disorders (eg, Sjögren syndrome) can present with a POTS-like manifestation.

Research into the role of autoantibodies in the pathophysiology of POTS offers the potential to develop novel therapeutic targets. Auto­antibodies that have been reported in POTS include those against M1 to M3 muscarinic receptors (present in over 87% of patients with POTS),27 cardiac lipid raft-associated proteins,28 adrenergic G-protein coupled receptors, alpha-1-adrenergic receptors, and beta-1- and beta-2-adrenergic receptors.29 Although commercial enzyme-linked immunosorbent assays can assess for these antibody fragments, it is not known whether targeting the antibodies improves outcomes. At this time, antibody testing for POTS should be confined to the research setting.

LINKS TO OTHER SYNDROMES

POTS is often associated with other conditions whose symptoms cannot be explained by postural intolerance or tachycardia.

Ehlers-Danlos syndromes are a group of inherited heterogeneous disorders involving joint hypermobility, skin hyperextensibility, and tissue fragility.30 The hypermobile subtype is most commonly associated with POTS, with patients often having symptoms of autonomic dysregulation and autonomic test abnormalities.31–33 Patients with POTS may have a history of joint subluxations, joint pain, cervical instability, and spontaneous epidural leaks. The reason for the overlap between the two syndromes is not clear.

Chronic fatigue syndrome is characterized by persistent fatigue that does not resolve with rest and is not necessarily associated with orthostatic changes. More than 75% of patients with POTS report general fatigue as a major complaint, and up to 23% meet the full criteria for chronic fatigue syndrome.34

 

 

DIAGNOSTIC STRATEGY

A patient presenting with symptoms suggestive of POTS should first undergo a detailed history and physical examination. Other causes of sinus tachycardia should be considered. 

Detailed history, symptom review

The history should focus on determining symptom burden, including tachycardia onset, frequency, severity, and triggers; the presence of syncope; and the impact of symptoms on daily function and quality of life.

lei_posturaltachycardiasyndrome_t1.jpg
POTS-associated orthostatic intolerance manifests with cardiac and noncardiac symptoms (Table 1).

Presyncope and its associated symptoms occur in less than one-third of patients with POTS, and syncope is not a principal feature.4 If syncope is the predominant complaint, alternative causes should be investigated. The usual cause of syncope in the general population is thought to be vasovagal.

In addition to orthostatic intolerance, gastrointestinal disturbances are common in POTS, presenting as abdominal pain, heartburn, irregular bowel movements, diarrhea, or constipation. Symptoms of gastroparesis are less common. Gastrointestinal symptoms tend to be prolonged, lasting hours and occurring multiple times a week. They tend not to improve in the supine position.35 

POTS-associated symptoms may develop insidiously, but patients often report onset after an acute stressor such as pregnancy, major surgery, or a presumed viral illness.4 Whether these putative triggers are causative or coincidental is unknown. Symptoms of orthostatic intolerance tend to be exacerbated by dehydration, heat, alcohol, exercise, and menstruation.36,37

Consider the family history: 1 in 8 patients with POTS reports familial orthostatic intolerance,38 suggesting a genetic role in some patients. Inquire about symptoms or a previous diagnosis of Ehlers-Danlos syndrome and mast cell activation syndrome.

Consider other conditions

lei_posturaltachycardiasyndrome_t2.jpg
Other causes of orthostatic tachycardia are listed in Table 2.39–41 Most can be diagnosed with a careful history, physical examination, and laboratory tests. Two of the more challenging diagnoses are described below. 

Pheochromocytoma causes hyperadrenergic symptoms (eg, palpitations, lightheadedness) like those in POTS, but patients with pheochromocytoma typically have these symptoms while supine. Pheochromocytoma is also characterized by plasma norepinephrine levels much higher than in POTS.4 Plasma metanephrine testing helps diagnose or rule out pheochromocytoma.5

Inappropriate sinus tachycardia, like pheochromocytoma, also has clinical features similar to those of POTS, as well as tachycardia present when supine. It involves higher sympathetic tone and lower parasympathetic tone compared with POTS; patients commonly have a daytime resting heart rate of at least 100 bpm or a 24-hour mean heart rate of at least 90 bpm.1,42 While the intrinsic heart rate is heightened in inappropriate sinus tachycardia, it is not different between POTS patients and healthy individuals.42,43 Distinguishing POTS from inappropriate sinus tachycardia is further complicated by the broad inclusion criteria of most studies of inappropriate sinus tachycardia, which failed to exclude patients with POTS.44 The Heart Rhythm Society recently adopted distinct definitions for the 2 conditions.1

Physical examination: Focus on vital signs

lei_posturaltachycardiasyndrome_f1.jpg
%3Cp%3EFigure%201.%20Results%20of%20head-up%20tilt-table%20(HUT)%20testing%20in%20a%20healthy%20person%20(top)%20and%20in%20a%20patient%20with%20postural%20tachycardia%20syndrome%20(POTS)%20(bottom).%20Upon%20passive%20head-up%20tilting%2C%20the%20heart%20rate%20increases%20in%20POTS%20by%20at%20least%2030%20bpm%20but%20remains%20largely%20stable%20in%20healthy%20individuals.%20Orthostatic%20hypotension%20(a%20fall%20in%20blood%20pressure%20of%20%E2%89%A5%2020%2F10%20mm%20Hg)%20does%20not%20occur%20in%20either%20patient.%3C%2Fp%3E
The most critical component of the physical examination is thorough measurement of orthostatic vital signs (Figure 1). Blood pressure and heart rate should be measured while the patient has been supine for at least 5 minutes, and again after being upright for 1, 3, 5, and 10 minutes. These measurements determine if orthostatic hypotension is present and whether the patient meets the heart rate criteria for POTS. Patients with POTS tend to experience greater orthostatic tachycardia in the morning, so evaluation early in the day optimizes diagnostic sensitivity.5

Dependent acrocyanosis—dark red-blue discoloration of the lower legs that is cold to the touch—occurs in about half of patients with POTS upon standing.4 Dependent acrocyanosis is associated with joint hypermobility and Ehlers-Danlos syndrome, so these conditions should also be considered if findings are positive.

Laboratory testing for other causes

Laboratory testing is used mainly to detect primary causes of sinus tachycardia. Tests should include:

  • Complete blood cell count with hematocrit (for severe anemia)
  • Thyroid-stimulating hormone level (for hyperthyroidism)
  • Electrolyte panel (for significant electrolyte disturbances).

Evidence is insufficient to support routinely measuring the vitamin B12 level, iron indices, and serum markers for celiac disease, although these may be done if the history or physical examination suggests related problems.4 Sicca symptoms (severe dry eye or dry mouth) should trigger evaluation for Sjögren syndrome.

Electrocardiography needed

Electrocardiography should be performed to investigate for cardiac conduction abnormalities as well as for resting markers of a supraventricular tachyarrhythmia. Extended ambulatory (Holter) monitoring may be useful to evaluate for a transient reentrant tachyarrhythmia4; however, it does not record body position, so it can be difficult to determine if detected episodes of tachycardia are related to posture.

Additional testing for select cases

Further investigation is usually not needed to diagnose POTS but should be considered in some cases. Advanced tests are typically performed at a tertiary care referral center and include: 


  • Quantitative sensory testing to evaluate for small-fiber neuropathy (ie, Quantitative Sudomotor Axon Reflex Test, or QSART), which occurs in the neuropathic POTS subtype
  • Formal autonomic function testing to characterize neurovascular responsiveness  
  • Supine and standing plasma norepinephrine levels (fractionated catecholamines) to characterize the net activation of the sympathetic nervous system
  • Blood volume assessments to assess hypovolemia 
  • Formal exercise testing to objectively quantify exercise capacity.

 

 

GRADED MANAGEMENT

No single universal gold-standard therapy exists for POTS, and management should be individually determined with the primary goals of treating symptoms and restoring function. A graded approach should be used, starting with conservative nonpharmacologic therapies and adding medications as needed.

While the disease course varies substantially from patient to patient, proper management is strongly associated with eventual symptom improvement.1

NONPHARMACOLOGIC STEPS FIRST

lei_posturaltachycardiasyndrome_t3.jpg
A multipronged nonpharmacologic approach should be used for all patients before resorting to medications (Table 3). In an observational study, most patients reported that such interventions were more helpful than medications.45 The following elements are recommended:

Education

Patients should be informed of the nature of their condition and referred to appropriate healthcare personnel. POTS is a chronic illness requiring individualized coping strategies, intensive physician interaction, and support of a multidisciplinary team. Patients and family members can be reassured that most symptoms improve over time with appropriate diagnosis and treatment.1 Patients should be advised to avoid aggravating triggers and activities.

Exercise

Exercise programs are encouraged but should be introduced gradually, as physical activity can exacerbate symptoms, especially at the outset. Several studies have reported benefits from a short-term (3-month) program, in which the patient gradually progresses from non-upright exercise (eg, rowing machine, recumbent cycle, swimming) to upright endurance exercises. At the end of these programs, significant cardiac remodeling, improved quality of life, and reduced heart rate responses to standing have been reported, and benefits have been reported to persist in patients who continued exercising after the 3-month study period.46,47

Despite the benefits of exercise interventions, compliance is low.46,47 To prevent early discouragement, patients should be advised that it can take 4 to 6 weeks of continued exercise before benefits appear. Patients are encouraged to exercise every other day for 30 minutes or more. Regimens should primarily focus on aerobic conditioning, but resistance training, concentrating on thigh muscles, can also help. Exercise is a treatment and not a cure, and benefits can rapidly disappear if regular activity (at least 3 times per week) is stopped.48

Compression stockings

Compression stockings help reduce peripheral venous pooling and enhance venous return to the heart. Waist-high stockings with compression of at least 30 to 40 mm Hg offer the best results. 

Diet

Increased fluid and salt intake is advisable for patients with suspected hypovolemia. At least 2 to 3 L of water accompanied by 10 to 12 g of daily sodium intake is recommended.1 This can usually be accomplished with diet and salt added to food, but salt tablets can be used if the patient prefers. The resultant plasma volume expansion may help reduce the reflex tachycardia upon standing.49

Check medications

lei_posturaltachycardiasyndrome_t4.jpg
The clinician should review—and perhaps discontinue—medications the patient is already taking that may exacerbate tachycardia or related symptoms (Table 4).50 Venodilators decrease preload, thereby reducing cardiac output and blood pressure, which triggers compensatory tachycardia. Diuretics can reduce effective blood volume and lower preload, leading to worsened symptoms mediated by hypovolemia.

Rescue therapy with saline infusion

Intravenous saline infusion can augment blood volume in patients who are clinically decompensated and present with severe symptoms.1 Intermittent infusion of 1 L of normal saline has been found to significantly reduce orthostatic tachycardia and related symptoms in patients with POTS, contributing to improved quality of life.51,52

Chronic saline infusions are not recommended for long-term care because of the risk of access complications and infection.1 Moak et al53 reported a high rate of bacteremia in a cohort of children with POTS with regular saline infusions, most of whom had a central line. On the other hand, Ruzieh et al54 reported significantly improved symptoms with regular saline infusions without a high rate of complications, but patients in this study received infusions for only a few months and through a peripheral intravenous catheter.

 

 

DRUG THERAPY

lei_posturaltachycardiasyndrome_t5.jpg
Drug therapy for POTS should be used only if nonpharmacologic interventions do not adequately relieve symptoms. Given the heterogeneity of POTS, treatment should be tailored to the patient’s underlying pathophysiology, key clinical features, and comorbidities. These considerations should guide the initial selection of medications, with adjustments as needed to alleviate adverse effects (Table 5).

No medications are approved by the US Food and Drug Administration (FDA) or Health Canada specifically for treating POTS, making all pharmacologic recommendations off-label. Although the drugs discussed below have been evaluated for POTS in controlled laboratory settings, they have yet to be tested in robust clinical trials.

Blood volume expansion

Several drugs expand blood volume, which may reduce orthostatic tachycardia.

Fludrocortisone is a synthetic aldosterone analogue that enhances sodium and water retention. Although one observational study found that it normalizes hemodynamic changes in response to orthostatic stress, no high-level evidence exists for its effectiveness for POTS.55 It is generally well tolerated, although possible adverse effects include hyperkalemia, hypertension, fatigue, nausea, headache, and edema.5,56

Desmopressin is a synthetic version of a natural antidiuretic hormone that increases kidney-mediated free-water reabsorption without sodium retention. It significantly reduces upright heart rate in patients with POTS and improves symptom burden. Although potential adverse effects include edema and headache, hyponatremia is the primary concern with daily use, especially with the increased water intake advised for POTS.57 Patients should be advised to use desmopressin no more than once a week for the acute improvement of symptoms. Intermittent monitoring of serum sodium levels is recommended for safety.

Erythropoietin replacement has been suggested for treating POTS to address the significant deficit in red blood cell volume. Although erythropoietin therapy has a direct vasoconstrictive effect and largely improves red blood cell volume in patients with POTS, it does not expand plasma volume, so orthostatic tachycardia is not itself reduced.22 Nevertheless, it may significantly improve POTS symptoms refractory to more common methods of treatment, and it should be reserved for such cases. In addition to the lack of effect on orthostatic tachycardia, drawbacks to using erythropoietin include its high cost, the need for subcutaneous administration, and the risk of life-threatening complications such as myocardial infarction and stroke.58,59

Heart rate-lowering agents

Propranolol, a nonselective beta-adrenergic antagonist, can significantly reduce standing heart rate and improve symptoms at low dosages (10–20 mg). Higher dosages can further restrain orthostatic tachycardia but are not as well tolerated, mainly due to hypotension and worsening of existing symptoms such as fatigue.60 Regular-acting propranolol works for about 4 to 5 hours per dose, so full-day coverage often requires dosing 4 times per day.

Ivabradine is a selective blocker of the  “funny” (If) channel that reduces the sinus node firing rate without affecting blood pressure, so it slows heart rate without causing supine hypertension or orthostatic hypotension.

A retrospective case series found that 60% of patients with POTS treated with ivabradine reported symptomatic improvement, and all patients experienced reduced tachycardia with continued use.61 Ivabradine has not been compared with placebo or propranolol in a randomized controlled trial, and it has not been well studied in pregnancy and so should be avoided because of potential teratogenic effects.

When prescribing ivabradine for women of childbearing age, a negative pregnancy test may be documented prior to initiation of therapy, and the use of highly effective methods of contraception is recommended. Ivabradine should be avoided in women contemplating pregnancy. Insurance coverage can limit access to ivabradine in the United States.

Central nervous system sympatholytics

Patients with prominent hyperadrenergic features may benefit from central sympatholytic agents. However, these drugs may not be well tolerated in patients with neuropathic POTS because of the effects of reduced systemic vascular resistance5 and the possible exacerbation of drowsiness, fatigue, and mental clouding.4 Patients can be extremely sensitive to these medications, so they should initially be prescribed at the lowest dose, then gradually increased as tolerated.

Clonidine, an alpha-2-adrenergic agonist, decreases central sympathetic tone. In hyperadrenergic patients, clonidine can stabilize heart rate and blood pressure, thereby reducing orthostatic symptoms.62

Methyldopa has effects similar to those of clonidine but is easier to titrate owing to its longer half-life.63 Methyldopa is typically started at 125 mg at bedtime and increased to 125 mg twice daily, if tolerated.             

 

 

Other agents

Midodrine is a prodrug. The active form, an alpha-1-adrenergic agonist, constricts peripheral veins and arteries to increase vascular resistance and venous return, thereby reducing orthostatic tachycardia.52 It is most useful in patients with impaired peripheral vasoconstriction (eg, neuropathic POTS) and may be less effective in those with hyperadrenergic POTS.64 Major limitations of midodrine include worsening supine hypertension and possible urinary retention.39

Because of midodrine’s short half-life, frequent dosing is required during daytime hours (eg, 8 AM, noon, and 4 PM), but it should not be taken within 4 to 5 hours of sleep because of the risk of supine hypertension. Midodrine is typically started at 2.5 to 5 mg per dose and can be titrated up to 15 mg per dose.

Midodrine is an FDA pregnancy category  C drug (adverse effects in pregnancy seen in animal models, but evidence lacking in humans). While ideally it should be avoided, we have used it safely in pregnant women with disabling POTS symptoms.

Pyridostigmine, an acetylcholinesterase inhibitor, increases cardiovagal tone and possibly sympathetic tone. It has been reported to significantly reduce standing heart rate and improve symptom burden in patients with POTS.65 However, pyridostigmine increases gastrointestinal mobility, leading to severe adverse effects in over 20% of patients, including abdominal cramps, nausea, and diarrhea.66

Droxidopa, a synthetic amino acid precursor of norepinephrine, improves dizziness and fatigue in POTS with minimal effects on blood pressure.67

Modafinil, a psychostimulant, may improve POTS-associated cognitive symptoms.4 It also raises upright blood pressure without significantly worsening standing heart rate or acute orthostatic symptoms.68

EFFECTS OF COMORBID DISORDERS ON MANAGEMENT

Ehlers-Danlos syndrome

Pharmacologic approaches to POTS should not be altered based on the presence of Ehlers-Danlos syndrome, but because many of these patients are prone to joint dislocation, exercise prescriptions may need adjusting.

A medical genetics consult is recommended for patients with Ehlers-Danlos syndrome. Although the hypermobile type (the form most commonly associated with POTS) is not associated with aortopathy, it can be confused with classical and vascular Ehlers-Danlos syndromes, which require serial aortic screening.30

Mast cell activation syndrome

Consultation with an allergist or immunologist may help patients with severe symptoms.

Autoantibodies and autoimmunity

Treatment of the underlying disorder is recommended and can result in significantly improved POTS symptoms.

SPECIALTY CARE REFERRAL

POTS can be challenging to manage. Given the range of physiologic, emotional, and functional distress patients experience, it often requires significant physician time and multidisciplinary care. Patients with continued severe or debilitating symptoms may benefit from referral to a tertiary-care center with experience in autonomic nervous system disorders.

PROGNOSIS

Limited data are available on the long-term prognosis of POTS, and more studies are needed in pediatric and adult populations. No deaths have been reported in the handful of published cases of POTS in patients older than 50.1 Some pediatric studies suggest that some teenagers “outgrow” their POTS. However, these data are not robust, and an alternative explanation is that as they get older, they see adult physicians for their POTS symptoms and so are lost to study follow-up.6,44,69 

We have not often seen POTS simply resolve without ongoing treatment. However, in our experience, most patients have improved symptoms and function with multimodal treatment (ie, exercise, salt, water, stockings, and some medications) and time.

References
  1. Sheldon RS, Grubb BP 2nd, Olshansky B, et al. 2015 Heart Rhythm Society expert consensus statement on the diagnosis and treatment of postural tachycardia syndrome, inappropriate sinus tachycardia, and vasovagal syncope. Heart Rhythm 2015; 12(6):e41–e63. doi:10.1016/j.hrthm.2015.03.029
  2. Bagai K, Song Y, Ling JF, et al. Sleep disturbances and diminished quality of life in postural tachycardia syndrome. J Clin Sleep Med 2011; 7(2):204–210. pmid:21509337
  3. Benrud-Larson LM, Dewar MS, Sandroni P, Rummans TA, Haythornthwaite JA, Low PA. Quality of life in patients with postural tachycardia syndrome. Mayo Clin Proc 2002; 77(6):531–537. doi:10.4065/77.6.531
  4. Raj SR. Postural tachycardia syndrome (POTS). Circulation 2013; 127(23):2336–2342. doi:10.1161/CIRCULATIONAHA.112.144501
  5. Raj SR. The postural tachycardia syndrome (POTS): pathophysiology, diagnosis & management. Indian Pacing Electrophysiol J 2006; 6(2):84–99. pmid:16943900
  6. Singer W, Sletten DM, Opfer-Gehrking TL, Brands CK, Fischer PR, Low PA. Postural tachycardia in children and adolescents: what is abnormal? J Pediatr 2012; 160(2):222–226. doi:10.1016/j.jpeds.2011.08.054
  7. Mar PL, Raj SR. Neuronal and hormonal perturbations in postural tachycardia syndrome. Front Physiol 2014; 5:220. doi:10.3389/fphys.2014.00220
  8. Garland EM, Raj SR, Black BK, Harris PA, Robertson D. The hemodynamic and neurohumoral phenotype of postural tachycardia syndrome. Neurology 2007; 69(8):790–798. doi:10.1212/01.wnl.0000267663.05398.40
  9. Da Costa JM. On irritable heart: a clinical study of a form of functional cardiac disorder and its consequences. Am J Med Sci 1871; 61(121):2–52.
  10. Lewis T. The tolerance of physical exertion, as shown by soldiers suffering from so-called “irritable heart.” Br Med J 1918; 1(2987):363–365. pmid:20768980
  11. Wood P. Da Costa’s syndrome (or effort syndrome): lecture I. Br Med J 1941; 1(4194):767–772. pmid:20783672
  12. Rosen SG, Cryer PE. Postural tachycardia syndrome. Reversal of sympathetic hyperresponsiveness and clinical improvement during sodium loading. Am J Med 1982; 72(5):847–850.
  13. Fouad FM, Tadena-Thome L, Bravo EL, Tarazi RC. Idiopathic hypovolemia. Ann Intern Med 1986; 104(3):298–303. pmid:3511818
  14. Schondorf R, Low PA. Idiopathic postural orthostatic tachycardia syndrome: an attenuated form of acute pandysautonomia? Neurology 1993; 43(1):132–137. pmid:8423877
  15. Vernino S, Low PA, Fealey RD, Stewart JD, Farrugia G, Lennon VA. Autoantibodies to ganglionic acetylcholine receptors in autoimmune autonomic neuropathies. N Engl J Med 2000; 343(12):847–855. doi:10.1056/NEJM200009213431204
  16. Raj SR, Robertson D. Blood volume perturbations in the postural tachycardia syndrome. Am J Med Sci 2007; 334(1):57–60. doi:10.1097/MAJ.0b013e318063c6c0
  17. Jacob G, Costa F, Shannon JR, et al. The neuropathic postural tachycardia syndrome. N Engl J Med 2000; 343(14):1008–1014. doi:10.1056/NEJM200010053431404
  18. Shannon JR, Flattem NL, Jordan J, et al. Orthostatic intolerance and tachycardia associated with norepinephrine-transporter deficiency. N Engl J Med 2000; 342(8):541–549. doi:10.1056/NEJM200002243420803
  19. Jones PK, Shaw BH, Raj SR. Clinical challenges in the diagnosis and management of postural tachycardia syndrome. Pract Neurol 2016; 16(6):431–438. doi:10.1136/practneurol-2016-001405
  20. Gunning WT, Karabin BL, Blomquist TM, Grubb BP. Postural orthostatic tachycardia syndrome is associated with platelet storage pool deficiency. Medicine (Baltimore) 2016; 95(37):e4849. doi:10.1097/MD.0000000000004849
  21. Kanjwal K, Sheikh M, Karabin B, Kanjwal Y, Grubb BP. Neurocardiogenic syncope coexisting with postural orthostatic tachycardia syndrome in patients suffering from orthostatic intolerance: a combined form of autonomic dysfunction. Pacing Clin Electrophysiol 2011; 34(5):549–554. doi:10.1111/j.1540-8159.2010.02994.x
  22. Raj SR, Biaggioni I, Yamhure PC, et al. Renin-aldosterone paradox and perturbed blood volume regulation underlying postural tachycardia syndrome. Circulation 2005; 111(13):1574–1582. doi:10.1161/01.CIR.0000160356.97313.5D
  23. Gibbons CH, Bonyhay I, Benson A, Wang N, Freeman R. Structural and functional small fiber abnormalities in the neuropathic postural tachycardia syndrome. PLoS One 2013; 8(12):e84716. doi:10.1371/journal.pone.0084716
  24. Low PA, Sandroni P, Joyner M, Shen WK. Postural tachycardia syndrome (POTS). J Cardiovasc Electrophysiol 2009; 20(3):352–358. doi:10.1111/j.1540-8167.2008.01407.x
  25. Kanjwal K, Saeed B, Karabin B, Kanjwal Y, Grubb BP. Clinical presentation and management of patients with hyperadrenergic postural orthostatic tachycardia syndrome. A single center experience. Cardiol J 2011; 18(5):527–531. pmid:21947988
  26. Shibao C, Arzubiaga C, Roberts J, et al. Hyperadrenergic postural tachycardia syndrome in mast cell activation disorders. Hypertension 2005; 45(3):385–390. doi:10.1161/01.HYP.0000158259.68614.40
  27. Dubey D, Hopkins S, Vernino S. M1 and M2 muscarinic receptor antibodies among patients with postural orthostatic tachycardia syndrome: potential disease biomarker [abstract]. J Clin Neuromuscul Dis 2016; 17(3):179S.
  28. Wang XL, Ling TY, Charlesworth MC, et al. Autoimmunoreactive IgGs against cardiac lipid raft-associated proteins in patients with postural orthostatic tachycardia syndrome. Transl Res 2013; 162(1):34–44. doi:10.1016/j.trsl.2013.03.002
  29. Li H, Yu X, Liles C, et al. Autoimmune basis for postural tachycardia syndrome. J Am Heart Assoc 2014; 3(1):e000755. doi:10.1161/JAHA.113.000755
  30. Malfait F, Francomano C, Byers P, et al. The 2017 international classification of the Ehlers-Danlos syndromes. Am J Med Genet C Semin Med Genet 2017; 175(1):8–26. doi:10.1002/ajmg.c.31552
  31. Wallman D, Weinberg J, Hohler AD. Ehlers-Danlos syndrome and postural tachycardia syndrome: a relationship study. J Neurol Sci 2014; 340(1-2):99–102. doi:10.1016/j.jns.2014.03.002
  32. De Wandele I, Calders P, Peersman W, et al. Autonomic symptom burden in the hypermobility type of Ehlers-Danlos syndrome: a comparative study with two other EDS types, fibromyalgia, and healthy controls. Semin Arthritis Rheum 2014; 44(3):353–361. doi:10.1016/j.semarthrit.2014.05.013
  33. Gazit Y, Nahir AM, Grahame R, Jacob G. Dysautonomia in the joint hypermobility syndrome. Am J Med 2003; 115(1):33–40. pmid:12867232
  34. Okamoto LE, Raj SR, Peltier A, et al. Neurohumoral and haemodynamic profile in postural tachycardia and chronic fatigue syndromes. Clin Sci (Lond) 2012; 122(4):183–192. doi:10.1042/CS20110200
  35. Wang LB, Culbertson CJ, Deb A, Morgenshtern K, Huang H, Hohler AD. Gastrointestinal dysfunction in postural tachycardia syndrome. J Neurol Sci 2015; 359(1-2):193–196. doi:10.1016/j.jns.2015.10.052
  36. Raj S, Sheldon R. Management of postural tachycardia syndrome, inappropriate sinus tachycardia and vasovagal syncope. Arrhythm Electrophysiol Rev 2016; 5(2):122–129. doi:10.15420/AER.2016.7.2
  37. Peggs KJ, Nguyen H, Enayat D, Keller NR, Al-Hendy A, Raj SR. Gynecologic disorders and menstrual cycle lightheadedness in postural tachycardia syndrome. Int J Gynaecol Obstet 2012; 118(3):242–246. doi:10.1016/j.ijgo.2012.04.014
  38. Thieben MJ, Sandroni P, Sletten DM, et al. Postural orthostatic tachycardia syndrome: the Mayo Clinic experience. Mayo Clin Proc 2007; 82(3):308–313. doi:10.4065/82.3.308
  39. Deb A, Morgenshtern K, Culbertson CJ, Wang LB, Hohler AD. A survey-based analysis of symptoms in patients with postural orthostatic tachycardia syndrome. J Am Coll Cardiol 2015; 28(7):157–159. pmid:25829642
  40. Ertek S, Cicero AF. Hyperthyroidism and cardiovascular complications: a narrative review on the basis of pathophysiology. Arch Med Sci 2013; 9(5):944–952. doi:10.5114/aoms.2013.38685
  41. Rangno RE, Langlois S. Comparison of withdrawal phenomena after propranolol, metoprolol and pindolol. Br J Clin Pharmacol 1982; 13(suppl 2):345S–351S. pmid:6125187
  42. Nwazue VC, Paranjape SY, Black BK, et al. Postural tachycardia syndrome and inappropriate sinus tachycardia: role of autonomic modulation and sinus node automaticity. J Am Heart Assoc 2014; 3(2):e000700. doi:10.1161/JAHA.113.000700
  43. Morillo CA, Klein GJ, Thakur RK, Li H, Zardini M, Yee R. Mechanism of “inappropriate” sinus tachycardia. Role of sympathovagal balance. Circulation 1994; 90(2):873–877. pmid:7913886
  44. Grubb BP. Postural tachycardia syndrome. Circulation 2008; 117(21):2814–2817. doi:10.1161/CIRCULATIONAHA.107.761643
  45. Bhatia R, Kizilbash SJ, Ahrens SP, et al. Outcomes of adolescent-onset postural orthostatic tachycardia syndrome. J Pediatr 2016; 173:149–153. doi:10.1016/j.jpeds.2016.02.035
  46. George SA, Bivens TB, Howden EJ, et al. The international POTS registry: evaluating the efficacy of an exercise training intervention in a community setting. Heart Rhythm 2016; 13(4):943–950. doi:10.1016/j.hrthm.2015.12.012
  47. Fu Q, VanGundy TB, Galbreath MM, et al. Cardiac origins of the postural orthostatic tachycardia syndrome. J Am Coll Cardiol 2010; 55(25):2858–2868. doi:10.1016/j.jacc.2010.02.043
  48. Raj SR. Row, row, row your way to treating postural tachycardia syndrome. Heart Rhythm 2016; 13(4):951–952. doi:10.1016/j.hrthm.2015.12.039
  49. Celedonio JE, Garland EM, Nwazue VC, et al. Effects of high sodium intake on blood volume and catecholamines in patients with postural tachycardia syndrome and healthy females [abstract]. Clin Auton Res 2014; 24:211.
  50. Garland EM, Celedonio JE, Raj SR. Postural tachycardia syndrome: beyond orthostatic intolerance. Curr Neurol Neurosci Rep 2015; 15(9):60. doi:10.1007/s11910-015-0583-8
  51. Gordon VM, Opfer-Gehrking TL, Novak V, Low PA. Hemodynamic and symptomatic effects of acute interventions on tilt in patients with postural tachycardia syndrome. Clin Auton Res 2000; 10:29–33. pmid:10750641
  52. Jacob G, Shannon JR, Black B, et al. Effects of volume loading and pressor agents in idiopathic orthostatic tachycardia. Circulation 1997; 96(2):575–580. pmid:9244228
  53. Moak JP, Leong D, Fabian R, et al. Intravenous hydration for management of medication-resistant orthostatic intolerance in the adolescent and young adult. Pediatr Cardiol 2016; 37(2):278–282. doi:10.1007/s00246-015-1274-6
  54. Ruzieh M, Baugh A, Dasa O, et al. Effects of intermittent intravenous saline infusions in patients with medication-refractory postural tachycardia syndrome. J Interv Card Electrophysiol 2017; 48(3):255–260. doi:10.1007/s10840-017-0225-y
  55. Freitas J, Santos R, Azevedo E, Costa O, Carvalho M, de Freitas AF. Clinical improvement in patients with orthostatic intolerance after treatment with bisoprolol and fludrocortisone. Clin Auton Res 2000; 10(5):293–299. pmid:11198485
  56. Lee AK, Krahn AD. Evaluation of syncope: focus on diagnosis and treatment of neurally mediated syncope. Expert Rev Cardiovasc Ther 2016; 14(6):725–736. doi:10.1586/14779072.2016.1164034
  57. Coffin ST, Black BK, Biaggioni I, et al. Desmopressin acutely decreases tachycardia and improves symptoms in the postural tachycardia syndrome. Heart Rhythm 2012; 9(9):1484–1490. doi:10.1016/j.hrthm.2012.05.002
  58. Kanjwal K, Saeed B, Karabin B, Kanjwal Y, Sheikh M, Grubb BP. Erythropoietin in the treatment of postural orthostatic tachycardia syndrome. Am J Ther 2012; 19(2):92–95. doi:10.1097/MJT.0b013e3181ef621a
  59. Hoeldtke RD, Horvath GG, Bryner KD. Treatment of orthostatic tachycardia with erythropoietin. Am J Med 1995; 99(5):525–529. pmid:7485211
  60. Raj SR, Black BK, Biaggioni I, et al. Propranolol decreases tachycardia and improves symptoms in the postural tachycardia syndrome: less is more. Circulation 2009; 120(9):725–734. doi:10.1161/CIRCULATIONAHA.108.846501
  61. McDonald C, Frith J, Newton JL. Single centre experience of ivabradine in postural orthostatic tachycardia syndrome. Europace 2011; 13(3):427–430. doi:10.1093/europace/euq390
  62. Gaffney FA, Lane LB, Pettinger W, Blomqvist G. Effects of long-term clonidine administration on the hemodynamic and neuroendocrine postural responses of patients with dysautonomia. Chest 1983; 83(suppl 2):436–438. pmid:6295714
  63. Jacob G, Biaggioni I. Idiopathic orthostatic intolerance and postural tachycardia syndromes. Am J Med Sci 1999; 317(2):88–101. pmid:10037112
  64. Ross AJ, Ocon AJ, Medow MS, Stewart JM. A double-blind placebo-controlled cross-over study of the vascular effects of midodrine in neuropathic compared with hyperadrenergic postural tachycardia syndrome. Clin Sci (Lond) 2014; 126(4):289–296. doi:10.1042/CS20130222
  65. Raj SR, Black BK, Biaggioni I, Harris PA, Robertson D. Acetylcholinesterase inhibition improves tachycardia in postural tachycardia syndrome. Circulation 2005; 111(21):2734–2340. doi:10.1161/CIRCULATIONAHA.104.497594
  66. Kanjwal K, Karabin B, Sheikh M, et al. Pyridostigmine in the treatment of postural orthostatic tachycardia: A single-center experience. Pacing Clin Electrophysiol 2011; 34(6):750–755. doi:10.1111/j.1540-8159.2011.03047.x
  67. Ruzieh M, Dasa O, Pacenta A, Karabin B, Grubb B. Droxidopa in the treatment of postural orthostatic tachycardia syndrome. Am J Ther 2017; 24(2):e157–e161. doi:10.1097/MJT.0000000000000468
  68. Kpaeyeh AG Jr, Mar PL, Raj V, et al. Hemodynamic profiles and tolerability of modafinil in the treatment of POTS: a randomized placebo-controlled trial. J Clin Psychopharmacol 2014; 34(6):738–741. doi:10.1097/JCP.0000000000000221
  69. Lai CC, Fischer PR, Brands CK, et al. Outcomes in adolescents with postural orthostatic tachycardia syndrome treated with midodrine and beta-blockers. Pacing Clin Electrophysiol 2009; 32(2):234–238. doi:10.1111/j.1540-8159.2008.02207.x
References
  1. Sheldon RS, Grubb BP 2nd, Olshansky B, et al. 2015 Heart Rhythm Society expert consensus statement on the diagnosis and treatment of postural tachycardia syndrome, inappropriate sinus tachycardia, and vasovagal syncope. Heart Rhythm 2015; 12(6):e41–e63. doi:10.1016/j.hrthm.2015.03.029
  2. Bagai K, Song Y, Ling JF, et al. Sleep disturbances and diminished quality of life in postural tachycardia syndrome. J Clin Sleep Med 2011; 7(2):204–210. pmid:21509337
  3. Benrud-Larson LM, Dewar MS, Sandroni P, Rummans TA, Haythornthwaite JA, Low PA. Quality of life in patients with postural tachycardia syndrome. Mayo Clin Proc 2002; 77(6):531–537. doi:10.4065/77.6.531
  4. Raj SR. Postural tachycardia syndrome (POTS). Circulation 2013; 127(23):2336–2342. doi:10.1161/CIRCULATIONAHA.112.144501
  5. Raj SR. The postural tachycardia syndrome (POTS): pathophysiology, diagnosis & management. Indian Pacing Electrophysiol J 2006; 6(2):84–99. pmid:16943900
  6. Singer W, Sletten DM, Opfer-Gehrking TL, Brands CK, Fischer PR, Low PA. Postural tachycardia in children and adolescents: what is abnormal? J Pediatr 2012; 160(2):222–226. doi:10.1016/j.jpeds.2011.08.054
  7. Mar PL, Raj SR. Neuronal and hormonal perturbations in postural tachycardia syndrome. Front Physiol 2014; 5:220. doi:10.3389/fphys.2014.00220
  8. Garland EM, Raj SR, Black BK, Harris PA, Robertson D. The hemodynamic and neurohumoral phenotype of postural tachycardia syndrome. Neurology 2007; 69(8):790–798. doi:10.1212/01.wnl.0000267663.05398.40
  9. Da Costa JM. On irritable heart: a clinical study of a form of functional cardiac disorder and its consequences. Am J Med Sci 1871; 61(121):2–52.
  10. Lewis T. The tolerance of physical exertion, as shown by soldiers suffering from so-called “irritable heart.” Br Med J 1918; 1(2987):363–365. pmid:20768980
  11. Wood P. Da Costa’s syndrome (or effort syndrome): lecture I. Br Med J 1941; 1(4194):767–772. pmid:20783672
  12. Rosen SG, Cryer PE. Postural tachycardia syndrome. Reversal of sympathetic hyperresponsiveness and clinical improvement during sodium loading. Am J Med 1982; 72(5):847–850.
  13. Fouad FM, Tadena-Thome L, Bravo EL, Tarazi RC. Idiopathic hypovolemia. Ann Intern Med 1986; 104(3):298–303. pmid:3511818
  14. Schondorf R, Low PA. Idiopathic postural orthostatic tachycardia syndrome: an attenuated form of acute pandysautonomia? Neurology 1993; 43(1):132–137. pmid:8423877
  15. Vernino S, Low PA, Fealey RD, Stewart JD, Farrugia G, Lennon VA. Autoantibodies to ganglionic acetylcholine receptors in autoimmune autonomic neuropathies. N Engl J Med 2000; 343(12):847–855. doi:10.1056/NEJM200009213431204
  16. Raj SR, Robertson D. Blood volume perturbations in the postural tachycardia syndrome. Am J Med Sci 2007; 334(1):57–60. doi:10.1097/MAJ.0b013e318063c6c0
  17. Jacob G, Costa F, Shannon JR, et al. The neuropathic postural tachycardia syndrome. N Engl J Med 2000; 343(14):1008–1014. doi:10.1056/NEJM200010053431404
  18. Shannon JR, Flattem NL, Jordan J, et al. Orthostatic intolerance and tachycardia associated with norepinephrine-transporter deficiency. N Engl J Med 2000; 342(8):541–549. doi:10.1056/NEJM200002243420803
  19. Jones PK, Shaw BH, Raj SR. Clinical challenges in the diagnosis and management of postural tachycardia syndrome. Pract Neurol 2016; 16(6):431–438. doi:10.1136/practneurol-2016-001405
  20. Gunning WT, Karabin BL, Blomquist TM, Grubb BP. Postural orthostatic tachycardia syndrome is associated with platelet storage pool deficiency. Medicine (Baltimore) 2016; 95(37):e4849. doi:10.1097/MD.0000000000004849
  21. Kanjwal K, Sheikh M, Karabin B, Kanjwal Y, Grubb BP. Neurocardiogenic syncope coexisting with postural orthostatic tachycardia syndrome in patients suffering from orthostatic intolerance: a combined form of autonomic dysfunction. Pacing Clin Electrophysiol 2011; 34(5):549–554. doi:10.1111/j.1540-8159.2010.02994.x
  22. Raj SR, Biaggioni I, Yamhure PC, et al. Renin-aldosterone paradox and perturbed blood volume regulation underlying postural tachycardia syndrome. Circulation 2005; 111(13):1574–1582. doi:10.1161/01.CIR.0000160356.97313.5D
  23. Gibbons CH, Bonyhay I, Benson A, Wang N, Freeman R. Structural and functional small fiber abnormalities in the neuropathic postural tachycardia syndrome. PLoS One 2013; 8(12):e84716. doi:10.1371/journal.pone.0084716
  24. Low PA, Sandroni P, Joyner M, Shen WK. Postural tachycardia syndrome (POTS). J Cardiovasc Electrophysiol 2009; 20(3):352–358. doi:10.1111/j.1540-8167.2008.01407.x
  25. Kanjwal K, Saeed B, Karabin B, Kanjwal Y, Grubb BP. Clinical presentation and management of patients with hyperadrenergic postural orthostatic tachycardia syndrome. A single center experience. Cardiol J 2011; 18(5):527–531. pmid:21947988
  26. Shibao C, Arzubiaga C, Roberts J, et al. Hyperadrenergic postural tachycardia syndrome in mast cell activation disorders. Hypertension 2005; 45(3):385–390. doi:10.1161/01.HYP.0000158259.68614.40
  27. Dubey D, Hopkins S, Vernino S. M1 and M2 muscarinic receptor antibodies among patients with postural orthostatic tachycardia syndrome: potential disease biomarker [abstract]. J Clin Neuromuscul Dis 2016; 17(3):179S.
  28. Wang XL, Ling TY, Charlesworth MC, et al. Autoimmunoreactive IgGs against cardiac lipid raft-associated proteins in patients with postural orthostatic tachycardia syndrome. Transl Res 2013; 162(1):34–44. doi:10.1016/j.trsl.2013.03.002
  29. Li H, Yu X, Liles C, et al. Autoimmune basis for postural tachycardia syndrome. J Am Heart Assoc 2014; 3(1):e000755. doi:10.1161/JAHA.113.000755
  30. Malfait F, Francomano C, Byers P, et al. The 2017 international classification of the Ehlers-Danlos syndromes. Am J Med Genet C Semin Med Genet 2017; 175(1):8–26. doi:10.1002/ajmg.c.31552
  31. Wallman D, Weinberg J, Hohler AD. Ehlers-Danlos syndrome and postural tachycardia syndrome: a relationship study. J Neurol Sci 2014; 340(1-2):99–102. doi:10.1016/j.jns.2014.03.002
  32. De Wandele I, Calders P, Peersman W, et al. Autonomic symptom burden in the hypermobility type of Ehlers-Danlos syndrome: a comparative study with two other EDS types, fibromyalgia, and healthy controls. Semin Arthritis Rheum 2014; 44(3):353–361. doi:10.1016/j.semarthrit.2014.05.013
  33. Gazit Y, Nahir AM, Grahame R, Jacob G. Dysautonomia in the joint hypermobility syndrome. Am J Med 2003; 115(1):33–40. pmid:12867232
  34. Okamoto LE, Raj SR, Peltier A, et al. Neurohumoral and haemodynamic profile in postural tachycardia and chronic fatigue syndromes. Clin Sci (Lond) 2012; 122(4):183–192. doi:10.1042/CS20110200
  35. Wang LB, Culbertson CJ, Deb A, Morgenshtern K, Huang H, Hohler AD. Gastrointestinal dysfunction in postural tachycardia syndrome. J Neurol Sci 2015; 359(1-2):193–196. doi:10.1016/j.jns.2015.10.052
  36. Raj S, Sheldon R. Management of postural tachycardia syndrome, inappropriate sinus tachycardia and vasovagal syncope. Arrhythm Electrophysiol Rev 2016; 5(2):122–129. doi:10.15420/AER.2016.7.2
  37. Peggs KJ, Nguyen H, Enayat D, Keller NR, Al-Hendy A, Raj SR. Gynecologic disorders and menstrual cycle lightheadedness in postural tachycardia syndrome. Int J Gynaecol Obstet 2012; 118(3):242–246. doi:10.1016/j.ijgo.2012.04.014
  38. Thieben MJ, Sandroni P, Sletten DM, et al. Postural orthostatic tachycardia syndrome: the Mayo Clinic experience. Mayo Clin Proc 2007; 82(3):308–313. doi:10.4065/82.3.308
  39. Deb A, Morgenshtern K, Culbertson CJ, Wang LB, Hohler AD. A survey-based analysis of symptoms in patients with postural orthostatic tachycardia syndrome. J Am Coll Cardiol 2015; 28(7):157–159. pmid:25829642
  40. Ertek S, Cicero AF. Hyperthyroidism and cardiovascular complications: a narrative review on the basis of pathophysiology. Arch Med Sci 2013; 9(5):944–952. doi:10.5114/aoms.2013.38685
  41. Rangno RE, Langlois S. Comparison of withdrawal phenomena after propranolol, metoprolol and pindolol. Br J Clin Pharmacol 1982; 13(suppl 2):345S–351S. pmid:6125187
  42. Nwazue VC, Paranjape SY, Black BK, et al. Postural tachycardia syndrome and inappropriate sinus tachycardia: role of autonomic modulation and sinus node automaticity. J Am Heart Assoc 2014; 3(2):e000700. doi:10.1161/JAHA.113.000700
  43. Morillo CA, Klein GJ, Thakur RK, Li H, Zardini M, Yee R. Mechanism of “inappropriate” sinus tachycardia. Role of sympathovagal balance. Circulation 1994; 90(2):873–877. pmid:7913886
  44. Grubb BP. Postural tachycardia syndrome. Circulation 2008; 117(21):2814–2817. doi:10.1161/CIRCULATIONAHA.107.761643
  45. Bhatia R, Kizilbash SJ, Ahrens SP, et al. Outcomes of adolescent-onset postural orthostatic tachycardia syndrome. J Pediatr 2016; 173:149–153. doi:10.1016/j.jpeds.2016.02.035
  46. George SA, Bivens TB, Howden EJ, et al. The international POTS registry: evaluating the efficacy of an exercise training intervention in a community setting. Heart Rhythm 2016; 13(4):943–950. doi:10.1016/j.hrthm.2015.12.012
  47. Fu Q, VanGundy TB, Galbreath MM, et al. Cardiac origins of the postural orthostatic tachycardia syndrome. J Am Coll Cardiol 2010; 55(25):2858–2868. doi:10.1016/j.jacc.2010.02.043
  48. Raj SR. Row, row, row your way to treating postural tachycardia syndrome. Heart Rhythm 2016; 13(4):951–952. doi:10.1016/j.hrthm.2015.12.039
  49. Celedonio JE, Garland EM, Nwazue VC, et al. Effects of high sodium intake on blood volume and catecholamines in patients with postural tachycardia syndrome and healthy females [abstract]. Clin Auton Res 2014; 24:211.
  50. Garland EM, Celedonio JE, Raj SR. Postural tachycardia syndrome: beyond orthostatic intolerance. Curr Neurol Neurosci Rep 2015; 15(9):60. doi:10.1007/s11910-015-0583-8
  51. Gordon VM, Opfer-Gehrking TL, Novak V, Low PA. Hemodynamic and symptomatic effects of acute interventions on tilt in patients with postural tachycardia syndrome. Clin Auton Res 2000; 10:29–33. pmid:10750641
  52. Jacob G, Shannon JR, Black B, et al. Effects of volume loading and pressor agents in idiopathic orthostatic tachycardia. Circulation 1997; 96(2):575–580. pmid:9244228
  53. Moak JP, Leong D, Fabian R, et al. Intravenous hydration for management of medication-resistant orthostatic intolerance in the adolescent and young adult. Pediatr Cardiol 2016; 37(2):278–282. doi:10.1007/s00246-015-1274-6
  54. Ruzieh M, Baugh A, Dasa O, et al. Effects of intermittent intravenous saline infusions in patients with medication-refractory postural tachycardia syndrome. J Interv Card Electrophysiol 2017; 48(3):255–260. doi:10.1007/s10840-017-0225-y
  55. Freitas J, Santos R, Azevedo E, Costa O, Carvalho M, de Freitas AF. Clinical improvement in patients with orthostatic intolerance after treatment with bisoprolol and fludrocortisone. Clin Auton Res 2000; 10(5):293–299. pmid:11198485
  56. Lee AK, Krahn AD. Evaluation of syncope: focus on diagnosis and treatment of neurally mediated syncope. Expert Rev Cardiovasc Ther 2016; 14(6):725–736. doi:10.1586/14779072.2016.1164034
  57. Coffin ST, Black BK, Biaggioni I, et al. Desmopressin acutely decreases tachycardia and improves symptoms in the postural tachycardia syndrome. Heart Rhythm 2012; 9(9):1484–1490. doi:10.1016/j.hrthm.2012.05.002
  58. Kanjwal K, Saeed B, Karabin B, Kanjwal Y, Sheikh M, Grubb BP. Erythropoietin in the treatment of postural orthostatic tachycardia syndrome. Am J Ther 2012; 19(2):92–95. doi:10.1097/MJT.0b013e3181ef621a
  59. Hoeldtke RD, Horvath GG, Bryner KD. Treatment of orthostatic tachycardia with erythropoietin. Am J Med 1995; 99(5):525–529. pmid:7485211
  60. Raj SR, Black BK, Biaggioni I, et al. Propranolol decreases tachycardia and improves symptoms in the postural tachycardia syndrome: less is more. Circulation 2009; 120(9):725–734. doi:10.1161/CIRCULATIONAHA.108.846501
  61. McDonald C, Frith J, Newton JL. Single centre experience of ivabradine in postural orthostatic tachycardia syndrome. Europace 2011; 13(3):427–430. doi:10.1093/europace/euq390
  62. Gaffney FA, Lane LB, Pettinger W, Blomqvist G. Effects of long-term clonidine administration on the hemodynamic and neuroendocrine postural responses of patients with dysautonomia. Chest 1983; 83(suppl 2):436–438. pmid:6295714
  63. Jacob G, Biaggioni I. Idiopathic orthostatic intolerance and postural tachycardia syndromes. Am J Med Sci 1999; 317(2):88–101. pmid:10037112
  64. Ross AJ, Ocon AJ, Medow MS, Stewart JM. A double-blind placebo-controlled cross-over study of the vascular effects of midodrine in neuropathic compared with hyperadrenergic postural tachycardia syndrome. Clin Sci (Lond) 2014; 126(4):289–296. doi:10.1042/CS20130222
  65. Raj SR, Black BK, Biaggioni I, Harris PA, Robertson D. Acetylcholinesterase inhibition improves tachycardia in postural tachycardia syndrome. Circulation 2005; 111(21):2734–2340. doi:10.1161/CIRCULATIONAHA.104.497594
  66. Kanjwal K, Karabin B, Sheikh M, et al. Pyridostigmine in the treatment of postural orthostatic tachycardia: A single-center experience. Pacing Clin Electrophysiol 2011; 34(6):750–755. doi:10.1111/j.1540-8159.2011.03047.x
  67. Ruzieh M, Dasa O, Pacenta A, Karabin B, Grubb B. Droxidopa in the treatment of postural orthostatic tachycardia syndrome. Am J Ther 2017; 24(2):e157–e161. doi:10.1097/MJT.0000000000000468
  68. Kpaeyeh AG Jr, Mar PL, Raj V, et al. Hemodynamic profiles and tolerability of modafinil in the treatment of POTS: a randomized placebo-controlled trial. J Clin Psychopharmacol 2014; 34(6):738–741. doi:10.1097/JCP.0000000000000221
  69. Lai CC, Fischer PR, Brands CK, et al. Outcomes in adolescents with postural orthostatic tachycardia syndrome treated with midodrine and beta-blockers. Pacing Clin Electrophysiol 2009; 32(2):234–238. doi:10.1111/j.1540-8159.2008.02207.x
Issue
Cleveland Clinic Journal of Medicine - 86(5)
Issue
Cleveland Clinic Journal of Medicine - 86(5)
Page Number
333-344
Page Number
333-344
Publications
Publications
Topics
Article Type
Display Headline
Evaluating and managing postural tachycardia syndrome
Display Headline
Evaluating and managing postural tachycardia syndrome
Legacy Keywords
postural tachycardia syndrome, POTS, autonomic nervous system, hypovolemia, hyperadrenergic, norepinephrine, mast cell activation syndrome, Ehlers-Danlos syndromes, tilt table, chronic fatigue syndrome, syncope, Lucy Lei, Derek Chew, Robert Sheldon, Satish Raj
Legacy Keywords
postural tachycardia syndrome, POTS, autonomic nervous system, hypovolemia, hyperadrenergic, norepinephrine, mast cell activation syndrome, Ehlers-Danlos syndromes, tilt table, chronic fatigue syndrome, syncope, Lucy Lei, Derek Chew, Robert Sheldon, Satish Raj
Sections
Inside the Article

KEY POINTS

  • Several POTS subtypes have been recognized, including hypovolemic, neuro­pathic, and hyperadrenergic forms, overlapping with Ehlers-Danlos syndrome, mast cell activation, and autoimmune syndromes. 
  • Treatment should take a graded approach, beginning with increasing salt and water intake, exercise, and compression stockings.
  • If needed, consider medications to expand blood volume, slow heart rate, or reduce central sympathetic tone.
  • Certain medications, including venodilators, diuretics, and serotonin-norepinephrine reuptake inhibitors, can exacerbate symptoms and should be avoided.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Wed, 04/24/2019 - 07:45
Un-Gate On Date
Wed, 04/24/2019 - 07:45
Use ProPublica
CFC Schedule Remove Status
Wed, 04/24/2019 - 07:45
Hide sidebar & use full width
render the right sidebar.
Article PDF Media

Gastric outlet obstruction: A red flag, potentially manageable

Article Type
Changed
Tue, 05/03/2022 - 15:14
Display Headline
Gastric outlet obstruction: A red flag, potentially manageable

A 72-year-old woman presents to the emergency department with progressive nausea and vomiting. One week earlier, she developed early satiety and nausea with vomiting after eating solid food. Three days later her symptoms progressed, and she became unable to take anything by mouth. The patient also experienced a 40-lb weight loss in the previous 3 months. She denies symptoms of abdominal pain, hematemesis, or melena. Her medical history includes cholecystectomy and type 2 diabetes mellitus, diagnosed 1 year ago. She has no family history of gastrointestinal malignancy. She says she smoked 1 pack a day in her 20s. She does not consume alcohol.

On physical examination, she is normotensive with a heart rate of 105 beats per minute. The oral mucosa is dry, and the abdomen is mildly distended and tender to palpation in the epigastrium. Laboratory evaluation reveals hypokalemia and metabolic alkalosis.

Computed tomography (CT) reveals a mass 3 cm by 4 cm in the pancreatic head. The mass has invaded the medial wall of the duodenum, with obstruction of the pancreatic and common bile ducts and extension into and occlusion of the superior mesenteric vein, with soft-tissue expansion around the superior mesenteric artery. CT also reveals retained stomach contents and an air-fluid level consistent with gastric outlet obstruction.

INTRINSIC OR EXTRINSIC BLOCKAGE

Gastric outlet obstruction, also called pyloric obstruction, is caused by intrinsic or extrinsic mechanical blockage of gastric emptying, generally in the distal stomach, pyloric channel, or duodenum, with associated symptoms of nausea, vomiting, abdominal pain, and early satiety. It is encountered in both the clinic and the hospital.

Here, we review the causes, diagnosis, and management of this disorder.

BENIGN AND MALIGNANT CAUSES

koop_gastricoutletobstruction_t1.jpg
Causes of obstruction are classified as either benign or malignant (Table 1). However, all cases of gastric outlet obstruction should be assumed to be due to underlying malignancy unless proven otherwise.1

In a retrospective study of 76 patients hospitalized with gastric outlet obstruction between 2006 and 2015 at our institution,2 29 cases (38%) were due to malignancy and 47 (62%) were due to benign causes. Pancreatic adenocarcinoma accounted for 13 cases (17%), while gastric adenocarcinoma accounted for 5 cases (7%); less common malignant causes were cholangiocarcinoma, cancer of the ampulla of Vater, duodenal adenocarcinoma, hepatocellular carcinoma, and metastatic disease. Of the benign causes, the most common were peptic ulcer disease (13 cases, 17%) and postoperative strictures or adhesions (11 cases, 14%).

These numbers reflect general trends around the world.

Less gastric cancer, more pancreatic cancer

The last several decades have seen a trend toward more cases due to cancer and fewer due to benign causes.3–14

In earlier studies in both developed and developing countries, gastric adenocarcinoma was the most common malignant cause of gastric outlet obstruction. Since then, it has become less common in Western countries, although it remains more common in Asia and Africa.7–14 This trend likely reflects environmental factors, including decreased prevalence of Helicobacter pylori infection, a major risk factor for gastric cancer, in Western countries.15–17

At the same time, pancreatic cancer is on the rise,16 and up to 20% of patients with pancreatic cancer develop gastric outlet obstruction.18 In a prospective observational study of 108 patients with malignant gastric outlet obstruction undergoing endoscopic stenting, pancreatic cancer was by far the most common malignancy, occurring in 54% of patients, followed by gastric cancer in 13%.19

Less peptic ulcer disease, but still common

Peptic ulcer disease used to account for up to 90% of cases of gastric outlet obstruction, and it is still the most common benign cause.

In 1990, gastric outlet obstruction was estimated to occur in 5% to 10% of all hospital admissions for ulcer-related complications, accounting for 2,000 operations annually.20,21 Gastric outlet obstruction now occurs in fewer than 5% of patients with duodenal ulcer disease and fewer than 2% of patients with gastric ulcer disease.22

Peptic ulcer disease remains an important cause of obstruction in countries with poor access to acid-suppressing drugs.23

Gastric outlet obstruction occurs in both acute and chronic peptic ulcer disease. In acute peptic ulcer disease, tissue inflammation and edema result in mechanical obstruction. Chronic peptic ulcer disease results in tissue scarring and fibrosis with strictures.20

Environmental factors, including improved diet, hygiene, physical activity, and the decreased prevalence of H pylori infection, also contribute to the decreased prevalence of peptic ulcer disease and its complications, including gastric outlet obstruction.3 The continued occurrence of peptic ulcer disease is associated with widespread use of low-dose aspirin and nonsteroidal anti-inflammatory drugs (NSAIDs), the most common causes of peptic ulcer disease in Western countries.24,25

Other nonmalignant causes of gastric outlet obstruction are diverse and less common. They include caustic ingestion, postsurgical strictures, benign tumors of the gastrointestinal tract, Crohn disease, and pancreatic disorders including acute pancreatitis, pancreatic pseudocyst, chronic pancreatitis, and annular pancreas. Intramural duodenal hematoma may cause obstruction after blunt abdominal trauma, endoscopic biopsy, or gastrostomy tube migration, especially in the setting of a bleeding disorder or anticoagulation.26

Tuberculosis should be suspected in countries in which it is common.7 In a prospective study of 64 patients with benign gastric outlet obstruction in India,27 16 (25%) had corrosive injury, 16 (25%) had tuberculosis, and 15 (23%) had peptic ulcer disease. Compared with patients with corrosive injury and peptic ulcer disease, patients with gastroduodenal tuberculosis had the best outcomes with appropriate treatment.

Other reported causes include Bouveret syndrome (an impacted gallstone in the proximal duodenum), phytobezoar, diaphragmatic hernia, gastric volvulus, and Ladd bands (peritoneal bands associated with intestinal malrotation).7,28,29

 

 

PRESENTING SYMPTOMS

Symptoms of gastric outlet obstruction include nausea, nonbilious vomiting, epigastric pain, early satiety, abdominal distention, and weight loss.

In our patients, the most common presenting symptoms were nausea and vomiting (80%), followed by abdominal pain (72%); weight loss (15%), abdominal distention (15%), and early satiety (9%) were less common.2

Patients with gastric outlet obstruction secondary to malignancy generally present with a shorter duration of symptoms than those with peptic ulcer disease and are more likely to be older.8,13 Other conditions with an acute onset of symptoms include gastric polyp prolapse, percutaneous endoscopic gastrostomy tube migration, gastric volvulus, and gallstone impaction.

Patients with gastric outlet obstruction associated with peptic ulcer disease generally have a long-standing history of symptoms, including dyspepsia and weight loss over several years.4

SIGNS ON EXAMINATION

On examination, look for signs of chronic gastric obstruction and its consequences, such as malnutrition, cachexia, volume depletion, and dental erosions.

A succussion splash may suggest gastric outlet obstruction. This is elicited by rocking the patient back and forth by the hips or abdomen while listening over the stomach for a splash, which may be heard without a stethoscope. The test is considered positive if present 3 or more hours after drinking fluids and suggests retention of gastric materials.30,31

In thin individuals, chronic gastric outlet obstruction makes the stomach dilate and hypertrophy, which may be evident by a palpably thickened stomach with visible gastric peristalsis.4

Other notable findings on physical examination may include a palpable abdominal mass, epigastric pain, or an abnormality suggestive of metastatic gastric cancer, such as an enlarged left supraclavicular lymph node (Virchow node) or periumbilical lymph node (Sister Mary Joseph nodule). The Virchow node is at the junction of the thoracic duct and the left subclavian vein where the lymphatic circulation from the body drains into the systemic circulation, and it may be the first sign of gastric cancer.32 Sister Mary Joseph nodule (named after a surgical assistant to Dr. William James Mayo) refers to a palpable mass at the umbilicus, generally resulting from metastasis of an abdominal malignancy.33

SIGNS ON FURTHER STUDIES

Laboratory evaluation may show signs of poor oral intake and electrolyte abnormalities secondary to chronic nausea, vomiting, and dehydration, including hypochloremic metabolic alkalosis and hypokalemia.

The underlying cause of gastric outlet obstruction has major implications for treatment and prognosis and cannot be differentiated by clinical presentation alone.1,9 Diagnosis is based on clinical features and radiologic or endoscopic evaluation consistent with gastric outlet obstruction.

Plain radiography may reveal an enlarged gastric bubble, and contrast studies may be useful to determine whether the obstruction is partial or complete, depending on whether the contrast passes into the small bowel.

koop_gastricoutletobstruction_f1.jpg
%3Cp%3EFigure%201.%20Computed%20tomography%20of%20the%20abdomen%20in%20the%20axial%20plane%20shows%20gastric%20distention%20(A%2C%20arrow)%20and%20a%203.9-cm%20mass%20at%20the%20pancreatic%20head%2C%20with%20compression%20of%20the%20descending%20duodenum%20(B%2C%20arrow)%2C%20resulting%20in%20gastric%20outlet%20obstruction.%20The%20patient%2C%20a%2072-year-old%20woman%2C%20presented%20with%201%20week%20of%20nausea%20and%20vomiting%20and%20was%20found%20to%20have%20pancreatic%20cancer.%20She%20was%20treated%20with%20endoscopic%20stenting.%3C%2Fp%3E
CT or magnetic resonance imaging may show gastric distention with retained stomach contents, suggesting a gastric, pyloric, duodenal, or pancreatic mass (Figure 1).

Upper endoscopy is often needed to establish the diagnosis and cause. Emptying the stomach with a nasogastric tube is recommended before endoscopy to minimize the risk of aspiration during the procedure, and endotracheal intubation should be considered for airway protection.34 Findings of gastric outlet obstruction on upper endoscopy include retained food and liquid. Endoscopic biopsy is important to differentiate between benign and malignant causes. For patients with malignancy, endoscopic ultrasonography is useful for diagnosis via tissue sampling with fine-needle aspiration and locoregional staging.35

A strategy. Most patients whose clinical presentation suggests gastric outlet obstruction require cross-sectional radiologic imaging, upper endoscopy, or both.36 CT is the preferred imaging study to evaluate for intestinal obstruction.36,37 Patients with suspected complete obstruction or perforation should undergo CT before upper endoscopy. Oral contrast may interfere with endoscopy and should be avoided if endoscopy is planned. Additionally, giving oral contrast may worsen patient discomfort and increase the risk of nausea, vomiting, and aspiration.36,37

Following radiographic evaluation, upper endoscopy can be performed after gastric decompression to identify the location and extent of the obstruction and to potentially provide a definitive diagnosis with biopsy.36

DIFFERENTIATE FROM GASTROPARESIS

Gastroparesis is a chronic neuromuscular disorder characterized by delayed gastric emptying without mechanical obstruction.38 The most common causes are diabetes, surgery, and idiopathy. Other causes include viral infection, connective tissue diseases, ischemia, infiltrative disorders, radiation, neurologic disorders, and paraneoplastic syndromes.39,40

Gastric outlet obstruction and gastroparesis share clinical symptoms including nausea, vomiting, abdominal pain, early satiety, and weight loss and are important to differentiate.36,38 Although abdominal pain may be present in both gastric outlet obstruction and gastroparesis, in gastroparesis it tends not to be the dominant symptom.40

Gastric scintigraphy is most commonly used to objectively quantify delayed gastric emptying.39 Upper endoscopy is imperative to exclude mechanical obstruction.39

 

 

MANAGEMENT

Initially, patients with signs and symptoms of gastric outlet obstruction should be given:

  • Nothing by mouth (NPO)
  • Intravenous fluids to correct volume depletion and electrolyte abnormalities
  • A nasogastric tube for gastric decompression and symptom relief if symptoms persist despite being NPO
  • A parenteral proton pump inhibitor, regardless of the cause of obstruction, to decrease gastric secretions41
  • Medications for pain and nausea, if needed.

Definitive treatment of gastric outlet obstruction depends on the underlying cause, whether benign or malignant.

Management of benign gastric outlet obstruction

Symptoms of gastric outlet obstruction resolve spontaneously in about half of cases caused by acute peptic ulcer disease, as acute inflammation resolves.9,22

Endoscopic dilation is an important option in patients with benign gastric outlet obstruction, including peptic ulcer disease. Peptic ulcer disease-induced gastric outlet obstruction can be safely treated with endoscopic balloon dilation. This treatment almost always relieves symptoms immediately; however, the long-term response has varied from 16% to 100%, and patients may require more than 1 dilation procedure.25,42,43 The need for 2 or more dilation procedures may predict need for surgery.44 Gastric outlet obstruction after caustic ingestion or endoscopic submucosal dissection may also respond to endoscopic balloon dilation.36

Eradication of H pylori may be effective and lead to complete resolution of symptoms in patients with gastric outlet obstruction due to this infection.45–47

NSAIDs should be discontinued in patients with peptic ulcer disease and gastric outlet obstruction. These drugs damage the gastrointestinal mucosa by inhibiting cyclo-oxygenase (COX) enzymes and decreasing synthesis of prostaglandins, which are important for mucosal defense.48 Patients may be unaware of NSAIDs contained in over-the-counter medications and may have difficulty discontinuing NSAIDs taken for pain.49

These drugs are an important cause of refractory peptic ulcer disease and can be detected by platelet COX activity testing, although this test is not widely available. In a study of patients with peptic ulcer disease without definite NSAID use or H pylori infection, up to one-third had evidence of surreptitious NSAID use as detected by platelet COX activity testing.50 In another study,51 platelet COX activity testing discovered over 20% more aspirin users than clinical history alone.

Surgery for patients with benign gastric outlet obstruction is used only when medical management and endoscopic dilation fail. Ideally, surgery should relieve the obstruction and target the underlying cause, such as peptic ulcer disease. Laparoscopic surgery is generally preferred to open surgery because patients can resume oral intake sooner, have a shorter hospital stay, and have less intraoperative blood loss.52 The simplest surgical procedure to relieve obstruction is laparoscopic gastrojejunostomy.

Patients with gastric outlet obstruction and peptic ulcer disease warrant laparoscopic vagotomy and antrectomy or distal gastrectomy. This removes the obstruction and the stimulus for gastric secretion.53 An alternative is vagotomy with a drainage procedure (pyloroplasty or gastrojejunostomy), which has a similar postoperative course and reduction in gastric acid secretion compared with antrectomy or distal gastrectomy.53,54

Daily proton pump inhibitors can be used for patients with benign gastric outlet obstruction not associated with peptic ulcer disease or risk factors; for such cases, vagotomy is not required.

Management of malignant gastric outlet obstruction

Patients with malignant gastric outlet obstruction may have intractable nausea and abdominal pain secondary to retention of gastric contents. The major goal of therapy is to improve symptoms and restore tolerance of an oral diet. The short-term prognosis of malignant gastric outlet obstruction is poor, with a median survival of 3 to 4 months, as these patients often have unresectable disease.55

Surgical bypass used to be the standard of care for palliation of malignant gastric obstruction, but that was before endoscopic stenting was developed.

Endoscopic stenting allows patients to resume oral intake and get out of the hospital sooner with fewer complications than with open surgical bypass. It may be a more appropriate option for palliation of symptoms in patients with malignant obstruction who have a poor prognosis and prefer a less invasive intervention.55,56

koop_gastricoutletobstruction_f2.jpg
%3Cp%3EFigure%202.%20Esophagogastroduodenoscopy%20(A)%20shows%20a%20large%20submucosal%20mass%20in%20the%20duodenal%20bulb%20(upper%20arrow)%2C%20with%20localized%20erosions%20(lower%20arrow).%20The%20mass%20was%2040%20%C3%97%2041%20mm%20in%20cross-sectional%20diameter%20on%20endoscopic%20ultrasonography.%20Fine-needle%20aspiration%20and%20pathology%20study%20revealed%20pancreatic%20adenocarcinoma.%20The%20obstruction%20was%20successfully%20opened%20(B)%20with%20a%2022-mm%20%C3%97%2012-cm%20WallFlex%20stent%20(Boston%20Scientific).%20The%20patient%20tolerated%20a%20liquid%20diet%20after%20the%20procedure.%3C%2Fp%3E

Endoscopic duodenal stenting of malignant gastric outlet obstruction has a success rate of greater than 90%, and most patients can tolerate a mechanical soft diet afterward.34 The procedure is usually performed with a 9-cm or 12-cm self-expanding duodenal stent, 22 mm in diameter, placed over a guide wire under endoscopic and fluoroscopic guidance (Figure 2). The stent is placed by removing the outer catheter, with distal-to-proximal stent deployment.

Patients who also have biliary obstruction may require biliary stent placement, which is generally performed before duodenal stenting. For patients with an endoscopic stent who develop biliary obstruction, endoscopic retrograde cholangiopancreatography can be attempted with placement of a biliary stent; however, these patients may require biliary drain placement by percutaneous transhepatic cholangiography or by endoscopic ultrasonographically guided transduodenal or transgastric biliary drainage.

From 20% to 30% of patients require repeated endoscopic stent placement, although most patients die within several months after stenting.34 Surgical options for patients who do not respond to endoscopic stenting include open or laparoscopic gastrojejunostomy.55

Laparoscopic gastrojejunostomy may provide better long-term outcomes than duodenal stenting for patients with malignant gastric outlet obstruction and a life expectancy longer than a few months.

A 2017 retrospective study of 155 patients with gastric outlet obstruction secondary to unresectable gastric cancer suggested that those who underwent laparoscopic gastrojejunostomy had better oral intake, better tolerance of chemotherapy, and longer overall survival than those who underwent duodenal stenting. Postsurgical complications were more common in the laparoscopic gastrojejunostomy group (16%) than in the duodenal stenting group (0%).57

In most of the studies comparing endoscopic stenting with surgery, the surgery was open gastrojejunostomy; there are limited data directly comparing stenting with laparoscopic gastrojejunostomy.55 Endoscopic stenting is estimated to be significantly less costly than surgery, with a median cost of $12,000 less than gastrojejunostomy.58 As an alternative to enteral stenting and surgical gastrojejunostomy, ultrasonography-guided endoscopic gastrojejunostomy or gastroenterostomy with placement of a lumen-apposing metal stent is emerging as a third treatment option and is under active investigation.59

Patients with malignancy that is potentially curable by resection should undergo surgical evaluation before consideration of endoscopic stenting. For patients who are not candidates for surgery or endoscopic stenting, a percutaneous gastrostomy tube can be considered for gastric decompression and symptom relief.

CASE CONCLUDED

The patient underwent esophagogastroduodenoscopy with endoscopic ultrasonography for evaluation of her pancreatic mass. Before the procedure, she was intubated to minimize the risk of aspiration due to persistent nausea and retained gastric contents. A large submucosal mass was found in the duodenal bulb. Endoscopic ultrasonography showed a mass within the pancreatic head with pancreatic duct obstruction. Fine-needle aspiration biopsy was performed, and pathology study revealed pancreatic adenocarcinoma. The patient underwent stenting with a 22-mm by 12-cm WallFlex stent (Boston Scientific), which led to resolution of nausea and advancement to a mechanical soft diet on hospital discharge.

She was scheduled for follow-up in the outpatient clinic for treatment of pancreatic cancer.

References
  1. Johnson CD. Gastric outlet obstruction malignant until proved otherwise. Am J Gastroenterol 1995; 90(10):1740. pmid:7572886
  2. Koop AH, Palmer WC, Mareth K, Burton MC, Bowman A, Stancampiano F. Tu1335 - Pancreatic cancer most common cause of malignant gastric outlet obstruction at a tertiary referral center: a 10 year retrospective study [abstract]. Gastroenterology 2018; 154(6, suppl 1):S-1343.
  3. Hall R, Royston C, Bardhan KD. The scars of time: the disappearance of peptic ulcer-related pyloric stenosis through the 20th century. J R Coll Physicians Edinb 2014; 44(3):201–208. doi:10.4997/JRCPE.2014.303
  4. Kreel L, Ellis H. Pyloric stenosis in adults: a clinical and radiological study of 100 consecutive patients. Gut 1965; 6(3):253–261. pmid:18668780
  5. Shone DN, Nikoomanesh P, Smith-Meek MM, Bender JS. Malignancy is the most common cause of gastric outlet obstruction in the era of H2 blockers. Am J Gastroenterol 1995; 90(10):1769–1770. pmid:7572891
  6. Ellis H. The diagnosis of benign and malignant pyloric obstruction. Clin Oncol 1976; 2(1):11–15. pmid:1277618
  7. Samad A, Khanzada TW, Shoukat I. Gastric outlet obstruction: change in etiology. Pak J Surg 2007; 23(1):29–32.
  8. Chowdhury A, Dhali GK, Banerjee PK. Etiology of gastric outlet obstruction. Am J Gastroenterol 1996; 91(8):1679. pmid:8759707
  9. Johnson CD, Ellis H. Gastric outlet obstruction now predicts malignancy. Br J Surg 1990; 77(9):1023–1024. pmid:2207566
  10. Misra SP, Dwivedi M, Misra V. Malignancy is the most common cause of gastric outlet obstruction even in a developing country. Endoscopy 1998; 30(5):484–486. doi:10.1055/s-2007-1001313
  11. Essoun SD, Dakubo JCB. Update of aetiological patterns of adult gastric outlet obstruction in Accra, Ghana. Int J Clin Med 2014; 5(17):1059–1064. doi:10.4236/ijcm.2014.517136
  12. Jaka H, Mchembe MD, Rambau PF, Chalya PL. Gastric outlet obstruction at Bugando Medical Centre in Northwestern Tanzania: a prospective review of 184 cases. BMC Surg 2013; 13:41. doi:10.1186/1471-2482-13-41
  13. Sukumar V, Ravindran C, Prasad RV. Demographic and etiological patterns of gastric outlet obstruction in Kerala, South India. N Am J Med Sci 2015; 7(9):403–406. doi:10.4103/1947-2714.166220
  14. Yoursef M, Mirza MR, Khan S. Gastric outlet obstruction. Pak J Surg 2005; 10(4):48–50.
  15. Ferlay J, Soerjomataram I, Dikshit R, et al. Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer 2015; 136(5):E359–E386. doi:10.1002/ijc.29210
  16. Parkin DM, Stjernsward J, Muir CS. Estimates of the worldwide frequency of twelve major cancers. Bull World Health Organ 1984; 62(2):163–182. pmid:6610488
  17. Karimi P, Islami F, Anandasabapathy S, Freedman ND, Kamangar F. Gastric cancer: descriptive epidemiology, risk factors, screening, and prevention. Cancer Epidemiol Biomarkers Prev 2014; 23(5):700–713. doi:10.1158/1055-9965.EPI-13-1057
  18. Jeurnink SM, Steyerberg EW, van Hooft JE, et al; Dutch SUSTENT Study Group. Surgical gastrojejunostomy or endoscopic stent placement for the palliation of malignant gastric outlet obstruction (SUSTENT) study): a multicenter randomized trial. Gastrointest Endosc 2010; 71(3):490–499. doi:10.1016/j.gie.2009.09.042
  19. Tringali A, Didden P, Repici A, et al. Endoscopic treatment of malignant gastric and duodenal strictures: a prospective, multicenter study. Gastrointest Endosc 2014; 79(1):66–75. doi:10.1016/j.gie.2013.06.032
  20. Malfertheiner P, Chan FK, McColl KE. Peptic ulcer disease. Lancet 2009; 374(9699):1449–1461. doi:10.1016/S0140-6736(09)60938-7
  21. Gibson JB, Behrman SW, Fabian TC, Britt LG. Gastric outlet obstruction resulting from peptic ulcer disease requiring surgical intervention is infrequently associated with Helicobacter pylori infection. J Am Coll Surg 2000; 191(1):32–37. pmid:10898181
  22. Kochhar R, Kochhar S. Endoscopic balloon dilation for benign gastric outlet obstruction in adults. World J Gastrointest Endosc 2010; 2(1):29–35. doi:10.4253/wjge.v2.i1.29
  23. Kotisso R. Gastric outlet obstruction in Northwestern Ethiopia. East Cent Afr J Surg 2000; 5(2):25-29.
  24. Hamzaoui L, Bouassida M, Ben Mansour I, et al. Balloon dilatation in patients with gastric outlet obstruction related to peptic ulcer disease. Arab J Gastroenterol 2015; 16(3–4):121–124. doi:10.1016/j.ajg.2015.07.004
  25. Najm WI. Peptic ulcer disease. Prim Care 2011; 38(3):383–394. doi:10.1016/j.pop.2011.05.001
  26. Veloso N, Amaro P, Ferreira M, Romaozinho JM, Sofia C. Acute pancreatitis associated with a nontraumatic, intramural duodenal hematoma. Endoscopy 2013; 45(suppl 2):E51–E52. doi:10.1055/s-0032-1325969
  27. Maharshi S, Puri AS, Sachdeva S, Kumar A, Dalal A, Gupta M. Aetiological spectrum of benign gastric outlet obstruction in India: new trends. Trop Doct 2016; 46(4):186–191. doi:10.1177/0049475515626032
  28. Sala MA, Ligabo AN, de Arruda MC, Indiani JM, Nacif MS. Intestinal malrotation associated with duodenal obstruction secondary to Ladd’s bands. Radiol Bras 2016; 49(4):271–272. doi:10.1590/0100-3984.2015.0106
  29. Alibegovic E, Kurtcehajic A, Hujdurovic A, Mujagic S, Alibegovic J, Kurtcehajic D. Bouveret syndrome or gallstone ileus. Am J Med 2018; 131(4):e175. doi:10.1016/j.amjmed.2017.10.044
  30. Lau JY, Chung SC, Sung JJ, et al. Through-the-scope balloon dilation for pyloric stenosis: long-term results. Gastrointest Endosc 1996; 43(2 Pt 1):98–101. pmid:8635729
  31. Ray K, Snowden C, Khatri K, McFall M. Gastric outlet obstruction from a caecal volvulus, herniated through epiploic foramen: a case report. BMJ Case Rep 2009; pii:bcr05.2009.1880. doi:10.1136/bcr.05.2009.1880
  32. Baumgart DC, Fischer A. Virchow’s node. Lancet 2007; 370(9598):1568. doi:10.1016/S0140-6736(07)61661-4
  33. Dar IH, Kamili MA, Dar SH, Kuchaai FA. Sister Mary Joseph nodule—a case report with review of literature. J Res Med Sci 2009; 14(6):385–387. pmid:21772912
  34. Tang SJ. Endoscopic stent placement for gastric outlet obstruction. Video Journal and Encyclopedia of GI Endoscopy 2013; 1(1):133–136.
  35. Valero M, Robles-Medranda C. Endoscopic ultrasound in oncology: an update of clinical applications in the gastrointestinal tract. World J Gastrointest Endosc 2017; 9(6):243–254.
  36. ASGE Standards of Practice Committee; Fukami N, Anderson MA, Khan K, et al. The role of endoscopy in gastroduodenal obstruction and gastroparesis. Gastrointest Endosc 2011; 74(1):13–21. doi:10.1016/j.gie.2010.12.003
  37. Ros PR, Huprich JE. ACR appropriateness criteria on suspected small-bowel obstruction. J Am Coll Radiol 2006; 3(11):838–841. doi:10.1016/j.jacr.2006.09.018
  38. Pasricha PJ, Parkman HP. Gastroparesis: definitions and diagnosis. Gastroenterol Clin North Am 2015; 44(1):1–7. doi:10.1016/j.gtc.2014.11.001
  39. Stein B, Everhart KK, Lacy BE. Gastroparesis: a review of current diagnosis and treatment options. J Clin Gastroenterol 2015; 49(7):550–558. doi:10.1097/MCG.0000000000000320
  40. Camilleri M, Parkman HP, Shafi MA, Abell TL, Gerson L; American College of Gastroenterology. Clinical guideline: management of gastroparesis. Am J Gastroenterol 2013; 108(1):18–37.
  41. Gursoy O, Memis D, Sut N. Effect of proton pump inhibitors on gastric juice volume, gastric pH and gastric intramucosal pH in critically ill patients: a randomized, double-blind, placebo-controlled study. Clin Drug Investig 2008; 28(12):777–782. doi:10.2165/0044011-200828120-00005
  42. Kuwada SK, Alexander GL. Long-term outcome of endoscopic dilation of nonmalignant pyloric stenosis. Gastrointest Endosc 1995; 41(1):15–17. pmid:7698619
  43. Kochhar R, Sethy PK, Nagi B, Wig JD. Endoscopic balloon dilatation of benign gastric outlet obstruction. J Gastroenterol Hepatol 2004; 19(4):418–422. pmid:15012779
  44. Perng CL, Lin HJ, Lo WC, Lai CR, Guo WS, Lee SD. Characteristics of patients with benign gastric outlet obstruction requiring surgery after endoscopic balloon dilation. Am J Gastroenterol 1996; 91(5):987–990. pmid:8633593
  45. Taskin V, Gurer I, Ozyilkan E, Sare M, Hilmioglu F. Effect of Helicobacter pylori eradication on peptic ulcer disease complicated with outlet obstruction. Helicobacter 2000; 5(1):38–40. pmid:10672050
  46. de Boer WA, Driessen WM. Resolution of gastric outlet obstruction after eradication of Helicobacter pylori. J Clin Gastroenterol 1995; 21(4):329–330. pmid:8583113
  47. Tursi A, Cammarota G, Papa A, Montalto M, Fedeli G, Gasbarrini G. Helicobacter pylori eradication helps resolve pyloric and duodenal stenosis. J Clin Gastroenterol 1996; 23(2):157–158. pmid:8877648
  48. Schmassmann A. Mechanisms of ulcer healing and effects of nonsteroidal anti-inflammatory drugs. Am J Med 1998; 104(3A):43S–51S; discussion 79S–80S. pmid:9572320
  49. Kim HU. Diagnostic and treatment approaches for refractory peptic ulcers. Clin Endosc 2015; 48(4):285–290. doi:10.5946/ce.2015.48.4.285
  50. Ong TZ, Hawkey CJ, Ho KY. Nonsteroidal anti-inflammatory drug use is a significant cause of peptic ulcer disease in a tertiary hospital in Singapore: a prospective study. J Clin Gastroenterol 2006; 40(9):795–800. doi:10.1097/01.mcg.0000225610.41105.7f
  51. Lanas A, Sekar MC, Hirschowitz BI. Objective evidence of aspirin use in both ulcer and nonulcer upper and lower gastrointestinal bleeding. Gastroenterology 1992; 103(3):862–869. pmid:1499936
  52. Zhang LP, Tabrizian P, Nguyen S, Telem D, Divino C. Laparoscopic gastrojejunostomy for the treatment of gastric outlet obstruction. JSLS 2011; 15(2):169–173. doi:10.4293/108680811X13022985132074
  53. Lagoo J, Pappas TN, Perez A. A relic or still relevant: the narrowing role for vagotomy in the treatment of peptic ulcer disease. Am J Surg 2014; 207(1):120–126. doi:10.1016/j.amjsurg.2013.02.012
  54. Csendes A, Maluenda F, Braghetto I, Schutte H, Burdiles P, Diaz JC. Prospective randomized study comparing three surgical techniques for the treatment of gastric outlet obstruction secondary to duodenal ulcer. Am J Surg 1993; 166(1):45–49. pmid:8101050
  55. Ly J, O’Grady G, Mittal A, Plank L, Windsor JA. A systematic review of methods to palliate malignant gastric outlet obstruction. Surg Endosc 2010; 24(2):290–297. doi:10.1007/s00464-009-0577-1
  56. Goldberg EM. Palliative treatment of gastric outlet obstruction in terminal patients: SEMS. Stent every malignant stricture! Gastrointest Endosc 2014; 79(1):76–78. doi:10.1016/j.gie.2013.07.056
  57. Min SH, Son SY, Jung DH, et al. Laparoscopic gastrojejunostomy versus duodenal stenting in unresectable gastric cancer with gastric outlet obstruction. Ann Surg Treat Res 2017; 93(3):130–136. doi:10.4174/astr.2017.93.3.130
  58. Roy A, Kim M, Christein J, Varadarajulu S. Stenting versus gastrojejunostomy for management of malignant gastric outlet obstruction: comparison of clinical outcomes and costs. Surg Endosc 2012; 26(11):3114–119. doi:10.1007/s00464-012-2301-9
  59. Amin S, Sethi A. Endoscopic ultrasound-guided gastrojejunostomy. Gastrointest Endosc Clin N Am 2017; 27(4):707–713. doi:10.1016/j.giec.2017.06.009
Article PDF
Author and Disclosure Information

Andree H. Koop, MD
Division of Community Internal Medicine, Mayo Clinic, Jacksonville, FL

William C. Palmer, MD
Division of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, FL

Fernando F. Stancampiano, MD
Division of Community Internal Medicine, Mayo Clinic, Jacksonville, FL

Address: Fernando F. Stancampiano, MD, Division of Community Internal Medicine, Mayo Clinic, 4500 San Pablo Road S, Jacksonville, FL 32224; stancampiano.f@mayo.edu

Issue
Cleveland Clinic Journal of Medicine - 86(5)
Publications
Topics
Page Number
345-353
Legacy Keywords
gastric outlet obstruction, GOO, gastric cancer, pancreatic cancer, endoscopic stenting, peptic ulcer disease, Andree Koop, William Palmer, Fernando Stancampiano
Sections
Author and Disclosure Information

Andree H. Koop, MD
Division of Community Internal Medicine, Mayo Clinic, Jacksonville, FL

William C. Palmer, MD
Division of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, FL

Fernando F. Stancampiano, MD
Division of Community Internal Medicine, Mayo Clinic, Jacksonville, FL

Address: Fernando F. Stancampiano, MD, Division of Community Internal Medicine, Mayo Clinic, 4500 San Pablo Road S, Jacksonville, FL 32224; stancampiano.f@mayo.edu

Author and Disclosure Information

Andree H. Koop, MD
Division of Community Internal Medicine, Mayo Clinic, Jacksonville, FL

William C. Palmer, MD
Division of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, FL

Fernando F. Stancampiano, MD
Division of Community Internal Medicine, Mayo Clinic, Jacksonville, FL

Address: Fernando F. Stancampiano, MD, Division of Community Internal Medicine, Mayo Clinic, 4500 San Pablo Road S, Jacksonville, FL 32224; stancampiano.f@mayo.edu

Article PDF
Article PDF
Related Articles

A 72-year-old woman presents to the emergency department with progressive nausea and vomiting. One week earlier, she developed early satiety and nausea with vomiting after eating solid food. Three days later her symptoms progressed, and she became unable to take anything by mouth. The patient also experienced a 40-lb weight loss in the previous 3 months. She denies symptoms of abdominal pain, hematemesis, or melena. Her medical history includes cholecystectomy and type 2 diabetes mellitus, diagnosed 1 year ago. She has no family history of gastrointestinal malignancy. She says she smoked 1 pack a day in her 20s. She does not consume alcohol.

On physical examination, she is normotensive with a heart rate of 105 beats per minute. The oral mucosa is dry, and the abdomen is mildly distended and tender to palpation in the epigastrium. Laboratory evaluation reveals hypokalemia and metabolic alkalosis.

Computed tomography (CT) reveals a mass 3 cm by 4 cm in the pancreatic head. The mass has invaded the medial wall of the duodenum, with obstruction of the pancreatic and common bile ducts and extension into and occlusion of the superior mesenteric vein, with soft-tissue expansion around the superior mesenteric artery. CT also reveals retained stomach contents and an air-fluid level consistent with gastric outlet obstruction.

INTRINSIC OR EXTRINSIC BLOCKAGE

Gastric outlet obstruction, also called pyloric obstruction, is caused by intrinsic or extrinsic mechanical blockage of gastric emptying, generally in the distal stomach, pyloric channel, or duodenum, with associated symptoms of nausea, vomiting, abdominal pain, and early satiety. It is encountered in both the clinic and the hospital.

Here, we review the causes, diagnosis, and management of this disorder.

BENIGN AND MALIGNANT CAUSES

koop_gastricoutletobstruction_t1.jpg
Causes of obstruction are classified as either benign or malignant (Table 1). However, all cases of gastric outlet obstruction should be assumed to be due to underlying malignancy unless proven otherwise.1

In a retrospective study of 76 patients hospitalized with gastric outlet obstruction between 2006 and 2015 at our institution,2 29 cases (38%) were due to malignancy and 47 (62%) were due to benign causes. Pancreatic adenocarcinoma accounted for 13 cases (17%), while gastric adenocarcinoma accounted for 5 cases (7%); less common malignant causes were cholangiocarcinoma, cancer of the ampulla of Vater, duodenal adenocarcinoma, hepatocellular carcinoma, and metastatic disease. Of the benign causes, the most common were peptic ulcer disease (13 cases, 17%) and postoperative strictures or adhesions (11 cases, 14%).

These numbers reflect general trends around the world.

Less gastric cancer, more pancreatic cancer

The last several decades have seen a trend toward more cases due to cancer and fewer due to benign causes.3–14

In earlier studies in both developed and developing countries, gastric adenocarcinoma was the most common malignant cause of gastric outlet obstruction. Since then, it has become less common in Western countries, although it remains more common in Asia and Africa.7–14 This trend likely reflects environmental factors, including decreased prevalence of Helicobacter pylori infection, a major risk factor for gastric cancer, in Western countries.15–17

At the same time, pancreatic cancer is on the rise,16 and up to 20% of patients with pancreatic cancer develop gastric outlet obstruction.18 In a prospective observational study of 108 patients with malignant gastric outlet obstruction undergoing endoscopic stenting, pancreatic cancer was by far the most common malignancy, occurring in 54% of patients, followed by gastric cancer in 13%.19

Less peptic ulcer disease, but still common

Peptic ulcer disease used to account for up to 90% of cases of gastric outlet obstruction, and it is still the most common benign cause.

In 1990, gastric outlet obstruction was estimated to occur in 5% to 10% of all hospital admissions for ulcer-related complications, accounting for 2,000 operations annually.20,21 Gastric outlet obstruction now occurs in fewer than 5% of patients with duodenal ulcer disease and fewer than 2% of patients with gastric ulcer disease.22

Peptic ulcer disease remains an important cause of obstruction in countries with poor access to acid-suppressing drugs.23

Gastric outlet obstruction occurs in both acute and chronic peptic ulcer disease. In acute peptic ulcer disease, tissue inflammation and edema result in mechanical obstruction. Chronic peptic ulcer disease results in tissue scarring and fibrosis with strictures.20

Environmental factors, including improved diet, hygiene, physical activity, and the decreased prevalence of H pylori infection, also contribute to the decreased prevalence of peptic ulcer disease and its complications, including gastric outlet obstruction.3 The continued occurrence of peptic ulcer disease is associated with widespread use of low-dose aspirin and nonsteroidal anti-inflammatory drugs (NSAIDs), the most common causes of peptic ulcer disease in Western countries.24,25

Other nonmalignant causes of gastric outlet obstruction are diverse and less common. They include caustic ingestion, postsurgical strictures, benign tumors of the gastrointestinal tract, Crohn disease, and pancreatic disorders including acute pancreatitis, pancreatic pseudocyst, chronic pancreatitis, and annular pancreas. Intramural duodenal hematoma may cause obstruction after blunt abdominal trauma, endoscopic biopsy, or gastrostomy tube migration, especially in the setting of a bleeding disorder or anticoagulation.26

Tuberculosis should be suspected in countries in which it is common.7 In a prospective study of 64 patients with benign gastric outlet obstruction in India,27 16 (25%) had corrosive injury, 16 (25%) had tuberculosis, and 15 (23%) had peptic ulcer disease. Compared with patients with corrosive injury and peptic ulcer disease, patients with gastroduodenal tuberculosis had the best outcomes with appropriate treatment.

Other reported causes include Bouveret syndrome (an impacted gallstone in the proximal duodenum), phytobezoar, diaphragmatic hernia, gastric volvulus, and Ladd bands (peritoneal bands associated with intestinal malrotation).7,28,29

 

 

PRESENTING SYMPTOMS

Symptoms of gastric outlet obstruction include nausea, nonbilious vomiting, epigastric pain, early satiety, abdominal distention, and weight loss.

In our patients, the most common presenting symptoms were nausea and vomiting (80%), followed by abdominal pain (72%); weight loss (15%), abdominal distention (15%), and early satiety (9%) were less common.2

Patients with gastric outlet obstruction secondary to malignancy generally present with a shorter duration of symptoms than those with peptic ulcer disease and are more likely to be older.8,13 Other conditions with an acute onset of symptoms include gastric polyp prolapse, percutaneous endoscopic gastrostomy tube migration, gastric volvulus, and gallstone impaction.

Patients with gastric outlet obstruction associated with peptic ulcer disease generally have a long-standing history of symptoms, including dyspepsia and weight loss over several years.4

SIGNS ON EXAMINATION

On examination, look for signs of chronic gastric obstruction and its consequences, such as malnutrition, cachexia, volume depletion, and dental erosions.

A succussion splash may suggest gastric outlet obstruction. This is elicited by rocking the patient back and forth by the hips or abdomen while listening over the stomach for a splash, which may be heard without a stethoscope. The test is considered positive if present 3 or more hours after drinking fluids and suggests retention of gastric materials.30,31

In thin individuals, chronic gastric outlet obstruction makes the stomach dilate and hypertrophy, which may be evident by a palpably thickened stomach with visible gastric peristalsis.4

Other notable findings on physical examination may include a palpable abdominal mass, epigastric pain, or an abnormality suggestive of metastatic gastric cancer, such as an enlarged left supraclavicular lymph node (Virchow node) or periumbilical lymph node (Sister Mary Joseph nodule). The Virchow node is at the junction of the thoracic duct and the left subclavian vein where the lymphatic circulation from the body drains into the systemic circulation, and it may be the first sign of gastric cancer.32 Sister Mary Joseph nodule (named after a surgical assistant to Dr. William James Mayo) refers to a palpable mass at the umbilicus, generally resulting from metastasis of an abdominal malignancy.33

SIGNS ON FURTHER STUDIES

Laboratory evaluation may show signs of poor oral intake and electrolyte abnormalities secondary to chronic nausea, vomiting, and dehydration, including hypochloremic metabolic alkalosis and hypokalemia.

The underlying cause of gastric outlet obstruction has major implications for treatment and prognosis and cannot be differentiated by clinical presentation alone.1,9 Diagnosis is based on clinical features and radiologic or endoscopic evaluation consistent with gastric outlet obstruction.

Plain radiography may reveal an enlarged gastric bubble, and contrast studies may be useful to determine whether the obstruction is partial or complete, depending on whether the contrast passes into the small bowel.

koop_gastricoutletobstruction_f1.jpg
%3Cp%3EFigure%201.%20Computed%20tomography%20of%20the%20abdomen%20in%20the%20axial%20plane%20shows%20gastric%20distention%20(A%2C%20arrow)%20and%20a%203.9-cm%20mass%20at%20the%20pancreatic%20head%2C%20with%20compression%20of%20the%20descending%20duodenum%20(B%2C%20arrow)%2C%20resulting%20in%20gastric%20outlet%20obstruction.%20The%20patient%2C%20a%2072-year-old%20woman%2C%20presented%20with%201%20week%20of%20nausea%20and%20vomiting%20and%20was%20found%20to%20have%20pancreatic%20cancer.%20She%20was%20treated%20with%20endoscopic%20stenting.%3C%2Fp%3E
CT or magnetic resonance imaging may show gastric distention with retained stomach contents, suggesting a gastric, pyloric, duodenal, or pancreatic mass (Figure 1).

Upper endoscopy is often needed to establish the diagnosis and cause. Emptying the stomach with a nasogastric tube is recommended before endoscopy to minimize the risk of aspiration during the procedure, and endotracheal intubation should be considered for airway protection.34 Findings of gastric outlet obstruction on upper endoscopy include retained food and liquid. Endoscopic biopsy is important to differentiate between benign and malignant causes. For patients with malignancy, endoscopic ultrasonography is useful for diagnosis via tissue sampling with fine-needle aspiration and locoregional staging.35

A strategy. Most patients whose clinical presentation suggests gastric outlet obstruction require cross-sectional radiologic imaging, upper endoscopy, or both.36 CT is the preferred imaging study to evaluate for intestinal obstruction.36,37 Patients with suspected complete obstruction or perforation should undergo CT before upper endoscopy. Oral contrast may interfere with endoscopy and should be avoided if endoscopy is planned. Additionally, giving oral contrast may worsen patient discomfort and increase the risk of nausea, vomiting, and aspiration.36,37

Following radiographic evaluation, upper endoscopy can be performed after gastric decompression to identify the location and extent of the obstruction and to potentially provide a definitive diagnosis with biopsy.36

DIFFERENTIATE FROM GASTROPARESIS

Gastroparesis is a chronic neuromuscular disorder characterized by delayed gastric emptying without mechanical obstruction.38 The most common causes are diabetes, surgery, and idiopathy. Other causes include viral infection, connective tissue diseases, ischemia, infiltrative disorders, radiation, neurologic disorders, and paraneoplastic syndromes.39,40

Gastric outlet obstruction and gastroparesis share clinical symptoms including nausea, vomiting, abdominal pain, early satiety, and weight loss and are important to differentiate.36,38 Although abdominal pain may be present in both gastric outlet obstruction and gastroparesis, in gastroparesis it tends not to be the dominant symptom.40

Gastric scintigraphy is most commonly used to objectively quantify delayed gastric emptying.39 Upper endoscopy is imperative to exclude mechanical obstruction.39

 

 

MANAGEMENT

Initially, patients with signs and symptoms of gastric outlet obstruction should be given:

  • Nothing by mouth (NPO)
  • Intravenous fluids to correct volume depletion and electrolyte abnormalities
  • A nasogastric tube for gastric decompression and symptom relief if symptoms persist despite being NPO
  • A parenteral proton pump inhibitor, regardless of the cause of obstruction, to decrease gastric secretions41
  • Medications for pain and nausea, if needed.

Definitive treatment of gastric outlet obstruction depends on the underlying cause, whether benign or malignant.

Management of benign gastric outlet obstruction

Symptoms of gastric outlet obstruction resolve spontaneously in about half of cases caused by acute peptic ulcer disease, as acute inflammation resolves.9,22

Endoscopic dilation is an important option in patients with benign gastric outlet obstruction, including peptic ulcer disease. Peptic ulcer disease-induced gastric outlet obstruction can be safely treated with endoscopic balloon dilation. This treatment almost always relieves symptoms immediately; however, the long-term response has varied from 16% to 100%, and patients may require more than 1 dilation procedure.25,42,43 The need for 2 or more dilation procedures may predict need for surgery.44 Gastric outlet obstruction after caustic ingestion or endoscopic submucosal dissection may also respond to endoscopic balloon dilation.36

Eradication of H pylori may be effective and lead to complete resolution of symptoms in patients with gastric outlet obstruction due to this infection.45–47

NSAIDs should be discontinued in patients with peptic ulcer disease and gastric outlet obstruction. These drugs damage the gastrointestinal mucosa by inhibiting cyclo-oxygenase (COX) enzymes and decreasing synthesis of prostaglandins, which are important for mucosal defense.48 Patients may be unaware of NSAIDs contained in over-the-counter medications and may have difficulty discontinuing NSAIDs taken for pain.49

These drugs are an important cause of refractory peptic ulcer disease and can be detected by platelet COX activity testing, although this test is not widely available. In a study of patients with peptic ulcer disease without definite NSAID use or H pylori infection, up to one-third had evidence of surreptitious NSAID use as detected by platelet COX activity testing.50 In another study,51 platelet COX activity testing discovered over 20% more aspirin users than clinical history alone.

Surgery for patients with benign gastric outlet obstruction is used only when medical management and endoscopic dilation fail. Ideally, surgery should relieve the obstruction and target the underlying cause, such as peptic ulcer disease. Laparoscopic surgery is generally preferred to open surgery because patients can resume oral intake sooner, have a shorter hospital stay, and have less intraoperative blood loss.52 The simplest surgical procedure to relieve obstruction is laparoscopic gastrojejunostomy.

Patients with gastric outlet obstruction and peptic ulcer disease warrant laparoscopic vagotomy and antrectomy or distal gastrectomy. This removes the obstruction and the stimulus for gastric secretion.53 An alternative is vagotomy with a drainage procedure (pyloroplasty or gastrojejunostomy), which has a similar postoperative course and reduction in gastric acid secretion compared with antrectomy or distal gastrectomy.53,54

Daily proton pump inhibitors can be used for patients with benign gastric outlet obstruction not associated with peptic ulcer disease or risk factors; for such cases, vagotomy is not required.

Management of malignant gastric outlet obstruction

Patients with malignant gastric outlet obstruction may have intractable nausea and abdominal pain secondary to retention of gastric contents. The major goal of therapy is to improve symptoms and restore tolerance of an oral diet. The short-term prognosis of malignant gastric outlet obstruction is poor, with a median survival of 3 to 4 months, as these patients often have unresectable disease.55

Surgical bypass used to be the standard of care for palliation of malignant gastric obstruction, but that was before endoscopic stenting was developed.

Endoscopic stenting allows patients to resume oral intake and get out of the hospital sooner with fewer complications than with open surgical bypass. It may be a more appropriate option for palliation of symptoms in patients with malignant obstruction who have a poor prognosis and prefer a less invasive intervention.55,56

koop_gastricoutletobstruction_f2.jpg
%3Cp%3EFigure%202.%20Esophagogastroduodenoscopy%20(A)%20shows%20a%20large%20submucosal%20mass%20in%20the%20duodenal%20bulb%20(upper%20arrow)%2C%20with%20localized%20erosions%20(lower%20arrow).%20The%20mass%20was%2040%20%C3%97%2041%20mm%20in%20cross-sectional%20diameter%20on%20endoscopic%20ultrasonography.%20Fine-needle%20aspiration%20and%20pathology%20study%20revealed%20pancreatic%20adenocarcinoma.%20The%20obstruction%20was%20successfully%20opened%20(B)%20with%20a%2022-mm%20%C3%97%2012-cm%20WallFlex%20stent%20(Boston%20Scientific).%20The%20patient%20tolerated%20a%20liquid%20diet%20after%20the%20procedure.%3C%2Fp%3E

Endoscopic duodenal stenting of malignant gastric outlet obstruction has a success rate of greater than 90%, and most patients can tolerate a mechanical soft diet afterward.34 The procedure is usually performed with a 9-cm or 12-cm self-expanding duodenal stent, 22 mm in diameter, placed over a guide wire under endoscopic and fluoroscopic guidance (Figure 2). The stent is placed by removing the outer catheter, with distal-to-proximal stent deployment.

Patients who also have biliary obstruction may require biliary stent placement, which is generally performed before duodenal stenting. For patients with an endoscopic stent who develop biliary obstruction, endoscopic retrograde cholangiopancreatography can be attempted with placement of a biliary stent; however, these patients may require biliary drain placement by percutaneous transhepatic cholangiography or by endoscopic ultrasonographically guided transduodenal or transgastric biliary drainage.

From 20% to 30% of patients require repeated endoscopic stent placement, although most patients die within several months after stenting.34 Surgical options for patients who do not respond to endoscopic stenting include open or laparoscopic gastrojejunostomy.55

Laparoscopic gastrojejunostomy may provide better long-term outcomes than duodenal stenting for patients with malignant gastric outlet obstruction and a life expectancy longer than a few months.

A 2017 retrospective study of 155 patients with gastric outlet obstruction secondary to unresectable gastric cancer suggested that those who underwent laparoscopic gastrojejunostomy had better oral intake, better tolerance of chemotherapy, and longer overall survival than those who underwent duodenal stenting. Postsurgical complications were more common in the laparoscopic gastrojejunostomy group (16%) than in the duodenal stenting group (0%).57

In most of the studies comparing endoscopic stenting with surgery, the surgery was open gastrojejunostomy; there are limited data directly comparing stenting with laparoscopic gastrojejunostomy.55 Endoscopic stenting is estimated to be significantly less costly than surgery, with a median cost of $12,000 less than gastrojejunostomy.58 As an alternative to enteral stenting and surgical gastrojejunostomy, ultrasonography-guided endoscopic gastrojejunostomy or gastroenterostomy with placement of a lumen-apposing metal stent is emerging as a third treatment option and is under active investigation.59

Patients with malignancy that is potentially curable by resection should undergo surgical evaluation before consideration of endoscopic stenting. For patients who are not candidates for surgery or endoscopic stenting, a percutaneous gastrostomy tube can be considered for gastric decompression and symptom relief.

CASE CONCLUDED

The patient underwent esophagogastroduodenoscopy with endoscopic ultrasonography for evaluation of her pancreatic mass. Before the procedure, she was intubated to minimize the risk of aspiration due to persistent nausea and retained gastric contents. A large submucosal mass was found in the duodenal bulb. Endoscopic ultrasonography showed a mass within the pancreatic head with pancreatic duct obstruction. Fine-needle aspiration biopsy was performed, and pathology study revealed pancreatic adenocarcinoma. The patient underwent stenting with a 22-mm by 12-cm WallFlex stent (Boston Scientific), which led to resolution of nausea and advancement to a mechanical soft diet on hospital discharge.

She was scheduled for follow-up in the outpatient clinic for treatment of pancreatic cancer.

A 72-year-old woman presents to the emergency department with progressive nausea and vomiting. One week earlier, she developed early satiety and nausea with vomiting after eating solid food. Three days later her symptoms progressed, and she became unable to take anything by mouth. The patient also experienced a 40-lb weight loss in the previous 3 months. She denies symptoms of abdominal pain, hematemesis, or melena. Her medical history includes cholecystectomy and type 2 diabetes mellitus, diagnosed 1 year ago. She has no family history of gastrointestinal malignancy. She says she smoked 1 pack a day in her 20s. She does not consume alcohol.

On physical examination, she is normotensive with a heart rate of 105 beats per minute. The oral mucosa is dry, and the abdomen is mildly distended and tender to palpation in the epigastrium. Laboratory evaluation reveals hypokalemia and metabolic alkalosis.

Computed tomography (CT) reveals a mass 3 cm by 4 cm in the pancreatic head. The mass has invaded the medial wall of the duodenum, with obstruction of the pancreatic and common bile ducts and extension into and occlusion of the superior mesenteric vein, with soft-tissue expansion around the superior mesenteric artery. CT also reveals retained stomach contents and an air-fluid level consistent with gastric outlet obstruction.

INTRINSIC OR EXTRINSIC BLOCKAGE

Gastric outlet obstruction, also called pyloric obstruction, is caused by intrinsic or extrinsic mechanical blockage of gastric emptying, generally in the distal stomach, pyloric channel, or duodenum, with associated symptoms of nausea, vomiting, abdominal pain, and early satiety. It is encountered in both the clinic and the hospital.

Here, we review the causes, diagnosis, and management of this disorder.

BENIGN AND MALIGNANT CAUSES

koop_gastricoutletobstruction_t1.jpg
Causes of obstruction are classified as either benign or malignant (Table 1). However, all cases of gastric outlet obstruction should be assumed to be due to underlying malignancy unless proven otherwise.1

In a retrospective study of 76 patients hospitalized with gastric outlet obstruction between 2006 and 2015 at our institution,2 29 cases (38%) were due to malignancy and 47 (62%) were due to benign causes. Pancreatic adenocarcinoma accounted for 13 cases (17%), while gastric adenocarcinoma accounted for 5 cases (7%); less common malignant causes were cholangiocarcinoma, cancer of the ampulla of Vater, duodenal adenocarcinoma, hepatocellular carcinoma, and metastatic disease. Of the benign causes, the most common were peptic ulcer disease (13 cases, 17%) and postoperative strictures or adhesions (11 cases, 14%).

These numbers reflect general trends around the world.

Less gastric cancer, more pancreatic cancer

The last several decades have seen a trend toward more cases due to cancer and fewer due to benign causes.3–14

In earlier studies in both developed and developing countries, gastric adenocarcinoma was the most common malignant cause of gastric outlet obstruction. Since then, it has become less common in Western countries, although it remains more common in Asia and Africa.7–14 This trend likely reflects environmental factors, including decreased prevalence of Helicobacter pylori infection, a major risk factor for gastric cancer, in Western countries.15–17

At the same time, pancreatic cancer is on the rise,16 and up to 20% of patients with pancreatic cancer develop gastric outlet obstruction.18 In a prospective observational study of 108 patients with malignant gastric outlet obstruction undergoing endoscopic stenting, pancreatic cancer was by far the most common malignancy, occurring in 54% of patients, followed by gastric cancer in 13%.19

Less peptic ulcer disease, but still common

Peptic ulcer disease used to account for up to 90% of cases of gastric outlet obstruction, and it is still the most common benign cause.

In 1990, gastric outlet obstruction was estimated to occur in 5% to 10% of all hospital admissions for ulcer-related complications, accounting for 2,000 operations annually.20,21 Gastric outlet obstruction now occurs in fewer than 5% of patients with duodenal ulcer disease and fewer than 2% of patients with gastric ulcer disease.22

Peptic ulcer disease remains an important cause of obstruction in countries with poor access to acid-suppressing drugs.23

Gastric outlet obstruction occurs in both acute and chronic peptic ulcer disease. In acute peptic ulcer disease, tissue inflammation and edema result in mechanical obstruction. Chronic peptic ulcer disease results in tissue scarring and fibrosis with strictures.20

Environmental factors, including improved diet, hygiene, physical activity, and the decreased prevalence of H pylori infection, also contribute to the decreased prevalence of peptic ulcer disease and its complications, including gastric outlet obstruction.3 The continued occurrence of peptic ulcer disease is associated with widespread use of low-dose aspirin and nonsteroidal anti-inflammatory drugs (NSAIDs), the most common causes of peptic ulcer disease in Western countries.24,25

Other nonmalignant causes of gastric outlet obstruction are diverse and less common. They include caustic ingestion, postsurgical strictures, benign tumors of the gastrointestinal tract, Crohn disease, and pancreatic disorders including acute pancreatitis, pancreatic pseudocyst, chronic pancreatitis, and annular pancreas. Intramural duodenal hematoma may cause obstruction after blunt abdominal trauma, endoscopic biopsy, or gastrostomy tube migration, especially in the setting of a bleeding disorder or anticoagulation.26

Tuberculosis should be suspected in countries in which it is common.7 In a prospective study of 64 patients with benign gastric outlet obstruction in India,27 16 (25%) had corrosive injury, 16 (25%) had tuberculosis, and 15 (23%) had peptic ulcer disease. Compared with patients with corrosive injury and peptic ulcer disease, patients with gastroduodenal tuberculosis had the best outcomes with appropriate treatment.

Other reported causes include Bouveret syndrome (an impacted gallstone in the proximal duodenum), phytobezoar, diaphragmatic hernia, gastric volvulus, and Ladd bands (peritoneal bands associated with intestinal malrotation).7,28,29

 

 

PRESENTING SYMPTOMS

Symptoms of gastric outlet obstruction include nausea, nonbilious vomiting, epigastric pain, early satiety, abdominal distention, and weight loss.

In our patients, the most common presenting symptoms were nausea and vomiting (80%), followed by abdominal pain (72%); weight loss (15%), abdominal distention (15%), and early satiety (9%) were less common.2

Patients with gastric outlet obstruction secondary to malignancy generally present with a shorter duration of symptoms than those with peptic ulcer disease and are more likely to be older.8,13 Other conditions with an acute onset of symptoms include gastric polyp prolapse, percutaneous endoscopic gastrostomy tube migration, gastric volvulus, and gallstone impaction.

Patients with gastric outlet obstruction associated with peptic ulcer disease generally have a long-standing history of symptoms, including dyspepsia and weight loss over several years.4

SIGNS ON EXAMINATION

On examination, look for signs of chronic gastric obstruction and its consequences, such as malnutrition, cachexia, volume depletion, and dental erosions.

A succussion splash may suggest gastric outlet obstruction. This is elicited by rocking the patient back and forth by the hips or abdomen while listening over the stomach for a splash, which may be heard without a stethoscope. The test is considered positive if present 3 or more hours after drinking fluids and suggests retention of gastric materials.30,31

In thin individuals, chronic gastric outlet obstruction makes the stomach dilate and hypertrophy, which may be evident by a palpably thickened stomach with visible gastric peristalsis.4

Other notable findings on physical examination may include a palpable abdominal mass, epigastric pain, or an abnormality suggestive of metastatic gastric cancer, such as an enlarged left supraclavicular lymph node (Virchow node) or periumbilical lymph node (Sister Mary Joseph nodule). The Virchow node is at the junction of the thoracic duct and the left subclavian vein where the lymphatic circulation from the body drains into the systemic circulation, and it may be the first sign of gastric cancer.32 Sister Mary Joseph nodule (named after a surgical assistant to Dr. William James Mayo) refers to a palpable mass at the umbilicus, generally resulting from metastasis of an abdominal malignancy.33

SIGNS ON FURTHER STUDIES

Laboratory evaluation may show signs of poor oral intake and electrolyte abnormalities secondary to chronic nausea, vomiting, and dehydration, including hypochloremic metabolic alkalosis and hypokalemia.

The underlying cause of gastric outlet obstruction has major implications for treatment and prognosis and cannot be differentiated by clinical presentation alone.1,9 Diagnosis is based on clinical features and radiologic or endoscopic evaluation consistent with gastric outlet obstruction.

Plain radiography may reveal an enlarged gastric bubble, and contrast studies may be useful to determine whether the obstruction is partial or complete, depending on whether the contrast passes into the small bowel.

koop_gastricoutletobstruction_f1.jpg
%3Cp%3EFigure%201.%20Computed%20tomography%20of%20the%20abdomen%20in%20the%20axial%20plane%20shows%20gastric%20distention%20(A%2C%20arrow)%20and%20a%203.9-cm%20mass%20at%20the%20pancreatic%20head%2C%20with%20compression%20of%20the%20descending%20duodenum%20(B%2C%20arrow)%2C%20resulting%20in%20gastric%20outlet%20obstruction.%20The%20patient%2C%20a%2072-year-old%20woman%2C%20presented%20with%201%20week%20of%20nausea%20and%20vomiting%20and%20was%20found%20to%20have%20pancreatic%20cancer.%20She%20was%20treated%20with%20endoscopic%20stenting.%3C%2Fp%3E
CT or magnetic resonance imaging may show gastric distention with retained stomach contents, suggesting a gastric, pyloric, duodenal, or pancreatic mass (Figure 1).

Upper endoscopy is often needed to establish the diagnosis and cause. Emptying the stomach with a nasogastric tube is recommended before endoscopy to minimize the risk of aspiration during the procedure, and endotracheal intubation should be considered for airway protection.34 Findings of gastric outlet obstruction on upper endoscopy include retained food and liquid. Endoscopic biopsy is important to differentiate between benign and malignant causes. For patients with malignancy, endoscopic ultrasonography is useful for diagnosis via tissue sampling with fine-needle aspiration and locoregional staging.35

A strategy. Most patients whose clinical presentation suggests gastric outlet obstruction require cross-sectional radiologic imaging, upper endoscopy, or both.36 CT is the preferred imaging study to evaluate for intestinal obstruction.36,37 Patients with suspected complete obstruction or perforation should undergo CT before upper endoscopy. Oral contrast may interfere with endoscopy and should be avoided if endoscopy is planned. Additionally, giving oral contrast may worsen patient discomfort and increase the risk of nausea, vomiting, and aspiration.36,37

Following radiographic evaluation, upper endoscopy can be performed after gastric decompression to identify the location and extent of the obstruction and to potentially provide a definitive diagnosis with biopsy.36

DIFFERENTIATE FROM GASTROPARESIS

Gastroparesis is a chronic neuromuscular disorder characterized by delayed gastric emptying without mechanical obstruction.38 The most common causes are diabetes, surgery, and idiopathy. Other causes include viral infection, connective tissue diseases, ischemia, infiltrative disorders, radiation, neurologic disorders, and paraneoplastic syndromes.39,40

Gastric outlet obstruction and gastroparesis share clinical symptoms including nausea, vomiting, abdominal pain, early satiety, and weight loss and are important to differentiate.36,38 Although abdominal pain may be present in both gastric outlet obstruction and gastroparesis, in gastroparesis it tends not to be the dominant symptom.40

Gastric scintigraphy is most commonly used to objectively quantify delayed gastric emptying.39 Upper endoscopy is imperative to exclude mechanical obstruction.39

 

 

MANAGEMENT

Initially, patients with signs and symptoms of gastric outlet obstruction should be given:

  • Nothing by mouth (NPO)
  • Intravenous fluids to correct volume depletion and electrolyte abnormalities
  • A nasogastric tube for gastric decompression and symptom relief if symptoms persist despite being NPO
  • A parenteral proton pump inhibitor, regardless of the cause of obstruction, to decrease gastric secretions41
  • Medications for pain and nausea, if needed.

Definitive treatment of gastric outlet obstruction depends on the underlying cause, whether benign or malignant.

Management of benign gastric outlet obstruction

Symptoms of gastric outlet obstruction resolve spontaneously in about half of cases caused by acute peptic ulcer disease, as acute inflammation resolves.9,22

Endoscopic dilation is an important option in patients with benign gastric outlet obstruction, including peptic ulcer disease. Peptic ulcer disease-induced gastric outlet obstruction can be safely treated with endoscopic balloon dilation. This treatment almost always relieves symptoms immediately; however, the long-term response has varied from 16% to 100%, and patients may require more than 1 dilation procedure.25,42,43 The need for 2 or more dilation procedures may predict need for surgery.44 Gastric outlet obstruction after caustic ingestion or endoscopic submucosal dissection may also respond to endoscopic balloon dilation.36

Eradication of H pylori may be effective and lead to complete resolution of symptoms in patients with gastric outlet obstruction due to this infection.45–47

NSAIDs should be discontinued in patients with peptic ulcer disease and gastric outlet obstruction. These drugs damage the gastrointestinal mucosa by inhibiting cyclo-oxygenase (COX) enzymes and decreasing synthesis of prostaglandins, which are important for mucosal defense.48 Patients may be unaware of NSAIDs contained in over-the-counter medications and may have difficulty discontinuing NSAIDs taken for pain.49

These drugs are an important cause of refractory peptic ulcer disease and can be detected by platelet COX activity testing, although this test is not widely available. In a study of patients with peptic ulcer disease without definite NSAID use or H pylori infection, up to one-third had evidence of surreptitious NSAID use as detected by platelet COX activity testing.50 In another study,51 platelet COX activity testing discovered over 20% more aspirin users than clinical history alone.

Surgery for patients with benign gastric outlet obstruction is used only when medical management and endoscopic dilation fail. Ideally, surgery should relieve the obstruction and target the underlying cause, such as peptic ulcer disease. Laparoscopic surgery is generally preferred to open surgery because patients can resume oral intake sooner, have a shorter hospital stay, and have less intraoperative blood loss.52 The simplest surgical procedure to relieve obstruction is laparoscopic gastrojejunostomy.

Patients with gastric outlet obstruction and peptic ulcer disease warrant laparoscopic vagotomy and antrectomy or distal gastrectomy. This removes the obstruction and the stimulus for gastric secretion.53 An alternative is vagotomy with a drainage procedure (pyloroplasty or gastrojejunostomy), which has a similar postoperative course and reduction in gastric acid secretion compared with antrectomy or distal gastrectomy.53,54

Daily proton pump inhibitors can be used for patients with benign gastric outlet obstruction not associated with peptic ulcer disease or risk factors; for such cases, vagotomy is not required.

Management of malignant gastric outlet obstruction

Patients with malignant gastric outlet obstruction may have intractable nausea and abdominal pain secondary to retention of gastric contents. The major goal of therapy is to improve symptoms and restore tolerance of an oral diet. The short-term prognosis of malignant gastric outlet obstruction is poor, with a median survival of 3 to 4 months, as these patients often have unresectable disease.55

Surgical bypass used to be the standard of care for palliation of malignant gastric obstruction, but that was before endoscopic stenting was developed.

Endoscopic stenting allows patients to resume oral intake and get out of the hospital sooner with fewer complications than with open surgical bypass. It may be a more appropriate option for palliation of symptoms in patients with malignant obstruction who have a poor prognosis and prefer a less invasive intervention.55,56

koop_gastricoutletobstruction_f2.jpg
%3Cp%3EFigure%202.%20Esophagogastroduodenoscopy%20(A)%20shows%20a%20large%20submucosal%20mass%20in%20the%20duodenal%20bulb%20(upper%20arrow)%2C%20with%20localized%20erosions%20(lower%20arrow).%20The%20mass%20was%2040%20%C3%97%2041%20mm%20in%20cross-sectional%20diameter%20on%20endoscopic%20ultrasonography.%20Fine-needle%20aspiration%20and%20pathology%20study%20revealed%20pancreatic%20adenocarcinoma.%20The%20obstruction%20was%20successfully%20opened%20(B)%20with%20a%2022-mm%20%C3%97%2012-cm%20WallFlex%20stent%20(Boston%20Scientific).%20The%20patient%20tolerated%20a%20liquid%20diet%20after%20the%20procedure.%3C%2Fp%3E

Endoscopic duodenal stenting of malignant gastric outlet obstruction has a success rate of greater than 90%, and most patients can tolerate a mechanical soft diet afterward.34 The procedure is usually performed with a 9-cm or 12-cm self-expanding duodenal stent, 22 mm in diameter, placed over a guide wire under endoscopic and fluoroscopic guidance (Figure 2). The stent is placed by removing the outer catheter, with distal-to-proximal stent deployment.

Patients who also have biliary obstruction may require biliary stent placement, which is generally performed before duodenal stenting. For patients with an endoscopic stent who develop biliary obstruction, endoscopic retrograde cholangiopancreatography can be attempted with placement of a biliary stent; however, these patients may require biliary drain placement by percutaneous transhepatic cholangiography or by endoscopic ultrasonographically guided transduodenal or transgastric biliary drainage.

From 20% to 30% of patients require repeated endoscopic stent placement, although most patients die within several months after stenting.34 Surgical options for patients who do not respond to endoscopic stenting include open or laparoscopic gastrojejunostomy.55

Laparoscopic gastrojejunostomy may provide better long-term outcomes than duodenal stenting for patients with malignant gastric outlet obstruction and a life expectancy longer than a few months.

A 2017 retrospective study of 155 patients with gastric outlet obstruction secondary to unresectable gastric cancer suggested that those who underwent laparoscopic gastrojejunostomy had better oral intake, better tolerance of chemotherapy, and longer overall survival than those who underwent duodenal stenting. Postsurgical complications were more common in the laparoscopic gastrojejunostomy group (16%) than in the duodenal stenting group (0%).57

In most of the studies comparing endoscopic stenting with surgery, the surgery was open gastrojejunostomy; there are limited data directly comparing stenting with laparoscopic gastrojejunostomy.55 Endoscopic stenting is estimated to be significantly less costly than surgery, with a median cost of $12,000 less than gastrojejunostomy.58 As an alternative to enteral stenting and surgical gastrojejunostomy, ultrasonography-guided endoscopic gastrojejunostomy or gastroenterostomy with placement of a lumen-apposing metal stent is emerging as a third treatment option and is under active investigation.59

Patients with malignancy that is potentially curable by resection should undergo surgical evaluation before consideration of endoscopic stenting. For patients who are not candidates for surgery or endoscopic stenting, a percutaneous gastrostomy tube can be considered for gastric decompression and symptom relief.

CASE CONCLUDED

The patient underwent esophagogastroduodenoscopy with endoscopic ultrasonography for evaluation of her pancreatic mass. Before the procedure, she was intubated to minimize the risk of aspiration due to persistent nausea and retained gastric contents. A large submucosal mass was found in the duodenal bulb. Endoscopic ultrasonography showed a mass within the pancreatic head with pancreatic duct obstruction. Fine-needle aspiration biopsy was performed, and pathology study revealed pancreatic adenocarcinoma. The patient underwent stenting with a 22-mm by 12-cm WallFlex stent (Boston Scientific), which led to resolution of nausea and advancement to a mechanical soft diet on hospital discharge.

She was scheduled for follow-up in the outpatient clinic for treatment of pancreatic cancer.

References
  1. Johnson CD. Gastric outlet obstruction malignant until proved otherwise. Am J Gastroenterol 1995; 90(10):1740. pmid:7572886
  2. Koop AH, Palmer WC, Mareth K, Burton MC, Bowman A, Stancampiano F. Tu1335 - Pancreatic cancer most common cause of malignant gastric outlet obstruction at a tertiary referral center: a 10 year retrospective study [abstract]. Gastroenterology 2018; 154(6, suppl 1):S-1343.
  3. Hall R, Royston C, Bardhan KD. The scars of time: the disappearance of peptic ulcer-related pyloric stenosis through the 20th century. J R Coll Physicians Edinb 2014; 44(3):201–208. doi:10.4997/JRCPE.2014.303
  4. Kreel L, Ellis H. Pyloric stenosis in adults: a clinical and radiological study of 100 consecutive patients. Gut 1965; 6(3):253–261. pmid:18668780
  5. Shone DN, Nikoomanesh P, Smith-Meek MM, Bender JS. Malignancy is the most common cause of gastric outlet obstruction in the era of H2 blockers. Am J Gastroenterol 1995; 90(10):1769–1770. pmid:7572891
  6. Ellis H. The diagnosis of benign and malignant pyloric obstruction. Clin Oncol 1976; 2(1):11–15. pmid:1277618
  7. Samad A, Khanzada TW, Shoukat I. Gastric outlet obstruction: change in etiology. Pak J Surg 2007; 23(1):29–32.
  8. Chowdhury A, Dhali GK, Banerjee PK. Etiology of gastric outlet obstruction. Am J Gastroenterol 1996; 91(8):1679. pmid:8759707
  9. Johnson CD, Ellis H. Gastric outlet obstruction now predicts malignancy. Br J Surg 1990; 77(9):1023–1024. pmid:2207566
  10. Misra SP, Dwivedi M, Misra V. Malignancy is the most common cause of gastric outlet obstruction even in a developing country. Endoscopy 1998; 30(5):484–486. doi:10.1055/s-2007-1001313
  11. Essoun SD, Dakubo JCB. Update of aetiological patterns of adult gastric outlet obstruction in Accra, Ghana. Int J Clin Med 2014; 5(17):1059–1064. doi:10.4236/ijcm.2014.517136
  12. Jaka H, Mchembe MD, Rambau PF, Chalya PL. Gastric outlet obstruction at Bugando Medical Centre in Northwestern Tanzania: a prospective review of 184 cases. BMC Surg 2013; 13:41. doi:10.1186/1471-2482-13-41
  13. Sukumar V, Ravindran C, Prasad RV. Demographic and etiological patterns of gastric outlet obstruction in Kerala, South India. N Am J Med Sci 2015; 7(9):403–406. doi:10.4103/1947-2714.166220
  14. Yoursef M, Mirza MR, Khan S. Gastric outlet obstruction. Pak J Surg 2005; 10(4):48–50.
  15. Ferlay J, Soerjomataram I, Dikshit R, et al. Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer 2015; 136(5):E359–E386. doi:10.1002/ijc.29210
  16. Parkin DM, Stjernsward J, Muir CS. Estimates of the worldwide frequency of twelve major cancers. Bull World Health Organ 1984; 62(2):163–182. pmid:6610488
  17. Karimi P, Islami F, Anandasabapathy S, Freedman ND, Kamangar F. Gastric cancer: descriptive epidemiology, risk factors, screening, and prevention. Cancer Epidemiol Biomarkers Prev 2014; 23(5):700–713. doi:10.1158/1055-9965.EPI-13-1057
  18. Jeurnink SM, Steyerberg EW, van Hooft JE, et al; Dutch SUSTENT Study Group. Surgical gastrojejunostomy or endoscopic stent placement for the palliation of malignant gastric outlet obstruction (SUSTENT) study): a multicenter randomized trial. Gastrointest Endosc 2010; 71(3):490–499. doi:10.1016/j.gie.2009.09.042
  19. Tringali A, Didden P, Repici A, et al. Endoscopic treatment of malignant gastric and duodenal strictures: a prospective, multicenter study. Gastrointest Endosc 2014; 79(1):66–75. doi:10.1016/j.gie.2013.06.032
  20. Malfertheiner P, Chan FK, McColl KE. Peptic ulcer disease. Lancet 2009; 374(9699):1449–1461. doi:10.1016/S0140-6736(09)60938-7
  21. Gibson JB, Behrman SW, Fabian TC, Britt LG. Gastric outlet obstruction resulting from peptic ulcer disease requiring surgical intervention is infrequently associated with Helicobacter pylori infection. J Am Coll Surg 2000; 191(1):32–37. pmid:10898181
  22. Kochhar R, Kochhar S. Endoscopic balloon dilation for benign gastric outlet obstruction in adults. World J Gastrointest Endosc 2010; 2(1):29–35. doi:10.4253/wjge.v2.i1.29
  23. Kotisso R. Gastric outlet obstruction in Northwestern Ethiopia. East Cent Afr J Surg 2000; 5(2):25-29.
  24. Hamzaoui L, Bouassida M, Ben Mansour I, et al. Balloon dilatation in patients with gastric outlet obstruction related to peptic ulcer disease. Arab J Gastroenterol 2015; 16(3–4):121–124. doi:10.1016/j.ajg.2015.07.004
  25. Najm WI. Peptic ulcer disease. Prim Care 2011; 38(3):383–394. doi:10.1016/j.pop.2011.05.001
  26. Veloso N, Amaro P, Ferreira M, Romaozinho JM, Sofia C. Acute pancreatitis associated with a nontraumatic, intramural duodenal hematoma. Endoscopy 2013; 45(suppl 2):E51–E52. doi:10.1055/s-0032-1325969
  27. Maharshi S, Puri AS, Sachdeva S, Kumar A, Dalal A, Gupta M. Aetiological spectrum of benign gastric outlet obstruction in India: new trends. Trop Doct 2016; 46(4):186–191. doi:10.1177/0049475515626032
  28. Sala MA, Ligabo AN, de Arruda MC, Indiani JM, Nacif MS. Intestinal malrotation associated with duodenal obstruction secondary to Ladd’s bands. Radiol Bras 2016; 49(4):271–272. doi:10.1590/0100-3984.2015.0106
  29. Alibegovic E, Kurtcehajic A, Hujdurovic A, Mujagic S, Alibegovic J, Kurtcehajic D. Bouveret syndrome or gallstone ileus. Am J Med 2018; 131(4):e175. doi:10.1016/j.amjmed.2017.10.044
  30. Lau JY, Chung SC, Sung JJ, et al. Through-the-scope balloon dilation for pyloric stenosis: long-term results. Gastrointest Endosc 1996; 43(2 Pt 1):98–101. pmid:8635729
  31. Ray K, Snowden C, Khatri K, McFall M. Gastric outlet obstruction from a caecal volvulus, herniated through epiploic foramen: a case report. BMJ Case Rep 2009; pii:bcr05.2009.1880. doi:10.1136/bcr.05.2009.1880
  32. Baumgart DC, Fischer A. Virchow’s node. Lancet 2007; 370(9598):1568. doi:10.1016/S0140-6736(07)61661-4
  33. Dar IH, Kamili MA, Dar SH, Kuchaai FA. Sister Mary Joseph nodule—a case report with review of literature. J Res Med Sci 2009; 14(6):385–387. pmid:21772912
  34. Tang SJ. Endoscopic stent placement for gastric outlet obstruction. Video Journal and Encyclopedia of GI Endoscopy 2013; 1(1):133–136.
  35. Valero M, Robles-Medranda C. Endoscopic ultrasound in oncology: an update of clinical applications in the gastrointestinal tract. World J Gastrointest Endosc 2017; 9(6):243–254.
  36. ASGE Standards of Practice Committee; Fukami N, Anderson MA, Khan K, et al. The role of endoscopy in gastroduodenal obstruction and gastroparesis. Gastrointest Endosc 2011; 74(1):13–21. doi:10.1016/j.gie.2010.12.003
  37. Ros PR, Huprich JE. ACR appropriateness criteria on suspected small-bowel obstruction. J Am Coll Radiol 2006; 3(11):838–841. doi:10.1016/j.jacr.2006.09.018
  38. Pasricha PJ, Parkman HP. Gastroparesis: definitions and diagnosis. Gastroenterol Clin North Am 2015; 44(1):1–7. doi:10.1016/j.gtc.2014.11.001
  39. Stein B, Everhart KK, Lacy BE. Gastroparesis: a review of current diagnosis and treatment options. J Clin Gastroenterol 2015; 49(7):550–558. doi:10.1097/MCG.0000000000000320
  40. Camilleri M, Parkman HP, Shafi MA, Abell TL, Gerson L; American College of Gastroenterology. Clinical guideline: management of gastroparesis. Am J Gastroenterol 2013; 108(1):18–37.
  41. Gursoy O, Memis D, Sut N. Effect of proton pump inhibitors on gastric juice volume, gastric pH and gastric intramucosal pH in critically ill patients: a randomized, double-blind, placebo-controlled study. Clin Drug Investig 2008; 28(12):777–782. doi:10.2165/0044011-200828120-00005
  42. Kuwada SK, Alexander GL. Long-term outcome of endoscopic dilation of nonmalignant pyloric stenosis. Gastrointest Endosc 1995; 41(1):15–17. pmid:7698619
  43. Kochhar R, Sethy PK, Nagi B, Wig JD. Endoscopic balloon dilatation of benign gastric outlet obstruction. J Gastroenterol Hepatol 2004; 19(4):418–422. pmid:15012779
  44. Perng CL, Lin HJ, Lo WC, Lai CR, Guo WS, Lee SD. Characteristics of patients with benign gastric outlet obstruction requiring surgery after endoscopic balloon dilation. Am J Gastroenterol 1996; 91(5):987–990. pmid:8633593
  45. Taskin V, Gurer I, Ozyilkan E, Sare M, Hilmioglu F. Effect of Helicobacter pylori eradication on peptic ulcer disease complicated with outlet obstruction. Helicobacter 2000; 5(1):38–40. pmid:10672050
  46. de Boer WA, Driessen WM. Resolution of gastric outlet obstruction after eradication of Helicobacter pylori. J Clin Gastroenterol 1995; 21(4):329–330. pmid:8583113
  47. Tursi A, Cammarota G, Papa A, Montalto M, Fedeli G, Gasbarrini G. Helicobacter pylori eradication helps resolve pyloric and duodenal stenosis. J Clin Gastroenterol 1996; 23(2):157–158. pmid:8877648
  48. Schmassmann A. Mechanisms of ulcer healing and effects of nonsteroidal anti-inflammatory drugs. Am J Med 1998; 104(3A):43S–51S; discussion 79S–80S. pmid:9572320
  49. Kim HU. Diagnostic and treatment approaches for refractory peptic ulcers. Clin Endosc 2015; 48(4):285–290. doi:10.5946/ce.2015.48.4.285
  50. Ong TZ, Hawkey CJ, Ho KY. Nonsteroidal anti-inflammatory drug use is a significant cause of peptic ulcer disease in a tertiary hospital in Singapore: a prospective study. J Clin Gastroenterol 2006; 40(9):795–800. doi:10.1097/01.mcg.0000225610.41105.7f
  51. Lanas A, Sekar MC, Hirschowitz BI. Objective evidence of aspirin use in both ulcer and nonulcer upper and lower gastrointestinal bleeding. Gastroenterology 1992; 103(3):862–869. pmid:1499936
  52. Zhang LP, Tabrizian P, Nguyen S, Telem D, Divino C. Laparoscopic gastrojejunostomy for the treatment of gastric outlet obstruction. JSLS 2011; 15(2):169–173. doi:10.4293/108680811X13022985132074
  53. Lagoo J, Pappas TN, Perez A. A relic or still relevant: the narrowing role for vagotomy in the treatment of peptic ulcer disease. Am J Surg 2014; 207(1):120–126. doi:10.1016/j.amjsurg.2013.02.012
  54. Csendes A, Maluenda F, Braghetto I, Schutte H, Burdiles P, Diaz JC. Prospective randomized study comparing three surgical techniques for the treatment of gastric outlet obstruction secondary to duodenal ulcer. Am J Surg 1993; 166(1):45–49. pmid:8101050
  55. Ly J, O’Grady G, Mittal A, Plank L, Windsor JA. A systematic review of methods to palliate malignant gastric outlet obstruction. Surg Endosc 2010; 24(2):290–297. doi:10.1007/s00464-009-0577-1
  56. Goldberg EM. Palliative treatment of gastric outlet obstruction in terminal patients: SEMS. Stent every malignant stricture! Gastrointest Endosc 2014; 79(1):76–78. doi:10.1016/j.gie.2013.07.056
  57. Min SH, Son SY, Jung DH, et al. Laparoscopic gastrojejunostomy versus duodenal stenting in unresectable gastric cancer with gastric outlet obstruction. Ann Surg Treat Res 2017; 93(3):130–136. doi:10.4174/astr.2017.93.3.130
  58. Roy A, Kim M, Christein J, Varadarajulu S. Stenting versus gastrojejunostomy for management of malignant gastric outlet obstruction: comparison of clinical outcomes and costs. Surg Endosc 2012; 26(11):3114–119. doi:10.1007/s00464-012-2301-9
  59. Amin S, Sethi A. Endoscopic ultrasound-guided gastrojejunostomy. Gastrointest Endosc Clin N Am 2017; 27(4):707–713. doi:10.1016/j.giec.2017.06.009
References
  1. Johnson CD. Gastric outlet obstruction malignant until proved otherwise. Am J Gastroenterol 1995; 90(10):1740. pmid:7572886
  2. Koop AH, Palmer WC, Mareth K, Burton MC, Bowman A, Stancampiano F. Tu1335 - Pancreatic cancer most common cause of malignant gastric outlet obstruction at a tertiary referral center: a 10 year retrospective study [abstract]. Gastroenterology 2018; 154(6, suppl 1):S-1343.
  3. Hall R, Royston C, Bardhan KD. The scars of time: the disappearance of peptic ulcer-related pyloric stenosis through the 20th century. J R Coll Physicians Edinb 2014; 44(3):201–208. doi:10.4997/JRCPE.2014.303
  4. Kreel L, Ellis H. Pyloric stenosis in adults: a clinical and radiological study of 100 consecutive patients. Gut 1965; 6(3):253–261. pmid:18668780
  5. Shone DN, Nikoomanesh P, Smith-Meek MM, Bender JS. Malignancy is the most common cause of gastric outlet obstruction in the era of H2 blockers. Am J Gastroenterol 1995; 90(10):1769–1770. pmid:7572891
  6. Ellis H. The diagnosis of benign and malignant pyloric obstruction. Clin Oncol 1976; 2(1):11–15. pmid:1277618
  7. Samad A, Khanzada TW, Shoukat I. Gastric outlet obstruction: change in etiology. Pak J Surg 2007; 23(1):29–32.
  8. Chowdhury A, Dhali GK, Banerjee PK. Etiology of gastric outlet obstruction. Am J Gastroenterol 1996; 91(8):1679. pmid:8759707
  9. Johnson CD, Ellis H. Gastric outlet obstruction now predicts malignancy. Br J Surg 1990; 77(9):1023–1024. pmid:2207566
  10. Misra SP, Dwivedi M, Misra V. Malignancy is the most common cause of gastric outlet obstruction even in a developing country. Endoscopy 1998; 30(5):484–486. doi:10.1055/s-2007-1001313
  11. Essoun SD, Dakubo JCB. Update of aetiological patterns of adult gastric outlet obstruction in Accra, Ghana. Int J Clin Med 2014; 5(17):1059–1064. doi:10.4236/ijcm.2014.517136
  12. Jaka H, Mchembe MD, Rambau PF, Chalya PL. Gastric outlet obstruction at Bugando Medical Centre in Northwestern Tanzania: a prospective review of 184 cases. BMC Surg 2013; 13:41. doi:10.1186/1471-2482-13-41
  13. Sukumar V, Ravindran C, Prasad RV. Demographic and etiological patterns of gastric outlet obstruction in Kerala, South India. N Am J Med Sci 2015; 7(9):403–406. doi:10.4103/1947-2714.166220
  14. Yoursef M, Mirza MR, Khan S. Gastric outlet obstruction. Pak J Surg 2005; 10(4):48–50.
  15. Ferlay J, Soerjomataram I, Dikshit R, et al. Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer 2015; 136(5):E359–E386. doi:10.1002/ijc.29210
  16. Parkin DM, Stjernsward J, Muir CS. Estimates of the worldwide frequency of twelve major cancers. Bull World Health Organ 1984; 62(2):163–182. pmid:6610488
  17. Karimi P, Islami F, Anandasabapathy S, Freedman ND, Kamangar F. Gastric cancer: descriptive epidemiology, risk factors, screening, and prevention. Cancer Epidemiol Biomarkers Prev 2014; 23(5):700–713. doi:10.1158/1055-9965.EPI-13-1057
  18. Jeurnink SM, Steyerberg EW, van Hooft JE, et al; Dutch SUSTENT Study Group. Surgical gastrojejunostomy or endoscopic stent placement for the palliation of malignant gastric outlet obstruction (SUSTENT) study): a multicenter randomized trial. Gastrointest Endosc 2010; 71(3):490–499. doi:10.1016/j.gie.2009.09.042
  19. Tringali A, Didden P, Repici A, et al. Endoscopic treatment of malignant gastric and duodenal strictures: a prospective, multicenter study. Gastrointest Endosc 2014; 79(1):66–75. doi:10.1016/j.gie.2013.06.032
  20. Malfertheiner P, Chan FK, McColl KE. Peptic ulcer disease. Lancet 2009; 374(9699):1449–1461. doi:10.1016/S0140-6736(09)60938-7
  21. Gibson JB, Behrman SW, Fabian TC, Britt LG. Gastric outlet obstruction resulting from peptic ulcer disease requiring surgical intervention is infrequently associated with Helicobacter pylori infection. J Am Coll Surg 2000; 191(1):32–37. pmid:10898181
  22. Kochhar R, Kochhar S. Endoscopic balloon dilation for benign gastric outlet obstruction in adults. World J Gastrointest Endosc 2010; 2(1):29–35. doi:10.4253/wjge.v2.i1.29
  23. Kotisso R. Gastric outlet obstruction in Northwestern Ethiopia. East Cent Afr J Surg 2000; 5(2):25-29.
  24. Hamzaoui L, Bouassida M, Ben Mansour I, et al. Balloon dilatation in patients with gastric outlet obstruction related to peptic ulcer disease. Arab J Gastroenterol 2015; 16(3–4):121–124. doi:10.1016/j.ajg.2015.07.004
  25. Najm WI. Peptic ulcer disease. Prim Care 2011; 38(3):383–394. doi:10.1016/j.pop.2011.05.001
  26. Veloso N, Amaro P, Ferreira M, Romaozinho JM, Sofia C. Acute pancreatitis associated with a nontraumatic, intramural duodenal hematoma. Endoscopy 2013; 45(suppl 2):E51–E52. doi:10.1055/s-0032-1325969
  27. Maharshi S, Puri AS, Sachdeva S, Kumar A, Dalal A, Gupta M. Aetiological spectrum of benign gastric outlet obstruction in India: new trends. Trop Doct 2016; 46(4):186–191. doi:10.1177/0049475515626032
  28. Sala MA, Ligabo AN, de Arruda MC, Indiani JM, Nacif MS. Intestinal malrotation associated with duodenal obstruction secondary to Ladd’s bands. Radiol Bras 2016; 49(4):271–272. doi:10.1590/0100-3984.2015.0106
  29. Alibegovic E, Kurtcehajic A, Hujdurovic A, Mujagic S, Alibegovic J, Kurtcehajic D. Bouveret syndrome or gallstone ileus. Am J Med 2018; 131(4):e175. doi:10.1016/j.amjmed.2017.10.044
  30. Lau JY, Chung SC, Sung JJ, et al. Through-the-scope balloon dilation for pyloric stenosis: long-term results. Gastrointest Endosc 1996; 43(2 Pt 1):98–101. pmid:8635729
  31. Ray K, Snowden C, Khatri K, McFall M. Gastric outlet obstruction from a caecal volvulus, herniated through epiploic foramen: a case report. BMJ Case Rep 2009; pii:bcr05.2009.1880. doi:10.1136/bcr.05.2009.1880
  32. Baumgart DC, Fischer A. Virchow’s node. Lancet 2007; 370(9598):1568. doi:10.1016/S0140-6736(07)61661-4
  33. Dar IH, Kamili MA, Dar SH, Kuchaai FA. Sister Mary Joseph nodule—a case report with review of literature. J Res Med Sci 2009; 14(6):385–387. pmid:21772912
  34. Tang SJ. Endoscopic stent placement for gastric outlet obstruction. Video Journal and Encyclopedia of GI Endoscopy 2013; 1(1):133–136.
  35. Valero M, Robles-Medranda C. Endoscopic ultrasound in oncology: an update of clinical applications in the gastrointestinal tract. World J Gastrointest Endosc 2017; 9(6):243–254.
  36. ASGE Standards of Practice Committee; Fukami N, Anderson MA, Khan K, et al. The role of endoscopy in gastroduodenal obstruction and gastroparesis. Gastrointest Endosc 2011; 74(1):13–21. doi:10.1016/j.gie.2010.12.003
  37. Ros PR, Huprich JE. ACR appropriateness criteria on suspected small-bowel obstruction. J Am Coll Radiol 2006; 3(11):838–841. doi:10.1016/j.jacr.2006.09.018
  38. Pasricha PJ, Parkman HP. Gastroparesis: definitions and diagnosis. Gastroenterol Clin North Am 2015; 44(1):1–7. doi:10.1016/j.gtc.2014.11.001
  39. Stein B, Everhart KK, Lacy BE. Gastroparesis: a review of current diagnosis and treatment options. J Clin Gastroenterol 2015; 49(7):550–558. doi:10.1097/MCG.0000000000000320
  40. Camilleri M, Parkman HP, Shafi MA, Abell TL, Gerson L; American College of Gastroenterology. Clinical guideline: management of gastroparesis. Am J Gastroenterol 2013; 108(1):18–37.
  41. Gursoy O, Memis D, Sut N. Effect of proton pump inhibitors on gastric juice volume, gastric pH and gastric intramucosal pH in critically ill patients: a randomized, double-blind, placebo-controlled study. Clin Drug Investig 2008; 28(12):777–782. doi:10.2165/0044011-200828120-00005
  42. Kuwada SK, Alexander GL. Long-term outcome of endoscopic dilation of nonmalignant pyloric stenosis. Gastrointest Endosc 1995; 41(1):15–17. pmid:7698619
  43. Kochhar R, Sethy PK, Nagi B, Wig JD. Endoscopic balloon dilatation of benign gastric outlet obstruction. J Gastroenterol Hepatol 2004; 19(4):418–422. pmid:15012779
  44. Perng CL, Lin HJ, Lo WC, Lai CR, Guo WS, Lee SD. Characteristics of patients with benign gastric outlet obstruction requiring surgery after endoscopic balloon dilation. Am J Gastroenterol 1996; 91(5):987–990. pmid:8633593
  45. Taskin V, Gurer I, Ozyilkan E, Sare M, Hilmioglu F. Effect of Helicobacter pylori eradication on peptic ulcer disease complicated with outlet obstruction. Helicobacter 2000; 5(1):38–40. pmid:10672050
  46. de Boer WA, Driessen WM. Resolution of gastric outlet obstruction after eradication of Helicobacter pylori. J Clin Gastroenterol 1995; 21(4):329–330. pmid:8583113
  47. Tursi A, Cammarota G, Papa A, Montalto M, Fedeli G, Gasbarrini G. Helicobacter pylori eradication helps resolve pyloric and duodenal stenosis. J Clin Gastroenterol 1996; 23(2):157–158. pmid:8877648
  48. Schmassmann A. Mechanisms of ulcer healing and effects of nonsteroidal anti-inflammatory drugs. Am J Med 1998; 104(3A):43S–51S; discussion 79S–80S. pmid:9572320
  49. Kim HU. Diagnostic and treatment approaches for refractory peptic ulcers. Clin Endosc 2015; 48(4):285–290. doi:10.5946/ce.2015.48.4.285
  50. Ong TZ, Hawkey CJ, Ho KY. Nonsteroidal anti-inflammatory drug use is a significant cause of peptic ulcer disease in a tertiary hospital in Singapore: a prospective study. J Clin Gastroenterol 2006; 40(9):795–800. doi:10.1097/01.mcg.0000225610.41105.7f
  51. Lanas A, Sekar MC, Hirschowitz BI. Objective evidence of aspirin use in both ulcer and nonulcer upper and lower gastrointestinal bleeding. Gastroenterology 1992; 103(3):862–869. pmid:1499936
  52. Zhang LP, Tabrizian P, Nguyen S, Telem D, Divino C. Laparoscopic gastrojejunostomy for the treatment of gastric outlet obstruction. JSLS 2011; 15(2):169–173. doi:10.4293/108680811X13022985132074
  53. Lagoo J, Pappas TN, Perez A. A relic or still relevant: the narrowing role for vagotomy in the treatment of peptic ulcer disease. Am J Surg 2014; 207(1):120–126. doi:10.1016/j.amjsurg.2013.02.012
  54. Csendes A, Maluenda F, Braghetto I, Schutte H, Burdiles P, Diaz JC. Prospective randomized study comparing three surgical techniques for the treatment of gastric outlet obstruction secondary to duodenal ulcer. Am J Surg 1993; 166(1):45–49. pmid:8101050
  55. Ly J, O’Grady G, Mittal A, Plank L, Windsor JA. A systematic review of methods to palliate malignant gastric outlet obstruction. Surg Endosc 2010; 24(2):290–297. doi:10.1007/s00464-009-0577-1
  56. Goldberg EM. Palliative treatment of gastric outlet obstruction in terminal patients: SEMS. Stent every malignant stricture! Gastrointest Endosc 2014; 79(1):76–78. doi:10.1016/j.gie.2013.07.056
  57. Min SH, Son SY, Jung DH, et al. Laparoscopic gastrojejunostomy versus duodenal stenting in unresectable gastric cancer with gastric outlet obstruction. Ann Surg Treat Res 2017; 93(3):130–136. doi:10.4174/astr.2017.93.3.130
  58. Roy A, Kim M, Christein J, Varadarajulu S. Stenting versus gastrojejunostomy for management of malignant gastric outlet obstruction: comparison of clinical outcomes and costs. Surg Endosc 2012; 26(11):3114–119. doi:10.1007/s00464-012-2301-9
  59. Amin S, Sethi A. Endoscopic ultrasound-guided gastrojejunostomy. Gastrointest Endosc Clin N Am 2017; 27(4):707–713. doi:10.1016/j.giec.2017.06.009
Issue
Cleveland Clinic Journal of Medicine - 86(5)
Issue
Cleveland Clinic Journal of Medicine - 86(5)
Page Number
345-353
Page Number
345-353
Publications
Publications
Topics
Article Type
Display Headline
Gastric outlet obstruction: A red flag, potentially manageable
Display Headline
Gastric outlet obstruction: A red flag, potentially manageable
Legacy Keywords
gastric outlet obstruction, GOO, gastric cancer, pancreatic cancer, endoscopic stenting, peptic ulcer disease, Andree Koop, William Palmer, Fernando Stancampiano
Legacy Keywords
gastric outlet obstruction, GOO, gastric cancer, pancreatic cancer, endoscopic stenting, peptic ulcer disease, Andree Koop, William Palmer, Fernando Stancampiano
Sections
Inside the Article

KEY POINTS

  • Causes of gastric outlet obstruction fall into 2 categories: benign and malignant. The cause should be presumed to be malignant until proven otherwise.
  • Peptic ulcer disease, a benign cause, used to account for most cases of gastric outlet obstruction. It is still common but has declined in frequency with the development of acid-suppressing drugs.
  • Gastric cancer used to be the most common malignant cause but has declined in frequency in Western countries with treatment for Helicobacter pylori infection. Now, pancreatic cancer predominates.
  • Endoscopic stenting is an effective, minimally invasive treatment for patients with malignant gastric outlet obstruction and poor prognosis, allowing resumption of oral intake and improving quality of life.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Thu, 04/25/2019 - 12:00
Un-Gate On Date
Thu, 04/25/2019 - 12:00
Use ProPublica
CFC Schedule Remove Status
Thu, 04/25/2019 - 12:00
Hide sidebar & use full width
render the right sidebar.
Article PDF Media

Acute kidney injury after hip or knee replacement: Can we lower the risk?

Article Type
Changed
Mon, 04/01/2019 - 08:59
Display Headline
Acute kidney injury after hip or knee replacement: Can we lower the risk?

Total hip or knee replacement (also called total joint arthroplasty) is highly successful at relieving pain and restoring function, but at the risk of acute kidney injury, which is a sudden loss of renal function. Various factors have been associated with this risk, some of which are potentially modifiable, notably, the use of nephrotoxic antibiotics and other drugs.

This review examines the incidence of acute kidney injury using current criteria in total joint arthroplasty of the hip or knee in general, and in the setting of revision surgery for prosthetic joint infection in particular, in which the risk is higher. We identify risk factors for acute kidney injury and propose ways to lower the risk.

MILLIONS OF PROCEDURES ANNUALLY

Total replacement of the hip1,2 or knee3 is being done more and more. Kurtz et al4 estimate that by the year 2030, we will see approximately 3.5 million primary total knee and 500,000 primary total hip replacements every year. In addition, revision total knee procedures are expected to exceed 250,000 per year, and revision total hip procedures are expected to exceed 90,000 per year.4

Chronic infection may complicate up to 2% of these procedures and is associated with significant morbidity, death, and financial costs. Currently, it may be the reason for 25% of total joint arthroplasty revisions,5 but by the year 2030, it is projected to account for 66% of revision total knee arthroplasties and 48% of revision total hip arthroplasties.6

PRIMARY TOTAL JOINT ARTHROPLASTY AND ACUTE KIDNEY INJURY

filippone_acutekidneyinjuryafterarthroplasty_t1.jpg
We searched Ovid MEDLINE for articles on acute kidney injury and either arthroplasty or antibiotic-loaded cement spacers. We found 22 studies, with a total of 72,850 patients, that assessed the incidence of acute kidney injury after primary or revision total joint arthroplasty of the hip or knee, or both, using current criteria7–28 (Table 1), and 3 additional studies that used discharge diagnosis coding.29–31

Study designs, findings varied widely

The incidence of acute kidney injury varied markedly among the studies of primary total joint arthroplasty or revision for aseptic reasons. Numerous factors explain this heterogeneity.

Designs ranged from single-center studies with relatively small numbers of patients to large regional and national samples based on administrative data.

filippone_acutekidneyinjuryafterarthroplasty_t2.jpg
The definition of acute kidney injury also varied, although many used current criteria, specifically the RIFLE (risk, injury, failure, loss, end-stage renal disease),32 AKIN (Acute Kidney Injury Network),33 and KDIGO (Kidney Disease Improving Global Outcomes)34 creatinine criteria (Table 2). Some studies considered only higher stages of acute kidney injury (equivalent to KDIGO stage 2 or 3), ignoring the most common stage, ie, stage 1. No study considered urine output criteria.

Almost all of the studies were retrospective. We are not aware of any randomized controlled trials.

Discharge diagnosis may miss many cases

Several studies based the diagnosis of acute kidney injury on International Classification of Diseases, Ninth Revision (ICD-9) coding from hospital discharge summaries.

Nadkarni et al,29 in the largest study published to date, used the nationwide inpatient sample database of more than 7 million total joint arthroplasties and found an incidence of acute kidney injury based on ICD-9 coding of 1.3% over the years 2002 to 2012, although this increased to 1.8% to 1.9% from 2010 to 2012.

Lopez-de-Andres et al,30 in a similar study using the Spanish national hospital discharge database, evaluated 20,188 patients who underwent revision total hip or knee arthroplasty and found an overall incidence of acute kidney injury of 0.94%, also using ICD-9 coding.

Gharaibeh et al31 used similar methods to diagnose acute kidney injury in a single-center study of 8,949 patients and found an incidence of 1.1%.

Although these 3 studies suggest that the incidence of acute kidney injury is relatively low, Grams et al35 found the sensitivity of ICD-9 coding from hospital records for the diagnosis of acute kidney injury to be only 11.7% compared with KDIGO serum creatinine and urine output criteria. This suggests that the true incidence in these studies may be many times higher, possibly near 10%.

Do all stages of kidney injury count?

Jafari et al,7 in a large series from a single medical center, used only the “I” (injury) and “F” (failure) levels of the RIFLE criteria (corresponding to stages 2 and 3 of the KDIGO criteria) and found an incidence of 0.55% in more than 17,000 total joint arthroplasties.

Jamsa et al8 used the same criteria for acute kidney injury (only “I” and “F”) and found 58 cases in 5,609 patients in whom postoperative serum creatinine was measured, for an incidence of 1%; the remaining 14,966 patients in their cohort did not have serum creatinine measured, and it was assumed they did not have acute kidney injury. Neither of these studies included the most common “R” (risk) stage of acute kidney injury.

Parr et al36 recently studied a nationwide sample of 657,840 hospitalized veterans and found that of 90,614 who developed acute kidney injury based on KDIGO creatinine criteria, 84% reached only stage R. This suggests that if all stages were considered, the true incidence of acute kidney injury would have been higher—possibly 4% in the Jafari series and possibly 7% in the Jamsa series.

Smaller studies had higher rates

Smaller, single-center series reported much higher incidences of acute kidney injury.

Kimmel et al11 found an incidence of 14.8% in 425 total joint arthroplasties using RIFLE creatinine criteria.

Johansson et al25 found an incidence of 19.9% in 136 total joint arthroplasties using KDIGO creatinine criteria.

Sehgal et al9 found an incidence of 21.9% in 659 total joint arthroplasties using AKIN creatinine criteria.

Challagundla et al24 found an incidence of 23.7% in 198 procedures using RIFLE creatinine criteria.

Weingarten et al,10 in a single-center series of 7,463 total joint arthroplasties, found an incidence of acute kidney injury of only 2.2% using AKIN criteria, although 12% of the patients with acute kidney injury did not return to their baseline serum creatinine levels by 3 months.

Our estimate: Nearly 10%

In total, in the 20 studies in Table 1 that included all stages of acute kidney injury, there were 1,909 cases of acute kidney injury in 34,337 patients, for an incidence of 5.6%. Considering that all studies but one were retrospective and none considered urine output criteria for acute kidney injury, we believe that using current KDIGO criteria, the true incidence of acute kidney injury complicating primary lower-extremity total joint arthroplasties is really closer to 10%.

 

 

RISK FACTORS FOR ACUTE KIDNEY INJURY

Various factors have been associated with development of acute kidney injury by multivariate analysis in these studies. Some are modifiable, while others are not, at least in the short term.

Nonmodifiable risk factors

Older age is often significant in studies assessing primary total joint arthroplasty or revision total joint arthroplasty not specifically for infection.11,12,16,17,26,28

Obesity is also a major factor in the development of acute kidney injury,7,10–12,17,18 and, along with age, is a major factor contributing to the need for joint replacement in the first place.

Male sex may increase risk.29

Diabetes mellitus was identified as a risk factor in several studies,10,12,17,20 and hypertension in a few.7,10,24

Other comorbidities and factors such as cardiovascular disease,7,10 liver disease,7 pulmonary disease,7 high American Society of Anesthesiology score,8,19 and benign heart murmurs preoperatively by routine physical examination have also been linked to acute kidney injury after joint arthroplasty.28

Chronic kidney disease as a risk factor

Chronic kidney disease at baseline was associated with acute kidney injury in several of these series.7,11–13,15,19,29

Warth et al12 studied 1,038 patients and found an incidence of acute kidney injury of 11% in the 135 with chronic kidney disease (defined as serum creatinine > 1.2 mg/dL) and who received acetaminophen or narcotics for pain control, compared with 4.8% in the remaining 903 patients without chronic kidney disease, who received ketorolac or celecoxib.

Perregaard et al13 studied 3,410 patients who underwent total hip arthroplasty and found an incidence of acute kidney injury (per KDIGO creatinine criteria) of 2.2% overall, but 7% in the 134 patients with chronic kidney disease based on KDIGO creatinine criteria.

Nowicka et al15 found an incidence of acute kidney injury of 16.7% in the 48 patients with chronic kidney disease (defined as a glomerular filtration rate estimated by the Cockroft-Gault formula of less than 60 mL/min/1.73 m2), compared with 4.5% in the remaining 289.

Modifiable risk factors

Modifiable risk factors that should be considered in high-risk cases include anemia, perioperative blood transfusion, perioperative use of renin-angiotensin-aldosterone system inhibitors such as angiotensin-converting enzyme (ACE) inhibitors and angiotensin II receptor blockers (ARBs), particular antibiotics used for prophylaxis, and nonsteroidal anti-inflammatory drugs used postoperatively.

Anemia and blood transfusion

Preoperative anemia has been associated with postoperative acute kidney injury in various surgical settings such as cardiac surgery.37,38 Perioperative red blood cell transfusions have also been associated with acute kidney injury in cardiac surgery; similar results may apply to total joint arthroplasty.

Choi et al,17 in 2,467 patients undergoing hip replacement, found a significant risk for acute kidney injury if postoperative hemoglobin was consistently below 10 g/dL compared with consistently above this level, with an inverse probability-of-treatment weighted odds ratio of 1.817 (P = .011).

Others have found a significant association of perioperative blood transfusion with acute kidney injury in total joint arthroplasty.10,29

Nadkarni et al,29 for example, used the nationwide inpatient sample database and found by multivariate analysis that perioperative blood transfusion was strongly associated with acute kidney injury, with an adjusted odds ratio of 2.28 (95% confidence interval [CI] 2.15–2.42, P < .0001).

Comment. A higher incidence of acute kidney injury may represent confounding by indication bias, as sicker patients or complicated surgeries may require transfusion, and this risk may not be completely accounted for by multivariate analysis. It is also possible, however, that transfusions per se may contribute to acute kidney injury. Possible direct or indirect mechanisms mediating acute kidney injury include hemolytic reactions, circulatory overload, acute lung injury, and immunomodulatory effects.39

Preoperative transfusion in anemic patients undergoing cardiac surgery may also reduce the incidence of postoperative acute kidney injury both by correcting the anemia and by limiting the need for perioperative transfusions.40 It remains to be determined whether elective preoperative transfusion to correct anemia would reduce postoperative development of acute kidney injury in total joint arthroplasty. As an aside, perioperative transfusion has also been linked to development of periprosthetic joint infection.41

Renin-angiotensin-aldosterone system inhibitors

Several studies found perioperative use of renin-angiotensin-aldosterone system inhibitors to be a risk factor for acute kidney injury.

Kimmel et al11 reported adjusted odds ratios of 2.70 (95% CI 1.12–6.48) for ACE inhibitor use and 2.64 (95% CI 1.18–5.93) for ARB use in a study of 425 primary total joint arthroplasties.

Challagundla et al24 found an odds ratio of 3.07 (95% CI 1.40–6.74) with ACE inhibitor or ARB use by multivariate analysis in 198 total joint arthroplasties.

Nielson et al18 studied 798 patients who underwent total joint arthroplasty and found that preoperative use of renin-angiotensin system inhibitors was associated with a significantly higher rate of postoperative acute kidney injury (8.3% vs 1.7% without inhibition), which was statistically significant by multivariate analysis (odds ratio 2.6, 95% CI 1.04–6.51).

We recommend holding renin-angiotensin-aldosterone system inhibitors 7 days before surgery through the postoperative period in high-risk cases.

Aminoglycoside use as a risk factor

Prophylactic administration of systemic antibiotics is the standard of care. In a systematic review of 26 studies and meta-analysis of 7 studies (3,065 patients), prophylactic antibiotics reduced the relative risk of wound infection by 81% with an absolute risk reduction of 8%.42

A modifiable risk factor for acute kidney injury is the specific antibiotic used for prophylaxis. Multiple studies assessed the risk of acute kidney injury comparing regimens containing an aminoglycoside (typically gentamicin) with regimens lacking these agents.20–26 In general, these studies found a significantly higher risk of acute kidney injury when gentamicin was used.

Challagundla et al24 found an incidence of acute kidney injury of 52% using RIFLE creatinine criteria in 52 patients receiving 8 g total of flucloxacillin plus 160 mg of gentamicin (120 mg if they weighed less than 60 kg) compared with 8% in 48 patients given cefuroxime (3 g total) and 14% in an additional 52 patients also given cefuroxime.

Johansson et al25 found an incidence of KDIGO creatinine-based acute kidney injury of 13% in 70 patients given dicloxacillin alone prophylactically compared with 27% given dicloxacillin and gentamicin, with a relative risk of 3.

Bell et al,21 in a large registry-based analysis from Scotland involving 7,666 elective orthopedic procedures, found that use of flucloxacillin 2 g plus a single dose of gentamicin 4 mg/kg was significantly associated with a 94% higher risk of acute kidney injury (KDIGO creatinine criteria) compared with a cefuroxime-based regimen, with absolute rates increasing from 6.2% to 10.8%.

Dubrovskaya et al20 and Ferguson et al,26 in contrast, found no increased risk with addition of gentamicin.

We recommend avoiding aminoglycosides for prophylaxis in primary lower-extremity total joint arthroplasty in patients at higher risk unless required for specific microbiologic reasons.

Vancomycin may also increase risk

Courtney et al19 assessed the risk of adding vancomycin to cefazolin for routine prophylaxis in a retrospective series of 1,828 total hip or knee arthroplasties and found a significantly higher rate of acute kidney injury, using AKIN criteria (13% vs 8%, odds ratio by multivariate analysis 1.82, P = .002).19

Other agents shown to be effective in treating periprosthetic joint infections or complicated skin and soft-tissue infections with resistant organisms include daptomycin43 and linezolid.44 These nonnephrotoxic alternatives to vancomycin may be a consideration if prophylaxis for methicillin-resistant Staphylococcus aureus is deemed necessary in patients at risk for acute kidney injury.

 

 

PROSTHETIC JOINT INFECTIONS AND ANTIBIOTIC-LOADED CEMENT

Deep infection may complicate nearly 1% of total hip45 and 2% of total knee arthroplasties.46 Kurtz et al4,6 have projected that by 2030, infection will be the cause of two-thirds of the estimated 268,000 revision total knee arthroplasties and about half of the estimated 96,700 revision total hip arthroplasties.

The most common method of treating a chronically infected replacement joint is a 2-stage procedure.5 First, the prosthesis is removed, all infected bone and soft tissue is debrided, and an antibiotic-loaded cement spacer is implanted. Systemic antibiotics are given concurrently, typically for about 6 weeks. After the infection is brought under control, perhaps 2 to 3 months later, the spacer is removed and a new joint is implanted with antibiotic-loaded cement. A 1-stage procedure may be an option in selected cases and would obviate the need for an antibiotic-loaded cement spacer.47,48

Of obvious relevance to development of acute kidney injury is the choice and amount of antibiotics embedded in the cement used for spacers and in implantation. Very high antibiotic levels are achieved within the joint space, usually with little systemic absorption, although significant systemic exposure has been documented in some cases.

The polymethylmethacrylate cement used for these purposes comes in 40-g bags. Multiple bags are typically required per joint, perhaps 2 to 4.49

The rate of elution of antibiotics is determined by several factors, including surface area, porosity, and the number of antibiotics. In general, elution is greatest early on, with exponential decline lasting perhaps 1 week,  followed by slow, sustained release over weeks to months.50 However, several in vitro studies have indicated that only about 5%50,51 of the total antibiotic actually elutes over time.

Initially, multiple antibiotic-laden cement beads were used to fill the joint space, but this significantly limited function and mobility.52 Now, cement spacers are used, and they can be nonarticulating or articulating for maximal joint mobility.53 Although much greater antibiotic elution occurs from beads due to their high surface area-to-volume ratio, spacers still provide an adequate dose.

ANTIBIOTIC-LOADED CEMENT: DOSAGE AND ELUTION CHARACTERISTICS

Antibiotic-loaded cement can be either low-dose or high-dose.

Low-dose cement

Low-dose cement typically consists of 0.5 to 1.0 g of antibiotic per 40-g bag of cement, usually an aminoglycoside (gentamicin or tobramycin) or vancomycin, and can be purchased premixed by the manufacturer. Such cement is only used prophylactically with primary total joint arthroplasty or revision for aseptic reasons, a practice common in Europe but less so in the United States. Some American authors propose antibiotic-loaded cement prophylaxis for patients at high risk, eg, those with immunosuppression, inflammatory cause of arthritis, or diabetes.54

Vrabec et al,55 in a study of low-dose tobramycin-loaded cement used for primary total knee arthroplasty, found a peak median intra-articular tobramycin concentration of 32 mg/L at 6 hours, declining to 6 mg/L at 48 hours with all serum levels 0.3 mg/L or less (unmeasureable) at similar time points.

Sterling et al,56 studying primary total hip arthroplasties with low-dose tobramycin-loaded cement, found mean levels in drainage fluid of 103 mg/L at 6 hours, declining to 15 mg/L at 48 hours. Serum levels peaked at 0.94 mg/L at 3 hours, declining to 0.2 mg/L by 48 hours.

Although most of the antibiotic elution occurs early (within the first week), antibiotic can be found in joint aspirates up to 20 years later.57 We are unaware of any well-documented cases of acute kidney injury ascribable to low-dose antibiotic-loaded cement used prophylactically. One case report making this assertion did not determine serum levels of aminoglycoside.58

High-dose cement

High-dose antibiotic-loaded cement typically contains about 4 to 8 g of antibiotic per 40-g bag of cement and is used in the treatment of prosthetic joint infection to form the spacers. The antibiotic must be mixed into the cement powder by the surgeon in the operating room.

There is no standard combination or dosage. The choice of antibiotic can be tailored to the infecting organism if known. Otherwise, gram-positive organisms are most common, and vancomycin and aminoglycosides are often used together. This particular combination will enhance the elution of both antibiotics when studied in vitro, a process termed “passive opportunism.”59 Other antibiotics in use include aztreonam, piperacillin, teicoplanin, fluoroquinolones, cephalosporins, and daptomycin, among others.

About 8 g of antibiotic total per 40-g bag is the maximum to allow easy molding.52 As an example, this may include 4 g of vancomycin and 3.6 g of tobramycin per 40 g. Given that 3 to 4 such bags are often used per joint, there is significant risk of systemic exposure.

Kalil et al60 studied 8 patients who received high-dose tobramycin-loaded cement to treat periprosthetic joint infections of the hip or knee and found that 7 had detectable serum levels (mean 0.84 mg/L, highest 2.0 mg/L), including 1 with a level of 0.9 mg/L on day 38; 4 of these 8 developed acute kidney injury by AKIN criteria, although other risk factors for acute kidney injury existed. Nearly all had concomitant vancomycin (3 to 8 g) added to the cement as well.

Hsieh et al61 studied 46 patients with infected total hip arthroplasties treated with high-dose antibiotic-loaded cement spacers (vancomycin 4 g and aztreonam 4 g per 40-g bag) and found vancomycin levels in joint drainage higher than 1,500 mg/L on day 1, decreasing to 571 mg/L on day 7; serum levels were low (range 0.1–1.6 mg/L at 24 hours), falling to undetectable by 72 hours.

 

 

ANTIBIOTIC-LOADED CEMENT SPACERS AND ACUTE KIDNEY INJURY

Case reports have associated high-dose antibiotic-loaded cement spacers with acute kidney injury.

Curtis et al62 described an 85-year-old patient with stage 3 chronic kidney disease who was treated for an infected total knee arthroplasty with an antibiotic-loaded cement spacer (containing 3.6 g of tobramycin and 3 g of cefazolin per 40-g bag, 3 bags total) and developed stage 3 acute kidney injury. After 16 days and 3 hemodialysis sessions, the patient’s serum tobramycin level was still 2 mg/L despite receiving no systemic tobramycin.

Wu et al63 reported a case of acute kidney injury that required dialysis after implantation of a tobramycin- and vancomycin-loaded spacer, with persistent serum tobramycin levels despite repeated hemodialysis sessions until the spacer was removed.

Chalmers et al64 described 2 patients with acute kidney injury and persistently elevated serum tobramycin levels (3.9 mg/L on day 39 in 1 patient and 2.0 mg/L on day 24 in the other patient) despite no systemic administration.

In these and other case reports,65–67 dialysis and spacer explantation were usually required. 


Comment. It is intuitive that acute kidney injury would more likely complicate revision total joint arthroplasties for infection than for primary total joint arthroplasties or revisions for aseptic reasons, given the systemic effects of infection and exposure to nephrotoxic or allergenic antibiotics. And the available data suggest that the risk of acute kidney injury is higher with revision for prosthetic joint infection than with revision for aseptic reasons. However, many of the studies were retrospective, relatively small, single-center series and used different definitions of acute kidney injury.

filippone_acutekidneyinjuryafterarthroplasty_t3.jpg
We are aware of 17 studies specifically addressing acute kidney injury or postoperative complications in general that may have included acute kidney injury.50,52,61,68–81 Ten of these studies found at least 1 case of acute kidney injury (Table 3). Of note, 7 studies totaling 219 patients reported no cases of acute kidney injury, although acute kidney injury per se was not mentioned and no definition of it was provided.50,61,76,77,79,80,82

Luu et al83 performed a systematic review of studies published between January 1989 and June 2012 reporting systemic complications (including acute kidney injury) of 2-stage revision arthroplasties including placement of an antibiotic-loaded cement spacer for treatment of periprosthetic joint infection. Overall, 10 studies were identified with 544 total patients. Five of these studies, with 409 patients, reported at least 1 case of acute kidney injury for a total of 27 patients, giving an incidence of 6.6% in these studies.68–71 The remaining 5 studies, totaling 135 patients, did not report any cases of acute kidney injury,50,61,76–78 although that was not the primary focus of any of those trials.

Most notable from this systematic review, the study of Menge et al69 retrospectively determined the incidence of acute kidney injury (defined as a 50% rise in serum creatinine to > 1.4 mg/dL within 90 days of surgery) to be 17% in 84 patients with infected total knee arthroplasties treated with antibiotic-loaded cement spacers. A mean of 3.5 bags of cement per spacer were used in the 35 articulating spacers, compared with 2.9 per nonarticulating spacer. These spacers contained vancomycin in 82% (median 4.0 g, range 1–16 g) and tobramycin in 94% (median 4.8 g, range 1–12 g), among others in small percentages. The dose of tobramycin in the spacer considered either as a dichotomous variable (> 4.8 g, OR 5.87) or linearly (OR 1.24 per 1-g increase) was significantly associated with acute kidney injury, although systemic administration of aminoglycosides or vancomycin was not.

Additional single-center series that were published subsequent to this review have generally used more current diagnostic criteria.

Noto et al72 found that 10 of 46 patients treated with antibiotic-loaded cement spacers had a greater than 50% rise in serum creatinine (average increase 260%). All spacers contained tobramycin (mean dose 8.2 g), and 9 of 10 also contained vancomycin (mean 7.6 g). All of the 9 patients with acute kidney injury with follow-up data recovered renal function.

Reed et al75 found 26 cases of acute kidney injury (based on RIFLE creatinine criteria) in 306 patients with antibiotic-loaded cement spacers treating various periprosthetic joint infections (including hips, knees, shoulders, and digits) and compared them with 74 controls who did not develop acute kidney injury. By multivariable analysis, receipt of an ACE inhibitor within 7 days of surgery and receipt of piperacillin-tazobactam within 7 days after surgery were both significantly more common in cases with acute kidney injury than in controls without acute kidney injury.

Aeng et al73 prospectively studied 50 consecutive patients receiving antibiotic-loaded spacers containing tobramycin (with or without vancomycin) for treatment of infected hip or knee replacements. Using RIFLE creatinine criteria, they found an incidence of acute kidney injury of 20% (10 of 50). Factors significantly associated with acute kidney injury included cement premixed by the manufacturer with gentamicin (0.5 g per 40-g bag) in addition to the tobramycin they added, intraoperative blood transfusions, and postoperative use of nonsteroidal anti-inflammatory drugs.

Geller et al,74 in a multicenter retrospective study of 247 patients with prosthetic joint infections (156 knees and 91 hips) undergoing antibiotic-loaded cement spacer placement, found an incidence of acute kidney injury of 26% based on KDIGO creatinine criteria. Significant risk factors included higher body mass index, lower preoperative hemoglobin level, drop in hemoglobin after surgery, and comorbidity (hypertension, diabetes, chronic kidney disease, or cardiovascular disease). Most of the spacers contained a combination of vancomycin and either tobramycin (81%) or gentamicin (13%). The spacers contained an average of 5.3 g (range 0.6–18 g) of vancomycin (average 2.65 g per 40-g bag) and an average of 5.2 g (range 0.5–16.4 g) of tobramycin (average 2.6 g per bag).

As in Menge et al,69 this study illustrates the wide range of antibiotic dosages in use and the lack of standardization. In contrast to the study by Menge et al, however, development of acute kidney injury was not related to the amount of vancomycin or tobramycin contained in the spacers. Eventual clearance of infection (at 1 and 2 years) was significantly related to increasing amounts of vancomycin. Multiple different systemic antibiotics were used, most commonly vancomycin (44%), and systemic vancomycin was not associated with acute kidney injury.

Yadav et al,81 in a study of 3,129 consecutive revision procedures of the knee or hip, found an incidence of acute kidney injury by RIFLE creatinine criteria of 29% in the 197 patients who received antibiotic-loaded cement spacers for periprosthetic joint infection compared with 3.4% in the 2,848 who underwent revision for aseptic reasons. In 84 patients with prosthetic joint infection having various surgeries not including placement of a spacer, the acute kidney injury rate at some point in their course was an alarmingly high 82%. In the group that received spacers, only age and comorbidity as assessed by Charlson comorbidity index were independently associated with acute kidney injury by multivariate analysis. Surprisingly, modest renal impairment was protective, possibly because physicians of patients with chronic kidney disease were more vigilant and took appropriate measures to prevent acute kidney injury.

Overall, the risk of acute kidney injury appears to be much higher during treatment of prosthetic joint infection with a 2-stage procedure using an antibiotic-loaded cement spacer than after primary total joint arthroplasty or revision for aseptic reasons, and may complicate up to one-third of cases.

 

 

REDUCING RISK DURING TREATMENT OF INFECTED REPLACEMENT JOINTS

filippone_acutekidneyinjuryafterarthroplasty_t4.jpg
Due to lack of appropriate data, how best to mitigate the risk of acute kidney injury is uncertain. In our opinion, however, the following measures should be considered (Table 4).

As in primary total joint arthroplasty in general, higher-risk cases should be identified based on age, body mass index, chronic kidney disease, comorbidities (hypertension, diabetes, established cardiovascular disease), and anemia.

Preoperative transfusion can be considered case by case depending on degree of anemia and associated risk factors.

All renin-angiotensin-aldosterone system inhibitors should be withheld starting 1 week before surgery.

Both nonselective and cyclooxygenase-2 selective nonsteroidal anti-inflammatory drugs should be avoided, if possible.

Strict attention should be paid to adequate intraoperative and postoperative fluid resuscitation.

Kidney function should be monitored closely in the early postoperative period, including urine output and daily creatinine for at least 72 hours.

Systemic administration of potentially nephrotoxic antibiotics should be minimized, especially the combination of vancomycin with piperacillin-tazobactam.84 Daptomycin is a consideration.43

If acute kidney injury should develop, serum levels of vancomycin or aminoglycosides should be measured if the spacer contains these antibiotics. The spacer may need to be removed if toxic serum levels persist.

TAKE-HOME POINTS

Acute kidney injury may complicate up to 10% of primary lower-extremity total joint arthroplasties and up to 25% of periprosthetic joint infections treated with a 2-stage procedure including placement of an antibiotic-loaded cement spacer in the first stage.

Risk factors for acute kidney injury include older age, obesity, chronic kidney disease, and overall comorbidity. Potentially modifiable risk factors include anemia, perioperative transfusions, aminoglycoside prophylaxis, perioperative renin-angiotensin system blockade, and postoperative nonsteroidal anti-inflammatory drugs. These should be mitigated when possible.

In patients with periprosthetic joint infection who receive antibiotic-loaded cement spacers, especially patients  with additional risk factors for acute kidney injury, strict attention should be paid to the dose of antibiotic in the spacer, with levels checked postoperatively if necessary. Nonnephrotoxic antibiotics should be chosen for systemic administration when possible.

Prospective randomized controlled trials are needed to guide therapy after total joint arthroplasty, and to verify the adverse long-term outcomes of acute kidney injury in this setting.

References
  1. Learmonth ID, Young C, Rorabeck C. The operation of the century: total hip replacement. Lancet 2007; 370(9597):1508–1519. doi:10.1016/S0140-6736(07)60457-7
  2. Pivec R, Johnson AJ, Mears SC, Mont MA. Hip arthroplasty. Lancet 2012; 380(9855):1768–1777. doi:10.1016/S0140-6736(12)60607-2
  3. Carr AJ, Robertsson O, Graves S, et al. Knee replacement. Lancet 2012; 379(9823):1331–1340. doi:10.1016/S0140-6736(11)60752-6
  4. Kurtz S, Ong K, Lau E, Mowat F, Halpern M. Projections of primary and revision hip and knee arthroplasty in the United States from 2005 to 2030. J Bone Joint Surg Am 2007; 89(4):780–785. doi:10.2106/JBJS.F.00222
  5. Kapadia BH, Berg RA, Daley JA, Fritz J, Bhave A, Mont MA. Periprosthetic joint infection. Lancet 2016; 387(10016):386–394. doi:10.1016/S0140-6736(14)61798-0
  6. Kurtz SM, Ong KL, Schmier J, et al. Future clinical and economic impact of revision total hip and knee arthroplasty. J Bone Joint Surg Am 2007; 89(suppl 3):144–151. doi:10.2106/JBJS.G.00587
  7. Jafari SM, Huang R, Joshi A, Parvizi J, Hozack WJ. Renal impairment following total joint arthroplasty: who is at risk? J Arthroplasty 2010; 25(6 suppl):49–53, 53.e1–2. doi:10.1016/j.arth.2010.04.008
  8. Jamsa P, Jamsen E, Lyytikainen LP, Kalliovalkama J, Eskelinen A, Oksala N. Risk factors associated with acute kidney injury in a cohort of 20,575 arthroplasty patients. Acta Orthop 2017; 88(4):370–376. doi:10.1080/17453674.2017.1301743
  9. Sehgal V, Bajwa SJ, Sehgal R, Eagan J, Reddy P, Lesko SM. Predictors of acute kidney injury in geriatric patients undergoing total knee replacement surgery. Int J Endocrinol Metab 2014; 12(3):e16713. doi:10.5812/ijem.16713
  10. Weingarten TN, Gurrieri C, Jarett PD, et al. Acute kidney injury following total joint arthroplasty: retrospective analysis. Can J Anaesth 2012; 59(12):1111–1118. doi:10.1007/s12630-012-9797-2
  11. Kimmel LA, Wilson S, Janardan JD, Liew SM, Walker RG. Incidence of acute kidney injury following total joint arthroplasty: a retrospective review by RIFLE criteria. Clin Kidney J 2014; 7(6):546–551. doi:10.1093/ckj/sfu108
  12. Warth LC, Noiseux NO, Hogue MH, Klaassen AL, Liu SS, Callaghan JJ. Risk of acute kidney injury after primary and revision total hip arthroplasty and total knee arthroplasty using a multimodal approach to perioperative pain control including ketorolac and celecoxib. J Arthroplasty 2016; 31(1):253–255. doi:10.1016/j.arth.2015.08.012
  13. Perregaard H, Damholt MB, Solgaard S, Petersen MB. Renal function after elective total hip replacement. Acta Orthop 2016; 87(3):235–238. doi:10.3109/17453674.2016.1155130
  14. Hassan BK, Sahlström A, Dessau RB. Risk factors for renal dysfunction after total hip joint replacement; a retrospective cohort study. J Orthop Surg Res 2015; 10:158. doi:10.1186/s13018-015-0299-0
  15. Nowicka A, Selvaraj T. Incidence of acute kidney injury after elective lower limb arthroplasty. J Clin Anesth 2016; 34:520–523. doi:10.1016/j.jclinane.2016.06.010
  16. Kim HJ, Koh WU, Kim SG, et al. Early postoperative albumin level following total knee arthroplasty is associated with acute kidney injury: a retrospective analysis of 1309 consecutive patients based on kidney disease improving global outcomes criteria. Medicine (Baltimore) 2016; 95(31):e4489. doi:10.1097/MD.0000000000004489
  17. Choi YJ, Kim S, Sim JH, Hahm K. Postoperative anemia is associated with acute kidney injury in patients undergoing total hip replacement arthroplasty: a retrospective study. Anesth Analg 2016; 122(6):1923–1928. doi:10.1213/ANE.0000000000001003
  18. Nielson E, Hennrikus E, Lehman E, Mets B. Angiotensin axis blockade, hypotension, and acute kidney injury in elective major orthopedic surgery. J Hosp Med 2014; 9(5):283–288. doi:10.1002/jhm.2155
  19. Courtney PM, Melnic CM, Zimmer Z, Anari J, Lee GC. Addition of vancomycin to cefazolin prophylaxis is associated with acute kidney injury after primary joint arthroplasty. Clin Orthop Relat Res 2015; 473(7):2197–2203. doi:10.1007/s11999-014-4062-3
  20. Dubrovskaya Y, Tejada R, Bosco J 3rd, et al. Single high dose gentamicin for perioperative prophylaxis in orthopedic surgery: evaluation of nephrotoxicity. SAGE Open Med 2015; 3:2050312115612803. doi:10.1177/2050312115612803
  21. Bell S, Davey P, Nathwani D, et al. Risk of AKI with gentamicin as surgical prophylaxis. J Am Soc Nephrol 2014; 25(11):2625–2632. doi:10.1681/ASN.2014010035
  22. Ross AD, Boscainos PJ, Malhas A, Wigderowitz C. Peri-operative renal morbidity secondary to gentamicin and flucloxacillin chemoprophylaxis for hip and knee arthroplasty. Scott Med J 2013; 58(4):209–212. doi:10.1177/0036933013507850
  23. Bailey O, Torkington MS, Anthony I, Wells J, Blyth M, Jones B. Antibiotic-related acute kidney injury in patients undergoing elective joint replacement. Bone Joint J 2014; 96-B(3):395–398. doi:10.1302/0301-620X.96B3.32745
  24. Challagundla SR, Knox D, Hawkins A, et al. Renal impairment after high-dose flucloxacillin and single-dose gentamicin prophylaxis in patients undergoing elective hip and knee replacement. Nephrol Dial Transplant 2013; 28(3):612–619. doi:10.1093/ndt/gfs458
  25. Johansson S, Christensen OM, Thorsmark AH. A retrospective study of acute kidney injury in hip arthroplasty patients receiving gentamicin and dicloxacillin. Acta Orthop 2016; 87(6):589–591. doi:10.1080/17453674.2016.1231008
  26. Ferguson KB, Winter A, Russo L, et al. Acute kidney injury following primary hip and knee arthroplasty surgery. Ann R Coll Surg Eng 2017; 99(4):307–312. doi:10.1308/rcsann.2016.0324
  27. Bjerregaard LS, Jorgensen CC, Kehlet H; Lundbeck Foundation Centre for Fast-Track Hip and Knee Replacement Collaborative Group. Serious renal and urological complications in fast-track primary total hip and knee arthroplasty; a detailed observational cohort study. Minerva Anestesiol 2016; 82(7):767–776. pmid:27028450
  28. Friedman JM, Couso R, Kitchens M, et al. Benign heart murmurs as a predictor for complications following total joint arthroplasty. J Orthop 2017; 14(4):470–474. doi:10.1016/j.jor.2017.07.009
  29. Nadkarni GN, Patel AA, Ahuja Y, et al. Incidence, risk factors, and outcome trends of acute kidney injury in elective total hip and knee arthroplasty. Am J Orthop (Belle Mead NJ) 2016; 45(1):E12–E19. pmid:26761921
  30. Lopez-de-Andres A, Hernandez-Barrera V, Martinez-Huedo MA, Villanueva-Martinez M, Jimenez-Trujillo I, Jimenez-Garcia R. Type 2 diabetes and in-hospital complications after revision of total hip and knee arthroplasty. PLoS One 2017; 12(8):e0183796. doi:10.1371/journal.pone.0183796
  31. Gharaibeh KA, Hamadah AM, Sierra RJ, Leung N, Kremers WK, El-Zoghby ZM. The rate of acute kidney injury after total hip arthroplasty is low but increases significantly in patients with specific comorbidities. J Bone Joint Surg Am 2017; 99(21):1819–1826. doi:10.2106/JBJS.16.01027
  32. Bellomo R, Ronco C, Kellum JA, Mehta RL, Palevsky P; Acute Dialysis Quality Initiative Workgroup. Acute renal failure—definition, outcome measures, animal models, fluid therapy and information technology needs: the Second International Consensus Conference of the Acute Dialysis Quality Initiative (ADQI) Group. Crit Care 2004; 8(4):R204–R212. doi:10.1186/cc2872
  33. Mehta RL, Kellum JA, Shah SV, et al; Acute Kidney Injury Network. Acute Kidney Injury Network: report of an initiative to improve outcomes in acute kidney injury. Crit Care 2007; 11(2):R31. doi:10.1186/cc5713
  34. Section 2: AKI Definition. Kidney Int Suppl (2011) 2012; 2(1):19–36. doi:10.1038/kisup.2011.32
  35. Grams ME, Waikar SS, MacMahon B, Whelton S, Ballew SH, Coresh J. Performance and limitations of administrative data in the identification of AKI. Clin J Am Soc Nephrol 2014; 9(4):682–689. doi:10.2215/CJN.07650713
  36. Parr SK, Matheny ME, Abdel-Kader K, et al. Acute kidney injury is a risk factor for subsequent proteinuria. Kidney Int 2018; 93(2):460–469. doi:10.1016/j.kint.2017.07.007
  37. Karkouti K, Wijeysundera DN, Yau TM, et al. Acute kidney injury after cardiac surgery: focus on modifiable risk factors. Circulation 2009; 119(4):495–502. doi:10.1161/CIRCULATIONAHA.108.786913
  38. Karkouti K, Grocott HP, Hall R, et al. Interrelationship of preoperative anemia, intraoperative anemia, and red blood cell transfusion as potentially modifiable risk factors for acute kidney injury in cardiac surgery: a historical multicentre cohort study. Can J Anaesth 2015; 62(4):377–384. doi:10.1007/s12630-014-0302-y
  39. Carson JL, Triulzi DJ, Ness PM. Indications for and adverse effects of red-cell transfusion. N Engl J Med 2017; 377(13):1261–1272. doi:10.1056/NEJMra1612789
  40. Karkouti K, Wijeysundera DN, Yau TM, et al. Advance targeted transfusion in anemic cardiac surgical patients for kidney protection: an unblinded randomized pilot clinical trial. Anesthesiology 2012; 116(3):613–621. doi:10.1097/ALN.0b013e3182475e39
  41. Newman ET, Watters TS, Lewis JS, et al. Impact of perioperative allogeneic and autologous blood transfusion on acute wound infection following total knee and total hip arthroplasty. J Bone Joint Surg Am 2014; 96(4):279–284. doi:10.2106/JBJS.L.01041
  42. AlBuhairan B, Hind D, Hutchinson A. Antibiotic prophylaxis for wound infections in total joint arthroplasty: a systematic review. J Bone Joint Surg Br 2008; 90(7):915–919. doi:10.1302/0301-620X.90B7.20498
  43. Corona Pérez-Cardona PS, Barro Ojeda V, Rodriguez Pardo D, et al. Clinical experience with daptomycin for the treatment of patients with knee and hip periprosthetic joint infections. J Antimicrob Chemother 2012; 67(7):1749–1754. doi:10.1093/jac/dks119
  44. Itani KM, Biswas P, Reisman A, Bhattacharyya H, Baruch AM. Clinical efficacy of oral linezolid compared with intravenous vancomycin for the treatment of methicillin-resistant Staphylococcus aureus-complicated skin and soft tissue infections: a retrospective, propensity score-matched, case-control analysis. Clin Ther 2012; 34(8):1667–1673.e1. doi:10.1016/j.clinthera.2012.06.018
  45. Dale H, Hallan G, Hallan G, Espehaug B, Havelin LI, Engesaeter LB. Increasing risk of revision due to deep infection after hip arthroplasty. Acta Orthop 2009; 80(6):639–645. doi:10.3109/17453670903506658
  46. Kurtz SM, Ong KL, Lau E, Bozic KJ, Berry D, Parvizi J. Prosthetic joint infection risk after TKA in the Medicare population. Clin Orthop Relat Res 2010; 468(1):52–56. doi:10.1007/s11999-009-1013-5
  47. Kunutsor SK, Whitehouse MR, Lenguerrand E, Blom AW, Beswick AD; INFORM Team. Re-infection outcomes following one- and two-stage surgical revision of infected knee prosthesis: a systematic review and meta-analysis. PLoS One 2016; 11(3):e0151537. doi:10.1371/journal.pone.0151537
  48. Negus JJ, Gifford PB, Haddad FS. Single-stage revision arthroplasty for infection—an underutilized treatment strategy. J Arthroplasty 2017; 32(7):2051–2055. doi:10.1016/j.arth.2017.02.059
  49. Stevens CM, Tetsworth KD, Calhoun JH, Mader JT. An articulated antibiotic spacer used for infected total knee arthroplasty: a comparative in vitro elution study of Simplex and Palacos bone cements. J Orthop Res 2005; 23(1):27–33. doi:10.1016/j.orthres.2004.03.003
  50. Chohfi M, Langlais F, Fourastier J, Minet J, Thomazeau H, Cormier M. Pharmacokinetics, uses, and limitations of vancomycin-loaded bone cement. Int Orthop 1998; 22(3):171–177. pmid:9728311
  51. Amin TJ, Lamping JW, Hendricks KJ, McIff TE. Increasing the elution of vancomycin from high-dose antibiotic-loaded bone cement: a novel preparation technique. J Bone Joint Surg Am 2012; 94(21):1946–1951. doi:10.2106/JBJS.L.00014
  52. Hsieh PH, Chen LH, Chen CH, Lee MS, Yang WE, Shih CH. Two-stage revision hip arthroplasty for infection with a custom-made, antibiotic-loaded, cement prosthesis as an interim spacer. J Trauma 2004; 56(6):1247–1252. pmid:15211133
  53. Cui Q, Mihalko WM, Shields JS, Ries M, Saleh KJ. Antibiotic-impregnated cement spacers for the treatment of infection associated with total hip or knee arthroplasty. J Bone Joint Surg Am 2007; 89(4):871–882. doi:10.2106/JBJS.E.01070
  54. Jiranek WA, Hanssen AD, Greenwald AS. Antibiotic-loaded bone cement for infection prophylaxis in total joint replacement. J Bone Joint Surg Am 2006; 88(11):2487–2500. doi:10.2106/JBJS.E.01126
  55. Vrabec G, Stevenson W, Elguizaoui S, Kirsch M, Pinkowski J. What is the intraarticular concentration of tobramycin using low-dose tobramycin bone cement in TKA: an in vivo analysis? Clin Orthop Relat Res 2016; 474(11):2441–2447. doi:10.1007/s11999-016-5006-x
  56. Sterling GJ, Crawford S, Potter JH, Koerbin G, Crawford R. The pharmacokinetics of Simplex-tobramycin bone cement. J Bone Joint Surg Br 2003; 85(5):646–649. pmid:12892183
  57. Fletcher MD, Spencer RF, Langkamer VG, Lovering AM. Gentamicin concentrations in diagnostic aspirates from 25 patients with hip and knee arthroplasties. Acta Orthop Scand 2004; 75(2):173–176. doi:10.1080/00016470412331294425
  58. Lau BP, Kumar VP. Acute kidney injury (AKI) with the use of antibiotic-impregnated bone cement in primary total knee arthroplasty. Ann Acad Med Singapore 2013; 42(12):692–695. pmid:24463833
  59. Penner MJ, Masri BA, Duncan CP. Elution characteristics of vancomycin and tobramycin combined in acrylic bone-cement. J Arthroplasty 1996; 11(8):939–944. pmid:8986572
  60. Kalil GZ, Ernst EJ, Johnson SJ, et al. Systemic exposure to aminoglycosides following knee and hip arthroplasty with aminoglycoside-loaded bone cement implants. Ann Pharmacother 2012; 46(7–8):929–934. doi:10.1345/aph.1R049
  61. Hsieh PH, Chang YH, Chen SH, Ueng SW, Shih CH. High concentration and bioactivity of vancomycin and aztreonam eluted from simplex cement spacers in two-stage revision of infected hip implants: a study of 46 patients at an average follow-up of 107 days. J Orthop Res 2006; 24(8):1615–1621. doi:10.1002/jor.20214
  62. Curtis JM, Sternhagen V, Batts D. Acute renal failure after placement of tobramycin-impregnated bone cement in an infected total knee arthroplasty. Pharmacotherapy 2005; 25(6):876–880. pmid:15927906
  63. Wu IM, Marin EP, Kashgarian M, Brewster UC. A case of an acute kidney injury secondary to an implanted aminoglycoside. Kidney Int 2009; 75(10):1109–1112. doi:10.1038/ki.2008.386
  64. Chalmers PN, Frank J, Sporer SM. Acute postoperative renal failure following insertion of an antibiotic-impregnated cement spacer in revision total joint arthroplasty: two case reports. JBJS Case Connect 2012; 2(1):e12. doi:10.2106/JBJS.CC.K.00094
  65. Patrick BN, Rivey MP, Allington DR. Acute renal failure associated with vancomycin- and tobramycin-laden cement in total hip arthroplasty. Ann Pharmacother 2006; 40(11):2037–2042. doi:10.1345/aph.1H173
  66. Dovas S, Liakopoulos V, Papatheodorou L, et al. Acute renal failure after antibiotic-impregnated bone cement treatment of an infected total knee arthroplasty. Clin Nephrol 2008; 69(3):207–212. pmid:18397720
  67. McGlothan KR, Gosmanova EO. A case report of acute interstitial nephritis associated with antibiotic-impregnated orthopedic bone-cement spacer. Tenn Med 2012; 105(9):37–40, 42. pmid:23097958
  68. Jung J, Schmid NV, Kelm J, Schmitt E, Anagnostakos K. Complications after spacer implantation in the treatment of hip joint infections. Int J Med Sci 2009; 6(5):265–273. pmid:19834592
  69. Menge TJ, Koethe JR, Jenkins CA, et al. Acute kidney injury after placement of an antibiotic-impregnated cement spacer during revision total knee arthroplasty. J Arthroplasty 2012; 27(6):1221–1227.e1–2. doi:10.1016/j.arth.2011.12.005
  70. Gooding CR, Masri BA, Duncan CP, Greidanus NV, Garbuz DS. Durable infection control and function with the PROSTALAC spacer in two-stage revision for infected knee arthroplasty. Clin Orthop Relat Res 2011; 469(4):985–993. doi:10.1007/s11999-010-1579-y
  71. Springer BD, Lee GC, Osmon D, Haidukewych GJ, Hanssen AD, Jacofsky DJ. Systemic safety of high-dose antibiotic-loaded cement spacers after resection of an infected total knee arthroplasty. Clin Orthop Relat Res 2004; 427:47–51. pmid:15552135
  72. Noto MJ, Koethe JR, Miller G, Wright PW. Detectable serum tobramycin levels in patients with renal dysfunction and recent placement of antibiotic-impregnated cement knee or hip spacers. Clin Infect Dis 2014; 58(12):1783–1784. doi:10.1093/cid/ciu159
  73. Aeng ES, Shalansky KF, Lau TT, et al. Acute kidney injury with tobramycin-impregnated bone cement spacers in prosthetic joint infections. Ann Pharmacother 2015; 49(11):1207–1213. doi:10.1177/1060028015600176
  74. Geller JA, Cunn G, Herschmiller T, Murtaugh T, Chen A. Acute kidney injury after first-stage joint revision for infection: Risk factors and the impact of antibiotic dosing. J Arthroplasty 2017; 32(10):3120–3125. doi:10.1016/j.arth.2017.04.054
  75. Reed EE, Johnston J, Severing J, Stevenson KB, Deutscher M. Nephrotoxicity risk factors and intravenous vancomycin dosing in the immediate postoperative period following antibiotic-impregnated cement spacer placement. Ann Pharmacother 2014; 48(8):962–969. doi:10.1177/1060028014535360
  76. Koo KH, Yang JW, Cho SH, et al. Impregnation of vancomycin, gentamicin, and cefotaxime in a cement spacer for two-stage cementless reconstruction in infected total hip arthroplasty. J Arthroplasty 2001; 16(7):882–892. doi:10.1054/arth.2001.24444
  77. Forsythe ME, Crawford S, Sterling GJ, Whitehouse SL, Crawford R. Safeness of simplex-tobramycin bone cement in patients with renal dysfunction undergoing total hip replacement. J Orthop Surg (Hong Kong) 2006; 14(1):38–42. doi:10.1177/230949900601400109
  78. Hsieh PH, Huang KC, Tai CL. Liquid gentamicin in bone cement spacers: in vivo antibiotic release and systemic safety in two-stage revision of infected hip arthroplasty. J Trauma 2009; 66(3):804–808. doi:10.1097/TA.0b013e31818896cc
  79. Hofmann AA, Goldberg T, Tanner AM, Kurtin SM. Treatment of infected total knee arthroplasty using an articulating spacer: 2- to 12-year experience. Clin Orthop Relat Res 2005; 430:125–131. pmid:15662313
  80. Evans RP. Successful treatment of total hip and knee infection with articulating antibiotic components: a modified treatment method. Clin Orthop Relat Res 2004; 427:37–46. pmid:15552134
  81. Yadav A, Alijanipour P, Ackerman CT, Karanth S, Hozack WJ, Filippone EJ. Acute kidney injury following failed total hip and knee arthroplasty. J Arthroplasty 2018; 33(10):3297–3303. doi:10.1016/j.arth.2018.06.019
  82. Hsieh PH, Huang KC, Lee PC, Lee MS. Two-stage revision of infected hip arthroplasty using an antibiotic-loaded spacer: retrospective comparison between short-term and prolonged antibiotic therapy. J Antimicrob Chemother 2009; 64(2):392–397. doi:10.1093/jac/dkp177
  83. Luu A, Syed F, Raman G, et al. Two-stage arthroplasty for prosthetic joint infection: a systematic review of acute kidney injury, systemic toxicity and infection control. J Arthroplasty 2013; 28(9):1490–1498.e1. doi:10.1016/j.arth.2013.02.035
  84. Filippone EJ, Kraft WK, Farber JL. The nephrotoxicity of vancomycin. Clin Pharmacol Ther 2017; 102(3):459–469. doi:10.1002/cpt.726
Article PDF
Author and Disclosure Information

Edward J. Filippone, MD, FASN
Clinical Associate Professor of Medicine, Department of Medicine, Division of Nephrology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA

Anju Yadav, MD
Assistant Professor, Department of Medicine, Division of Nephrology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA

Address: Edward J. Filippone, MD, FASN, Department of Medicine, Division of Nephrology, Sidney Kimmel Medical College, Thomas Jefferson University, 2228 South Broad Street, Philadelphia, PA 19145; kidneys@comcast.net

Issue
Cleveland Clinic Journal of Medicine - 86(4)
Publications
Topics
Page Number
263-276
Legacy Keywords
acute kidney injury, AKI, total joint arthroplasty, TJA, hip replacement, knee replacement, antibiotic, aminoglycoside, cement, prosthetic joint infections, antibiotic-loaded cement, gentamicin, tobramycin, vancomycin, Edward Filippone, Anju Yadav
Sections
Author and Disclosure Information

Edward J. Filippone, MD, FASN
Clinical Associate Professor of Medicine, Department of Medicine, Division of Nephrology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA

Anju Yadav, MD
Assistant Professor, Department of Medicine, Division of Nephrology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA

Address: Edward J. Filippone, MD, FASN, Department of Medicine, Division of Nephrology, Sidney Kimmel Medical College, Thomas Jefferson University, 2228 South Broad Street, Philadelphia, PA 19145; kidneys@comcast.net

Author and Disclosure Information

Edward J. Filippone, MD, FASN
Clinical Associate Professor of Medicine, Department of Medicine, Division of Nephrology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA

Anju Yadav, MD
Assistant Professor, Department of Medicine, Division of Nephrology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA

Address: Edward J. Filippone, MD, FASN, Department of Medicine, Division of Nephrology, Sidney Kimmel Medical College, Thomas Jefferson University, 2228 South Broad Street, Philadelphia, PA 19145; kidneys@comcast.net

Article PDF
Article PDF
Related Articles

Total hip or knee replacement (also called total joint arthroplasty) is highly successful at relieving pain and restoring function, but at the risk of acute kidney injury, which is a sudden loss of renal function. Various factors have been associated with this risk, some of which are potentially modifiable, notably, the use of nephrotoxic antibiotics and other drugs.

This review examines the incidence of acute kidney injury using current criteria in total joint arthroplasty of the hip or knee in general, and in the setting of revision surgery for prosthetic joint infection in particular, in which the risk is higher. We identify risk factors for acute kidney injury and propose ways to lower the risk.

MILLIONS OF PROCEDURES ANNUALLY

Total replacement of the hip1,2 or knee3 is being done more and more. Kurtz et al4 estimate that by the year 2030, we will see approximately 3.5 million primary total knee and 500,000 primary total hip replacements every year. In addition, revision total knee procedures are expected to exceed 250,000 per year, and revision total hip procedures are expected to exceed 90,000 per year.4

Chronic infection may complicate up to 2% of these procedures and is associated with significant morbidity, death, and financial costs. Currently, it may be the reason for 25% of total joint arthroplasty revisions,5 but by the year 2030, it is projected to account for 66% of revision total knee arthroplasties and 48% of revision total hip arthroplasties.6

PRIMARY TOTAL JOINT ARTHROPLASTY AND ACUTE KIDNEY INJURY

filippone_acutekidneyinjuryafterarthroplasty_t1.jpg
We searched Ovid MEDLINE for articles on acute kidney injury and either arthroplasty or antibiotic-loaded cement spacers. We found 22 studies, with a total of 72,850 patients, that assessed the incidence of acute kidney injury after primary or revision total joint arthroplasty of the hip or knee, or both, using current criteria7–28 (Table 1), and 3 additional studies that used discharge diagnosis coding.29–31

Study designs, findings varied widely

The incidence of acute kidney injury varied markedly among the studies of primary total joint arthroplasty or revision for aseptic reasons. Numerous factors explain this heterogeneity.

Designs ranged from single-center studies with relatively small numbers of patients to large regional and national samples based on administrative data.

filippone_acutekidneyinjuryafterarthroplasty_t2.jpg
The definition of acute kidney injury also varied, although many used current criteria, specifically the RIFLE (risk, injury, failure, loss, end-stage renal disease),32 AKIN (Acute Kidney Injury Network),33 and KDIGO (Kidney Disease Improving Global Outcomes)34 creatinine criteria (Table 2). Some studies considered only higher stages of acute kidney injury (equivalent to KDIGO stage 2 or 3), ignoring the most common stage, ie, stage 1. No study considered urine output criteria.

Almost all of the studies were retrospective. We are not aware of any randomized controlled trials.

Discharge diagnosis may miss many cases

Several studies based the diagnosis of acute kidney injury on International Classification of Diseases, Ninth Revision (ICD-9) coding from hospital discharge summaries.

Nadkarni et al,29 in the largest study published to date, used the nationwide inpatient sample database of more than 7 million total joint arthroplasties and found an incidence of acute kidney injury based on ICD-9 coding of 1.3% over the years 2002 to 2012, although this increased to 1.8% to 1.9% from 2010 to 2012.

Lopez-de-Andres et al,30 in a similar study using the Spanish national hospital discharge database, evaluated 20,188 patients who underwent revision total hip or knee arthroplasty and found an overall incidence of acute kidney injury of 0.94%, also using ICD-9 coding.

Gharaibeh et al31 used similar methods to diagnose acute kidney injury in a single-center study of 8,949 patients and found an incidence of 1.1%.

Although these 3 studies suggest that the incidence of acute kidney injury is relatively low, Grams et al35 found the sensitivity of ICD-9 coding from hospital records for the diagnosis of acute kidney injury to be only 11.7% compared with KDIGO serum creatinine and urine output criteria. This suggests that the true incidence in these studies may be many times higher, possibly near 10%.

Do all stages of kidney injury count?

Jafari et al,7 in a large series from a single medical center, used only the “I” (injury) and “F” (failure) levels of the RIFLE criteria (corresponding to stages 2 and 3 of the KDIGO criteria) and found an incidence of 0.55% in more than 17,000 total joint arthroplasties.

Jamsa et al8 used the same criteria for acute kidney injury (only “I” and “F”) and found 58 cases in 5,609 patients in whom postoperative serum creatinine was measured, for an incidence of 1%; the remaining 14,966 patients in their cohort did not have serum creatinine measured, and it was assumed they did not have acute kidney injury. Neither of these studies included the most common “R” (risk) stage of acute kidney injury.

Parr et al36 recently studied a nationwide sample of 657,840 hospitalized veterans and found that of 90,614 who developed acute kidney injury based on KDIGO creatinine criteria, 84% reached only stage R. This suggests that if all stages were considered, the true incidence of acute kidney injury would have been higher—possibly 4% in the Jafari series and possibly 7% in the Jamsa series.

Smaller studies had higher rates

Smaller, single-center series reported much higher incidences of acute kidney injury.

Kimmel et al11 found an incidence of 14.8% in 425 total joint arthroplasties using RIFLE creatinine criteria.

Johansson et al25 found an incidence of 19.9% in 136 total joint arthroplasties using KDIGO creatinine criteria.

Sehgal et al9 found an incidence of 21.9% in 659 total joint arthroplasties using AKIN creatinine criteria.

Challagundla et al24 found an incidence of 23.7% in 198 procedures using RIFLE creatinine criteria.

Weingarten et al,10 in a single-center series of 7,463 total joint arthroplasties, found an incidence of acute kidney injury of only 2.2% using AKIN criteria, although 12% of the patients with acute kidney injury did not return to their baseline serum creatinine levels by 3 months.

Our estimate: Nearly 10%

In total, in the 20 studies in Table 1 that included all stages of acute kidney injury, there were 1,909 cases of acute kidney injury in 34,337 patients, for an incidence of 5.6%. Considering that all studies but one were retrospective and none considered urine output criteria for acute kidney injury, we believe that using current KDIGO criteria, the true incidence of acute kidney injury complicating primary lower-extremity total joint arthroplasties is really closer to 10%.

 

 

RISK FACTORS FOR ACUTE KIDNEY INJURY

Various factors have been associated with development of acute kidney injury by multivariate analysis in these studies. Some are modifiable, while others are not, at least in the short term.

Nonmodifiable risk factors

Older age is often significant in studies assessing primary total joint arthroplasty or revision total joint arthroplasty not specifically for infection.11,12,16,17,26,28

Obesity is also a major factor in the development of acute kidney injury,7,10–12,17,18 and, along with age, is a major factor contributing to the need for joint replacement in the first place.

Male sex may increase risk.29

Diabetes mellitus was identified as a risk factor in several studies,10,12,17,20 and hypertension in a few.7,10,24

Other comorbidities and factors such as cardiovascular disease,7,10 liver disease,7 pulmonary disease,7 high American Society of Anesthesiology score,8,19 and benign heart murmurs preoperatively by routine physical examination have also been linked to acute kidney injury after joint arthroplasty.28

Chronic kidney disease as a risk factor

Chronic kidney disease at baseline was associated with acute kidney injury in several of these series.7,11–13,15,19,29

Warth et al12 studied 1,038 patients and found an incidence of acute kidney injury of 11% in the 135 with chronic kidney disease (defined as serum creatinine > 1.2 mg/dL) and who received acetaminophen or narcotics for pain control, compared with 4.8% in the remaining 903 patients without chronic kidney disease, who received ketorolac or celecoxib.

Perregaard et al13 studied 3,410 patients who underwent total hip arthroplasty and found an incidence of acute kidney injury (per KDIGO creatinine criteria) of 2.2% overall, but 7% in the 134 patients with chronic kidney disease based on KDIGO creatinine criteria.

Nowicka et al15 found an incidence of acute kidney injury of 16.7% in the 48 patients with chronic kidney disease (defined as a glomerular filtration rate estimated by the Cockroft-Gault formula of less than 60 mL/min/1.73 m2), compared with 4.5% in the remaining 289.

Modifiable risk factors

Modifiable risk factors that should be considered in high-risk cases include anemia, perioperative blood transfusion, perioperative use of renin-angiotensin-aldosterone system inhibitors such as angiotensin-converting enzyme (ACE) inhibitors and angiotensin II receptor blockers (ARBs), particular antibiotics used for prophylaxis, and nonsteroidal anti-inflammatory drugs used postoperatively.

Anemia and blood transfusion

Preoperative anemia has been associated with postoperative acute kidney injury in various surgical settings such as cardiac surgery.37,38 Perioperative red blood cell transfusions have also been associated with acute kidney injury in cardiac surgery; similar results may apply to total joint arthroplasty.

Choi et al,17 in 2,467 patients undergoing hip replacement, found a significant risk for acute kidney injury if postoperative hemoglobin was consistently below 10 g/dL compared with consistently above this level, with an inverse probability-of-treatment weighted odds ratio of 1.817 (P = .011).

Others have found a significant association of perioperative blood transfusion with acute kidney injury in total joint arthroplasty.10,29

Nadkarni et al,29 for example, used the nationwide inpatient sample database and found by multivariate analysis that perioperative blood transfusion was strongly associated with acute kidney injury, with an adjusted odds ratio of 2.28 (95% confidence interval [CI] 2.15–2.42, P < .0001).

Comment. A higher incidence of acute kidney injury may represent confounding by indication bias, as sicker patients or complicated surgeries may require transfusion, and this risk may not be completely accounted for by multivariate analysis. It is also possible, however, that transfusions per se may contribute to acute kidney injury. Possible direct or indirect mechanisms mediating acute kidney injury include hemolytic reactions, circulatory overload, acute lung injury, and immunomodulatory effects.39

Preoperative transfusion in anemic patients undergoing cardiac surgery may also reduce the incidence of postoperative acute kidney injury both by correcting the anemia and by limiting the need for perioperative transfusions.40 It remains to be determined whether elective preoperative transfusion to correct anemia would reduce postoperative development of acute kidney injury in total joint arthroplasty. As an aside, perioperative transfusion has also been linked to development of periprosthetic joint infection.41

Renin-angiotensin-aldosterone system inhibitors

Several studies found perioperative use of renin-angiotensin-aldosterone system inhibitors to be a risk factor for acute kidney injury.

Kimmel et al11 reported adjusted odds ratios of 2.70 (95% CI 1.12–6.48) for ACE inhibitor use and 2.64 (95% CI 1.18–5.93) for ARB use in a study of 425 primary total joint arthroplasties.

Challagundla et al24 found an odds ratio of 3.07 (95% CI 1.40–6.74) with ACE inhibitor or ARB use by multivariate analysis in 198 total joint arthroplasties.

Nielson et al18 studied 798 patients who underwent total joint arthroplasty and found that preoperative use of renin-angiotensin system inhibitors was associated with a significantly higher rate of postoperative acute kidney injury (8.3% vs 1.7% without inhibition), which was statistically significant by multivariate analysis (odds ratio 2.6, 95% CI 1.04–6.51).

We recommend holding renin-angiotensin-aldosterone system inhibitors 7 days before surgery through the postoperative period in high-risk cases.

Aminoglycoside use as a risk factor

Prophylactic administration of systemic antibiotics is the standard of care. In a systematic review of 26 studies and meta-analysis of 7 studies (3,065 patients), prophylactic antibiotics reduced the relative risk of wound infection by 81% with an absolute risk reduction of 8%.42

A modifiable risk factor for acute kidney injury is the specific antibiotic used for prophylaxis. Multiple studies assessed the risk of acute kidney injury comparing regimens containing an aminoglycoside (typically gentamicin) with regimens lacking these agents.20–26 In general, these studies found a significantly higher risk of acute kidney injury when gentamicin was used.

Challagundla et al24 found an incidence of acute kidney injury of 52% using RIFLE creatinine criteria in 52 patients receiving 8 g total of flucloxacillin plus 160 mg of gentamicin (120 mg if they weighed less than 60 kg) compared with 8% in 48 patients given cefuroxime (3 g total) and 14% in an additional 52 patients also given cefuroxime.

Johansson et al25 found an incidence of KDIGO creatinine-based acute kidney injury of 13% in 70 patients given dicloxacillin alone prophylactically compared with 27% given dicloxacillin and gentamicin, with a relative risk of 3.

Bell et al,21 in a large registry-based analysis from Scotland involving 7,666 elective orthopedic procedures, found that use of flucloxacillin 2 g plus a single dose of gentamicin 4 mg/kg was significantly associated with a 94% higher risk of acute kidney injury (KDIGO creatinine criteria) compared with a cefuroxime-based regimen, with absolute rates increasing from 6.2% to 10.8%.

Dubrovskaya et al20 and Ferguson et al,26 in contrast, found no increased risk with addition of gentamicin.

We recommend avoiding aminoglycosides for prophylaxis in primary lower-extremity total joint arthroplasty in patients at higher risk unless required for specific microbiologic reasons.

Vancomycin may also increase risk

Courtney et al19 assessed the risk of adding vancomycin to cefazolin for routine prophylaxis in a retrospective series of 1,828 total hip or knee arthroplasties and found a significantly higher rate of acute kidney injury, using AKIN criteria (13% vs 8%, odds ratio by multivariate analysis 1.82, P = .002).19

Other agents shown to be effective in treating periprosthetic joint infections or complicated skin and soft-tissue infections with resistant organisms include daptomycin43 and linezolid.44 These nonnephrotoxic alternatives to vancomycin may be a consideration if prophylaxis for methicillin-resistant Staphylococcus aureus is deemed necessary in patients at risk for acute kidney injury.

 

 

PROSTHETIC JOINT INFECTIONS AND ANTIBIOTIC-LOADED CEMENT

Deep infection may complicate nearly 1% of total hip45 and 2% of total knee arthroplasties.46 Kurtz et al4,6 have projected that by 2030, infection will be the cause of two-thirds of the estimated 268,000 revision total knee arthroplasties and about half of the estimated 96,700 revision total hip arthroplasties.

The most common method of treating a chronically infected replacement joint is a 2-stage procedure.5 First, the prosthesis is removed, all infected bone and soft tissue is debrided, and an antibiotic-loaded cement spacer is implanted. Systemic antibiotics are given concurrently, typically for about 6 weeks. After the infection is brought under control, perhaps 2 to 3 months later, the spacer is removed and a new joint is implanted with antibiotic-loaded cement. A 1-stage procedure may be an option in selected cases and would obviate the need for an antibiotic-loaded cement spacer.47,48

Of obvious relevance to development of acute kidney injury is the choice and amount of antibiotics embedded in the cement used for spacers and in implantation. Very high antibiotic levels are achieved within the joint space, usually with little systemic absorption, although significant systemic exposure has been documented in some cases.

The polymethylmethacrylate cement used for these purposes comes in 40-g bags. Multiple bags are typically required per joint, perhaps 2 to 4.49

The rate of elution of antibiotics is determined by several factors, including surface area, porosity, and the number of antibiotics. In general, elution is greatest early on, with exponential decline lasting perhaps 1 week,  followed by slow, sustained release over weeks to months.50 However, several in vitro studies have indicated that only about 5%50,51 of the total antibiotic actually elutes over time.

Initially, multiple antibiotic-laden cement beads were used to fill the joint space, but this significantly limited function and mobility.52 Now, cement spacers are used, and they can be nonarticulating or articulating for maximal joint mobility.53 Although much greater antibiotic elution occurs from beads due to their high surface area-to-volume ratio, spacers still provide an adequate dose.

ANTIBIOTIC-LOADED CEMENT: DOSAGE AND ELUTION CHARACTERISTICS

Antibiotic-loaded cement can be either low-dose or high-dose.

Low-dose cement

Low-dose cement typically consists of 0.5 to 1.0 g of antibiotic per 40-g bag of cement, usually an aminoglycoside (gentamicin or tobramycin) or vancomycin, and can be purchased premixed by the manufacturer. Such cement is only used prophylactically with primary total joint arthroplasty or revision for aseptic reasons, a practice common in Europe but less so in the United States. Some American authors propose antibiotic-loaded cement prophylaxis for patients at high risk, eg, those with immunosuppression, inflammatory cause of arthritis, or diabetes.54

Vrabec et al,55 in a study of low-dose tobramycin-loaded cement used for primary total knee arthroplasty, found a peak median intra-articular tobramycin concentration of 32 mg/L at 6 hours, declining to 6 mg/L at 48 hours with all serum levels 0.3 mg/L or less (unmeasureable) at similar time points.

Sterling et al,56 studying primary total hip arthroplasties with low-dose tobramycin-loaded cement, found mean levels in drainage fluid of 103 mg/L at 6 hours, declining to 15 mg/L at 48 hours. Serum levels peaked at 0.94 mg/L at 3 hours, declining to 0.2 mg/L by 48 hours.

Although most of the antibiotic elution occurs early (within the first week), antibiotic can be found in joint aspirates up to 20 years later.57 We are unaware of any well-documented cases of acute kidney injury ascribable to low-dose antibiotic-loaded cement used prophylactically. One case report making this assertion did not determine serum levels of aminoglycoside.58

High-dose cement

High-dose antibiotic-loaded cement typically contains about 4 to 8 g of antibiotic per 40-g bag of cement and is used in the treatment of prosthetic joint infection to form the spacers. The antibiotic must be mixed into the cement powder by the surgeon in the operating room.

There is no standard combination or dosage. The choice of antibiotic can be tailored to the infecting organism if known. Otherwise, gram-positive organisms are most common, and vancomycin and aminoglycosides are often used together. This particular combination will enhance the elution of both antibiotics when studied in vitro, a process termed “passive opportunism.”59 Other antibiotics in use include aztreonam, piperacillin, teicoplanin, fluoroquinolones, cephalosporins, and daptomycin, among others.

About 8 g of antibiotic total per 40-g bag is the maximum to allow easy molding.52 As an example, this may include 4 g of vancomycin and 3.6 g of tobramycin per 40 g. Given that 3 to 4 such bags are often used per joint, there is significant risk of systemic exposure.

Kalil et al60 studied 8 patients who received high-dose tobramycin-loaded cement to treat periprosthetic joint infections of the hip or knee and found that 7 had detectable serum levels (mean 0.84 mg/L, highest 2.0 mg/L), including 1 with a level of 0.9 mg/L on day 38; 4 of these 8 developed acute kidney injury by AKIN criteria, although other risk factors for acute kidney injury existed. Nearly all had concomitant vancomycin (3 to 8 g) added to the cement as well.

Hsieh et al61 studied 46 patients with infected total hip arthroplasties treated with high-dose antibiotic-loaded cement spacers (vancomycin 4 g and aztreonam 4 g per 40-g bag) and found vancomycin levels in joint drainage higher than 1,500 mg/L on day 1, decreasing to 571 mg/L on day 7; serum levels were low (range 0.1–1.6 mg/L at 24 hours), falling to undetectable by 72 hours.

 

 

ANTIBIOTIC-LOADED CEMENT SPACERS AND ACUTE KIDNEY INJURY

Case reports have associated high-dose antibiotic-loaded cement spacers with acute kidney injury.

Curtis et al62 described an 85-year-old patient with stage 3 chronic kidney disease who was treated for an infected total knee arthroplasty with an antibiotic-loaded cement spacer (containing 3.6 g of tobramycin and 3 g of cefazolin per 40-g bag, 3 bags total) and developed stage 3 acute kidney injury. After 16 days and 3 hemodialysis sessions, the patient’s serum tobramycin level was still 2 mg/L despite receiving no systemic tobramycin.

Wu et al63 reported a case of acute kidney injury that required dialysis after implantation of a tobramycin- and vancomycin-loaded spacer, with persistent serum tobramycin levels despite repeated hemodialysis sessions until the spacer was removed.

Chalmers et al64 described 2 patients with acute kidney injury and persistently elevated serum tobramycin levels (3.9 mg/L on day 39 in 1 patient and 2.0 mg/L on day 24 in the other patient) despite no systemic administration.

In these and other case reports,65–67 dialysis and spacer explantation were usually required. 


Comment. It is intuitive that acute kidney injury would more likely complicate revision total joint arthroplasties for infection than for primary total joint arthroplasties or revisions for aseptic reasons, given the systemic effects of infection and exposure to nephrotoxic or allergenic antibiotics. And the available data suggest that the risk of acute kidney injury is higher with revision for prosthetic joint infection than with revision for aseptic reasons. However, many of the studies were retrospective, relatively small, single-center series and used different definitions of acute kidney injury.

filippone_acutekidneyinjuryafterarthroplasty_t3.jpg
We are aware of 17 studies specifically addressing acute kidney injury or postoperative complications in general that may have included acute kidney injury.50,52,61,68–81 Ten of these studies found at least 1 case of acute kidney injury (Table 3). Of note, 7 studies totaling 219 patients reported no cases of acute kidney injury, although acute kidney injury per se was not mentioned and no definition of it was provided.50,61,76,77,79,80,82

Luu et al83 performed a systematic review of studies published between January 1989 and June 2012 reporting systemic complications (including acute kidney injury) of 2-stage revision arthroplasties including placement of an antibiotic-loaded cement spacer for treatment of periprosthetic joint infection. Overall, 10 studies were identified with 544 total patients. Five of these studies, with 409 patients, reported at least 1 case of acute kidney injury for a total of 27 patients, giving an incidence of 6.6% in these studies.68–71 The remaining 5 studies, totaling 135 patients, did not report any cases of acute kidney injury,50,61,76–78 although that was not the primary focus of any of those trials.

Most notable from this systematic review, the study of Menge et al69 retrospectively determined the incidence of acute kidney injury (defined as a 50% rise in serum creatinine to > 1.4 mg/dL within 90 days of surgery) to be 17% in 84 patients with infected total knee arthroplasties treated with antibiotic-loaded cement spacers. A mean of 3.5 bags of cement per spacer were used in the 35 articulating spacers, compared with 2.9 per nonarticulating spacer. These spacers contained vancomycin in 82% (median 4.0 g, range 1–16 g) and tobramycin in 94% (median 4.8 g, range 1–12 g), among others in small percentages. The dose of tobramycin in the spacer considered either as a dichotomous variable (> 4.8 g, OR 5.87) or linearly (OR 1.24 per 1-g increase) was significantly associated with acute kidney injury, although systemic administration of aminoglycosides or vancomycin was not.

Additional single-center series that were published subsequent to this review have generally used more current diagnostic criteria.

Noto et al72 found that 10 of 46 patients treated with antibiotic-loaded cement spacers had a greater than 50% rise in serum creatinine (average increase 260%). All spacers contained tobramycin (mean dose 8.2 g), and 9 of 10 also contained vancomycin (mean 7.6 g). All of the 9 patients with acute kidney injury with follow-up data recovered renal function.

Reed et al75 found 26 cases of acute kidney injury (based on RIFLE creatinine criteria) in 306 patients with antibiotic-loaded cement spacers treating various periprosthetic joint infections (including hips, knees, shoulders, and digits) and compared them with 74 controls who did not develop acute kidney injury. By multivariable analysis, receipt of an ACE inhibitor within 7 days of surgery and receipt of piperacillin-tazobactam within 7 days after surgery were both significantly more common in cases with acute kidney injury than in controls without acute kidney injury.

Aeng et al73 prospectively studied 50 consecutive patients receiving antibiotic-loaded spacers containing tobramycin (with or without vancomycin) for treatment of infected hip or knee replacements. Using RIFLE creatinine criteria, they found an incidence of acute kidney injury of 20% (10 of 50). Factors significantly associated with acute kidney injury included cement premixed by the manufacturer with gentamicin (0.5 g per 40-g bag) in addition to the tobramycin they added, intraoperative blood transfusions, and postoperative use of nonsteroidal anti-inflammatory drugs.

Geller et al,74 in a multicenter retrospective study of 247 patients with prosthetic joint infections (156 knees and 91 hips) undergoing antibiotic-loaded cement spacer placement, found an incidence of acute kidney injury of 26% based on KDIGO creatinine criteria. Significant risk factors included higher body mass index, lower preoperative hemoglobin level, drop in hemoglobin after surgery, and comorbidity (hypertension, diabetes, chronic kidney disease, or cardiovascular disease). Most of the spacers contained a combination of vancomycin and either tobramycin (81%) or gentamicin (13%). The spacers contained an average of 5.3 g (range 0.6–18 g) of vancomycin (average 2.65 g per 40-g bag) and an average of 5.2 g (range 0.5–16.4 g) of tobramycin (average 2.6 g per bag).

As in Menge et al,69 this study illustrates the wide range of antibiotic dosages in use and the lack of standardization. In contrast to the study by Menge et al, however, development of acute kidney injury was not related to the amount of vancomycin or tobramycin contained in the spacers. Eventual clearance of infection (at 1 and 2 years) was significantly related to increasing amounts of vancomycin. Multiple different systemic antibiotics were used, most commonly vancomycin (44%), and systemic vancomycin was not associated with acute kidney injury.

Yadav et al,81 in a study of 3,129 consecutive revision procedures of the knee or hip, found an incidence of acute kidney injury by RIFLE creatinine criteria of 29% in the 197 patients who received antibiotic-loaded cement spacers for periprosthetic joint infection compared with 3.4% in the 2,848 who underwent revision for aseptic reasons. In 84 patients with prosthetic joint infection having various surgeries not including placement of a spacer, the acute kidney injury rate at some point in their course was an alarmingly high 82%. In the group that received spacers, only age and comorbidity as assessed by Charlson comorbidity index were independently associated with acute kidney injury by multivariate analysis. Surprisingly, modest renal impairment was protective, possibly because physicians of patients with chronic kidney disease were more vigilant and took appropriate measures to prevent acute kidney injury.

Overall, the risk of acute kidney injury appears to be much higher during treatment of prosthetic joint infection with a 2-stage procedure using an antibiotic-loaded cement spacer than after primary total joint arthroplasty or revision for aseptic reasons, and may complicate up to one-third of cases.

 

 

REDUCING RISK DURING TREATMENT OF INFECTED REPLACEMENT JOINTS

filippone_acutekidneyinjuryafterarthroplasty_t4.jpg
Due to lack of appropriate data, how best to mitigate the risk of acute kidney injury is uncertain. In our opinion, however, the following measures should be considered (Table 4).

As in primary total joint arthroplasty in general, higher-risk cases should be identified based on age, body mass index, chronic kidney disease, comorbidities (hypertension, diabetes, established cardiovascular disease), and anemia.

Preoperative transfusion can be considered case by case depending on degree of anemia and associated risk factors.

All renin-angiotensin-aldosterone system inhibitors should be withheld starting 1 week before surgery.

Both nonselective and cyclooxygenase-2 selective nonsteroidal anti-inflammatory drugs should be avoided, if possible.

Strict attention should be paid to adequate intraoperative and postoperative fluid resuscitation.

Kidney function should be monitored closely in the early postoperative period, including urine output and daily creatinine for at least 72 hours.

Systemic administration of potentially nephrotoxic antibiotics should be minimized, especially the combination of vancomycin with piperacillin-tazobactam.84 Daptomycin is a consideration.43

If acute kidney injury should develop, serum levels of vancomycin or aminoglycosides should be measured if the spacer contains these antibiotics. The spacer may need to be removed if toxic serum levels persist.

TAKE-HOME POINTS

Acute kidney injury may complicate up to 10% of primary lower-extremity total joint arthroplasties and up to 25% of periprosthetic joint infections treated with a 2-stage procedure including placement of an antibiotic-loaded cement spacer in the first stage.

Risk factors for acute kidney injury include older age, obesity, chronic kidney disease, and overall comorbidity. Potentially modifiable risk factors include anemia, perioperative transfusions, aminoglycoside prophylaxis, perioperative renin-angiotensin system blockade, and postoperative nonsteroidal anti-inflammatory drugs. These should be mitigated when possible.

In patients with periprosthetic joint infection who receive antibiotic-loaded cement spacers, especially patients  with additional risk factors for acute kidney injury, strict attention should be paid to the dose of antibiotic in the spacer, with levels checked postoperatively if necessary. Nonnephrotoxic antibiotics should be chosen for systemic administration when possible.

Prospective randomized controlled trials are needed to guide therapy after total joint arthroplasty, and to verify the adverse long-term outcomes of acute kidney injury in this setting.

Total hip or knee replacement (also called total joint arthroplasty) is highly successful at relieving pain and restoring function, but at the risk of acute kidney injury, which is a sudden loss of renal function. Various factors have been associated with this risk, some of which are potentially modifiable, notably, the use of nephrotoxic antibiotics and other drugs.

This review examines the incidence of acute kidney injury using current criteria in total joint arthroplasty of the hip or knee in general, and in the setting of revision surgery for prosthetic joint infection in particular, in which the risk is higher. We identify risk factors for acute kidney injury and propose ways to lower the risk.

MILLIONS OF PROCEDURES ANNUALLY

Total replacement of the hip1,2 or knee3 is being done more and more. Kurtz et al4 estimate that by the year 2030, we will see approximately 3.5 million primary total knee and 500,000 primary total hip replacements every year. In addition, revision total knee procedures are expected to exceed 250,000 per year, and revision total hip procedures are expected to exceed 90,000 per year.4

Chronic infection may complicate up to 2% of these procedures and is associated with significant morbidity, death, and financial costs. Currently, it may be the reason for 25% of total joint arthroplasty revisions,5 but by the year 2030, it is projected to account for 66% of revision total knee arthroplasties and 48% of revision total hip arthroplasties.6

PRIMARY TOTAL JOINT ARTHROPLASTY AND ACUTE KIDNEY INJURY

filippone_acutekidneyinjuryafterarthroplasty_t1.jpg
We searched Ovid MEDLINE for articles on acute kidney injury and either arthroplasty or antibiotic-loaded cement spacers. We found 22 studies, with a total of 72,850 patients, that assessed the incidence of acute kidney injury after primary or revision total joint arthroplasty of the hip or knee, or both, using current criteria7–28 (Table 1), and 3 additional studies that used discharge diagnosis coding.29–31

Study designs, findings varied widely

The incidence of acute kidney injury varied markedly among the studies of primary total joint arthroplasty or revision for aseptic reasons. Numerous factors explain this heterogeneity.

Designs ranged from single-center studies with relatively small numbers of patients to large regional and national samples based on administrative data.

filippone_acutekidneyinjuryafterarthroplasty_t2.jpg
The definition of acute kidney injury also varied, although many used current criteria, specifically the RIFLE (risk, injury, failure, loss, end-stage renal disease),32 AKIN (Acute Kidney Injury Network),33 and KDIGO (Kidney Disease Improving Global Outcomes)34 creatinine criteria (Table 2). Some studies considered only higher stages of acute kidney injury (equivalent to KDIGO stage 2 or 3), ignoring the most common stage, ie, stage 1. No study considered urine output criteria.

Almost all of the studies were retrospective. We are not aware of any randomized controlled trials.

Discharge diagnosis may miss many cases

Several studies based the diagnosis of acute kidney injury on International Classification of Diseases, Ninth Revision (ICD-9) coding from hospital discharge summaries.

Nadkarni et al,29 in the largest study published to date, used the nationwide inpatient sample database of more than 7 million total joint arthroplasties and found an incidence of acute kidney injury based on ICD-9 coding of 1.3% over the years 2002 to 2012, although this increased to 1.8% to 1.9% from 2010 to 2012.

Lopez-de-Andres et al,30 in a similar study using the Spanish national hospital discharge database, evaluated 20,188 patients who underwent revision total hip or knee arthroplasty and found an overall incidence of acute kidney injury of 0.94%, also using ICD-9 coding.

Gharaibeh et al31 used similar methods to diagnose acute kidney injury in a single-center study of 8,949 patients and found an incidence of 1.1%.

Although these 3 studies suggest that the incidence of acute kidney injury is relatively low, Grams et al35 found the sensitivity of ICD-9 coding from hospital records for the diagnosis of acute kidney injury to be only 11.7% compared with KDIGO serum creatinine and urine output criteria. This suggests that the true incidence in these studies may be many times higher, possibly near 10%.

Do all stages of kidney injury count?

Jafari et al,7 in a large series from a single medical center, used only the “I” (injury) and “F” (failure) levels of the RIFLE criteria (corresponding to stages 2 and 3 of the KDIGO criteria) and found an incidence of 0.55% in more than 17,000 total joint arthroplasties.

Jamsa et al8 used the same criteria for acute kidney injury (only “I” and “F”) and found 58 cases in 5,609 patients in whom postoperative serum creatinine was measured, for an incidence of 1%; the remaining 14,966 patients in their cohort did not have serum creatinine measured, and it was assumed they did not have acute kidney injury. Neither of these studies included the most common “R” (risk) stage of acute kidney injury.

Parr et al36 recently studied a nationwide sample of 657,840 hospitalized veterans and found that of 90,614 who developed acute kidney injury based on KDIGO creatinine criteria, 84% reached only stage R. This suggests that if all stages were considered, the true incidence of acute kidney injury would have been higher—possibly 4% in the Jafari series and possibly 7% in the Jamsa series.

Smaller studies had higher rates

Smaller, single-center series reported much higher incidences of acute kidney injury.

Kimmel et al11 found an incidence of 14.8% in 425 total joint arthroplasties using RIFLE creatinine criteria.

Johansson et al25 found an incidence of 19.9% in 136 total joint arthroplasties using KDIGO creatinine criteria.

Sehgal et al9 found an incidence of 21.9% in 659 total joint arthroplasties using AKIN creatinine criteria.

Challagundla et al24 found an incidence of 23.7% in 198 procedures using RIFLE creatinine criteria.

Weingarten et al,10 in a single-center series of 7,463 total joint arthroplasties, found an incidence of acute kidney injury of only 2.2% using AKIN criteria, although 12% of the patients with acute kidney injury did not return to their baseline serum creatinine levels by 3 months.

Our estimate: Nearly 10%

In total, in the 20 studies in Table 1 that included all stages of acute kidney injury, there were 1,909 cases of acute kidney injury in 34,337 patients, for an incidence of 5.6%. Considering that all studies but one were retrospective and none considered urine output criteria for acute kidney injury, we believe that using current KDIGO criteria, the true incidence of acute kidney injury complicating primary lower-extremity total joint arthroplasties is really closer to 10%.

 

 

RISK FACTORS FOR ACUTE KIDNEY INJURY

Various factors have been associated with development of acute kidney injury by multivariate analysis in these studies. Some are modifiable, while others are not, at least in the short term.

Nonmodifiable risk factors

Older age is often significant in studies assessing primary total joint arthroplasty or revision total joint arthroplasty not specifically for infection.11,12,16,17,26,28

Obesity is also a major factor in the development of acute kidney injury,7,10–12,17,18 and, along with age, is a major factor contributing to the need for joint replacement in the first place.

Male sex may increase risk.29

Diabetes mellitus was identified as a risk factor in several studies,10,12,17,20 and hypertension in a few.7,10,24

Other comorbidities and factors such as cardiovascular disease,7,10 liver disease,7 pulmonary disease,7 high American Society of Anesthesiology score,8,19 and benign heart murmurs preoperatively by routine physical examination have also been linked to acute kidney injury after joint arthroplasty.28

Chronic kidney disease as a risk factor

Chronic kidney disease at baseline was associated with acute kidney injury in several of these series.7,11–13,15,19,29

Warth et al12 studied 1,038 patients and found an incidence of acute kidney injury of 11% in the 135 with chronic kidney disease (defined as serum creatinine > 1.2 mg/dL) and who received acetaminophen or narcotics for pain control, compared with 4.8% in the remaining 903 patients without chronic kidney disease, who received ketorolac or celecoxib.

Perregaard et al13 studied 3,410 patients who underwent total hip arthroplasty and found an incidence of acute kidney injury (per KDIGO creatinine criteria) of 2.2% overall, but 7% in the 134 patients with chronic kidney disease based on KDIGO creatinine criteria.

Nowicka et al15 found an incidence of acute kidney injury of 16.7% in the 48 patients with chronic kidney disease (defined as a glomerular filtration rate estimated by the Cockroft-Gault formula of less than 60 mL/min/1.73 m2), compared with 4.5% in the remaining 289.

Modifiable risk factors

Modifiable risk factors that should be considered in high-risk cases include anemia, perioperative blood transfusion, perioperative use of renin-angiotensin-aldosterone system inhibitors such as angiotensin-converting enzyme (ACE) inhibitors and angiotensin II receptor blockers (ARBs), particular antibiotics used for prophylaxis, and nonsteroidal anti-inflammatory drugs used postoperatively.

Anemia and blood transfusion

Preoperative anemia has been associated with postoperative acute kidney injury in various surgical settings such as cardiac surgery.37,38 Perioperative red blood cell transfusions have also been associated with acute kidney injury in cardiac surgery; similar results may apply to total joint arthroplasty.

Choi et al,17 in 2,467 patients undergoing hip replacement, found a significant risk for acute kidney injury if postoperative hemoglobin was consistently below 10 g/dL compared with consistently above this level, with an inverse probability-of-treatment weighted odds ratio of 1.817 (P = .011).

Others have found a significant association of perioperative blood transfusion with acute kidney injury in total joint arthroplasty.10,29

Nadkarni et al,29 for example, used the nationwide inpatient sample database and found by multivariate analysis that perioperative blood transfusion was strongly associated with acute kidney injury, with an adjusted odds ratio of 2.28 (95% confidence interval [CI] 2.15–2.42, P < .0001).

Comment. A higher incidence of acute kidney injury may represent confounding by indication bias, as sicker patients or complicated surgeries may require transfusion, and this risk may not be completely accounted for by multivariate analysis. It is also possible, however, that transfusions per se may contribute to acute kidney injury. Possible direct or indirect mechanisms mediating acute kidney injury include hemolytic reactions, circulatory overload, acute lung injury, and immunomodulatory effects.39

Preoperative transfusion in anemic patients undergoing cardiac surgery may also reduce the incidence of postoperative acute kidney injury both by correcting the anemia and by limiting the need for perioperative transfusions.40 It remains to be determined whether elective preoperative transfusion to correct anemia would reduce postoperative development of acute kidney injury in total joint arthroplasty. As an aside, perioperative transfusion has also been linked to development of periprosthetic joint infection.41

Renin-angiotensin-aldosterone system inhibitors

Several studies found perioperative use of renin-angiotensin-aldosterone system inhibitors to be a risk factor for acute kidney injury.

Kimmel et al11 reported adjusted odds ratios of 2.70 (95% CI 1.12–6.48) for ACE inhibitor use and 2.64 (95% CI 1.18–5.93) for ARB use in a study of 425 primary total joint arthroplasties.

Challagundla et al24 found an odds ratio of 3.07 (95% CI 1.40–6.74) with ACE inhibitor or ARB use by multivariate analysis in 198 total joint arthroplasties.

Nielson et al18 studied 798 patients who underwent total joint arthroplasty and found that preoperative use of renin-angiotensin system inhibitors was associated with a significantly higher rate of postoperative acute kidney injury (8.3% vs 1.7% without inhibition), which was statistically significant by multivariate analysis (odds ratio 2.6, 95% CI 1.04–6.51).

We recommend holding renin-angiotensin-aldosterone system inhibitors 7 days before surgery through the postoperative period in high-risk cases.

Aminoglycoside use as a risk factor

Prophylactic administration of systemic antibiotics is the standard of care. In a systematic review of 26 studies and meta-analysis of 7 studies (3,065 patients), prophylactic antibiotics reduced the relative risk of wound infection by 81% with an absolute risk reduction of 8%.42

A modifiable risk factor for acute kidney injury is the specific antibiotic used for prophylaxis. Multiple studies assessed the risk of acute kidney injury comparing regimens containing an aminoglycoside (typically gentamicin) with regimens lacking these agents.20–26 In general, these studies found a significantly higher risk of acute kidney injury when gentamicin was used.

Challagundla et al24 found an incidence of acute kidney injury of 52% using RIFLE creatinine criteria in 52 patients receiving 8 g total of flucloxacillin plus 160 mg of gentamicin (120 mg if they weighed less than 60 kg) compared with 8% in 48 patients given cefuroxime (3 g total) and 14% in an additional 52 patients also given cefuroxime.

Johansson et al25 found an incidence of KDIGO creatinine-based acute kidney injury of 13% in 70 patients given dicloxacillin alone prophylactically compared with 27% given dicloxacillin and gentamicin, with a relative risk of 3.

Bell et al,21 in a large registry-based analysis from Scotland involving 7,666 elective orthopedic procedures, found that use of flucloxacillin 2 g plus a single dose of gentamicin 4 mg/kg was significantly associated with a 94% higher risk of acute kidney injury (KDIGO creatinine criteria) compared with a cefuroxime-based regimen, with absolute rates increasing from 6.2% to 10.8%.

Dubrovskaya et al20 and Ferguson et al,26 in contrast, found no increased risk with addition of gentamicin.

We recommend avoiding aminoglycosides for prophylaxis in primary lower-extremity total joint arthroplasty in patients at higher risk unless required for specific microbiologic reasons.

Vancomycin may also increase risk

Courtney et al19 assessed the risk of adding vancomycin to cefazolin for routine prophylaxis in a retrospective series of 1,828 total hip or knee arthroplasties and found a significantly higher rate of acute kidney injury, using AKIN criteria (13% vs 8%, odds ratio by multivariate analysis 1.82, P = .002).19

Other agents shown to be effective in treating periprosthetic joint infections or complicated skin and soft-tissue infections with resistant organisms include daptomycin43 and linezolid.44 These nonnephrotoxic alternatives to vancomycin may be a consideration if prophylaxis for methicillin-resistant Staphylococcus aureus is deemed necessary in patients at risk for acute kidney injury.

 

 

PROSTHETIC JOINT INFECTIONS AND ANTIBIOTIC-LOADED CEMENT

Deep infection may complicate nearly 1% of total hip45 and 2% of total knee arthroplasties.46 Kurtz et al4,6 have projected that by 2030, infection will be the cause of two-thirds of the estimated 268,000 revision total knee arthroplasties and about half of the estimated 96,700 revision total hip arthroplasties.

The most common method of treating a chronically infected replacement joint is a 2-stage procedure.5 First, the prosthesis is removed, all infected bone and soft tissue is debrided, and an antibiotic-loaded cement spacer is implanted. Systemic antibiotics are given concurrently, typically for about 6 weeks. After the infection is brought under control, perhaps 2 to 3 months later, the spacer is removed and a new joint is implanted with antibiotic-loaded cement. A 1-stage procedure may be an option in selected cases and would obviate the need for an antibiotic-loaded cement spacer.47,48

Of obvious relevance to development of acute kidney injury is the choice and amount of antibiotics embedded in the cement used for spacers and in implantation. Very high antibiotic levels are achieved within the joint space, usually with little systemic absorption, although significant systemic exposure has been documented in some cases.

The polymethylmethacrylate cement used for these purposes comes in 40-g bags. Multiple bags are typically required per joint, perhaps 2 to 4.49

The rate of elution of antibiotics is determined by several factors, including surface area, porosity, and the number of antibiotics. In general, elution is greatest early on, with exponential decline lasting perhaps 1 week,  followed by slow, sustained release over weeks to months.50 However, several in vitro studies have indicated that only about 5%50,51 of the total antibiotic actually elutes over time.

Initially, multiple antibiotic-laden cement beads were used to fill the joint space, but this significantly limited function and mobility.52 Now, cement spacers are used, and they can be nonarticulating or articulating for maximal joint mobility.53 Although much greater antibiotic elution occurs from beads due to their high surface area-to-volume ratio, spacers still provide an adequate dose.

ANTIBIOTIC-LOADED CEMENT: DOSAGE AND ELUTION CHARACTERISTICS

Antibiotic-loaded cement can be either low-dose or high-dose.

Low-dose cement

Low-dose cement typically consists of 0.5 to 1.0 g of antibiotic per 40-g bag of cement, usually an aminoglycoside (gentamicin or tobramycin) or vancomycin, and can be purchased premixed by the manufacturer. Such cement is only used prophylactically with primary total joint arthroplasty or revision for aseptic reasons, a practice common in Europe but less so in the United States. Some American authors propose antibiotic-loaded cement prophylaxis for patients at high risk, eg, those with immunosuppression, inflammatory cause of arthritis, or diabetes.54

Vrabec et al,55 in a study of low-dose tobramycin-loaded cement used for primary total knee arthroplasty, found a peak median intra-articular tobramycin concentration of 32 mg/L at 6 hours, declining to 6 mg/L at 48 hours with all serum levels 0.3 mg/L or less (unmeasureable) at similar time points.

Sterling et al,56 studying primary total hip arthroplasties with low-dose tobramycin-loaded cement, found mean levels in drainage fluid of 103 mg/L at 6 hours, declining to 15 mg/L at 48 hours. Serum levels peaked at 0.94 mg/L at 3 hours, declining to 0.2 mg/L by 48 hours.

Although most of the antibiotic elution occurs early (within the first week), antibiotic can be found in joint aspirates up to 20 years later.57 We are unaware of any well-documented cases of acute kidney injury ascribable to low-dose antibiotic-loaded cement used prophylactically. One case report making this assertion did not determine serum levels of aminoglycoside.58

High-dose cement

High-dose antibiotic-loaded cement typically contains about 4 to 8 g of antibiotic per 40-g bag of cement and is used in the treatment of prosthetic joint infection to form the spacers. The antibiotic must be mixed into the cement powder by the surgeon in the operating room.

There is no standard combination or dosage. The choice of antibiotic can be tailored to the infecting organism if known. Otherwise, gram-positive organisms are most common, and vancomycin and aminoglycosides are often used together. This particular combination will enhance the elution of both antibiotics when studied in vitro, a process termed “passive opportunism.”59 Other antibiotics in use include aztreonam, piperacillin, teicoplanin, fluoroquinolones, cephalosporins, and daptomycin, among others.

About 8 g of antibiotic total per 40-g bag is the maximum to allow easy molding.52 As an example, this may include 4 g of vancomycin and 3.6 g of tobramycin per 40 g. Given that 3 to 4 such bags are often used per joint, there is significant risk of systemic exposure.

Kalil et al60 studied 8 patients who received high-dose tobramycin-loaded cement to treat periprosthetic joint infections of the hip or knee and found that 7 had detectable serum levels (mean 0.84 mg/L, highest 2.0 mg/L), including 1 with a level of 0.9 mg/L on day 38; 4 of these 8 developed acute kidney injury by AKIN criteria, although other risk factors for acute kidney injury existed. Nearly all had concomitant vancomycin (3 to 8 g) added to the cement as well.

Hsieh et al61 studied 46 patients with infected total hip arthroplasties treated with high-dose antibiotic-loaded cement spacers (vancomycin 4 g and aztreonam 4 g per 40-g bag) and found vancomycin levels in joint drainage higher than 1,500 mg/L on day 1, decreasing to 571 mg/L on day 7; serum levels were low (range 0.1–1.6 mg/L at 24 hours), falling to undetectable by 72 hours.

 

 

ANTIBIOTIC-LOADED CEMENT SPACERS AND ACUTE KIDNEY INJURY

Case reports have associated high-dose antibiotic-loaded cement spacers with acute kidney injury.

Curtis et al62 described an 85-year-old patient with stage 3 chronic kidney disease who was treated for an infected total knee arthroplasty with an antibiotic-loaded cement spacer (containing 3.6 g of tobramycin and 3 g of cefazolin per 40-g bag, 3 bags total) and developed stage 3 acute kidney injury. After 16 days and 3 hemodialysis sessions, the patient’s serum tobramycin level was still 2 mg/L despite receiving no systemic tobramycin.

Wu et al63 reported a case of acute kidney injury that required dialysis after implantation of a tobramycin- and vancomycin-loaded spacer, with persistent serum tobramycin levels despite repeated hemodialysis sessions until the spacer was removed.

Chalmers et al64 described 2 patients with acute kidney injury and persistently elevated serum tobramycin levels (3.9 mg/L on day 39 in 1 patient and 2.0 mg/L on day 24 in the other patient) despite no systemic administration.

In these and other case reports,65–67 dialysis and spacer explantation were usually required. 


Comment. It is intuitive that acute kidney injury would more likely complicate revision total joint arthroplasties for infection than for primary total joint arthroplasties or revisions for aseptic reasons, given the systemic effects of infection and exposure to nephrotoxic or allergenic antibiotics. And the available data suggest that the risk of acute kidney injury is higher with revision for prosthetic joint infection than with revision for aseptic reasons. However, many of the studies were retrospective, relatively small, single-center series and used different definitions of acute kidney injury.

filippone_acutekidneyinjuryafterarthroplasty_t3.jpg
We are aware of 17 studies specifically addressing acute kidney injury or postoperative complications in general that may have included acute kidney injury.50,52,61,68–81 Ten of these studies found at least 1 case of acute kidney injury (Table 3). Of note, 7 studies totaling 219 patients reported no cases of acute kidney injury, although acute kidney injury per se was not mentioned and no definition of it was provided.50,61,76,77,79,80,82

Luu et al83 performed a systematic review of studies published between January 1989 and June 2012 reporting systemic complications (including acute kidney injury) of 2-stage revision arthroplasties including placement of an antibiotic-loaded cement spacer for treatment of periprosthetic joint infection. Overall, 10 studies were identified with 544 total patients. Five of these studies, with 409 patients, reported at least 1 case of acute kidney injury for a total of 27 patients, giving an incidence of 6.6% in these studies.68–71 The remaining 5 studies, totaling 135 patients, did not report any cases of acute kidney injury,50,61,76–78 although that was not the primary focus of any of those trials.

Most notable from this systematic review, the study of Menge et al69 retrospectively determined the incidence of acute kidney injury (defined as a 50% rise in serum creatinine to > 1.4 mg/dL within 90 days of surgery) to be 17% in 84 patients with infected total knee arthroplasties treated with antibiotic-loaded cement spacers. A mean of 3.5 bags of cement per spacer were used in the 35 articulating spacers, compared with 2.9 per nonarticulating spacer. These spacers contained vancomycin in 82% (median 4.0 g, range 1–16 g) and tobramycin in 94% (median 4.8 g, range 1–12 g), among others in small percentages. The dose of tobramycin in the spacer considered either as a dichotomous variable (> 4.8 g, OR 5.87) or linearly (OR 1.24 per 1-g increase) was significantly associated with acute kidney injury, although systemic administration of aminoglycosides or vancomycin was not.

Additional single-center series that were published subsequent to this review have generally used more current diagnostic criteria.

Noto et al72 found that 10 of 46 patients treated with antibiotic-loaded cement spacers had a greater than 50% rise in serum creatinine (average increase 260%). All spacers contained tobramycin (mean dose 8.2 g), and 9 of 10 also contained vancomycin (mean 7.6 g). All of the 9 patients with acute kidney injury with follow-up data recovered renal function.

Reed et al75 found 26 cases of acute kidney injury (based on RIFLE creatinine criteria) in 306 patients with antibiotic-loaded cement spacers treating various periprosthetic joint infections (including hips, knees, shoulders, and digits) and compared them with 74 controls who did not develop acute kidney injury. By multivariable analysis, receipt of an ACE inhibitor within 7 days of surgery and receipt of piperacillin-tazobactam within 7 days after surgery were both significantly more common in cases with acute kidney injury than in controls without acute kidney injury.

Aeng et al73 prospectively studied 50 consecutive patients receiving antibiotic-loaded spacers containing tobramycin (with or without vancomycin) for treatment of infected hip or knee replacements. Using RIFLE creatinine criteria, they found an incidence of acute kidney injury of 20% (10 of 50). Factors significantly associated with acute kidney injury included cement premixed by the manufacturer with gentamicin (0.5 g per 40-g bag) in addition to the tobramycin they added, intraoperative blood transfusions, and postoperative use of nonsteroidal anti-inflammatory drugs.

Geller et al,74 in a multicenter retrospective study of 247 patients with prosthetic joint infections (156 knees and 91 hips) undergoing antibiotic-loaded cement spacer placement, found an incidence of acute kidney injury of 26% based on KDIGO creatinine criteria. Significant risk factors included higher body mass index, lower preoperative hemoglobin level, drop in hemoglobin after surgery, and comorbidity (hypertension, diabetes, chronic kidney disease, or cardiovascular disease). Most of the spacers contained a combination of vancomycin and either tobramycin (81%) or gentamicin (13%). The spacers contained an average of 5.3 g (range 0.6–18 g) of vancomycin (average 2.65 g per 40-g bag) and an average of 5.2 g (range 0.5–16.4 g) of tobramycin (average 2.6 g per bag).

As in Menge et al,69 this study illustrates the wide range of antibiotic dosages in use and the lack of standardization. In contrast to the study by Menge et al, however, development of acute kidney injury was not related to the amount of vancomycin or tobramycin contained in the spacers. Eventual clearance of infection (at 1 and 2 years) was significantly related to increasing amounts of vancomycin. Multiple different systemic antibiotics were used, most commonly vancomycin (44%), and systemic vancomycin was not associated with acute kidney injury.

Yadav et al,81 in a study of 3,129 consecutive revision procedures of the knee or hip, found an incidence of acute kidney injury by RIFLE creatinine criteria of 29% in the 197 patients who received antibiotic-loaded cement spacers for periprosthetic joint infection compared with 3.4% in the 2,848 who underwent revision for aseptic reasons. In 84 patients with prosthetic joint infection having various surgeries not including placement of a spacer, the acute kidney injury rate at some point in their course was an alarmingly high 82%. In the group that received spacers, only age and comorbidity as assessed by Charlson comorbidity index were independently associated with acute kidney injury by multivariate analysis. Surprisingly, modest renal impairment was protective, possibly because physicians of patients with chronic kidney disease were more vigilant and took appropriate measures to prevent acute kidney injury.

Overall, the risk of acute kidney injury appears to be much higher during treatment of prosthetic joint infection with a 2-stage procedure using an antibiotic-loaded cement spacer than after primary total joint arthroplasty or revision for aseptic reasons, and may complicate up to one-third of cases.

 

 

REDUCING RISK DURING TREATMENT OF INFECTED REPLACEMENT JOINTS

filippone_acutekidneyinjuryafterarthroplasty_t4.jpg
Due to lack of appropriate data, how best to mitigate the risk of acute kidney injury is uncertain. In our opinion, however, the following measures should be considered (Table 4).

As in primary total joint arthroplasty in general, higher-risk cases should be identified based on age, body mass index, chronic kidney disease, comorbidities (hypertension, diabetes, established cardiovascular disease), and anemia.

Preoperative transfusion can be considered case by case depending on degree of anemia and associated risk factors.

All renin-angiotensin-aldosterone system inhibitors should be withheld starting 1 week before surgery.

Both nonselective and cyclooxygenase-2 selective nonsteroidal anti-inflammatory drugs should be avoided, if possible.

Strict attention should be paid to adequate intraoperative and postoperative fluid resuscitation.

Kidney function should be monitored closely in the early postoperative period, including urine output and daily creatinine for at least 72 hours.

Systemic administration of potentially nephrotoxic antibiotics should be minimized, especially the combination of vancomycin with piperacillin-tazobactam.84 Daptomycin is a consideration.43

If acute kidney injury should develop, serum levels of vancomycin or aminoglycosides should be measured if the spacer contains these antibiotics. The spacer may need to be removed if toxic serum levels persist.

TAKE-HOME POINTS

Acute kidney injury may complicate up to 10% of primary lower-extremity total joint arthroplasties and up to 25% of periprosthetic joint infections treated with a 2-stage procedure including placement of an antibiotic-loaded cement spacer in the first stage.

Risk factors for acute kidney injury include older age, obesity, chronic kidney disease, and overall comorbidity. Potentially modifiable risk factors include anemia, perioperative transfusions, aminoglycoside prophylaxis, perioperative renin-angiotensin system blockade, and postoperative nonsteroidal anti-inflammatory drugs. These should be mitigated when possible.

In patients with periprosthetic joint infection who receive antibiotic-loaded cement spacers, especially patients  with additional risk factors for acute kidney injury, strict attention should be paid to the dose of antibiotic in the spacer, with levels checked postoperatively if necessary. Nonnephrotoxic antibiotics should be chosen for systemic administration when possible.

Prospective randomized controlled trials are needed to guide therapy after total joint arthroplasty, and to verify the adverse long-term outcomes of acute kidney injury in this setting.

References
  1. Learmonth ID, Young C, Rorabeck C. The operation of the century: total hip replacement. Lancet 2007; 370(9597):1508–1519. doi:10.1016/S0140-6736(07)60457-7
  2. Pivec R, Johnson AJ, Mears SC, Mont MA. Hip arthroplasty. Lancet 2012; 380(9855):1768–1777. doi:10.1016/S0140-6736(12)60607-2
  3. Carr AJ, Robertsson O, Graves S, et al. Knee replacement. Lancet 2012; 379(9823):1331–1340. doi:10.1016/S0140-6736(11)60752-6
  4. Kurtz S, Ong K, Lau E, Mowat F, Halpern M. Projections of primary and revision hip and knee arthroplasty in the United States from 2005 to 2030. J Bone Joint Surg Am 2007; 89(4):780–785. doi:10.2106/JBJS.F.00222
  5. Kapadia BH, Berg RA, Daley JA, Fritz J, Bhave A, Mont MA. Periprosthetic joint infection. Lancet 2016; 387(10016):386–394. doi:10.1016/S0140-6736(14)61798-0
  6. Kurtz SM, Ong KL, Schmier J, et al. Future clinical and economic impact of revision total hip and knee arthroplasty. J Bone Joint Surg Am 2007; 89(suppl 3):144–151. doi:10.2106/JBJS.G.00587
  7. Jafari SM, Huang R, Joshi A, Parvizi J, Hozack WJ. Renal impairment following total joint arthroplasty: who is at risk? J Arthroplasty 2010; 25(6 suppl):49–53, 53.e1–2. doi:10.1016/j.arth.2010.04.008
  8. Jamsa P, Jamsen E, Lyytikainen LP, Kalliovalkama J, Eskelinen A, Oksala N. Risk factors associated with acute kidney injury in a cohort of 20,575 arthroplasty patients. Acta Orthop 2017; 88(4):370–376. doi:10.1080/17453674.2017.1301743
  9. Sehgal V, Bajwa SJ, Sehgal R, Eagan J, Reddy P, Lesko SM. Predictors of acute kidney injury in geriatric patients undergoing total knee replacement surgery. Int J Endocrinol Metab 2014; 12(3):e16713. doi:10.5812/ijem.16713
  10. Weingarten TN, Gurrieri C, Jarett PD, et al. Acute kidney injury following total joint arthroplasty: retrospective analysis. Can J Anaesth 2012; 59(12):1111–1118. doi:10.1007/s12630-012-9797-2
  11. Kimmel LA, Wilson S, Janardan JD, Liew SM, Walker RG. Incidence of acute kidney injury following total joint arthroplasty: a retrospective review by RIFLE criteria. Clin Kidney J 2014; 7(6):546–551. doi:10.1093/ckj/sfu108
  12. Warth LC, Noiseux NO, Hogue MH, Klaassen AL, Liu SS, Callaghan JJ. Risk of acute kidney injury after primary and revision total hip arthroplasty and total knee arthroplasty using a multimodal approach to perioperative pain control including ketorolac and celecoxib. J Arthroplasty 2016; 31(1):253–255. doi:10.1016/j.arth.2015.08.012
  13. Perregaard H, Damholt MB, Solgaard S, Petersen MB. Renal function after elective total hip replacement. Acta Orthop 2016; 87(3):235–238. doi:10.3109/17453674.2016.1155130
  14. Hassan BK, Sahlström A, Dessau RB. Risk factors for renal dysfunction after total hip joint replacement; a retrospective cohort study. J Orthop Surg Res 2015; 10:158. doi:10.1186/s13018-015-0299-0
  15. Nowicka A, Selvaraj T. Incidence of acute kidney injury after elective lower limb arthroplasty. J Clin Anesth 2016; 34:520–523. doi:10.1016/j.jclinane.2016.06.010
  16. Kim HJ, Koh WU, Kim SG, et al. Early postoperative albumin level following total knee arthroplasty is associated with acute kidney injury: a retrospective analysis of 1309 consecutive patients based on kidney disease improving global outcomes criteria. Medicine (Baltimore) 2016; 95(31):e4489. doi:10.1097/MD.0000000000004489
  17. Choi YJ, Kim S, Sim JH, Hahm K. Postoperative anemia is associated with acute kidney injury in patients undergoing total hip replacement arthroplasty: a retrospective study. Anesth Analg 2016; 122(6):1923–1928. doi:10.1213/ANE.0000000000001003
  18. Nielson E, Hennrikus E, Lehman E, Mets B. Angiotensin axis blockade, hypotension, and acute kidney injury in elective major orthopedic surgery. J Hosp Med 2014; 9(5):283–288. doi:10.1002/jhm.2155
  19. Courtney PM, Melnic CM, Zimmer Z, Anari J, Lee GC. Addition of vancomycin to cefazolin prophylaxis is associated with acute kidney injury after primary joint arthroplasty. Clin Orthop Relat Res 2015; 473(7):2197–2203. doi:10.1007/s11999-014-4062-3
  20. Dubrovskaya Y, Tejada R, Bosco J 3rd, et al. Single high dose gentamicin for perioperative prophylaxis in orthopedic surgery: evaluation of nephrotoxicity. SAGE Open Med 2015; 3:2050312115612803. doi:10.1177/2050312115612803
  21. Bell S, Davey P, Nathwani D, et al. Risk of AKI with gentamicin as surgical prophylaxis. J Am Soc Nephrol 2014; 25(11):2625–2632. doi:10.1681/ASN.2014010035
  22. Ross AD, Boscainos PJ, Malhas A, Wigderowitz C. Peri-operative renal morbidity secondary to gentamicin and flucloxacillin chemoprophylaxis for hip and knee arthroplasty. Scott Med J 2013; 58(4):209–212. doi:10.1177/0036933013507850
  23. Bailey O, Torkington MS, Anthony I, Wells J, Blyth M, Jones B. Antibiotic-related acute kidney injury in patients undergoing elective joint replacement. Bone Joint J 2014; 96-B(3):395–398. doi:10.1302/0301-620X.96B3.32745
  24. Challagundla SR, Knox D, Hawkins A, et al. Renal impairment after high-dose flucloxacillin and single-dose gentamicin prophylaxis in patients undergoing elective hip and knee replacement. Nephrol Dial Transplant 2013; 28(3):612–619. doi:10.1093/ndt/gfs458
  25. Johansson S, Christensen OM, Thorsmark AH. A retrospective study of acute kidney injury in hip arthroplasty patients receiving gentamicin and dicloxacillin. Acta Orthop 2016; 87(6):589–591. doi:10.1080/17453674.2016.1231008
  26. Ferguson KB, Winter A, Russo L, et al. Acute kidney injury following primary hip and knee arthroplasty surgery. Ann R Coll Surg Eng 2017; 99(4):307–312. doi:10.1308/rcsann.2016.0324
  27. Bjerregaard LS, Jorgensen CC, Kehlet H; Lundbeck Foundation Centre for Fast-Track Hip and Knee Replacement Collaborative Group. Serious renal and urological complications in fast-track primary total hip and knee arthroplasty; a detailed observational cohort study. Minerva Anestesiol 2016; 82(7):767–776. pmid:27028450
  28. Friedman JM, Couso R, Kitchens M, et al. Benign heart murmurs as a predictor for complications following total joint arthroplasty. J Orthop 2017; 14(4):470–474. doi:10.1016/j.jor.2017.07.009
  29. Nadkarni GN, Patel AA, Ahuja Y, et al. Incidence, risk factors, and outcome trends of acute kidney injury in elective total hip and knee arthroplasty. Am J Orthop (Belle Mead NJ) 2016; 45(1):E12–E19. pmid:26761921
  30. Lopez-de-Andres A, Hernandez-Barrera V, Martinez-Huedo MA, Villanueva-Martinez M, Jimenez-Trujillo I, Jimenez-Garcia R. Type 2 diabetes and in-hospital complications after revision of total hip and knee arthroplasty. PLoS One 2017; 12(8):e0183796. doi:10.1371/journal.pone.0183796
  31. Gharaibeh KA, Hamadah AM, Sierra RJ, Leung N, Kremers WK, El-Zoghby ZM. The rate of acute kidney injury after total hip arthroplasty is low but increases significantly in patients with specific comorbidities. J Bone Joint Surg Am 2017; 99(21):1819–1826. doi:10.2106/JBJS.16.01027
  32. Bellomo R, Ronco C, Kellum JA, Mehta RL, Palevsky P; Acute Dialysis Quality Initiative Workgroup. Acute renal failure—definition, outcome measures, animal models, fluid therapy and information technology needs: the Second International Consensus Conference of the Acute Dialysis Quality Initiative (ADQI) Group. Crit Care 2004; 8(4):R204–R212. doi:10.1186/cc2872
  33. Mehta RL, Kellum JA, Shah SV, et al; Acute Kidney Injury Network. Acute Kidney Injury Network: report of an initiative to improve outcomes in acute kidney injury. Crit Care 2007; 11(2):R31. doi:10.1186/cc5713
  34. Section 2: AKI Definition. Kidney Int Suppl (2011) 2012; 2(1):19–36. doi:10.1038/kisup.2011.32
  35. Grams ME, Waikar SS, MacMahon B, Whelton S, Ballew SH, Coresh J. Performance and limitations of administrative data in the identification of AKI. Clin J Am Soc Nephrol 2014; 9(4):682–689. doi:10.2215/CJN.07650713
  36. Parr SK, Matheny ME, Abdel-Kader K, et al. Acute kidney injury is a risk factor for subsequent proteinuria. Kidney Int 2018; 93(2):460–469. doi:10.1016/j.kint.2017.07.007
  37. Karkouti K, Wijeysundera DN, Yau TM, et al. Acute kidney injury after cardiac surgery: focus on modifiable risk factors. Circulation 2009; 119(4):495–502. doi:10.1161/CIRCULATIONAHA.108.786913
  38. Karkouti K, Grocott HP, Hall R, et al. Interrelationship of preoperative anemia, intraoperative anemia, and red blood cell transfusion as potentially modifiable risk factors for acute kidney injury in cardiac surgery: a historical multicentre cohort study. Can J Anaesth 2015; 62(4):377–384. doi:10.1007/s12630-014-0302-y
  39. Carson JL, Triulzi DJ, Ness PM. Indications for and adverse effects of red-cell transfusion. N Engl J Med 2017; 377(13):1261–1272. doi:10.1056/NEJMra1612789
  40. Karkouti K, Wijeysundera DN, Yau TM, et al. Advance targeted transfusion in anemic cardiac surgical patients for kidney protection: an unblinded randomized pilot clinical trial. Anesthesiology 2012; 116(3):613–621. doi:10.1097/ALN.0b013e3182475e39
  41. Newman ET, Watters TS, Lewis JS, et al. Impact of perioperative allogeneic and autologous blood transfusion on acute wound infection following total knee and total hip arthroplasty. J Bone Joint Surg Am 2014; 96(4):279–284. doi:10.2106/JBJS.L.01041
  42. AlBuhairan B, Hind D, Hutchinson A. Antibiotic prophylaxis for wound infections in total joint arthroplasty: a systematic review. J Bone Joint Surg Br 2008; 90(7):915–919. doi:10.1302/0301-620X.90B7.20498
  43. Corona Pérez-Cardona PS, Barro Ojeda V, Rodriguez Pardo D, et al. Clinical experience with daptomycin for the treatment of patients with knee and hip periprosthetic joint infections. J Antimicrob Chemother 2012; 67(7):1749–1754. doi:10.1093/jac/dks119
  44. Itani KM, Biswas P, Reisman A, Bhattacharyya H, Baruch AM. Clinical efficacy of oral linezolid compared with intravenous vancomycin for the treatment of methicillin-resistant Staphylococcus aureus-complicated skin and soft tissue infections: a retrospective, propensity score-matched, case-control analysis. Clin Ther 2012; 34(8):1667–1673.e1. doi:10.1016/j.clinthera.2012.06.018
  45. Dale H, Hallan G, Hallan G, Espehaug B, Havelin LI, Engesaeter LB. Increasing risk of revision due to deep infection after hip arthroplasty. Acta Orthop 2009; 80(6):639–645. doi:10.3109/17453670903506658
  46. Kurtz SM, Ong KL, Lau E, Bozic KJ, Berry D, Parvizi J. Prosthetic joint infection risk after TKA in the Medicare population. Clin Orthop Relat Res 2010; 468(1):52–56. doi:10.1007/s11999-009-1013-5
  47. Kunutsor SK, Whitehouse MR, Lenguerrand E, Blom AW, Beswick AD; INFORM Team. Re-infection outcomes following one- and two-stage surgical revision of infected knee prosthesis: a systematic review and meta-analysis. PLoS One 2016; 11(3):e0151537. doi:10.1371/journal.pone.0151537
  48. Negus JJ, Gifford PB, Haddad FS. Single-stage revision arthroplasty for infection—an underutilized treatment strategy. J Arthroplasty 2017; 32(7):2051–2055. doi:10.1016/j.arth.2017.02.059
  49. Stevens CM, Tetsworth KD, Calhoun JH, Mader JT. An articulated antibiotic spacer used for infected total knee arthroplasty: a comparative in vitro elution study of Simplex and Palacos bone cements. J Orthop Res 2005; 23(1):27–33. doi:10.1016/j.orthres.2004.03.003
  50. Chohfi M, Langlais F, Fourastier J, Minet J, Thomazeau H, Cormier M. Pharmacokinetics, uses, and limitations of vancomycin-loaded bone cement. Int Orthop 1998; 22(3):171–177. pmid:9728311
  51. Amin TJ, Lamping JW, Hendricks KJ, McIff TE. Increasing the elution of vancomycin from high-dose antibiotic-loaded bone cement: a novel preparation technique. J Bone Joint Surg Am 2012; 94(21):1946–1951. doi:10.2106/JBJS.L.00014
  52. Hsieh PH, Chen LH, Chen CH, Lee MS, Yang WE, Shih CH. Two-stage revision hip arthroplasty for infection with a custom-made, antibiotic-loaded, cement prosthesis as an interim spacer. J Trauma 2004; 56(6):1247–1252. pmid:15211133
  53. Cui Q, Mihalko WM, Shields JS, Ries M, Saleh KJ. Antibiotic-impregnated cement spacers for the treatment of infection associated with total hip or knee arthroplasty. J Bone Joint Surg Am 2007; 89(4):871–882. doi:10.2106/JBJS.E.01070
  54. Jiranek WA, Hanssen AD, Greenwald AS. Antibiotic-loaded bone cement for infection prophylaxis in total joint replacement. J Bone Joint Surg Am 2006; 88(11):2487–2500. doi:10.2106/JBJS.E.01126
  55. Vrabec G, Stevenson W, Elguizaoui S, Kirsch M, Pinkowski J. What is the intraarticular concentration of tobramycin using low-dose tobramycin bone cement in TKA: an in vivo analysis? Clin Orthop Relat Res 2016; 474(11):2441–2447. doi:10.1007/s11999-016-5006-x
  56. Sterling GJ, Crawford S, Potter JH, Koerbin G, Crawford R. The pharmacokinetics of Simplex-tobramycin bone cement. J Bone Joint Surg Br 2003; 85(5):646–649. pmid:12892183
  57. Fletcher MD, Spencer RF, Langkamer VG, Lovering AM. Gentamicin concentrations in diagnostic aspirates from 25 patients with hip and knee arthroplasties. Acta Orthop Scand 2004; 75(2):173–176. doi:10.1080/00016470412331294425
  58. Lau BP, Kumar VP. Acute kidney injury (AKI) with the use of antibiotic-impregnated bone cement in primary total knee arthroplasty. Ann Acad Med Singapore 2013; 42(12):692–695. pmid:24463833
  59. Penner MJ, Masri BA, Duncan CP. Elution characteristics of vancomycin and tobramycin combined in acrylic bone-cement. J Arthroplasty 1996; 11(8):939–944. pmid:8986572
  60. Kalil GZ, Ernst EJ, Johnson SJ, et al. Systemic exposure to aminoglycosides following knee and hip arthroplasty with aminoglycoside-loaded bone cement implants. Ann Pharmacother 2012; 46(7–8):929–934. doi:10.1345/aph.1R049
  61. Hsieh PH, Chang YH, Chen SH, Ueng SW, Shih CH. High concentration and bioactivity of vancomycin and aztreonam eluted from simplex cement spacers in two-stage revision of infected hip implants: a study of 46 patients at an average follow-up of 107 days. J Orthop Res 2006; 24(8):1615–1621. doi:10.1002/jor.20214
  62. Curtis JM, Sternhagen V, Batts D. Acute renal failure after placement of tobramycin-impregnated bone cement in an infected total knee arthroplasty. Pharmacotherapy 2005; 25(6):876–880. pmid:15927906
  63. Wu IM, Marin EP, Kashgarian M, Brewster UC. A case of an acute kidney injury secondary to an implanted aminoglycoside. Kidney Int 2009; 75(10):1109–1112. doi:10.1038/ki.2008.386
  64. Chalmers PN, Frank J, Sporer SM. Acute postoperative renal failure following insertion of an antibiotic-impregnated cement spacer in revision total joint arthroplasty: two case reports. JBJS Case Connect 2012; 2(1):e12. doi:10.2106/JBJS.CC.K.00094
  65. Patrick BN, Rivey MP, Allington DR. Acute renal failure associated with vancomycin- and tobramycin-laden cement in total hip arthroplasty. Ann Pharmacother 2006; 40(11):2037–2042. doi:10.1345/aph.1H173
  66. Dovas S, Liakopoulos V, Papatheodorou L, et al. Acute renal failure after antibiotic-impregnated bone cement treatment of an infected total knee arthroplasty. Clin Nephrol 2008; 69(3):207–212. pmid:18397720
  67. McGlothan KR, Gosmanova EO. A case report of acute interstitial nephritis associated with antibiotic-impregnated orthopedic bone-cement spacer. Tenn Med 2012; 105(9):37–40, 42. pmid:23097958
  68. Jung J, Schmid NV, Kelm J, Schmitt E, Anagnostakos K. Complications after spacer implantation in the treatment of hip joint infections. Int J Med Sci 2009; 6(5):265–273. pmid:19834592
  69. Menge TJ, Koethe JR, Jenkins CA, et al. Acute kidney injury after placement of an antibiotic-impregnated cement spacer during revision total knee arthroplasty. J Arthroplasty 2012; 27(6):1221–1227.e1–2. doi:10.1016/j.arth.2011.12.005
  70. Gooding CR, Masri BA, Duncan CP, Greidanus NV, Garbuz DS. Durable infection control and function with the PROSTALAC spacer in two-stage revision for infected knee arthroplasty. Clin Orthop Relat Res 2011; 469(4):985–993. doi:10.1007/s11999-010-1579-y
  71. Springer BD, Lee GC, Osmon D, Haidukewych GJ, Hanssen AD, Jacofsky DJ. Systemic safety of high-dose antibiotic-loaded cement spacers after resection of an infected total knee arthroplasty. Clin Orthop Relat Res 2004; 427:47–51. pmid:15552135
  72. Noto MJ, Koethe JR, Miller G, Wright PW. Detectable serum tobramycin levels in patients with renal dysfunction and recent placement of antibiotic-impregnated cement knee or hip spacers. Clin Infect Dis 2014; 58(12):1783–1784. doi:10.1093/cid/ciu159
  73. Aeng ES, Shalansky KF, Lau TT, et al. Acute kidney injury with tobramycin-impregnated bone cement spacers in prosthetic joint infections. Ann Pharmacother 2015; 49(11):1207–1213. doi:10.1177/1060028015600176
  74. Geller JA, Cunn G, Herschmiller T, Murtaugh T, Chen A. Acute kidney injury after first-stage joint revision for infection: Risk factors and the impact of antibiotic dosing. J Arthroplasty 2017; 32(10):3120–3125. doi:10.1016/j.arth.2017.04.054
  75. Reed EE, Johnston J, Severing J, Stevenson KB, Deutscher M. Nephrotoxicity risk factors and intravenous vancomycin dosing in the immediate postoperative period following antibiotic-impregnated cement spacer placement. Ann Pharmacother 2014; 48(8):962–969. doi:10.1177/1060028014535360
  76. Koo KH, Yang JW, Cho SH, et al. Impregnation of vancomycin, gentamicin, and cefotaxime in a cement spacer for two-stage cementless reconstruction in infected total hip arthroplasty. J Arthroplasty 2001; 16(7):882–892. doi:10.1054/arth.2001.24444
  77. Forsythe ME, Crawford S, Sterling GJ, Whitehouse SL, Crawford R. Safeness of simplex-tobramycin bone cement in patients with renal dysfunction undergoing total hip replacement. J Orthop Surg (Hong Kong) 2006; 14(1):38–42. doi:10.1177/230949900601400109
  78. Hsieh PH, Huang KC, Tai CL. Liquid gentamicin in bone cement spacers: in vivo antibiotic release and systemic safety in two-stage revision of infected hip arthroplasty. J Trauma 2009; 66(3):804–808. doi:10.1097/TA.0b013e31818896cc
  79. Hofmann AA, Goldberg T, Tanner AM, Kurtin SM. Treatment of infected total knee arthroplasty using an articulating spacer: 2- to 12-year experience. Clin Orthop Relat Res 2005; 430:125–131. pmid:15662313
  80. Evans RP. Successful treatment of total hip and knee infection with articulating antibiotic components: a modified treatment method. Clin Orthop Relat Res 2004; 427:37–46. pmid:15552134
  81. Yadav A, Alijanipour P, Ackerman CT, Karanth S, Hozack WJ, Filippone EJ. Acute kidney injury following failed total hip and knee arthroplasty. J Arthroplasty 2018; 33(10):3297–3303. doi:10.1016/j.arth.2018.06.019
  82. Hsieh PH, Huang KC, Lee PC, Lee MS. Two-stage revision of infected hip arthroplasty using an antibiotic-loaded spacer: retrospective comparison between short-term and prolonged antibiotic therapy. J Antimicrob Chemother 2009; 64(2):392–397. doi:10.1093/jac/dkp177
  83. Luu A, Syed F, Raman G, et al. Two-stage arthroplasty for prosthetic joint infection: a systematic review of acute kidney injury, systemic toxicity and infection control. J Arthroplasty 2013; 28(9):1490–1498.e1. doi:10.1016/j.arth.2013.02.035
  84. Filippone EJ, Kraft WK, Farber JL. The nephrotoxicity of vancomycin. Clin Pharmacol Ther 2017; 102(3):459–469. doi:10.1002/cpt.726
References
  1. Learmonth ID, Young C, Rorabeck C. The operation of the century: total hip replacement. Lancet 2007; 370(9597):1508–1519. doi:10.1016/S0140-6736(07)60457-7
  2. Pivec R, Johnson AJ, Mears SC, Mont MA. Hip arthroplasty. Lancet 2012; 380(9855):1768–1777. doi:10.1016/S0140-6736(12)60607-2
  3. Carr AJ, Robertsson O, Graves S, et al. Knee replacement. Lancet 2012; 379(9823):1331–1340. doi:10.1016/S0140-6736(11)60752-6
  4. Kurtz S, Ong K, Lau E, Mowat F, Halpern M. Projections of primary and revision hip and knee arthroplasty in the United States from 2005 to 2030. J Bone Joint Surg Am 2007; 89(4):780–785. doi:10.2106/JBJS.F.00222
  5. Kapadia BH, Berg RA, Daley JA, Fritz J, Bhave A, Mont MA. Periprosthetic joint infection. Lancet 2016; 387(10016):386–394. doi:10.1016/S0140-6736(14)61798-0
  6. Kurtz SM, Ong KL, Schmier J, et al. Future clinical and economic impact of revision total hip and knee arthroplasty. J Bone Joint Surg Am 2007; 89(suppl 3):144–151. doi:10.2106/JBJS.G.00587
  7. Jafari SM, Huang R, Joshi A, Parvizi J, Hozack WJ. Renal impairment following total joint arthroplasty: who is at risk? J Arthroplasty 2010; 25(6 suppl):49–53, 53.e1–2. doi:10.1016/j.arth.2010.04.008
  8. Jamsa P, Jamsen E, Lyytikainen LP, Kalliovalkama J, Eskelinen A, Oksala N. Risk factors associated with acute kidney injury in a cohort of 20,575 arthroplasty patients. Acta Orthop 2017; 88(4):370–376. doi:10.1080/17453674.2017.1301743
  9. Sehgal V, Bajwa SJ, Sehgal R, Eagan J, Reddy P, Lesko SM. Predictors of acute kidney injury in geriatric patients undergoing total knee replacement surgery. Int J Endocrinol Metab 2014; 12(3):e16713. doi:10.5812/ijem.16713
  10. Weingarten TN, Gurrieri C, Jarett PD, et al. Acute kidney injury following total joint arthroplasty: retrospective analysis. Can J Anaesth 2012; 59(12):1111–1118. doi:10.1007/s12630-012-9797-2
  11. Kimmel LA, Wilson S, Janardan JD, Liew SM, Walker RG. Incidence of acute kidney injury following total joint arthroplasty: a retrospective review by RIFLE criteria. Clin Kidney J 2014; 7(6):546–551. doi:10.1093/ckj/sfu108
  12. Warth LC, Noiseux NO, Hogue MH, Klaassen AL, Liu SS, Callaghan JJ. Risk of acute kidney injury after primary and revision total hip arthroplasty and total knee arthroplasty using a multimodal approach to perioperative pain control including ketorolac and celecoxib. J Arthroplasty 2016; 31(1):253–255. doi:10.1016/j.arth.2015.08.012
  13. Perregaard H, Damholt MB, Solgaard S, Petersen MB. Renal function after elective total hip replacement. Acta Orthop 2016; 87(3):235–238. doi:10.3109/17453674.2016.1155130
  14. Hassan BK, Sahlström A, Dessau RB. Risk factors for renal dysfunction after total hip joint replacement; a retrospective cohort study. J Orthop Surg Res 2015; 10:158. doi:10.1186/s13018-015-0299-0
  15. Nowicka A, Selvaraj T. Incidence of acute kidney injury after elective lower limb arthroplasty. J Clin Anesth 2016; 34:520–523. doi:10.1016/j.jclinane.2016.06.010
  16. Kim HJ, Koh WU, Kim SG, et al. Early postoperative albumin level following total knee arthroplasty is associated with acute kidney injury: a retrospective analysis of 1309 consecutive patients based on kidney disease improving global outcomes criteria. Medicine (Baltimore) 2016; 95(31):e4489. doi:10.1097/MD.0000000000004489
  17. Choi YJ, Kim S, Sim JH, Hahm K. Postoperative anemia is associated with acute kidney injury in patients undergoing total hip replacement arthroplasty: a retrospective study. Anesth Analg 2016; 122(6):1923–1928. doi:10.1213/ANE.0000000000001003
  18. Nielson E, Hennrikus E, Lehman E, Mets B. Angiotensin axis blockade, hypotension, and acute kidney injury in elective major orthopedic surgery. J Hosp Med 2014; 9(5):283–288. doi:10.1002/jhm.2155
  19. Courtney PM, Melnic CM, Zimmer Z, Anari J, Lee GC. Addition of vancomycin to cefazolin prophylaxis is associated with acute kidney injury after primary joint arthroplasty. Clin Orthop Relat Res 2015; 473(7):2197–2203. doi:10.1007/s11999-014-4062-3
  20. Dubrovskaya Y, Tejada R, Bosco J 3rd, et al. Single high dose gentamicin for perioperative prophylaxis in orthopedic surgery: evaluation of nephrotoxicity. SAGE Open Med 2015; 3:2050312115612803. doi:10.1177/2050312115612803
  21. Bell S, Davey P, Nathwani D, et al. Risk of AKI with gentamicin as surgical prophylaxis. J Am Soc Nephrol 2014; 25(11):2625–2632. doi:10.1681/ASN.2014010035
  22. Ross AD, Boscainos PJ, Malhas A, Wigderowitz C. Peri-operative renal morbidity secondary to gentamicin and flucloxacillin chemoprophylaxis for hip and knee arthroplasty. Scott Med J 2013; 58(4):209–212. doi:10.1177/0036933013507850
  23. Bailey O, Torkington MS, Anthony I, Wells J, Blyth M, Jones B. Antibiotic-related acute kidney injury in patients undergoing elective joint replacement. Bone Joint J 2014; 96-B(3):395–398. doi:10.1302/0301-620X.96B3.32745
  24. Challagundla SR, Knox D, Hawkins A, et al. Renal impairment after high-dose flucloxacillin and single-dose gentamicin prophylaxis in patients undergoing elective hip and knee replacement. Nephrol Dial Transplant 2013; 28(3):612–619. doi:10.1093/ndt/gfs458
  25. Johansson S, Christensen OM, Thorsmark AH. A retrospective study of acute kidney injury in hip arthroplasty patients receiving gentamicin and dicloxacillin. Acta Orthop 2016; 87(6):589–591. doi:10.1080/17453674.2016.1231008
  26. Ferguson KB, Winter A, Russo L, et al. Acute kidney injury following primary hip and knee arthroplasty surgery. Ann R Coll Surg Eng 2017; 99(4):307–312. doi:10.1308/rcsann.2016.0324
  27. Bjerregaard LS, Jorgensen CC, Kehlet H; Lundbeck Foundation Centre for Fast-Track Hip and Knee Replacement Collaborative Group. Serious renal and urological complications in fast-track primary total hip and knee arthroplasty; a detailed observational cohort study. Minerva Anestesiol 2016; 82(7):767–776. pmid:27028450
  28. Friedman JM, Couso R, Kitchens M, et al. Benign heart murmurs as a predictor for complications following total joint arthroplasty. J Orthop 2017; 14(4):470–474. doi:10.1016/j.jor.2017.07.009
  29. Nadkarni GN, Patel AA, Ahuja Y, et al. Incidence, risk factors, and outcome trends of acute kidney injury in elective total hip and knee arthroplasty. Am J Orthop (Belle Mead NJ) 2016; 45(1):E12–E19. pmid:26761921
  30. Lopez-de-Andres A, Hernandez-Barrera V, Martinez-Huedo MA, Villanueva-Martinez M, Jimenez-Trujillo I, Jimenez-Garcia R. Type 2 diabetes and in-hospital complications after revision of total hip and knee arthroplasty. PLoS One 2017; 12(8):e0183796. doi:10.1371/journal.pone.0183796
  31. Gharaibeh KA, Hamadah AM, Sierra RJ, Leung N, Kremers WK, El-Zoghby ZM. The rate of acute kidney injury after total hip arthroplasty is low but increases significantly in patients with specific comorbidities. J Bone Joint Surg Am 2017; 99(21):1819–1826. doi:10.2106/JBJS.16.01027
  32. Bellomo R, Ronco C, Kellum JA, Mehta RL, Palevsky P; Acute Dialysis Quality Initiative Workgroup. Acute renal failure—definition, outcome measures, animal models, fluid therapy and information technology needs: the Second International Consensus Conference of the Acute Dialysis Quality Initiative (ADQI) Group. Crit Care 2004; 8(4):R204–R212. doi:10.1186/cc2872
  33. Mehta RL, Kellum JA, Shah SV, et al; Acute Kidney Injury Network. Acute Kidney Injury Network: report of an initiative to improve outcomes in acute kidney injury. Crit Care 2007; 11(2):R31. doi:10.1186/cc5713
  34. Section 2: AKI Definition. Kidney Int Suppl (2011) 2012; 2(1):19–36. doi:10.1038/kisup.2011.32
  35. Grams ME, Waikar SS, MacMahon B, Whelton S, Ballew SH, Coresh J. Performance and limitations of administrative data in the identification of AKI. Clin J Am Soc Nephrol 2014; 9(4):682–689. doi:10.2215/CJN.07650713
  36. Parr SK, Matheny ME, Abdel-Kader K, et al. Acute kidney injury is a risk factor for subsequent proteinuria. Kidney Int 2018; 93(2):460–469. doi:10.1016/j.kint.2017.07.007
  37. Karkouti K, Wijeysundera DN, Yau TM, et al. Acute kidney injury after cardiac surgery: focus on modifiable risk factors. Circulation 2009; 119(4):495–502. doi:10.1161/CIRCULATIONAHA.108.786913
  38. Karkouti K, Grocott HP, Hall R, et al. Interrelationship of preoperative anemia, intraoperative anemia, and red blood cell transfusion as potentially modifiable risk factors for acute kidney injury in cardiac surgery: a historical multicentre cohort study. Can J Anaesth 2015; 62(4):377–384. doi:10.1007/s12630-014-0302-y
  39. Carson JL, Triulzi DJ, Ness PM. Indications for and adverse effects of red-cell transfusion. N Engl J Med 2017; 377(13):1261–1272. doi:10.1056/NEJMra1612789
  40. Karkouti K, Wijeysundera DN, Yau TM, et al. Advance targeted transfusion in anemic cardiac surgical patients for kidney protection: an unblinded randomized pilot clinical trial. Anesthesiology 2012; 116(3):613–621. doi:10.1097/ALN.0b013e3182475e39
  41. Newman ET, Watters TS, Lewis JS, et al. Impact of perioperative allogeneic and autologous blood transfusion on acute wound infection following total knee and total hip arthroplasty. J Bone Joint Surg Am 2014; 96(4):279–284. doi:10.2106/JBJS.L.01041
  42. AlBuhairan B, Hind D, Hutchinson A. Antibiotic prophylaxis for wound infections in total joint arthroplasty: a systematic review. J Bone Joint Surg Br 2008; 90(7):915–919. doi:10.1302/0301-620X.90B7.20498
  43. Corona Pérez-Cardona PS, Barro Ojeda V, Rodriguez Pardo D, et al. Clinical experience with daptomycin for the treatment of patients with knee and hip periprosthetic joint infections. J Antimicrob Chemother 2012; 67(7):1749–1754. doi:10.1093/jac/dks119
  44. Itani KM, Biswas P, Reisman A, Bhattacharyya H, Baruch AM. Clinical efficacy of oral linezolid compared with intravenous vancomycin for the treatment of methicillin-resistant Staphylococcus aureus-complicated skin and soft tissue infections: a retrospective, propensity score-matched, case-control analysis. Clin Ther 2012; 34(8):1667–1673.e1. doi:10.1016/j.clinthera.2012.06.018
  45. Dale H, Hallan G, Hallan G, Espehaug B, Havelin LI, Engesaeter LB. Increasing risk of revision due to deep infection after hip arthroplasty. Acta Orthop 2009; 80(6):639–645. doi:10.3109/17453670903506658
  46. Kurtz SM, Ong KL, Lau E, Bozic KJ, Berry D, Parvizi J. Prosthetic joint infection risk after TKA in the Medicare population. Clin Orthop Relat Res 2010; 468(1):52–56. doi:10.1007/s11999-009-1013-5
  47. Kunutsor SK, Whitehouse MR, Lenguerrand E, Blom AW, Beswick AD; INFORM Team. Re-infection outcomes following one- and two-stage surgical revision of infected knee prosthesis: a systematic review and meta-analysis. PLoS One 2016; 11(3):e0151537. doi:10.1371/journal.pone.0151537
  48. Negus JJ, Gifford PB, Haddad FS. Single-stage revision arthroplasty for infection—an underutilized treatment strategy. J Arthroplasty 2017; 32(7):2051–2055. doi:10.1016/j.arth.2017.02.059
  49. Stevens CM, Tetsworth KD, Calhoun JH, Mader JT. An articulated antibiotic spacer used for infected total knee arthroplasty: a comparative in vitro elution study of Simplex and Palacos bone cements. J Orthop Res 2005; 23(1):27–33. doi:10.1016/j.orthres.2004.03.003
  50. Chohfi M, Langlais F, Fourastier J, Minet J, Thomazeau H, Cormier M. Pharmacokinetics, uses, and limitations of vancomycin-loaded bone cement. Int Orthop 1998; 22(3):171–177. pmid:9728311
  51. Amin TJ, Lamping JW, Hendricks KJ, McIff TE. Increasing the elution of vancomycin from high-dose antibiotic-loaded bone cement: a novel preparation technique. J Bone Joint Surg Am 2012; 94(21):1946–1951. doi:10.2106/JBJS.L.00014
  52. Hsieh PH, Chen LH, Chen CH, Lee MS, Yang WE, Shih CH. Two-stage revision hip arthroplasty for infection with a custom-made, antibiotic-loaded, cement prosthesis as an interim spacer. J Trauma 2004; 56(6):1247–1252. pmid:15211133
  53. Cui Q, Mihalko WM, Shields JS, Ries M, Saleh KJ. Antibiotic-impregnated cement spacers for the treatment of infection associated with total hip or knee arthroplasty. J Bone Joint Surg Am 2007; 89(4):871–882. doi:10.2106/JBJS.E.01070
  54. Jiranek WA, Hanssen AD, Greenwald AS. Antibiotic-loaded bone cement for infection prophylaxis in total joint replacement. J Bone Joint Surg Am 2006; 88(11):2487–2500. doi:10.2106/JBJS.E.01126
  55. Vrabec G, Stevenson W, Elguizaoui S, Kirsch M, Pinkowski J. What is the intraarticular concentration of tobramycin using low-dose tobramycin bone cement in TKA: an in vivo analysis? Clin Orthop Relat Res 2016; 474(11):2441–2447. doi:10.1007/s11999-016-5006-x
  56. Sterling GJ, Crawford S, Potter JH, Koerbin G, Crawford R. The pharmacokinetics of Simplex-tobramycin bone cement. J Bone Joint Surg Br 2003; 85(5):646–649. pmid:12892183
  57. Fletcher MD, Spencer RF, Langkamer VG, Lovering AM. Gentamicin concentrations in diagnostic aspirates from 25 patients with hip and knee arthroplasties. Acta Orthop Scand 2004; 75(2):173–176. doi:10.1080/00016470412331294425
  58. Lau BP, Kumar VP. Acute kidney injury (AKI) with the use of antibiotic-impregnated bone cement in primary total knee arthroplasty. Ann Acad Med Singapore 2013; 42(12):692–695. pmid:24463833
  59. Penner MJ, Masri BA, Duncan CP. Elution characteristics of vancomycin and tobramycin combined in acrylic bone-cement. J Arthroplasty 1996; 11(8):939–944. pmid:8986572
  60. Kalil GZ, Ernst EJ, Johnson SJ, et al. Systemic exposure to aminoglycosides following knee and hip arthroplasty with aminoglycoside-loaded bone cement implants. Ann Pharmacother 2012; 46(7–8):929–934. doi:10.1345/aph.1R049
  61. Hsieh PH, Chang YH, Chen SH, Ueng SW, Shih CH. High concentration and bioactivity of vancomycin and aztreonam eluted from simplex cement spacers in two-stage revision of infected hip implants: a study of 46 patients at an average follow-up of 107 days. J Orthop Res 2006; 24(8):1615–1621. doi:10.1002/jor.20214
  62. Curtis JM, Sternhagen V, Batts D. Acute renal failure after placement of tobramycin-impregnated bone cement in an infected total knee arthroplasty. Pharmacotherapy 2005; 25(6):876–880. pmid:15927906
  63. Wu IM, Marin EP, Kashgarian M, Brewster UC. A case of an acute kidney injury secondary to an implanted aminoglycoside. Kidney Int 2009; 75(10):1109–1112. doi:10.1038/ki.2008.386
  64. Chalmers PN, Frank J, Sporer SM. Acute postoperative renal failure following insertion of an antibiotic-impregnated cement spacer in revision total joint arthroplasty: two case reports. JBJS Case Connect 2012; 2(1):e12. doi:10.2106/JBJS.CC.K.00094
  65. Patrick BN, Rivey MP, Allington DR. Acute renal failure associated with vancomycin- and tobramycin-laden cement in total hip arthroplasty. Ann Pharmacother 2006; 40(11):2037–2042. doi:10.1345/aph.1H173
  66. Dovas S, Liakopoulos V, Papatheodorou L, et al. Acute renal failure after antibiotic-impregnated bone cement treatment of an infected total knee arthroplasty. Clin Nephrol 2008; 69(3):207–212. pmid:18397720
  67. McGlothan KR, Gosmanova EO. A case report of acute interstitial nephritis associated with antibiotic-impregnated orthopedic bone-cement spacer. Tenn Med 2012; 105(9):37–40, 42. pmid:23097958
  68. Jung J, Schmid NV, Kelm J, Schmitt E, Anagnostakos K. Complications after spacer implantation in the treatment of hip joint infections. Int J Med Sci 2009; 6(5):265–273. pmid:19834592
  69. Menge TJ, Koethe JR, Jenkins CA, et al. Acute kidney injury after placement of an antibiotic-impregnated cement spacer during revision total knee arthroplasty. J Arthroplasty 2012; 27(6):1221–1227.e1–2. doi:10.1016/j.arth.2011.12.005
  70. Gooding CR, Masri BA, Duncan CP, Greidanus NV, Garbuz DS. Durable infection control and function with the PROSTALAC spacer in two-stage revision for infected knee arthroplasty. Clin Orthop Relat Res 2011; 469(4):985–993. doi:10.1007/s11999-010-1579-y
  71. Springer BD, Lee GC, Osmon D, Haidukewych GJ, Hanssen AD, Jacofsky DJ. Systemic safety of high-dose antibiotic-loaded cement spacers after resection of an infected total knee arthroplasty. Clin Orthop Relat Res 2004; 427:47–51. pmid:15552135
  72. Noto MJ, Koethe JR, Miller G, Wright PW. Detectable serum tobramycin levels in patients with renal dysfunction and recent placement of antibiotic-impregnated cement knee or hip spacers. Clin Infect Dis 2014; 58(12):1783–1784. doi:10.1093/cid/ciu159
  73. Aeng ES, Shalansky KF, Lau TT, et al. Acute kidney injury with tobramycin-impregnated bone cement spacers in prosthetic joint infections. Ann Pharmacother 2015; 49(11):1207–1213. doi:10.1177/1060028015600176
  74. Geller JA, Cunn G, Herschmiller T, Murtaugh T, Chen A. Acute kidney injury after first-stage joint revision for infection: Risk factors and the impact of antibiotic dosing. J Arthroplasty 2017; 32(10):3120–3125. doi:10.1016/j.arth.2017.04.054
  75. Reed EE, Johnston J, Severing J, Stevenson KB, Deutscher M. Nephrotoxicity risk factors and intravenous vancomycin dosing in the immediate postoperative period following antibiotic-impregnated cement spacer placement. Ann Pharmacother 2014; 48(8):962–969. doi:10.1177/1060028014535360
  76. Koo KH, Yang JW, Cho SH, et al. Impregnation of vancomycin, gentamicin, and cefotaxime in a cement spacer for two-stage cementless reconstruction in infected total hip arthroplasty. J Arthroplasty 2001; 16(7):882–892. doi:10.1054/arth.2001.24444
  77. Forsythe ME, Crawford S, Sterling GJ, Whitehouse SL, Crawford R. Safeness of simplex-tobramycin bone cement in patients with renal dysfunction undergoing total hip replacement. J Orthop Surg (Hong Kong) 2006; 14(1):38–42. doi:10.1177/230949900601400109
  78. Hsieh PH, Huang KC, Tai CL. Liquid gentamicin in bone cement spacers: in vivo antibiotic release and systemic safety in two-stage revision of infected hip arthroplasty. J Trauma 2009; 66(3):804–808. doi:10.1097/TA.0b013e31818896cc
  79. Hofmann AA, Goldberg T, Tanner AM, Kurtin SM. Treatment of infected total knee arthroplasty using an articulating spacer: 2- to 12-year experience. Clin Orthop Relat Res 2005; 430:125–131. pmid:15662313
  80. Evans RP. Successful treatment of total hip and knee infection with articulating antibiotic components: a modified treatment method. Clin Orthop Relat Res 2004; 427:37–46. pmid:15552134
  81. Yadav A, Alijanipour P, Ackerman CT, Karanth S, Hozack WJ, Filippone EJ. Acute kidney injury following failed total hip and knee arthroplasty. J Arthroplasty 2018; 33(10):3297–3303. doi:10.1016/j.arth.2018.06.019
  82. Hsieh PH, Huang KC, Lee PC, Lee MS. Two-stage revision of infected hip arthroplasty using an antibiotic-loaded spacer: retrospective comparison between short-term and prolonged antibiotic therapy. J Antimicrob Chemother 2009; 64(2):392–397. doi:10.1093/jac/dkp177
  83. Luu A, Syed F, Raman G, et al. Two-stage arthroplasty for prosthetic joint infection: a systematic review of acute kidney injury, systemic toxicity and infection control. J Arthroplasty 2013; 28(9):1490–1498.e1. doi:10.1016/j.arth.2013.02.035
  84. Filippone EJ, Kraft WK, Farber JL. The nephrotoxicity of vancomycin. Clin Pharmacol Ther 2017; 102(3):459–469. doi:10.1002/cpt.726
Issue
Cleveland Clinic Journal of Medicine - 86(4)
Issue
Cleveland Clinic Journal of Medicine - 86(4)
Page Number
263-276
Page Number
263-276
Publications
Publications
Topics
Article Type
Display Headline
Acute kidney injury after hip or knee replacement: Can we lower the risk?
Display Headline
Acute kidney injury after hip or knee replacement: Can we lower the risk?
Legacy Keywords
acute kidney injury, AKI, total joint arthroplasty, TJA, hip replacement, knee replacement, antibiotic, aminoglycoside, cement, prosthetic joint infections, antibiotic-loaded cement, gentamicin, tobramycin, vancomycin, Edward Filippone, Anju Yadav
Legacy Keywords
acute kidney injury, AKI, total joint arthroplasty, TJA, hip replacement, knee replacement, antibiotic, aminoglycoside, cement, prosthetic joint infections, antibiotic-loaded cement, gentamicin, tobramycin, vancomycin, Edward Filippone, Anju Yadav
Sections
Inside the Article

KEY POINTS

  • Using current diagnostic criteria, the incidence of acute kidney injury complicating primary total joint arthroplasty may be nearly 10%, and 25% after placement of an antibiotic-loaded cement spacer to treat infection.
  • In primary total joint arthroplasty, significant risk factors include older age, higher body mass index, chronic kidney disease, comorbidity, anemia, perioperative transfusion, aminoglycoside prophylaxis and treatment, preoperative heart murmur, and renin-angiotensin-aldosterone system blockade.
  • Acute kidney injury may arise from infection, systemic administration of nephrotoxic antibiotics, and elution of antibiotics from antibiotic-loaded cement.
  • No randomized controlled trial aimed at reducing acute kidney injury in these settings has been published; however, suggestions for practice modification are made based on the available data.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Tue, 03/26/2019 - 08:15
Un-Gate On Date
Tue, 03/26/2019 - 08:15
Use ProPublica
CFC Schedule Remove Status
Tue, 03/26/2019 - 08:15
Hide sidebar & use full width
render the right sidebar.
Article PDF Media

A woman, age 35, with new-onset ascites

Article Type
Changed
Mon, 04/01/2019 - 08:54
Display Headline
A woman, age 35, with new-onset ascites

A 35-year-old woman is admitted to the hospital with a 5-day history of abdominal distention and jaundice. She reports no history of fever, chills, night sweats, abdominal pain, nausea, vomiting, diarrhea, changes in urine color, change in stool color, weight loss, weight gain, or loss of appetite.

She is petite, with a body mass index of 19.4 kg/m2. She has no known history of medical conditions or surgery and is not taking any medications. Her family history is unremarkable, and she denies current or past tobacco, alcohol, or illicit drug use.

RECENT TRAVEL

She says that during a trip to Central America several months ago, she had suffered a seizure and was taken to a local hospital, where laboratory testing revealed elevated aspartate aminotransferase (AST) and alanine aminotransferase (ALT) levels. She says that the rest of the workup at that time was normal.

About 1 week after that incident, she returned home and saw her primary care physician, who ordered further testing, which showed mild hyperbilirubinemia and mild elevation of AST and ALT levels. Her physician attributed the elevations to atovaquone, which she had been taking for malaria prophylaxis, as repeat testing 2 weeks later showed improvement in AST and ALT levels.

The patient says she returned to her normal state of health until about 5 days ago, when she noticed jaundice and abdominal distention, but without abdominal pain, dark urine, or clay-colored stools. She became concerned and went to her local hospital. Testing there noted mild elevation of AST and ALT, as well as an elevated international normalized ratio (INR) and hyperbilirubinemia. Computed tomography of the abdomen and pelvis showed hepatomegaly with possible fatty liver. Because of these results, the patient was transferred to our institution for further evaluation.

EVALUATION AT OUR INSTITUTION

On examination at our institution, she is afebrile, and vital signs are within normal ranges. She has bilateral scleral icterus and diffuse jaundice, but no other skin finding such as rash or spider angioma. She has no lymphadenopathy. Her abdomen is distended, with tense ascites, and her liver is tender to palpation. The tip of the spleen is not palpable.

lippert_new-onsetascites_t1.jpg

The cardiovascular examination reveals no murmurs, rubs, or gallops, but she has jugular venous distention and +2 pitting edema of both lower extremities.

On respiratory examination, there is dullness to percussion, with slight crackles on auscultation at the right lung base. The neurologic examination is normal.

Table 1 shows the results of initial laboratory testing.

1. Which study would provide the most information on the cause of ascites?

  • Abdominal ultrasonography
  • Abdominal paracentesis with ascitic fluid analysis
  • Chest radiography
  • Echocardiography
  • Urine protein-to-creatinine ratio

Abdominal paracentesis with ascitic fluid analysis is the essential study for any patient with clinically apparent new-onset ascites.1–3 It is the study that provides the most information on the cause of ascites.

In our patient, abdominal paracentesis yields 1,000 mL of straw-colored ascitic fluid, and analysis shows 86 nucleated cells, 28 of which are polymorphonuclear cells, and 0 red blood cells, with negative Gram stain and culture. The ascitic albumin level is 0.85 g/dL, with an ascitic protein of 1.1 g/dL.

Abdominal ultrasonography shows a diffusely echogenic liver, no focal lesions, moderate ascites, normal portal vein flow, no intrahepatic or extrahepatic biliary duct dilation, normal kidney sizes, no hydronephrosis, and no intra-abdominal mass. Chest radiography is clear with no sign of consolidation, edema, or effusion. Echocardiography shows a normal left ventricular ejection fraction with no valvular disease or pericardial effusion. A random urine protein-creatinine ratio is normal at 0.1 (reference range < 0.2).

 

 

2. What is the most likely cause of her ascites based on the workup to this point?

  • Cirrhosis
  • Heart failure
  • Nephrotic syndrome
  • Portal vein thrombus
  • Abdominal malignancy
  • Malaria

lippert_new-onsetascites_f1.jpg
%3Cp%3EFigure%201.%20Interpreting%20the%20serum-ascites%20albumin%20gradient%20(SAAG)%20and%20ascitic%20protein%20levels.%3C%2Fp%3E

An initial approach to ascitic fluid analysis is to calculate the serum-ascites albumin gradient (SAAG). The SAAG is calculated as the serum albumin level minus the ascitic fluid albumin level.4,5 This is useful in determining the cause of the ascites (Figure 1).4,5 A gradient of 1.1 g/dL or higher indicates portal hypertension.4,5

Common causes of portal hypertension include cirrhosis, alcoholic hepatitis, heart failure, vascular occlusion syndromes (eg, Budd-Chiari syndrome, portal vein thrombosis), idiopathic portal fibrosis, and metastatic liver disease.5,6

If portal hypertension is present based on the SAAG, the next step is to review the ascitic protein level to help distinguish between a hepatic and a cardiac etiology of the ascites. An ascitic protein level less than 2.5 g/dL indicates a primary liver pathology (eg, cirrhosis). An ascitic protein level of 2.5 g/dL or greater typically indicates a cardiac condition (eg, heart failure, pericardial disease) with secondary congestive hepatopathy.5,6

If the SAAG is less than 1.1 g/dL, the ascites is likely not from portal hypertension. Typical causes of a low SAAG include infection, malignancy, pancreatic ascites, and nephrotic syndrome.5,6

In our patient, the SAAG is 1.35 g/dL (2.2 g/dL minus 0.85 g/dL), ie, elevated and due to portal hypertension. With an SAAG of 1.1 g/dL or greater and an ascitic fluid protein level less than 2.5 g/dL, as in our patient, the most likely cause is cirrhosis.

Heart failure is unlikely based on her normal brain natriuretic peptide level, an ascitic fluid protein level below 2.5 g/dL, and normal results on echocardiography. Nephrotic syndrome is also very unlikely based on the patient’s normal random urine protein-creatinine ratio. Portal vein thrombus and abdominal malignancy are essentially ruled out by the negative results of Doppler abdominal ultrasonography, with normal venous flow and no intra-abdominal mass and coupled with an elevated SAAG.

Although the patient has a history of travel, the incubation period for malaria would not fit the time frame of presentation. Also, she did not have typical malarial symptoms, her rapid malaria test was negative, and a peripheral blood smear for blood parasites was negative. It should be noted, however, that Plasmodium malariae infection classically presents with flulike symptoms and can resemble nephrotic syndrome, including peripheral edema, ascites, heavy proteinuria, hypoalbuminemia, and hyperlipidemia.7

3. In which patients is antibiotic prophylaxis against spontaneous bacterial peritonitis (SBP) appropriate?

  • Any patient with cirrhosis
  • Any patient with cirrhosis who is hospitalized
  • Any patient with cirrhosis and an ascitic fluid protein level below 2.0 g/dL
  • Any patient with cirrhosis and a history of SBP

Any patient with cirrhosis and a history of SBP should receive prophylactic antibiotics,8 as should any patient deemed at high risk of SBP. It is indicated in the following patients:

  • Patients with cirrhosis and gastrointestinal bleeding9,10
  • Patients with cirrhosis and a previous episode of SBP8
  • Patients with cirrhosis and an ascitic fluid protein level less than 1.5 g/dL with either impaired renal function (creatinine ≥ 1.2 mg/dL, blood urea nitrogen level ≥ 25 mg/dL, or serum sodium ≤ 130 mmol/L) or liver failure (Child-Pugh score ≥ 9 and a bilirubin ≥ 3 mg/dL)9
  • Patients with cirrhosis who are hospitalized for other reasons and have an ascitic protein level < 1.0 g/dL.9

Our patient has no signs or symptoms of gastrointestinal bleeding and no history of SBP. Her ascitic fluid protein level is 1.1 g/dL, and she has normal renal function. However, her Child-Pugh score is 12 (3 points for total bilirubin > 3 mg/dL, 3 points for serum albumin < 2.8 g/dL, 2 points for an INR 1.7 to 2.2, 3 points for moderate ascites, and 1 point for no encephalopathy), with a bilirubin of 17.0 mg/dL. Based on this, she is placed on antibiotic prophylaxis for SBP.

Our patient then undergoes an extensive workup for liver disease. Results of tests for toxins, autoimmune diseases, and inheritable diseases are all within normal limits. At this point, despite the patient’s reported negative alcohol history, our leading diagnosis is alcoholic hepatitis.

lippert_new-onsetascites_f2.jpg
%3Cp%3EFigure%202.%20Transjugular%20biopsy%20study%20(top)%20shows%20severe%20steatosis%20from%20lipid%20accumulation%20in%20hepatocytes%20(arrow)%20(hematoxylin%20and%20eosin%2C%20%C3%97%20100).%20Higher%20magnification%20(bottom)%20shows%20inflammatory%20cells%20(neutrophils%2C%20arrows)%20surrounding%20Mallory-Denk%20bodies%2C%20which%20are%20hyaline%20(eosinophilic)%20inclusions%20(hematoxylin%20and%20eosin%2C%20%C3%97%20400).%3C%2Fp%3E

To confirm this diagnosis, she subsequently undergoes transjugular liver biopsy, considered the gold standard for the diagnosis of alcoholic hepatitis. During the procedure, the hepatic venous pressure gradient is measured at 18 mm Hg (reference range 1–5 mm Hg), suggestive of portal hypertension. The pathology study shows severe fatty change, active steatohepatitis with ballooning degeneration, easily identifiable Mallory-Denk bodies, and prominent neutrophilic infiltration, as well as extensive bridging fibrosis (Figure 2). These findings point to alcoholic hepatitis.

After the biopsy results, we speak with the patient further about her alcohol habits. At this point, she informs us that she has consumed significant amounts of alcohol since the age of 18 (6 to 12 alcoholic beverages per day, including beer and hard liquor). Therefore, based on this new information, on her jaundice and ascites, and on results of laboratory testing and biopsy, we confirmed our diagnosis of alcoholic hepatitis.

 

 

4. When is drug treatment appropriate for alcoholic hepatitis?

  • Model for End-stage Liver Disease (MELD) score greater than 12
  • MELD score greater than 15
  • Maddrey Discriminant Function score greater than 25
  • Maddrey Discriminant Function score greater than 32
  • Glasgow score greater than 5
  • Glasgow score greater than 7

The best answer is a Maddrey Discriminant Function score greater than 32. A variety of scoring systems have been used to assess the severity of alcoholic hepatitis and to guide treatment, including the Maddrey Discriminant Function score, the MELD score, and the Glasgow score.11–16 They share similar laboratory values in their calculations, including prothrombin time (or INR) and total bilirubin.11–16 Typically, a Maddrey Discriminant Function score greater than 32, a Glasgow score of greater than 9, or a MELD score greater than 21 is used to determine whether pharmacologic treatment is indicated.11–16

lippert_new-onsetascites_f3.jpg
%3Cp%3EFigure%203.%20Algorithm%20for%20the%20management%20of%20alcoholic%20hepatitis.%3C%2Fp%3E

The typical treatment is prednisolone or pentoxifylline.11,17–21 The Lille score is designed to help decide whether to stop corticosteroids after 1 week of administration due to lack of treatment response.22 It predicts mortality rates within 6 months; a score of 0.45 or less indicates a good prognosis, and corticosteroid therapy should continue for 28 days (Figure 3).22

Our patient’s discriminant function score is 50, her Glasgow score is 10, and her MELD score is 28; thus, she begins treatment with oral prednisolone. Her Lille score at 1 week is 0.119, indicating a good prognosis, and her corticosteroids are continued for a total of 28 days.

It should be highlighted that the most important treatment is abstinence from alcohol.11 Recent literature suggests that any benefit of prednisolone or pentoxifylline in terms of mortality rates is questionable,19–20 and there is evidence that giving both drugs simultaneously may improve mortality rates,11,21 but the evidence remains conflicting at this time.

ALCOHOLIC HEPATITIS

Alcoholic hepatitis is a clinical syndrome of jaundice and liver failure, often in the setting of heavy alcohol use for decades.11,12 The incidence is unknown, but the typical age of presentation is between 40 and 50.11,12 The chief sign is a rapid onset of jaundice (< 3 months); common signs and symptoms include fever, ascites, proximal muscle loss, and an enlarged, tender liver.12 Encephalopathy may be seen in severe alcoholic hepatitis.12

Our patient is 35 years old. She has jaundice with rapid onset, as well as ascites and a tender liver.

The diagnosis of alcoholic hepatitis must take into account the patient’s history, physical examination, and laboratory findings. Until proven otherwise, the diagnosis should be presumed in the following scenario: ascites and jaundice on examination (usually with a duration < 3 months); a history of heavy alcohol use; neutrophilic leukocytosis; an AST level that is elevated but below 300 U/L; an ALT level above the normal range but below 300 U/L; an AST-ALT ratio greater than 2; a total serum bilirubin level above 5 mg/dL; and an elevated INR.11,12 Liver biopsy is the gold standard for diagnosis. Though not routinely done because of risks associated with the procedure, it may help confirm the diagnosis if it is in question.

CASE CONCLUDED

We start our patient on oral prednisolone 40 mg daily for alcoholic hepatitis. Her symptoms and laboratory testing results including bilirubin improve. Her Lille score at 7 days indicates a good prognosis, prompting continuation of corticosteroid treatment for the full 28 days.

She is referred to an outpatient alcohol rehabilitation program and has remained sober as of the last outpatient note.

Alcoholic hepatitis is extremely difficult to diagnose, and no single blood test or imaging study confirms the diagnosis. The history, physical examination findings, and laboratory findings are crucial. If the diagnosis is still in doubt, liver biopsy may help confirm the diagnosis.

References
  1. Ruyon BA; AASLD Practice Guidelines Committee. Management of adult patients with ascites due to cirrhosis: an update. Hepatology 2009; 49(6):2087–2107. doi:10.1002/hep.22853
  2. Hoefs JC, Canawati HN, Sapico FL, Hopkins RR, Weiner J, Montgomerie JZ. Spontaneous bacterial peritonitis. Hepatology 1982; 2(4):399–407. pmid:7095741
  3. Ginès P, Cárdenas A, Arroyo V, Rodés J. Management of cirrhosis and ascites. N Engl J Med 2004; 350(16):1646–1654. doi:10.1056/NEJMra035021
  4. Runyon BA, Montano AA, Akriviadis EA, Antillon MR, Irving MA, McHutchison JG. The serum-ascites albumin gradient is superior to the exudate-transudate concept in the differential diagnosis of ascites. Ann Intern Med 1992; 117(3):215–220. pmid:1616215
  5. Hernaez R, Hamilton JP. Unexplained ascites. Clin Liver Dis 2016; 7(3):53–56. https://aasldpubs.onlinelibrary.wiley.com/doi/epdf/10.1002/cld.537
  6. Huang LL, Xia HH, Zhu SL. Ascitic fluid analysis in the differential diagnosis of ascites: focus on cirrhotic ascites. J Clin Transl Hepatol 2014; 2(1):58–64. doi:10.14218/JCTH.2013.00010
  7. Bartoloni A, Zammarchi L. Clinical aspects of uncomplicated and severe malaria. Mediterr J Hematol Infect Dis 2012; 4(1):e2012026. doi:10.4084/MJHID.2012.026
  8. Titó L, Rimola A, Ginès P, Llach J, Arroyo V, Rodés J. Recurrence of spontaneous bacterial peritonitis in cirrhosis: frequency and predictive factors. Hepatology 1988; 8(1):27–31. pmid:3257456
  9. Fernández J, Ruiz del Arbol L, Gómez C, et al. Norfloxacin vs ceftriaxone in the prophylaxis of infections in patients with advanced cirrhosis and hemorrhage. Gastroenterology 2006; 131(4):1049–1056. doi:10.1053/j.gastro.2006.07.010
  10. Runyon B; The American Association for the Study of Liver Diseases (AASLD). Management of adult patients with ascites due to cirrhosis: update 2012. https://www.aasld.org/sites/default/files/guideline_documents/141020_Guideline_Ascites_4UFb_2015.pdf. Accessed September 4, 2018.
  11. Sidhu SS, Goyal O, Kishore H, Sidhu S. New paradigms in management of alcoholic hepatitis: a review. Hepatol Int 2017; 11(3):255–267. doi:10.1007/s12072-017-9790-5
  12. Lucey MR, Mathurin P, Morgan TR. Alcoholic hepatitis. N Engl J Med 2009; 360(26):2758–2769. doi:10.1056/NEJMra0805786
  13. Maddrey WC, Boitnott JK, Bedine MS, Weber FL Jr, Mezey E, White RI Jr. Corticosteroid therapy of alcoholic hepatitis. Gastroenterology 1978; 75(2):193–199. pmid:352788
  14. Forrest EH, Evans CD, Stewart S, et al. Analysis of factors predictive of mortality in alcoholic hepatitis and derivation and validation of the Glasgow alcoholic hepatitis score. Gut 2005; 54(8):1174–1179. doi:10.1136/gut.2004.050781
  15. Dunn W, Jamil LH, Brown LS, et al. MELD accurately predicts mortality in patients with alcoholic hepatitis. Hepatology 2005; 41(2):353–358. doi:10.1002/hep.20503
  16. Sheth M, Riggs M, Patel T. Utility of the Mayo end-stage liver disease (MELD) score in assessing prognosis of patients with alcoholic hepatitis. BMC Gastroenterol 2002; 2:2. pmid:11835693
  17. Akriviadis E, Botla R, Briggs W, Han S, Reynolds T, Shakil O. Pentoxifylline improves short-term survival in severe acute alcoholic hepatitis: a double-blind, placebo-controlled trial. Gastroenterology 2000; 119(6):1637–1648. pmid:11113085
  18. Mathurin P, O’Grady J, Carithers RL, et al. Corticosteroids improve short-term survival in patients with severe alcoholic hepatitis: meta-analysis of individual patient data. Gut 2011; 60(2):255–260. doi:10.1136/gut.2010.224097
  19. Thursz MR, Richardson P, Allison M, et al; STOPAH Trial. Prednisolone or pentoxifylline for alcoholic hepatitis. N Engl J Med 2015; 372(17):1619–1628. doi:10.1056/NEJMoa1412278
  20. Thursz M, Forrest E, Roderick P, et al. The clinical effectiveness and cost-effectiveness of steroids or pentoxifylline for alcoholic hepatitis (STOPAH): a 2 × 2 factorial randomised controlled trial. Health Technol Assess 2015; 19(102):1–104. doi:10.3310/hta191020
  21. Lee YS, Kim HJ, Kim JH, et al. Treatment of severe alcoholic hepatitis with corticosteroid, pentoxifylline, or dual therapy: a systematic review and meta-analysis. J Clin Gastroenterol 2017; 51(4):364–377. doi:10.1097/MCG.0000000000000674
  22. Louvet A, Naveau S, Abdelnour M, et al. The Lille model: a new tool for therapeutic strategy in patients with severe alcoholic hepatitis treated with steroids. Hepatology 2007; 45(6):1348–1354. doi:10.1002/hep.21607
Article PDF
Author and Disclosure Information

William C. Lippert, MD, MPH
Instructor of Internal Medicine, Department of Internal Medicine, Section of Hospital Medicine, Wake Forest Baptist Medical Center, Winston-Salem, NC

Eun Y. Lee, MD
Professor and Director of Anatomic Pathology, Department of Pathology and Laboratory Medicine, University of Kentucky Medical Center, Lexington, KY

Aibek E. Mirrakhimov, MD
Assistant Professor of Medicine, Division of Hospital Medicine, University of Kentucky Medical Center, Lexington, KY

Address: William C. Lippert, MD, MPH, Section of Hospital Medicine, Department of Internal Medicine, Wake Forest Baptist Medical Center, 1 Medical Center Boulevard, Winston-Salem, NC 27157;
wlippert@wakehealth.edu

Issue
Cleveland Clinic Journal of Medicine - 86(4)
Publications
Topics
Page Number
257-262
Legacy Keywords
ascites, jaundice, cirrhosis, portal hypertension, liver function tests, alanine aminotransferase, ALT, aspartate aminotransferase, AST, hepatitis, serum-ascites albumin gradient, SAAG, alcoholism, steatosis, liver biopsy, Model for End-stage Liver Disease score, MELD, Maddrey Discriminant Function score, DF score, alcoholic hepatitis, William Lippert, Eun Lee, Aibek Mirrakhimov
Sections
Author and Disclosure Information

William C. Lippert, MD, MPH
Instructor of Internal Medicine, Department of Internal Medicine, Section of Hospital Medicine, Wake Forest Baptist Medical Center, Winston-Salem, NC

Eun Y. Lee, MD
Professor and Director of Anatomic Pathology, Department of Pathology and Laboratory Medicine, University of Kentucky Medical Center, Lexington, KY

Aibek E. Mirrakhimov, MD
Assistant Professor of Medicine, Division of Hospital Medicine, University of Kentucky Medical Center, Lexington, KY

Address: William C. Lippert, MD, MPH, Section of Hospital Medicine, Department of Internal Medicine, Wake Forest Baptist Medical Center, 1 Medical Center Boulevard, Winston-Salem, NC 27157;
wlippert@wakehealth.edu

Author and Disclosure Information

William C. Lippert, MD, MPH
Instructor of Internal Medicine, Department of Internal Medicine, Section of Hospital Medicine, Wake Forest Baptist Medical Center, Winston-Salem, NC

Eun Y. Lee, MD
Professor and Director of Anatomic Pathology, Department of Pathology and Laboratory Medicine, University of Kentucky Medical Center, Lexington, KY

Aibek E. Mirrakhimov, MD
Assistant Professor of Medicine, Division of Hospital Medicine, University of Kentucky Medical Center, Lexington, KY

Address: William C. Lippert, MD, MPH, Section of Hospital Medicine, Department of Internal Medicine, Wake Forest Baptist Medical Center, 1 Medical Center Boulevard, Winston-Salem, NC 27157;
wlippert@wakehealth.edu

Article PDF
Article PDF
Related Articles

A 35-year-old woman is admitted to the hospital with a 5-day history of abdominal distention and jaundice. She reports no history of fever, chills, night sweats, abdominal pain, nausea, vomiting, diarrhea, changes in urine color, change in stool color, weight loss, weight gain, or loss of appetite.

She is petite, with a body mass index of 19.4 kg/m2. She has no known history of medical conditions or surgery and is not taking any medications. Her family history is unremarkable, and she denies current or past tobacco, alcohol, or illicit drug use.

RECENT TRAVEL

She says that during a trip to Central America several months ago, she had suffered a seizure and was taken to a local hospital, where laboratory testing revealed elevated aspartate aminotransferase (AST) and alanine aminotransferase (ALT) levels. She says that the rest of the workup at that time was normal.

About 1 week after that incident, she returned home and saw her primary care physician, who ordered further testing, which showed mild hyperbilirubinemia and mild elevation of AST and ALT levels. Her physician attributed the elevations to atovaquone, which she had been taking for malaria prophylaxis, as repeat testing 2 weeks later showed improvement in AST and ALT levels.

The patient says she returned to her normal state of health until about 5 days ago, when she noticed jaundice and abdominal distention, but without abdominal pain, dark urine, or clay-colored stools. She became concerned and went to her local hospital. Testing there noted mild elevation of AST and ALT, as well as an elevated international normalized ratio (INR) and hyperbilirubinemia. Computed tomography of the abdomen and pelvis showed hepatomegaly with possible fatty liver. Because of these results, the patient was transferred to our institution for further evaluation.

EVALUATION AT OUR INSTITUTION

On examination at our institution, she is afebrile, and vital signs are within normal ranges. She has bilateral scleral icterus and diffuse jaundice, but no other skin finding such as rash or spider angioma. She has no lymphadenopathy. Her abdomen is distended, with tense ascites, and her liver is tender to palpation. The tip of the spleen is not palpable.

lippert_new-onsetascites_t1.jpg

The cardiovascular examination reveals no murmurs, rubs, or gallops, but she has jugular venous distention and +2 pitting edema of both lower extremities.

On respiratory examination, there is dullness to percussion, with slight crackles on auscultation at the right lung base. The neurologic examination is normal.

Table 1 shows the results of initial laboratory testing.

1. Which study would provide the most information on the cause of ascites?

  • Abdominal ultrasonography
  • Abdominal paracentesis with ascitic fluid analysis
  • Chest radiography
  • Echocardiography
  • Urine protein-to-creatinine ratio

Abdominal paracentesis with ascitic fluid analysis is the essential study for any patient with clinically apparent new-onset ascites.1–3 It is the study that provides the most information on the cause of ascites.

In our patient, abdominal paracentesis yields 1,000 mL of straw-colored ascitic fluid, and analysis shows 86 nucleated cells, 28 of which are polymorphonuclear cells, and 0 red blood cells, with negative Gram stain and culture. The ascitic albumin level is 0.85 g/dL, with an ascitic protein of 1.1 g/dL.

Abdominal ultrasonography shows a diffusely echogenic liver, no focal lesions, moderate ascites, normal portal vein flow, no intrahepatic or extrahepatic biliary duct dilation, normal kidney sizes, no hydronephrosis, and no intra-abdominal mass. Chest radiography is clear with no sign of consolidation, edema, or effusion. Echocardiography shows a normal left ventricular ejection fraction with no valvular disease or pericardial effusion. A random urine protein-creatinine ratio is normal at 0.1 (reference range < 0.2).

 

 

2. What is the most likely cause of her ascites based on the workup to this point?

  • Cirrhosis
  • Heart failure
  • Nephrotic syndrome
  • Portal vein thrombus
  • Abdominal malignancy
  • Malaria

lippert_new-onsetascites_f1.jpg
%3Cp%3EFigure%201.%20Interpreting%20the%20serum-ascites%20albumin%20gradient%20(SAAG)%20and%20ascitic%20protein%20levels.%3C%2Fp%3E

An initial approach to ascitic fluid analysis is to calculate the serum-ascites albumin gradient (SAAG). The SAAG is calculated as the serum albumin level minus the ascitic fluid albumin level.4,5 This is useful in determining the cause of the ascites (Figure 1).4,5 A gradient of 1.1 g/dL or higher indicates portal hypertension.4,5

Common causes of portal hypertension include cirrhosis, alcoholic hepatitis, heart failure, vascular occlusion syndromes (eg, Budd-Chiari syndrome, portal vein thrombosis), idiopathic portal fibrosis, and metastatic liver disease.5,6

If portal hypertension is present based on the SAAG, the next step is to review the ascitic protein level to help distinguish between a hepatic and a cardiac etiology of the ascites. An ascitic protein level less than 2.5 g/dL indicates a primary liver pathology (eg, cirrhosis). An ascitic protein level of 2.5 g/dL or greater typically indicates a cardiac condition (eg, heart failure, pericardial disease) with secondary congestive hepatopathy.5,6

If the SAAG is less than 1.1 g/dL, the ascites is likely not from portal hypertension. Typical causes of a low SAAG include infection, malignancy, pancreatic ascites, and nephrotic syndrome.5,6

In our patient, the SAAG is 1.35 g/dL (2.2 g/dL minus 0.85 g/dL), ie, elevated and due to portal hypertension. With an SAAG of 1.1 g/dL or greater and an ascitic fluid protein level less than 2.5 g/dL, as in our patient, the most likely cause is cirrhosis.

Heart failure is unlikely based on her normal brain natriuretic peptide level, an ascitic fluid protein level below 2.5 g/dL, and normal results on echocardiography. Nephrotic syndrome is also very unlikely based on the patient’s normal random urine protein-creatinine ratio. Portal vein thrombus and abdominal malignancy are essentially ruled out by the negative results of Doppler abdominal ultrasonography, with normal venous flow and no intra-abdominal mass and coupled with an elevated SAAG.

Although the patient has a history of travel, the incubation period for malaria would not fit the time frame of presentation. Also, she did not have typical malarial symptoms, her rapid malaria test was negative, and a peripheral blood smear for blood parasites was negative. It should be noted, however, that Plasmodium malariae infection classically presents with flulike symptoms and can resemble nephrotic syndrome, including peripheral edema, ascites, heavy proteinuria, hypoalbuminemia, and hyperlipidemia.7

3. In which patients is antibiotic prophylaxis against spontaneous bacterial peritonitis (SBP) appropriate?

  • Any patient with cirrhosis
  • Any patient with cirrhosis who is hospitalized
  • Any patient with cirrhosis and an ascitic fluid protein level below 2.0 g/dL
  • Any patient with cirrhosis and a history of SBP

Any patient with cirrhosis and a history of SBP should receive prophylactic antibiotics,8 as should any patient deemed at high risk of SBP. It is indicated in the following patients:

  • Patients with cirrhosis and gastrointestinal bleeding9,10
  • Patients with cirrhosis and a previous episode of SBP8
  • Patients with cirrhosis and an ascitic fluid protein level less than 1.5 g/dL with either impaired renal function (creatinine ≥ 1.2 mg/dL, blood urea nitrogen level ≥ 25 mg/dL, or serum sodium ≤ 130 mmol/L) or liver failure (Child-Pugh score ≥ 9 and a bilirubin ≥ 3 mg/dL)9
  • Patients with cirrhosis who are hospitalized for other reasons and have an ascitic protein level < 1.0 g/dL.9

Our patient has no signs or symptoms of gastrointestinal bleeding and no history of SBP. Her ascitic fluid protein level is 1.1 g/dL, and she has normal renal function. However, her Child-Pugh score is 12 (3 points for total bilirubin > 3 mg/dL, 3 points for serum albumin < 2.8 g/dL, 2 points for an INR 1.7 to 2.2, 3 points for moderate ascites, and 1 point for no encephalopathy), with a bilirubin of 17.0 mg/dL. Based on this, she is placed on antibiotic prophylaxis for SBP.

Our patient then undergoes an extensive workup for liver disease. Results of tests for toxins, autoimmune diseases, and inheritable diseases are all within normal limits. At this point, despite the patient’s reported negative alcohol history, our leading diagnosis is alcoholic hepatitis.

lippert_new-onsetascites_f2.jpg
%3Cp%3EFigure%202.%20Transjugular%20biopsy%20study%20(top)%20shows%20severe%20steatosis%20from%20lipid%20accumulation%20in%20hepatocytes%20(arrow)%20(hematoxylin%20and%20eosin%2C%20%C3%97%20100).%20Higher%20magnification%20(bottom)%20shows%20inflammatory%20cells%20(neutrophils%2C%20arrows)%20surrounding%20Mallory-Denk%20bodies%2C%20which%20are%20hyaline%20(eosinophilic)%20inclusions%20(hematoxylin%20and%20eosin%2C%20%C3%97%20400).%3C%2Fp%3E

To confirm this diagnosis, she subsequently undergoes transjugular liver biopsy, considered the gold standard for the diagnosis of alcoholic hepatitis. During the procedure, the hepatic venous pressure gradient is measured at 18 mm Hg (reference range 1–5 mm Hg), suggestive of portal hypertension. The pathology study shows severe fatty change, active steatohepatitis with ballooning degeneration, easily identifiable Mallory-Denk bodies, and prominent neutrophilic infiltration, as well as extensive bridging fibrosis (Figure 2). These findings point to alcoholic hepatitis.

After the biopsy results, we speak with the patient further about her alcohol habits. At this point, she informs us that she has consumed significant amounts of alcohol since the age of 18 (6 to 12 alcoholic beverages per day, including beer and hard liquor). Therefore, based on this new information, on her jaundice and ascites, and on results of laboratory testing and biopsy, we confirmed our diagnosis of alcoholic hepatitis.

 

 

4. When is drug treatment appropriate for alcoholic hepatitis?

  • Model for End-stage Liver Disease (MELD) score greater than 12
  • MELD score greater than 15
  • Maddrey Discriminant Function score greater than 25
  • Maddrey Discriminant Function score greater than 32
  • Glasgow score greater than 5
  • Glasgow score greater than 7

The best answer is a Maddrey Discriminant Function score greater than 32. A variety of scoring systems have been used to assess the severity of alcoholic hepatitis and to guide treatment, including the Maddrey Discriminant Function score, the MELD score, and the Glasgow score.11–16 They share similar laboratory values in their calculations, including prothrombin time (or INR) and total bilirubin.11–16 Typically, a Maddrey Discriminant Function score greater than 32, a Glasgow score of greater than 9, or a MELD score greater than 21 is used to determine whether pharmacologic treatment is indicated.11–16

lippert_new-onsetascites_f3.jpg
%3Cp%3EFigure%203.%20Algorithm%20for%20the%20management%20of%20alcoholic%20hepatitis.%3C%2Fp%3E

The typical treatment is prednisolone or pentoxifylline.11,17–21 The Lille score is designed to help decide whether to stop corticosteroids after 1 week of administration due to lack of treatment response.22 It predicts mortality rates within 6 months; a score of 0.45 or less indicates a good prognosis, and corticosteroid therapy should continue for 28 days (Figure 3).22

Our patient’s discriminant function score is 50, her Glasgow score is 10, and her MELD score is 28; thus, she begins treatment with oral prednisolone. Her Lille score at 1 week is 0.119, indicating a good prognosis, and her corticosteroids are continued for a total of 28 days.

It should be highlighted that the most important treatment is abstinence from alcohol.11 Recent literature suggests that any benefit of prednisolone or pentoxifylline in terms of mortality rates is questionable,19–20 and there is evidence that giving both drugs simultaneously may improve mortality rates,11,21 but the evidence remains conflicting at this time.

ALCOHOLIC HEPATITIS

Alcoholic hepatitis is a clinical syndrome of jaundice and liver failure, often in the setting of heavy alcohol use for decades.11,12 The incidence is unknown, but the typical age of presentation is between 40 and 50.11,12 The chief sign is a rapid onset of jaundice (< 3 months); common signs and symptoms include fever, ascites, proximal muscle loss, and an enlarged, tender liver.12 Encephalopathy may be seen in severe alcoholic hepatitis.12

Our patient is 35 years old. She has jaundice with rapid onset, as well as ascites and a tender liver.

The diagnosis of alcoholic hepatitis must take into account the patient’s history, physical examination, and laboratory findings. Until proven otherwise, the diagnosis should be presumed in the following scenario: ascites and jaundice on examination (usually with a duration < 3 months); a history of heavy alcohol use; neutrophilic leukocytosis; an AST level that is elevated but below 300 U/L; an ALT level above the normal range but below 300 U/L; an AST-ALT ratio greater than 2; a total serum bilirubin level above 5 mg/dL; and an elevated INR.11,12 Liver biopsy is the gold standard for diagnosis. Though not routinely done because of risks associated with the procedure, it may help confirm the diagnosis if it is in question.

CASE CONCLUDED

We start our patient on oral prednisolone 40 mg daily for alcoholic hepatitis. Her symptoms and laboratory testing results including bilirubin improve. Her Lille score at 7 days indicates a good prognosis, prompting continuation of corticosteroid treatment for the full 28 days.

She is referred to an outpatient alcohol rehabilitation program and has remained sober as of the last outpatient note.

Alcoholic hepatitis is extremely difficult to diagnose, and no single blood test or imaging study confirms the diagnosis. The history, physical examination findings, and laboratory findings are crucial. If the diagnosis is still in doubt, liver biopsy may help confirm the diagnosis.

A 35-year-old woman is admitted to the hospital with a 5-day history of abdominal distention and jaundice. She reports no history of fever, chills, night sweats, abdominal pain, nausea, vomiting, diarrhea, changes in urine color, change in stool color, weight loss, weight gain, or loss of appetite.

She is petite, with a body mass index of 19.4 kg/m2. She has no known history of medical conditions or surgery and is not taking any medications. Her family history is unremarkable, and she denies current or past tobacco, alcohol, or illicit drug use.

RECENT TRAVEL

She says that during a trip to Central America several months ago, she had suffered a seizure and was taken to a local hospital, where laboratory testing revealed elevated aspartate aminotransferase (AST) and alanine aminotransferase (ALT) levels. She says that the rest of the workup at that time was normal.

About 1 week after that incident, she returned home and saw her primary care physician, who ordered further testing, which showed mild hyperbilirubinemia and mild elevation of AST and ALT levels. Her physician attributed the elevations to atovaquone, which she had been taking for malaria prophylaxis, as repeat testing 2 weeks later showed improvement in AST and ALT levels.

The patient says she returned to her normal state of health until about 5 days ago, when she noticed jaundice and abdominal distention, but without abdominal pain, dark urine, or clay-colored stools. She became concerned and went to her local hospital. Testing there noted mild elevation of AST and ALT, as well as an elevated international normalized ratio (INR) and hyperbilirubinemia. Computed tomography of the abdomen and pelvis showed hepatomegaly with possible fatty liver. Because of these results, the patient was transferred to our institution for further evaluation.

EVALUATION AT OUR INSTITUTION

On examination at our institution, she is afebrile, and vital signs are within normal ranges. She has bilateral scleral icterus and diffuse jaundice, but no other skin finding such as rash or spider angioma. She has no lymphadenopathy. Her abdomen is distended, with tense ascites, and her liver is tender to palpation. The tip of the spleen is not palpable.

lippert_new-onsetascites_t1.jpg

The cardiovascular examination reveals no murmurs, rubs, or gallops, but she has jugular venous distention and +2 pitting edema of both lower extremities.

On respiratory examination, there is dullness to percussion, with slight crackles on auscultation at the right lung base. The neurologic examination is normal.

Table 1 shows the results of initial laboratory testing.

1. Which study would provide the most information on the cause of ascites?

  • Abdominal ultrasonography
  • Abdominal paracentesis with ascitic fluid analysis
  • Chest radiography
  • Echocardiography
  • Urine protein-to-creatinine ratio

Abdominal paracentesis with ascitic fluid analysis is the essential study for any patient with clinically apparent new-onset ascites.1–3 It is the study that provides the most information on the cause of ascites.

In our patient, abdominal paracentesis yields 1,000 mL of straw-colored ascitic fluid, and analysis shows 86 nucleated cells, 28 of which are polymorphonuclear cells, and 0 red blood cells, with negative Gram stain and culture. The ascitic albumin level is 0.85 g/dL, with an ascitic protein of 1.1 g/dL.

Abdominal ultrasonography shows a diffusely echogenic liver, no focal lesions, moderate ascites, normal portal vein flow, no intrahepatic or extrahepatic biliary duct dilation, normal kidney sizes, no hydronephrosis, and no intra-abdominal mass. Chest radiography is clear with no sign of consolidation, edema, or effusion. Echocardiography shows a normal left ventricular ejection fraction with no valvular disease or pericardial effusion. A random urine protein-creatinine ratio is normal at 0.1 (reference range < 0.2).

 

 

2. What is the most likely cause of her ascites based on the workup to this point?

  • Cirrhosis
  • Heart failure
  • Nephrotic syndrome
  • Portal vein thrombus
  • Abdominal malignancy
  • Malaria

lippert_new-onsetascites_f1.jpg
%3Cp%3EFigure%201.%20Interpreting%20the%20serum-ascites%20albumin%20gradient%20(SAAG)%20and%20ascitic%20protein%20levels.%3C%2Fp%3E

An initial approach to ascitic fluid analysis is to calculate the serum-ascites albumin gradient (SAAG). The SAAG is calculated as the serum albumin level minus the ascitic fluid albumin level.4,5 This is useful in determining the cause of the ascites (Figure 1).4,5 A gradient of 1.1 g/dL or higher indicates portal hypertension.4,5

Common causes of portal hypertension include cirrhosis, alcoholic hepatitis, heart failure, vascular occlusion syndromes (eg, Budd-Chiari syndrome, portal vein thrombosis), idiopathic portal fibrosis, and metastatic liver disease.5,6

If portal hypertension is present based on the SAAG, the next step is to review the ascitic protein level to help distinguish between a hepatic and a cardiac etiology of the ascites. An ascitic protein level less than 2.5 g/dL indicates a primary liver pathology (eg, cirrhosis). An ascitic protein level of 2.5 g/dL or greater typically indicates a cardiac condition (eg, heart failure, pericardial disease) with secondary congestive hepatopathy.5,6

If the SAAG is less than 1.1 g/dL, the ascites is likely not from portal hypertension. Typical causes of a low SAAG include infection, malignancy, pancreatic ascites, and nephrotic syndrome.5,6

In our patient, the SAAG is 1.35 g/dL (2.2 g/dL minus 0.85 g/dL), ie, elevated and due to portal hypertension. With an SAAG of 1.1 g/dL or greater and an ascitic fluid protein level less than 2.5 g/dL, as in our patient, the most likely cause is cirrhosis.

Heart failure is unlikely based on her normal brain natriuretic peptide level, an ascitic fluid protein level below 2.5 g/dL, and normal results on echocardiography. Nephrotic syndrome is also very unlikely based on the patient’s normal random urine protein-creatinine ratio. Portal vein thrombus and abdominal malignancy are essentially ruled out by the negative results of Doppler abdominal ultrasonography, with normal venous flow and no intra-abdominal mass and coupled with an elevated SAAG.

Although the patient has a history of travel, the incubation period for malaria would not fit the time frame of presentation. Also, she did not have typical malarial symptoms, her rapid malaria test was negative, and a peripheral blood smear for blood parasites was negative. It should be noted, however, that Plasmodium malariae infection classically presents with flulike symptoms and can resemble nephrotic syndrome, including peripheral edema, ascites, heavy proteinuria, hypoalbuminemia, and hyperlipidemia.7

3. In which patients is antibiotic prophylaxis against spontaneous bacterial peritonitis (SBP) appropriate?

  • Any patient with cirrhosis
  • Any patient with cirrhosis who is hospitalized
  • Any patient with cirrhosis and an ascitic fluid protein level below 2.0 g/dL
  • Any patient with cirrhosis and a history of SBP

Any patient with cirrhosis and a history of SBP should receive prophylactic antibiotics,8 as should any patient deemed at high risk of SBP. It is indicated in the following patients:

  • Patients with cirrhosis and gastrointestinal bleeding9,10
  • Patients with cirrhosis and a previous episode of SBP8
  • Patients with cirrhosis and an ascitic fluid protein level less than 1.5 g/dL with either impaired renal function (creatinine ≥ 1.2 mg/dL, blood urea nitrogen level ≥ 25 mg/dL, or serum sodium ≤ 130 mmol/L) or liver failure (Child-Pugh score ≥ 9 and a bilirubin ≥ 3 mg/dL)9
  • Patients with cirrhosis who are hospitalized for other reasons and have an ascitic protein level < 1.0 g/dL.9

Our patient has no signs or symptoms of gastrointestinal bleeding and no history of SBP. Her ascitic fluid protein level is 1.1 g/dL, and she has normal renal function. However, her Child-Pugh score is 12 (3 points for total bilirubin > 3 mg/dL, 3 points for serum albumin < 2.8 g/dL, 2 points for an INR 1.7 to 2.2, 3 points for moderate ascites, and 1 point for no encephalopathy), with a bilirubin of 17.0 mg/dL. Based on this, she is placed on antibiotic prophylaxis for SBP.

Our patient then undergoes an extensive workup for liver disease. Results of tests for toxins, autoimmune diseases, and inheritable diseases are all within normal limits. At this point, despite the patient’s reported negative alcohol history, our leading diagnosis is alcoholic hepatitis.

lippert_new-onsetascites_f2.jpg
%3Cp%3EFigure%202.%20Transjugular%20biopsy%20study%20(top)%20shows%20severe%20steatosis%20from%20lipid%20accumulation%20in%20hepatocytes%20(arrow)%20(hematoxylin%20and%20eosin%2C%20%C3%97%20100).%20Higher%20magnification%20(bottom)%20shows%20inflammatory%20cells%20(neutrophils%2C%20arrows)%20surrounding%20Mallory-Denk%20bodies%2C%20which%20are%20hyaline%20(eosinophilic)%20inclusions%20(hematoxylin%20and%20eosin%2C%20%C3%97%20400).%3C%2Fp%3E

To confirm this diagnosis, she subsequently undergoes transjugular liver biopsy, considered the gold standard for the diagnosis of alcoholic hepatitis. During the procedure, the hepatic venous pressure gradient is measured at 18 mm Hg (reference range 1–5 mm Hg), suggestive of portal hypertension. The pathology study shows severe fatty change, active steatohepatitis with ballooning degeneration, easily identifiable Mallory-Denk bodies, and prominent neutrophilic infiltration, as well as extensive bridging fibrosis (Figure 2). These findings point to alcoholic hepatitis.

After the biopsy results, we speak with the patient further about her alcohol habits. At this point, she informs us that she has consumed significant amounts of alcohol since the age of 18 (6 to 12 alcoholic beverages per day, including beer and hard liquor). Therefore, based on this new information, on her jaundice and ascites, and on results of laboratory testing and biopsy, we confirmed our diagnosis of alcoholic hepatitis.

 

 

4. When is drug treatment appropriate for alcoholic hepatitis?

  • Model for End-stage Liver Disease (MELD) score greater than 12
  • MELD score greater than 15
  • Maddrey Discriminant Function score greater than 25
  • Maddrey Discriminant Function score greater than 32
  • Glasgow score greater than 5
  • Glasgow score greater than 7

The best answer is a Maddrey Discriminant Function score greater than 32. A variety of scoring systems have been used to assess the severity of alcoholic hepatitis and to guide treatment, including the Maddrey Discriminant Function score, the MELD score, and the Glasgow score.11–16 They share similar laboratory values in their calculations, including prothrombin time (or INR) and total bilirubin.11–16 Typically, a Maddrey Discriminant Function score greater than 32, a Glasgow score of greater than 9, or a MELD score greater than 21 is used to determine whether pharmacologic treatment is indicated.11–16

lippert_new-onsetascites_f3.jpg
%3Cp%3EFigure%203.%20Algorithm%20for%20the%20management%20of%20alcoholic%20hepatitis.%3C%2Fp%3E

The typical treatment is prednisolone or pentoxifylline.11,17–21 The Lille score is designed to help decide whether to stop corticosteroids after 1 week of administration due to lack of treatment response.22 It predicts mortality rates within 6 months; a score of 0.45 or less indicates a good prognosis, and corticosteroid therapy should continue for 28 days (Figure 3).22

Our patient’s discriminant function score is 50, her Glasgow score is 10, and her MELD score is 28; thus, she begins treatment with oral prednisolone. Her Lille score at 1 week is 0.119, indicating a good prognosis, and her corticosteroids are continued for a total of 28 days.

It should be highlighted that the most important treatment is abstinence from alcohol.11 Recent literature suggests that any benefit of prednisolone or pentoxifylline in terms of mortality rates is questionable,19–20 and there is evidence that giving both drugs simultaneously may improve mortality rates,11,21 but the evidence remains conflicting at this time.

ALCOHOLIC HEPATITIS

Alcoholic hepatitis is a clinical syndrome of jaundice and liver failure, often in the setting of heavy alcohol use for decades.11,12 The incidence is unknown, but the typical age of presentation is between 40 and 50.11,12 The chief sign is a rapid onset of jaundice (< 3 months); common signs and symptoms include fever, ascites, proximal muscle loss, and an enlarged, tender liver.12 Encephalopathy may be seen in severe alcoholic hepatitis.12

Our patient is 35 years old. She has jaundice with rapid onset, as well as ascites and a tender liver.

The diagnosis of alcoholic hepatitis must take into account the patient’s history, physical examination, and laboratory findings. Until proven otherwise, the diagnosis should be presumed in the following scenario: ascites and jaundice on examination (usually with a duration < 3 months); a history of heavy alcohol use; neutrophilic leukocytosis; an AST level that is elevated but below 300 U/L; an ALT level above the normal range but below 300 U/L; an AST-ALT ratio greater than 2; a total serum bilirubin level above 5 mg/dL; and an elevated INR.11,12 Liver biopsy is the gold standard for diagnosis. Though not routinely done because of risks associated with the procedure, it may help confirm the diagnosis if it is in question.

CASE CONCLUDED

We start our patient on oral prednisolone 40 mg daily for alcoholic hepatitis. Her symptoms and laboratory testing results including bilirubin improve. Her Lille score at 7 days indicates a good prognosis, prompting continuation of corticosteroid treatment for the full 28 days.

She is referred to an outpatient alcohol rehabilitation program and has remained sober as of the last outpatient note.

Alcoholic hepatitis is extremely difficult to diagnose, and no single blood test or imaging study confirms the diagnosis. The history, physical examination findings, and laboratory findings are crucial. If the diagnosis is still in doubt, liver biopsy may help confirm the diagnosis.

References
  1. Ruyon BA; AASLD Practice Guidelines Committee. Management of adult patients with ascites due to cirrhosis: an update. Hepatology 2009; 49(6):2087–2107. doi:10.1002/hep.22853
  2. Hoefs JC, Canawati HN, Sapico FL, Hopkins RR, Weiner J, Montgomerie JZ. Spontaneous bacterial peritonitis. Hepatology 1982; 2(4):399–407. pmid:7095741
  3. Ginès P, Cárdenas A, Arroyo V, Rodés J. Management of cirrhosis and ascites. N Engl J Med 2004; 350(16):1646–1654. doi:10.1056/NEJMra035021
  4. Runyon BA, Montano AA, Akriviadis EA, Antillon MR, Irving MA, McHutchison JG. The serum-ascites albumin gradient is superior to the exudate-transudate concept in the differential diagnosis of ascites. Ann Intern Med 1992; 117(3):215–220. pmid:1616215
  5. Hernaez R, Hamilton JP. Unexplained ascites. Clin Liver Dis 2016; 7(3):53–56. https://aasldpubs.onlinelibrary.wiley.com/doi/epdf/10.1002/cld.537
  6. Huang LL, Xia HH, Zhu SL. Ascitic fluid analysis in the differential diagnosis of ascites: focus on cirrhotic ascites. J Clin Transl Hepatol 2014; 2(1):58–64. doi:10.14218/JCTH.2013.00010
  7. Bartoloni A, Zammarchi L. Clinical aspects of uncomplicated and severe malaria. Mediterr J Hematol Infect Dis 2012; 4(1):e2012026. doi:10.4084/MJHID.2012.026
  8. Titó L, Rimola A, Ginès P, Llach J, Arroyo V, Rodés J. Recurrence of spontaneous bacterial peritonitis in cirrhosis: frequency and predictive factors. Hepatology 1988; 8(1):27–31. pmid:3257456
  9. Fernández J, Ruiz del Arbol L, Gómez C, et al. Norfloxacin vs ceftriaxone in the prophylaxis of infections in patients with advanced cirrhosis and hemorrhage. Gastroenterology 2006; 131(4):1049–1056. doi:10.1053/j.gastro.2006.07.010
  10. Runyon B; The American Association for the Study of Liver Diseases (AASLD). Management of adult patients with ascites due to cirrhosis: update 2012. https://www.aasld.org/sites/default/files/guideline_documents/141020_Guideline_Ascites_4UFb_2015.pdf. Accessed September 4, 2018.
  11. Sidhu SS, Goyal O, Kishore H, Sidhu S. New paradigms in management of alcoholic hepatitis: a review. Hepatol Int 2017; 11(3):255–267. doi:10.1007/s12072-017-9790-5
  12. Lucey MR, Mathurin P, Morgan TR. Alcoholic hepatitis. N Engl J Med 2009; 360(26):2758–2769. doi:10.1056/NEJMra0805786
  13. Maddrey WC, Boitnott JK, Bedine MS, Weber FL Jr, Mezey E, White RI Jr. Corticosteroid therapy of alcoholic hepatitis. Gastroenterology 1978; 75(2):193–199. pmid:352788
  14. Forrest EH, Evans CD, Stewart S, et al. Analysis of factors predictive of mortality in alcoholic hepatitis and derivation and validation of the Glasgow alcoholic hepatitis score. Gut 2005; 54(8):1174–1179. doi:10.1136/gut.2004.050781
  15. Dunn W, Jamil LH, Brown LS, et al. MELD accurately predicts mortality in patients with alcoholic hepatitis. Hepatology 2005; 41(2):353–358. doi:10.1002/hep.20503
  16. Sheth M, Riggs M, Patel T. Utility of the Mayo end-stage liver disease (MELD) score in assessing prognosis of patients with alcoholic hepatitis. BMC Gastroenterol 2002; 2:2. pmid:11835693
  17. Akriviadis E, Botla R, Briggs W, Han S, Reynolds T, Shakil O. Pentoxifylline improves short-term survival in severe acute alcoholic hepatitis: a double-blind, placebo-controlled trial. Gastroenterology 2000; 119(6):1637–1648. pmid:11113085
  18. Mathurin P, O’Grady J, Carithers RL, et al. Corticosteroids improve short-term survival in patients with severe alcoholic hepatitis: meta-analysis of individual patient data. Gut 2011; 60(2):255–260. doi:10.1136/gut.2010.224097
  19. Thursz MR, Richardson P, Allison M, et al; STOPAH Trial. Prednisolone or pentoxifylline for alcoholic hepatitis. N Engl J Med 2015; 372(17):1619–1628. doi:10.1056/NEJMoa1412278
  20. Thursz M, Forrest E, Roderick P, et al. The clinical effectiveness and cost-effectiveness of steroids or pentoxifylline for alcoholic hepatitis (STOPAH): a 2 × 2 factorial randomised controlled trial. Health Technol Assess 2015; 19(102):1–104. doi:10.3310/hta191020
  21. Lee YS, Kim HJ, Kim JH, et al. Treatment of severe alcoholic hepatitis with corticosteroid, pentoxifylline, or dual therapy: a systematic review and meta-analysis. J Clin Gastroenterol 2017; 51(4):364–377. doi:10.1097/MCG.0000000000000674
  22. Louvet A, Naveau S, Abdelnour M, et al. The Lille model: a new tool for therapeutic strategy in patients with severe alcoholic hepatitis treated with steroids. Hepatology 2007; 45(6):1348–1354. doi:10.1002/hep.21607
References
  1. Ruyon BA; AASLD Practice Guidelines Committee. Management of adult patients with ascites due to cirrhosis: an update. Hepatology 2009; 49(6):2087–2107. doi:10.1002/hep.22853
  2. Hoefs JC, Canawati HN, Sapico FL, Hopkins RR, Weiner J, Montgomerie JZ. Spontaneous bacterial peritonitis. Hepatology 1982; 2(4):399–407. pmid:7095741
  3. Ginès P, Cárdenas A, Arroyo V, Rodés J. Management of cirrhosis and ascites. N Engl J Med 2004; 350(16):1646–1654. doi:10.1056/NEJMra035021
  4. Runyon BA, Montano AA, Akriviadis EA, Antillon MR, Irving MA, McHutchison JG. The serum-ascites albumin gradient is superior to the exudate-transudate concept in the differential diagnosis of ascites. Ann Intern Med 1992; 117(3):215–220. pmid:1616215
  5. Hernaez R, Hamilton JP. Unexplained ascites. Clin Liver Dis 2016; 7(3):53–56. https://aasldpubs.onlinelibrary.wiley.com/doi/epdf/10.1002/cld.537
  6. Huang LL, Xia HH, Zhu SL. Ascitic fluid analysis in the differential diagnosis of ascites: focus on cirrhotic ascites. J Clin Transl Hepatol 2014; 2(1):58–64. doi:10.14218/JCTH.2013.00010
  7. Bartoloni A, Zammarchi L. Clinical aspects of uncomplicated and severe malaria. Mediterr J Hematol Infect Dis 2012; 4(1):e2012026. doi:10.4084/MJHID.2012.026
  8. Titó L, Rimola A, Ginès P, Llach J, Arroyo V, Rodés J. Recurrence of spontaneous bacterial peritonitis in cirrhosis: frequency and predictive factors. Hepatology 1988; 8(1):27–31. pmid:3257456
  9. Fernández J, Ruiz del Arbol L, Gómez C, et al. Norfloxacin vs ceftriaxone in the prophylaxis of infections in patients with advanced cirrhosis and hemorrhage. Gastroenterology 2006; 131(4):1049–1056. doi:10.1053/j.gastro.2006.07.010
  10. Runyon B; The American Association for the Study of Liver Diseases (AASLD). Management of adult patients with ascites due to cirrhosis: update 2012. https://www.aasld.org/sites/default/files/guideline_documents/141020_Guideline_Ascites_4UFb_2015.pdf. Accessed September 4, 2018.
  11. Sidhu SS, Goyal O, Kishore H, Sidhu S. New paradigms in management of alcoholic hepatitis: a review. Hepatol Int 2017; 11(3):255–267. doi:10.1007/s12072-017-9790-5
  12. Lucey MR, Mathurin P, Morgan TR. Alcoholic hepatitis. N Engl J Med 2009; 360(26):2758–2769. doi:10.1056/NEJMra0805786
  13. Maddrey WC, Boitnott JK, Bedine MS, Weber FL Jr, Mezey E, White RI Jr. Corticosteroid therapy of alcoholic hepatitis. Gastroenterology 1978; 75(2):193–199. pmid:352788
  14. Forrest EH, Evans CD, Stewart S, et al. Analysis of factors predictive of mortality in alcoholic hepatitis and derivation and validation of the Glasgow alcoholic hepatitis score. Gut 2005; 54(8):1174–1179. doi:10.1136/gut.2004.050781
  15. Dunn W, Jamil LH, Brown LS, et al. MELD accurately predicts mortality in patients with alcoholic hepatitis. Hepatology 2005; 41(2):353–358. doi:10.1002/hep.20503
  16. Sheth M, Riggs M, Patel T. Utility of the Mayo end-stage liver disease (MELD) score in assessing prognosis of patients with alcoholic hepatitis. BMC Gastroenterol 2002; 2:2. pmid:11835693
  17. Akriviadis E, Botla R, Briggs W, Han S, Reynolds T, Shakil O. Pentoxifylline improves short-term survival in severe acute alcoholic hepatitis: a double-blind, placebo-controlled trial. Gastroenterology 2000; 119(6):1637–1648. pmid:11113085
  18. Mathurin P, O’Grady J, Carithers RL, et al. Corticosteroids improve short-term survival in patients with severe alcoholic hepatitis: meta-analysis of individual patient data. Gut 2011; 60(2):255–260. doi:10.1136/gut.2010.224097
  19. Thursz MR, Richardson P, Allison M, et al; STOPAH Trial. Prednisolone or pentoxifylline for alcoholic hepatitis. N Engl J Med 2015; 372(17):1619–1628. doi:10.1056/NEJMoa1412278
  20. Thursz M, Forrest E, Roderick P, et al. The clinical effectiveness and cost-effectiveness of steroids or pentoxifylline for alcoholic hepatitis (STOPAH): a 2 × 2 factorial randomised controlled trial. Health Technol Assess 2015; 19(102):1–104. doi:10.3310/hta191020
  21. Lee YS, Kim HJ, Kim JH, et al. Treatment of severe alcoholic hepatitis with corticosteroid, pentoxifylline, or dual therapy: a systematic review and meta-analysis. J Clin Gastroenterol 2017; 51(4):364–377. doi:10.1097/MCG.0000000000000674
  22. Louvet A, Naveau S, Abdelnour M, et al. The Lille model: a new tool for therapeutic strategy in patients with severe alcoholic hepatitis treated with steroids. Hepatology 2007; 45(6):1348–1354. doi:10.1002/hep.21607
Issue
Cleveland Clinic Journal of Medicine - 86(4)
Issue
Cleveland Clinic Journal of Medicine - 86(4)
Page Number
257-262
Page Number
257-262
Publications
Publications
Topics
Article Type
Display Headline
A woman, age 35, with new-onset ascites
Display Headline
A woman, age 35, with new-onset ascites
Legacy Keywords
ascites, jaundice, cirrhosis, portal hypertension, liver function tests, alanine aminotransferase, ALT, aspartate aminotransferase, AST, hepatitis, serum-ascites albumin gradient, SAAG, alcoholism, steatosis, liver biopsy, Model for End-stage Liver Disease score, MELD, Maddrey Discriminant Function score, DF score, alcoholic hepatitis, William Lippert, Eun Lee, Aibek Mirrakhimov
Legacy Keywords
ascites, jaundice, cirrhosis, portal hypertension, liver function tests, alanine aminotransferase, ALT, aspartate aminotransferase, AST, hepatitis, serum-ascites albumin gradient, SAAG, alcoholism, steatosis, liver biopsy, Model for End-stage Liver Disease score, MELD, Maddrey Discriminant Function score, DF score, alcoholic hepatitis, William Lippert, Eun Lee, Aibek Mirrakhimov
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Tue, 03/26/2019 - 11:00
Un-Gate On Date
Tue, 03/26/2019 - 11:00
Use ProPublica
CFC Schedule Remove Status
Tue, 03/26/2019 - 11:00
Hide sidebar & use full width
render the right sidebar.
Article PDF Media

Human papillomavirus in 2019: An update on cervical cancer prevention and screening guidelines

Article Type
Changed
Wed, 04/24/2019 - 12:36
Display Headline
Human papillomavirus in 2019: An update on cervical cancer prevention and screening guidelines

About 12% of women worldwide are infected with human papillomavirus (HPV).1 Persistent HPV infection with high-risk strains such as HPV 6, 11, 16, and 18 cause nearly all cases of cervical cancer and some anal, vaginal, penile, and oropharyngeal cancers.2 An estimated 13,000 cases of invasive cervical cancer will be diagnosed this year in the United States alone.3

Up to 70% of HPV-related cervical cancer cases can be prevented with vaccination. A number of changes have been made to the vaccination schedule within the past few years—patients younger than 15 need only 2 rather than 3 doses, and the vaccine itself can be used in adults up to age 45.

Vaccination and routine cervical cancer screening are both necessary to prevent this disease3 along with effective family and patient counseling. Here, we discuss the most up-to-date HPV vaccination recommendations, current cervical cancer screening guidelines, counseling techniques that increase vaccination acceptance rates, and follow-up protocols for abnormal cervical cancer screening results.

TYPES OF HPV VACCINES

HPV immunization can prevent up to 70% of cases of cervical cancer due to HPV as well as 90% of genital warts.4 The US Food and Drug Administration (FDA) has approved 3 HPV vaccines:

  • Gardasil 9 targets HPV types 6, 11, 16, and 18 along with 31, 33, 45, 52, 58—these cause 90% of cervical cancer cases and most cases of genital warts5—making it the most effective vaccine available; Gardasil 9 is the only HPV vaccine currently available in the United States
  • The bivalent vaccine (Cervarix) targeted HPV 16 and 18 only, and was discontinued in the United States in 2016
  • The quadrivalent HPV vaccine (Gardasil) targeted HPV 16 and 18 as well as 6 and 11, which cause most cases of genital warts; the last available doses in the United States expired in May 2017; it has been replaced by Gardasil 9.

The incidence of cervical cancer in the United States dropped 29% among 15- to 24-year-olds from 2003–2006 when HPV vaccination first started to 2011–2014.6

VACCINE DOSING RECOMMENDATIONS FOR PRIMARY PREVENTION

zhang_hpv_t1.jpg

The Advisory Committee on Immunization Practices (ACIP) revised its HPV vaccine schedule in 2016, when it decreased the necessary doses from 3 to 2 for patients under age 15 and addressed the needs of special patient populations.7 In late 2018, the FDA approved the use of the vaccine in men and women up to age 45. However, no change in guidelines have yet been made (Table 1).

In females, the ACIP recommends starting HPV vaccination at age 11 or 12, but it can be given as early as age 9. A 2-dose schedule is recommended for the 9-valent vaccine before the patient’s 15th birthday (the second dose 6 to 12 months after the first).7 For females who initiate HPV vaccination between ages 15 and 45, a 3-dose schedule is necessary (at 0, 1 to 2, and 6 months).7,8

The change to a 2-dose schedule was prompted by an evaluation of girls ages 9 to 13 randomized to receive either a 2- or 3-dose schedule. Antibody responses with a 2-dose schedule were not inferior to those of young women (ages 16 to 26) who received all 3 doses.9 The geometric mean titer ratios remained noninferior throughout the study period of 36 months.

However, a loss of noninferiority was noted for HPV-18 by 24 months and for HPV-6 by 36 months.9 Thus, further studies are needed to understand the duration of protection with a 2-dose schedule. Nevertheless, decreasing the number of doses makes it a more convenient and cost-effective option for many families.

The recommendations are the same for males except for one notable difference: in males ages 21 to 26, vaccination is not routinely recommended by the ACIP, but rather it is considered a “permissive use” recommendation: ie, the vaccine should be offered and final decisions on administration be made after individualized discussion with the patient.10 Permissive-use status also means the vaccine may not be covered by health insurance. Even though the vaccine is now available to men and women until age 45, many insurance plans do not cover it after age 26.

Children of either sex with a history of sexual abuse should receive their first vaccine dose beginning at age 9.7

Immunocompromised patients should follow the 3-dose schedule regardless of their sex or the age when vaccination was initiated.10

For transgender patients and for men not previously vaccinated who have sex with men, the 3-dose schedule vaccine should be given by the age of 26 (this is a routine recommendation, not a permissive one).8

 

 

CHALLENGES OF VACCINATION

Effective patient and family counseling is important. Even though the first HPV vaccine was approved in 2006, only 34.9% of US adolescents were fully vaccinated by 2015. This was in part because providers did not recommend it, were unfamiliar with it, or had concerns about its safety,11,12 and in part because some parents refused it.

The physician must address any myths regarding HPV vaccination and ensure that parents and patients understand that HPV vaccine is safe and effective. Studies have shown that with high-quality recommendations (ie, the care provider strongly endorses the HPV vaccine, encourages same-day vaccination, and discusses cancer prevention), patients are 9 times more likely to start the HPV vaccination schedule and 3 times more likely to follow through with subsequent doses.13

Providing good family and patient education does not necessarily require spending more counseling time. A recent study showed that spending less time discussing the HPV vaccine can lead to better vaccine coverage.14 The study compared parent HPV vaccine counseling techniques and found that simply informing patients and their families that the HPV vaccine was due was associated with a higher vaccine acceptance rate than inviting conversations about it.14 When providers announced that the vaccine was due, assuming the parents were ready to vaccinate, there was a 5.4% increase in HPV vaccination coverage.14

zhang_hpv_t2.jpg

Conversely, physicians who engaged parents in open-ended discussions about the HPV vaccine did not improve HPV vaccination coverage.14 The authors suggested that providers approach HPV vaccination as if they were counseling patients and families about the need to avoid second-hand smoke or the need to use car seats. If parents or patients resist the presumptive announcement approach, expanded counseling and shared decision-making are appropriate. This includes addressing misconceptions that parents and patients may have about the HPV vaccine. The American Cancer Society lists 8 facts to reference (Table 2).15

SECONDARY PREVENTION: CERVICAL CANCER SCREENING

Since the introduction of the Papanicolaou (Pap) test, US cervical cancer incidence rates have decreased by more than 60%.16 Because almost all cervical cancer is preventable with proper screening, all women ages 21 to 65 should be screened.

zhang_hpv_t3.jpg

Currently, there are 3 options available for cervical cancer screening: the Pap-only test, the Pap-HPV cotest, and the high-risk HPV-only test (Table 3). The latter 2 options detect high-risk HPV genotypes.

Several organizations have screening algorithms that recommend when to use these tests, but the 3 that shape today’s standard of care in cervical cancer screening come from the American College of Obstetricians and Gynecologists (ACOG), the American Society for Colposcopy and Cervical Pathology (ASCCP), and US Preventive Services Task Force (USPSTF).17–19

Pap-only testing is performed every 3 years to screen for cervical neoplasia that might indicate premalignancy.

Pap-HPV cotesting is performed every 5 years in women older than 30 with past normal screening. Until 2018, all 3 organizations recommended cotesting as the preferred screening algorithm for women ages 30 to 65.17–19 Patients with a history of abnormal test results require more frequent testing as recommended by the ASCCP.18

The high-risk HPV-only test utilizes real-time polymerase chain reaction to detect HPV 16, HPV 18, and 12 other HPV genotypes. Only 2 tests are approved by the FDA as stand-alone cervical cancer screening tests—the Roche Cobas HPV test approved in 2014 and the Becton Dickinson Onclarity HPV assay approved in 2018. Other HPV tests that are used in a cotesting strategy should not be used for high-risk HPV-only testing because their performance characteristics may differ.

In 2015, the Addressing the Need for Advanced HPV Diagnostics (ATHENA) study showed that 1 round of high-risk HPV-only screening for women older than 25 was more sensitive than Pap-only or cotesting for stage 3 cervical intraepithelial neoplasia or more severe disease (after 3 years of follow-up).20 Current guidelines from ASCCP18 and ACOG17 state that the high-risk HPV test can be repeated every 3 years (when used to screen by itself) if the woman is older than 25 and has had a normal test result.

zhang_hpv_f1.jpg
%3Cp%3EFigure%201.%3C%2Fp%3E

If the HPV test result is positive for high-risk HPV 16 or 18 genotypes, then immediate colposcopy is indicated; women who test positive for one of the other 12 high-risk subtypes will need to undergo a Pap test to determine the appropriate follow-up (Figure 1).18,21

In 2018, the USPSTF updated its recommendations, noting that for women age 30 to 65, Pap-only testing every 3 years, cotesting every 5 years, or high-risk HPV-only testing every 5 years are all appropriate screening strategies, with the Pap-only or high-risk HPV-only screenings being preferred.19 This is in contrast to ACOG and ASCCP recommendations for cotesting every 5 years, with alternative options of Pap-only or HPV-only testing being done every 3 years.17,18

 

 

Is there a best screening protocol?

The USPSTF reviewed large randomized and observational studies to summarize the effectiveness of the 3 screening strategies and commissioned a decision analysis model to compare the risks, benefits, and costs of the 3 screening algorithms. The guideline statement notes both cotesting and high-risk HPV testing offer similar cancer detection rates: each prevents 1 additional cancer per 1,000 women screened as opposed to Pap-only testing.19

Also, tests that incorporate high-risk HPV screening may offer better detection of cervical adenocarcinoma (which has a worse prognosis than the more common squamous cell carcinoma type). However, both HPV-based screening strategies are more likely to require additional colposcopies for follow-up than Pap-only screening (1,630 colposcopies required for each cancer prevented with high-risk HPV alone, 1,635 with cotesting). Colposcopy is a simple office procedure that causes minimal discomfort to the patient.

The USPSTF guideline also differs in the recommended frequency of high-risk HPV-only testing; a high-risk HPV result should be repeated every 5 years if normal (as opposed to every 3 years as recommended by ACOG and ASCCP).19 The 5-year recommendation is based on analysis modeling, which suggests that performing high-risk HPV-only testing more frequently is unlikely to improve detection rates but will increase the number of screening tests and colposcopies.19

No trial has directly compared cotesting with high-risk HPV testing for more than 2 rounds of screening. The updated USPSTF recommendations are based on modeling estimates and expert opinion, which assesses cost and benefit vs harm in the long term. Also, no high-risk HPV test is currently FDA-approved for every-5-year screening when used by itself.

All 3 cervical cancer screening methods provide highly effective cancer prevention, so it is important for providers to choose the strategy that best fits their practice. The most critical aspect of screening is getting all women screened, no matter which method is used.

It is critical to remember that the screening intervals are intended for patients without symptoms. Those who have new concerns such as bleeding should have a diagnostic Pap done to evaluate their symptoms.

Follow-up of abnormal results

Regardless of the pathway chosen, appropriate follow-up of any abnormal test result is critical to the early detection of cancer. Established follow-up guidelines exist,22,23 but accessing this information can be difficult for the busy clinician. The ASCCP has a mobile phone application that outlines the action steps corresponding to the patient’s age and results of any combination of Pap or HPV testing. The app also includes the best screening algorithms for a particular patient.24

All guidelines agree that cervical cancer screening should start at age 21, regardless of HPV vaccination status or age of sexual initiation.17,18,25 Screening can be discontinued at age 65 for women with normal screening results in the prior decade (3 consecutive negative Pap results or 2 consecutive negative cotest results).23

For women who have had a total hysterectomy and no history of cervical neoplasia, screening should be stopped immediately after the procedure. However, several high-risk groups of women will need continued screening past the age of 65, or after a hysterectomy.

For a woman with a history of stage 2 cervical intraepithelial neoplasia or higher grade lesions, routine screening is continued for an additional 20 years, even if she is over age 65. Pap-only testing every 3 years is acceptable, because the role of HPV testing is unclear after hysterectomy.23 Prior guidelines suggested annual screening in these patients, so the change to every 3 years is notable. Many gynecologic oncologists will recommend that women with a history of cervical cancer continue annual screening indefinitely.

Within the first 2 to 3 years after treatment for high-grade dysplastic changes, annual follow-up is done by the gynecologic oncology team. Providers who offer follow-up during this time frame should keep in communication with the oncology team to ensure appropriate, individualized care. These recommendations are based on expert opinion, so variations in clinical practice may be seen.

Women infected with the human immunodeficiency virus can have Pap-only testing every 3 years, after a series of 3 normal annual Pap results.26 But screening does not stop at age 65.23,26 For patients who are immunosuppressed or have a history of diethylstilbestrol exposure, screening should be done annually indefinitely.23

References
  1. Bruni L, Diaz M, Castellsagué X, Ferrer E, Bosch FX, de Sanjosé S. Cervical human papillomavirus prevalence in 5 continents: meta-analysis of 1 million women with normal cytological findings. J Infect Dis 2010; 202(12):1789–1799. doi:10.1086/657321
  2. de Martel C, Ferlay J, Franceschi S, et al. Global burden of cancer attributable to infections in 2008: a review and synthetic analysis. Lancet Oncol 2012; 13(6):607–615. doi:10.1016/S1470-2045(12)70137-7
  3. American Cancer Society. Key statistics for cervical cancer. www.cancer.org/cancer/cervical-cancer/about/key-statistics.html. Accessed February 14, 2019.
  4. Thaxton L, Waxman AG. Cervical cancer prevention: immunization and screening 2015. Med Clin North Am 2015; 99(3):469–477. doi:10.1016/j.mcna.2015.01.003
  5. McNamara M, Batur P, Walsh JME, Johnson KM. HPV update: vaccination, screening, and associated disease. J Gen Intern Med 2016; 31(11):1360–1366. doi:10.1007/s11606-016-3725-z
  6. Guo F, Cofie LE, Berenson AB. Cervical cancer incidence in young US females after human papillomavirus vaccine introduction. Am J Prev Med 2018; 55(2):197–204. doi:10.1016/j.amepre.2018.03.013
  7. Meites E, Kempe A, Markowitz LE. Use of a 2-dose schedule for human papillomavirus vaccination—updated recommendations of the Advisory Committee on Immunization Practices. MMWR Morb Mortal Wkly Rep 2016; 65(49):1405–1408. doi:10.15585/mmwr.mm6549a5
  8. Centers for Disease Control and Prevention (CDC). Supplemental information and guidance for vaccination providers regarding use of 9-valent HPV vaccine Information for persons who started an HPV vaccination series with quadrivalent or bivalent HPV vaccine. www.cdc.gov/hpv/downloads/9vhpv-guidance.pdf. Accessed February 14, 2019.
  9. Dobson SR, McNeil S, Dionne M, et al. Immunogenicity of 2 doses of HPV vaccine in younger adolescents vs 3 doses in young women: a randomized clinical trial. JAMA 2013; 309(17):1793–1802. doi:10.1001/jama.2013.1625
  10. Markowitz LE, Dunne EF, Saraiya M, et al; Centers for Disease Control and Prevention (CDC). Human papillomavirus vaccination: recommendations of the Advisory Committee on Immunization Practices (ACIP). MMWR Recomm Rep 2014; 63(RR-05):1–30. pmid:25167164
  11. Thompson EL, Rosen BL, Vamos CA, Kadono M, Daley EM. Human papillomavirus vaccination: what are the reasons for nonvaccination among US adolescents? J Adolesc Health 2017; 61(3):288–293. doi:10.1016/j.jadohealth.2017.05.015
  12. Reagan-Steiner S, Yankey D, Jeyarajah J, et al. National, regional, state, and selected local area vaccination coverage among adolescents aged 13-17 years—United States, 2015. MMWR Morb Mortal Wkly Rep 2016; 65(33):850–858. doi:10.15585/mmwr.mm6533a4
  13. Gilkey MB, Calo WA, Moss JL, Shah PD, Marciniak MW, Brewer NT. Provider communication and HPV vaccination: The impact of recommendation quality. Vaccine 2016; 34(9):1187–1192. doi:10.1016/j.vaccine.2016.01.023
  14. Brewer NT, Hall ME, Malo TL, Gilkey MB, Quinn B, Lathren C. Announcements versus conversations to improve HPV vaccination coverage: a randomized trial. Pediatrics 2017; 139(1):e20161764. doi:10.1542/peds.2016-1764
  15. American Cancer Society. HPV vaccine facts. www.cancer.org/cancer/cancer-causes/infectious-agents/hpv/hpv-vaccine-facts-and-fears.html. Accessed February 14, 2019.
  16. National Cancer Institute; Chasan R, Manrow R. Cervical cancer. https://report.nih.gov/nihfactsheets/viewfactsheet.aspx?csid=76. Accessed February 14, 2019.
  17. The American College of Obstetricians and Gynecologists (ACOG). Frequently asked questions. Cervical cancer screening. www.acog.org/Patients/FAQs/Cervical-Cancer-Screening. Accessed February 14, 2019.
  18. Saslow D, Solomon D, Lawson HW, et al; American Cancer Society; American Society for Colposcopy and Cervical Pathology; American Society for Clinical Pathology. American Cancer Society, American Society for Colposcopy and Cervical Pathology, and American Society for Clinical Pathology screening guidelines for the prevention and early detection of cervical cancer. Am J Clin Pathol 2012; 137(4):516–542. doi:10.1309/AJCPTGD94EVRSJCG
  19. US Preventive Services Task Force; Curry SJ, Krist AH, Owens DK, et al. Screening for cervical cancer: US Preventive Services Task Force Recommendation Statement. JAMA 2018; 320(7):674–686. doi:10.1001/jama.2018.10897
  20. Wright TC, Stoler MH, Behrens CM, Sharma A, Zhang G, Wright TL. Primary cervical cancer screening with human papillomavirus: end of study results from the ATHENA study using HPV as the first-line screening test. Gynecol Oncol 2015; 136(2):189–197. doi:10.1016/j.ygyno.2014.11.076
  21. Huh WK, Ault KA, Chelmow D, et al. Use of primary high-risk human papillomavirus testing for cervical cancer screening: interim clinical guidance. Obstet Gynecol 2015; 125(2):330–337. doi:10.1097/AOG.0000000000000669
  22. Massad LS, Einstein MH, Huh WK, et al; 2012 ASCCP Consensus Guidelines Conference. 2012 updated consensus guidelines for the management of abnormal cervical cancer screening tests and cancer precursors. Obstet Gynecol 2013; 121(4):829–846. doi:10.1097/AOG.0b013e3182883a34
  23. Committee on Practice Bulletins—Gynecology. Practice Bulletin No. 168: cervical cancer screening and prevention. Obstet Gynecol 2016; 128(4):e111–e130. doi:10.1097/AOG.0000000000001708
  24. ASCCP. Mobile app. http://www.asccp.org/store-detail2/asccp-mobile-app. Accessed February 14, 2019.
  25. USPSTF. Draft recommendation: cervical cancer: screening. www.uspreventiveservicestaskforce.org/Page/Document/draft-recommendation-statement/cervical-cancer-screening2. Accessed February 14, 2019.
  26. Masur H, Brooks JT, Benson CA, Holmes KK, Pau AK, Kaplan JE; National Institutes of Health; Centers for Disease Control and Prevention; HIV Medicine Association of the Infectious Diseases Society of America. Prevention and treatment of opportunistic infections in HIV-infected adults and adolescents: Updated guidelines from the Centers for Disease Control and Prevention, National Institutes of Health, and HIV Medicine Association of the Infectious Diseases Society of America. Clin Infect Dis 2014; 58(9):1308–1311. doi:10.1093/cid/ciu094
Article PDF
Author and Disclosure Information

Salina Zhang, BS
Case Western Reserve University School of Medicine, Cleveland, OH

Pelin Batur, MD, FACP, NCMP, CCD
Department of Obstetrics and Gynecology, Women’s Health Institute, Cleveland Clinic; Associate Professor of Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH; Deputy Editor, Cleveland Clinic Journal of Medicine, Working Group Member of the US Cervical Cancer Screening Risk-Based Management Guidelines Committee

Address: Pelin Batur, MD, FACP, NCMP, CCD, Department of Obstetrics and Gynecology, Women’s Health Institute, A81, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195; baturp@ccf.org

Issue
Cleveland Clinic Journal of Medicine - 86(3)
Publications
Topics
Page Number
173-178
Legacy Keywords
human papillomavirus, HPV, cervical cancer, screening, immunization, vaccination, HPV vaccine, Gardasil, Papanicolaou test, Pap test, HPV test, screening, Salina Zhang, Pelin Batur
Sections
Author and Disclosure Information

Salina Zhang, BS
Case Western Reserve University School of Medicine, Cleveland, OH

Pelin Batur, MD, FACP, NCMP, CCD
Department of Obstetrics and Gynecology, Women’s Health Institute, Cleveland Clinic; Associate Professor of Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH; Deputy Editor, Cleveland Clinic Journal of Medicine, Working Group Member of the US Cervical Cancer Screening Risk-Based Management Guidelines Committee

Address: Pelin Batur, MD, FACP, NCMP, CCD, Department of Obstetrics and Gynecology, Women’s Health Institute, A81, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195; baturp@ccf.org

Author and Disclosure Information

Salina Zhang, BS
Case Western Reserve University School of Medicine, Cleveland, OH

Pelin Batur, MD, FACP, NCMP, CCD
Department of Obstetrics and Gynecology, Women’s Health Institute, Cleveland Clinic; Associate Professor of Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH; Deputy Editor, Cleveland Clinic Journal of Medicine, Working Group Member of the US Cervical Cancer Screening Risk-Based Management Guidelines Committee

Address: Pelin Batur, MD, FACP, NCMP, CCD, Department of Obstetrics and Gynecology, Women’s Health Institute, A81, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195; baturp@ccf.org

Article PDF
Article PDF
Related Articles

About 12% of women worldwide are infected with human papillomavirus (HPV).1 Persistent HPV infection with high-risk strains such as HPV 6, 11, 16, and 18 cause nearly all cases of cervical cancer and some anal, vaginal, penile, and oropharyngeal cancers.2 An estimated 13,000 cases of invasive cervical cancer will be diagnosed this year in the United States alone.3

Up to 70% of HPV-related cervical cancer cases can be prevented with vaccination. A number of changes have been made to the vaccination schedule within the past few years—patients younger than 15 need only 2 rather than 3 doses, and the vaccine itself can be used in adults up to age 45.

Vaccination and routine cervical cancer screening are both necessary to prevent this disease3 along with effective family and patient counseling. Here, we discuss the most up-to-date HPV vaccination recommendations, current cervical cancer screening guidelines, counseling techniques that increase vaccination acceptance rates, and follow-up protocols for abnormal cervical cancer screening results.

TYPES OF HPV VACCINES

HPV immunization can prevent up to 70% of cases of cervical cancer due to HPV as well as 90% of genital warts.4 The US Food and Drug Administration (FDA) has approved 3 HPV vaccines:

  • Gardasil 9 targets HPV types 6, 11, 16, and 18 along with 31, 33, 45, 52, 58—these cause 90% of cervical cancer cases and most cases of genital warts5—making it the most effective vaccine available; Gardasil 9 is the only HPV vaccine currently available in the United States
  • The bivalent vaccine (Cervarix) targeted HPV 16 and 18 only, and was discontinued in the United States in 2016
  • The quadrivalent HPV vaccine (Gardasil) targeted HPV 16 and 18 as well as 6 and 11, which cause most cases of genital warts; the last available doses in the United States expired in May 2017; it has been replaced by Gardasil 9.

The incidence of cervical cancer in the United States dropped 29% among 15- to 24-year-olds from 2003–2006 when HPV vaccination first started to 2011–2014.6

VACCINE DOSING RECOMMENDATIONS FOR PRIMARY PREVENTION

zhang_hpv_t1.jpg

The Advisory Committee on Immunization Practices (ACIP) revised its HPV vaccine schedule in 2016, when it decreased the necessary doses from 3 to 2 for patients under age 15 and addressed the needs of special patient populations.7 In late 2018, the FDA approved the use of the vaccine in men and women up to age 45. However, no change in guidelines have yet been made (Table 1).

In females, the ACIP recommends starting HPV vaccination at age 11 or 12, but it can be given as early as age 9. A 2-dose schedule is recommended for the 9-valent vaccine before the patient’s 15th birthday (the second dose 6 to 12 months after the first).7 For females who initiate HPV vaccination between ages 15 and 45, a 3-dose schedule is necessary (at 0, 1 to 2, and 6 months).7,8

The change to a 2-dose schedule was prompted by an evaluation of girls ages 9 to 13 randomized to receive either a 2- or 3-dose schedule. Antibody responses with a 2-dose schedule were not inferior to those of young women (ages 16 to 26) who received all 3 doses.9 The geometric mean titer ratios remained noninferior throughout the study period of 36 months.

However, a loss of noninferiority was noted for HPV-18 by 24 months and for HPV-6 by 36 months.9 Thus, further studies are needed to understand the duration of protection with a 2-dose schedule. Nevertheless, decreasing the number of doses makes it a more convenient and cost-effective option for many families.

The recommendations are the same for males except for one notable difference: in males ages 21 to 26, vaccination is not routinely recommended by the ACIP, but rather it is considered a “permissive use” recommendation: ie, the vaccine should be offered and final decisions on administration be made after individualized discussion with the patient.10 Permissive-use status also means the vaccine may not be covered by health insurance. Even though the vaccine is now available to men and women until age 45, many insurance plans do not cover it after age 26.

Children of either sex with a history of sexual abuse should receive their first vaccine dose beginning at age 9.7

Immunocompromised patients should follow the 3-dose schedule regardless of their sex or the age when vaccination was initiated.10

For transgender patients and for men not previously vaccinated who have sex with men, the 3-dose schedule vaccine should be given by the age of 26 (this is a routine recommendation, not a permissive one).8

 

 

CHALLENGES OF VACCINATION

Effective patient and family counseling is important. Even though the first HPV vaccine was approved in 2006, only 34.9% of US adolescents were fully vaccinated by 2015. This was in part because providers did not recommend it, were unfamiliar with it, or had concerns about its safety,11,12 and in part because some parents refused it.

The physician must address any myths regarding HPV vaccination and ensure that parents and patients understand that HPV vaccine is safe and effective. Studies have shown that with high-quality recommendations (ie, the care provider strongly endorses the HPV vaccine, encourages same-day vaccination, and discusses cancer prevention), patients are 9 times more likely to start the HPV vaccination schedule and 3 times more likely to follow through with subsequent doses.13

Providing good family and patient education does not necessarily require spending more counseling time. A recent study showed that spending less time discussing the HPV vaccine can lead to better vaccine coverage.14 The study compared parent HPV vaccine counseling techniques and found that simply informing patients and their families that the HPV vaccine was due was associated with a higher vaccine acceptance rate than inviting conversations about it.14 When providers announced that the vaccine was due, assuming the parents were ready to vaccinate, there was a 5.4% increase in HPV vaccination coverage.14

zhang_hpv_t2.jpg

Conversely, physicians who engaged parents in open-ended discussions about the HPV vaccine did not improve HPV vaccination coverage.14 The authors suggested that providers approach HPV vaccination as if they were counseling patients and families about the need to avoid second-hand smoke or the need to use car seats. If parents or patients resist the presumptive announcement approach, expanded counseling and shared decision-making are appropriate. This includes addressing misconceptions that parents and patients may have about the HPV vaccine. The American Cancer Society lists 8 facts to reference (Table 2).15

SECONDARY PREVENTION: CERVICAL CANCER SCREENING

Since the introduction of the Papanicolaou (Pap) test, US cervical cancer incidence rates have decreased by more than 60%.16 Because almost all cervical cancer is preventable with proper screening, all women ages 21 to 65 should be screened.

zhang_hpv_t3.jpg

Currently, there are 3 options available for cervical cancer screening: the Pap-only test, the Pap-HPV cotest, and the high-risk HPV-only test (Table 3). The latter 2 options detect high-risk HPV genotypes.

Several organizations have screening algorithms that recommend when to use these tests, but the 3 that shape today’s standard of care in cervical cancer screening come from the American College of Obstetricians and Gynecologists (ACOG), the American Society for Colposcopy and Cervical Pathology (ASCCP), and US Preventive Services Task Force (USPSTF).17–19

Pap-only testing is performed every 3 years to screen for cervical neoplasia that might indicate premalignancy.

Pap-HPV cotesting is performed every 5 years in women older than 30 with past normal screening. Until 2018, all 3 organizations recommended cotesting as the preferred screening algorithm for women ages 30 to 65.17–19 Patients with a history of abnormal test results require more frequent testing as recommended by the ASCCP.18

The high-risk HPV-only test utilizes real-time polymerase chain reaction to detect HPV 16, HPV 18, and 12 other HPV genotypes. Only 2 tests are approved by the FDA as stand-alone cervical cancer screening tests—the Roche Cobas HPV test approved in 2014 and the Becton Dickinson Onclarity HPV assay approved in 2018. Other HPV tests that are used in a cotesting strategy should not be used for high-risk HPV-only testing because their performance characteristics may differ.

In 2015, the Addressing the Need for Advanced HPV Diagnostics (ATHENA) study showed that 1 round of high-risk HPV-only screening for women older than 25 was more sensitive than Pap-only or cotesting for stage 3 cervical intraepithelial neoplasia or more severe disease (after 3 years of follow-up).20 Current guidelines from ASCCP18 and ACOG17 state that the high-risk HPV test can be repeated every 3 years (when used to screen by itself) if the woman is older than 25 and has had a normal test result.

zhang_hpv_f1.jpg
%3Cp%3EFigure%201.%3C%2Fp%3E

If the HPV test result is positive for high-risk HPV 16 or 18 genotypes, then immediate colposcopy is indicated; women who test positive for one of the other 12 high-risk subtypes will need to undergo a Pap test to determine the appropriate follow-up (Figure 1).18,21

In 2018, the USPSTF updated its recommendations, noting that for women age 30 to 65, Pap-only testing every 3 years, cotesting every 5 years, or high-risk HPV-only testing every 5 years are all appropriate screening strategies, with the Pap-only or high-risk HPV-only screenings being preferred.19 This is in contrast to ACOG and ASCCP recommendations for cotesting every 5 years, with alternative options of Pap-only or HPV-only testing being done every 3 years.17,18

 

 

Is there a best screening protocol?

The USPSTF reviewed large randomized and observational studies to summarize the effectiveness of the 3 screening strategies and commissioned a decision analysis model to compare the risks, benefits, and costs of the 3 screening algorithms. The guideline statement notes both cotesting and high-risk HPV testing offer similar cancer detection rates: each prevents 1 additional cancer per 1,000 women screened as opposed to Pap-only testing.19

Also, tests that incorporate high-risk HPV screening may offer better detection of cervical adenocarcinoma (which has a worse prognosis than the more common squamous cell carcinoma type). However, both HPV-based screening strategies are more likely to require additional colposcopies for follow-up than Pap-only screening (1,630 colposcopies required for each cancer prevented with high-risk HPV alone, 1,635 with cotesting). Colposcopy is a simple office procedure that causes minimal discomfort to the patient.

The USPSTF guideline also differs in the recommended frequency of high-risk HPV-only testing; a high-risk HPV result should be repeated every 5 years if normal (as opposed to every 3 years as recommended by ACOG and ASCCP).19 The 5-year recommendation is based on analysis modeling, which suggests that performing high-risk HPV-only testing more frequently is unlikely to improve detection rates but will increase the number of screening tests and colposcopies.19

No trial has directly compared cotesting with high-risk HPV testing for more than 2 rounds of screening. The updated USPSTF recommendations are based on modeling estimates and expert opinion, which assesses cost and benefit vs harm in the long term. Also, no high-risk HPV test is currently FDA-approved for every-5-year screening when used by itself.

All 3 cervical cancer screening methods provide highly effective cancer prevention, so it is important for providers to choose the strategy that best fits their practice. The most critical aspect of screening is getting all women screened, no matter which method is used.

It is critical to remember that the screening intervals are intended for patients without symptoms. Those who have new concerns such as bleeding should have a diagnostic Pap done to evaluate their symptoms.

Follow-up of abnormal results

Regardless of the pathway chosen, appropriate follow-up of any abnormal test result is critical to the early detection of cancer. Established follow-up guidelines exist,22,23 but accessing this information can be difficult for the busy clinician. The ASCCP has a mobile phone application that outlines the action steps corresponding to the patient’s age and results of any combination of Pap or HPV testing. The app also includes the best screening algorithms for a particular patient.24

All guidelines agree that cervical cancer screening should start at age 21, regardless of HPV vaccination status or age of sexual initiation.17,18,25 Screening can be discontinued at age 65 for women with normal screening results in the prior decade (3 consecutive negative Pap results or 2 consecutive negative cotest results).23

For women who have had a total hysterectomy and no history of cervical neoplasia, screening should be stopped immediately after the procedure. However, several high-risk groups of women will need continued screening past the age of 65, or after a hysterectomy.

For a woman with a history of stage 2 cervical intraepithelial neoplasia or higher grade lesions, routine screening is continued for an additional 20 years, even if she is over age 65. Pap-only testing every 3 years is acceptable, because the role of HPV testing is unclear after hysterectomy.23 Prior guidelines suggested annual screening in these patients, so the change to every 3 years is notable. Many gynecologic oncologists will recommend that women with a history of cervical cancer continue annual screening indefinitely.

Within the first 2 to 3 years after treatment for high-grade dysplastic changes, annual follow-up is done by the gynecologic oncology team. Providers who offer follow-up during this time frame should keep in communication with the oncology team to ensure appropriate, individualized care. These recommendations are based on expert opinion, so variations in clinical practice may be seen.

Women infected with the human immunodeficiency virus can have Pap-only testing every 3 years, after a series of 3 normal annual Pap results.26 But screening does not stop at age 65.23,26 For patients who are immunosuppressed or have a history of diethylstilbestrol exposure, screening should be done annually indefinitely.23

About 12% of women worldwide are infected with human papillomavirus (HPV).1 Persistent HPV infection with high-risk strains such as HPV 6, 11, 16, and 18 cause nearly all cases of cervical cancer and some anal, vaginal, penile, and oropharyngeal cancers.2 An estimated 13,000 cases of invasive cervical cancer will be diagnosed this year in the United States alone.3

Up to 70% of HPV-related cervical cancer cases can be prevented with vaccination. A number of changes have been made to the vaccination schedule within the past few years—patients younger than 15 need only 2 rather than 3 doses, and the vaccine itself can be used in adults up to age 45.

Vaccination and routine cervical cancer screening are both necessary to prevent this disease3 along with effective family and patient counseling. Here, we discuss the most up-to-date HPV vaccination recommendations, current cervical cancer screening guidelines, counseling techniques that increase vaccination acceptance rates, and follow-up protocols for abnormal cervical cancer screening results.

TYPES OF HPV VACCINES

HPV immunization can prevent up to 70% of cases of cervical cancer due to HPV as well as 90% of genital warts.4 The US Food and Drug Administration (FDA) has approved 3 HPV vaccines:

  • Gardasil 9 targets HPV types 6, 11, 16, and 18 along with 31, 33, 45, 52, 58—these cause 90% of cervical cancer cases and most cases of genital warts5—making it the most effective vaccine available; Gardasil 9 is the only HPV vaccine currently available in the United States
  • The bivalent vaccine (Cervarix) targeted HPV 16 and 18 only, and was discontinued in the United States in 2016
  • The quadrivalent HPV vaccine (Gardasil) targeted HPV 16 and 18 as well as 6 and 11, which cause most cases of genital warts; the last available doses in the United States expired in May 2017; it has been replaced by Gardasil 9.

The incidence of cervical cancer in the United States dropped 29% among 15- to 24-year-olds from 2003–2006 when HPV vaccination first started to 2011–2014.6

VACCINE DOSING RECOMMENDATIONS FOR PRIMARY PREVENTION

zhang_hpv_t1.jpg

The Advisory Committee on Immunization Practices (ACIP) revised its HPV vaccine schedule in 2016, when it decreased the necessary doses from 3 to 2 for patients under age 15 and addressed the needs of special patient populations.7 In late 2018, the FDA approved the use of the vaccine in men and women up to age 45. However, no change in guidelines have yet been made (Table 1).

In females, the ACIP recommends starting HPV vaccination at age 11 or 12, but it can be given as early as age 9. A 2-dose schedule is recommended for the 9-valent vaccine before the patient’s 15th birthday (the second dose 6 to 12 months after the first).7 For females who initiate HPV vaccination between ages 15 and 45, a 3-dose schedule is necessary (at 0, 1 to 2, and 6 months).7,8

The change to a 2-dose schedule was prompted by an evaluation of girls ages 9 to 13 randomized to receive either a 2- or 3-dose schedule. Antibody responses with a 2-dose schedule were not inferior to those of young women (ages 16 to 26) who received all 3 doses.9 The geometric mean titer ratios remained noninferior throughout the study period of 36 months.

However, a loss of noninferiority was noted for HPV-18 by 24 months and for HPV-6 by 36 months.9 Thus, further studies are needed to understand the duration of protection with a 2-dose schedule. Nevertheless, decreasing the number of doses makes it a more convenient and cost-effective option for many families.

The recommendations are the same for males except for one notable difference: in males ages 21 to 26, vaccination is not routinely recommended by the ACIP, but rather it is considered a “permissive use” recommendation: ie, the vaccine should be offered and final decisions on administration be made after individualized discussion with the patient.10 Permissive-use status also means the vaccine may not be covered by health insurance. Even though the vaccine is now available to men and women until age 45, many insurance plans do not cover it after age 26.

Children of either sex with a history of sexual abuse should receive their first vaccine dose beginning at age 9.7

Immunocompromised patients should follow the 3-dose schedule regardless of their sex or the age when vaccination was initiated.10

For transgender patients and for men not previously vaccinated who have sex with men, the 3-dose schedule vaccine should be given by the age of 26 (this is a routine recommendation, not a permissive one).8

 

 

CHALLENGES OF VACCINATION

Effective patient and family counseling is important. Even though the first HPV vaccine was approved in 2006, only 34.9% of US adolescents were fully vaccinated by 2015. This was in part because providers did not recommend it, were unfamiliar with it, or had concerns about its safety,11,12 and in part because some parents refused it.

The physician must address any myths regarding HPV vaccination and ensure that parents and patients understand that HPV vaccine is safe and effective. Studies have shown that with high-quality recommendations (ie, the care provider strongly endorses the HPV vaccine, encourages same-day vaccination, and discusses cancer prevention), patients are 9 times more likely to start the HPV vaccination schedule and 3 times more likely to follow through with subsequent doses.13

Providing good family and patient education does not necessarily require spending more counseling time. A recent study showed that spending less time discussing the HPV vaccine can lead to better vaccine coverage.14 The study compared parent HPV vaccine counseling techniques and found that simply informing patients and their families that the HPV vaccine was due was associated with a higher vaccine acceptance rate than inviting conversations about it.14 When providers announced that the vaccine was due, assuming the parents were ready to vaccinate, there was a 5.4% increase in HPV vaccination coverage.14

zhang_hpv_t2.jpg

Conversely, physicians who engaged parents in open-ended discussions about the HPV vaccine did not improve HPV vaccination coverage.14 The authors suggested that providers approach HPV vaccination as if they were counseling patients and families about the need to avoid second-hand smoke or the need to use car seats. If parents or patients resist the presumptive announcement approach, expanded counseling and shared decision-making are appropriate. This includes addressing misconceptions that parents and patients may have about the HPV vaccine. The American Cancer Society lists 8 facts to reference (Table 2).15

SECONDARY PREVENTION: CERVICAL CANCER SCREENING

Since the introduction of the Papanicolaou (Pap) test, US cervical cancer incidence rates have decreased by more than 60%.16 Because almost all cervical cancer is preventable with proper screening, all women ages 21 to 65 should be screened.

zhang_hpv_t3.jpg

Currently, there are 3 options available for cervical cancer screening: the Pap-only test, the Pap-HPV cotest, and the high-risk HPV-only test (Table 3). The latter 2 options detect high-risk HPV genotypes.

Several organizations have screening algorithms that recommend when to use these tests, but the 3 that shape today’s standard of care in cervical cancer screening come from the American College of Obstetricians and Gynecologists (ACOG), the American Society for Colposcopy and Cervical Pathology (ASCCP), and US Preventive Services Task Force (USPSTF).17–19

Pap-only testing is performed every 3 years to screen for cervical neoplasia that might indicate premalignancy.

Pap-HPV cotesting is performed every 5 years in women older than 30 with past normal screening. Until 2018, all 3 organizations recommended cotesting as the preferred screening algorithm for women ages 30 to 65.17–19 Patients with a history of abnormal test results require more frequent testing as recommended by the ASCCP.18

The high-risk HPV-only test utilizes real-time polymerase chain reaction to detect HPV 16, HPV 18, and 12 other HPV genotypes. Only 2 tests are approved by the FDA as stand-alone cervical cancer screening tests—the Roche Cobas HPV test approved in 2014 and the Becton Dickinson Onclarity HPV assay approved in 2018. Other HPV tests that are used in a cotesting strategy should not be used for high-risk HPV-only testing because their performance characteristics may differ.

In 2015, the Addressing the Need for Advanced HPV Diagnostics (ATHENA) study showed that 1 round of high-risk HPV-only screening for women older than 25 was more sensitive than Pap-only or cotesting for stage 3 cervical intraepithelial neoplasia or more severe disease (after 3 years of follow-up).20 Current guidelines from ASCCP18 and ACOG17 state that the high-risk HPV test can be repeated every 3 years (when used to screen by itself) if the woman is older than 25 and has had a normal test result.

zhang_hpv_f1.jpg
%3Cp%3EFigure%201.%3C%2Fp%3E

If the HPV test result is positive for high-risk HPV 16 or 18 genotypes, then immediate colposcopy is indicated; women who test positive for one of the other 12 high-risk subtypes will need to undergo a Pap test to determine the appropriate follow-up (Figure 1).18,21

In 2018, the USPSTF updated its recommendations, noting that for women age 30 to 65, Pap-only testing every 3 years, cotesting every 5 years, or high-risk HPV-only testing every 5 years are all appropriate screening strategies, with the Pap-only or high-risk HPV-only screenings being preferred.19 This is in contrast to ACOG and ASCCP recommendations for cotesting every 5 years, with alternative options of Pap-only or HPV-only testing being done every 3 years.17,18

 

 

Is there a best screening protocol?

The USPSTF reviewed large randomized and observational studies to summarize the effectiveness of the 3 screening strategies and commissioned a decision analysis model to compare the risks, benefits, and costs of the 3 screening algorithms. The guideline statement notes both cotesting and high-risk HPV testing offer similar cancer detection rates: each prevents 1 additional cancer per 1,000 women screened as opposed to Pap-only testing.19

Also, tests that incorporate high-risk HPV screening may offer better detection of cervical adenocarcinoma (which has a worse prognosis than the more common squamous cell carcinoma type). However, both HPV-based screening strategies are more likely to require additional colposcopies for follow-up than Pap-only screening (1,630 colposcopies required for each cancer prevented with high-risk HPV alone, 1,635 with cotesting). Colposcopy is a simple office procedure that causes minimal discomfort to the patient.

The USPSTF guideline also differs in the recommended frequency of high-risk HPV-only testing; a high-risk HPV result should be repeated every 5 years if normal (as opposed to every 3 years as recommended by ACOG and ASCCP).19 The 5-year recommendation is based on analysis modeling, which suggests that performing high-risk HPV-only testing more frequently is unlikely to improve detection rates but will increase the number of screening tests and colposcopies.19

No trial has directly compared cotesting with high-risk HPV testing for more than 2 rounds of screening. The updated USPSTF recommendations are based on modeling estimates and expert opinion, which assesses cost and benefit vs harm in the long term. Also, no high-risk HPV test is currently FDA-approved for every-5-year screening when used by itself.

All 3 cervical cancer screening methods provide highly effective cancer prevention, so it is important for providers to choose the strategy that best fits their practice. The most critical aspect of screening is getting all women screened, no matter which method is used.

It is critical to remember that the screening intervals are intended for patients without symptoms. Those who have new concerns such as bleeding should have a diagnostic Pap done to evaluate their symptoms.

Follow-up of abnormal results

Regardless of the pathway chosen, appropriate follow-up of any abnormal test result is critical to the early detection of cancer. Established follow-up guidelines exist,22,23 but accessing this information can be difficult for the busy clinician. The ASCCP has a mobile phone application that outlines the action steps corresponding to the patient’s age and results of any combination of Pap or HPV testing. The app also includes the best screening algorithms for a particular patient.24

All guidelines agree that cervical cancer screening should start at age 21, regardless of HPV vaccination status or age of sexual initiation.17,18,25 Screening can be discontinued at age 65 for women with normal screening results in the prior decade (3 consecutive negative Pap results or 2 consecutive negative cotest results).23

For women who have had a total hysterectomy and no history of cervical neoplasia, screening should be stopped immediately after the procedure. However, several high-risk groups of women will need continued screening past the age of 65, or after a hysterectomy.

For a woman with a history of stage 2 cervical intraepithelial neoplasia or higher grade lesions, routine screening is continued for an additional 20 years, even if she is over age 65. Pap-only testing every 3 years is acceptable, because the role of HPV testing is unclear after hysterectomy.23 Prior guidelines suggested annual screening in these patients, so the change to every 3 years is notable. Many gynecologic oncologists will recommend that women with a history of cervical cancer continue annual screening indefinitely.

Within the first 2 to 3 years after treatment for high-grade dysplastic changes, annual follow-up is done by the gynecologic oncology team. Providers who offer follow-up during this time frame should keep in communication with the oncology team to ensure appropriate, individualized care. These recommendations are based on expert opinion, so variations in clinical practice may be seen.

Women infected with the human immunodeficiency virus can have Pap-only testing every 3 years, after a series of 3 normal annual Pap results.26 But screening does not stop at age 65.23,26 For patients who are immunosuppressed or have a history of diethylstilbestrol exposure, screening should be done annually indefinitely.23

References
  1. Bruni L, Diaz M, Castellsagué X, Ferrer E, Bosch FX, de Sanjosé S. Cervical human papillomavirus prevalence in 5 continents: meta-analysis of 1 million women with normal cytological findings. J Infect Dis 2010; 202(12):1789–1799. doi:10.1086/657321
  2. de Martel C, Ferlay J, Franceschi S, et al. Global burden of cancer attributable to infections in 2008: a review and synthetic analysis. Lancet Oncol 2012; 13(6):607–615. doi:10.1016/S1470-2045(12)70137-7
  3. American Cancer Society. Key statistics for cervical cancer. www.cancer.org/cancer/cervical-cancer/about/key-statistics.html. Accessed February 14, 2019.
  4. Thaxton L, Waxman AG. Cervical cancer prevention: immunization and screening 2015. Med Clin North Am 2015; 99(3):469–477. doi:10.1016/j.mcna.2015.01.003
  5. McNamara M, Batur P, Walsh JME, Johnson KM. HPV update: vaccination, screening, and associated disease. J Gen Intern Med 2016; 31(11):1360–1366. doi:10.1007/s11606-016-3725-z
  6. Guo F, Cofie LE, Berenson AB. Cervical cancer incidence in young US females after human papillomavirus vaccine introduction. Am J Prev Med 2018; 55(2):197–204. doi:10.1016/j.amepre.2018.03.013
  7. Meites E, Kempe A, Markowitz LE. Use of a 2-dose schedule for human papillomavirus vaccination—updated recommendations of the Advisory Committee on Immunization Practices. MMWR Morb Mortal Wkly Rep 2016; 65(49):1405–1408. doi:10.15585/mmwr.mm6549a5
  8. Centers for Disease Control and Prevention (CDC). Supplemental information and guidance for vaccination providers regarding use of 9-valent HPV vaccine Information for persons who started an HPV vaccination series with quadrivalent or bivalent HPV vaccine. www.cdc.gov/hpv/downloads/9vhpv-guidance.pdf. Accessed February 14, 2019.
  9. Dobson SR, McNeil S, Dionne M, et al. Immunogenicity of 2 doses of HPV vaccine in younger adolescents vs 3 doses in young women: a randomized clinical trial. JAMA 2013; 309(17):1793–1802. doi:10.1001/jama.2013.1625
  10. Markowitz LE, Dunne EF, Saraiya M, et al; Centers for Disease Control and Prevention (CDC). Human papillomavirus vaccination: recommendations of the Advisory Committee on Immunization Practices (ACIP). MMWR Recomm Rep 2014; 63(RR-05):1–30. pmid:25167164
  11. Thompson EL, Rosen BL, Vamos CA, Kadono M, Daley EM. Human papillomavirus vaccination: what are the reasons for nonvaccination among US adolescents? J Adolesc Health 2017; 61(3):288–293. doi:10.1016/j.jadohealth.2017.05.015
  12. Reagan-Steiner S, Yankey D, Jeyarajah J, et al. National, regional, state, and selected local area vaccination coverage among adolescents aged 13-17 years—United States, 2015. MMWR Morb Mortal Wkly Rep 2016; 65(33):850–858. doi:10.15585/mmwr.mm6533a4
  13. Gilkey MB, Calo WA, Moss JL, Shah PD, Marciniak MW, Brewer NT. Provider communication and HPV vaccination: The impact of recommendation quality. Vaccine 2016; 34(9):1187–1192. doi:10.1016/j.vaccine.2016.01.023
  14. Brewer NT, Hall ME, Malo TL, Gilkey MB, Quinn B, Lathren C. Announcements versus conversations to improve HPV vaccination coverage: a randomized trial. Pediatrics 2017; 139(1):e20161764. doi:10.1542/peds.2016-1764
  15. American Cancer Society. HPV vaccine facts. www.cancer.org/cancer/cancer-causes/infectious-agents/hpv/hpv-vaccine-facts-and-fears.html. Accessed February 14, 2019.
  16. National Cancer Institute; Chasan R, Manrow R. Cervical cancer. https://report.nih.gov/nihfactsheets/viewfactsheet.aspx?csid=76. Accessed February 14, 2019.
  17. The American College of Obstetricians and Gynecologists (ACOG). Frequently asked questions. Cervical cancer screening. www.acog.org/Patients/FAQs/Cervical-Cancer-Screening. Accessed February 14, 2019.
  18. Saslow D, Solomon D, Lawson HW, et al; American Cancer Society; American Society for Colposcopy and Cervical Pathology; American Society for Clinical Pathology. American Cancer Society, American Society for Colposcopy and Cervical Pathology, and American Society for Clinical Pathology screening guidelines for the prevention and early detection of cervical cancer. Am J Clin Pathol 2012; 137(4):516–542. doi:10.1309/AJCPTGD94EVRSJCG
  19. US Preventive Services Task Force; Curry SJ, Krist AH, Owens DK, et al. Screening for cervical cancer: US Preventive Services Task Force Recommendation Statement. JAMA 2018; 320(7):674–686. doi:10.1001/jama.2018.10897
  20. Wright TC, Stoler MH, Behrens CM, Sharma A, Zhang G, Wright TL. Primary cervical cancer screening with human papillomavirus: end of study results from the ATHENA study using HPV as the first-line screening test. Gynecol Oncol 2015; 136(2):189–197. doi:10.1016/j.ygyno.2014.11.076
  21. Huh WK, Ault KA, Chelmow D, et al. Use of primary high-risk human papillomavirus testing for cervical cancer screening: interim clinical guidance. Obstet Gynecol 2015; 125(2):330–337. doi:10.1097/AOG.0000000000000669
  22. Massad LS, Einstein MH, Huh WK, et al; 2012 ASCCP Consensus Guidelines Conference. 2012 updated consensus guidelines for the management of abnormal cervical cancer screening tests and cancer precursors. Obstet Gynecol 2013; 121(4):829–846. doi:10.1097/AOG.0b013e3182883a34
  23. Committee on Practice Bulletins—Gynecology. Practice Bulletin No. 168: cervical cancer screening and prevention. Obstet Gynecol 2016; 128(4):e111–e130. doi:10.1097/AOG.0000000000001708
  24. ASCCP. Mobile app. http://www.asccp.org/store-detail2/asccp-mobile-app. Accessed February 14, 2019.
  25. USPSTF. Draft recommendation: cervical cancer: screening. www.uspreventiveservicestaskforce.org/Page/Document/draft-recommendation-statement/cervical-cancer-screening2. Accessed February 14, 2019.
  26. Masur H, Brooks JT, Benson CA, Holmes KK, Pau AK, Kaplan JE; National Institutes of Health; Centers for Disease Control and Prevention; HIV Medicine Association of the Infectious Diseases Society of America. Prevention and treatment of opportunistic infections in HIV-infected adults and adolescents: Updated guidelines from the Centers for Disease Control and Prevention, National Institutes of Health, and HIV Medicine Association of the Infectious Diseases Society of America. Clin Infect Dis 2014; 58(9):1308–1311. doi:10.1093/cid/ciu094
References
  1. Bruni L, Diaz M, Castellsagué X, Ferrer E, Bosch FX, de Sanjosé S. Cervical human papillomavirus prevalence in 5 continents: meta-analysis of 1 million women with normal cytological findings. J Infect Dis 2010; 202(12):1789–1799. doi:10.1086/657321
  2. de Martel C, Ferlay J, Franceschi S, et al. Global burden of cancer attributable to infections in 2008: a review and synthetic analysis. Lancet Oncol 2012; 13(6):607–615. doi:10.1016/S1470-2045(12)70137-7
  3. American Cancer Society. Key statistics for cervical cancer. www.cancer.org/cancer/cervical-cancer/about/key-statistics.html. Accessed February 14, 2019.
  4. Thaxton L, Waxman AG. Cervical cancer prevention: immunization and screening 2015. Med Clin North Am 2015; 99(3):469–477. doi:10.1016/j.mcna.2015.01.003
  5. McNamara M, Batur P, Walsh JME, Johnson KM. HPV update: vaccination, screening, and associated disease. J Gen Intern Med 2016; 31(11):1360–1366. doi:10.1007/s11606-016-3725-z
  6. Guo F, Cofie LE, Berenson AB. Cervical cancer incidence in young US females after human papillomavirus vaccine introduction. Am J Prev Med 2018; 55(2):197–204. doi:10.1016/j.amepre.2018.03.013
  7. Meites E, Kempe A, Markowitz LE. Use of a 2-dose schedule for human papillomavirus vaccination—updated recommendations of the Advisory Committee on Immunization Practices. MMWR Morb Mortal Wkly Rep 2016; 65(49):1405–1408. doi:10.15585/mmwr.mm6549a5
  8. Centers for Disease Control and Prevention (CDC). Supplemental information and guidance for vaccination providers regarding use of 9-valent HPV vaccine Information for persons who started an HPV vaccination series with quadrivalent or bivalent HPV vaccine. www.cdc.gov/hpv/downloads/9vhpv-guidance.pdf. Accessed February 14, 2019.
  9. Dobson SR, McNeil S, Dionne M, et al. Immunogenicity of 2 doses of HPV vaccine in younger adolescents vs 3 doses in young women: a randomized clinical trial. JAMA 2013; 309(17):1793–1802. doi:10.1001/jama.2013.1625
  10. Markowitz LE, Dunne EF, Saraiya M, et al; Centers for Disease Control and Prevention (CDC). Human papillomavirus vaccination: recommendations of the Advisory Committee on Immunization Practices (ACIP). MMWR Recomm Rep 2014; 63(RR-05):1–30. pmid:25167164
  11. Thompson EL, Rosen BL, Vamos CA, Kadono M, Daley EM. Human papillomavirus vaccination: what are the reasons for nonvaccination among US adolescents? J Adolesc Health 2017; 61(3):288–293. doi:10.1016/j.jadohealth.2017.05.015
  12. Reagan-Steiner S, Yankey D, Jeyarajah J, et al. National, regional, state, and selected local area vaccination coverage among adolescents aged 13-17 years—United States, 2015. MMWR Morb Mortal Wkly Rep 2016; 65(33):850–858. doi:10.15585/mmwr.mm6533a4
  13. Gilkey MB, Calo WA, Moss JL, Shah PD, Marciniak MW, Brewer NT. Provider communication and HPV vaccination: The impact of recommendation quality. Vaccine 2016; 34(9):1187–1192. doi:10.1016/j.vaccine.2016.01.023
  14. Brewer NT, Hall ME, Malo TL, Gilkey MB, Quinn B, Lathren C. Announcements versus conversations to improve HPV vaccination coverage: a randomized trial. Pediatrics 2017; 139(1):e20161764. doi:10.1542/peds.2016-1764
  15. American Cancer Society. HPV vaccine facts. www.cancer.org/cancer/cancer-causes/infectious-agents/hpv/hpv-vaccine-facts-and-fears.html. Accessed February 14, 2019.
  16. National Cancer Institute; Chasan R, Manrow R. Cervical cancer. https://report.nih.gov/nihfactsheets/viewfactsheet.aspx?csid=76. Accessed February 14, 2019.
  17. The American College of Obstetricians and Gynecologists (ACOG). Frequently asked questions. Cervical cancer screening. www.acog.org/Patients/FAQs/Cervical-Cancer-Screening. Accessed February 14, 2019.
  18. Saslow D, Solomon D, Lawson HW, et al; American Cancer Society; American Society for Colposcopy and Cervical Pathology; American Society for Clinical Pathology. American Cancer Society, American Society for Colposcopy and Cervical Pathology, and American Society for Clinical Pathology screening guidelines for the prevention and early detection of cervical cancer. Am J Clin Pathol 2012; 137(4):516–542. doi:10.1309/AJCPTGD94EVRSJCG
  19. US Preventive Services Task Force; Curry SJ, Krist AH, Owens DK, et al. Screening for cervical cancer: US Preventive Services Task Force Recommendation Statement. JAMA 2018; 320(7):674–686. doi:10.1001/jama.2018.10897
  20. Wright TC, Stoler MH, Behrens CM, Sharma A, Zhang G, Wright TL. Primary cervical cancer screening with human papillomavirus: end of study results from the ATHENA study using HPV as the first-line screening test. Gynecol Oncol 2015; 136(2):189–197. doi:10.1016/j.ygyno.2014.11.076
  21. Huh WK, Ault KA, Chelmow D, et al. Use of primary high-risk human papillomavirus testing for cervical cancer screening: interim clinical guidance. Obstet Gynecol 2015; 125(2):330–337. doi:10.1097/AOG.0000000000000669
  22. Massad LS, Einstein MH, Huh WK, et al; 2012 ASCCP Consensus Guidelines Conference. 2012 updated consensus guidelines for the management of abnormal cervical cancer screening tests and cancer precursors. Obstet Gynecol 2013; 121(4):829–846. doi:10.1097/AOG.0b013e3182883a34
  23. Committee on Practice Bulletins—Gynecology. Practice Bulletin No. 168: cervical cancer screening and prevention. Obstet Gynecol 2016; 128(4):e111–e130. doi:10.1097/AOG.0000000000001708
  24. ASCCP. Mobile app. http://www.asccp.org/store-detail2/asccp-mobile-app. Accessed February 14, 2019.
  25. USPSTF. Draft recommendation: cervical cancer: screening. www.uspreventiveservicestaskforce.org/Page/Document/draft-recommendation-statement/cervical-cancer-screening2. Accessed February 14, 2019.
  26. Masur H, Brooks JT, Benson CA, Holmes KK, Pau AK, Kaplan JE; National Institutes of Health; Centers for Disease Control and Prevention; HIV Medicine Association of the Infectious Diseases Society of America. Prevention and treatment of opportunistic infections in HIV-infected adults and adolescents: Updated guidelines from the Centers for Disease Control and Prevention, National Institutes of Health, and HIV Medicine Association of the Infectious Diseases Society of America. Clin Infect Dis 2014; 58(9):1308–1311. doi:10.1093/cid/ciu094
Issue
Cleveland Clinic Journal of Medicine - 86(3)
Issue
Cleveland Clinic Journal of Medicine - 86(3)
Page Number
173-178
Page Number
173-178
Publications
Publications
Topics
Article Type
Display Headline
Human papillomavirus in 2019: An update on cervical cancer prevention and screening guidelines
Display Headline
Human papillomavirus in 2019: An update on cervical cancer prevention and screening guidelines
Legacy Keywords
human papillomavirus, HPV, cervical cancer, screening, immunization, vaccination, HPV vaccine, Gardasil, Papanicolaou test, Pap test, HPV test, screening, Salina Zhang, Pelin Batur
Legacy Keywords
human papillomavirus, HPV, cervical cancer, screening, immunization, vaccination, HPV vaccine, Gardasil, Papanicolaou test, Pap test, HPV test, screening, Salina Zhang, Pelin Batur
Sections
Inside the Article

KEY POINTS

  • Immunization against HPV can prevent up to 70% of HPV-related cervical cancer cases.
  • Gardasil 9 is the only HPV vaccine currently available in the United States and is now approved for use in males and females between the ages of 9 and 45.
  • In girls and boys younger than 15, a 2-dose schedule is recommended; patients ages 15 through 45 require 3 doses.
  • Vaccine acceptance rates are highest when primary care providers announce that the vaccine is due rather than invite open-ended discussions.
  • Regular cervical cancer screening is an important preventive tool and should be performed using the Papanicolaou (Pap) test, the high-risk HPV-only test, or the Pap-HPV cotest.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Thu, 02/28/2019 - 12:15
Un-Gate On Date
Thu, 02/28/2019 - 12:15
Use ProPublica
CFC Schedule Remove Status
Thu, 02/28/2019 - 12:15
Hide sidebar & use full width
render the right sidebar.
Article PDF Media

Three neglected numbers in the CBC: The RDW, MPV, and NRBC count

Article Type
Changed
Wed, 07/03/2019 - 10:18
Display Headline
Three neglected numbers in the CBC: The RDW, MPV, and NRBC count

The complete blood cell count (CBC) is one of the most frequently ordered laboratory tests in both the inpatient and outpatient settings. Not long ago, the CBC required peering through a microscope and counting the red blood cells, white blood cells, and platelets. These 3 numbers are still the primary purpose of the test.

Now, with automated counters, the CBC report also contains other numbers that delineate characteristics of each cell type. For example:

The mean corpuscular volume is the average volume of red blood cells. Providers use it to classify anemia as either microcytic, normocytic, or macrocytic, each with its own differential diagnosis.

The differential white blood cell count provides absolute counts and relative percentages of each type of leukocyte. For example, the absolute neutrophil count is an important measure of immunocompetence.

But other values in the CBC may be overlooked, even though they can provide important information. Here, we highlight 3 of them:

  • The red blood cell distribution width (RDW)
  • The mean platelet volume (MPV)
  • The nucleated red blood cell (NRBC) count.

In addition to describing their diagnostic utility, we also discuss emerging evidence of their potential prognostic significance in hematologic and nonhematologic disorders. By incorporating an awareness of their value in clinical practice, providers can maximize the usefulness of the CBC.

RED BLOOD CELL DISTRIBUTION WIDTH

may_interpretingthecbc_f1.jpg
%3Cp%3EFigure%201.%20A%3A%20Example%20of%20a%20normal%20red%20blood%20cell%20distribution%20width%20(RDW)%20of%2013.5%25%20(red%20line)%20in%20a%20patient%20with%20a%20normal%20complete%20blood%20cell%20count.%20B%3A%20Example%20of%20an%20increased%20RDW%20of%2028.8%25%20in%20a%20patient%20with%20iron%20deficiency%20shortly%20after%20initiation%20of%20iron%20supplementation.%3C%2Fp%3E

The RDW is a measure of variation (anisocytosis) in the size of the circulating red cells. The term “width” is misleading, as the value is not derived from the width of the red blood cell, but rather from the width of the distribution curve of the corpuscular volume (Figure 1). Therefore, a normal RDW means that the cells are all about the same size, while a high RDW means they vary widely in size.

The RDW can be calculated either as a coefficient of variation, with a reference range of 11% to 16% depending on the laboratory, or, less often, as a standard deviation, with a reference range of 39 to 46 fL.

The RDW can differentiate between causes of anemia

A high RDW is often found in nutritional deficiencies of iron, vitamin B12, and folate. This information is helpful in differentiating the cause of microcytic anemia, as a high RDW suggests iron-deficiency anemia while a normal RDW suggests thalassemia.1 In iron deficiency, the RDW often rises before the mean corpuscular volume falls, serving as an early diagnostic clue.

The RDW can also be high after recent hemorrhage or rapid hemolysis, as the acute drop in hemoglobin results in increased production of reticulocytes, which are larger than mature erythrocytes.

Because a range of disorders can elevate the RDW, reviewing the peripheral blood smear is an important next step in the diagnostic evaluation, specifically looking for reticulocytes, microspherocytes, and other abnormal red blood cells contributing to the RDW elevation.

A normal RDW is less diagnostically useful. It indicates the red blood cells are of uniform size, but they may be uniformly small or large depending on how long the anemia has persisted. Since red cells circulate for only about 120 days, patients who have severe iron-deficiency anemia for months to years are expected to have a normal rather than a high RDW, as their red cells of normal size have all been replaced by microcytes.

A low RDW is not consistently associated with any hematologic disorder.

RDW may have prognostic value

Emerging data suggest that the RDW may also have prognostic value in nonhematologic diseases. In a retrospective study of 15,852 adult participants in the Third National Health and Nutrition Examination Survey (1988–1994), a higher RDW was associated with a higher risk of death, with the all-cause mortality rate increasing by 23% for every 1% increment in RDW.2

This correlation is particularly prominent in cardiac disorders. In 2 large retrospective studies of patients with symptomatic heart failure, a higher RDW was a strong predictor of morbidity and death (hazard ratio 1.17 per 1-standard deviation increase, P < .001), even stronger than more commonly used variables such as ejection fraction, New York Heart Association functional class, and renal function.3

In a retrospective analysis of 4,111 patients with myocardial infarction, the degree of RDW elevation correlated with the risk of repeat nonfatal myocardial infarction, coronary death, new symptomatic heart failure, and stroke.4

It is hypothesized that high RDW may reflect poor cell membrane integrity from altered cholesterol content, which in turn has deleterious effects on multiple organ systems and is therefore associated with adverse outcomes.5

Currently, using the RDW to assess prognosis remains investigational, and how best to interpret it in daily practice requires further study.

 

 

MEAN PLATELET VOLUME

The MPV, ie, the average size of platelets, is reported in femtoliters (fL). Because the MPV varies depending on the instrument used, each laboratory has a unique reference range, usually about 8 to 12 fL. The MPV must be interpreted in conjunction with the platelet count; the product of the MPV and platelet count is called the total platelet mass.

Using the MPV to find the cause of thrombocytopenia

The MPV can be used to help narrow the differential diagnosis of thrombocytopenia. For example, it is high in thrombocytopenia resulting from peripheral destruction, as in immune thrombocytopenic purpura. This is because as platelets are lost, thrombopoietin production increases and new, larger platelets are released from healthy megakaryocytes in an attempt to increase the total platelet mass.

may_interpretingthecbc_f2.jpg
%3Cp%3EFigure%202.%20Giant%20platelets%20(thin%20arrows)%2C%20normal%20sized%20platelets%20(dotted%20arrows)%2C%20and%20a%20nucleated%20red%20blood%20cell%20(thick%20arrow)%20in%20a%20patient%20with%20myelofibrosis%20and%20extensive%20extramedullary%20hematopoiesis.%3C%2Fp%3E

In contrast, the MPV is low in patients with thrombocytopenia due to megakaryocyte hypoplasia, as malfunctioning megakaryocytes cannot maintain the total platelet mass, and any platelets produced remain small. This distinction can be obscured in the setting of splenomegaly, as larger platelets are more easily sequestered in the spleen and the MPV may therefore be low or normal.

The MPV can also be used to differentiate congenital thrombocytopenic disorders, which can be characterized by either a high MPV (eg, gray platelet syndrome, Bernard-Soulier syndrome) or a low MPV (eg, Wiskott-Aldrich syndrome) (Figure 2).

MPV may have prognostic value

Evidence suggests that the MPV also has potential prognostic value, particularly in vascular disease, as larger platelets are hypothesized to have increased hemostatic potential.

In a large meta-analysis of patients with coronary artery disease, a high MPV was associated with worse outcomes; the risk of death or myocardial infarction was 17% higher in those with a high MPV (the threshold ranged from 8.4 to 11.7 fL in the different studies) than in those with a low MPV.6

In a study of 213 patients with non-ST-segment elevation myocardial infarction, the risk of significant coronary artery disease was 4.18 times higher in patients with a high MPV and a high troponin level than in patients with a normal MPV and a high troponin.7 The authors suggested that a high MPV may help identify patients at highest risk of significant coronary artery disease who would benefit from invasive studies (ie, coronary angiography).

This correlation has also been observed in other forms of vascular disease. In 261 patients who underwent carotid angioplasty and stenting, an MPV higher than 10.1 fL was associated with a risk of in-stent restenosis more than 3 times higher.8

The MPV has also been found to be higher in patients with type 2 diabetes than in controls, particularly in those with microvascular complications such as retinopathy or microalbuminuria.9

Conversely, in patients with cancer, a low MPV appears to be associated with a poor prognosis. In a retrospective analysis of 236 patients with esophageal cancer, those who had an MPV of 7.4 fL or less had significantly shorter overall survival than patients with an MPV higher than 7.4 fL.10

A low MPV has also been associated with an increased risk of venous thromoboembolism in patients with cancer. In a prospective observational cohort study of 1,544 patients, the 2-year probability of venous thromboembolism was 9% in patients with an MPV less than 10.8 fL, compared with 5.5% in those with higher MPV values. The 2-year overall survival rate was also higher in patients with high MPV than in those with low MPV, at 64.7% vs 55.7%, respectively (P = .001).11

But the MPV is far from a perfect clinical metric. Since its measurement is subject to significant laboratory variation, an abnormal value should always be confirmed with evaluation of a peripheral blood smear. Furthermore, it is unclear why a high MPV portends poor prognosis in patients without cancer, whereas the opposite is true in patients with cancer. Therefore, its role in prognostication remains investigational, and further studies are essential to determine its appropriate usefulness in clinical practice.12

NUCLEATED RED BLOOD CELL COUNT

NRBCs are immature red blood cell precursors not present in the circulation of healthy adults. During erythropoiesis, the common myeloid progenitor cell first differentiates into a proerythroblast; subsequently, the chromatin in the nucleus of the proerythroblast gradually condenses until it becomes an orthochromatic erythroblast, also known as a nucleated red cell (Figure 2). Once the nucleus is expelled, the cell is known as a reticulocyte, which ultimately becomes a mature erythrocyte.

Healthy newborns have circulating NRBCs that rapidly disappear within a few weeks of birth. However, NRBCs can return to the circulation in a variety of disease states.

Causes of NRBCs

Brisk hemolysis or rapid blood loss can cause NRBCs to be released into the blood as erythropoiesis increases in an attempt to compensate for acute anemia.

Damage or stress to the bone marrow also causes NRBCs to be released into the peripheral blood, as is often the case in hematologic diseases. In a study of 478 patients with hematologic diseases, the frequency of NRBC positivity at diagnosis was highest in patients with chronic myeloid leukemia (100%), acute leukemia (62%), and myelodysplastic syndromes (45%).13 NRBCs also appeared at higher frequencies during chemotherapy in other hematologic conditions, such as hemophagocytic lymphohistiocytosis.

The mechanism by which NRBCs are expelled from the bone marrow is unclear, though studies have suggested that inflammation or hypoxia or both cause increased hematopoietic stress, resulting in the release of immature red cells. Increased concentrations of inflammatory cytokines (interleukin 6 and interleukin 3) and erythropoietin in the plasma and decreased arterial oxygen partial tension have been reported in patients with circulating NRBCs.14,15

Because they are associated with hematologic disorders, the finding of NRBCs should prompt evaluation of a peripheral smear to assess for abnormalities in other cell lines.

The NRBC count and prognosis

In critically ill patients, peripheral NRBCs can also indicate life-threatening conditions.

In a study of 421 adult intensive care patients, the in-hospital mortality rate was 42% in those with peripheral NRBCs vs 5.9% in those without them.16 Further, the higher the NRBC count and the more days that NRBCs were reported in the CBC, the higher the risk of death.

In adults with acute respiratory distress syndrome, the finding of any NRBCs in the peripheral blood was an independent risk factor for death, and an NRBC count higher than 220 cells/µL was associated with a more than 3-fold higher risk of death.17

Daily screening in patients in surgical intensive care units revealed that NRBCs appeared an average of 9 days before death, consistent with an early marker of impending decline.18

In another study,19 the risk of death within 90 days of hospital discharge was higher in NRBC-positive patients, reaching 21.9% in those who had a count higher than 200 cells/µL. The risk of unplanned hospital readmission within 30 days was also increased.

Leukoerythroblastosis

The combination of NRBCs and immature white blood cells (eg, myelocytes, metamyelocytes) is called leukoerythroblastosis.

Leukoerythroblastosis is classically seen in myelophthisic anemias in which hematopoietic cells in the marrow are displaced by fibrosis, tumor, or other space-occupying processes, but it can also occur in any situation of acute marrow stress, including critical illness.

In addition, leukoerythroblastosis appears in a rare complication of sickle cell hemoglobinopathies: bone marrow necrosis with fat embolism syndrome.20,21 As the marrow necroses, fat emboli are released in the systemic circulation causing micro- and macrovascular occlusions and multiorgan failure. The largest case series in the literature reports 58 patients with bone marrow necrosis with fat embolism syndrome.22

At our institution, we have seen 18 patients with this condition in the past 8 years, with the frequency of diagnosis increasing with heightened awareness of the disorder. We have found that leukoerythroblastosis is often an early marker of this unrecognized syndrome and can prompt emergency red cell exchange, which is considered to be lifesaving in this condition.22

These examples and many others show that the presence of NRBCs in the CBC can serve as an important clinical warning.

OLD TESTS CAN STILL BE USEFUL

The CBC provides much more than simple cell counts; it is a rich collection of information related to each blood cell. These days, with new diagnostic tests and prognostic tools based on molecular analysis, it is important to not overlook the value of the tests clinicians have been ordering for generations.

The RDW, MPV, and NRBC count will not likely provide definitive or flawless diagnostic or prognostic information, but when understood and used correctly, they provide readily available, cost-effective, and useful data that can supplement and guide clinical decision-making. By understanding the CBC more fully, providers can maximize the truly complete nature of this routine laboratory test.

References
  1. Lima CS, Reis AR, Grotto HZ, Saad ST, Costa FF. Comparison of red cell distribution width and a red cell discriminant function incorporating volume dispersion for distinguishing iron deficiency from beta thalassemia trait in patients with microcytosis. Sao Paulo Med J 1996; 114(5):1265–1269. pmid:9239926
  2. Perlstein TS, Weuve J, Pfeffer MA, Beckman JA. Red blood cell distribution width and mortality risk in a community-based prospective cohort. Arch Intern Med 2009; 169(6):588–594. doi:10.1001/archinternmed.2009.55
  3. Felker GM, Allen LA, Pocock SJ, et al; CHARM Investigators. Red cell distribution width as a novel prognostic marker in heart failure: data from the CHARM Program and the Duke Databank. J Am Coll Cardiol 2007; 50(1):40–47. doi:10.1016/j.jacc.2007.02.067
  4. Tonelli M, Sacks F, Arnold M, Moye L, Davis B, Pfeffer M; for the Cholesterol and Recurrent Events (CARE) Trial Investigators. Relation between red blood cell distribution width and cardiovascular event rate in people with coronary disease. Circulation 2008; 117(2):163–168. doi:10.1161/CIRCULATIONAHA.107.727545
  5. Goldstein MR, Mascitelli L, Pezzetta F. Is red cell distribution width a marker of overall membrane integrity? [Letter] Arch Intern Med 2009; 169(16):1539–1540. doi:10.1001/archinternmed.2009.275
  6. Sansanaydhu N, Numthavaj P, Muntham D, et al. Prognostic effect of mean platelet volume in patients with coronary artery disease. A systematic review and meta-analysis. Thromb Haemost 2015; 114(6):1299–1309. doi:10.1160/TH15-04-0280
  7. Taskesen T, Sekhon H, Wroblewski I, et al. Usefulness of mean platelet volume to predict significant coronary artery disease in patients with non-ST-elevation acute coronary syndromes. Am J Cardiol 2017; 119(2):192–196. doi:10.1016/j.amjcard.2016.09.042
  8. Dai Z, Gao J, Li S, et al. Mean platelet volume as a predictor for restenosis after carotid angioplasty and stenting. Stroke 2018; 49(4):872–876. doi:10.1161/STROKEAHA.117.019748
  9. Papanas N, Symeonidis G, Maltezos E, et al. Mean platelet volume in patients with type 2 diabetes mellitus. Platelets 2004; 15(8):475–478. doi:10.1080/0953710042000267707
  10. Shen W, Cui MM, Wang X, Wang RT. Reduced mean platelet volume is associated with poor prognosis in esophageal cancer. Cancer Biomark 2018; 22(3):559–563. doi:10.3233/CBM-181231
  11. Riedl J, Kaider A, Reitter EM, et al. Association of mean platelet volume with risk of venous thromboembolism and mortality in patients with cancer. Results from the Vienna Cancer and Thrombosis Study (CATS). Thromb Haemost 2014; 111(4):670–678. doi:10.1160/TH13-07-0603
  12. Tsiara S, Elisaf M, Jagroop IA, Mikhailidis DP. Platelets as predictors of vascular risk: is there a practical index of platelet activity? Clin Appl Thromb Hemost 2003; 9(3):177–190. pmid:14507105
  13. Danise P, Maconi M, Barrella F, et al. Evaluation of nucleated red blood cells in the peripheral blood of hematological diseases. Clin Chem Lab Med 2011; 50(2):357–360. doi:10.1515/CCLM.2011.766
  14. Stachon A, Bolulul O, Holland-Letz T, Krieg M. Association between nucleated red blood cells in blood and the levels of erythropoietin, interleukin 3, interleukin 6, and interleukin 12p70. Shock 2005; 24(1):34–39. pmid:15988318
  15. Kuert S, Holland-Letz T, Friese J, Stachon A. Association of nucleated red blood cells in blood and arterial oxygen partial tension. Clin Chem Lab Med 2011; 49(2):257–263. doi:10.1515/CCLM.2011.041
  16. Stachon A, Holland-Letz T, Krieg M. In-hospital mortality of intensive care patients with nucleated red blood cells in blood. Clin Chem Lab Med 2004; 42(8):933–938. doi:10.1515/CCLM.2004.151
  17. Menk M, Giebelhäuser L, Vorderwülbecke G, et al. Nucleated red blood cells as predictors of mortality in patients with acute respiratory distress syndrome (ARDS): an observational study. Ann Intensive Care 2018; 8(1):42. doi:10.1186/s13613-018-0387-5
  18. Stachon A, Kempf R, Holland-Letz T, Friese J, Becker A, Krieg M. Daily monitoring of nucleated red blood cells in the blood of surgical intensive care patients. Clin Chim Acta 2006; 366(1–2):329–335. doi:10.1016/j.cca.2005.11.022
  19. Purtle SW, Horkan CM, Moromizato T, Gibbons FK, Christopher KB. Nucleated red blood cells, critical illness survivors and postdischarge outcomes: a cohort study. Crit Care 2017; 21(1):154. doi:10.1186/s13054-017-1724-z
  20. May J, Sullivan JC, LaVie D, LaVie K, Marques MB. Inside out: bone marrow necrosis and fat embolism complicating sickle-beta+ thalassemia. Am J Med 2016; 129(12):e321–e324. doi:10.1016/j.amjmed.2016.05.027
  21. Gangaraju R, Reddy VV, Marques MB. Fat embolism syndrome secondary to bone marrow necrosis in patients with hemoglobinopathies. South Med J 2016; 109(9):549–553. doi:10.14423/SMJ.0000000000000520
  22. Tsitsikas DA, Gallinella G, Patel S, Seligman H, Greaves P, Amos RJ. Bone marrow necrosis and fat embolism syndrome in sickle cell disease: increased susceptibility of patients with non-SS genotypes and a possible association with human parvovirus B19 infection. Blood Rev 2014; 28(1):23–30. doi:10.1016/j.blre.2013.12.002
Article PDF
Author and Disclosure Information

Jori E. May, MD
Department of Medicine, University of Alabama, Birmingham

Marisa B. Marques, MD
Department of Pathology, University of Alabama, Birmingham

Vishnu V.B. Reddy, MD
Department of Pathology, University of Alabama, Birmingham

Radhika Gangaraju, MD
Department of Medicine, University of Alabama, Birmingham

Address: Jori E. May, MD, Department of Medicine, University of Alabama, 1720 2nd Avenue South, NP 2565, Birmingham, AL 35294; jemay@uabmc.edu

Issue
Cleveland Clinic Journal of Medicine - 86(3)
Publications
Topics
Page Number
167-172
Legacy Keywords
complete blood cell count, CBC, red cell distribution width, RDW, mean platelet volume, MPV, nucleated red blood cell count, NRBC, anemia, thrombocytopenia, iron deficiency, thalassemia, blood test, prognosis, leukoerythroblastosis, Jori May, Marisa Marques, Vishnu Reddy, Radhika Gangaraju
Sections
Author and Disclosure Information

Jori E. May, MD
Department of Medicine, University of Alabama, Birmingham

Marisa B. Marques, MD
Department of Pathology, University of Alabama, Birmingham

Vishnu V.B. Reddy, MD
Department of Pathology, University of Alabama, Birmingham

Radhika Gangaraju, MD
Department of Medicine, University of Alabama, Birmingham

Address: Jori E. May, MD, Department of Medicine, University of Alabama, 1720 2nd Avenue South, NP 2565, Birmingham, AL 35294; jemay@uabmc.edu

Author and Disclosure Information

Jori E. May, MD
Department of Medicine, University of Alabama, Birmingham

Marisa B. Marques, MD
Department of Pathology, University of Alabama, Birmingham

Vishnu V.B. Reddy, MD
Department of Pathology, University of Alabama, Birmingham

Radhika Gangaraju, MD
Department of Medicine, University of Alabama, Birmingham

Address: Jori E. May, MD, Department of Medicine, University of Alabama, 1720 2nd Avenue South, NP 2565, Birmingham, AL 35294; jemay@uabmc.edu

Article PDF
Article PDF
Related Articles

The complete blood cell count (CBC) is one of the most frequently ordered laboratory tests in both the inpatient and outpatient settings. Not long ago, the CBC required peering through a microscope and counting the red blood cells, white blood cells, and platelets. These 3 numbers are still the primary purpose of the test.

Now, with automated counters, the CBC report also contains other numbers that delineate characteristics of each cell type. For example:

The mean corpuscular volume is the average volume of red blood cells. Providers use it to classify anemia as either microcytic, normocytic, or macrocytic, each with its own differential diagnosis.

The differential white blood cell count provides absolute counts and relative percentages of each type of leukocyte. For example, the absolute neutrophil count is an important measure of immunocompetence.

But other values in the CBC may be overlooked, even though they can provide important information. Here, we highlight 3 of them:

  • The red blood cell distribution width (RDW)
  • The mean platelet volume (MPV)
  • The nucleated red blood cell (NRBC) count.

In addition to describing their diagnostic utility, we also discuss emerging evidence of their potential prognostic significance in hematologic and nonhematologic disorders. By incorporating an awareness of their value in clinical practice, providers can maximize the usefulness of the CBC.

RED BLOOD CELL DISTRIBUTION WIDTH

may_interpretingthecbc_f1.jpg
%3Cp%3EFigure%201.%20A%3A%20Example%20of%20a%20normal%20red%20blood%20cell%20distribution%20width%20(RDW)%20of%2013.5%25%20(red%20line)%20in%20a%20patient%20with%20a%20normal%20complete%20blood%20cell%20count.%20B%3A%20Example%20of%20an%20increased%20RDW%20of%2028.8%25%20in%20a%20patient%20with%20iron%20deficiency%20shortly%20after%20initiation%20of%20iron%20supplementation.%3C%2Fp%3E

The RDW is a measure of variation (anisocytosis) in the size of the circulating red cells. The term “width” is misleading, as the value is not derived from the width of the red blood cell, but rather from the width of the distribution curve of the corpuscular volume (Figure 1). Therefore, a normal RDW means that the cells are all about the same size, while a high RDW means they vary widely in size.

The RDW can be calculated either as a coefficient of variation, with a reference range of 11% to 16% depending on the laboratory, or, less often, as a standard deviation, with a reference range of 39 to 46 fL.

The RDW can differentiate between causes of anemia

A high RDW is often found in nutritional deficiencies of iron, vitamin B12, and folate. This information is helpful in differentiating the cause of microcytic anemia, as a high RDW suggests iron-deficiency anemia while a normal RDW suggests thalassemia.1 In iron deficiency, the RDW often rises before the mean corpuscular volume falls, serving as an early diagnostic clue.

The RDW can also be high after recent hemorrhage or rapid hemolysis, as the acute drop in hemoglobin results in increased production of reticulocytes, which are larger than mature erythrocytes.

Because a range of disorders can elevate the RDW, reviewing the peripheral blood smear is an important next step in the diagnostic evaluation, specifically looking for reticulocytes, microspherocytes, and other abnormal red blood cells contributing to the RDW elevation.

A normal RDW is less diagnostically useful. It indicates the red blood cells are of uniform size, but they may be uniformly small or large depending on how long the anemia has persisted. Since red cells circulate for only about 120 days, patients who have severe iron-deficiency anemia for months to years are expected to have a normal rather than a high RDW, as their red cells of normal size have all been replaced by microcytes.

A low RDW is not consistently associated with any hematologic disorder.

RDW may have prognostic value

Emerging data suggest that the RDW may also have prognostic value in nonhematologic diseases. In a retrospective study of 15,852 adult participants in the Third National Health and Nutrition Examination Survey (1988–1994), a higher RDW was associated with a higher risk of death, with the all-cause mortality rate increasing by 23% for every 1% increment in RDW.2

This correlation is particularly prominent in cardiac disorders. In 2 large retrospective studies of patients with symptomatic heart failure, a higher RDW was a strong predictor of morbidity and death (hazard ratio 1.17 per 1-standard deviation increase, P < .001), even stronger than more commonly used variables such as ejection fraction, New York Heart Association functional class, and renal function.3

In a retrospective analysis of 4,111 patients with myocardial infarction, the degree of RDW elevation correlated with the risk of repeat nonfatal myocardial infarction, coronary death, new symptomatic heart failure, and stroke.4

It is hypothesized that high RDW may reflect poor cell membrane integrity from altered cholesterol content, which in turn has deleterious effects on multiple organ systems and is therefore associated with adverse outcomes.5

Currently, using the RDW to assess prognosis remains investigational, and how best to interpret it in daily practice requires further study.

 

 

MEAN PLATELET VOLUME

The MPV, ie, the average size of platelets, is reported in femtoliters (fL). Because the MPV varies depending on the instrument used, each laboratory has a unique reference range, usually about 8 to 12 fL. The MPV must be interpreted in conjunction with the platelet count; the product of the MPV and platelet count is called the total platelet mass.

Using the MPV to find the cause of thrombocytopenia

The MPV can be used to help narrow the differential diagnosis of thrombocytopenia. For example, it is high in thrombocytopenia resulting from peripheral destruction, as in immune thrombocytopenic purpura. This is because as platelets are lost, thrombopoietin production increases and new, larger platelets are released from healthy megakaryocytes in an attempt to increase the total platelet mass.

may_interpretingthecbc_f2.jpg
%3Cp%3EFigure%202.%20Giant%20platelets%20(thin%20arrows)%2C%20normal%20sized%20platelets%20(dotted%20arrows)%2C%20and%20a%20nucleated%20red%20blood%20cell%20(thick%20arrow)%20in%20a%20patient%20with%20myelofibrosis%20and%20extensive%20extramedullary%20hematopoiesis.%3C%2Fp%3E

In contrast, the MPV is low in patients with thrombocytopenia due to megakaryocyte hypoplasia, as malfunctioning megakaryocytes cannot maintain the total platelet mass, and any platelets produced remain small. This distinction can be obscured in the setting of splenomegaly, as larger platelets are more easily sequestered in the spleen and the MPV may therefore be low or normal.

The MPV can also be used to differentiate congenital thrombocytopenic disorders, which can be characterized by either a high MPV (eg, gray platelet syndrome, Bernard-Soulier syndrome) or a low MPV (eg, Wiskott-Aldrich syndrome) (Figure 2).

MPV may have prognostic value

Evidence suggests that the MPV also has potential prognostic value, particularly in vascular disease, as larger platelets are hypothesized to have increased hemostatic potential.

In a large meta-analysis of patients with coronary artery disease, a high MPV was associated with worse outcomes; the risk of death or myocardial infarction was 17% higher in those with a high MPV (the threshold ranged from 8.4 to 11.7 fL in the different studies) than in those with a low MPV.6

In a study of 213 patients with non-ST-segment elevation myocardial infarction, the risk of significant coronary artery disease was 4.18 times higher in patients with a high MPV and a high troponin level than in patients with a normal MPV and a high troponin.7 The authors suggested that a high MPV may help identify patients at highest risk of significant coronary artery disease who would benefit from invasive studies (ie, coronary angiography).

This correlation has also been observed in other forms of vascular disease. In 261 patients who underwent carotid angioplasty and stenting, an MPV higher than 10.1 fL was associated with a risk of in-stent restenosis more than 3 times higher.8

The MPV has also been found to be higher in patients with type 2 diabetes than in controls, particularly in those with microvascular complications such as retinopathy or microalbuminuria.9

Conversely, in patients with cancer, a low MPV appears to be associated with a poor prognosis. In a retrospective analysis of 236 patients with esophageal cancer, those who had an MPV of 7.4 fL or less had significantly shorter overall survival than patients with an MPV higher than 7.4 fL.10

A low MPV has also been associated with an increased risk of venous thromoboembolism in patients with cancer. In a prospective observational cohort study of 1,544 patients, the 2-year probability of venous thromboembolism was 9% in patients with an MPV less than 10.8 fL, compared with 5.5% in those with higher MPV values. The 2-year overall survival rate was also higher in patients with high MPV than in those with low MPV, at 64.7% vs 55.7%, respectively (P = .001).11

But the MPV is far from a perfect clinical metric. Since its measurement is subject to significant laboratory variation, an abnormal value should always be confirmed with evaluation of a peripheral blood smear. Furthermore, it is unclear why a high MPV portends poor prognosis in patients without cancer, whereas the opposite is true in patients with cancer. Therefore, its role in prognostication remains investigational, and further studies are essential to determine its appropriate usefulness in clinical practice.12

NUCLEATED RED BLOOD CELL COUNT

NRBCs are immature red blood cell precursors not present in the circulation of healthy adults. During erythropoiesis, the common myeloid progenitor cell first differentiates into a proerythroblast; subsequently, the chromatin in the nucleus of the proerythroblast gradually condenses until it becomes an orthochromatic erythroblast, also known as a nucleated red cell (Figure 2). Once the nucleus is expelled, the cell is known as a reticulocyte, which ultimately becomes a mature erythrocyte.

Healthy newborns have circulating NRBCs that rapidly disappear within a few weeks of birth. However, NRBCs can return to the circulation in a variety of disease states.

Causes of NRBCs

Brisk hemolysis or rapid blood loss can cause NRBCs to be released into the blood as erythropoiesis increases in an attempt to compensate for acute anemia.

Damage or stress to the bone marrow also causes NRBCs to be released into the peripheral blood, as is often the case in hematologic diseases. In a study of 478 patients with hematologic diseases, the frequency of NRBC positivity at diagnosis was highest in patients with chronic myeloid leukemia (100%), acute leukemia (62%), and myelodysplastic syndromes (45%).13 NRBCs also appeared at higher frequencies during chemotherapy in other hematologic conditions, such as hemophagocytic lymphohistiocytosis.

The mechanism by which NRBCs are expelled from the bone marrow is unclear, though studies have suggested that inflammation or hypoxia or both cause increased hematopoietic stress, resulting in the release of immature red cells. Increased concentrations of inflammatory cytokines (interleukin 6 and interleukin 3) and erythropoietin in the plasma and decreased arterial oxygen partial tension have been reported in patients with circulating NRBCs.14,15

Because they are associated with hematologic disorders, the finding of NRBCs should prompt evaluation of a peripheral smear to assess for abnormalities in other cell lines.

The NRBC count and prognosis

In critically ill patients, peripheral NRBCs can also indicate life-threatening conditions.

In a study of 421 adult intensive care patients, the in-hospital mortality rate was 42% in those with peripheral NRBCs vs 5.9% in those without them.16 Further, the higher the NRBC count and the more days that NRBCs were reported in the CBC, the higher the risk of death.

In adults with acute respiratory distress syndrome, the finding of any NRBCs in the peripheral blood was an independent risk factor for death, and an NRBC count higher than 220 cells/µL was associated with a more than 3-fold higher risk of death.17

Daily screening in patients in surgical intensive care units revealed that NRBCs appeared an average of 9 days before death, consistent with an early marker of impending decline.18

In another study,19 the risk of death within 90 days of hospital discharge was higher in NRBC-positive patients, reaching 21.9% in those who had a count higher than 200 cells/µL. The risk of unplanned hospital readmission within 30 days was also increased.

Leukoerythroblastosis

The combination of NRBCs and immature white blood cells (eg, myelocytes, metamyelocytes) is called leukoerythroblastosis.

Leukoerythroblastosis is classically seen in myelophthisic anemias in which hematopoietic cells in the marrow are displaced by fibrosis, tumor, or other space-occupying processes, but it can also occur in any situation of acute marrow stress, including critical illness.

In addition, leukoerythroblastosis appears in a rare complication of sickle cell hemoglobinopathies: bone marrow necrosis with fat embolism syndrome.20,21 As the marrow necroses, fat emboli are released in the systemic circulation causing micro- and macrovascular occlusions and multiorgan failure. The largest case series in the literature reports 58 patients with bone marrow necrosis with fat embolism syndrome.22

At our institution, we have seen 18 patients with this condition in the past 8 years, with the frequency of diagnosis increasing with heightened awareness of the disorder. We have found that leukoerythroblastosis is often an early marker of this unrecognized syndrome and can prompt emergency red cell exchange, which is considered to be lifesaving in this condition.22

These examples and many others show that the presence of NRBCs in the CBC can serve as an important clinical warning.

OLD TESTS CAN STILL BE USEFUL

The CBC provides much more than simple cell counts; it is a rich collection of information related to each blood cell. These days, with new diagnostic tests and prognostic tools based on molecular analysis, it is important to not overlook the value of the tests clinicians have been ordering for generations.

The RDW, MPV, and NRBC count will not likely provide definitive or flawless diagnostic or prognostic information, but when understood and used correctly, they provide readily available, cost-effective, and useful data that can supplement and guide clinical decision-making. By understanding the CBC more fully, providers can maximize the truly complete nature of this routine laboratory test.

The complete blood cell count (CBC) is one of the most frequently ordered laboratory tests in both the inpatient and outpatient settings. Not long ago, the CBC required peering through a microscope and counting the red blood cells, white blood cells, and platelets. These 3 numbers are still the primary purpose of the test.

Now, with automated counters, the CBC report also contains other numbers that delineate characteristics of each cell type. For example:

The mean corpuscular volume is the average volume of red blood cells. Providers use it to classify anemia as either microcytic, normocytic, or macrocytic, each with its own differential diagnosis.

The differential white blood cell count provides absolute counts and relative percentages of each type of leukocyte. For example, the absolute neutrophil count is an important measure of immunocompetence.

But other values in the CBC may be overlooked, even though they can provide important information. Here, we highlight 3 of them:

  • The red blood cell distribution width (RDW)
  • The mean platelet volume (MPV)
  • The nucleated red blood cell (NRBC) count.

In addition to describing their diagnostic utility, we also discuss emerging evidence of their potential prognostic significance in hematologic and nonhematologic disorders. By incorporating an awareness of their value in clinical practice, providers can maximize the usefulness of the CBC.

RED BLOOD CELL DISTRIBUTION WIDTH

may_interpretingthecbc_f1.jpg
%3Cp%3EFigure%201.%20A%3A%20Example%20of%20a%20normal%20red%20blood%20cell%20distribution%20width%20(RDW)%20of%2013.5%25%20(red%20line)%20in%20a%20patient%20with%20a%20normal%20complete%20blood%20cell%20count.%20B%3A%20Example%20of%20an%20increased%20RDW%20of%2028.8%25%20in%20a%20patient%20with%20iron%20deficiency%20shortly%20after%20initiation%20of%20iron%20supplementation.%3C%2Fp%3E

The RDW is a measure of variation (anisocytosis) in the size of the circulating red cells. The term “width” is misleading, as the value is not derived from the width of the red blood cell, but rather from the width of the distribution curve of the corpuscular volume (Figure 1). Therefore, a normal RDW means that the cells are all about the same size, while a high RDW means they vary widely in size.

The RDW can be calculated either as a coefficient of variation, with a reference range of 11% to 16% depending on the laboratory, or, less often, as a standard deviation, with a reference range of 39 to 46 fL.

The RDW can differentiate between causes of anemia

A high RDW is often found in nutritional deficiencies of iron, vitamin B12, and folate. This information is helpful in differentiating the cause of microcytic anemia, as a high RDW suggests iron-deficiency anemia while a normal RDW suggests thalassemia.1 In iron deficiency, the RDW often rises before the mean corpuscular volume falls, serving as an early diagnostic clue.

The RDW can also be high after recent hemorrhage or rapid hemolysis, as the acute drop in hemoglobin results in increased production of reticulocytes, which are larger than mature erythrocytes.

Because a range of disorders can elevate the RDW, reviewing the peripheral blood smear is an important next step in the diagnostic evaluation, specifically looking for reticulocytes, microspherocytes, and other abnormal red blood cells contributing to the RDW elevation.

A normal RDW is less diagnostically useful. It indicates the red blood cells are of uniform size, but they may be uniformly small or large depending on how long the anemia has persisted. Since red cells circulate for only about 120 days, patients who have severe iron-deficiency anemia for months to years are expected to have a normal rather than a high RDW, as their red cells of normal size have all been replaced by microcytes.

A low RDW is not consistently associated with any hematologic disorder.

RDW may have prognostic value

Emerging data suggest that the RDW may also have prognostic value in nonhematologic diseases. In a retrospective study of 15,852 adult participants in the Third National Health and Nutrition Examination Survey (1988–1994), a higher RDW was associated with a higher risk of death, with the all-cause mortality rate increasing by 23% for every 1% increment in RDW.2

This correlation is particularly prominent in cardiac disorders. In 2 large retrospective studies of patients with symptomatic heart failure, a higher RDW was a strong predictor of morbidity and death (hazard ratio 1.17 per 1-standard deviation increase, P < .001), even stronger than more commonly used variables such as ejection fraction, New York Heart Association functional class, and renal function.3

In a retrospective analysis of 4,111 patients with myocardial infarction, the degree of RDW elevation correlated with the risk of repeat nonfatal myocardial infarction, coronary death, new symptomatic heart failure, and stroke.4

It is hypothesized that high RDW may reflect poor cell membrane integrity from altered cholesterol content, which in turn has deleterious effects on multiple organ systems and is therefore associated with adverse outcomes.5

Currently, using the RDW to assess prognosis remains investigational, and how best to interpret it in daily practice requires further study.

 

 

MEAN PLATELET VOLUME

The MPV, ie, the average size of platelets, is reported in femtoliters (fL). Because the MPV varies depending on the instrument used, each laboratory has a unique reference range, usually about 8 to 12 fL. The MPV must be interpreted in conjunction with the platelet count; the product of the MPV and platelet count is called the total platelet mass.

Using the MPV to find the cause of thrombocytopenia

The MPV can be used to help narrow the differential diagnosis of thrombocytopenia. For example, it is high in thrombocytopenia resulting from peripheral destruction, as in immune thrombocytopenic purpura. This is because as platelets are lost, thrombopoietin production increases and new, larger platelets are released from healthy megakaryocytes in an attempt to increase the total platelet mass.

may_interpretingthecbc_f2.jpg
%3Cp%3EFigure%202.%20Giant%20platelets%20(thin%20arrows)%2C%20normal%20sized%20platelets%20(dotted%20arrows)%2C%20and%20a%20nucleated%20red%20blood%20cell%20(thick%20arrow)%20in%20a%20patient%20with%20myelofibrosis%20and%20extensive%20extramedullary%20hematopoiesis.%3C%2Fp%3E

In contrast, the MPV is low in patients with thrombocytopenia due to megakaryocyte hypoplasia, as malfunctioning megakaryocytes cannot maintain the total platelet mass, and any platelets produced remain small. This distinction can be obscured in the setting of splenomegaly, as larger platelets are more easily sequestered in the spleen and the MPV may therefore be low or normal.

The MPV can also be used to differentiate congenital thrombocytopenic disorders, which can be characterized by either a high MPV (eg, gray platelet syndrome, Bernard-Soulier syndrome) or a low MPV (eg, Wiskott-Aldrich syndrome) (Figure 2).

MPV may have prognostic value

Evidence suggests that the MPV also has potential prognostic value, particularly in vascular disease, as larger platelets are hypothesized to have increased hemostatic potential.

In a large meta-analysis of patients with coronary artery disease, a high MPV was associated with worse outcomes; the risk of death or myocardial infarction was 17% higher in those with a high MPV (the threshold ranged from 8.4 to 11.7 fL in the different studies) than in those with a low MPV.6

In a study of 213 patients with non-ST-segment elevation myocardial infarction, the risk of significant coronary artery disease was 4.18 times higher in patients with a high MPV and a high troponin level than in patients with a normal MPV and a high troponin.7 The authors suggested that a high MPV may help identify patients at highest risk of significant coronary artery disease who would benefit from invasive studies (ie, coronary angiography).

This correlation has also been observed in other forms of vascular disease. In 261 patients who underwent carotid angioplasty and stenting, an MPV higher than 10.1 fL was associated with a risk of in-stent restenosis more than 3 times higher.8

The MPV has also been found to be higher in patients with type 2 diabetes than in controls, particularly in those with microvascular complications such as retinopathy or microalbuminuria.9

Conversely, in patients with cancer, a low MPV appears to be associated with a poor prognosis. In a retrospective analysis of 236 patients with esophageal cancer, those who had an MPV of 7.4 fL or less had significantly shorter overall survival than patients with an MPV higher than 7.4 fL.10

A low MPV has also been associated with an increased risk of venous thromoboembolism in patients with cancer. In a prospective observational cohort study of 1,544 patients, the 2-year probability of venous thromboembolism was 9% in patients with an MPV less than 10.8 fL, compared with 5.5% in those with higher MPV values. The 2-year overall survival rate was also higher in patients with high MPV than in those with low MPV, at 64.7% vs 55.7%, respectively (P = .001).11

But the MPV is far from a perfect clinical metric. Since its measurement is subject to significant laboratory variation, an abnormal value should always be confirmed with evaluation of a peripheral blood smear. Furthermore, it is unclear why a high MPV portends poor prognosis in patients without cancer, whereas the opposite is true in patients with cancer. Therefore, its role in prognostication remains investigational, and further studies are essential to determine its appropriate usefulness in clinical practice.12

NUCLEATED RED BLOOD CELL COUNT

NRBCs are immature red blood cell precursors not present in the circulation of healthy adults. During erythropoiesis, the common myeloid progenitor cell first differentiates into a proerythroblast; subsequently, the chromatin in the nucleus of the proerythroblast gradually condenses until it becomes an orthochromatic erythroblast, also known as a nucleated red cell (Figure 2). Once the nucleus is expelled, the cell is known as a reticulocyte, which ultimately becomes a mature erythrocyte.

Healthy newborns have circulating NRBCs that rapidly disappear within a few weeks of birth. However, NRBCs can return to the circulation in a variety of disease states.

Causes of NRBCs

Brisk hemolysis or rapid blood loss can cause NRBCs to be released into the blood as erythropoiesis increases in an attempt to compensate for acute anemia.

Damage or stress to the bone marrow also causes NRBCs to be released into the peripheral blood, as is often the case in hematologic diseases. In a study of 478 patients with hematologic diseases, the frequency of NRBC positivity at diagnosis was highest in patients with chronic myeloid leukemia (100%), acute leukemia (62%), and myelodysplastic syndromes (45%).13 NRBCs also appeared at higher frequencies during chemotherapy in other hematologic conditions, such as hemophagocytic lymphohistiocytosis.

The mechanism by which NRBCs are expelled from the bone marrow is unclear, though studies have suggested that inflammation or hypoxia or both cause increased hematopoietic stress, resulting in the release of immature red cells. Increased concentrations of inflammatory cytokines (interleukin 6 and interleukin 3) and erythropoietin in the plasma and decreased arterial oxygen partial tension have been reported in patients with circulating NRBCs.14,15

Because they are associated with hematologic disorders, the finding of NRBCs should prompt evaluation of a peripheral smear to assess for abnormalities in other cell lines.

The NRBC count and prognosis

In critically ill patients, peripheral NRBCs can also indicate life-threatening conditions.

In a study of 421 adult intensive care patients, the in-hospital mortality rate was 42% in those with peripheral NRBCs vs 5.9% in those without them.16 Further, the higher the NRBC count and the more days that NRBCs were reported in the CBC, the higher the risk of death.

In adults with acute respiratory distress syndrome, the finding of any NRBCs in the peripheral blood was an independent risk factor for death, and an NRBC count higher than 220 cells/µL was associated with a more than 3-fold higher risk of death.17

Daily screening in patients in surgical intensive care units revealed that NRBCs appeared an average of 9 days before death, consistent with an early marker of impending decline.18

In another study,19 the risk of death within 90 days of hospital discharge was higher in NRBC-positive patients, reaching 21.9% in those who had a count higher than 200 cells/µL. The risk of unplanned hospital readmission within 30 days was also increased.

Leukoerythroblastosis

The combination of NRBCs and immature white blood cells (eg, myelocytes, metamyelocytes) is called leukoerythroblastosis.

Leukoerythroblastosis is classically seen in myelophthisic anemias in which hematopoietic cells in the marrow are displaced by fibrosis, tumor, or other space-occupying processes, but it can also occur in any situation of acute marrow stress, including critical illness.

In addition, leukoerythroblastosis appears in a rare complication of sickle cell hemoglobinopathies: bone marrow necrosis with fat embolism syndrome.20,21 As the marrow necroses, fat emboli are released in the systemic circulation causing micro- and macrovascular occlusions and multiorgan failure. The largest case series in the literature reports 58 patients with bone marrow necrosis with fat embolism syndrome.22

At our institution, we have seen 18 patients with this condition in the past 8 years, with the frequency of diagnosis increasing with heightened awareness of the disorder. We have found that leukoerythroblastosis is often an early marker of this unrecognized syndrome and can prompt emergency red cell exchange, which is considered to be lifesaving in this condition.22

These examples and many others show that the presence of NRBCs in the CBC can serve as an important clinical warning.

OLD TESTS CAN STILL BE USEFUL

The CBC provides much more than simple cell counts; it is a rich collection of information related to each blood cell. These days, with new diagnostic tests and prognostic tools based on molecular analysis, it is important to not overlook the value of the tests clinicians have been ordering for generations.

The RDW, MPV, and NRBC count will not likely provide definitive or flawless diagnostic or prognostic information, but when understood and used correctly, they provide readily available, cost-effective, and useful data that can supplement and guide clinical decision-making. By understanding the CBC more fully, providers can maximize the truly complete nature of this routine laboratory test.

References
  1. Lima CS, Reis AR, Grotto HZ, Saad ST, Costa FF. Comparison of red cell distribution width and a red cell discriminant function incorporating volume dispersion for distinguishing iron deficiency from beta thalassemia trait in patients with microcytosis. Sao Paulo Med J 1996; 114(5):1265–1269. pmid:9239926
  2. Perlstein TS, Weuve J, Pfeffer MA, Beckman JA. Red blood cell distribution width and mortality risk in a community-based prospective cohort. Arch Intern Med 2009; 169(6):588–594. doi:10.1001/archinternmed.2009.55
  3. Felker GM, Allen LA, Pocock SJ, et al; CHARM Investigators. Red cell distribution width as a novel prognostic marker in heart failure: data from the CHARM Program and the Duke Databank. J Am Coll Cardiol 2007; 50(1):40–47. doi:10.1016/j.jacc.2007.02.067
  4. Tonelli M, Sacks F, Arnold M, Moye L, Davis B, Pfeffer M; for the Cholesterol and Recurrent Events (CARE) Trial Investigators. Relation between red blood cell distribution width and cardiovascular event rate in people with coronary disease. Circulation 2008; 117(2):163–168. doi:10.1161/CIRCULATIONAHA.107.727545
  5. Goldstein MR, Mascitelli L, Pezzetta F. Is red cell distribution width a marker of overall membrane integrity? [Letter] Arch Intern Med 2009; 169(16):1539–1540. doi:10.1001/archinternmed.2009.275
  6. Sansanaydhu N, Numthavaj P, Muntham D, et al. Prognostic effect of mean platelet volume in patients with coronary artery disease. A systematic review and meta-analysis. Thromb Haemost 2015; 114(6):1299–1309. doi:10.1160/TH15-04-0280
  7. Taskesen T, Sekhon H, Wroblewski I, et al. Usefulness of mean platelet volume to predict significant coronary artery disease in patients with non-ST-elevation acute coronary syndromes. Am J Cardiol 2017; 119(2):192–196. doi:10.1016/j.amjcard.2016.09.042
  8. Dai Z, Gao J, Li S, et al. Mean platelet volume as a predictor for restenosis after carotid angioplasty and stenting. Stroke 2018; 49(4):872–876. doi:10.1161/STROKEAHA.117.019748
  9. Papanas N, Symeonidis G, Maltezos E, et al. Mean platelet volume in patients with type 2 diabetes mellitus. Platelets 2004; 15(8):475–478. doi:10.1080/0953710042000267707
  10. Shen W, Cui MM, Wang X, Wang RT. Reduced mean platelet volume is associated with poor prognosis in esophageal cancer. Cancer Biomark 2018; 22(3):559–563. doi:10.3233/CBM-181231
  11. Riedl J, Kaider A, Reitter EM, et al. Association of mean platelet volume with risk of venous thromboembolism and mortality in patients with cancer. Results from the Vienna Cancer and Thrombosis Study (CATS). Thromb Haemost 2014; 111(4):670–678. doi:10.1160/TH13-07-0603
  12. Tsiara S, Elisaf M, Jagroop IA, Mikhailidis DP. Platelets as predictors of vascular risk: is there a practical index of platelet activity? Clin Appl Thromb Hemost 2003; 9(3):177–190. pmid:14507105
  13. Danise P, Maconi M, Barrella F, et al. Evaluation of nucleated red blood cells in the peripheral blood of hematological diseases. Clin Chem Lab Med 2011; 50(2):357–360. doi:10.1515/CCLM.2011.766
  14. Stachon A, Bolulul O, Holland-Letz T, Krieg M. Association between nucleated red blood cells in blood and the levels of erythropoietin, interleukin 3, interleukin 6, and interleukin 12p70. Shock 2005; 24(1):34–39. pmid:15988318
  15. Kuert S, Holland-Letz T, Friese J, Stachon A. Association of nucleated red blood cells in blood and arterial oxygen partial tension. Clin Chem Lab Med 2011; 49(2):257–263. doi:10.1515/CCLM.2011.041
  16. Stachon A, Holland-Letz T, Krieg M. In-hospital mortality of intensive care patients with nucleated red blood cells in blood. Clin Chem Lab Med 2004; 42(8):933–938. doi:10.1515/CCLM.2004.151
  17. Menk M, Giebelhäuser L, Vorderwülbecke G, et al. Nucleated red blood cells as predictors of mortality in patients with acute respiratory distress syndrome (ARDS): an observational study. Ann Intensive Care 2018; 8(1):42. doi:10.1186/s13613-018-0387-5
  18. Stachon A, Kempf R, Holland-Letz T, Friese J, Becker A, Krieg M. Daily monitoring of nucleated red blood cells in the blood of surgical intensive care patients. Clin Chim Acta 2006; 366(1–2):329–335. doi:10.1016/j.cca.2005.11.022
  19. Purtle SW, Horkan CM, Moromizato T, Gibbons FK, Christopher KB. Nucleated red blood cells, critical illness survivors and postdischarge outcomes: a cohort study. Crit Care 2017; 21(1):154. doi:10.1186/s13054-017-1724-z
  20. May J, Sullivan JC, LaVie D, LaVie K, Marques MB. Inside out: bone marrow necrosis and fat embolism complicating sickle-beta+ thalassemia. Am J Med 2016; 129(12):e321–e324. doi:10.1016/j.amjmed.2016.05.027
  21. Gangaraju R, Reddy VV, Marques MB. Fat embolism syndrome secondary to bone marrow necrosis in patients with hemoglobinopathies. South Med J 2016; 109(9):549–553. doi:10.14423/SMJ.0000000000000520
  22. Tsitsikas DA, Gallinella G, Patel S, Seligman H, Greaves P, Amos RJ. Bone marrow necrosis and fat embolism syndrome in sickle cell disease: increased susceptibility of patients with non-SS genotypes and a possible association with human parvovirus B19 infection. Blood Rev 2014; 28(1):23–30. doi:10.1016/j.blre.2013.12.002
References
  1. Lima CS, Reis AR, Grotto HZ, Saad ST, Costa FF. Comparison of red cell distribution width and a red cell discriminant function incorporating volume dispersion for distinguishing iron deficiency from beta thalassemia trait in patients with microcytosis. Sao Paulo Med J 1996; 114(5):1265–1269. pmid:9239926
  2. Perlstein TS, Weuve J, Pfeffer MA, Beckman JA. Red blood cell distribution width and mortality risk in a community-based prospective cohort. Arch Intern Med 2009; 169(6):588–594. doi:10.1001/archinternmed.2009.55
  3. Felker GM, Allen LA, Pocock SJ, et al; CHARM Investigators. Red cell distribution width as a novel prognostic marker in heart failure: data from the CHARM Program and the Duke Databank. J Am Coll Cardiol 2007; 50(1):40–47. doi:10.1016/j.jacc.2007.02.067
  4. Tonelli M, Sacks F, Arnold M, Moye L, Davis B, Pfeffer M; for the Cholesterol and Recurrent Events (CARE) Trial Investigators. Relation between red blood cell distribution width and cardiovascular event rate in people with coronary disease. Circulation 2008; 117(2):163–168. doi:10.1161/CIRCULATIONAHA.107.727545
  5. Goldstein MR, Mascitelli L, Pezzetta F. Is red cell distribution width a marker of overall membrane integrity? [Letter] Arch Intern Med 2009; 169(16):1539–1540. doi:10.1001/archinternmed.2009.275
  6. Sansanaydhu N, Numthavaj P, Muntham D, et al. Prognostic effect of mean platelet volume in patients with coronary artery disease. A systematic review and meta-analysis. Thromb Haemost 2015; 114(6):1299–1309. doi:10.1160/TH15-04-0280
  7. Taskesen T, Sekhon H, Wroblewski I, et al. Usefulness of mean platelet volume to predict significant coronary artery disease in patients with non-ST-elevation acute coronary syndromes. Am J Cardiol 2017; 119(2):192–196. doi:10.1016/j.amjcard.2016.09.042
  8. Dai Z, Gao J, Li S, et al. Mean platelet volume as a predictor for restenosis after carotid angioplasty and stenting. Stroke 2018; 49(4):872–876. doi:10.1161/STROKEAHA.117.019748
  9. Papanas N, Symeonidis G, Maltezos E, et al. Mean platelet volume in patients with type 2 diabetes mellitus. Platelets 2004; 15(8):475–478. doi:10.1080/0953710042000267707
  10. Shen W, Cui MM, Wang X, Wang RT. Reduced mean platelet volume is associated with poor prognosis in esophageal cancer. Cancer Biomark 2018; 22(3):559–563. doi:10.3233/CBM-181231
  11. Riedl J, Kaider A, Reitter EM, et al. Association of mean platelet volume with risk of venous thromboembolism and mortality in patients with cancer. Results from the Vienna Cancer and Thrombosis Study (CATS). Thromb Haemost 2014; 111(4):670–678. doi:10.1160/TH13-07-0603
  12. Tsiara S, Elisaf M, Jagroop IA, Mikhailidis DP. Platelets as predictors of vascular risk: is there a practical index of platelet activity? Clin Appl Thromb Hemost 2003; 9(3):177–190. pmid:14507105
  13. Danise P, Maconi M, Barrella F, et al. Evaluation of nucleated red blood cells in the peripheral blood of hematological diseases. Clin Chem Lab Med 2011; 50(2):357–360. doi:10.1515/CCLM.2011.766
  14. Stachon A, Bolulul O, Holland-Letz T, Krieg M. Association between nucleated red blood cells in blood and the levels of erythropoietin, interleukin 3, interleukin 6, and interleukin 12p70. Shock 2005; 24(1):34–39. pmid:15988318
  15. Kuert S, Holland-Letz T, Friese J, Stachon A. Association of nucleated red blood cells in blood and arterial oxygen partial tension. Clin Chem Lab Med 2011; 49(2):257–263. doi:10.1515/CCLM.2011.041
  16. Stachon A, Holland-Letz T, Krieg M. In-hospital mortality of intensive care patients with nucleated red blood cells in blood. Clin Chem Lab Med 2004; 42(8):933–938. doi:10.1515/CCLM.2004.151
  17. Menk M, Giebelhäuser L, Vorderwülbecke G, et al. Nucleated red blood cells as predictors of mortality in patients with acute respiratory distress syndrome (ARDS): an observational study. Ann Intensive Care 2018; 8(1):42. doi:10.1186/s13613-018-0387-5
  18. Stachon A, Kempf R, Holland-Letz T, Friese J, Becker A, Krieg M. Daily monitoring of nucleated red blood cells in the blood of surgical intensive care patients. Clin Chim Acta 2006; 366(1–2):329–335. doi:10.1016/j.cca.2005.11.022
  19. Purtle SW, Horkan CM, Moromizato T, Gibbons FK, Christopher KB. Nucleated red blood cells, critical illness survivors and postdischarge outcomes: a cohort study. Crit Care 2017; 21(1):154. doi:10.1186/s13054-017-1724-z
  20. May J, Sullivan JC, LaVie D, LaVie K, Marques MB. Inside out: bone marrow necrosis and fat embolism complicating sickle-beta+ thalassemia. Am J Med 2016; 129(12):e321–e324. doi:10.1016/j.amjmed.2016.05.027
  21. Gangaraju R, Reddy VV, Marques MB. Fat embolism syndrome secondary to bone marrow necrosis in patients with hemoglobinopathies. South Med J 2016; 109(9):549–553. doi:10.14423/SMJ.0000000000000520
  22. Tsitsikas DA, Gallinella G, Patel S, Seligman H, Greaves P, Amos RJ. Bone marrow necrosis and fat embolism syndrome in sickle cell disease: increased susceptibility of patients with non-SS genotypes and a possible association with human parvovirus B19 infection. Blood Rev 2014; 28(1):23–30. doi:10.1016/j.blre.2013.12.002
Issue
Cleveland Clinic Journal of Medicine - 86(3)
Issue
Cleveland Clinic Journal of Medicine - 86(3)
Page Number
167-172
Page Number
167-172
Publications
Publications
Topics
Article Type
Display Headline
Three neglected numbers in the CBC: The RDW, MPV, and NRBC count
Display Headline
Three neglected numbers in the CBC: The RDW, MPV, and NRBC count
Legacy Keywords
complete blood cell count, CBC, red cell distribution width, RDW, mean platelet volume, MPV, nucleated red blood cell count, NRBC, anemia, thrombocytopenia, iron deficiency, thalassemia, blood test, prognosis, leukoerythroblastosis, Jori May, Marisa Marques, Vishnu Reddy, Radhika Gangaraju
Legacy Keywords
complete blood cell count, CBC, red cell distribution width, RDW, mean platelet volume, MPV, nucleated red blood cell count, NRBC, anemia, thrombocytopenia, iron deficiency, thalassemia, blood test, prognosis, leukoerythroblastosis, Jori May, Marisa Marques, Vishnu Reddy, Radhika Gangaraju
Sections
Inside the Article

KEY POINTS

  • The RDW can help differentiate the cause of anemia: eg, a high RDW suggests iron-deficiency anemia, while a normal RDW suggests thalassemia. Studies also suggest that a high RDW may be associated with an increased rate of all-cause mortality and may predict a poor prognosis in several cardiac diseases.
  • The MPV can be used in the evaluation of thrombocytopenia. Furthermore, emerging evidence suggests that high MPV is associated with worse outcomes in cardiovascular disorders.
  • An elevated NRBC count may predict poor outcomes in a number of critical care settings. It can also indicate a serious underlying hematologic disorder.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Wed, 02/27/2019 - 14:45
Un-Gate On Date
Wed, 02/27/2019 - 14:45
Use ProPublica
CFC Schedule Remove Status
Wed, 02/27/2019 - 14:45
Hide sidebar & use full width
render the right sidebar.
Article PDF Media

Assessing liver fibrosis without biopsy in patients with HCV or NAFLD

Article Type
Changed
Fri, 03/01/2019 - 15:25
Display Headline
Assessing liver fibrosis without biopsy in patients with HCV or NAFLD

Staging of liver fibrosis, important for determining prognosis in patients with chronic liver disease and for the need to start screening for complications of cirrhosis, was traditionally done only by liver biopsy. While biopsy is still the gold standard method to stage fibrosis, noninvasive methods have been developed that can also assess disease severity.

This article briefly reviews the epidemiology and physiology of chronic liver disease and the traditional role of liver biopsy. Pros and cons of alternative fibrosis assessment methods are discussed, with a focus on their utility for patients with nonalcoholic fatty liver disease (NAFLD) and hepatitis C virus (HCV) infection.

CHRONIC LIVER DISEASE: A HUGE HEALTH BURDEN

Chronic liver disease is associated with enormous health and financial costs in the United States. Its prevalence is about 15%,1 and it is the 12th leading cause of death.2 Hospital costs are estimated at about $4 billion annually.3

The most common causes of chronic liver disease are NAFLD (which may be present in up to one-third of the US population and is increasing with the epidemic of obesity), its aggressive variant, nonalcoholic steatohepatitis (NASH) (present in about 3% of the population), and HCV infection (1%).4,5

Since direct-acting antiviral agents were introduced, HCV infection dropped from being the leading cause of liver transplant to third place.6 But at the same time, the number of patients on the transplant waiting list who have NASH has risen faster than for any other cause of chronic liver disease.7

FIBROSIS: A KEY INDICATOR OF DISEASE SEVERITY

singh_assessingliverfibrosis_t1.jpg
With any form of liver disease, collagen is deposited in hepatic lobules over time, a process called fibrosis. Both HCV infection and NASH involve necroinflammation in the liver, hepatocyte apoptosis, and activation of stellate cells, leading to progressive collagen deposition in hepatic lobules. Fibrosis typically starts in the region of the central vein and portal tracts and eventually extends to other areas of the lobule.

singh_assessingliverfibrosis_f1.jpg
%3Cp%3EFigure%201.%20Findings%20on%20liver%20biopsy%20in%20nonalcoholic%20fatty%20liver%20disease%20and%20hepatitis%20C%20virus%20infection.%3C%2Fp%3E
Determining fibrosis severity is critical when a patient is diagnosed with chronic liver disease, as it predicts long-term clinical outcomes and death in HCV8 and NAFLD.9 Different staging systems have been developed to reflect the degree of fibrosis, based on its distribution as seen on liver biopsy (Table 1, Figure 1).

In HCV infection, advanced fibrosis is defined as either stage 4 to 6 using the Ishak system10 or stage 3 to 4 using the Meta-analysis of Histological Data in Viral Hepatitis (METAVIR) system.11

In NAFLD, advanced fibrosis is defined as stage 3 to 4 using the NASH Clinical Research Network system.12

Staging fibrosis is also important so that patients with cirrhosis can be identified early to begin screening for hepatocellular carcinoma and esophageal varices to reduce the risks of illness and death. In addition, insurance companies often require documentation of fibrosis stage before treating HCV with the new direct-acting antiviral agents.

LIVER BIOPSY IS STILL THE GOLD STANDARD

Although invasive, liver biopsy remains the gold standard for determining fibrosis stage. Liver biopsies were performed “blindly” (without imaging) until the 1990s, but imaging-guided biopsy using ultrasonography was then developed, which entailed less pain and lower complication and hospitalization rates. Slightly more hepatic tissue is obtained with guided liver biopsy, but the difference was deemed clinically insignificant.13 Concern initially arose about the added cost involved with imaging, but imaging-guided biopsy was actually found to be more cost-effective.14

In the 2000s, transjugular liver biopsy via the right internal jugular vein became available. This method was originally used primarily in patients with ascites or significant coagulopathy. At first, there were concerns about the adequacy of specimens obtained to make an accurate diagnosis or establish fibrosis stage, but this limitation was overcome with improved techniques.15,16 Transjugular liver biopsy has the additional advantage of enabling one to measure the hepatic venous pressure gradient, which also has prognostic significance; a gradient greater than 10 mm Hg is associated with worse prognosis.17

Disadvantages of biopsy: Complications, sampling errors

Liver biopsy has disadvantages. Reported rates of complications necessitating hospitalization using the blind method were as high as 6% in the 1970s,18 dropping to 3.2% in a 1993 study.19 Bleeding remains the most worrisome complication. With the transjugular method, major and minor complication rates are less than 1% and 7%, respectively.15,16 Complication rates with imaging-guided biopsy are also low.

Liver biopsy is also prone to sampling error. The number of portal tracts obtained in the biopsy correlates with the accuracy of fibrosis staging, and smaller samples may lead to underestimating fibrosis stage. In patients with HCV, samples more than 15 mm long led to accurate staging diagnosis in 65% of patients, and those longer than 25 mm conferred 75% accuracy.20 Also, different stages can be diagnosed from samples obtained from separate locations in the liver, although rarely is the difference more than a single stage.21

Histologic evaluation of liver biopsies is operator-dependent. Although significant interobserver variation has been reported for degree of inflammation, there tends to be good concordance for fibrosis staging.22,23

 

 

STAGING BASED ON DEMOGRAPHIC AND LABORATORY VARIABLES

Several scores based on patient characteristics and laboratory values have been developed for assessing liver fibrosis and have been specifically validated for HCV infection, NAFLD, or both. They can serve as inexpensive initial screening tests for the presence or absence of advanced fibrosis.

FIB-4 index for HCV, NAFLD

The FIB-4 index predicts the presence of advanced fibrosis using, as its name indicates, a combination of 4 factors in fibrosis: age, platelet count, and the levels of aspartate aminotransferase (AST) and alanine aminotransferase (ALT), according to the formula:

FIB-4 index = (age × AST [U/L]) /
(platelet count [× 109/L] × √ALT [U/L]).

The index was derived from data from 832 patients co-infected with HCV and human immunodeficiency virus.24 The Ishak staging system10 for fibrosis on liver biopsy was used for confirmation, with stage 4 to 6 defined as advanced fibrosis. A cutoff value of more than 3.25 had a positive predictive value of 65% for advanced fibrosis, and to exclude advanced fibrosis, a cutoff value of less than 1.45 had a negative predictive value of 90%.

The FIB-4 index has since been validated in patients with HCV infection25 and NAFLD.26 In a subsequent study in 142 patients with NAFLD, the FIB-4 index was more accurate in diagnosing advanced fibrosis than the other noninvasive prediction models discussed below.27

NAFLD fibrosis score

The NAFLD fibrosis score, constructed and validated only in patients with biopsy-confirmed NAFLD, incorporates age, body mass index, presence of diabetes or prediabetes, albumin level, platelet count, and AST and ALT levels.

A group of 480 patients was used to construct the score, and 253 patients were used to validate it. Using the high cutoff value of 0.676, the presence of advanced fibrosis was diagnosed with a positive predictive value of 90% in the group used to construct the model (82% in the validation group). Using the low cutoff score of –1.455, advanced fibrosis could be excluded with a negative predictive value of 93% in the construction group and 88% in the validation group.28 A score between the cutoff values merits liver biopsy to determine fibrosis stage. The score is more accurate in patients with diabetes.29 When used by primary care physicians, the NAFLD fibrosis score is more cost-effective than transient elastography and liver biopsy for accurately predicting advanced fibrosis.30

AST-to-platelet ratio index score for HCV, NAFLD

The AST-to-platelet ratio index (APRI) score was developed in 2003 using a cohort of 270 patients with HCV and liver biopsy as the standard. A cutoff value of less than or equal to 0.5 had a negative predictive value of 86% for the absence of significant fibrosis, while a score of more than 1.5 detected the presence of significant fibrosis with a positive predictive value of 88%.31 The APRI score was subsequently validated for NAFLD.27,32

FibroSure uses a patented formula

FibroSure (LabCorp; labcorp.com) uses a patented mathematical formula that takes into account age, sex, and levels of gamma-glutamyl transferase, total bilirubin, haptoglobin, apolipoprotein-A, and alpha-2 macroglobulin to assess fibrosis. Developed in 2001 for use in patients with HCV infection, it was reported to have a positive predictive value of greater than 90% and a negative predictive value of 100% for clinically significant fibrosis, defined as stage 2 to 4 based on the METAVIR staging system in the prediction model.33 The use of FibroSure in patients with HCV was subsequently validated in various meta-analyses and systematic reviews.34,35 It is less accurate in patients with normal ALT levels.36

FibroSure also has good accuracy for predicting fibrosis stage in chronic liver disease due to other causes, including NAFLD.37

The prediction models discussed above use routine laboratory tests for chronic liver disease and thus are inexpensive. The high cost of additional testing needed for FibroSure, coupled with the risk of misdiagnosis, makes its cost-effectiveness questionable.38

 

 

IMAGING TO PREDICT FIBROSIS STAGE

Conventional ultrasonography (with or without vascular imaging) and computed tomography can detect cirrhosis on the basis of certain imaging characteristics,39,40 including the nodular contour of the liver, caudate lobe hypertrophy, ascites, reversal of blood flow in the portal vein, and splenomegaly. However, they cannot detect fibrosis in its early stages.

The 3 methods discussed below provide more accurate fibrosis staging by measuring the velocity of shear waves sent across hepatic tissue. Because shear-wave velocity increases with liver stiffness, the fibrosis stage can be estimated from this information.41

Transient elastography

Transient elastography uses a special ultrasound transducer. It is highly accurate for predicting advanced fibrosis for almost all causes of chronic liver disease, including HCV infection42,43 and NAFLD.44 The cutoff values of wave velocity to estimate fibrosis stage differ by liver disease etiology.

Transient elastography should not be used to evaluate fibrosis in patients with acute hepatitis, which transiently increases liver stiffness, resulting in a falsely high fibrosis stage diagnosis.45 It is also not a good method for evaluating fibrosis in patients with biliary obstruction or extrahepatic venous congestion. Because liver stiffness can increase after eating,46 the test should be done under fasting conditions.

A significant limitation of transient elastography has been its poor accuracy in patients with obesity.47 This has been largely overcome with the use of a more powerful (XL) probe but is still a limitation for those with morbid obesity.48 Because many patients with NAFLD are obese, this limitation can be significant.

Transient elastography has gained popularity for evaluating fibrosis in patients with chronic liver disease for multiple reasons: it is cost-effective and results are highly reproducible, with low variation in results among different observers and in individual observers.49 Combined with a platelet count, it can also be used to detect the development of clinically significant portal hypertension in patients with cirrhosis, thus determining the need to screen for esophageal varices using endoscopy.50 Screening endoscopy can be avoided in patients whose liver stiffness remains below 20 kPa or whose platelet count is above 150 × 109/L.

Acoustic radiation force imaging

Unlike transient elastography, which requires a separate transducer probe to assess shear- wave velocity, acoustic radiation force imaging uses the same transducer for both this function and imaging. Different image modes are available when testing for liver stiffness, so a region of interest that is optimal for avoiding vascular structures or masses can be selected, increasing accuracy.51

Acoustic radiation force imaging has been tested in different causes of chronic liver disease, including HCV and NAFLD,52 with accuracy similar to that of transient elastography.53 For overweight and obese patients, acoustic radiation force imaging is more accurate than transient elastography using the XL probe.54 However, this method is still new, and we need more data to support using one method over the other.

Magnetic resonance elastography

Magnetic resonance elastography uses a special transducer placed under the rib cage to transmit shear waves concurrently with magnetic resonance imaging. It has been tested in patients with HCV and NAFLD and has been found to have better diagnostic accuracy than transient elastography and acoustic radiation force imaging.55,56 Patients must be fasting for better diagnostic accuracy57 and must hold their breath while elastography is performed. The need for breath-holding and the high cost limit the use of this method for assessing fibrosis.

BOTTOM LINE FOR ASSESSING FIBROSIS

singh_assessingliverfibrosis_f2.jpg
%3Cp%3EFigure%202.%20Algorithm%20to%20determine%20fibrosis%20stage%20for%20nonalcoholic%20fatty%20liver%20disease.%3C%2Fp%3E
Although liver biopsy remains the gold standard for accurately determining fibrosis stage, noninvasive methods, especially imaging techniques, are fast evolving. Guidelines recommend using transient elastography to determine fibrosis stage noninvasively in patients with HCV,58 but a similar recommendation cannot be made for NAFLD with available data. For NAFLD, combined elastography and NAFLD fibrosis score are recommended to determine the need for a liver biopsy (Figure 2).59 Currently, we recommend using a combination of the scores discussed above and the imaging tests.

References
  1. Younossi ZM, Stepanova M, Afendy M, et al. Changes in the prevalence of the most common causes of chronic liver diseases in the United States from 1988 to 2008. Clin Gastroenterol Hepatol 2011; 9(6):524–530.e1. doi:10.1016/j.cgh.2011.03.020
  2. Kochanek KD, Xu J, Murphy SL, Miniño AM, Kung H-C. Deaths: final data for 2009. Natl Vital Stat Rep 2011; 60(3):1–116. pmid:24974587
  3. Volk ML, Tocco RS, Bazick J, Rakoski MO, Lok AS. Hospital readmissions among patients with decompensated cirrhosis. Am J Gastroenterol 2012; 107(2):247–252. doi:10.1038/ajg.2011.314
  4. Vernon G, Baranova A, Younossi ZM. Systematic review: the epidemiology and natural history of non-alcoholic fatty liver disease and non-alcoholic steatohepatitis in adults. Aliment Pharmacol Ther 2011; 34(3):274–285. doi:10.1111/j.1365-2036.2011.04724.x
  5. Udompap P, Kim D, Kim WR. Current and future burden of chronic nonmalignant liver disease. Clin Gastroenterol Hepatol 2015; 13(12):2031–2041. doi:10.1016/j.cgh.2015.08.015
  6. Kim WR, Lake JR, Smith JM, et al. OPTN/SRTR 2016 annual data report: liver. Am J Transplant 2018; 18(suppl 1):172–253. doi:10.1111/ajt.14559
  7. Wong RJ, Aguilar M, Cheung R, et al. Nonalcoholic steatohepatitis is the second leading etiology of liver disease among adults awaiting liver transplantation in the United States. Gastroenterology 2015; 148(3):547–555. doi:10.1053/j.gastro.2014.11.039
  8. Ishak K, Baptista A, Bianchi L, et al. Histological grading and staging of chronic hepatitis. J Hepatol 1995; 22(6):696–699. pmid:7560864
  9. Bedossa P, Poynard T. An algorithm for the grading of activity in chronic hepatitis C. Hepatology 1996; 24(2):289–293. doi:10.1002/hep.510240201
  10. Kleiner DE, Brunt EM, Van Natta M, et al; Nonalcoholic Steatohepatitis Clinical Research Network. Design and validation of a histological scoring system for nonalcoholic fatty liver disease. Hepatology 2005; 41(6):1313–1321. doi:10.1002/hep.20701
  11. Everhart JE, Wright EC, Goodman ZD, et al; HALT-C Trial Group. Prognostic value of Ishak fibrosis stage: findings from the hepatitis C antiviral long-term treatment against cirrhosis trial. Hepatology 2010; 51(2):585–594. doi:10.1002/hep.23315
  12. Angulo P, Kleiner DE, Dam-Larsen S, et al. Liver fibrosis, but no other histologic features, is associated with long-term outcomes of patients with nonalcoholic fatty liver disease. Gastroenterology 2015; 149(2):389–397.e10. doi:10.1053/j.gastro.2015.04.043
  13. Lindor KD, Bru C, Jorgensen RA, et al. The role of ultrasonography and automatic-needle biopsy in outpatient percutaneous liver biopsy. Hepatology 1996; 23(5):1079–1083. doi:10.1002/hep.510230522
  14. Pasha T, Gabriel S, Therneau T, Dickson ER, Lindor KD. Cost-effectiveness of ultrasound-guided liver biopsy. Hepatology 1998; 27(5):1220–1226. doi:10.1002/hep.510270506
  15. Alessandria C, Debernardi-Venon W, Rizzetto M, Marzano A. Transjugular liver biopsy: a relatively simple procedure with an indefinite past and an expected brilliant future. J Hepatol 2008; 48(1):171–173. doi:10.1016/j.jhep.2007.10.001
  16. Kalambokis G, Manousou P, Vibhakorn S, et al. Transjugular liver biopsy—indications, adequacy, quality of specimens, and complications—a systematic review. J Hepatol 2007; 47(2):284–294. doi:10.1016/j.jhep.2007.05.001
  17. Ripoll C, Groszmann R, Garcia-Tsao G, et al; Portal Hypertension Collaborative Group. Hepatic venous pressure gradient predicts clinical decompensation in patients with compensated cirrhosis. Gastroenterology 2007; 133(2):481–488. doi:10.1053/j.gastro.2007.05.024
  18. Perrault J, McGill DB, Ott BJ, Taylor WF. Liver biopsy: complications in 1000 inpatients and outpatients. Gastroenterology 1978; 74(1):103–106. pmid:618417
  19. Janes CH, Lindor KD. Outcome of patients hospitalized for complications after outpatient liver biopsy. Ann Intern Med 1993; 118(2):96–98. pmid:8416324
  20. Bedossa P, Dargere D, Paradis V. Sampling variability of liver fibrosis in chronic hepatitis C. Hepatology 2003; 38(6):1449–1457. doi:10.1016/j.hep.2003.09.022
  21. Regev A, Berho M, Jeffers LJ, et al. Sampling error and intraobserver variation in liver biopsy in patients with chronic HCV infection. Am J Gastroenterol 2002; 97(10):2614–2618. doi:10.1111/j.1572-0241.2002.06038.x
  22. Goldin RD, Goldin JG, Burt AD, et al. Intra-observer and inter-observer variation in the histopathological assessment of chronic viral hepatitis. J Hepatol 1996; 25(5):649–654. pmid:8938541
  23. Intraobserver and interobserver variations in liver biopsy interpretation in patients with chronic hepatitis C. The French METAVIR Cooperative Study Group. Hepatology 1994; 20(1 Pt 1):15–20. pmid:8020885
  24. Sterling RK, Lissen E, Clumeck N, et al; APRICOT Clinical Investigators. Development of a simple noninvasive index to predict significant fibrosis in patients with HIV/HCV coinfection. Hepatology 2006; 43(6):1317–1325. doi:10.1002/hep.21178
  25. Vallet-Pichard A, Mallet V, Nalpas B, et al. FIB-4: an inexpensive and accurate marker of fibrosis in HCV infection. comparison with liver biopsy and fibrotest. Hepatology 2007; 46(1):32–36. doi:10.1002/hep.21669
  26. Shah AG, Lydecker A, Murray K, Tetri BN, Contos MJ, Sanyal AJ; Nash Clinical Research Network. Comparison of noninvasive markers of fibrosis in patients with nonalcoholic fatty liver disease. Clin Gastroenterol Hepatol 2009; 7(10):1104–1112. doi:10.1016/j.cgh.2009.05.033
  27. McPherson S, Stewart SF, Henderson E, Burt AD, Day CP. Simple non-invasive fibrosis scoring systems can reliably exclude advanced fibrosis in patients with non-alcoholic fatty liver disease. Gut 2010; 59(9):1265–1269. doi:10.1136/gut.2010.216077
  28. Angulo P, Hui JM, Marchesini G, et al. The NAFLD fibrosis score: A noninvasive system that identifies liver fibrosis in patients with NAFLD. Hepatology 2007; 45(4):846–854. doi:10.1002/hep.21496
  29. Goh GB, Pagadala MR, Dasarathy J, et al. Clinical spectrum of non-alcoholic fatty liver disease in diabetic and non-diabetic patients. BBA Clin 2015; 3:141–145. doi:10.1016/j.bbacli.2014.09.001
  30. Tapper EB, Hunink MG, Afdhal NH, Lai M, Sengupta N. Cost-effectiveness analysis: risk stratification of nonalcoholic fatty liver disease (NAFLD) by the primary care physician using the NAFLD fibrosis score. PLoS One 2016; 11(2):e0147237. doi:10.1371/journal.pone.0147237
  31. Wai CT, Greenson JK, Fontana RJ, et al. A simple noninvasive index can predict both significant fibrosis and cirrhosis in patients with chronic hepatitis C. Hepatology 2003; 38(2):518–526. doi:10.1053/jhep.2003.50346
  32. Calès P, Lainé F, Boursier J, et al. Comparison of blood tests for liver fibrosis specific or not to NAFLD. J Hepatol 2009; 50(1):165–173. doi:10.1016/j.jhep.2008.07.035
  33. Imbert-Bismut F, Ratziu V, Pieroni L, Charlotte F, Benhamou Y, Poynard T; MULTIVIRC Group. Biochemical markers of liver fibrosis in patients with hepatitis C virus infection: a prospective study. Lancet 2001; 357(9262):1069–1075. doi:10.1016/S0140-6736(00)04258-6
  34. Shaheen AA, Wan AF, Myers RP. FibroTest and FibroScan for the prediction of hepatitis C-related fibrosis: a systematic review of diagnostic test accuracy. Am J Gastroenterol 2007; 102(11):2589–2600. doi:10.1111/j.1572-0241.2007.01466.x
  35. Smith JO, Sterling RK. Systematic review: non-invasive methods of fibrosis analysis in chronic hepatitis C. Aliment Pharmacol Ther 2009; 30(6):557–576. doi:10.1111/j.1365-2036.2009.04062.x
  36. Sebastiani G, Vario A, Guido M, Alberti A. Performance of noninvasive markers for liver fibrosis is reduced in chronic hepatitis C with normal transaminases. J Viral Hepat 2007; 15(3):212–218. doi:10.1111/j.1365-2893.2007.00932.x
  37. Poynard T, Morra R, Halfon P, et al. Meta-analyses of FibroTest diagnostic value in chronic liver disease. BMC Gastroenterol 2007; 7:40. doi:10.1186/1471-230X-7-40
  38. Carlson JJ, Kowdley KV, Sullivan SD, Ramsey SD, Veenstra DL. An evaluation of the potential cost-effectiveness of non-invasive testing strategies in the diagnosis of significant liver fibrosis. J Gastroenterol Hepatol 2009; 24(5):786–791. doi:10.1111/j.1440-1746.2009.05778.x
  39. Aubé C, Oberti F, Korali N, et al. Ultrasonographic diagnosis of hepatic fibrosis or cirrhosis. J Hepatol 1999; 30(3):472–478. pmid:10190731
  40. Di Lelio A, Cestari C, Lomazzi A, Beretta L. Cirrhosis: diagnosis with sonographic study of the liver surface. Radiology 1989; 172(2):389–392. doi:10.1148/radiology.172.2.2526349
  41. Wong VW, Chan HL. Transient elastography. J Gastroenterol Hepatol 2010; 25(11):1726–1731. doi:10.1111/j.1440-1746.2010.06437.x
  42. Arena U, Vizzutti F, Abraldes JG, et al. Reliability of transient elastography for the diagnosis of advanced fibrosis in chronic hepatitis C. Gut 2008; 57(9):1288–1293. doi:10.1136/gut.2008.149708
  43. Ziol M, Handra-Luca A, Kettaneh A, et al. Noninvasive assessment of liver fibrosis by measurement of stiffness in patients with chronic hepatitis C. Hepatology 2005; 41(1):48–54. doi:10.1002/hep.20506
  44. Wong VW, Vergniol J, Wong GL, et al. Diagnosis of fibrosis and cirrhosis using liver stiffness measurement in nonalcoholic fatty liver disease. Hepatology 2010; 51(2):454–462. doi:10.1002/hep.23312
  45. Sagir A, Erhardt A, Schmitt M, Häussinger D. Transient elastography is unreliable for detection of cirrhosis in patients with acute liver damage. Hepatology 2007; 48(2):592–595. doi:10.1002/hep.22056
  46. Mederacke I, Wursthorn K, Kirschner J, et al. Food intake increases liver stiffness in patients with chronic or resolved hepatitis C virus infection. Liver Int 2009; 29(10):1500–1506. doi:10.1111/j.1478-3231.2009.02100.x
  47. Castéra L, Foucher J, Bernard PH, et al. Pitfalls of liver stiffness measurement: a 5-year prospective study of 13,369 examinations. Hepatology 2010; 51(3):828–835. doi:10.1002/hep.23425
  48. Wong VW, Vergniol J, Wong GL, et al. Liver stiffness measurement using XL probe in patients with nonalcoholic fatty liver disease. Am J Gastroenterol 2012; 107(12):1862–1871. doi:10.1038/ajg.2012.331
  49. Fraquelli M, Rigamonti C, Casazza G, et al. Reproducibility of transient elastography in the evaluation of liver fibrosis in patients with chronic liver disease. Gut 2007; 56(7):968–973. doi:10.1136/gut.2006.111302
  50. de Franchis R; Baveno VI Faculty. Expanding consensus in portal hypertension: report of the Baveno VI Consensus Workshop: stratifying risk and individualizing care for portal hypertension. J Hepatol 2015; 63(3):743–752. doi:10.1016/j.jhep.2015.05.022
  51. Friedrich-Rust M, Wunder K, Kriener S, et al. Liver fibrosis in viral hepatitis: noninvasive assessment with acoustic radiation force impulse imaging versus transient elastography. Radiology 2009; 252(2):595–604. doi:10.1148/radiol.2523081928
  52. Yoneda M, Suzuki K, Kato S, et al. Nonalcoholic fatty liver disease: US-based acoustic radiation force impulse elastography. Radiology 2010; 256(2):640–647. doi:10.1148/radiol.10091662
  53. Bota S, Herkner H, Sporea I, et al. Meta-analysis: ARFI elastography versus transient elastography for the evaluation of liver fibrosis. Liver Int 2013; 33(8):1138–1147. doi:10.1111/liv.12240
  54. Attia D, Bantel H, Lenzen H, Manns MP, Gebel MJ, Potthoff A. Liver stiffness measurement using acoustic radiation force impulse elastography in overweight and obese patients. Aliment Pharmacol Ther 2016; 44(4):366–379. doi:10.1111/apt.13710
  55. Cui J, Heba E, Hernandez C, et al. Magnetic resonance elastography is superior to acoustic radiation force impulse for the diagnosis of fibrosis in patients with biopsy-proven nonalcoholic fatty liver disease: a prospective study. Hepatology 2016; 63(2):453–461. doi:10.1002/hep.28337
  56. Huwart L, Sempoux C, Vicaut E, et al. Magnetic resonance elastography for the noninvasive staging of liver fibrosis. Gastroenterology 2008; 135(1):32–40. doi:10.1053/j.gastro.2008.03.076
  57. Jajamovich GH, Dyvorne H, Donnerhack C, Taouli B. Quantitative liver MRI combining phase contrast imaging, elastography, and DWI: assessment of reproducibility and postprandial effect at 3.0 T. PLoS One 2014; 9(5):e97355. doi:10.1371/journal.pone.0097355
  58. Lim JK, Flamm SL, Singh S, Falck-Ytter YT; Clinical Guidelines Committee of the American Gastroenterological Association. American Gastroenterological Association Institute guideline on the role of elastography in the evaluation of liver fibrosis. Gastroenterology 2017; 152(6):1536–1543. doi:10.1053/j.gastro.2017.03.017
  59. N, Feldstein AE. Noninvasive diagnosis of nonalcoholic fatty liver disease: are we there yet? Metabolism 2016; 65(8):1087–1095. doi:10.1016/j.metabol.2016.01.013
Article PDF
Author and Disclosure Information

Tavankit Singh, MD
Department of Gastroenterology and Hepatology, Cleveland Clinic

Daniela S. Allende, MD
Director, Hepatobiliary Pathology, Department of Pathology, Cleveland Clinic; Associate
Professor, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH

Arthur J. McCullough, MD
Departments of Gastroenterology and Hepatology and Pathobiology and Transplantation Center, Cleveland Clinic; Professor, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH

Address: Arthur J. McCullough, MD, Department of Gastroenterology and Hepatology, A30, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195; mcculla@ccf.org

Issue
Cleveland Clinic Journal of Medicine - 86(3)
Publications
Topics
Page Number
179-186
Legacy Keywords
liver, fibrosis, nonalcoholic fatty liver disease, NAFLD, nonalcoholic steatohepatitis, NASH, cirrhosis, hepatitis C virus, HCV, biopsy, staging, Ishak, METAVIR, FIB-4 index, NAFLD fibrosis score, AST-to-platelet raio index, APRI, FibroSure, ultrasonography, transient elastography, acoustic radiation force imaging, liver stiffness measurement, magnetic resonance elastography, Tavankit Singh, Daniela Allende, Arthur McCullough
Sections
Author and Disclosure Information

Tavankit Singh, MD
Department of Gastroenterology and Hepatology, Cleveland Clinic

Daniela S. Allende, MD
Director, Hepatobiliary Pathology, Department of Pathology, Cleveland Clinic; Associate
Professor, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH

Arthur J. McCullough, MD
Departments of Gastroenterology and Hepatology and Pathobiology and Transplantation Center, Cleveland Clinic; Professor, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH

Address: Arthur J. McCullough, MD, Department of Gastroenterology and Hepatology, A30, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195; mcculla@ccf.org

Author and Disclosure Information

Tavankit Singh, MD
Department of Gastroenterology and Hepatology, Cleveland Clinic

Daniela S. Allende, MD
Director, Hepatobiliary Pathology, Department of Pathology, Cleveland Clinic; Associate
Professor, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH

Arthur J. McCullough, MD
Departments of Gastroenterology and Hepatology and Pathobiology and Transplantation Center, Cleveland Clinic; Professor, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH

Address: Arthur J. McCullough, MD, Department of Gastroenterology and Hepatology, A30, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195; mcculla@ccf.org

Article PDF
Article PDF
Related Articles

Staging of liver fibrosis, important for determining prognosis in patients with chronic liver disease and for the need to start screening for complications of cirrhosis, was traditionally done only by liver biopsy. While biopsy is still the gold standard method to stage fibrosis, noninvasive methods have been developed that can also assess disease severity.

This article briefly reviews the epidemiology and physiology of chronic liver disease and the traditional role of liver biopsy. Pros and cons of alternative fibrosis assessment methods are discussed, with a focus on their utility for patients with nonalcoholic fatty liver disease (NAFLD) and hepatitis C virus (HCV) infection.

CHRONIC LIVER DISEASE: A HUGE HEALTH BURDEN

Chronic liver disease is associated with enormous health and financial costs in the United States. Its prevalence is about 15%,1 and it is the 12th leading cause of death.2 Hospital costs are estimated at about $4 billion annually.3

The most common causes of chronic liver disease are NAFLD (which may be present in up to one-third of the US population and is increasing with the epidemic of obesity), its aggressive variant, nonalcoholic steatohepatitis (NASH) (present in about 3% of the population), and HCV infection (1%).4,5

Since direct-acting antiviral agents were introduced, HCV infection dropped from being the leading cause of liver transplant to third place.6 But at the same time, the number of patients on the transplant waiting list who have NASH has risen faster than for any other cause of chronic liver disease.7

FIBROSIS: A KEY INDICATOR OF DISEASE SEVERITY

singh_assessingliverfibrosis_t1.jpg
With any form of liver disease, collagen is deposited in hepatic lobules over time, a process called fibrosis. Both HCV infection and NASH involve necroinflammation in the liver, hepatocyte apoptosis, and activation of stellate cells, leading to progressive collagen deposition in hepatic lobules. Fibrosis typically starts in the region of the central vein and portal tracts and eventually extends to other areas of the lobule.

singh_assessingliverfibrosis_f1.jpg
%3Cp%3EFigure%201.%20Findings%20on%20liver%20biopsy%20in%20nonalcoholic%20fatty%20liver%20disease%20and%20hepatitis%20C%20virus%20infection.%3C%2Fp%3E
Determining fibrosis severity is critical when a patient is diagnosed with chronic liver disease, as it predicts long-term clinical outcomes and death in HCV8 and NAFLD.9 Different staging systems have been developed to reflect the degree of fibrosis, based on its distribution as seen on liver biopsy (Table 1, Figure 1).

In HCV infection, advanced fibrosis is defined as either stage 4 to 6 using the Ishak system10 or stage 3 to 4 using the Meta-analysis of Histological Data in Viral Hepatitis (METAVIR) system.11

In NAFLD, advanced fibrosis is defined as stage 3 to 4 using the NASH Clinical Research Network system.12

Staging fibrosis is also important so that patients with cirrhosis can be identified early to begin screening for hepatocellular carcinoma and esophageal varices to reduce the risks of illness and death. In addition, insurance companies often require documentation of fibrosis stage before treating HCV with the new direct-acting antiviral agents.

LIVER BIOPSY IS STILL THE GOLD STANDARD

Although invasive, liver biopsy remains the gold standard for determining fibrosis stage. Liver biopsies were performed “blindly” (without imaging) until the 1990s, but imaging-guided biopsy using ultrasonography was then developed, which entailed less pain and lower complication and hospitalization rates. Slightly more hepatic tissue is obtained with guided liver biopsy, but the difference was deemed clinically insignificant.13 Concern initially arose about the added cost involved with imaging, but imaging-guided biopsy was actually found to be more cost-effective.14

In the 2000s, transjugular liver biopsy via the right internal jugular vein became available. This method was originally used primarily in patients with ascites or significant coagulopathy. At first, there were concerns about the adequacy of specimens obtained to make an accurate diagnosis or establish fibrosis stage, but this limitation was overcome with improved techniques.15,16 Transjugular liver biopsy has the additional advantage of enabling one to measure the hepatic venous pressure gradient, which also has prognostic significance; a gradient greater than 10 mm Hg is associated with worse prognosis.17

Disadvantages of biopsy: Complications, sampling errors

Liver biopsy has disadvantages. Reported rates of complications necessitating hospitalization using the blind method were as high as 6% in the 1970s,18 dropping to 3.2% in a 1993 study.19 Bleeding remains the most worrisome complication. With the transjugular method, major and minor complication rates are less than 1% and 7%, respectively.15,16 Complication rates with imaging-guided biopsy are also low.

Liver biopsy is also prone to sampling error. The number of portal tracts obtained in the biopsy correlates with the accuracy of fibrosis staging, and smaller samples may lead to underestimating fibrosis stage. In patients with HCV, samples more than 15 mm long led to accurate staging diagnosis in 65% of patients, and those longer than 25 mm conferred 75% accuracy.20 Also, different stages can be diagnosed from samples obtained from separate locations in the liver, although rarely is the difference more than a single stage.21

Histologic evaluation of liver biopsies is operator-dependent. Although significant interobserver variation has been reported for degree of inflammation, there tends to be good concordance for fibrosis staging.22,23

 

 

STAGING BASED ON DEMOGRAPHIC AND LABORATORY VARIABLES

Several scores based on patient characteristics and laboratory values have been developed for assessing liver fibrosis and have been specifically validated for HCV infection, NAFLD, or both. They can serve as inexpensive initial screening tests for the presence or absence of advanced fibrosis.

FIB-4 index for HCV, NAFLD

The FIB-4 index predicts the presence of advanced fibrosis using, as its name indicates, a combination of 4 factors in fibrosis: age, platelet count, and the levels of aspartate aminotransferase (AST) and alanine aminotransferase (ALT), according to the formula:

FIB-4 index = (age × AST [U/L]) /
(platelet count [× 109/L] × √ALT [U/L]).

The index was derived from data from 832 patients co-infected with HCV and human immunodeficiency virus.24 The Ishak staging system10 for fibrosis on liver biopsy was used for confirmation, with stage 4 to 6 defined as advanced fibrosis. A cutoff value of more than 3.25 had a positive predictive value of 65% for advanced fibrosis, and to exclude advanced fibrosis, a cutoff value of less than 1.45 had a negative predictive value of 90%.

The FIB-4 index has since been validated in patients with HCV infection25 and NAFLD.26 In a subsequent study in 142 patients with NAFLD, the FIB-4 index was more accurate in diagnosing advanced fibrosis than the other noninvasive prediction models discussed below.27

NAFLD fibrosis score

The NAFLD fibrosis score, constructed and validated only in patients with biopsy-confirmed NAFLD, incorporates age, body mass index, presence of diabetes or prediabetes, albumin level, platelet count, and AST and ALT levels.

A group of 480 patients was used to construct the score, and 253 patients were used to validate it. Using the high cutoff value of 0.676, the presence of advanced fibrosis was diagnosed with a positive predictive value of 90% in the group used to construct the model (82% in the validation group). Using the low cutoff score of –1.455, advanced fibrosis could be excluded with a negative predictive value of 93% in the construction group and 88% in the validation group.28 A score between the cutoff values merits liver biopsy to determine fibrosis stage. The score is more accurate in patients with diabetes.29 When used by primary care physicians, the NAFLD fibrosis score is more cost-effective than transient elastography and liver biopsy for accurately predicting advanced fibrosis.30

AST-to-platelet ratio index score for HCV, NAFLD

The AST-to-platelet ratio index (APRI) score was developed in 2003 using a cohort of 270 patients with HCV and liver biopsy as the standard. A cutoff value of less than or equal to 0.5 had a negative predictive value of 86% for the absence of significant fibrosis, while a score of more than 1.5 detected the presence of significant fibrosis with a positive predictive value of 88%.31 The APRI score was subsequently validated for NAFLD.27,32

FibroSure uses a patented formula

FibroSure (LabCorp; labcorp.com) uses a patented mathematical formula that takes into account age, sex, and levels of gamma-glutamyl transferase, total bilirubin, haptoglobin, apolipoprotein-A, and alpha-2 macroglobulin to assess fibrosis. Developed in 2001 for use in patients with HCV infection, it was reported to have a positive predictive value of greater than 90% and a negative predictive value of 100% for clinically significant fibrosis, defined as stage 2 to 4 based on the METAVIR staging system in the prediction model.33 The use of FibroSure in patients with HCV was subsequently validated in various meta-analyses and systematic reviews.34,35 It is less accurate in patients with normal ALT levels.36

FibroSure also has good accuracy for predicting fibrosis stage in chronic liver disease due to other causes, including NAFLD.37

The prediction models discussed above use routine laboratory tests for chronic liver disease and thus are inexpensive. The high cost of additional testing needed for FibroSure, coupled with the risk of misdiagnosis, makes its cost-effectiveness questionable.38

 

 

IMAGING TO PREDICT FIBROSIS STAGE

Conventional ultrasonography (with or without vascular imaging) and computed tomography can detect cirrhosis on the basis of certain imaging characteristics,39,40 including the nodular contour of the liver, caudate lobe hypertrophy, ascites, reversal of blood flow in the portal vein, and splenomegaly. However, they cannot detect fibrosis in its early stages.

The 3 methods discussed below provide more accurate fibrosis staging by measuring the velocity of shear waves sent across hepatic tissue. Because shear-wave velocity increases with liver stiffness, the fibrosis stage can be estimated from this information.41

Transient elastography

Transient elastography uses a special ultrasound transducer. It is highly accurate for predicting advanced fibrosis for almost all causes of chronic liver disease, including HCV infection42,43 and NAFLD.44 The cutoff values of wave velocity to estimate fibrosis stage differ by liver disease etiology.

Transient elastography should not be used to evaluate fibrosis in patients with acute hepatitis, which transiently increases liver stiffness, resulting in a falsely high fibrosis stage diagnosis.45 It is also not a good method for evaluating fibrosis in patients with biliary obstruction or extrahepatic venous congestion. Because liver stiffness can increase after eating,46 the test should be done under fasting conditions.

A significant limitation of transient elastography has been its poor accuracy in patients with obesity.47 This has been largely overcome with the use of a more powerful (XL) probe but is still a limitation for those with morbid obesity.48 Because many patients with NAFLD are obese, this limitation can be significant.

Transient elastography has gained popularity for evaluating fibrosis in patients with chronic liver disease for multiple reasons: it is cost-effective and results are highly reproducible, with low variation in results among different observers and in individual observers.49 Combined with a platelet count, it can also be used to detect the development of clinically significant portal hypertension in patients with cirrhosis, thus determining the need to screen for esophageal varices using endoscopy.50 Screening endoscopy can be avoided in patients whose liver stiffness remains below 20 kPa or whose platelet count is above 150 × 109/L.

Acoustic radiation force imaging

Unlike transient elastography, which requires a separate transducer probe to assess shear- wave velocity, acoustic radiation force imaging uses the same transducer for both this function and imaging. Different image modes are available when testing for liver stiffness, so a region of interest that is optimal for avoiding vascular structures or masses can be selected, increasing accuracy.51

Acoustic radiation force imaging has been tested in different causes of chronic liver disease, including HCV and NAFLD,52 with accuracy similar to that of transient elastography.53 For overweight and obese patients, acoustic radiation force imaging is more accurate than transient elastography using the XL probe.54 However, this method is still new, and we need more data to support using one method over the other.

Magnetic resonance elastography

Magnetic resonance elastography uses a special transducer placed under the rib cage to transmit shear waves concurrently with magnetic resonance imaging. It has been tested in patients with HCV and NAFLD and has been found to have better diagnostic accuracy than transient elastography and acoustic radiation force imaging.55,56 Patients must be fasting for better diagnostic accuracy57 and must hold their breath while elastography is performed. The need for breath-holding and the high cost limit the use of this method for assessing fibrosis.

BOTTOM LINE FOR ASSESSING FIBROSIS

singh_assessingliverfibrosis_f2.jpg
%3Cp%3EFigure%202.%20Algorithm%20to%20determine%20fibrosis%20stage%20for%20nonalcoholic%20fatty%20liver%20disease.%3C%2Fp%3E
Although liver biopsy remains the gold standard for accurately determining fibrosis stage, noninvasive methods, especially imaging techniques, are fast evolving. Guidelines recommend using transient elastography to determine fibrosis stage noninvasively in patients with HCV,58 but a similar recommendation cannot be made for NAFLD with available data. For NAFLD, combined elastography and NAFLD fibrosis score are recommended to determine the need for a liver biopsy (Figure 2).59 Currently, we recommend using a combination of the scores discussed above and the imaging tests.

Staging of liver fibrosis, important for determining prognosis in patients with chronic liver disease and for the need to start screening for complications of cirrhosis, was traditionally done only by liver biopsy. While biopsy is still the gold standard method to stage fibrosis, noninvasive methods have been developed that can also assess disease severity.

This article briefly reviews the epidemiology and physiology of chronic liver disease and the traditional role of liver biopsy. Pros and cons of alternative fibrosis assessment methods are discussed, with a focus on their utility for patients with nonalcoholic fatty liver disease (NAFLD) and hepatitis C virus (HCV) infection.

CHRONIC LIVER DISEASE: A HUGE HEALTH BURDEN

Chronic liver disease is associated with enormous health and financial costs in the United States. Its prevalence is about 15%,1 and it is the 12th leading cause of death.2 Hospital costs are estimated at about $4 billion annually.3

The most common causes of chronic liver disease are NAFLD (which may be present in up to one-third of the US population and is increasing with the epidemic of obesity), its aggressive variant, nonalcoholic steatohepatitis (NASH) (present in about 3% of the population), and HCV infection (1%).4,5

Since direct-acting antiviral agents were introduced, HCV infection dropped from being the leading cause of liver transplant to third place.6 But at the same time, the number of patients on the transplant waiting list who have NASH has risen faster than for any other cause of chronic liver disease.7

FIBROSIS: A KEY INDICATOR OF DISEASE SEVERITY

singh_assessingliverfibrosis_t1.jpg
With any form of liver disease, collagen is deposited in hepatic lobules over time, a process called fibrosis. Both HCV infection and NASH involve necroinflammation in the liver, hepatocyte apoptosis, and activation of stellate cells, leading to progressive collagen deposition in hepatic lobules. Fibrosis typically starts in the region of the central vein and portal tracts and eventually extends to other areas of the lobule.

singh_assessingliverfibrosis_f1.jpg
%3Cp%3EFigure%201.%20Findings%20on%20liver%20biopsy%20in%20nonalcoholic%20fatty%20liver%20disease%20and%20hepatitis%20C%20virus%20infection.%3C%2Fp%3E
Determining fibrosis severity is critical when a patient is diagnosed with chronic liver disease, as it predicts long-term clinical outcomes and death in HCV8 and NAFLD.9 Different staging systems have been developed to reflect the degree of fibrosis, based on its distribution as seen on liver biopsy (Table 1, Figure 1).

In HCV infection, advanced fibrosis is defined as either stage 4 to 6 using the Ishak system10 or stage 3 to 4 using the Meta-analysis of Histological Data in Viral Hepatitis (METAVIR) system.11

In NAFLD, advanced fibrosis is defined as stage 3 to 4 using the NASH Clinical Research Network system.12

Staging fibrosis is also important so that patients with cirrhosis can be identified early to begin screening for hepatocellular carcinoma and esophageal varices to reduce the risks of illness and death. In addition, insurance companies often require documentation of fibrosis stage before treating HCV with the new direct-acting antiviral agents.

LIVER BIOPSY IS STILL THE GOLD STANDARD

Although invasive, liver biopsy remains the gold standard for determining fibrosis stage. Liver biopsies were performed “blindly” (without imaging) until the 1990s, but imaging-guided biopsy using ultrasonography was then developed, which entailed less pain and lower complication and hospitalization rates. Slightly more hepatic tissue is obtained with guided liver biopsy, but the difference was deemed clinically insignificant.13 Concern initially arose about the added cost involved with imaging, but imaging-guided biopsy was actually found to be more cost-effective.14

In the 2000s, transjugular liver biopsy via the right internal jugular vein became available. This method was originally used primarily in patients with ascites or significant coagulopathy. At first, there were concerns about the adequacy of specimens obtained to make an accurate diagnosis or establish fibrosis stage, but this limitation was overcome with improved techniques.15,16 Transjugular liver biopsy has the additional advantage of enabling one to measure the hepatic venous pressure gradient, which also has prognostic significance; a gradient greater than 10 mm Hg is associated with worse prognosis.17

Disadvantages of biopsy: Complications, sampling errors

Liver biopsy has disadvantages. Reported rates of complications necessitating hospitalization using the blind method were as high as 6% in the 1970s,18 dropping to 3.2% in a 1993 study.19 Bleeding remains the most worrisome complication. With the transjugular method, major and minor complication rates are less than 1% and 7%, respectively.15,16 Complication rates with imaging-guided biopsy are also low.

Liver biopsy is also prone to sampling error. The number of portal tracts obtained in the biopsy correlates with the accuracy of fibrosis staging, and smaller samples may lead to underestimating fibrosis stage. In patients with HCV, samples more than 15 mm long led to accurate staging diagnosis in 65% of patients, and those longer than 25 mm conferred 75% accuracy.20 Also, different stages can be diagnosed from samples obtained from separate locations in the liver, although rarely is the difference more than a single stage.21

Histologic evaluation of liver biopsies is operator-dependent. Although significant interobserver variation has been reported for degree of inflammation, there tends to be good concordance for fibrosis staging.22,23

 

 

STAGING BASED ON DEMOGRAPHIC AND LABORATORY VARIABLES

Several scores based on patient characteristics and laboratory values have been developed for assessing liver fibrosis and have been specifically validated for HCV infection, NAFLD, or both. They can serve as inexpensive initial screening tests for the presence or absence of advanced fibrosis.

FIB-4 index for HCV, NAFLD

The FIB-4 index predicts the presence of advanced fibrosis using, as its name indicates, a combination of 4 factors in fibrosis: age, platelet count, and the levels of aspartate aminotransferase (AST) and alanine aminotransferase (ALT), according to the formula:

FIB-4 index = (age × AST [U/L]) /
(platelet count [× 109/L] × √ALT [U/L]).

The index was derived from data from 832 patients co-infected with HCV and human immunodeficiency virus.24 The Ishak staging system10 for fibrosis on liver biopsy was used for confirmation, with stage 4 to 6 defined as advanced fibrosis. A cutoff value of more than 3.25 had a positive predictive value of 65% for advanced fibrosis, and to exclude advanced fibrosis, a cutoff value of less than 1.45 had a negative predictive value of 90%.

The FIB-4 index has since been validated in patients with HCV infection25 and NAFLD.26 In a subsequent study in 142 patients with NAFLD, the FIB-4 index was more accurate in diagnosing advanced fibrosis than the other noninvasive prediction models discussed below.27

NAFLD fibrosis score

The NAFLD fibrosis score, constructed and validated only in patients with biopsy-confirmed NAFLD, incorporates age, body mass index, presence of diabetes or prediabetes, albumin level, platelet count, and AST and ALT levels.

A group of 480 patients was used to construct the score, and 253 patients were used to validate it. Using the high cutoff value of 0.676, the presence of advanced fibrosis was diagnosed with a positive predictive value of 90% in the group used to construct the model (82% in the validation group). Using the low cutoff score of –1.455, advanced fibrosis could be excluded with a negative predictive value of 93% in the construction group and 88% in the validation group.28 A score between the cutoff values merits liver biopsy to determine fibrosis stage. The score is more accurate in patients with diabetes.29 When used by primary care physicians, the NAFLD fibrosis score is more cost-effective than transient elastography and liver biopsy for accurately predicting advanced fibrosis.30

AST-to-platelet ratio index score for HCV, NAFLD

The AST-to-platelet ratio index (APRI) score was developed in 2003 using a cohort of 270 patients with HCV and liver biopsy as the standard. A cutoff value of less than or equal to 0.5 had a negative predictive value of 86% for the absence of significant fibrosis, while a score of more than 1.5 detected the presence of significant fibrosis with a positive predictive value of 88%.31 The APRI score was subsequently validated for NAFLD.27,32

FibroSure uses a patented formula

FibroSure (LabCorp; labcorp.com) uses a patented mathematical formula that takes into account age, sex, and levels of gamma-glutamyl transferase, total bilirubin, haptoglobin, apolipoprotein-A, and alpha-2 macroglobulin to assess fibrosis. Developed in 2001 for use in patients with HCV infection, it was reported to have a positive predictive value of greater than 90% and a negative predictive value of 100% for clinically significant fibrosis, defined as stage 2 to 4 based on the METAVIR staging system in the prediction model.33 The use of FibroSure in patients with HCV was subsequently validated in various meta-analyses and systematic reviews.34,35 It is less accurate in patients with normal ALT levels.36

FibroSure also has good accuracy for predicting fibrosis stage in chronic liver disease due to other causes, including NAFLD.37

The prediction models discussed above use routine laboratory tests for chronic liver disease and thus are inexpensive. The high cost of additional testing needed for FibroSure, coupled with the risk of misdiagnosis, makes its cost-effectiveness questionable.38

 

 

IMAGING TO PREDICT FIBROSIS STAGE

Conventional ultrasonography (with or without vascular imaging) and computed tomography can detect cirrhosis on the basis of certain imaging characteristics,39,40 including the nodular contour of the liver, caudate lobe hypertrophy, ascites, reversal of blood flow in the portal vein, and splenomegaly. However, they cannot detect fibrosis in its early stages.

The 3 methods discussed below provide more accurate fibrosis staging by measuring the velocity of shear waves sent across hepatic tissue. Because shear-wave velocity increases with liver stiffness, the fibrosis stage can be estimated from this information.41

Transient elastography

Transient elastography uses a special ultrasound transducer. It is highly accurate for predicting advanced fibrosis for almost all causes of chronic liver disease, including HCV infection42,43 and NAFLD.44 The cutoff values of wave velocity to estimate fibrosis stage differ by liver disease etiology.

Transient elastography should not be used to evaluate fibrosis in patients with acute hepatitis, which transiently increases liver stiffness, resulting in a falsely high fibrosis stage diagnosis.45 It is also not a good method for evaluating fibrosis in patients with biliary obstruction or extrahepatic venous congestion. Because liver stiffness can increase after eating,46 the test should be done under fasting conditions.

A significant limitation of transient elastography has been its poor accuracy in patients with obesity.47 This has been largely overcome with the use of a more powerful (XL) probe but is still a limitation for those with morbid obesity.48 Because many patients with NAFLD are obese, this limitation can be significant.

Transient elastography has gained popularity for evaluating fibrosis in patients with chronic liver disease for multiple reasons: it is cost-effective and results are highly reproducible, with low variation in results among different observers and in individual observers.49 Combined with a platelet count, it can also be used to detect the development of clinically significant portal hypertension in patients with cirrhosis, thus determining the need to screen for esophageal varices using endoscopy.50 Screening endoscopy can be avoided in patients whose liver stiffness remains below 20 kPa or whose platelet count is above 150 × 109/L.

Acoustic radiation force imaging

Unlike transient elastography, which requires a separate transducer probe to assess shear- wave velocity, acoustic radiation force imaging uses the same transducer for both this function and imaging. Different image modes are available when testing for liver stiffness, so a region of interest that is optimal for avoiding vascular structures or masses can be selected, increasing accuracy.51

Acoustic radiation force imaging has been tested in different causes of chronic liver disease, including HCV and NAFLD,52 with accuracy similar to that of transient elastography.53 For overweight and obese patients, acoustic radiation force imaging is more accurate than transient elastography using the XL probe.54 However, this method is still new, and we need more data to support using one method over the other.

Magnetic resonance elastography

Magnetic resonance elastography uses a special transducer placed under the rib cage to transmit shear waves concurrently with magnetic resonance imaging. It has been tested in patients with HCV and NAFLD and has been found to have better diagnostic accuracy than transient elastography and acoustic radiation force imaging.55,56 Patients must be fasting for better diagnostic accuracy57 and must hold their breath while elastography is performed. The need for breath-holding and the high cost limit the use of this method for assessing fibrosis.

BOTTOM LINE FOR ASSESSING FIBROSIS

singh_assessingliverfibrosis_f2.jpg
%3Cp%3EFigure%202.%20Algorithm%20to%20determine%20fibrosis%20stage%20for%20nonalcoholic%20fatty%20liver%20disease.%3C%2Fp%3E
Although liver biopsy remains the gold standard for accurately determining fibrosis stage, noninvasive methods, especially imaging techniques, are fast evolving. Guidelines recommend using transient elastography to determine fibrosis stage noninvasively in patients with HCV,58 but a similar recommendation cannot be made for NAFLD with available data. For NAFLD, combined elastography and NAFLD fibrosis score are recommended to determine the need for a liver biopsy (Figure 2).59 Currently, we recommend using a combination of the scores discussed above and the imaging tests.

References
  1. Younossi ZM, Stepanova M, Afendy M, et al. Changes in the prevalence of the most common causes of chronic liver diseases in the United States from 1988 to 2008. Clin Gastroenterol Hepatol 2011; 9(6):524–530.e1. doi:10.1016/j.cgh.2011.03.020
  2. Kochanek KD, Xu J, Murphy SL, Miniño AM, Kung H-C. Deaths: final data for 2009. Natl Vital Stat Rep 2011; 60(3):1–116. pmid:24974587
  3. Volk ML, Tocco RS, Bazick J, Rakoski MO, Lok AS. Hospital readmissions among patients with decompensated cirrhosis. Am J Gastroenterol 2012; 107(2):247–252. doi:10.1038/ajg.2011.314
  4. Vernon G, Baranova A, Younossi ZM. Systematic review: the epidemiology and natural history of non-alcoholic fatty liver disease and non-alcoholic steatohepatitis in adults. Aliment Pharmacol Ther 2011; 34(3):274–285. doi:10.1111/j.1365-2036.2011.04724.x
  5. Udompap P, Kim D, Kim WR. Current and future burden of chronic nonmalignant liver disease. Clin Gastroenterol Hepatol 2015; 13(12):2031–2041. doi:10.1016/j.cgh.2015.08.015
  6. Kim WR, Lake JR, Smith JM, et al. OPTN/SRTR 2016 annual data report: liver. Am J Transplant 2018; 18(suppl 1):172–253. doi:10.1111/ajt.14559
  7. Wong RJ, Aguilar M, Cheung R, et al. Nonalcoholic steatohepatitis is the second leading etiology of liver disease among adults awaiting liver transplantation in the United States. Gastroenterology 2015; 148(3):547–555. doi:10.1053/j.gastro.2014.11.039
  8. Ishak K, Baptista A, Bianchi L, et al. Histological grading and staging of chronic hepatitis. J Hepatol 1995; 22(6):696–699. pmid:7560864
  9. Bedossa P, Poynard T. An algorithm for the grading of activity in chronic hepatitis C. Hepatology 1996; 24(2):289–293. doi:10.1002/hep.510240201
  10. Kleiner DE, Brunt EM, Van Natta M, et al; Nonalcoholic Steatohepatitis Clinical Research Network. Design and validation of a histological scoring system for nonalcoholic fatty liver disease. Hepatology 2005; 41(6):1313–1321. doi:10.1002/hep.20701
  11. Everhart JE, Wright EC, Goodman ZD, et al; HALT-C Trial Group. Prognostic value of Ishak fibrosis stage: findings from the hepatitis C antiviral long-term treatment against cirrhosis trial. Hepatology 2010; 51(2):585–594. doi:10.1002/hep.23315
  12. Angulo P, Kleiner DE, Dam-Larsen S, et al. Liver fibrosis, but no other histologic features, is associated with long-term outcomes of patients with nonalcoholic fatty liver disease. Gastroenterology 2015; 149(2):389–397.e10. doi:10.1053/j.gastro.2015.04.043
  13. Lindor KD, Bru C, Jorgensen RA, et al. The role of ultrasonography and automatic-needle biopsy in outpatient percutaneous liver biopsy. Hepatology 1996; 23(5):1079–1083. doi:10.1002/hep.510230522
  14. Pasha T, Gabriel S, Therneau T, Dickson ER, Lindor KD. Cost-effectiveness of ultrasound-guided liver biopsy. Hepatology 1998; 27(5):1220–1226. doi:10.1002/hep.510270506
  15. Alessandria C, Debernardi-Venon W, Rizzetto M, Marzano A. Transjugular liver biopsy: a relatively simple procedure with an indefinite past and an expected brilliant future. J Hepatol 2008; 48(1):171–173. doi:10.1016/j.jhep.2007.10.001
  16. Kalambokis G, Manousou P, Vibhakorn S, et al. Transjugular liver biopsy—indications, adequacy, quality of specimens, and complications—a systematic review. J Hepatol 2007; 47(2):284–294. doi:10.1016/j.jhep.2007.05.001
  17. Ripoll C, Groszmann R, Garcia-Tsao G, et al; Portal Hypertension Collaborative Group. Hepatic venous pressure gradient predicts clinical decompensation in patients with compensated cirrhosis. Gastroenterology 2007; 133(2):481–488. doi:10.1053/j.gastro.2007.05.024
  18. Perrault J, McGill DB, Ott BJ, Taylor WF. Liver biopsy: complications in 1000 inpatients and outpatients. Gastroenterology 1978; 74(1):103–106. pmid:618417
  19. Janes CH, Lindor KD. Outcome of patients hospitalized for complications after outpatient liver biopsy. Ann Intern Med 1993; 118(2):96–98. pmid:8416324
  20. Bedossa P, Dargere D, Paradis V. Sampling variability of liver fibrosis in chronic hepatitis C. Hepatology 2003; 38(6):1449–1457. doi:10.1016/j.hep.2003.09.022
  21. Regev A, Berho M, Jeffers LJ, et al. Sampling error and intraobserver variation in liver biopsy in patients with chronic HCV infection. Am J Gastroenterol 2002; 97(10):2614–2618. doi:10.1111/j.1572-0241.2002.06038.x
  22. Goldin RD, Goldin JG, Burt AD, et al. Intra-observer and inter-observer variation in the histopathological assessment of chronic viral hepatitis. J Hepatol 1996; 25(5):649–654. pmid:8938541
  23. Intraobserver and interobserver variations in liver biopsy interpretation in patients with chronic hepatitis C. The French METAVIR Cooperative Study Group. Hepatology 1994; 20(1 Pt 1):15–20. pmid:8020885
  24. Sterling RK, Lissen E, Clumeck N, et al; APRICOT Clinical Investigators. Development of a simple noninvasive index to predict significant fibrosis in patients with HIV/HCV coinfection. Hepatology 2006; 43(6):1317–1325. doi:10.1002/hep.21178
  25. Vallet-Pichard A, Mallet V, Nalpas B, et al. FIB-4: an inexpensive and accurate marker of fibrosis in HCV infection. comparison with liver biopsy and fibrotest. Hepatology 2007; 46(1):32–36. doi:10.1002/hep.21669
  26. Shah AG, Lydecker A, Murray K, Tetri BN, Contos MJ, Sanyal AJ; Nash Clinical Research Network. Comparison of noninvasive markers of fibrosis in patients with nonalcoholic fatty liver disease. Clin Gastroenterol Hepatol 2009; 7(10):1104–1112. doi:10.1016/j.cgh.2009.05.033
  27. McPherson S, Stewart SF, Henderson E, Burt AD, Day CP. Simple non-invasive fibrosis scoring systems can reliably exclude advanced fibrosis in patients with non-alcoholic fatty liver disease. Gut 2010; 59(9):1265–1269. doi:10.1136/gut.2010.216077
  28. Angulo P, Hui JM, Marchesini G, et al. The NAFLD fibrosis score: A noninvasive system that identifies liver fibrosis in patients with NAFLD. Hepatology 2007; 45(4):846–854. doi:10.1002/hep.21496
  29. Goh GB, Pagadala MR, Dasarathy J, et al. Clinical spectrum of non-alcoholic fatty liver disease in diabetic and non-diabetic patients. BBA Clin 2015; 3:141–145. doi:10.1016/j.bbacli.2014.09.001
  30. Tapper EB, Hunink MG, Afdhal NH, Lai M, Sengupta N. Cost-effectiveness analysis: risk stratification of nonalcoholic fatty liver disease (NAFLD) by the primary care physician using the NAFLD fibrosis score. PLoS One 2016; 11(2):e0147237. doi:10.1371/journal.pone.0147237
  31. Wai CT, Greenson JK, Fontana RJ, et al. A simple noninvasive index can predict both significant fibrosis and cirrhosis in patients with chronic hepatitis C. Hepatology 2003; 38(2):518–526. doi:10.1053/jhep.2003.50346
  32. Calès P, Lainé F, Boursier J, et al. Comparison of blood tests for liver fibrosis specific or not to NAFLD. J Hepatol 2009; 50(1):165–173. doi:10.1016/j.jhep.2008.07.035
  33. Imbert-Bismut F, Ratziu V, Pieroni L, Charlotte F, Benhamou Y, Poynard T; MULTIVIRC Group. Biochemical markers of liver fibrosis in patients with hepatitis C virus infection: a prospective study. Lancet 2001; 357(9262):1069–1075. doi:10.1016/S0140-6736(00)04258-6
  34. Shaheen AA, Wan AF, Myers RP. FibroTest and FibroScan for the prediction of hepatitis C-related fibrosis: a systematic review of diagnostic test accuracy. Am J Gastroenterol 2007; 102(11):2589–2600. doi:10.1111/j.1572-0241.2007.01466.x
  35. Smith JO, Sterling RK. Systematic review: non-invasive methods of fibrosis analysis in chronic hepatitis C. Aliment Pharmacol Ther 2009; 30(6):557–576. doi:10.1111/j.1365-2036.2009.04062.x
  36. Sebastiani G, Vario A, Guido M, Alberti A. Performance of noninvasive markers for liver fibrosis is reduced in chronic hepatitis C with normal transaminases. J Viral Hepat 2007; 15(3):212–218. doi:10.1111/j.1365-2893.2007.00932.x
  37. Poynard T, Morra R, Halfon P, et al. Meta-analyses of FibroTest diagnostic value in chronic liver disease. BMC Gastroenterol 2007; 7:40. doi:10.1186/1471-230X-7-40
  38. Carlson JJ, Kowdley KV, Sullivan SD, Ramsey SD, Veenstra DL. An evaluation of the potential cost-effectiveness of non-invasive testing strategies in the diagnosis of significant liver fibrosis. J Gastroenterol Hepatol 2009; 24(5):786–791. doi:10.1111/j.1440-1746.2009.05778.x
  39. Aubé C, Oberti F, Korali N, et al. Ultrasonographic diagnosis of hepatic fibrosis or cirrhosis. J Hepatol 1999; 30(3):472–478. pmid:10190731
  40. Di Lelio A, Cestari C, Lomazzi A, Beretta L. Cirrhosis: diagnosis with sonographic study of the liver surface. Radiology 1989; 172(2):389–392. doi:10.1148/radiology.172.2.2526349
  41. Wong VW, Chan HL. Transient elastography. J Gastroenterol Hepatol 2010; 25(11):1726–1731. doi:10.1111/j.1440-1746.2010.06437.x
  42. Arena U, Vizzutti F, Abraldes JG, et al. Reliability of transient elastography for the diagnosis of advanced fibrosis in chronic hepatitis C. Gut 2008; 57(9):1288–1293. doi:10.1136/gut.2008.149708
  43. Ziol M, Handra-Luca A, Kettaneh A, et al. Noninvasive assessment of liver fibrosis by measurement of stiffness in patients with chronic hepatitis C. Hepatology 2005; 41(1):48–54. doi:10.1002/hep.20506
  44. Wong VW, Vergniol J, Wong GL, et al. Diagnosis of fibrosis and cirrhosis using liver stiffness measurement in nonalcoholic fatty liver disease. Hepatology 2010; 51(2):454–462. doi:10.1002/hep.23312
  45. Sagir A, Erhardt A, Schmitt M, Häussinger D. Transient elastography is unreliable for detection of cirrhosis in patients with acute liver damage. Hepatology 2007; 48(2):592–595. doi:10.1002/hep.22056
  46. Mederacke I, Wursthorn K, Kirschner J, et al. Food intake increases liver stiffness in patients with chronic or resolved hepatitis C virus infection. Liver Int 2009; 29(10):1500–1506. doi:10.1111/j.1478-3231.2009.02100.x
  47. Castéra L, Foucher J, Bernard PH, et al. Pitfalls of liver stiffness measurement: a 5-year prospective study of 13,369 examinations. Hepatology 2010; 51(3):828–835. doi:10.1002/hep.23425
  48. Wong VW, Vergniol J, Wong GL, et al. Liver stiffness measurement using XL probe in patients with nonalcoholic fatty liver disease. Am J Gastroenterol 2012; 107(12):1862–1871. doi:10.1038/ajg.2012.331
  49. Fraquelli M, Rigamonti C, Casazza G, et al. Reproducibility of transient elastography in the evaluation of liver fibrosis in patients with chronic liver disease. Gut 2007; 56(7):968–973. doi:10.1136/gut.2006.111302
  50. de Franchis R; Baveno VI Faculty. Expanding consensus in portal hypertension: report of the Baveno VI Consensus Workshop: stratifying risk and individualizing care for portal hypertension. J Hepatol 2015; 63(3):743–752. doi:10.1016/j.jhep.2015.05.022
  51. Friedrich-Rust M, Wunder K, Kriener S, et al. Liver fibrosis in viral hepatitis: noninvasive assessment with acoustic radiation force impulse imaging versus transient elastography. Radiology 2009; 252(2):595–604. doi:10.1148/radiol.2523081928
  52. Yoneda M, Suzuki K, Kato S, et al. Nonalcoholic fatty liver disease: US-based acoustic radiation force impulse elastography. Radiology 2010; 256(2):640–647. doi:10.1148/radiol.10091662
  53. Bota S, Herkner H, Sporea I, et al. Meta-analysis: ARFI elastography versus transient elastography for the evaluation of liver fibrosis. Liver Int 2013; 33(8):1138–1147. doi:10.1111/liv.12240
  54. Attia D, Bantel H, Lenzen H, Manns MP, Gebel MJ, Potthoff A. Liver stiffness measurement using acoustic radiation force impulse elastography in overweight and obese patients. Aliment Pharmacol Ther 2016; 44(4):366–379. doi:10.1111/apt.13710
  55. Cui J, Heba E, Hernandez C, et al. Magnetic resonance elastography is superior to acoustic radiation force impulse for the diagnosis of fibrosis in patients with biopsy-proven nonalcoholic fatty liver disease: a prospective study. Hepatology 2016; 63(2):453–461. doi:10.1002/hep.28337
  56. Huwart L, Sempoux C, Vicaut E, et al. Magnetic resonance elastography for the noninvasive staging of liver fibrosis. Gastroenterology 2008; 135(1):32–40. doi:10.1053/j.gastro.2008.03.076
  57. Jajamovich GH, Dyvorne H, Donnerhack C, Taouli B. Quantitative liver MRI combining phase contrast imaging, elastography, and DWI: assessment of reproducibility and postprandial effect at 3.0 T. PLoS One 2014; 9(5):e97355. doi:10.1371/journal.pone.0097355
  58. Lim JK, Flamm SL, Singh S, Falck-Ytter YT; Clinical Guidelines Committee of the American Gastroenterological Association. American Gastroenterological Association Institute guideline on the role of elastography in the evaluation of liver fibrosis. Gastroenterology 2017; 152(6):1536–1543. doi:10.1053/j.gastro.2017.03.017
  59. N, Feldstein AE. Noninvasive diagnosis of nonalcoholic fatty liver disease: are we there yet? Metabolism 2016; 65(8):1087–1095. doi:10.1016/j.metabol.2016.01.013
References
  1. Younossi ZM, Stepanova M, Afendy M, et al. Changes in the prevalence of the most common causes of chronic liver diseases in the United States from 1988 to 2008. Clin Gastroenterol Hepatol 2011; 9(6):524–530.e1. doi:10.1016/j.cgh.2011.03.020
  2. Kochanek KD, Xu J, Murphy SL, Miniño AM, Kung H-C. Deaths: final data for 2009. Natl Vital Stat Rep 2011; 60(3):1–116. pmid:24974587
  3. Volk ML, Tocco RS, Bazick J, Rakoski MO, Lok AS. Hospital readmissions among patients with decompensated cirrhosis. Am J Gastroenterol 2012; 107(2):247–252. doi:10.1038/ajg.2011.314
  4. Vernon G, Baranova A, Younossi ZM. Systematic review: the epidemiology and natural history of non-alcoholic fatty liver disease and non-alcoholic steatohepatitis in adults. Aliment Pharmacol Ther 2011; 34(3):274–285. doi:10.1111/j.1365-2036.2011.04724.x
  5. Udompap P, Kim D, Kim WR. Current and future burden of chronic nonmalignant liver disease. Clin Gastroenterol Hepatol 2015; 13(12):2031–2041. doi:10.1016/j.cgh.2015.08.015
  6. Kim WR, Lake JR, Smith JM, et al. OPTN/SRTR 2016 annual data report: liver. Am J Transplant 2018; 18(suppl 1):172–253. doi:10.1111/ajt.14559
  7. Wong RJ, Aguilar M, Cheung R, et al. Nonalcoholic steatohepatitis is the second leading etiology of liver disease among adults awaiting liver transplantation in the United States. Gastroenterology 2015; 148(3):547–555. doi:10.1053/j.gastro.2014.11.039
  8. Ishak K, Baptista A, Bianchi L, et al. Histological grading and staging of chronic hepatitis. J Hepatol 1995; 22(6):696–699. pmid:7560864
  9. Bedossa P, Poynard T. An algorithm for the grading of activity in chronic hepatitis C. Hepatology 1996; 24(2):289–293. doi:10.1002/hep.510240201
  10. Kleiner DE, Brunt EM, Van Natta M, et al; Nonalcoholic Steatohepatitis Clinical Research Network. Design and validation of a histological scoring system for nonalcoholic fatty liver disease. Hepatology 2005; 41(6):1313–1321. doi:10.1002/hep.20701
  11. Everhart JE, Wright EC, Goodman ZD, et al; HALT-C Trial Group. Prognostic value of Ishak fibrosis stage: findings from the hepatitis C antiviral long-term treatment against cirrhosis trial. Hepatology 2010; 51(2):585–594. doi:10.1002/hep.23315
  12. Angulo P, Kleiner DE, Dam-Larsen S, et al. Liver fibrosis, but no other histologic features, is associated with long-term outcomes of patients with nonalcoholic fatty liver disease. Gastroenterology 2015; 149(2):389–397.e10. doi:10.1053/j.gastro.2015.04.043
  13. Lindor KD, Bru C, Jorgensen RA, et al. The role of ultrasonography and automatic-needle biopsy in outpatient percutaneous liver biopsy. Hepatology 1996; 23(5):1079–1083. doi:10.1002/hep.510230522
  14. Pasha T, Gabriel S, Therneau T, Dickson ER, Lindor KD. Cost-effectiveness of ultrasound-guided liver biopsy. Hepatology 1998; 27(5):1220–1226. doi:10.1002/hep.510270506
  15. Alessandria C, Debernardi-Venon W, Rizzetto M, Marzano A. Transjugular liver biopsy: a relatively simple procedure with an indefinite past and an expected brilliant future. J Hepatol 2008; 48(1):171–173. doi:10.1016/j.jhep.2007.10.001
  16. Kalambokis G, Manousou P, Vibhakorn S, et al. Transjugular liver biopsy—indications, adequacy, quality of specimens, and complications—a systematic review. J Hepatol 2007; 47(2):284–294. doi:10.1016/j.jhep.2007.05.001
  17. Ripoll C, Groszmann R, Garcia-Tsao G, et al; Portal Hypertension Collaborative Group. Hepatic venous pressure gradient predicts clinical decompensation in patients with compensated cirrhosis. Gastroenterology 2007; 133(2):481–488. doi:10.1053/j.gastro.2007.05.024
  18. Perrault J, McGill DB, Ott BJ, Taylor WF. Liver biopsy: complications in 1000 inpatients and outpatients. Gastroenterology 1978; 74(1):103–106. pmid:618417
  19. Janes CH, Lindor KD. Outcome of patients hospitalized for complications after outpatient liver biopsy. Ann Intern Med 1993; 118(2):96–98. pmid:8416324
  20. Bedossa P, Dargere D, Paradis V. Sampling variability of liver fibrosis in chronic hepatitis C. Hepatology 2003; 38(6):1449–1457. doi:10.1016/j.hep.2003.09.022
  21. Regev A, Berho M, Jeffers LJ, et al. Sampling error and intraobserver variation in liver biopsy in patients with chronic HCV infection. Am J Gastroenterol 2002; 97(10):2614–2618. doi:10.1111/j.1572-0241.2002.06038.x
  22. Goldin RD, Goldin JG, Burt AD, et al. Intra-observer and inter-observer variation in the histopathological assessment of chronic viral hepatitis. J Hepatol 1996; 25(5):649–654. pmid:8938541
  23. Intraobserver and interobserver variations in liver biopsy interpretation in patients with chronic hepatitis C. The French METAVIR Cooperative Study Group. Hepatology 1994; 20(1 Pt 1):15–20. pmid:8020885
  24. Sterling RK, Lissen E, Clumeck N, et al; APRICOT Clinical Investigators. Development of a simple noninvasive index to predict significant fibrosis in patients with HIV/HCV coinfection. Hepatology 2006; 43(6):1317–1325. doi:10.1002/hep.21178
  25. Vallet-Pichard A, Mallet V, Nalpas B, et al. FIB-4: an inexpensive and accurate marker of fibrosis in HCV infection. comparison with liver biopsy and fibrotest. Hepatology 2007; 46(1):32–36. doi:10.1002/hep.21669
  26. Shah AG, Lydecker A, Murray K, Tetri BN, Contos MJ, Sanyal AJ; Nash Clinical Research Network. Comparison of noninvasive markers of fibrosis in patients with nonalcoholic fatty liver disease. Clin Gastroenterol Hepatol 2009; 7(10):1104–1112. doi:10.1016/j.cgh.2009.05.033
  27. McPherson S, Stewart SF, Henderson E, Burt AD, Day CP. Simple non-invasive fibrosis scoring systems can reliably exclude advanced fibrosis in patients with non-alcoholic fatty liver disease. Gut 2010; 59(9):1265–1269. doi:10.1136/gut.2010.216077
  28. Angulo P, Hui JM, Marchesini G, et al. The NAFLD fibrosis score: A noninvasive system that identifies liver fibrosis in patients with NAFLD. Hepatology 2007; 45(4):846–854. doi:10.1002/hep.21496
  29. Goh GB, Pagadala MR, Dasarathy J, et al. Clinical spectrum of non-alcoholic fatty liver disease in diabetic and non-diabetic patients. BBA Clin 2015; 3:141–145. doi:10.1016/j.bbacli.2014.09.001
  30. Tapper EB, Hunink MG, Afdhal NH, Lai M, Sengupta N. Cost-effectiveness analysis: risk stratification of nonalcoholic fatty liver disease (NAFLD) by the primary care physician using the NAFLD fibrosis score. PLoS One 2016; 11(2):e0147237. doi:10.1371/journal.pone.0147237
  31. Wai CT, Greenson JK, Fontana RJ, et al. A simple noninvasive index can predict both significant fibrosis and cirrhosis in patients with chronic hepatitis C. Hepatology 2003; 38(2):518–526. doi:10.1053/jhep.2003.50346
  32. Calès P, Lainé F, Boursier J, et al. Comparison of blood tests for liver fibrosis specific or not to NAFLD. J Hepatol 2009; 50(1):165–173. doi:10.1016/j.jhep.2008.07.035
  33. Imbert-Bismut F, Ratziu V, Pieroni L, Charlotte F, Benhamou Y, Poynard T; MULTIVIRC Group. Biochemical markers of liver fibrosis in patients with hepatitis C virus infection: a prospective study. Lancet 2001; 357(9262):1069–1075. doi:10.1016/S0140-6736(00)04258-6
  34. Shaheen AA, Wan AF, Myers RP. FibroTest and FibroScan for the prediction of hepatitis C-related fibrosis: a systematic review of diagnostic test accuracy. Am J Gastroenterol 2007; 102(11):2589–2600. doi:10.1111/j.1572-0241.2007.01466.x
  35. Smith JO, Sterling RK. Systematic review: non-invasive methods of fibrosis analysis in chronic hepatitis C. Aliment Pharmacol Ther 2009; 30(6):557–576. doi:10.1111/j.1365-2036.2009.04062.x
  36. Sebastiani G, Vario A, Guido M, Alberti A. Performance of noninvasive markers for liver fibrosis is reduced in chronic hepatitis C with normal transaminases. J Viral Hepat 2007; 15(3):212–218. doi:10.1111/j.1365-2893.2007.00932.x
  37. Poynard T, Morra R, Halfon P, et al. Meta-analyses of FibroTest diagnostic value in chronic liver disease. BMC Gastroenterol 2007; 7:40. doi:10.1186/1471-230X-7-40
  38. Carlson JJ, Kowdley KV, Sullivan SD, Ramsey SD, Veenstra DL. An evaluation of the potential cost-effectiveness of non-invasive testing strategies in the diagnosis of significant liver fibrosis. J Gastroenterol Hepatol 2009; 24(5):786–791. doi:10.1111/j.1440-1746.2009.05778.x
  39. Aubé C, Oberti F, Korali N, et al. Ultrasonographic diagnosis of hepatic fibrosis or cirrhosis. J Hepatol 1999; 30(3):472–478. pmid:10190731
  40. Di Lelio A, Cestari C, Lomazzi A, Beretta L. Cirrhosis: diagnosis with sonographic study of the liver surface. Radiology 1989; 172(2):389–392. doi:10.1148/radiology.172.2.2526349
  41. Wong VW, Chan HL. Transient elastography. J Gastroenterol Hepatol 2010; 25(11):1726–1731. doi:10.1111/j.1440-1746.2010.06437.x
  42. Arena U, Vizzutti F, Abraldes JG, et al. Reliability of transient elastography for the diagnosis of advanced fibrosis in chronic hepatitis C. Gut 2008; 57(9):1288–1293. doi:10.1136/gut.2008.149708
  43. Ziol M, Handra-Luca A, Kettaneh A, et al. Noninvasive assessment of liver fibrosis by measurement of stiffness in patients with chronic hepatitis C. Hepatology 2005; 41(1):48–54. doi:10.1002/hep.20506
  44. Wong VW, Vergniol J, Wong GL, et al. Diagnosis of fibrosis and cirrhosis using liver stiffness measurement in nonalcoholic fatty liver disease. Hepatology 2010; 51(2):454–462. doi:10.1002/hep.23312
  45. Sagir A, Erhardt A, Schmitt M, Häussinger D. Transient elastography is unreliable for detection of cirrhosis in patients with acute liver damage. Hepatology 2007; 48(2):592–595. doi:10.1002/hep.22056
  46. Mederacke I, Wursthorn K, Kirschner J, et al. Food intake increases liver stiffness in patients with chronic or resolved hepatitis C virus infection. Liver Int 2009; 29(10):1500–1506. doi:10.1111/j.1478-3231.2009.02100.x
  47. Castéra L, Foucher J, Bernard PH, et al. Pitfalls of liver stiffness measurement: a 5-year prospective study of 13,369 examinations. Hepatology 2010; 51(3):828–835. doi:10.1002/hep.23425
  48. Wong VW, Vergniol J, Wong GL, et al. Liver stiffness measurement using XL probe in patients with nonalcoholic fatty liver disease. Am J Gastroenterol 2012; 107(12):1862–1871. doi:10.1038/ajg.2012.331
  49. Fraquelli M, Rigamonti C, Casazza G, et al. Reproducibility of transient elastography in the evaluation of liver fibrosis in patients with chronic liver disease. Gut 2007; 56(7):968–973. doi:10.1136/gut.2006.111302
  50. de Franchis R; Baveno VI Faculty. Expanding consensus in portal hypertension: report of the Baveno VI Consensus Workshop: stratifying risk and individualizing care for portal hypertension. J Hepatol 2015; 63(3):743–752. doi:10.1016/j.jhep.2015.05.022
  51. Friedrich-Rust M, Wunder K, Kriener S, et al. Liver fibrosis in viral hepatitis: noninvasive assessment with acoustic radiation force impulse imaging versus transient elastography. Radiology 2009; 252(2):595–604. doi:10.1148/radiol.2523081928
  52. Yoneda M, Suzuki K, Kato S, et al. Nonalcoholic fatty liver disease: US-based acoustic radiation force impulse elastography. Radiology 2010; 256(2):640–647. doi:10.1148/radiol.10091662
  53. Bota S, Herkner H, Sporea I, et al. Meta-analysis: ARFI elastography versus transient elastography for the evaluation of liver fibrosis. Liver Int 2013; 33(8):1138–1147. doi:10.1111/liv.12240
  54. Attia D, Bantel H, Lenzen H, Manns MP, Gebel MJ, Potthoff A. Liver stiffness measurement using acoustic radiation force impulse elastography in overweight and obese patients. Aliment Pharmacol Ther 2016; 44(4):366–379. doi:10.1111/apt.13710
  55. Cui J, Heba E, Hernandez C, et al. Magnetic resonance elastography is superior to acoustic radiation force impulse for the diagnosis of fibrosis in patients with biopsy-proven nonalcoholic fatty liver disease: a prospective study. Hepatology 2016; 63(2):453–461. doi:10.1002/hep.28337
  56. Huwart L, Sempoux C, Vicaut E, et al. Magnetic resonance elastography for the noninvasive staging of liver fibrosis. Gastroenterology 2008; 135(1):32–40. doi:10.1053/j.gastro.2008.03.076
  57. Jajamovich GH, Dyvorne H, Donnerhack C, Taouli B. Quantitative liver MRI combining phase contrast imaging, elastography, and DWI: assessment of reproducibility and postprandial effect at 3.0 T. PLoS One 2014; 9(5):e97355. doi:10.1371/journal.pone.0097355
  58. Lim JK, Flamm SL, Singh S, Falck-Ytter YT; Clinical Guidelines Committee of the American Gastroenterological Association. American Gastroenterological Association Institute guideline on the role of elastography in the evaluation of liver fibrosis. Gastroenterology 2017; 152(6):1536–1543. doi:10.1053/j.gastro.2017.03.017
  59. N, Feldstein AE. Noninvasive diagnosis of nonalcoholic fatty liver disease: are we there yet? Metabolism 2016; 65(8):1087–1095. doi:10.1016/j.metabol.2016.01.013
Issue
Cleveland Clinic Journal of Medicine - 86(3)
Issue
Cleveland Clinic Journal of Medicine - 86(3)
Page Number
179-186
Page Number
179-186
Publications
Publications
Topics
Article Type
Display Headline
Assessing liver fibrosis without biopsy in patients with HCV or NAFLD
Display Headline
Assessing liver fibrosis without biopsy in patients with HCV or NAFLD
Legacy Keywords
liver, fibrosis, nonalcoholic fatty liver disease, NAFLD, nonalcoholic steatohepatitis, NASH, cirrhosis, hepatitis C virus, HCV, biopsy, staging, Ishak, METAVIR, FIB-4 index, NAFLD fibrosis score, AST-to-platelet raio index, APRI, FibroSure, ultrasonography, transient elastography, acoustic radiation force imaging, liver stiffness measurement, magnetic resonance elastography, Tavankit Singh, Daniela Allende, Arthur McCullough
Legacy Keywords
liver, fibrosis, nonalcoholic fatty liver disease, NAFLD, nonalcoholic steatohepatitis, NASH, cirrhosis, hepatitis C virus, HCV, biopsy, staging, Ishak, METAVIR, FIB-4 index, NAFLD fibrosis score, AST-to-platelet raio index, APRI, FibroSure, ultrasonography, transient elastography, acoustic radiation force imaging, liver stiffness measurement, magnetic resonance elastography, Tavankit Singh, Daniela Allende, Arthur McCullough
Sections
Inside the Article

KEY POINTS

  • Liver biopsy remains the gold standard for determining fibrosis stage but is expensive and entails risk of complications.
  • For patients infected with HCV, fibrosis stage should be determined with transient elastography, a transthoracic ultrasonographic technique that measures shear-wave velocity.
  • For patients with cirrhosis, transient elastography combined with a platelet count can detect developing portal hypertension and determine whether to screen for esophageal varices.
  • For NAFLD, combined elastography and NAFLD fibrosis score—which incorporates patient characteristics and laboratory test results—should be used to determine the need for liver biopsy.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Tue, 02/26/2019 - 14:30
Un-Gate On Date
Tue, 02/26/2019 - 14:30
Use ProPublica
CFC Schedule Remove Status
Tue, 02/26/2019 - 14:30
Hide sidebar & use full width
render the right sidebar.
Article PDF Media